78
2017 UNIVERSIDADE DE LISBOA FACULDADE DE CIÊNCIAS DEPARTAMENTO DE BIOLOGIA VEGETAL Genomic deletions in Escherichia coli K-12 MG1655 for plasmid DNA production Diana Tamára Vaz Cipriano Mestrado em Microbiologia Aplicada Dissertação orientada por: Prof. Doutor Francisco Dionísio Prof. Doutor Gabriel Monteiro

Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

Embed Size (px)

Citation preview

Page 1: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

2017

UNIVERSIDADE DE LISBOA

FACULDADE DE CIÊNCIAS

DEPARTAMENTO DE BIOLOGIA VEGETAL

Genomic deletions in Escherichia coli K-12 MG1655 for

plasmid DNA production

Diana Tamára Vaz Cipriano

Mestrado em Microbiologia Aplicada

Dissertação orientada por:

Prof. Doutor Francisco Dionísio

Prof. Doutor Gabriel Monteiro

Page 2: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

Genomic deletions in Escherichia coli K-12 MG1655 for

plasmid DNA production

Diana Tamára Vaz Cipriano

2017

This thesis was fully performed at the Institute of Bioengineering and Biosciences in Instituto

Superior Técnico under the direct supervision of Professor Dr. Gabriel Monteiro in the scope of

the Master in Applied Microbiology of the Faculty of Sciences of the University of Lisbon.

Page 3: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

“Failure is not an option” – Eugene Francis Kranz

Page 4: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

iv

Acknowledgment

I would first like to thank my thesis extern supervisor Professor Doctor Gabriel Monteiro of the

Instituto Superior Técnico at Universidade de Lisboa, for having accepted me in your group, iBB

group. The door to Prof. Gabriel Monteiro office was always open whenever I ran into a trouble

spot or had a question about my research or writing. He consistently allowed this paper to be my

own work, but steered me in the right the direction whenever he thought I needed it.

I would also like to thank my intern supervisor Professor Doctor Francisco Dionísio for having

accepted to be my supervisor.

To the people of the iBB group, my sincere thanks, for all the sharing of knowledge at each lab

meeting, and for all the fun times.

Sofia Duarte, thank you so much for all the professional and personal teachings. You were,

without doubt, a great guide, who allowed me to concretize this project. Furthermore, thank you

for the sincere and fun friendship that we have created. You are the best.

Maria Martins and Cláudia Alves, thank you so much for your participation in this project, for

your help in my doubts and for all teachings. Thank you so much for your friendship.

Marisa Santos, thank you so much for all teachings about HPLC, was a good experience.

Thanks to Professora Leonilde Moreira and to Inês Silva, for all clarifications regarding

sequencing of strains.

Also, to Ricardo Pereira for his work and dedication to the 7th floor and Rosa Gonçalves for the

sympathy, affection and for maintaining the lab material clean and organized.

Soraia Guerreiro, one more time, we are together in this stage. Thank you for all your support and

help in hard moments.

The most important thanks, I must express my very profound gratitude to my parents, my brother

and all my family for providing me with unfailing support and continuous encouragement

throughout my years of study and through the process of researching and writing this thesis.

Thank you for hanging on to my unwanted absence. This accomplishment would not have been

possible without them. Thank you.

Last but not least, my friends. Thank you so much to the friends who accompanied me and

supported me in this stage of my life. The names are not important, the quantity much less. It

matters that I have the best with me.

Now it is time to celebrate.

Page 5: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

v

Abstract

The interest in plasmid DNA (pDNA) as a biopharmaceutical has been increasing over the last

few years, especially after the approval of the first DNA vaccines. Gene therapy and DNA

vaccines represent a promise in the treatment of diseases like viral infections, genetic and acquired

diseases. From the point of view of manufacture, it is important to establish a profitable process

with a low cost-production relation. So, over the last years the investigation has optimized and

improved the production conditions as well as genetically modifying some bacterial strains to turn

the production of hypothetical vectors profitable.

pDNA vectors offer considerable benefits over viral systems in gene-based therapy and

vaccination applications, including higher shelf stability, low immunogenicity and toxicity, and

simple manufacture in large scale. However, also shows low transfection efficacy, and requires

milligram scale of pharmaceutical-grade pDNA per patient, which implies extensive production

efforts. In order to fulfil high pDNA production requirements, genetically engineered strains

specially designed to achieve high pDNA yields are required.

This master thesis was a follow up on a previous work aiming at genetically modifying

Escherichia coli strains. To explore the effect of strain genetic background, a new pDNA

production strain, mutated in of three genes pgi, endA and recA (GALG20), was previously

constructed. However, as an unintentional genomic deletion of the 20 kb, derived from rac

prophage, appeared in GALG20, a new derivative from MG1655 deleted in pgi, endA and recA

genes was constructed (GALGNEW) using λ-Red System described by Datsenko and Wanner

[1]. This new strain was compared with GALG20, and with the wild-type strain MG1655.

However, contrary to expected, GALG20 and GALGNEW strains show slight differences in

growth kinetics (31.4 ± 2.0 and 32.6 ± 2.7, respectively) and plasmid production yields [169.2 ±

14.3 (mg/ L) and 204.3 ± 44.3 (mg/ L), respectively]. As expected, these two mutant strains show

higher volumetric and specific plasmid production yields, when comparing with E. coli MG1655.

An additional goal of this work was to construct zwf deletion mutants of GALGNEW, using the

CRISPR- Cas9 System instead of the λ-Red method. This last goal was not concluded.

Keywords:

Gene therapy

DNA vaccination

Plasmid DNA

Strain engineering

Escherichia coli

Page 6: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

vi

Resumo

A terapia genética e as vacinas de DNA representam um avanço no tratamento de doenças como

infeções virais, doenças genéticas e adquiridas. Do ponto de vista de produção, é importante

estabelecer um processo rentável com uma relação custo-produção reduzida. Assim, nos últimos

anos a investigação tem otimizado e melhorado as condições de produção, bem como modificado

geneticamente algumas estirpes bacterianas para tornar rentável a produção de vetores

hipotéticos.

Os vetores de DNA plasmídico (pDNA) proporcionam benefícios consideráveis em relação aos

sistemas virais em aplicações de terapia e vacinação baseadas em genes, incluindo maior

estabilidade, baixa imunogenicidade e toxicidade e produção simples em grande escala. No

entanto, também mostra baixa eficácia de transfeção e requer o uso à escala de miligramas de

pDNA de qualidade farmacêutica por paciente, o que implica esforços de produção extensivos. A

fim de satisfazer os requisitos de produção de pDNA elevada, são necessárias estirpes

geneticamente modificadas especialmente concebidas para alcançar rendimentos elevados na

produção de pDNA.

Esta dissertação de mestrado deu continuidade a um trabalho anterior com o objetivo de modificar

geneticamente estirpes de Escherichia coli (E. coli) MG1655 através da deleção de genes alvo

(pgi, endA e recA), recorrendo ao método λ-Red descrito por Datsenko e Wanner, e

posteriormente, comparar os seus padrões de cinética de crescimento com a estirpe original.

Os genes selecionados para deleção desempenham diferentes funções no metabolismo de E. coli

estando diretamente relacionados com a produção de nucleótidos e produção de DNA plasmídico.

O gene pgi é responsável por codificar a enzima fosfoglucose isomerase, a qual catalisa a

conversão da glucose-6-fosfato em frutose-6-fosfato, correspondendo à primeira etapa da

glicólise. A deleção deste gene conduz a um redireccionamento do fluxo de carbono para a via

dos fosfatos de pentose, o que leva a um aumento da síntese de nucleótidos (R5P e E4P),

necessários para a síntese de DNA plasmídico. Esta alteração fornece ainda elevadas quantidades

de poder redutor (NADPH). O gene endA é responsável por codificar uma endonuclease não

especifica, enquanto o gene recA participa no sistema de reparação da estirpe por recombinação

homóloga de sequências. A deleção dos genes endA e recA minimiza a digestão inespecífica do

DNA plasmídico bem como a sua recombinação. O resultado da deleção dos três genes descritos

anteriormente foi a criação da estirpe GALGNEW. Esta estirpe foi comparada com a estirpe

mutante previamente criada, GALG20, bem como com a estirpe original MG1655. Antes de

realizados os ensaios de crescimento, todas as estirpes foram transformadas com o plasmídeo

pVAX1GFP.

Neste projeto foi investigada a natureza e a frequência da deleção genómica não intencional de

20 kb, derivada do prófago rac. Esta deleção surge quando estirpes de E. coli K-12 MG1655 são

deletadas no gene pgi, usando o método λ-Red descrito por Datsenko e Wanner. Este fenómeno

foi verificado após a criação da estirpe GALG20 por sequenciação, não tendo sido encontrada

nenhuma leitura de amplificação “read” no genoma da estirpe naquele local. Através de métodos

de análise molecular, nomeadamente: reação em cadeia da polimerase (PCR) e sequenciação de

nova geração (NGS) MiSeq®, a presença da sequência rac foi confirmada no genoma da nova

estirpe GALGNEW. As reações de PCR realizadas envolveram pares de oligonucleótidos

sintetizados a montante, a jusante e na própria região de 20 kb.

Page 7: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

vii

Tal como descrito por Liu et al. (2015), a presença de genes de prófagos fornece múltiplos

benefícios ao hospedeiro, afim deste sobreviver em ambientes com condições adversas, sendo

exemplos: sob stresses oxidativo, ácido, osmótico e sob stress causado pela presença de

antibióticos no meio de crescimento. A deleção da sequência rac conduz a um decréscimo da

resistência do hospedeiro aos stresses mencionados, tornando a estirpe mais suscetível.

Um objetivo adicional deste trabalho foi a construção de uma nova estirpe deletada no gene zwf

a partir da estirpe GALGNEW, aplicando um método designado CRISPR- Cas9 descrito por

Reish e Prather. Este último objetivo não foi concluído.

Diversas diferenças foram encontradas entre os dois métodos de deleção de genes. Contrariamente

ao método λ-Red descrito por Datsenko e Wanner, este novo método (CRISPR-Cas9) não requer

a síntese por PCR de uma sequência portadora do gene que fornece resistência ao antibiótico

canamicina, designada cassete de canamicina, e são utilizados três plasmídeos para

transformação. O método CRISPR-Cas9 tem por base a transformação da estirpe hospedeira com

um plasmídeo que carrega o gene cas9, o qual codifica uma endonuclease responsável por fazer

um “nick” em cadeia simples. O local onde a enzima corta é específico e definido pela escolha de

uma região composta por um tripleto (NGG) designada PAM, a qual se localiza na região do gene

a deletar. No fim de confirmada a deleção do gene alvo, a estirpe é transformada com um terceiro

plasmídeo (pKDsg-p15) para curar o primeiro (pCas9cr4). Contudo, existem características

comuns a estes dois métodos, sendo exemplos: (a) a presença dos genes λ-Red (exo, bet e gam),

introduzidos num segundo plasmídeo (pKD46 no método λ-Red e pKDsg-xxx método CRISPR-

Cas9) e que vão permitir a recombinação entre o fragmento de interesse e o genoma da estirpe

hospedeira e (b) a termossensibilidade do segundo plasmídeo.

Relativamente ao gene zwf, este é responsável por codificar a enzima glucose-6-fosfato

desidrogenase (G6PDH), sendo esta fundamental no metabolismo central uma vez que se encontra

envolvida na divisão do carbono entre a glicólise e a via dos fosfatos de pentose. Com a deleção

deste gene é esperado uma restruturação do fluxo de carbono de E. coli para vias alternativas.

Apresentando dados mais concretos, as estirpes mutadas no gene zwf conseguem direcionar

98.9% e 87.0% do fluxo total de carbono através da primeira etapa da glicólise e do ciclo dos

ácidos tricarboxílicos (pela conversão de acetil-coenzima A em citrato). Quando feita a mesma

análise na estirpe original não mutada, verifica-se que o fluxo de carbono é menor na primeira

etapa da glicólise (78.6%) e na mesma etapa do ciclo dos ácidos tricarboxílicos (73.1%).

Todavia, as estirpes GALG20 e GALGNEW apresentam ligeiras diferenças ao nível de cinética

de crescimento (31.4 ± 2.0 and 32.6 ± 2.7, respectivamente) e de rendimento de produção de

plasmídeos [169.2 ± 14.3 (mg/ L) e 204.3 ± 44.3 (mg/ L), respetivamente]. Tal como era

expectável, estas duas estirpes mutantes mostram rendimentos volumétricos e de produção de

plasmídeo mais elevados, quando comparadas com a estirpe original E. coli MG1655.

Palavras-chave:

Terapia genética, Vacinas de DNA, DNA plasmídico, Engenharia de estirpes, Escherichia coli

Page 8: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

viii

Page 9: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

9

Contents

ACKNOWLEDGMENT ____________________________________________________________ IV

ABSTRACT ________________________________________________________________________ V

RESUMO __________________________________________________________________________ VI

LIST OF FIGURES ________________________________________________________________ 11

LIST OF ABBREVIATIONS ________________________________________________________ 15

THESIS MOTIVATION ____________________________________________________________ 16

1. INTRODUCTION____________________________________________________________ 17

1.1. Escherichia coli: The host strain _____________________________________________ 17 1.2. Plasmid DNA and Therapeutic Applications ___________________________________ 20

1.2.1. Types of Vaccines: Evolution and characteristics _____________________________ 21 1.2.2. Design of Plasmids for Gene Therapy and Vaccination ________________________ 23 1.2.3. Plasmid Structural Stability ______________________________________________ 26

1.2.3.1 Plasmid Size _________________________________________________________________ 27 1.2.3.2 DNA structure ________________________________________________________________ 28

1.2.4. Stability in Replication Process ___________________________________________ 28 1.2.5. Advantages of DNA vaccines ____________________________________________ 29 1.2.6. pVAX1-GFP plasmid ___________________________________________________ 29

1.3. Effect of plasmid DNA synthesis on E. coli central carbon metabolism ______________ 30 1.4. Relevant genes for E. coli strain engineering aiming to increase pDNA production _____ 31

2. MATERIALS AND METHODS ________________________________________________ 33

2.1. Media, Chemicals and Other Reagents ________________________________________ 33 2.2. Preparation of Competent cells_______________________________________________ 34

2.2.1. Electrocompetent cells __________________________________________________ 34 A. Transformation by electroporation_____________________________________________________ 34

2.2.2. Chemical competent cells ________________________________________________ 35 B. Transformation by heat shock ________________________________________________________ 35

2.3. Red Disruption System _____________________________________________________ 36 2.3.1. Strains and plasmids ____________________________________________________ 36 2.3.2. Oligonucleotides _______________________________________________________ 37 2.3.3. Generation of kanamycin cassette _________________________________________ 39 2.3.4. Gene disruption strategy _________________________________________________ 40

2.4. CRISPR Cas9-System method _______________________________________________ 42 2.4.1. Strains and plasmids ____________________________________________________ 42 2.4.2. Oligonucleotides _______________________________________________________ 43 2.4.3. Plasmid construction and protospacer design _________________________________ 43 2.4.4. Gene disruption strategy _________________________________________________ 46

2.5. Gel extraction and purification ________________________________________________ 46 2.6. Plasmid DNA purification ____________________________________________________ 47 2.7. Plasmid DNA restriction _____________________________________________________ 47 2.8. General PCR parameters _____________________________________________________ 48 2.9. Agarose electrophoresis ______________________________________________________ 48 2.10. Shake flask cultivation _______________________________________________________ 48 2.11. Measurement of glucose ______________________________________________________ 49 2.12. Plasmid DNA quantification __________________________________________________ 49 2.13. Cells banks preparations _____________________________________________________ 49 2.14. Genomic deletion analysis ____________________________________________________ 50 2.15. DNA sequencing ____________________________________________________________ 51

Page 10: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

10

3. RESULTS AND DISCUSSION _________________________________________________ 52

Plasmid DNA profile _________________________________________________________ 52 3.2. Knockouts by Red Disruption System _________________________________________ 54

3.2.1. Kanamycin cassette generation ______________________________________________ 54 3.2.2. pgi gene knockout ________________________________________________________ 55 3.2.3. recA gene knockout ______________________________________________________ 57 3.2.4. fnr and ralR genes ________________________________________________________ 59

3.3. CRISPR Cas9- System method _______________________________________________ 64 3.3.1. Construction of plasmid pKDsg-zwf _________________________________________ 64

3.4. Shake flask cultivation ______________________________________________________ 65 3.5. Measurement of glucose and plasmid DNA quantification _________________________ 68 3.6. Genomic deletion analysis____________________________________________________ 71 3.7. DNA sequencing results ____________________________________________________ 73

4. CONCLUSION AND FUTURE WORK _____________________________________________ 74

5. REFERENCES __________________________________________________________________ 75

Page 11: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

11

List of Figures

Figure 1. Phylogenetic tree based on glucose-6-phosphate isomerase encoded by pgi gene. The selected

sequences were aligned by Muscle Alignment Software. The topology was inferred using the Neighbor-

Joining method and the evolutionary distances were computed using the Poisson correction method

implemented in the software MEGA 7.0.18. The orange square ( ) in center represent the root of the

circular tree, the red circular forms ( ), the green squares ( ) and the yellow triangle ( ) represent 3 of

15 orders of class Gammaproteobacteria: Enterobacteriales, Vibrionales and Pseudomonadales,

respectively. _______________________________________________________________________ 17 Figure 2. E. coli K-12 and derivatives: creation of new strains and relationship between different strains.

Lineage of MG1655 and W3110, close relatives of wild-type E.coli K-12 (green box). Generation of

strains containing multiple mutations from MC1061, DH1 and JM101. Dark boxes represent commonly

used E. coli strains for plasmid DNA production and recent developments in E. coli strains designed for

high yield pDNA processes. Full line arrows represent the relationship between the strains and dashed

line arrows represent mutations carried from one strain to the other. Schematic representation adapted

from [9]–[11]. ______________________________________________________________________ 19 Figure 3. Indications addressed by gene therapy clinical trials. Data updated in August 2016 [19]. ___ 20 Figure 4. Geographical distribution of gene therapy clinical trials by continent. Data updated in August

2016 [19]. _________________________________________________________________________ 21 Figure 5. The various vaccine technologies developed over time. _____________________________ 22 Figure 6. Vectors used in Gene Therapy clinical trials. Data updated in August 2016 [19]. _________ 23 Figure 7. The main stages for pDNA vaccine design, production and vaccination. Adapted from [12],

[18], [32]. _________________________________________________________________________ 24 Figure 8. Genetic elements of a pDNA vector. The plasmid consists of a Plasmid Propagation Unit (PPU)

that operate in the microbial host and a Eukaryotic Expression Unit (EEU) that drives the protein

synthesis in the eukaryotic cells [28]. ____________________________________________________ 25 Figure 9. Ranking of alternative plasmid selection approaches according to plasmid size and

transformation efficiency. Adapted from [38]. _____________________________________________ 27 Figure 10. The Central Carbohydrate Metabolic Network [85]. _______________________________ 30 Figure 11. Schematic map of the plasmids used in Red system. (A) plasmid pKD13 (image created in

SnapGene® software) [5], (B) plasmid pKD46 [86] and (C) plasmid pCP20 [7].__________________ 36 Figure 12. Schematic representation of construction of the primers: to generate kanamycin cassette

(forward and reverse primers) and to confirm the insertion of the kanamycin cassette (primers check)

[51]. ______________________________________________________________________________ 38 Figure 13. Kanamycin resistance cassette generated by PCR: Schematic representation created in the

SnapGene software [36]. ______________________________________________________________ 39 Figure 14. Gene disruption strategy. H1 and H2 are the homology extensions or regions, P1 and P2 are

the priming sites. Strategy described by Datsenko and Wanner [53]. ___________________________ 40 Figure 15. Schematic map of the no-SCAR plasmids. (A) Plasmid pCas9cr4. (B) Plasmid pKDsg-xxx

[54]. ______________________________________________________________________________ 42 Figure 16. A schematic diagram of the proposed CPEC mechanism for cloning an individual gene. The

fragment 1 (orange line) and the fragment 2 (blue line) share overlapping regions at the ends. The

hybridized fragments extend using each other as a template until they complete a full circle (black line)

and reach their own 5’-ends. The assembled plasmid has two nicks, one on each strand. They can be used

for transformation with or without further purification. Adapted from [71], [87]. _________________ 44 Figure 17. Schematic representation of main steps of no-SCAR method. Adapted from [54]. _______ 46 Figure 18. Schematic representation of location of the oligonucleotides in the genome of MG1655 strain.

__________________________________________________________________________________ 50

Figure 19. Agarose gel expressing the band profile of plasmid pKD13 digested with BglII (lane 1) and

non- digested (lane 2).________________________________________________________________ 52 Figure 20. Qualitative analysis of plasmid pKD46: Plasmid DNA isoforms [88] and agarose gel analysis

of restriction digestion reactions of plasmid pKD46. Lane 1 – purified plasmid digested with EcoRI, lane

2 - purified plasmid digested with BamHI, lane 3 - purified plasmid undigested. The last lane (M)

corresponds to molecular weight marker NZYDNA ladder III. ________________________________ 53

Page 12: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

12

Figure 21. Agarose gel analysis of digestion reactions of pKDsg-ack, pKDsg-p15 and pCas9cr4 plasmids

with restriction enzymes. The first lane (M) is molecular weight marker ladder III. Lane 1 is a plasmid

pKDsg-ack non-digested, lane 2 is plasmid pKDsg-p15 digested with HindIII, lane 3 is plasmid pKDsg-

p15 non-digested, lane 4 is pKDsg-ack digested with HindIII, lane 5 is plasmid pCas9cr4 non-digested

and lane 6 is plasmid pCas9cr4 digested with BamHI. ______________________________________ 53 Figure 22. Agarose gel obtained from the PCR using to generate the KanR cassette for recA gene

knockout. In lane (1) PCR product and in lane (M) molecular weight marker NZYDNA Ladder III. __ 55 Figure 23. Agarose gel electrophoresis showing the result of colony PCR of strain MG1655 ΔendA::kan.

In the first lane (M) is molecular weight marker NZYDNA I from NZYTech. The lanes 1-6 correspond to

different six colonies analyzed. Lane 7 corresponds to a negative control performed without DNA. ___ 55 Figure 24. Agarose gel obtained from final colony PCR used to verify the knockout mutants and the

removal of the KanR cassette. In the first lane (M) is molecular weight marker NZYDNA Ladder III. In

the following lanes (1- 4) are the different colonies analized. The last lane correspond to the negative

control. ___________________________________________________________________________ 56 Figure 25. Agarose gel analysis of colony PCR to confirm the endA and pgi genes knockouts using check

primers. In the first lane (M) is molecular weight marker NZYDNA Ladder III. Lanes 1 and 2

corresponding to colonies analyzed with primers to check pgi gene knockout. Lane 3 is the positive

control. Lane 4 is the negative control using primers check for pgi gene knockout. Lanes 5 and 6

corresponding to colonies analyzed with primers to check endA gene knockout. Lane 7 is the positive

control. Lane 8 is the negative control using primers check for endA gene knockout. ______________ 57 Figure 26. Agarose gel with the amplified fragments from colony PCR to confirm the insertion of KanR

cassette. In first lane (M) is the molecular weight molecular NZYDNA ladder III and in following lanes

are the PCR products. The lanes 1-6 correspond to the colonies chosen from LB+ kan plate. In the lanes 7

and 8 are the positive control and the negative control, respectively. ___________________________ 58 Figure 27. Agarose gel analysis of colony PCR to confirm the recA gene disruption. The lanes 1 -6 are

the PCR products amplified using check primers for recA gene. The lane 7 correspond to the negative

control of the reaction. The last lane (M) is molecular weight marker NZYDNA ladder III. _________ 59

Figure 28. Agarose gel analysis of colony PCR to confirm the insertion of KanR cassette in MG1655

∆endA + pKD46 cells and to test the presence of rac. _______________________________________ 60 Figure 29. Agarose gel analysis of colony PCR to assess the cure of the plasmids and consequently

deletion of pgi gene in MG1655 ∆endAΔpgi cells and to test the presence of rac. _________________ 61 Figure 30. Agarose gel analyses of PCR reaction in order to test the presence of the rac sequence using

primers for fnr and ralR genes. In the first lane (M) is molecular weight marker ladder III. The lanes 1

and 2 are the amplified products with primers for the fnr gene. The lane 3 is the negative control prepared

with water and primers for fnr gene. The lanes 4 and 5 are the amplified products using primers for the

ralR gene. The lane 6 is the negative control prepared with water and primers for ralR gene. ________ 62 Figure 31. Agarose gel analyses of PCR reaction in order to test the presence of the rac sequence using

primers for fnr and ralR genes. _________________________________________________________ 62 Figure 32. Agarose gel analyses of PCR reaction in order to test the presence of the rac sequence, using

primers for fnr and ralR genes, and to assess the cure of the plasmids and consequently deletion of recA

gene. _____________________________________________________________________________ 63 Figure 33. Agarose gel analyses of PCR reaction in order to test the presence of the rac sequence, using

primers for ralR (A) and fnr (B) genes. __________________________________________________ 64 Figure 34. Agarose gel analyses: (A) result of PCR reaction to generate, separately, two fragments that

constitute the pKDsg-zwf plasmid. In the first lane (M) is molecular weight marker ladder III. The lanes 1

and 2 are the amplified products. (B) After CPEC reaction, the pKDsg-zwf was transformed into DH5α

cells, purified and quantified. 1,000 ng of purified plasmid was analized in 1% agarose gel. In the first

lane (M) is molecular weight marker ladder III. The lane 1 is the amplified product corresponding to the

pKDsg-zwf plasmid. _________________________________________________________________ 65 Figure 35. Effect of endA, recA and pgi genes knockout on biomass production and variation of medium

pH, following three strategies of shake flask cultivations (C1, C2 and C3). (A) Biomass produced in

GALG20 and GALGNEW strains following strategy C1. Optical density was measured at 0 h, 4 h, 8 h,

10 h and 24 h of growth. Plots depict mean values ± standard error of mean (SEM) of the four

independent experiments. (B) Variation of medium pH during growth of GALG20 and GALGNEW

strains following strategy C1. pH was measured at 0 h, 4 h, 8 h, 10 h and 24 h of growth. Plots depict

mean values ± standard error of mean (SEM) of the four independent experiments. (C) Biomass produced

Page 13: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

13

in GALG20 and GALGNEW strains following strategy C2. Optical density was measured at 24 h of

growth, during six days. (D) Variation of medium pH during growth of GALG20 and GALGNEW strains

following strategy C2. pH was measured at 24 h of growth. Plots depict mean values ± standard error of

mean (SEM) of the six independent experiments. (E) Biomass produced in GALG20, GALGNEW and

MG1655 strains following strategy C3. Optical density was measured at 0 h, 4 h, 8 h, 10 h, 17 h and 24 h

of growth. Plots depict mean values ± standard error of mean (SEM) of the one independent experiment.

(F) Variation of medium pH during growth of GALG20, GALGNEW and MG1655 strains following

strategy C3. pH was measured at 24 h of growth. Plots depict mean values ± standard error of mean

(SEM) of the one independent experiment. _______________________________________________ 67 Figure 36. Results of the quantification of glucose consumption throughout the growth versus biomass

(OD600 nm) for GALG20 and GALGNEW. The presented results derived from average values of 4 days

of growth. Glucose concentration was measured in duplicates by HPLC. ________________________ 69 Figure 37. Quantification of plasmid DNA yield volumetric (g/L) using two pgi mutant strains: GALG20

and GALGNEW grown in glucose, following strategy C1. Strains were grown for 24 h in shake flasks (37

°C, 250 rpm) with rich medium supplemented with 20 g/L of glucose. Plots depict mean values ±

standard error of mean (SEM) of the four independent experiments. ___________________________ 69 Figure 38. Quantification of plasmid DNA yield volumetric (g/L) using two pgi mutant strains: GALG20

and GALGNEW, and wild-type strain: MG1655, grown in glucose, following strategy C3. Strains were

grown for 24 h in shake flasks (37 °C, 250 rpm) with rich medium supplemented with 20 g/L of glucose.

Plots depict mean values of the one independent experiment. _________________________________ 70 Figure 39. Agarose gel analyses of PCR reaction to screen for the genomic deletion in GALG20 strain.

The ninth lane (L) is molecular weight marker ladder III. Lanes 1 -8 are amplified DNA from fresh

colony of one subculture. Lanes 9 -16 are amplified DNA from fresh colony of twelve subcultures. Lanes

17- 24 are negative controls prepared without DNA. Lane 25 is a positive control prepared with primers

for recA gene. ______________________________________________________________________ 71 Figure 40. Output of genomic DNA sequencing of mutant strains. Analysis of genomic deletion

sequence. Top figure: Segment of genomic DNA of GALGNEW strain. Bottom figure: Segment of

genomic DNA of GALG20 strain. ______________________________________________________ 73

Page 14: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

14

List of Tables

Table 1. New generation of vaccines: main characteristics. __________________________________ 22 Table 2. Overview of major factors affecting plasmid structural stability. Adapted from [39]. _______ 27 Table 3. Plasmid pVAX1-GFP: elements and their purposes. The schematic representation was created

with the SnapGene software. __________________________________________________________ 29 Table 4. Plasmid pVAX1-GFP and their main characteristics. ________________________________ 30 Table 5. Strains used in study and main characteristics. _____________________________________ 36

Table 6. Plasmids used in this work and main characteristics. ________________________________ 37 Table 7. Primer sequence and characteristics used to generate kan cassette (F and R) and to check

(check_F and check_R) for endA, pgi and recA genes knockouts.Lowercase letters represent the sequence

from the template plasmid pKD13 and uppercase letters correspond to the sequence from the genome of

wild-type strain. ____________________________________________________________________ 38 Table 8. Oligonucleotides sequences and characteristics used. ________________________________ 39 Table 9. PCR reaction and program used to generate KanR cassette in pgi, endA and recA genes

knockouts. _________________________________________________________________________ 39 Table 10. PCR reaction and respective program used in colony PCR. __________________________ 41 Table 11. Strains used in this study with method described by Reisch and Prather [55] . ___________ 42 Table 12. Plasmids used in this study and their main characteristics. ___________________________ 43 Table 13. Primer sequence and characteristics used to generate the pKDsg-zwf plasmid [69], to generate

homologous arms (E and F) and to check (G and H) for zwf gene knockout. Lowercase letters represent

the sequence from the template plasmid pKDsg-ack and uppercase letters correspond to the protospacer,

in the gene zwf, preceded for a PAM site. ________________________________________________ 43 Table 14. PCR reaction and program to generate the two fragments of pKDsg plasmid. ____________ 44 Table 15. PCR reaction and program to generate the pKDsg-zwf plasmid. ______________________ 45 Table 16. Restriction enzymes and their characteristics used in plasmids DNA digestions reactions. __ 47 Table 17. Composition of Restriction Enzyme Reaction Buffers [80]. _________________________ 48 Table 18. Synthesis of the differences between the various strategies of shake flask cultivation. _____ 49 Table 19. Oligonucleotides used to test the absence or presence of unintentional genomic deletion. Some

characteristics of these oligonucleotides are present. ________________________________________ 50 Table 20. PCR program and PCR reaction using to assess the absence of rac sequence in GALG20. __ 51 Table 21. Expected and obtained results for the PCR reaction to test the genomic deletion in GALG20

strain. _____________________________________________________________________________ 72

Page 15: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

15

List of Abbreviations

Amp Ampicillin

bp Basepairs

Cm Chloramphenicol

CMV Cytomegalovirus

DNA Deoxyribonucleic acid

FTR FLP recognition target

gDNA Genomic DNA

GFP Green Fluorescent Protein

Kan Kanamycin

kb Kilo basepairs

KmR Kanamycin Resistance

LB Luria- Bertani

OC DNA Open Circular DNA

OD Optical Density

Ori Origin of replication

PCR Polymerase Chain Reactions

pDNA Plasmid DNA

Primer F Primer forward

Primer R Primer reverse

RCF / G Relative centrifugal force / acceleration relative

RNA Ribonucleic acid

SC pDNA Super Coiled DNA

sgRNA Single-guide RNA

Spec Spectinomycin

Tris Trizma® (TRIS base)

˚C Degree Celsius

Page 16: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

16

Thesis motivation

The plasmid DNA production is becoming increasingly important as therapeutic approach make

their way into clinical trials and eventually into the pharmaceutical product. The numerous

clinical trials for plasmid DNA products have demonstrated the safety of the DNA vaccination

method and indicate the potential of this relatively new field of therapeutics. This powerful

bioproduct has become a viable option to treatment of cancer, as well as for the gene therapy and

even for bacterial and viral diseases. Thus, research community have focused on the development

integrate process between the upstream and downstream processing. However, the quality of final

product is ultimately determined by fermentation strategy.

For these reasons, the biomass yield, plasmid yield and plasmid quality improvement can be

reached through optimization of the growth environment and of the plasmid-producing organism.

Therefore, the objective of this work was quantify the yield of plasmid DNA by culture of

Escherichia coli K-12 with several deletions (∆pgi∆endA∆recA), named GALGNEW, comparing

with two other strains, wild-type and GALG20. These gene knockouts have a positive impact in

the yield of this bioproduct.

Page 17: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

17

1. INTRODUCTION

1.1. Escherichia coli: The host strain

The genus Escherichia is one of the key genera of enteric bacteria included in a total of 346 genera

within the Gammaproteobacteria [3] (Figure 1). Enteric bacteria group involves Gram-negative,

non-spore forming rod, facultative aerobic, oxidase-negative, catalase-positive, denitrifying

bacteria, glucose-fermenting bacteria and motile by peritrichous flagella or non-motile [4].

Although, many species of the genus Escherichia are pathogenic to humans and animals, they are

of extreme industrial importance [4].

Escherichia coli (E. coli) is a commensal bacterium found in the digestive tract of warm-blood

animals and humans. E. coli strains constitute about 1% of the bacterial population of the gut [5].

E. coli consists of a diverse group of bacteria. Most E. coli strains are harmless, however, some

E. coli strains are pathogenic to humans being categorized into pathotypes [5]. Six pathovars are

Figure 1. Phylogenetic tree based on glucose-6-phosphate isomerase encoded by pgi gene. The selected sequences were aligned by Muscle Alignment Software. The topology was inferred using the Neighbor-Joining method and the

evolutionary distances were computed using the Poisson correction method implemented in the software MEGA 7.0.18.

The orange square ( ) in center represent the root of the circular tree, the red circular forms ( ), the green squares ( )

and the yellow triangle ( ) represent 3 of 15 orders of class Gammaproteobacteria: Enterobacteriales, Vibrionales and Pseudomonadales, respectively.

Page 18: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

18

associated with enteric/ diarrhoeal disease, urinary tract infections (UTIs) and pulmonary system

infections (meningitis). These pathovars are: enteropathogenic E. coli (EPEC),

enterohaemorrhagic E. coli (EHEC), enterotoxigenic E. coli (ETEC), enteroaggregative E. coli

(EAEC), enteroinvasive E. coli (EIEC) and diffusely adherent E. coli (DAEC) [6].

Nevertheless, E. coli has an extreme medical and biotechnological importance. Around fifty

approved recombinant therapeutic proteins are produced in this model organism, for example:

insulin for diabetes treatment, monoclonal antibodies (mAbs), growth hormone to humans and

farm animals, interferons against viral diseases, erythropoietin used in patients with severe anemia

and deoxyribonuclease I (DNase I) commonly used in hereditary disease cystic fibrosis (CF).

Products of recombinant DNA technology from E. coli are very useful in production of vaccines

[7], [8].

E. coli K-12 was one of the first organisms suggested for whole genome sequencing after having

been isolated from diphtheria patient, in 1922 [9], [10]. Since that a lot of mutant strains have

been created such as MG1655 and W3110, closely related E. coli K-12 “wild types” [11], DH1,

DH5, DH5α, DH5α ∆fruR, DH10B, JM101 ∆pykF ∆pykA and JM108 strains, typically used for

pDNA production [12], [13].

The creation of these strains and relationship between them are illustrated in Figure 2.

Page 19: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

19

DH5α strain was constructed by sequential introduction of the ∆(lacZYAargF)U169 deletion from strain SH210 and Φ80dlacZ∆M15 from strain TB1.

Several advantages are known for the use of this bacterium in plasmid DNA (pDNA) production, such as, their ability to grow quickly under minimal

growth conditions, high pDNA yields, complete genome sequenced and easy manipulation at laboratory and industrial scales [13].

E. coli K-12 MG1655 and E. coli DH5α strains were used in this work as the hosts strains for the construction of the GALGNEW strain using the Datensko

and Wanner protocol, and for the construction of the deletion mutant (∆zwf) by CRIPSR Cas9 System method, respectively. The MG1655 strain also was

used for comparison purposes (plasmid DNA production and growth kinetics) with two mutant strains: GALG20 and GALGNEW.

“Cavalli Hfr λ-” Hfr

λ- relA1 spoT1 metB1

cre-510 PO2A

X

“W208 λ-” F

-

thr-1 leuB6 lacZ4 supE44

rfbD1 thi-1

Figure 2. E. coli K-12 and derivatives: creation of new strains and relationship between different strains. Lineage of MG1655 and W3110, close relatives of wild-type E.coli K-12 (green

box). Generation of strains containing multiple mutations from MC1061, DH1 and JM101. Dark boxes represent commonly used E. coli strains for plasmid DNA production and recent developments in E. coli strains designed for high yield pDNA processes. Full line arrows represent the relationship between the strains and dashed line arrows represent mutations carried

from one strain to the other. Schematic representation adapted from [11]–[13].

Page 20: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

20

1.2. Plasmid DNA and Therapeutic Applications

A plasmid is a small, circular and double-stranded DNA molecule that is separate from a cell’s

chromosomal DNA. This extrachromosomal DNA, occurs naturally in archaea, bacteria, yeast

and some higher eukaryotic cells. Another property of this fragment of DNA is the ability to self-

replicate [14], [15]. This ability is due to the presence of at least one origin of replication (ORI).

The plasmid requires some elements that allow the propagation of the plasmid within host, such

as: origin of replication and an antibiotic resistance gene or a selectable marker [16] .

Plasmids have taken on a crucial role in the biotechnology and pharmaceutical domains being

implemented for DNA manipulation, gene expression and heterologous proteins production.

Heterologous proteins should substitute proteins or provide a lost function in patient due to

defective or absent activity [14]. More recently, an alternative to treat diseases is the

administration of the gene of interest to the patient. The plasmids are used as vectors in the

immune system, carrying of antigen to elicit immune responses in higher order animals [14], [17].

This strategy is designated as gene therapy or genetic medicine. In the early 1990s the transfer of

genes to humans was reported and since then allowed as a new approach for vaccination. Plasmid

DNA (pDNA) is the base for promising DNA vaccines and gene therapies against many

infections, acquired, and genetic diseases, including HIV-AIDS, Ebola, Malaria, Dengue, and

different types of cancer (the highest percentage – 64.5%), enteric pathogens, and seasonal

influenza viruses [18] (Figure 3). In clinical, a particular application of DNA vaccines is the

generation of the therapeutic vaccines for tumor control, which are induced by human papilloma

virus. These improvements lead to increase safety without compromising efficacy [19].

In veterinary domain, some products are licensed for application [13]. This includes two

infectious disease vaccines for West Nile virus in horses, infectious haematopoietic necrosis virus

in salmon, a melanoma cancer vaccine for dogs and a growth hormone releasing factor therapy

for pigs [20].

Figure 3. Indications addressed by gene therapy clinical trials. Data updated in August 2016 [21].

Page 21: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

21

Last year, over 2400 clinical trials have been completed, are ongoing or have been approved

worldwide. More than 65% of the trials have been performed in America and almost 24% in

Europe, as represent in Figure 4 [21].

1.2.1. Types of Vaccines: Evolution and characteristics

In literature, the opinions about the types of vaccines organized in groups are variable. Some

authors regard as first generation vaccines for humans were developed against diseases with

high mortality rate, for example: smallpox, cholera, typhoid fever, plague, yellow fever, polio and

rubella, consisting mostly of killed/ inactivated or live/ attenuated microorganisms, very reactive

and in some cases, inefficient [19], [22], [23] (Figure 5). During the 20th century, with technology

advances, more new types of vaccines were developed leading to the second generation vaccines

including toxoid vaccines, polysaccharides vaccines and purified proteins vaccines [19]. The

subunit vaccines are considered as an extension of the toxoid approach and the third and newest

generation of vaccines, together DNA vaccines and recombinant vector vaccine [19], [23].

Other authors consider the subunit vaccines as the first approach of vaccines, together killed/

inactivated and live/ attenuated vaccines and the new generation of vaccines include DNA

vaccines to induce a more effective cellular and humoral responses [24]–[26].

Figure 4. Geographical distribution of gene therapy clinical trials by continent. Data updated in August 2016 [21].

Page 22: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

22

The main characteristics of the new generation of vaccines are presented in Table 1.

Table 1. New generation of vaccines: main characteristics.

Type of

vaccines Strategy Advantages Disadvantages Vaccines

Sub-unit

Production of recombinant

proteins in

heterologous

systems

Low adverse reactions;

Not infectious (safely

for immuno-

suppressed animals)

Antibodies may not recognize the antigens

(native structure);

Stimulation of

immune system inefficient

Influenza

Hepatitis B

HPV

Acellular

pertussis

DNA

recombi-

nant

Genetic

manipulation for insertion

of genes

encoding

antigens

Closely mimics a natural infection;

Resulting in a greater

immune response

Small chance that the

DNA vector could be integrated into the

host cells;

Still in experimental

stages

Rabies

Measles

HIV

Dengue fever*

BCG*

Typhoid

fever*

Adenovirus

*

DNA

vaccines

Immunization

with recombinant

plasmids

Do not contain any form of pathogen;

No risk of infection;

More stable;

More affordable costs;

Produces humoral and

cellular immune response

Production antibodies

against DNA

(hypothesis);

Chance of inducing

mutation; Plasmids may be

integrated into the cell

and can lead to

transfer of resistance gene

Toxoplasmosis

Leishmaniosis

Anaplasmosis

Malaria

Herpes

HIV

Influenza

Melanoma

vaccine**

Figure 5. The various vaccine technologies developed over time.

IPV – Inactivated Polio Vaccine; OPV – Oral Polio Vaccine; HPV – Human Papillomavirus [22].

* Vaccines not yet available for use in humans; ** Vaccine for use in dogs [19], [27]–[29] .

Page 23: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

23

Compared to conventional vaccines, DNA vaccines have many advantages such as high stability,

not being infectious, focusing the immune response to only those antigens desired for

immunization and long-term persistence of the vaccine protection.

DNA vaccines are constructed by one gene or by a combination of different genes encoding

different antigens [18]. Conventional vaccines are based on whole pathogens and typically induce

immune responses against a several components of the organism.

1.2.2. Design of Plasmids for Gene Therapy and Vaccination

Gene therapy and DNA vaccination involve the injection of vector in vivo, which contain the gene

of interest to the patient, to elicit an immune response to a protein encoded on the plasmid [14],

[19], [25], [26]. Viral vectors are considered as the most effective of all gene delivery methods

for in vivo gene transfer. There are several vectors available to introduce the gene of interest into

human cells, as shown in Figure 6.

Commonly used viral vectors for brain cancer gene therapy includes retrovirus (18.2%), herpes

simplex virus (3.6%), adenovirus (21.4%) and adeno-associated virus (7.0%). Apart from these,

the most used is pDNA (21.8%). Over 17% of the trials for human gene therapy have been based

on naked pDNA, whereas lipofection (which also requires pDNA production) counts for 4.6% of

the trials [21].

The biotechnological production of pDNA is performed into two parts: upstream and

downstream. Upstream (stage 1) is a stage processing during which pDNA is produced by

transformed cells and the downstream (stages 2, 3 and 4) is the isolation and purification of the

bioproduct, as represented in Figure 7.

Figure 6. Vectors used in Gene Therapy clinical trials. Data updated in August 2016 [21].

Page 24: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

24

The first stage in planning an efficient process of DNA vaccination (and also in gene therapy)

should be the identification of the target gene, the vector design [30], the cloning of target gene

into a vector (pDNA) and its transformation into a bacterial cell, typically E. coli [20].

The plasmid used as vector contains some elements that allow its propagation in the bacterial host

(plasmid propagation unit) and expression of the vaccine gene in the eukaryotic cells of the

recipient organism (eukaryotic expression unit. The organization of these elements reflects the

plasmid’s functionality [16]. The unit responsible for plasmid propagation (prokaryotic

expression) contains a replication region and a selection marker as an antibiotic resistance gene,

and the unit responsible for vaccine synthesis (eukaryotic expression) that comprises a target

gene encoding an antigen (transgene), a promoter, a terminator and sometimes sequences like

introns, signal sequences and immune stimulatory sequences (ISS) [16], [18] (Figure 8).

Figure 7. The main stages for pDNA vaccine design, production and vaccination. Adapted from [14], [20], [34].

Stage 4

Eukaryotic Expression

Immunogenicity

Stage 3

Delivery

Stage 2

Purification

Formulation

Stability

Stage 1

Vector Design

Bacterial Transformation

Bacterial Cultivation

Page 25: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

25

The replication region allows the maintenance and propagation of the plasmid (multiple copies)

in host cells [16]. For this purpose, the most commonly used plasmids are derived from the

pBR322 or pUC plasmids [31], being used origins of replication that provide large number of

copies of DNA plasmids in bacteria, such as the E. coli’s ColE1 origin of replication [14], [31].

The ColE1 origin of replication produces a relatively low copy number (20-40 copies per cell),

so some modifications were made, using pUC vector with ColE1 as origin of replication

increasing the copy number to 500 per cell [14]. Selectable markers such as, antibiotic resistance

genes, ensure a stable inheritance of plasmids during bacterial growth, also being a powerful

selector important in cloning steps [31].

The eukaryotic expression unit consists of the elements necessary for high-level expression and

targeting of the therapeutic component. Promoters are essential in plasmids to drive high

expression of the target gene in mammalian cells. Viral-derived promoters, such as the

cytomegalovirus (CMV), the simian virus 40 (SV40) and the Rous sarcoma virus (RSV) are the

most widely used promoters [31]. These plasmids also contain a terminator, a polyadenylation

signal sequence, coupled to termination and processing of the therapeutic gene [14]. The introns

are introduced to augment the promoter activity because demonstrate a beneficial effect on the in

vivo expression of the transgene. Intron A from CMV is widely used. A signal sequence is a

sequence that codes for a signal peptide with approximately 20-40 amino acids. This signal

peptide is responsible for secretion of the synthesized peptide to the extra-cellular milieu, and is

located upstream of the vaccine gene. The function of the ISS is increase the potency of a DNA

vaccine. These are nucleotide hexamers that interact with toll-like receptors and add adjuvanticity

[30]. The ability of the host to recognize bacterial DNA depends of unmethylated cytosine–

guanine(CpG) dinucleotides in particular sequences, called CpG motifs [32]. These dinucleotides

are covalently linked CG dinucleotides. The frequency of these motifs is extensively suppressed

in vertebrates, including mammals. The bacterial immunostimulatory DNA sequences (ISS) and

CpG motifs are synonyms, in which are defined functionally and structurally, respectively [33].

Figure 8. Genetic elements of a pDNA vector. The plasmid consists of a Plasmid Propagation Unit (PPU) that operate

in the microbial host and a Eukaryotic Expression Unit (EEU) that drives the protein synthesis in the eukaryotic cells [30].

Page 26: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

26

In the second stage of the process, the plasmid DNA is typically extracted using alkaline lysis,

purified to remove impurities such as protein, RNA, chromosomal DNA, and endotoxins to

acceptable levels and formulated for delivery. Of these impurities, chromosomal DNA is the most

difficult to remove due to similar properties to the plasmid product [34]. At formulation step, the

purified antigen is combined with adjuvants, stabilizers and preservatives to form the final vaccine

preparation. Adjuvants are common to most licensed vaccines. The role of the adjuvants is to

enhance the immune responses elicited by vaccination. Examples of adjuvants tested for plasmid

DNA vaccines are: poly-lactide coglycolide (PLG), poloxamers and Vaxfectin® [18]. The

stabilizers increase the storage life, and preservatives allow the use of multi dose vials and prevent

fungal and/or bacterial contaminations [35].

In the third step, plasmid is delivery to a eukaryotic cells and in finally step, the gene of interest

is expressed [20]. Relative to the quantity of plasmid DNA required, it will differ between

therapies, and each disease is likely to have specific needs. Generally, for DNA vaccination the

dose will range from micrograms to milligrams [36] . For instance, therapeutic doses required for

cardiovascular diseases are often 1 mg to 10 mg of pDNA. Regarding oncology genes, dosages

range from 10 μg to 10 mg of pDNA. In the case of Hepatitis B infections, HIV infections, malaria

and tuberculosis therapeutic doses range from 10 μg to 100 μg of pDNA [37]. DNA vaccines

consist of a plasmid DNA back-bone containing a target gene (an antigen-encoding gene) and a

strong mammalian promoter, which controls its expression, as shown in Figure 8. When injected

intramuscularly or intradermally, the antigen is transcribed, translated and presented to the

immune system [38]. The response of immune system is initiated and expanded by antigen

presentation. The antigen is taken up by antigen-presenting cells (macrophages and dendritic

cells). The role of these cells in the immune response is to present the peptide fragments of the

antigen on their surface in the context of major histocompatibility complex (MHC) class II

molecules, forming a peptide-class II complex. These complexes are transported to the surface

and are recognized by specific CD4+ T cells [32].

The structural characteristics of plasmids are important from a therapeutic point of view, since

stability and efficacy depend in part of the topology of the plasmid [14].

1.2.3. Plasmid Structural Stability

One of the major problems encountered during the design of a DNA vaccine is assurance of its

structural integrity [39]. The pDNA required for a therapeutic product should be homogeneous

relatively to structural form and DNA sequence. Some sequences can be harmful to the production

of high-quality supercoiled plasmid. Hence, some elements should be avoided as purine-

pyrimidine/oligopurine-oligopyrimidine tracts, Chi sequences, G-rich sequences, direct repeats,

inverted repeats, poly-A sequence, nuclease sensitive regions, regions similar to genomic DNA,

and insertion sequences [39]. The main consequences of these determinants are summarized in

Table 2.

Page 27: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

27

1.2.3.1 Plasmid Size

Plasmid size is a critical criterion in vector

design, because tends to correlate with a

higher propensity for intramolecular

recombination and/or genome integration

events [39]. The plasmids should be as small

as possible, containing only the essentials

elements for the therapeutic applications

[30]. The pDNA vectors for therapeutic

applications with reduced size have as

additional advantage the capacity of result in

higher amounts of pDNA produced by

bacterial cultivation. Another advantage of

the shorter plasmids is the increased of the

transfection efficiency making easier the

purification, especially at large scale [14].

Regarding on diffusion of DNA in the

cytoplasm of cells is strongly size

dependent, with little or no diffusion for

DNA upper 2,000 base pairs (bp).

Minicircles are an example of the successful

transformation, comparing with longer conventional plasmids (Figure 9). Minicircles are double-

stranded, supercoiled expression cassettes devoid of the bacterial pDNA backbone. The

transfection of cells by a minicircle 2.9 kb in size is 77 times more efficient than a plasmid 52.5

kb in length [40]. Therefore, the minicircles are also a promising tool regarding safety [40].

Figure 9. Ranking of alternative plasmid selection approaches according to plasmid size and transformation efficiency.

Adapted from [40].

Table 2. Overview of major factors affecting plasmid structural stability. Adapted from [39].

Page 28: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

28

1.2.3.2 DNA structure

Dynamic structure of DNA is also important characteristic since plasmids can assume several

conformations apart from the most common negatively supercoiled B-form [39]. Several non-

Watson- Crick DNA structures have been discovered such as, C-DNA, D-DNA, P-DNA and T-

DNA. However, the most prevalent of these is the Z-form. Examples of other unusual structures

are intrinsic bends, triplexes and quadriplexis [41]. These conformations usually derived from the

presence of repeated DNA motifs such as direct repeats, inverted repeats, purine-pyrimidine and

G-rich sequences (Table 2), which are prone to genetic rearrangements (deletions, duplications,

inversions, translocations and insertions) [39].

In order to determine the implications such unusual structural features have on plasmid

production, Cooke and co-workers evaluated the influence of specific non- Watson-Crick DNA

structures on the stability, yield and topology of the general cloning pBluescript vector [39], [41].

The authors found that plasmid containing Z-DNA forming regions is more unstable comparing

with other structures (triplexes, bends and quadruplexes). Furthermore, the triplexes structure

led to decreased amounts of supercoiled plasmid by 5% in the 3.8 kb plasmid [41].

Relative to the plasmid topology, it is knowing that plasmid DNAs that constitute DNA vaccines

are produced in bacteria as supercoiled (SC) or covalently close circular (CCC) plasmid DNA.

Single strand nicking results in the relaxation of supercoiled DNA into another isoform, named

open circular DNA (OC). Immunization studies done by Pillai et al. [42], reveal SC and OC

DNAs having different biological activities for in vivo immunizations, being the amount of SC

plasmid DNA in a vaccine preparation the best predictor, presented 3-times higher ability than

the OC DNA.

1.2.4. Stability in Replication Process

A plasmid is defined as a DNA molecule capable of autonomous replication. Many natural

plasmids are stably maintained at their characteristic copy number within the growing bacterial

population, controlling their concentration and regulating the rate of replication [43]. Some

elements can be considered within plasmid: (a) origin(s) of replication (ori), which is

characteristic of each replicon, (b) many plasmids encode a protein involved in the initiation of

replication, named Rep protein and (c) the genes involved in the control of replication [44]. The

plasmid copy number may vary depending on the host strain and on the growth conditions, but

any particular plasmid has a characteristic copy number. Replication by the Theta mechanism

has been most extensively studied among the prototype circular plasmids of gram-negative

bacteria. This mechanism involves melting of the parental strands, synthesis of a primer RNA

(pRNA), and initiation of DNA synthesis by covalent extension of the pRNA [44].

Page 29: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

29

1.2.5. Advantages of DNA vaccines

Whereas traditional vaccines rely on the production of antibodies through the injection of live

attenuated virus, killed viral particles or recombinant viral proteins, DNA vaccine plasmids are

non-live, non-replicating and nonspreading, there is little risk of either reversion to a disease-

causing form or secondary infection. Thus, DNA vaccination present many advantages in terms

of safety (not being infectious does not have capacity to revert to virulent forms, does not require

use of toxic treatments and no significant adverse events in any clinical trial), simple construction

and rapid production (synthetic and PCR methods allow simple engineering design modifications,

optimization of the plasmid depending of the target gene and rapid formulation, reproducible,

large-scale production), high stability (long shelf life and more temperature-stable),

immunogenicity (induction of T and B cell-specific antigen) and mobility (ease of storage and

transport) [14], [31], [45]. A significant advantage, mainly for emerging pathogens is that this

new generation of vaccines do not require the handling of potentially deadly infectious agents

[18].

1.2.6. pVAX1-GFP plasmid

pVAX1-GFP is a 3.7 kb plasmid vector designed for use in the development of DNA vaccines.

pVAX1 contains no eukaryotic or bacterial region optimizations, and consequently has relatively

low manufacturing yield and expression in vitro [34]. The elements that compose this vector, their

purposes and schematic representation of pVAX1-GFP plasmid are illustrated in Table 3 [46].

Table 3. Plasmid pVAX1-GFP: elements and their purposes. The schematic representation was created with the

SnapGene software.

Elements Purpose

Human

cytomegalovirus

immediate-early

(CMV) promoter

High-level expression in a wide range of

mammalian cells

Bovine growth

hormone

Polyadenylation signal for efficient

transcription termination and

polyadenylation of mRNA

Kanamycin

resistance gene Selection in E. coli (host strain)

pUC origin High-copy number replication and growth

in E. coli

Green Fluorescent

Protein (GFP)

The expression could be observed by

fluorescence microscopy or by flow

cytometry

Plasmid pVAX1GFP derived from pVAX1LacZ (Invitrogen, Carlsbad, CA). The plasmid

pVAX1GFP and their main features are in Table 4.

Page 30: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

30

Table 4. Plasmid pVAX1-GFP and their main characteristics.

1.3. Effect of plasmid DNA synthesis on E. coli central carbon metabolism

Plasmid DNA synthesis can perturb the E. coli global gene regulation, leading to significant

changes in central metabolic pathways and altering levels of gene expression, in the host. Many

researchers observed that plasmid maintenance retards host growth rate and biomass yield,

comparing with plasmid-free cells. For plasmid replication and expression of the antibiotic

resistance gene, the required nutrients and energy levels imposes a higher metabolic burden on

the host cells [13], [47]. In E. coli, the central metabolic pathways provide some important

elements, like precursors (nucleotide precursors), cofactors and energy, for the biosynthesis and

other metabolic processes [28]. Therefore, the central carbon metabolism is a target for genetic

engineering

strategies to

increase pDNA

yields [13].

The main

central

metabolic

pathways of E.

coli are

glycolysis, the

tricarboxylic

acid (TCA)

cycle and the

pentose

phosphate (PP)

(Figure 10).

Glycolysis is

the main

catabolic

pathway of

carbohydrates,

using glucose

for the

provision of

energy and building precursors. The final product of this pathway is pyruvate having as

intermediate product fructose diphosphate. Glycolysis is composed of ten reactions catalyzed by

Plasmid Size

(bp)

Antibiotic resistance

genes

Origin of

replication Purpose

pVAX1GFP 3697 Kanamycin pUC ori Bacterial

cultivations

Figure 10. The Central Carbohydrate Metabolic Network [85].

Page 31: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

31

specific enzymes that are coded by individual genes, which are downregulated, in plasmid-

bearing-cells, excepted gapA gene. The most important control of the glycolysis resides in only

two irreversible steps, catalyzed by phosphofructokinase and pyruvate kinase, coded by pfk and

pyk genes.

The pentose phosphate pathway (PPP) is the second major route for carbohydrate and performs

several functions such as: (1) catabolism of carbon sources including sugars like xylose and

ribose, (2) synthesis of reducing power (NADPH) and (3) biosynthesis of the nucleotide

precursors, nucleic acids, amino acids, vitamins and lipopolysaccharides [47]. Are examples of

nucleotide precursors: ribose-5-phosphate (R5P) and erythrose-4-phosphate (E4P) [13]. The

reducing power and nucleotides are essential for biomass and pDNA production and they are

directly related in this pathway. Plasmid-bearing cells carrying high copy number, require a level

of nucleotides higher and in this case the PP pathway may be insufficient considering the cell’s

metabolic needs [48].

The other pathway with important roles in metabolism of E. coli is tricarboxylic acid (TCA)

cycle, being essential for the complete oxidization of acetyl coenzyme A (CoA) from glycolysis,

occurring in eight reactions. Some intermediates of this cycle play an important role in amino

acids synthesis such as, oxaloacetate (OAA) and alpha-ketoglutarate (AKG). In plasmid-bearing

cells, most of the TCA cycle participants’ genes are up-regulated for different E. coli strains, such

as fumA, aceB, sucBCD and sdhCD genes. This up-regulation could indicate a greater

accumulation of carbon in the cycle due to the reduced growth rate [13], [47].

1.4. Relevant genes for E. coli strain engineering aiming to increase

pDNA production

Phosphoglucose isomerase (Pgi) is an enzyme coded by pgi gene and catalyzes the conversion of

glucose-6-phosphate into fructose-6-phosphate, being a reserve reaction of glycolysis (box with

dash blue line in Figure 10). The knockout of the pgi gene leads to the redirection of the carbon

flux into the PP pathway, increasing the synthesis of nucleotides (R5P and E4P) required for the

pDNA synthesis. Also provide high amounts of reducing power (NADPH) [49].

Other relevant gene is endA that encodes for DNA-specific endonuclease I. Endonuclease I is a

periplasmic enzyme that cleaves within duplex DNA [50]. A knockout of this gene leads to a

decrease non-specific digestion of plasmid, improving the quality of plasmid preparations [13].

In E. coli K-12 strain, the recA gene codes for a polypeptide essential for the recBCD pathway of

homologous recombination, more specifically, DNA strand exchange and recombination protein

with protease and nuclease activity [13]. The RecA protein performs many functions: (1)

catalyzes homologous pairing and strand exchange of DNA molecules necessary for DNA

recombination repair, (2) ATP and DNA-dependent co-proteolytic processing of effector proteins

and (3) interaction with mutagenic protein factors to facilitate error-prone DNA synthesis past

DNA lesions. Mutations in recA affect not only recombination, but also DNA repair, mutagenesis,

and cell division [51], having been observed to have a positive impact on pDNA yield [13].

Page 32: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

32

However, the effect of some mutations, such as ΔendA and ΔrecA, are very strain and/or plasmid

dependent [52].

Another mutant was constructed by disruption of zwf gene, which encodes glucose-6-phosphate

dehydrogenase (G6PDH) [53], following the CRISPR Cas9 system. G6PDH catalyzes the first

reaction of the PP pathway (represented into box with dash red line in Figure 10). This knockout

led to restructuring of the carbon flux through central metabolism in E. coli, allowing alternative

routes. Previous studies concluded that the distribution of carbon fluxes in the zwf mutants

showed that the anabolic requirements for nucleotide R5P and E4P were satisfied by the reverse

activity of the non-oxidative branch of the PP pathway, in which, ribulose-5-phosphate is

converted into fructose-6-phosphate and glyceraldehyde-3-phosphate. The consequence of this

restructuring of metabolic fluxes is a virtually unchanged rate of NADPH synthesis as compared

with wild-type [54]. More specifically, the zwf mutant directed 98.9% and 87.0% of the total

carbon flux through the first step of glycolysis (from G6P to F6P) and TCA cycle [from acetyl-

coenzyme A (AcA) to citrate (CIT)], respectively, whereas the parent strain showed an obviously

lower flux through the first step of the glycolysis (78.6%) and TCA cycle (73.1%) [53].

Page 33: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

33

2. MATERIALS AND METHODS

In this study two strategies were used for genome editing in E. coli: λ-Red-mediated gene

replacement technique described by Datsenko and Wanner [1] and the CRISCP Cas9-System

described by Reisch and Prather [55].

2.1. Media, Chemicals and Other Reagents

For cell cultures, LB (Luria-Bertani) broth (25 g/ L) from NZYTech was used. When required the

culture medium was supplemented with ampicillin [(100 µg/ mL) (Sigma-Aldrich®)], kanamycin

[(30 µg/ mL) (aMRESCO®)], chloramphenicol [(50 µg/ mL) (Sigma-Aldrich®)] or

spectinomycin [(50 µg/ mL) (Fluka® Analytical). The solid media were prepared using LB agar

(40 g/ L) from NZYTech.

To increase plasmid production a complex medium was used in shake flask cultivation. This

medium is composed by basal cultivation medium [Peptone (Fluka® Analytical) (10 g/ L), yeast

extract (10 g/ L) (Liofilchem®), (NH4)2SO4 (3 g/ L) (PanReac), K2HPO4 (3.5 g/ L) (PanReac),

KH2PO4 (3.5 g/ L) (PanReac), pH 7.1], trace elements solution [FeCl3∙6H2O (27 g/ L) (Sigma

Aldrich, ZnCl2 (2 g/ L) (Sigma-Aldrich), CoCl3∙6H2O (2 g/ L) (Sigma-Aldrich), Na2MoO4∙2H2O

(2 g/ L) (Fluka Analytical), CaCl2∙2H2O (1 g/ L) (Merck), CuCl2∙2H2O (1.3 g/ L) (Sigma-Aldrich),

N3BO3 (0.5 g/ L) (Fisher Scientific), HCl 1.2 M (Riedel-deHaën)], glucose solution [(20 g/ L)

(Fisher Scientific)] and seed supplement solution [MgSO4∙7H2O (240 g/ L) (Merck), thiamine (24

g/ L) (Sigma-Aldrich)] [56]. The trace elements solution and seed supplement solution were filter-

sterilized while basal cultivation medium was autoclaved in the shake flask. Glucose solution was

autoclaved. After sterilization, the remaining solutions were added to 250 mL shake flasks in the

following quantities per flask: 50 mL (basal cultivation medium), 50 µL (trace elements solution),

2 mL (glucose) and 415 µL (seed supplement solution). This complex medium was supplemented

with 50 µL kanamycin (30 µg /mL).

For polymerase chain reactions (PCR) three DNA polymerase kits were used: NovaTaq™ Hot

Start Master Mix Kit (Novagen®), KOD Hot Start DNA Polymerase (Novagen®) and Platinum®

PCR SuperMix High Fidelity (Invitrogen®). The termocyclers used were ThermoHybaid Px2 or

TGradient from Biometra.

All agarose gel electrophoreses, were performed with agarose (Low-EEO/Multi-Purpose) from

Fisher Scientific, in a concentration of 1% (w/ v).

The molecular weight marker used were NZYDNA ladder I (Size Range: 0.2 – 1.8 kb) NZYDNA

ladder III (Size Range: 0.2 – 10 kb) from NZYTech and HyperLadder™ 50 bp (Size Range: 0.05

– 2.0 kb) from Bioline.

To induce the recombinase expression, L-arabinose 20% w/ v was prepared (Merck) [1].

Chemical competent cells were prepared using the Transformation and Storage Solution (TSS)

buffer [MgCl2∙6H2O (1.670 g/ L) (Fagron), Poly(ethylene glycol) with average mol wt 8,000

(Sigma- Aldrich), LB broth (25 g/ L) (NZYTech)], pH 6.5, adjusted with HCl 1 M v/ v (Riedel-

deHaën) . This solution was filter-sterilized (0.22 µm). Dimethyl sulfoxide (DMSO) (Sigma-

Aldrich) was added to TSS in proportion 1:10 final volume.

Plasmid DNAs were digested using appropriate restriction enzymes with the respective buffers

from Promega.

Page 34: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

34

Glycerol 99%, ACROS Organics-Fisher Scientific was used to preserve the frozen cell banks at -

80 ˚C in a final concentration of 20%.

2.2. Preparation of Competent cells

2.2.1. Electrocompetent cells

Before the introduction of the KanR cassette in host cells, electrocompetent cells were prepared.

Shake flasks with autoclaved LB (50 mL) were inoculated with 30 µL from a frozen stock of the

desired strain and incubated with orbital shaking at 37 ˚C, 250 rpm. Cells were harvested at the

end of an overnight growth (OD600nm = 1-2). Each bacterial culture was divided into five Falcon

15 mL Conical Centrifuge Tubes and centrifuged at 6,000 g, 3 min, 4 ˚C.

Supernatant was discarded under aseptic conditions. Each pellet was re-suspended in 1 mL cold

sterile milli-Q H2O and transferred to 1.5 mL Eppendorf. The centrifugations (1 min on

microcentrifuge, MiniStar silverline from VWR) and washing steps were repeated four times. In

the last wash, the supernatant was discarded and bacterial pellet was re-suspended, in 100 μL cold

sterile milli-Q H2O. The bacterial suspension was ready to eletroporate.

A. Transformation by electroporation

Electroporation is a transfection method based on an electrical pulse to create temporary pores in

cell membranes. Through these pores, some substances like plasmids or other nucleic acids forms

can pass into cells [57]. Electroporation cuvettes with 2 mm gap width were used for this

efficiently process. Before using, the cuvettes were cleaned with ethanol 70% (v/ v) and dried

with absorbent paper. Electrocompetent cells and the selected DNA molecules were mixed in 1.5

mL Eppendorf and transferred to an electroporation cuvette and incubated 30 min on ice. A 2500

V electric shock was applied to cells by the electroporator ECM 399 from BTX. The

transformants were recovered after adding of 900 µL LB broth. The recover conditions were

optimized depending of the DNA molecule used to transform the cells.

In this work, the electroporation protocol was applied to transform electrocompetent cells as

suggested by Datsenko and Wanner strategy [1] and in third step of the CRISPR/Cas9 System

[55].

I. Transformation with KanR cassette: Transformants were recovered by incubating on

orbital shaking at 37 ˚C, 250 rpm, 2 h, allowing expression of antibiotic resistance gene.

100 µL of recovered cells was plated on LB- agar plate supplemented with kanamycin

(30 µg/ mL) and incubated overnight at 37 ˚C. (The remaining LB-cell mixture was left

on the laboratory bench overnight. On next day, if there were no transformants on plate,

the remaining LB-cell mixture was plated).

II. Transformation with pKD46 plasmid: Transformants were recovered by incubating on

orbital shaking at 30 ºC, during 1 h. The recovered cells were plated in LB supplemented

with ampicillin (100 µg/ mL) and incubated at 30 ˚C, overnight.

Page 35: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

35

III. Transformation with pCP20 plasmid: Transformants were recovered by incubating on

orbital shaking at 30 ˚C during 1 h. The recovered cells were centrifuged and the pellet

was plated on LB plate supplemented with chloramphenicol (50 µg/ mL) and grown

overnight at 30 ˚C.

IV. Transformation with oligonucleotides (dsDNA): Transformants were recovered by

incubating with orbital shaking at 30 ºC, during 1-2 h. Then, recovered cells were plated

on LB agar with chloramphenicol (50 µg/ mL), spectinomycin (50 µg/ mL) and

anhydrotetracycline (100 ng/ mL). Plates were incubated at 30 ºC, overnight.

2.2.2. Chemical competent cells

Chemical competent cells protocol was performed with DH5α strain. Falcon 15 mL Conical

Centrifuge Tube with 5 mL autoclaved LB was inoculated with a 30 µL of the aliquot of strain

and incubated on orbital shaking incubator at 37 ˚C, 250 rpm. On the next day, a shake flask with

autoclaved LB (20 mL) was inoculated with an OD600nm = 0.1 from pre-inoculate. The inoculated

shake flask was incubated on orbital shaking incubator at 37 ˚C, 250 rpm. Cells were harvested

at OD600nm = 1. Bacterial suspension was divided in two Falcon 15 mL Conical Centrifuge Tubes

and each was centrifuged at 1,000 g, 10 min, 4 ˚C.

Supernatant was discarded under sterile conditions. Each pellet was re-suspended in TSS +

DMSO solution (1,900 µL: 100 µL) and kept on ice for 10 min. Successively, aliquots were

prepared for cryopreservation.

B. Transformation by heat shock

This technique was used to introduce the pVAX-GFP plasmid and the three plasmids used in

CRISPR Cas9 System into chemically competent cells.

Each plasmid DNA was gently mixed with chilled cells and the mixture was incubated 30 min on

ice to allow the plasmid to come into close contact with the cells. The plasmid-cell mixture is then

briefly heated at 42 °C for 1 min, allowing the DNA to enter the cell through the transiently

disrupted membrane. The heated mixture is then placed back on ice for 2 min in order to retain

the plasmids inside the bacteria [58]. To recover, 900 µL of LB broth, was added, immediately.

The recover conditions were optimized depending on the selected molecule to transform the cells.

I. Transformation with pVAX-GFP: 10 ng of pVAX-GFP plasmid was used. Transformants

were recovered by incubating on orbital shaking at 37 ˚C, for 1 h. 100 µL of recovered cells

was plated on LB- agar plate supplemented with kanamycin (30 µg/ mL) and incubated

overnight at 37 ˚C.

II. Transformation with CRISPR Cas9 System Plasmids: Transformants were recovered by

incubating on orbital shaking at 30 ºC, for 1 h. After, the mixture was centrifuged and the

pellet was re-suspended in 100 µL of supernatant and plated on LB agar plate supplemented

with chloramphenicol (50 µg/ mL) and spectinomycin (50 µg/ mL), for pCas9cr4 or pKDsg

plasmids, respectively.

Page 36: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

36

2.3. Red Disruption System

2.3.1. Strains and plasmids

The bacterial strains used in this method and their genotypes are indicated in the Table 5.

Table 5. Strains used in study and main characteristics.

Strain Genotype References

K-12 MG1655 F- λ- ilvG rfb- 50 rph1 [59]

K-12 MG1655 ∆endA F- λ- ilvG rfb- 50 rph1 ΔendA [59]

K-12 MG1655 ∆endA∆pgi F- λ- ilvG rfb- 50 rph1 ΔendA Δpgi [59]

GALG20 F- λ- ilvG rfb- 50 rph1 ΔendA Δpgi ΔrecA rac- [48]

GALGNEW F- λ- ilvG rfb- 50 rph1 ΔendA Δpgi ΔrecA This study

(not published)

Bacterial strain E. coli K-12 MG1655 was from the Prather Lab, MIT. This strain was used as a

starting point for the derivative strains GALG20 and GALGNEW. Strain GALG20 was

constructed by Geisa Gonçalves, a former PhD student at IST. The difference between GALG20

and GALGNEW is an unintended secondary genomic deletion of approximately 20 kb. For gene

knockouts three plasmids were used: pKD13, pKD46 and pCP20 (Figure 11).

In pKD46 and pCP20 plasmids, the oriR101 and the repA101-ts are derived from pSC101

replication origin. These are low copy number plasmids vectors [60].

The pKD13 plasmid is the template plasmid for gene disruption. It contains a kanamycin

resistance gene (KanR) flanked by flippase recombination targets (FRT) and ampicillin resistance

gene (AmpR). This plasmid is used to make an insertion cassette containing kanamycin resistance

(B)

(C)

(A)

Figure 11. Schematic map of the plasmids used in Red system. (A) plasmid pKD13 (image created in SnapGene®

software) [5], (B) plasmid pKD46 [86] and (C) plasmid pCP20 [7].

Page 37: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

37

gene (KanR cassette) [1]. The pKD46 plasmid is a λ-Red recombinase expression plasmid, which

includes three genes: exo, β and γ, whose products are Exo, Beta and Gam, respectively [61]. Gam

inhibits the host RecBCD exonuclease V, which degrades linear DNA avoiding E. coli

transformation [1]. Thus, Beta and Exo can promote recombination. The λ-Red recombinase

genes are under the control of the araB promoter [1]. The Red recombinase expression is induced

by arabinose, promoting the integration of the cassette. This plasmid carries an ampicillin

resistance gene. Other feature of pKD46 is the temperature-sensitive replicon, so it can be cured

by raising the temperature [1].

pCP20 is an ampicillin and chloramphenicol resistant plasmid, is temperature-sensitive the level

of replication and thermal induction of FLP synthesis [1]. This helper plasmid expressing the FLP

recombinase can eliminate the resistance gene, acting on the directly repeated FRT sites flanking

the KanR cassette [1], [62]. Expression of the λ- Red recombinase or FLP recombinase is required

from helper plasmids such as pKD46 and pCP20, respectively [62].

The main characteristics of the used plasmids are summarized in Table 6.

Table 6. Plasmids used in this work and main characteristics.

Plasmids Size

(bp)

Antibiotic

resistance genes

Origin of

replication

Copy

Number Purpose

pKD13 3,434 Kanamycin

Ampicillin R6K gamma ~15-20

Construction by PCR of

insertion cassette

containing KanR

pKD46 6,329 Ampicillin oriR101 ~5 Promotes the action of

recombination

pCP20 9,332 Chloramphenicol

Ampicillin repA101ts ~5

Eliminates resistance

cassette

2.3.2. Oligonucleotides

Oligonucleotides were designed in ApE plasmid editor software and synthesized by Stabvida.

The schematic design of the primers is represented in Figure 12. The forward primer was

designed using a sequence homologous to the upstream region flanking the gene and the ‘priming

site 1’. For the design of the reverse primer it was used a sequence homologous to the downstream

region flanking the gene and the ‘priming site 2’. The priming sites are fragments homologous to

the pKD13 plasmid [63]. The 60 bp homology sites flanking the gene of interest just include the

primers to generate KanR cassette.

Page 38: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

38

Check primers were designed to confirm the total gene disruption.

The oligonucleotides used in Red Disruption System method are indicated in Table 7.

Table 7. Primer sequence and characteristics used to generate kan cassette (F and R) and to check (check_F and

check_R) for endA, pgi and recA genes knockouts.Lowercase letters represent the sequence from the template

plasmid pKD13 and uppercase letters correspond to the sequence from the genome of wild-type strain.

Gene Primer Sequence (5’ 3’) Size

(bp)

Tm

( ͦC)

%

G-C

endA

F AGGAACTTTCCTGATCTGGCTGATTGCATACCAAAACAG

CTTTCGCTACGTTGCTGGCTCgtgtaggctggagctgcttc 80 79 51

R TAGTTAAAAATCCGCGTCGTCTCCCCACGCGGTTGTACGC

GTGGGGTAGGGGTTAACAAAtccgtcgacctgcagtt 77 79 51

check

_F CGTCTATCGCTGTGTTCAC 19 54 53

check

_R CGCATTTATCATCCTGAACC 20 52 45

pgi

F ACTAAAACCATCACATTTTTCTGTGACTGGCGCTACAATC

TTCCAAAGTCACAATTCTCAgtgtaggctggagctgctt 80 75 44

R TAAGACGCGACCGCGTCGCATCAGGCATCGGTTGCCGGA

TGCGGCGTGAACGCCTTATCCtccgtcgacctgcagtt 77 84 62

check

_F AATGCTTCACTGCGCTAAGG 20 57 50

check

_R CGTCGGCATTGTTATTAAGG 20 43 55

recA

F ATACTGTATGAGCATACAGTATAATTGCTTCAACAGAAC

ATATTGACTATCCGGTATTACgtgtaggctggagctgcttc 80 73 40

R TGATTCTGTCATGGCATATCCTTACAACTTAAAAAAGCAA

AAGGGCCGCAGATGCGACCCtccgtcgacctgcagtt 77 78 48

check

_F TCGTCAGGCTACTGCGTATGC 21 60 57

check _R

CAGTGAGCAAGAACTGCGACG 21 60 57

Primers for genes fnr and ralR were also designed in order to distinguish the GALG20 and

GALGNEW strains (Table 8). These genes are included in an unintended secondary genomic

fragment of approximately 20 kb present in GALGNEW strain.

Homology to the

target (60 bp) Priming site

1 (20 bp) Homology to the

target (60 bp)

Priming site

2 (17 bp) Kanamycin cassette FRT FRT

Target Gene

Kanamycin cassette

Primer check F (~20 bp) Primer check R (~20 bp)

Figure 12. Schematic representation of construction of the primers: to generate kanamycin cassette (forward

and reverse primers) and to confirm the insertion of the kanamycin cassette (primers check) [51].

Page 39: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

39

Table 8. Oligonucleotides sequences and characteristics used.

Gene Primer Sequence (5’ 3’) Size

(bp)

Tm

( ͦC) %G+C

fnr F TCAGTCTGGCGGTTGTGCTATC 22 60 55

R AGAAACCATCAGCCGTCTGC 20 59 55

ralR F TTCAGCCTGGCGGTGTAATG 20 59 55

R AAGGTGGCACTCCTACTAAC 20 55 50

2.3.3. Generation of kanamycin cassette

For the construction of the KanR cassette, a PCR was performed using the different primers, to

make the linear recombination cassette (Table 7). The Figure 13 is a schematic representation of

KanR cassette.

The composition and conditions of PCR reactions are indicated in

Table 9. After the PCR reactions, the resulting products were analyzed by 1% (w/ v) agarose gel

electrophoresis, dividing the volume into 2 lanes (5 µL+ 15 µL). After electrophoresis, agarose

gel was cut into two parts. To minimize damage to the DNA, the right part of the gel (lane with

15 µL of PCR product) was not exposed to ethidium bromide solution and UV light. Cut out the

band was required to purify the DNA fragment. The excised fragment was transferred to a 15 mL

Falcon. The extraction of the fragment corresponding to KanR cassette from the gel was performed

using NZYGelpure Kit from NZYTech. The concentrations of the purified product were measured

using NanoVue Plus Spectrophotometer (GE Healthcare).

Table 9. PCR reaction and program used to generate KanR cassette in pgi, endA and recA genes knockouts.

PCR reaction PCR program

pgi

endA

recA

Platinum PCR Supermix High

Fidelity 22.5 μL

Incubation 94 ˚C 2 min

1

cycle

pKD13 plasmid 10 ng Denaturation 94 ˚C 5 s

35

cycl

es

Primer F 0.5 μL Annealing 55 ˚C 25 s

Primer R 0.5 μL Extension 72 ˚C 90 s

H2O Up to

25 μL

Figure 13. Kanamycin resistance cassette generated by PCR: Schematic representation created in the SnapGene software [36].

Page 40: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

40

2.3.4. Gene disruption strategy

The strategy used in this work was adapted from Red System described by Datsenko and Wanner,

performed in four steps. The basic strategy is to replace a chromosomal sequence with a selectable

antibiotic resistance gene. PCR is used to make an insertion cassette which contains a selectable

marker, usually kanamycin resistance, using primers with a homology extensions (H1 and H2)

(Step 1 in Figure 14) [1]. Antibiotic resistance is flanked by FTR sites to allow later excision of

the marker (Step 1 in Figure 14).

The first step was the generation of the cassette. Purification of pKD13 plasmid was made and

primers to amplify the selectable marker, FRT and custom mutation cassette were designed, as

represent in Table 7. Then a PCR was performed to generate the cassette (Table 9). Subsequently,

the PCR product was resolved in an agarose gel and the fragment was excised and purified.

The second step was the chromosomal integration. pKD46 plasmid (promotes the

recombination) was purified and electrocompetent cells were transformed by electroporation as

described in point A of section 2.2.1..

Transformants carrying pKD46 plasmid (recombineering-cells) were grown in 5 mL of LB

medium with ampicillin at 30 ˚C, 250 rpm. On next day, 50 mL of LB supplemented with

ampicillin in 250 mL shake flasks were inoculated to an OD600nm = 0.1. Cells were grown at 30

˚C, 250 rpm during 4 h. Then, 385 µL L-arabinose 20% (w/ v) was added to culture and incubated

at 30 ˚C, 250 rpm during 2 h. L-arabinose binds AraC, that is encoded by the gene araC present

in pKD46 plasmid. This protein allows the transcription of Lambda Red genes from the ParaB

promoter, expressing recombinase genes [64].

Figure 14. Gene disruption strategy. H1 and H2 are the homology extensions or regions, P1 and P2 are the priming sites. Strategy

described by Datsenko and Wanner [1].

Page 41: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

41

100 µL of electrocompetent transformants carrying the Red helper plasmid were electroporated,

as described in point A of section 2.2.1, with 1 μL of the PCR amplified KmR cassette.

Colony PCR analysis was the third step. Three to six freshly isolated colonies were suspended in

70 µL sterile water by pipetting. The DNA extraction was performed by thermal lysis placing 50

µL of the mixture at 99 ˚C for 5 min and immediately after in ice. The PCR reaction composition

and program are described in Table 10.

Table 10. PCR reaction and respective program used in colony PCR.

The last step was the elimination of the antibiotic resistance gene. A freshly KmR colony was

grown in 5 mL LB supplemented with kanamycin (30 µg/ ml). On next day, 50 mL of LB

supplemented with kanamycin in 250 mL Erlenmeyer flask were inoculated to an OD600nm = 0.1.

Cells were grown at 37 ˚C, 250 rpm. Cells were harvested at OD600nm = 1 and eletrocompetent

cells were prepared. The electrocompetent cells were transformed with pCP20 plasmid by

electroporation (point A of section 2.2.1). The pCP20 plasmid presents ampicillin and

chloramphenicol selection markers and shows temperature-sensitivity for replication. This

plasmid carries the gene flp encoding the FLP recombinase.

On the next day, four colonies were separately re-suspended, in 10 µL sterile water and plated on

LB plates without antibiotics. The plates were incubated at 43 ˚C, overnight. The KmR cassette

can be removed by FLP action, leaving behind a short nucleotide sequence with one FRT site,

[1]. The Red recombinase and FLP helper plasmids can be cured by growing cells at 43 ̊ C because

they have temperature-sensitive replicons [1].

After one overnight, the colony PCR was accomplished selecting one colony from each portion

of LB plates (4 colonies in total). The check primers and primers for genes fnr and ralR were

used. When the PCR result was the expected, 20 µL of 70 µL which were not used for DNA

extraction, were plated on LB plate without antibiotic, LB with chloramphenicol, LB with

kanamycin and LB with ampicillin, divided into 4 parts. The plates were incubated at 30 ˚C,

overnight. A final colony PCR analysis was made, selecting 2 colonies from LB plate. The

colonies having the expected profile were grown and cell banks were performed.

PCR reaction (Vfinal= 25 μL) PCR program

pgi

endA

recA

NovaTaq Polymerase 12.5 μL Incubation 95 ˚C 5min 1

cycle

DNA 10.0 μL Denaturation 94 ˚C 1 min

35

cycl

es

Primer F 1.0 μL Annealing 60 ˚C 1 min

Primer R 1.0 μL Extension 72 ˚C 2 min

Water 0.5 μL Final Extension 72 ˚C 10 min 1

cycle

Page 42: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

42

2.4. CRISPR Cas9-System method

2.4.1. Strains and plasmids

The bacterial strains used in this method and their genotypes are presented in the Table 11.

Table 11. Strains used in this study with method described by Reisch and Prather [55] .

Bacterial strain E. coli DH5α was obtained from Invitrogen. This strain was used as a starting

point for the construction of the single-guide RNA [55]. GALGNEW was transformed with three

plasmids: pCas9cr4, pKDsg-zwf and pKDsg-p15, which are represented in Figure 15 and were

obtained from Addgene. The plasmid pKDsg-zwf was constructed from pKDsg-ack in this work.

The main characteristics of these plasmids are indicated in Table 12. E. coli DH5α was used to

test the transformations with the several plasmids.

pCas9cr4 plasmid carries a Cas9 nuclease gene, which is under the control of Tet promoter, a tetR

constitutively expressed and a chloramphenicol resistance gene [55]. Cas9 is a endonuclease that

targets a specific DNA sequence and the only requirement of this nuclease is that the protospacer

be adjacent to the triplet NGG designated protospacer adjacent motif (PAM) [55].

Plasmid pKDsg-xxx which has the sgRNA expressed under control of the PTET promoter and has

λ-Red recombinase genes, under control of the arabinose inducible promoter ParaB. This plasmid

was the base for the construction of pKDsg-ack [66], pKDsg-zwf and pKDsg-p15 plasmids.

Plasmid pKDsg-p15 was created which targeted the p15A origin of replication of pCas9cr4, since

the plasmid pCas9cr4 does not possess the capacity that allow curing of the plasmid [67].

The origin of replication/replicon defines that these plasmids occur in low copy number, as shown

in Table 12 [68].

Strain Genotype Reference

E. coli DH5α F- Φ80lacZΔM15 Δ(lacZYA-argF) U169 recA1 endA1

hsdR17 (rK-, mK+) phoA supE44 thi-1 gyrA96 relA1 [65]

GALGNEW F- λ- ilvG rfb- 50 rph1 Δpgi ΔendA ΔrecA This work

(A) (B)

Figure 15. Schematic map of the no-SCAR plasmids. (A) Plasmid pCas9cr4. (B) Plasmid pKDsg-xxx [55].

Page 43: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

43

Table 12. Plasmids used in this study and their main characteristics.

2.4.2. Oligonucleotides

Oligonucleotides were designed using ApE plasmid editor software and synthesized by Stabvida.

The oligonucleotides used in this method are indicated in Table 13.

Table 13. Primer sequence and characteristics used to generate the pKDsg-zwf plasmid [69], to generate homologous arms (E and F) and to check (G and H) for zwf gene knockout. Lowercase letters represent the sequence from the

template plasmid pKDsg-ack and uppercase letters correspond to the protospacer, in the gene zwf, preceded for a

PAM site.

Name Primer Sequence Size

(bp)

T

(˚C)

%

G+C

zwf-

pfragfwd F CTTTCGCGCCGAAAATGACCgttttagagctagaaatagcaag 43 68 44

zwf-

pfragrev R GGTCATTTTCGGCGCGAAAGgtgctcagtatctctatcactga 43 71 49

pKDsgRNA

-frag2fwd F ccaattgtccatattgcatca 21 53 38

pKDsgRNA

-frag1rev R tcgagctctaaggaggttataaa 23 54 39

E F

TTAAGTACCGGGTTAGTTAACTTAAGGAGAATG

ACTATCTGCGCTTATCCTTTATGGTTATTTTACCGGT

70 71 37

F R ACCGGTAAAATAACCATAAAGGATAAGCGCAGA TAGTCATTCTCCTTAAGTTAACTAACCCGGTACT

TAA

70 71 37

G F TGACTGAAACGCCTGTAACC 20 56 50

H R CCTGTGTGCCGTGTTAATGA 20 56 50

2.4.3. Plasmid construction and protospacer design

All plasmids used in this work are listed in Table 12. The plasmid pKDsg-zwf was created using

circular polymerase extension cloning (CPEC) [55][69]. Two PCR reactions were performed to

amplify the plasmid in two fragments under the conditions presented in Table 14.

Plasmids Size

(bp)

Antibiotic

resistance genes

Origin of

replication

Copy

Number Purpose

pCas9cr4 6,770 Chloramphenicol p15A ~10

Cas9 nuclease under control of PTET

promoter with ssrA tag and constitutive

tetR

pKDsg-ack 6,959 Spectinomycin ori101 ~5 Template to create the plasmid pKDsg-zwf

pKDsg-zwf 6,959 Spectinomycin ori101 ~5

With homologous regions to the target

gene. Arabinose inducible λ-red and

anhydrotetracycline inducible sgRNA expression

pKDsg-p15 6,962 Spectinomycin ori101 ~5 Eliminate resistance of the plasmid

pCas9cr4

Page 44: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

44

Table 14. PCR reaction and program to generate the two fragments of pKDsg plasmid.

The two linear dsDNA products

were digested with restriction

enzyme DpnI for 1 h at 37ºC and then

resolved in agarose gel [55]. DpnI is

a restriction enzyme that cleaves

only when its recognition site is

methylated (Dam methylated),

therefore it digests the template

plasmid but not the PCR product

[70], [71].

The digested products were resolved

in 1% agarose gel and the correct

fragments were excised, purified and

quantified.

The two-purified linear ssDNA were

used as template for a CPEC

reaction. CPEC reaction is a method

to extend overlapping regions

between the insert and vector

fragments to form a complete

circular plasmid (Figure 16) [72]. In

this case, the two fragments that

result from the PCR reaction and

digestion with DpnI, share

overlapping sequences on both ends.

Therefore, these two fragments can

hybridize and extend using each

other as a template in order to form a

complete double- stranded plasmid

[72].

PCR reaction PCR program

KOD Hot Start DNA Polymerase 1.0 μL Incubation 95 ˚C 2

min

1

cycle MgSO4 2.3 μL

pDNA 10 ng Denaturation 95 ˚C 30 s

35 c

ycl

es Primer F 1.3 μL

Primer R 1.3 μL Annealing 59 ˚C 30 s

dNTPs 2.5 μL

10x buffer for KOD DNA Polymerase 1.0 μL Extension 70 ˚C 1

min Water Up to 25 μL

Figure 16. A schematic diagram of the proposed CPEC mechanism for

cloning an individual gene. The fragment 1 (orange line) and the fragment 2 (blue line) share overlapping regions at the ends. The hybridized

fragments extend using each other as a template until they complete a full

circle (black line) and reach their own 5’-ends. The assembled plasmid

has two nicks, one on each strand. They can be used for transformation with or without further purification. Adapted from [72], [87].

Page 45: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

45

CPEC reaction was carried out in a thermocycler. The PCR reaction and program are present in

Table 15.

Table 15. PCR reaction and program to generate the pKDsg-zwf plasmid.

PCR reaction PCR program

KOD Hot Start DNA Polymerase 1.0 µL Denaturation 95°C 1 min

35 c

ycl

es MgSO4 2.3 µL

Fragment 1 10 µL Annealing 55°C 30 s

Fragment 2 10 µL

dNTPs 2.5 µL Extension 70°C 1 min

10x buffer for KOD DNA Polymerase 2.5 µL

Water Up to 30 µL

4 µL of the CPEC reaction product were added to 100 μl of chemical competent DH5α cells

aliquot and transformed as described in section 2.2.2. A colony from LB + spectinomycin plate

was selected and grown in 5 mL LB broth supplemented with 25 µL spectinomycin, overnight.

On the next day, the bacterial suspension was centrifuged, the pellet was purified to collect the

plasmid. The purified plasmid was quantified and 1,000 ng of the constructed plasmid (pKDsg-

zwf) was analyzed in 1 % agarose gel. The protospacer (target) was designed from pKDsg-ack

plasmid. The protospacer (target) must precede a triplet NGG site known as the protospacer

adjacent motif (PAM) and consist in a 20 bp targeting sequence. The PAM is necessary for Cas9

nuclease to bind target DNA [55]. Primers for cloning the protospacer into the pKDsgRNA

plasmid are represent in Table 13.

Page 46: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

46

2.4.4. Gene disruption strategy

The strategy used in this work was adapted

from no-SCAR (Scarless Cas9 Assisted

Recombineering) system described by Reisch

and Prather [55]. It is a new tool that can edit

the genome of E. coli without chromosomal

markers, using λ-Red system, as used by

Datsenko and Wanner [1]. λ-Red system

facilitates genomic integration of donor DNA

and dsDNA cleavage by Cas9 nuclease to

countselect against wild-type cells [55]. The

plasmids used in this strategy (Table 12) have

temperature sensitive origin of replication can

easily be cured.

CRISPR/Cas9 system to deletion of the zwf

gene is composed by 4 main steps,

represented in Figure 17:

1) Transformation of E. coli strain

GALGNEW with pCas9cr4 plasmid.

2) Transformation of E. coli strain

GALGNEW with pKDsg-zwf

plasmid.

3) Recombineering.

4) Plasmid curing.

The first step of this method was the

transformation of pCas9cr4 plasmid into

chemically competent GALGNEW by heat

shock, as described in point B of section

2.2.2. On the next day, transformants were

grown in 5 mL LB broth supplemented with

25 µL of spectinomycin (50 µg/ mL) and

chemically competent cells were prepared.

The second step was the transformation of the chemically competent GALGNEW + pCas9cr4 by

heat shock with pKDsg-zwf plasmid which contains the protospacer (target gene).

The steps 3 and 4 were not performed.

2.5. Gel extraction and purification

When the product of PCR’s amplification is used to transform cells, the product was verified and

extracted from an agarose gel, dividing the volume into two lanes (5 µL + 15 µL). After

Figure 17. Schematic representation of main steps of no-SCAR method. Adapted from [55].

Page 47: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

47

electrophoresis, the agarose gel was cut into two parts. To minimize damage to the DNA, the right

part of the gel (lane with 15 µL of PCR product) was not exposed to ethidium bromide solution

and UV light. Cutting out the band was required to purify the DNA fragment. The excised

fragment was transferred to a 15 mL Falcon.

For the purification of DNA from TAE agarose gels, the commercial kit: QIAquick Gel Extraction

Kit from Qiagen was used. This kit can be used to purify DNA fragments from 70 bp to 10 kb

[73]. The steps 4 and 6 of the protocol are optional and were not performed.

2.6. Plasmid DNA purification

Plasmid DNA was purified from cells harvested at the end of overnight growth (OD600nm = 1-2)

and from cells harvested from shake flask cultivations (OD600nm =10), using the High Pure Plasmid

Isolation Kit (Roche) and their recommend protocol [74]. The optional steps were not performed.

DNA was eluted with water.

2.7. Plasmid DNA restriction

The purified plasmid DNAs were digested with one restriction enzyme (RE) selected to give a

distinct DNA band pattern or a band profile. The REs selected and respective buffers for the used

plasmids are illustrated in Table 16.

Each reaction was performed with 1,000 ng pDNA, 0.5 µL enzyme, respective buffer (10 % of

final volume) and milli-Q water up to the required final volume. The mixture was incubated at 37

˚C during 2 h. The digested products were separated in a 1 % agarose gel.

Table 16. Restriction enzymes and their characteristics used in plasmids DNA digestions reactions.

The composition of each restriction enzyme reaction buffer (1x) is present in Table 17.

Restriction

Enzymes

Buffer

(% Activity) Site Plasmid DNA Reference

BamHI E (100 %) 5’…A|GATCT…3’

3’… TCTAG|A…5’

pKD46 [75]

pCas9cr4

HindIII E (100 %) 5’…A|AGCTT…3’

3’…TTCGA|A…5’

pKDsg-p15

pKDsg-ack [76]

EcoRI H (100 %) 5’…G|AATTC…3’

3’…CTTAA|G…5’

pKD46 [77]

pVAX1-GFP

KpnI J (100 %) 5’…GGTAC|C…3’

3’…C|CATGG…5’ pCas9cr4 [78]

BglII D (100 %) 5’…A|GATCT…3’

3’…TCTAG|A…5’ pKD13 [79]

Page 48: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

48

Table 17. Composition of Restriction Enzyme Reaction Buffers [80].

Buffer pH

(at 37 °C)

Tris-HCl

(mM)

MgCl2

(mM)

NaCl

(mM) KCl (mM)

DTT

(mM)

D 7.9 6 6 150 — 1

E 7.5 6 6 100 — 1

H 7.5 90 10 50 — —

J 7.5 10 7 — 50 1

2.8. General PCR parameters

The PCR reactions were used to generate the kanamycin cassette and to show that all mutants

have the expected modification [1]. A freshly isolated colony was suspended in 70 µL sterile

water with a plastic tip. The DNA extraction was performed by thermal lysis placing 50 µL of the

mixture at 99 ˚C for 5 min and after kept in ice. The mixture was centrifuged at 12,000 g for 2

min. The reaction with all compounds was prepared. The PCR reactions were performed in a

thermal cycler Hybaid PX2 from Thermo Scientific. Each program was adapted depending on the

purpose.

2.9. Agarose electrophoresis

Gel electrophoresis analysis were performed using 1% (w/ v) agarose gel (500 bp to 1 kb of DNA)

in TAE buffer (40 mM Tris, 20 nM acetic acid and 1 mM EDTA, pH 8.0), in horizontal

electrophoresis tanks from VWR and Electrophoresis Power supply – EPS 301 from Amersham

Pharmacia Biotech. Electrophoresis was carried out for 1 h at 100 V (small agarose gel – 8 wells)

or 1 h 30 min at 120 V (medium or large agarose gels – 15 or 30 wells, respectively), using TAE

buffer 1x as the running buffer. Gels were stained with ethidium bromide (EtBr) (0.5 µg/ mL) and

visualized under UV light on an Eagle Eye II ® Stratagene trans-illuminator. The molecular

weight marker used was NZYDNA ladder III.

2.10. Shake flask cultivation

The inocula were prepared from frozen cell banks of transformed cells with pVAX1GFP plasmid)

in LB medium supplemented with kanamycin, grown overnight and then used to inoculate batch

culture to an initial OD600nm of approximately 0.1. Culture was grown at 37 ˚C for 24 h in 250 mL

shake flask containing 50 mL of the complex medium supplemented with kanamycin, at 250 rpm.

Glucose (20 g/ L) was used as the primary carbon source. Two replicates were prepared for each

strain. Samples were collected at 0, 4, 8, 10, 17 and 24 h to measure pH, glucose and bacterial

density (OD600nm).

Page 49: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

49

In this work, based on the above description, three strategies of shake flask cultivation were

performed. These strategies differ in the number of the strains used, in the number of assays

performed and in the harvest time (hours of growth) of the cells to purification and quantification

of the pDNA, as demonstrated in Table 18.

Table 18. Synthesis of the differences between the various strategies of shake flask cultivation.

Strategy Strains Number of

test days

Harvest Time

(OD600nm =10)

C1 GALG20/ GALGNEW 4 8 h

C2 GALG20/GALGNEW 6 24 h

C3 GALG20/GALGNEW/MG1655 1 17 h

2.11. Measurement of glucose

Culture broth samples with a volume equivalent to OD600nm =10 were centrifuged, at 13,000 g for

10 min, and the supernatants were filtered through a 0.22 µm-pore-size filter. Glucose level was

quantified in a high performance liquid chromatography (HPLC) system (Merck Hitachi,

Darmstadt, Germany) equipped with a refractive index detector (L-7490, Merck Hitachi,

Darmstadt, Germany) and Rezex ROA Organic Acid H+ (8%) column (300 mm x 7.8 mm,

Phenomenex), at 65 ˚C. Compounds were quantified from 20 µL sample injections. Sulfuric acid

[(5 mM) (ACROS)] was used as mobile phase at 0.5 mL/ min.

2.12. Plasmid DNA quantification

After plasmid DNA purification, the quantification of the plasmid DNA was performed using

NanoVue Plus Spectrophotometer (GE Healthcare ®). Plasmid quality was assessed by gel

electrophoresis, to determine which were the predominant isoforms.

2.13. Cells banks preparations

Throughout the study frozen cells banks was prepared in a proportion 80 µL cells for 20 µL

glycerol 99 %. The frozen cells banks were store at -80 ͦ C.

Page 50: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

50

2.14. Genomic deletion analysis

There are evidences showing that unintentional genomic deletions or mutations associated with

the λ -Red mutagenesis procedure such as those observed may be a feature of unstable genomic

regions in MG1655 that are centered around insertion sequence (IS) elements [81]. An example

of this feature is GALG20 strain. The presence of one unintended secondary genomic deletion of

approximately 20 kb in the genome of GALG20 strain was identified. This genomic element

coming from the prophage rac, which was the first prophage discovered in E. coli K-12 being

considered as a phage fossil, having been acquired over 4.5 million years ago. In E. coli K-12, rac

harbors 25 genes (intR, kilR, pinR, racC, racR, ralR, recE, recT, rzoR, sieB, stfR, tfaR, tmpR,

trkG, ydaC, ydaE, ydaF, ydaG, ydaQ, ydaS, ydaT, ydaU, ydaV, ynaE, ynaK) and 5 pseudogenes

(ydaW', rzpR', ydaY', ynaA', and lomR') [2], [82]. The elimination of this phagic element leads to

a decrease in resistance to acid stress, oxidate stress and antibiotic stress [2]. The nomenclature

allocated and which will be used throughout the work is rac sequence or genomic deletion.

To analyze this genomic deletion, several PCR reactions were performed throughout the

GALGNEW strain construction, using fnr and ralR oligonucleotides (Table 8). After completing

the elimination of endA, pgi and recA genes, new specific oligonucleotides were designed to test

the absence or presence of unintentional genomic deletion in GALG20 and GALGNEW (Table

19).

Table 19. Oligonucleotides used to test the absence or presence of unintentional genomic deletion. Some

characteristics of these oligonucleotides are present.

Name Primer Sequence (5’ 3’) Size Tm

(°C)

%

GC

GALG20_del1_fwd

(F1) F AGCCAGATACAAGGGGTTGCTGAA 24 62 50

GALG20_fwd2

(F2) F TGTAAATCCAGCTAAGAGGTGAGG 24 58 46

GALG20_del2_ rev

(R1) R CAATATTCCGCTGTCTGAGTGGAC 24 59 50

GALG20_del3_ rev

(R2) R ACTGTTCATAGCCTGCGCCATA 22 60 50

GALG20_rev3

(R3) R ACTCGGGCCTTGTCAGTTATTG 22 59 50

GALG20_rev4

(R4) R TTTCCGATATGCACCAGGCAC 21 59 52

These oligonucleotides were designed from the genome of MG1655 strain and are schematically

represented in Figure 18.

Figure 18. Schematic representation of location of the oligonucleotides in the genome of MG1655 strain.

Genomic DNA 5’ 3’

~19,880 bp

Page 51: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

51

The origin of tested GALG20 colonies were different. Some colonies were from one sub-culture

and others were from 12 sub-cultures. All colonies were tested with several combinations of

oligonucleotides. PCR program and reaction followed the indications presented in Table 20.

The combinations of oligonucleotides were: F1R1, F1R2, F1R3, F1R4, F2R1, F2R2, F2R3 and

F2R4. Some negative and positive controls were prepared. The negative controls were prepared

replacing DNA for sterile milli-Q water, and positive controls were prepared using DNA from

the same colonies but using oligonucleotides for recA gene.

After all these PCR reactions, mutant strains were again sent for sequencing.

Table 20. PCR program and PCR reaction using to assess the absence of rac sequence in GALG20.

2.15. DNA sequencing

Plasmid DNA was sequenced by Stabvida and genomic DNA purified were sequenced by

MiSeq® System from Illumina [83] in Instituto Gulbenkian de Ciência (Oeiras).

PCR reaction (Vfinal= 25 μL) PCR program

F1R1

F1R2

F1R3

F1R4

F2R1

F2R2

F2R3

F2R4

NovaTaq Polymerase 12.5 μL Incubation 95 ˚C 7 min 1

cycle

DNA 10.0 μL Denaturation 94 ˚C 1 min

35

cycl

es

Primer F 1.0 μL Annealing 55 ˚C 1 min

Primer R 1.0 μL Extension 72 ˚C 1 min

Water 0.5 μL Final Extension 72 ˚C 10 min 1

cycle

Page 52: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

52

3. RESULTS AND DISCUSSION

Plasmid DNA profile

To evaluate the distinct DNA band pattern or band profile of the several plasmids used in this

work, restriction analyses with restriction enzymes were made, as represented in Table 16.

Plasmid pCP20 was not evaluate because its sequence is not available.

The pKD13 plasmid is the template plasmid for gene disruption and was used to design a cassette

containing kanamycin resistance gene (KanR cassette). The band profile of the plasmid pKD13

was analyzed by restriction reaction with enzyme BglII, as represented in Figure 11. For this

reaction one single 3,434 bp fragment is expected. The result is represented in Figure 19.

Lane 1 shows a fragment with size between 3,000 and 4,000 bp, as expected. Lane 2 corresponds

to the non-digested purified plasmid that was used as template to generate the kanamycin cassette.

This demonstrates that the pKD13 plasmid’s sequence is correct.

The plasmid pKD46, as mentioned before, contains the gene that codes for the λ-Red recombinase

and was used to transform cells containing the KanR cassette. This plasmid is essential for the

homologous recombination between the KanR cassette and the genome to occur. Therefore, a

restriction analyses was performed with restriction enzymes BamHI and EcoRI (Figure 11 – B),

separately, to confirm the sequence of the plasmid. The plasmid pKD46 non- digested was also

analyzed in agarose gel.

In Figure 20 the restriction analyses of pKD46 plasmid is represented.

2 1 M

Figure 19. Agarose gel expressing the band profile of plasmid pKD13 digested with BglII (lane 1) and non- digested

(lane 2).

Page 53: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

53

The result of digestion with EcoRI should be the presence of two fragments with 4,820 bp and

1,509 bp. However, in lane 1, three fragments are present with approximately, 10,000 bp, 6,000

bp and 5,000 bp. Alternatively, digestion with BamHI is expected to linearize the plasmid into

one single 6,329 bp fragment. In lane 2, only one band with size between 6,000- 7,500 bp can be

seen, as expected. So, it is possible to assume that digestion was successful and the profile

analyzed correspond to pKD46.

Relative to CRISPR Cas9-System’s plasmids, as mentioned before, the pCas9cr4 carries a Cas9

nuclease gene encoding Cas9 endonuclease, pKDsg-ack was used as template for the construction

of plasmid pKDsg-zwf and pKDsg-p15 to cure the first plasmid. The bands pattern for these

plasmids were analyzed by digestion with restriction enzymes BamHI and HindIII, as depicted in

Table 16. The PCR products were tested by agarose gel electrophoresis (Figure 21). The three

purified plasmids non-digested were also analyzed in an agarose gel.

Figure 20. Qualitative analysis of plasmid pKD46: Plasmid DNA isoforms and agarose gel analysis of restriction

digestion reactions of plasmid pKD46. Lane 1 – purified plasmid digested with EcoRI, lane 2 - purified plasmid

digested with BamHI, lane 3 - purified plasmid undigested. The last lane (M) corresponds to molecular weight marker NZYDNA ladder III.

1 2 3 M

Figure 21. Agarose gel analysis of digestion reactions of pKDsg-ack, pKDsg-p15 and pCas9cr4 plasmids with

restriction enzymes. The first lane (M) is molecular weight marker ladder III. Lane 1 is a plasmid pKDsg-ack non-

digested, lane 2 is plasmid pKDsg-p15 digested with HindIII, lane 3 is plasmid pKDsg-p15 non-digested, lane 4 is pKDsg-ack digested with HindIII, lane 5 is plasmid pCas9cr4 non-digested and lane 6 is plasmid pCas9cr4 digested

with BamHI.

6 5 4 3 2 1 M

Page 54: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

54

The result of digestion of pCas9cr4 with BamHI, should be the presence of one fragment with

6,770 bp, as obtained in lane 6.

For the digestions of pKDsg-ack and pKDsg-p15 plasmids with HindIII were expected one single

6,959 bp and 6,962 bp fragments, respectively.

In lanes 2 and 4 a fragment between 6,000 bp and 7,500 bp can be seen, corresponding to the

expected size for the pKDsg-p15 and pKDsg-ack digested plasmids, respectively. Lane 6,

corresponds to the plasmid pCas9cr4 which was digested with BamHI. The possible fragment

between 6,000 bp and 7,500 bp can be observed, slightly below the obtained fragments for the

other two plasmids.

3.2. Knockouts by Red Disruption System

The protocol used is illustrated in Figure 14.

3.2.1. Kanamycin cassette generation

The first step for the endA (previously knockout by other members of our group), pgi and recA

genes knockouts by Red disruption system was the generation of a cassette with the KanR gene,

as described in section 2.3.3.. This KanR cassette is flanked by FLP recognition target (FRT).

PCR products were generated by using several pairs of 77- to 80-nt-long primers that included

60-nt homology extensions and 17- to 20-nt priming sequences for pKD13 as template,

represented in Table 7. The KanR cassette used to transform MG1655ΔendA to delete the pgi

gene was gently provided by Sofia Duarte, a former PhD student at IST, and Maria Martins, a

former MSc student at IST. The reaction and respective program PCR are represented in Table

9. The homology regions to the genes, the priming sites and the FRT regions in each extremity of

the KanR gene, compose the cassette, leading to a 1,414 bp sequence.

The PCR amplification was verified by agarose gel electrophoresis.

The result present in Figure 22 correspond to PCR reaction to generate KanR cassette for recA

gene knockout.

A band size of approximately 1,400 bp was obtained, corresponding to expected size for the

resistance cassette. The fragment contending KanR cassette present in right part of the agarose gel

was cut by comparison with left part of agarose gel (lane 1, Figure 19). Then, the excised fragment

was purified and the KanR cassette was used to transform MG1655ΔendAΔpgi recombineering-

ready cells.

Page 55: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

55

3.2.2. pgi gene knockout

As mentioned in section 1.4., the pgi gene codes for phosphoglucose isomerase (Pgi), an enzyme

that catalyzes the conversion of glucose-6-phosphate into fructose-6-phosphate. Therefore, the

elimination of Pgi aims to redirect the carbon flux into the PPP, enhance the synthesis of

nucleotides, and also provide high amounts of reducing cofactors, such as NADPH, and

consequently, an increase in pDNA production [48], [59].

After purification of the kanamycin cassette, it was transformed by electroporation into the

recombineering-ready cells from strain MG1655ΔendA. Red-recombinase expressed by plasmid

pKD46, allow the insertion of KanR cassette by exchange with the wild-type pgi gene. To check

for the insertion of the KanR cassette, 6 colonies were selected and a colony PCR was performed,

using the PCR program and the check

primers designed for this purpose (Table

9 and Table 7, respectively). The result

can be seen in Figure 23. Using the

check primers described in Table 7, the

PCR product should be 1,949 bp

fragment, in case of no insertion of the

KanR cassette (corresponding to the pgi

gene size) or a fragment with a size of

1,553 bp, in case of insertion of the KanR

cassette (MG1655 ΔendA::kan).

Figure 22. Agarose gel obtained from the PCR using to generate the KanR cassette for recA gene knockout. In lane (1)

PCR product and in lane (M) molecular weight marker NZYDNA Ladder III.

M 1

1 2 3 4 5 6 7

Figure 23. Agarose gel electrophoresis showing the result of colony PCR of strain MG1655 ΔendA::kan. In the first lane (M)

is molecular weight marker NZYDNA I from NZYTech. The

lanes 1-6 correspond to different six colonies analyzed. Lane 7

corresponds to a negative control performed without DNA.

M

Page 56: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

56

Only four colonies (lanes 3, 4, 5 and 6) exhibit the genotype corresponding to the insertion of the

KanR cassette. This means that the Red-mediated recombination was successful in these colonies

that correspond to these amplifications. The other two colonies (lanes 1 and 2) did not present any

amplification. However, in all lanes the band corresponding to the pgi gene amplification does

not appear. Considering that tested cells were previously selected for kanamycin resistance, after

transformation with KanR cassette, it was expected amplification of a fragment with size

corresponding to insertion of the KanR cassette.

The negative control has not shown amplification, as expected. One of these positive colonies

was grown in LB broth supplemented with kanamycin in order to prepare electrocompetent cells

as described in section 2.2.1. The eletrocompetent cells were electrotransformed with pCP20

plasmid that expresses FLP recombinase able to removes the KanR cassette. The cells transformed

were plated in LB plates supplemented with chloramphenicol. Six LB + Cm positive colonies

were plated in LB plates antibiotic-free and incubated overnight at 43 ˚C. The temperature rise

induces FLP recombinase expression and select for loss of pCP20. After this incubation, the cells

were tested in LB plate without antibiotics and LB plates supplemented with ampicillin,

kanamycin and chloramphenicol. This test is required to confirm complete loss of plasmid from

colonies. In case of cassette removal, the amplified product should be a fragment between 300-

400 bp, depending the cut location within the FRT sites as shown in Figure 24. This resultant

fragment is known as “scar”. As expected, all the colonies tested seem to have lost the KanR

cassette. The negative control has not shown amplification.

Cells banks from colony 2, represented in lane 2 of Figure 24, were made and stored at -80 ˚C.

Figure 24. Agarose gel obtained from final colony PCR used to verify the knockout mutants and the removal of the KanR cassette. In the first lane (M) is molecular weight marker NZYDNA Ladder III. In the following lanes (1- 4) are

the different colonies analized. The last lane correspond to the negative control.

1 2 3 4 5 M

Page 57: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

57

3.2.3. recA gene knockout

The recA gene codes for a polypeptide essential for the recBCD pathway of homologous

recombination (section 1.4.). Thus, recA mutants have less undesirable homologous

recombination than wild-type cells [13].

For the construction of the GALGNEW strain, the MG1655ΔendAΔpgi cells were used as

recipients, specifically the cells stored at -80°C from the colony correspondent to lane 2 in Figure

24. 30 µL aliquot of frozen MG1655ΔendAΔpgi cells was plated in LB without antibiotics. To

confirm endA and pgi genes knockouts, a colony PCR was performed from two colonies using

primers check represented in Table 7. The PCR reaction program is present in Table 10. Controls

were made with both primer pairs: a negative control without DNA and a positive control with

DNA from GALG20. The PCR products were resolved in agarose gel electrophoresis (Figure

25).

The gene disruption method for the recA gene was similar to the previously presented for pgi

gene knockout.

The KanR cassette was generated and purified and then used to transform recombineering-ready

cells from strain MG1655ΔendAΔpgi by electroporation. The transformed cells with KanR

cassette (MG1655 ΔendAΔpgi::kan) were inoculated in LB plate supplemented with kanamycin,

and six colonies were chosen for colony PCR screening. A negative control without DNA and a

positive control choosing a colony MG1655ΔendAΔpgi transformed with pKD46 were prepared.

A colony PCR was accomplished using check primers for recA gene (Table 7). The PCR reaction

and program are described in Table 10.

In case of insertion of the KanR cassette it is expected an amplicon with 1,527 bp. If the insertion

was not successful, the result would be a fragment with 1,371 bp, corresponding to amplification

of recA original gene.

Figure 25. Agarose gel analysis of colony PCR to confirm the endA and pgi genes knockouts using check primers. In the first lane (M) is molecular weight marker NZYDNA Ladder III. Lanes 1 and 2 corresponding to colonies analyzed

with primers to check pgi gene knockout. Lane 3 is the positive control. Lane 4 is the negative control using primers

check for pgi gene knockout. Lanes 5 and 6 corresponding to colonies analyzed with primers to check endA gene knockout. Lane 7 is the positive control. Lane 8 is the negative control using primers check for endA gene knockout.

1 2 3 4 5 6 7 8 M

Page 58: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

58

The size of the amplified fragment was screened by agarose gel electrophoresis (Figure 26).

Four attempts to insert the KanR cassette failed. Possible explanations are a contamination of the

primers, failure on the expression of recombinase encoded in plasmid pKD46 or due to the size

of KanR cassette (1,414 bp).

The obtained sizes were compared with the positive control (lane 7) that has 150 bp difference in

size comparing with length fragment of transformed with KanR cassette. The lane 6, for example,

shows that insertion of KanR cassette was successful. So the colony correspondent to the lane 6

was used to make the transformation with plasmid pCP20 protocol.

After transformation of cells with plasmid pCP20, the mixture was inoculated in LB

supplemented with chloramphenicol and then six colonies were streaked to LB plate without

antibiotics divided into six areas (one for each colony). This LB plate was incubated at 43 ˚C

overnight. On the next day, in order to confirm the mutation, one colony from each area was

selected for colony PCR analysis. For this reaction, the check primers for recA gene presented in

Table 7 were used. The PCR reaction conditions are represented in Table 10. A negative control

prepared with water was included in PCR reaction.

The PCR products were resolved in agarose gel electrophoresis (Figure 27).

Figure 26. Agarose gel with the amplified fragments from colony PCR to confirm the insertion of KanR cassette. In

first lane (M) is the molecular weight molecular NZYDNA ladder III and in following lanes are the PCR products. The lanes 1-6 correspond to the colonies chosen from LB+ kan plate. In the lanes 7 and 8 are the positive control and

the negative control, respectively.

8 7 6 5 4 3 2 1 M

Page 59: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

59

In case of gene deletion, the “scar” corresponds to a fragment with size between 300 bp and 400

bp.

It is possible to verify that the size of the obtained fragments corresponds to “scar”, in other words,

the disruption of recA gene was achieved. Relative to the negative control, can be observed a

slight amplification.

Subsequently, genomic DNA was extracted and sequenced for confirm endA, pgi and recA genes

removal. The new strain MG1655 ΔendAΔpgiΔrecA rac+ was designated GALGNEW.

3.2.4. fnr and ralR genes

Over the course of work, the difference between GALG20 and GALGNEW was tested. As

previously said, GALG20 strain has a genomic deletion with a size of approximately 19.8 kb (rac

sequence). Two of the genes belonging to the rac sequence are fnr and ralR genes, having been

created primers for these two genes (Table 8), which will amplify the regions of interest with

approximately 611 bp, for fnr primer and 294 bp, for ralR primer.

To assess the presence of the rac sequence during GALGNEW strain construction, colony PCRs

were performed several times.

Figure 27. Agarose gel analysis of colony PCR to confirm the recA gene disruption. The lanes 1 -6 are the PCR products amplified using check primers for recA gene. The lane 7 correspond to the negative control of the reaction. The last lane

(M) is molecular weight marker NZYDNA ladder III.

7 6 5 4 3 2 1 M

Page 60: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

60

a) Six colonies of MG1655ΔendA cells transformed with pKD46 from two LB plates

supplemented with kanamycin were selected.

The PCR products were resolved by agarose gel electrophoresis being possible to visualize the

result in the Figure 28.

Analyzing only the reactions used to test the presence of rac sequence, all colonies tested show a

positive result being represented a fragment of approximately 600 bp (lanes 8 to 13) and 400 bp

(lanes 15 to 20) which corresponds to the amplification of the fnr and ralR genes, correspondingly.

These results confirm the presence of rac sequence.

Figure 28. Agarose gel analysis of colony PCR to confirm the insertion of KanR cassette in MG1655 ∆endA + pKD46

cells and to test the presence of rac.

Lanes 1 -6 are the PCR products amplified using check primers for pgi gene.

Lane 7 corresponds to the negative control without DNA, using the same primers as in the previous samples. Lanes 8-13 and lanes 15-20 are the PCR products amplified using primers for fnr and ralR genes, respectively, in order

to confirm the presence of rac.

Lanes 14 and 21 correspond to the negative control of each of the combinations of primers.

Lanes M1, M2 and M3 are molecular weight marker NZYDNA ladder I, HyperLadder™ 50 bp and NZYDNA ladder III, respectively.

7 6 5 4 3 2 1 M2 M1 M3 8 9 10 11 12

M3 13 14 15 16 17 18 19 20 21

M1 M2

M3

Page 61: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

61

b) Four colonies of MG1655ΔendAΔpgi cells from LB plate without antibiotics after

curing the plasmids at 43 ºC were selected.

The PCR products were resolved by agarose gel electrophoresis. The results are represented in

Figure 29.

Analyzing only the reactions used to test the presence of rac sequence, it is possible to verify that

the results are identical to the previous test.

The four colonies tested show a positive result presenting a fragment of approximately 600 bp

(lanes 1 to 4) correspondent to the amplification of the fnr gene, and 400 bp (lanes 6 to 9)

correspondent to the amplification of the ralR gene. These results confirm the presence of rac

sequence.

Posteriorly, the colonies tested were plated onto LB plate without antibiotics, LB plate

supplemented with kanamycin, LB plate supplemented with chloramphenicol and LB plate

supplemented with ampicillin and incubated at 30 ºC. On next day, a colony PCR was performed

7 6 5 4 3 2 1 M M 8 9 10

M 11 12 13 14 15

Figure 29. Agarose gel analysis of colony PCR to assess the cure of the plasmids and consequently deletion of pgi

gene in MG1655 ∆endAΔpgi cells and to test the presence of rac.

Lanes 1 -4 and 6-9 are the PCR products amplified using primers for fnr and ralR genes, respectively, to confirm the

presence of rac.

Lanes 5 and 10 correspond to the negative control of each of the combinations of primers. Lanes 11-14 are the PCR products amplified with check primers for pgi gene.

Lane 15 corresponds to the negative control without DNA, using the same primers as in the previous samples.

Lanes M are molecular weight marker NZYDNA ladder I.

Page 62: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

62

to verify if the procedure was successful, selecting two colonies from LB plate. The resulting

product was analyzed in agarose gel electrophoresis, yielding the gel in Figure 30 .

The presence/ absence of rac sequence was assessed, one more time, using the colony

correspondent to lane 2 (MG1655ΔendAΔpgi cells) in Figure 24 and other one colony of

GALG20 strain (deletion confirmed by sequencing). A negative control without DNA was added

to PCR reaction for each primer pairs. The PCR products were resolved by agarose gel

electrophoresis (Figure 31). Therefore, it was expected that GALG20 strain does not have

amplify with primers for fnr and ralR genes.

Figure 30. Agarose gel analyses of PCR reaction to test the presence of the rac sequence using primers for fnr and

ralR genes. In the first lane (M) is molecular weight marker ladder III. The lanes 1 and 2 are the amplified products

with primers for the fnr gene. The lane 3 is the negative control prepared with water and primers for fnr gene. The

lanes 4 and 5 are the amplified products using primers for the ralR gene. The lane 6 is the negative control prepared with water and primers for ralR gene.

1 6 5 4 3 2 M

Figure 31. Agarose gel analyses of PCR reaction in order to test the presence of the rac sequence using primers for

fnr and ralR genes. In the first lane (M) is molecular weight marker ladder III.

Lanes 1 and 2 are the products resulting from amplification of DNA of the MG1655ΔendAΔpgi cells with primers

for the fnr gene.

Lane 3 is the product resulting from amplification of DNA of the GALG20 with primers for the fnr gene. Lane 4 is the negative control prepared with water and primers for fnr gene.

Lanes 5 and 6 are the amplified products using DNA of the MG1655ΔendAΔpgi cells and primers for the ralR gene.

Lane 7 is the product resulting from amplification of DNA of the MG1655ΔendAΔpgi cells and primers for ralR gene

Lane 8 is the negative control prepared with water and primers for ralR gene.

M 6 5 4 3 2 1 8 7

Page 63: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

63

Once again, the obtained results allow to infer that the rac sequence is present in the genome of

the strain under construction. As expected, the colony of GALG20 tested shows a negative result

correspondent to the absence of rac sequence, as seen in lanes 3 and 6 of Figure 31.

c) Six colonies of MG1655ΔendAΔpgiΔrecA cells from LB plate without antibiotics

after curing the plasmids at 43 ºC were selected.

The PCR products were resolved by agarose gel electrophoresis being possible to visualize the

result in the Figure 32.

Subsequently, the positive results, correspondent to the recA gene elimination (“scar”), were re-

streaked onto a new LB plate. After this step, a new colony PCR reaction was performed selecting

three colonies from LB plate and using fnr and ralR primers. The resulting product was analyzed

in agarose gel electrophoresis, yielding the gel in Figure 33.

M 6 5 4 3 2 1 8 7 10 9

M 16 15 14 13 12 11 18 17 20 21 19

Figure 32. Agarose gel analyses of PCR reaction to test the presence of the rac sequence, using primers for fnr and ralR genes, and to assess the cure of the plasmids and consequently deletion of recA gene.

Lanes M are molecular weight marker ladder III.

Lanes 1-7 are the products resulting from amplification of DNA of the MG1655ΔendAΔpgiΔrecA cells with check primers for the recA gene.

Lanes 8-13 are the products resulting from amplification of DNA of the MG1655ΔendAΔpgiΔrecA cells with primers for

the fnr gene.

Lane 14 is the negative control prepared with water and primers for fnr gene. Lanes 15-20 are the amplified products using DNA of the MG1655ΔendAΔpgiΔrecA cells and primers for the ralR gene.

Lane 21 is the negative control prepared with water and primers for ralR gene.

Page 64: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

64

As seen in Figure 32 and Figure 33, it is possible to conclude that the rac sequence is present in

new strain, GALGNEW, although a fragment is visible in the negative controls of each reaction

(Lane 14 in Figure 32 and lanes 4-(A) and 4-(B) in Figure 33).

These results also attest the difference between the two engineered strains (GALG20 and

GALGNEW), mentioned in section 2.14..

3.3. CRISPR Cas9- System method

3.3.1. Construction of plasmid pKDsg-zwf

As mentioned in section 2.4.3, the pKDsg-zwf plasmid was constructed from pKDsg-ack

plasmid, by cloning the specific target. For this, two PCR reactions and one CPEC reaction were

made. The two PCR products were separated by agarose gel electrophoresis. According to the

sequence of the pKDsg-ack plasmid analyzed by the ApE software, the expected fragment 1

should have 2,845 bp generated using oligonucleotides “zwf-pfragfwd” and “pKDsgRNA-

frag1rev”, and fragment 2 should have 4,414 bp, resulted from the amplification with

oligonucleotides “pKDsgRNA-frag2fwd” and “zwf-pfragrev”. These oligonucleotides are

presented in Table 13.

The obtained results were the expected and can be seen in Figure 34 – (A).

Figure 33. Agarose gel analyses of PCR reaction to test the presence of the rac sequence, using primers for ralR (A) and fnr (B) genes.

Lanes M are molecular weight marker ladder III.

Image (A): Lanes 1 to 3 are the products resulting from amplification of DNA of the MG1655ΔendAΔpgiΔrecA cells with primers for the ralR gene.

Lane 4 is the negative control prepared with water and primers for ralR gene.

Image (B): Lanes 1 to 3 are the products resulting from amplification of DNA of the MG1655ΔendAΔpgiΔrecA cells

with primers for the fnr gene.

Lane 4 is the negative control prepared with water and primers for fnr gene.

Page 65: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

65

After excision and purification of the fragments a quantification of fragments concentration was

made, 20.5 ng/ µL for the fragment 1 and 35.0 ng/ µL for the fragment 2. Subsequently, these two

fragments were ligated by a CPEC reaction (section 2.4.3) and transformed into chemically

competent DH5α cells. The transformed cells were plated onto LB plate supplemented with

spectinomycin. 48 hours later, three colonies in the inoculated LB plate, supplemented with

spectinomycin, were observed. After purification, the concentration of 109.0 ng/ µL for pKDsg-

zwf was obtained and the size of the plasmid analyzed in 1 % agarose gel. The result is represented

in Figure 34 – (B). The ligation of these two fragments generated a fragment with approximately

4,000 – 5,000 bp, size slightly smaller than expected (6,959 bp).

3.4. Shake flask cultivation

The pgi gene of the wild type E. coli strain MG1655 was knocked out with the goal of redirecting

the carbon flux into the pentose phosphate pathway to increase nucleotide synthesis (R5P and

E4P) required for the pDNA synthesis and NADPH generation. The elimination of endA and recA

genes were made in wild-type strain (MG1655) to minimize recombination and nonspecific

digestion of DNA, as mentioned in section 1.4.

After the construction of the new strain (GALGNEW), it was necessary to evaluate cell growth

behavior as well as pDNA production. To do that, shake flask cultivations were performed

following three strategies (C1, C2 and C3), using complex medium supplemented with glucose

(20 g/ L) and kanamycin (30 µg/ mL) at pH 7.1, as described in section 2.10. The growth

parameters (OD600nm and pH) of the mutant strains (GALG20 and GALGNEW) (all strategies)

(A) (B)

Figure 34. Agarose gel analyses: (A) result of PCR reaction to generate, separately, two fragments that constitute the pKDsg-zwf plasmid. In the first lane (M) is molecular weight marker ladder III. The lanes 1 and 2 are the amplified

products. (B) After CPEC reaction, the pKDsg-zwf was transformed into DH5α cells, purified and quantified. 1,000

ng of purified plasmid was analized in 1% agarose gel. In the first lane (M) is molecular weight marker ladder III. The

lane 1 is the amplified product corresponding to the pKDsg-zwf plasmid.

2 1 M M 1

Page 66: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

66

and wild type strain (MG1655) (strategy C3) were measured. All strains were transformed with

pVAX1GFP. In strategy C2, contrary to strategy C1, the growth parameters were measured only

at 24 h of growth. The results obtained in each of the strategies carried out were analyzed and

graphically represented Figure 35.

Page 67: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

67

Figure 35. Effect of endA, recA and pgi genes knockout on biomass production and variation of medium pH, following three strategies of shake flask cultivations (C1, C2 and C3). (A) Biomass

produced in GALG20 and GALGNEW strains following strategy C1. Optical density was measured at 0 h, 4 h, 8 h, 10 h and 24 h of growth. Plots depict mean values ± standard error of mean (SEM) of the four independent experiments. (B) Variation of medium pH during growth of GALG20 and GALGNEW strains following strategy C1. pH was measured at 0 h, 4 h, 8 h, 10 h and

24 h of growth. Plots depict mean values ± standard error of mean (SEM) of the four independent experiments at 10 h and 24 h. (C) Biomass produced in GALG20 and GALGNEW strains

following strategy C2. Optical density was measured at 24 h of growth, during six days. (D) Variation of medium pH during growth of GALG20 and GALGNEW strains following strategy C2.

pH was measured at 24 h of growth. Plots depict mean values ± standard error of mean (SEM) of the six independent experiments. (E) Biomass produced in GALG20, GALGNEW and MG1655 strains following strategy C3. Optical density was measured at 0 h, 4 h, 8 h, 10 h, 17 h and 24 h of growth. Plots depict mean values ± standard error of mean (SEM) of the one independent

experiment. (F) Variation of medium pH during growth of GALG20, GALGNEW and MG1655 strains following strategy C3. pH was measured at 24 h of growth. Plots depict mean values ±

standard error of mean (SEM) of the one independent experiment at 10 h, 17 h and 24 h.

C A B

F E D

Page 68: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

68

The data show that the mutant strains (GALG20 and GALGNEW) demonstrated to have a similar

growth behavior, reaching stationary phase after approximately 8 hours (Figure 35- A), as well

as the pH variation pattern of the medium (Figure 35- B). The optical density (OD600nm) of 26.9

was obtained for GALG20 after 10 h, value slightly below in comparison with previous studies,

which obtained an optical density of 30 for GALG20 [49]. The final OD600nm value achieved was

slightly higher for GALGNEW (32.6 ± 2.7) than for GALG20 (31.4 ± 2.0). These final OD600nm

values are similar to the values obtained by carrying out the C2 strategy (Figure 35- C), in which

it is possible verify higher values for the new strain (33.1 ± 1.0) comparing with GALG20 (30.8

± 0.9). Relatively to the pH analysis of the culture medium after 24 h of growth, a significant drop

(approximately 2%) between the strategy C1 (Figure 35- B) and C2 (Figure 35- D) is visible in

both of strains: (to 6.6 ± 0.07 for 6.45 ± 0.03, in GALGNEW, and to 6.4 ± 0.11 for 6.26 ± 0.02 in

GALG20).

To obtain a more complete evaluation of the behavior of the mutant strains in culture medium,

these were compared with MG1655, following the strategy C3. As seen in Figure 35- E, after 10

h, MG1655 did not surpass OD600nm values of 2, an unusual result when compared with previous

studies that indicate OD600nm value of approximately 10, for the same strain at the same time [49].

Nevertheless, the wild-type strain reached a final OD600nm value of 8.8 ± 0.2. As expected,

GALG20 demonstrates a higher growth kinetic when compared to MG1655 during the growth,

although the values obtained are much lower (OD600nm = 11 ± 0.8) than the expected values

(OD600nm = 30), at the 10 h of growth [49]. However, the GALGNEW surpassed the remaining

strains reaching OD600nm value at the same time (10 h) of 14.8 ± 1.2.

Analyzing the variation of pH in this strategy (Figure 35- F), it is possible observe a radically

decrease to 6.6 at 10 h for 4.8 at 24 h for MG1655. For the other two strains, the pH was constant

throughout the growth. As expected, the pH shows similar values between mutant strains because

both strains were deleted in pgi gene [49].

3.5. Measurement of glucose and plasmid DNA quantification

As mentioned in section 1.4., the knockout of the pgi gene redirects glycolytic flux, increasing

fluxes in the pentose phosphate pathway and enhancing nucleotide synthesis and NADPH

production. However, glycolysis would continue due to the generation of fructose-6-phosphate

and glyceraldeyde-3-phosphate [48]. Glucose is a preferred carbon source and was used in shake

flask cultivations. This experiment was conducted with an initial concentration of glucose of 20

g/ L. All samples were collected at 0, 4, 8, 10 and 24 h of growth during four days of independent

growth (strategy C1). As described in section 2.11, the samples were centrifuged and the glucose

level was quantified in a high performance liquid chromatography (HPLC) system and after the

glucose consumption was measured as a function of the cell density (OD600nm). Assays with the

wild-type strain were not performed. The results are represented in Figure 36.

Page 69: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

69

The glucose consumption and the biomass formation profiles are similar between the two pgi

mutant strains (GALG20 and GALGNEW). For the same concentration of glucose (20 g/ L),

MG1655 ∆endA∆recA strain did not surpass of biomass of 3.5 ± 0.3 [48].

The strains carrying high-copy pDNA require extra synthesis of nucleotides and that the carbon

flux into the PP pathway may not be sufficient to meet cellular needs. To increase the synthesis

of these elements (nucleotides and reducing cofactors), the pgi gene was knockout [48].

To compare the pDNA production potential of mutant strains (GALG20 and GALGNEW),

samples were collected to an OD600nm = 10 at hour 8 of shake flask cultivation (samples from

strategy C1). Collected samples were centrifuged for cell recovery and then purified using the

High Pure Plasmid Isolation Kit (Roche) and its recommended protocol. After determination of

the concentration using Nanodrop Spectrophotometer, the data were graphically represented in

Figure 37.

The volumetric plasmid yield (mg/ L) is relative to the total productivity of the cell culture, and,

as seen in Figure 37, GALG20 appears to have higher productivity in comparison with

Figure 36. Results of the quantification of glucose consumption throughout the growth versus biomass (OD600nm) for

GALG20 and GALGNEW. The presented results derived from average values of 4 days of growth. Glucose

concentration was measured in duplicates by HPLC.

Figure 37. Quantification of plasmid DNA yield volumetric (mg/ L) using two pgi mutant strains: GALG20 and

GALGNEW grown in glucose, following strategy C1. Strains were grown for 24 h in shake flasks (37 °C, 250 rpm)

with rich medium supplemented with 20 g/ L of glucose. Plots depict mean values ± standard error of mean (SEM)

of the four independent experiments.

8 h of growth

Page 70: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

70

GALGNEW, reaching values of 204.3 ± 44.3 (mg/ L) and 169.2 ± 14.3 (mg/ L), respectively.

The values of volumetric plasmid yield for GALG20 are much higher than the values described

in previous studies of 140.8 ± 0.8 (mg/ L) [48].

The pDNA production profiles between wild-type strains and mutant strains also were studied

(strategy C3). The main difference comparing the previous studies is the time of growth at

which the cells were harvested (hour 17 of growth). As is possible observe in Figure 36, it is

still possible to verify the presence of glucose in the growth medium. The results are present in

Figure 38.

Contrary to that observed in Figure 37, GALGNEW appears to have higher productivity in

comparison with GALG20. The expected result would be a higher plasmid DNA production in

GALG20 and in GALGNEW strains, in comparison to MG1655 cells. However, the differences

in pVAX1GFP production between the GALG20 and MG1655 strains are not as high as described

in previous studies, probably due to deletion of pgi gene.

According to the literature, the effect of glucose on pDNA production is not the same in all of

strains. In order to determine this effect, Gonçalves and co-workers [48] studied the influence of

the concentration of glucose on pDNA production in several strains, such as: MG1655

∆endA∆recA and GALG20, in three different conditions: 5 g/ L of glucose initially plus 10 g/ L

of glucose after 12 h, 10 g/ L of glucose initially plus 10 g/ L of glucose after 12 h and 20 g/ L

with no extra addition of glucose. The authors verified that MG1655 ∆endA∆recA produced 5-

fold more pVAX1GFP in 5 + 10 g/ L of glucose than 20 g/ L of glucose. However, the same result

was not obtained for GALG20, which appears to have higher productivity in 20 g/ L of glucose

[10.9 ± 0.2 (mg/ L)], when comparing with the other percentage of carbon sources. The deletion

of endA, recA and pgi genes contributed to an increase in pVAX1GFP production. A significant

difference between GALG20 and GALGNEW can be observed, where the new strain appears to

have higher productivity than other strains. This difference may be due to the presence of the rac

sequence in the genome of GALGNEW.

Figure 38. Quantification of plasmid DNA yield volumetric (mg/ L) using two pgi mutant strains: GALG20 and

GALGNEW, and wild-type strain: MG1655, grown in glucose, following strategy C3. Strains were grown for 24 h in shake flasks (37 °C, 250 rpm) with rich medium supplemented with 20 g/ L of glucose. Plots depict mean values of the

one independent experiment.

Page 71: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

71

3.6. Genomic deletion analysis

The presence of one unintended secondary genomic deletion of approximately 20 kb in the

genome of GALG20 strain (rac sequence) was identified. As mentioned in section 2.14, several

PCR reactions were realized using primers for fnr and ralR genes. Posteriorly, new confirmations

to assess the absence of rac sequence in GALG20 were performed after 12 subcultures and one

subculture in LB plate, to achieve purer colonies. Several combinations of primers presented

inside and outside of genomic deletion were using, as illustrated in Figure 18.

The results obtained are present in Figure 39. In order to facilitate the analysis of the results, a

table was created with the expected results and the results obtained for each sample (Table 21).

Figure 39. Agarose gel analyses of PCR reaction to screen for the genomic deletion in GALG20 strain. The ninth

lane (L) is molecular weight marker ladder III. Lanes 1 -8 are amplified DNA from fresh colony of one subculture.

Lanes 9 -16 are amplified DNA from fresh colony of twelve subcultures. Lanes 17- 24 are negative controls prepared without DNA. Lane 25 is a positive control prepared with primers for recA gene.

Page 72: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

72

Table 21. Expected and obtained results for the PCR reaction to test the genomic deletion in GALG20 strain.

When analyzing the obtained PCR

analysis results it is possible to verify

that the strain GALG20 presents a

dubious result (orange boxes) with

the possibility of mixed population in

the same colony, even after 12

continuous subcultures. These results

are contradictory with those obtained

in the first sequencing of GALG20,

which demonstrated the absence of

rac sequence.

To unravel this doubt, the genome of

the strain was again sequenced.

Relatively to GALGNEW, as

mentioned throughout the results

presented in section 3.2.4., the

presence of this phage sequence (rac

sequence) in its genome is

guaranteed.

Lanes

Combination

Primers

Expected

result

(bp)

Obtained

result (bp) G

AL

G2

0

(1 s

ub

cult

ure

)

1 F1R1 0 Inconclusive

2 F1R2 616 600

3 F1R3 0 Inconclusive

4 F1R4 859 >600

5 F2R1 0 >200

6 F2R2 0 800

7 F2R3 0 400

8 F2R4 0 0

GA

LG

20

(12

su

bcu

ltu

res)

9 F1R1 0 Inconclusive

10 F1R2 616 >600

11 F1R3 0 Inconclusive

12 F1R4 859 >600

13 F2R1 0 ~200

GA

LG

20

(12

subcu

ltu

res)

14 F2R2 0 0

15 F2R3 0 400

16 F2R4 0 0

Neg

ativ

e co

ntr

ols

17 F1R1 0 400-600

18 F1R2 0 600

19 F1R3 0 600-800

20 F1R4 0 600-800

21 F2R1 0 200

22 F2R2 0 0

23 F2R3 0 200-400

24 F2R4 0 0

Po

siti

ve

Co

ntr

ols

25 recA ~400 200-400

Page 73: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

73

3.7. DNA sequencing results

GALG20 and GALGNEW strains were sequenced by Instituto Gulbenkian de Ciência.

The sequenced genome was analyzed in Geneious 6.1.8 software by Professor Leonilde Moreira

and doctor Inês Silva Nunes.

The genomic deletion region is located, approximately, between the position 1,397,239 bp and

the position 1,417,119 bp. When this genomic fragment, with around 20 kb, is present as in case

of GALGNEW strain, many reads are observed in this region (Figure 40).

The sequencing of the GALGNEW strain (Figure 40 – A) allowed not only to confirm the correct

deletion of the endA, pgi and recA genes but also to validate the results obtained in PCR reactions,

in which fnr and ralR primers were used. According to the results obtained by sequencing, the rac

sequence is not present in the GALG20 genome (Figure 40 – B), although this result was not

obtained in all PCR reactions carried out.

Figure 40. Output of genomic DNA sequencing of mutant strains. Analysis of genomic deletion sequence. Top

figure: Segment of genomic DNA of GALGNEW strain. Bottom figure: Segment of genomic DNA of GALG20

strain.

A

B

Page 74: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

74

4. Conclusion and Future Work

Plasmid DNA market is expected to increase in the next years, being necessary to create a system

with high productivity and low costs. Several studies have been performed with the goal of

improving both upstream and downstream processes of plasmid DNA production process.

However, there is still no consensus on what strategy is the best in all parameters.

The goal of this project was to verify the influence of deleted target genes, such as pgi, endA and

recA, on plasmid DNA production on GALGNEW strain, comparing with GALG20 and

MG1655. There are a few previous studies that indicate that pgi gene deletion would have a

positive impact on pDNA production by increasing the reducing power available for pDNA [48],

[49]. This result was obtained when the three strains (MG1655, GALG20 and GALGNEW) were

grown under the same conditions and the plasmid DNA production yield was measured.

Differences in growth kinetics were also observed between wild-type strain and mutant strains.

The main difference found between mutant strains in the rac sequence, an unintended secondary

genomic deletion of approximately 20 kb was identified in the genome of GALG20 strain. The

presence of this phagic sequence was confirmed in the genome of GALGNEW strain. Rac

sequence may be the reason why the GALGNEW strain achieves OD600nm values and plasmid

produced values higher than the other two strains. As mentioned in Introduction, the deletion of

rac sequence leads to a decrease in resistance to acid stress, oxidate stress and antibiotic stress.

The second goal of this work was verify the influence of zwf gene deletion on plasmid DNA

production on GALGNEW strain. This objective has not been completed, but I hope to have the

opportunity to further develop it in the future. There are a few previous studies that indicate

several results, such as:

- That overexpression of this gene would have a positive impact on pDNA production by

increasing the reducing power available for pDNA [84].

- The flux distribution in the zwf- over -expressing mutant was similar to that obtained for

the wild-type parent strain [54].

In conclusion, there are still a lot of studies to perform in order to identify a strategy that increases

plasmid productivity and quality, reducing the production costs. In addition to modifying genes

related to plasmid productivity it is also necessary to increase stability and safety. It is known that

plasmid productivity reaches a plateau in which the cells cannot produce higher quantities.

Therefore, as future work, it is necessary to investigate the influence of simultaneous deletion of

endA, pgi, recA and zwf genes in MG1655 strains following CRISPR- Cas9 System protocol [55].

There are several feeding strategies that can be used in the process of pDNA production by

varying the fermentation strategies (batch or fed-batch fermentation), medium composition,

carbon sources at several concentrations (glucose or glycerol), genes to be deleted (pykF, pykA,

ack-pta) and test the overexpression of some genes (zwf and rpiA) using several strains beyond

the wild-type strain (MG1655), such as highly mutagenized genetic background (DH5α) [48].

The single and double knockouts of pykF and pykA leads to an increase of pDNA synthesis in

different E. coli strains, once this deletion reduces the acetate production and increase carbon flux

into the PPP [48].

Page 75: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

75

5. References

[1] K. A. Datsenko, B. L. Wanner, and J. Beckwith, “One-step inactivation of chromosomal genes in

Escherichia coli K-12 using PCR products.”

[2] X. Liu, Y. Li, Y. Guo, Z. Zeng, B. Li, T. K. Wood, X. Cai, and X. Wang, “Physiological Function

of Rac Prophage During Biofilm Formation and Regulation of Rac Excision in Escherichia coli

K-12.,” Sci. Rep., vol. 5, no. November, p. 16074, 2015.

[3] “Proteobacteria.” [Online]. Available: http://www.gbif.org/species/362. [Accessed: 22-Sep-2016].

[4] E. H. Halococcus, N. M. Methanocaldococcus, C. Archaeoglobus, M. Sulfolobus, M.

Methanospirillum, P. Thermococcus, P. Nanoarchaeum, M. Desulfurococcus, P. Thermoproteus,

and T. Ferroplasma, Biology of Microorganisms. .

[5] T. R. Callaway, M. A. Carr, T. S. Edrington, R. C. Anderson, D. J. Nisbet, F. Safety, and S. P.

Ag-, “Diet , Escherichia coli O157 : H7 , and Cattle : A Review After 10 Years,” vol. 1, no. 979,

pp. 67–80, 2007.

[6] J. B. Kaper, J. P. Nataro, and H. L. T. Mobley, “Pathogenic Escherichia coli,” Nat. Rev.

Microbiol., vol. 2, no. 2, pp. 123–140, 2004.

[7] C. H. Henry and L. Xiaoming, “Industrial production of recombinant therapeutics in Escherichia

coli and its recent advancements,” pp. 383–399, 2012.

[8] Nandkishor Jha, “Pharmaceutical Products of Recombinant DNA Technology.” [Online].

Available: http://www.biologydiscussion.com/dna/pharmaceutical-products-of-recombinant-dna-

technology/10015. [Accessed: 25-Sep-2016].

[9] B. J. Bachmann, “Pedigrees of some mutant strains of Escherichia coli K-12,” Bacteriol Rev, vol.

36, no. 4, pp. 525–557, 1972.

[10] F. R. Blattner, G. Plunkett, C. A. Bloch, N. T. Perna, V. Burland, M. Riley, J. Collado-vides, J. D.

Glasner, C. K. Rode, G. F. Mayhew, J. Gregor, N. W. Davis, H. A. Kirkpatrick, M. A. Goeden,

D. J. Rose, B. Mau, and Y. Shao, “The Complete Genome Sequence of Escherichia coli K-12,”

Sci. Mag, vol. 277, no. September, pp. 1453–1462, 1997.

[11] B. J. Bachmann, “Derivations and Genotypes of Some Mutant Derivatives of Escherichia coli K-

12,” Escherichia coli Salmonella typhimurium Cell. Mol. Biol., no. 143, pp. 2460–2488, 1996.

[12] K. Hayashi, N. Morooka, Y. Yamamoto, K. Fujita, K. Isono, S. Choi, E. Ohtsubo, T. Baba, B. L.

Wanner, H. Mori, and T. Horiuchi, “Highly accurate genome sequences of Escherichia coli K-12

strains MG1655 and W3110,” Mol. Syst. Biol., vol. 2, no. 1, p. 2006.0007, 2006.

[13] G. A. L. Gonçalves, D. M. Bower, D. M. F. Prazeres, G. A. Monteiro, and K. L. J. Prather,

“Rational engineering of Escherichia coli strains for plasmid biopharmaceutical manufacturing,”

Biotechnol. J., vol. 7, no. 2, pp. 251–261, 2012.

[14] A. R. L. and O. T. Ramírez, Plasmid DNA Production for Therapeutic Applications, vol. 267.

2004.

[15] et al. Lodish H, Berk A, Zipursky SL, “DNA Cloning with Plasmid Vectors,” Molecular Cell

Biology. 4th edition. New York: W. H. Freeman, 2000. [Online]. Available:

https://www.ncbi.nlm.nih.gov/books/NBK21498/. [Accessed: 08-Oct-2016].

[16] J. Glenting and S. Wessels, “Ensuring safety of DNA vaccines.,” Microb. Cell Fact., vol. 4, p. 26,

2005.

[17] D. L. Montgomery and K. J. Prather, “Design of plasmid DNA constructs for vaccines.,” Methods

Mol. Med., vol. 127, pp. 11–22, 2006.

[18] R. B. Moss, “Prospects for control of emerging infectious diseases with plasmid DNA vaccines.,”

J. Immune Based Ther. Vaccines, vol. 7, p. 3, 2009.

[19] L. Pasteur, “the Development of Vaccines,” vol. 24, no. 70, pp. 19–30, 2010.

[20] and C. P. H. James A Williamsa,*, Aaron E Carnesa, “Plasmid DNA Vaccine vector design:

impact on efficacy, safety and upstream production,” Clin. Lymphoma, vol. 9, no. 1, pp. 19–22,

2010.

[21] “Gene therapy clinical trials worldwide.” [Online]. Available:

http://www.wiley.com//legacy/wileychi/genmed/clinical/. [Accessed: 11-Oct-2016].

[22] P. Bonanni and J. I. Santos, “Vaccine evolution,” Perspect. Vaccinol., vol. 1, no. 1, pp. 1–24,

2011.

[23] J. Gonçalves, “Adaptive immune system,” 2015.

[24] O. Finco and R. Rappuoli, “Designing vaccines for the twenty-first century society,” Front.

Immunol., vol. 5, no. JAN, pp. 865–872, 2014.

Page 76: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

76

[25] B. Ferraro, M. P. Morrow, N. A. Hutnick, T. H. Shin, C. E. Lucke, and D. B. Weiner, “Clinical

applications of DNA vaccines: Current progress,” Clin. Infect. Dis., vol. 53, no. 3, pp. 296–302,

2011.

[26] R. Strugnell, F. Zepp, A. Cunningham, and T. Tantawichien, “Vaccine antigens,” Perspect.

Vaccinol., vol. 1, no. 1, pp. 61–88, 2011.

[27] K. D. and S. V. S. A.K. Singh, A.K. Verma, Neha , Ruchi Tiwari , K. Karthik, “Plant

Vaccine.Pdf,” Journal of Biological Sciences, vol. 14, no. 2. pp. 95–109, 2014.

[28] “UNDERSTANDING VACCINES.” [Online]. Available: http://www.publichealth.org/public-

awareness/understanding-vaccines/vaccines-work/. [Accessed: 08-Oct-2016].

[29] “Types of vaccines.” [Online]. Available: http://www.vaccines.gov/more_info/types/. [Accessed:

08-Oct-2016].

[30] A. Tejeda-Mansir and R. Montesinos, “Upstream Processing of Plasmid DNA for Vaccine and

Gene Therapy Applications,” Recent Pat. Biotechnol., vol. 2, no. 3, pp. 156–172, 2008.

[31] V. B. Pereira, M. Zurita-Turk, T. D. L. Saraiva, C. P. De Castro, B. M. Souza, P. Mancha Agresti,

F. A. Lima, V. N. Pfeiffer, M. S. P. Azevedo, C. S. Rocha, D. S. Pontes, V. Azevedo, and A.

Miyoshi, “DNA Vaccines Approach: From Concepts to Applications,” World J. Vaccines, vol. 4,

no. 2, pp. 50–71, 2014.

[32] D. A. Blanks, “Immunostimulatory sequences in immunotherapy,” Otolaryngol. Head Neck

Surg., pp. 281–285, 2007.

[33] J. H. Van Uden and E. Raz, “Immunostimulatory DNA Sequences,” Methods, vol. 29, pp. 145–

168.

[34] J. A. Williams, “Vector Design for Improved DNA Vaccine Efficacy, Safety and Production,”

Vaccines, vol. 1, pp. 225–249, 2013.

[35] “Vaccine components,” Natl. Cent. Immun. Res. Surveillanve, vol. 2013, no. May, pp. 1–5, 2013.

[36] M. S. Levy, R. D. O’Kennedy, P. Ayazi-Shamlou, and P. Dunnill, “Biochemical engineering

approaches to the challenges of producing pure plasmid DNA,” Trends Biotechnol., vol. 18, no. 7,

pp. 296–305, 2000.

[37] “Large Scale CGMP Manufacturing of pDNA for Gene Therapy.” [Online]. Available:

https://books.google.pt/books?id=iE-alhelixoC&lpg=PA151&ots=GfFJYmmEwt&dq=therapeutic

doses required for cardiovascular diseases are often 1 mg to 10 mg of pDNA&hl=pt-

PT&pg=PA151#v=onepage&q=therapeutic doses required for cardiovascular diseases are often.

[Accessed: 14-Oct-2016].

[38] G. Mor and M. Eliza, “Plasmid DNA Vaccines,” vol. 19, 2001.

[39] P. H. Oliveira, K. J. Prather, D. M. F. Prazeres, and G. A. Monteiro, “Structural instability of

plasmid biopharmaceuticals: challenges and implications,” Trends Biotechnol., vol. 27, no. 9, pp.

503–511, 2009.

[40] P. H. Oliveira and J. Mairhofer, “Marker-free plasmids for biotechnological applications -

implications and perspectives,” Trends Biotechnol., vol. 31, no. 9, pp. 539–547, 2013.

[41] J. R. Cooke, E. A. McKie, J. M. Ward, and E. Keshavarz-Moore, “Impact of intrinsic DNA

structure on processing of plasmids for gene therapy and DNA vaccines,” J. Biotechnol., vol. 114,

no. 3, pp. 239–254, 2004.

[42] V. B. Pillai, M. Hellerstein, T. Yul, R. R. Amara, and L. Robinson, “Comparative studies on in

vitro expression and in vivo immunogenicity of supercoiled and open circular forms of plasmid

DNA vaccines,” vol. 26, no. 8, pp. 1136–1141, 2009.

[43] A. Carattoli, “Plasmids in Gram negatives: Molecular typing of resistance plasmids,” Int. J. Med.

Microbiol., vol. 301, no. 8, pp. 654–658, 2011.

[44] G. del Solar, R. Giraldo, M. J. Ruiz-Echevarría, M. Espinosa, and R. Díaz-Orejas, “Replication

and control of circular bacterial plasmids.,” Microbiol. Mol. Biol. Rev., vol. 62, no. 2, pp. 434–

464, 1998.

[45] Michele A. Kutzler* and David B. Weiner‡, “DNA vaccines: ready for prime time?,” Biophys.

Chem., vol. 257, no. 5, pp. 2432–2437, 2005.

[46] “pVAX1.” [Online]. Available: http://www.yrgene.com/documents/vector/pvax1.pdf. [Accessed:

03-Oct-2016].

[47] D. S. W. Ow, P. M. Nissom, R. Philp, S. K. W. Oh, and M. G. S. Yap, “Global transcriptional

analysis of metabolic burden due to plasmid maintenance in Escherichia coli DH5α during batch

fermentation,” Enzyme Microb. Technol., vol. 39, no. 3, pp. 391–398, 2006.

[48] G. A. L. Gonçalves, D. M. F. Prazeres, G. A. Monteiro, and K. L. J. Prather, “De novo creation of

MG1655-derived E. coli strains specifically designed for plasmid DNA production,” Appl.

Page 77: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

77

Microbiol. Biotechnol., 2013.

[49] C. P. A. Alves, O. D. Duarte, A. Gabriel, and M. F. Prazeres, “Use of an E. coli pgi Knockout

Strain as a Plasmid Producer,” BioPharm Int., no. February, pp. 2–6, 2016.

[50] “endA gene.” [Online]. Available: http://ecocyc.org/gene?orgid=ECOLI&id=EG11336.

[Accessed: 13-Oct-2016].

[51] T. Shibata, “recA protein,” Tanpakushitsu Kakusan Koso., vol. 32, no. 1, pp. 69–76, 1987.

[52] S. Y. Yau, E. Keshavarz-Moore, and J. Ward, “Host strain influences on supercoiled plasmid

DNA production in Escherichia coli: Implications for efficient design of large-scale processes,”

Biotechnol. Bioeng., vol. 101, no. 3, pp. 529–544, 2008.

[53] J. Zhao, T. Baba, H. Mori, and K. Shimizu, “Effect of zwf gene knockout on the metabolism of

Escherichia coli grown on glucose or acetate,” Metab. Eng., vol. 6, no. 2, pp. 164–174, 2004.

[54] C. Nicolas, P. Kiefer, F. Letisse, J. Krömer, S. Massou, P. Soucaille, C. Wittmann, N. D. Lindley,

and J. C. Portais, “Response of the central metabolism of Escherichia coli to modified expression

of the gene encoding the glucose-6-phosphate dehydrogenase,” FEBS Lett., vol. 581, no. 20, pp.

3771–3776, 2007.

[55] C. R. Reisch and K. L. J. Prather, “The no-SCAR (Scarless Cas9 Assisted Recombineering)

system for genome editing in Escherichia coli,” Nat. Publ. Gr., 2015.

[56] W. Saltzman, H. Shen, and J. Brandsma, Eds., DNA Vaccines Methods and protocols, Second.

New Jersey, 2006.

[57] “Electroporation.” [Online]. Available:

https://www.thermofisher.com/pt/en/home/references/gibco-cell-culture-basics/transfection-

basics/transfection-methods/electroporation.html. [Accessed: 19-Nov-2016].

[58] “Transformation by Heat Shock.” [Online]. Available:

https://worldwide.promega.com/resources/pubhub/enotes/how-are-competent-bacterial-cells-

transformed-with-a-plasmid/. [Accessed: 19-Nov-2016].

[59] G. A. L. Gonçalves, K. L. J. Prather, G. A. Monteiro, A. E. Carnes, and D. M. F. Prazeres,

“Plasmid DNA production with Escherichia coli GALG20, a pgi-gene knockout strain:

Fermentation strategies and impact on downstream processing,” J. Biotechnol., vol. 186, pp. 119–

127, 2014.

[60] “oriR101&repA101ts.” [Online]. Available: http://partsregistry.org/Part:BBa_K524000.

[Accessed: 13-Sep-2016].

[61] “Red recombinase plasmid pKD46, complete sequence.” [Online]. Available:

http://www.ncbi.nlm.nih.gov/nuccore/AY048746.1. [Accessed: 01-Aug-2016].

[62] B. Doublet, G. Douard, H. Targant, D. Meunier, J. Y. Madec, and A. Cloeckaert, “Antibiotic

marker modifications of λ- Red and FLP helper plasmids, pKD46 and pCP20, for inactivation of

chromosomal genes using PCR products in multidrug-resistant strains,” J. Microbiol. Methods,

vol. 75, no. 2, pp. 359–361, 2008.

[63] T. Baba, T. Ara, M. Hasegawa, Y. Takai, Y. Okumura, M. Baba, K. A. Datsenko, M. Tomita, B.

L. Wanner, and H. Mori, “Construction of Escherichia coli K-12 in-frame, single-gene knockout

mutants: the Keio collection.,” Mol. Syst. Biol., vol. 2, p. 2006.0008, 2006.

[64] R. Abdoli, S. Amirthalingam, a Lillquist, and J. Nutt, “Effect of L-arabinose on the specific

homologous recombination efficiency using the Lambda Red Recombinase system for gene

disruption of lacI in Escherichia coli C29 cells,” J. Exp. Microbiol. Immunol., vol. 11, no. April,

pp. 120–124, 2007.

[65] “Invitrogen.” [Online]. Available: https://www.thermofisher.com/pt/en/home/life-

science/cloning/competent-cells-for-transformation/chemically-competent/dh5alpha-

genotypes.html. [Accessed: 02-Aug-2016].

[66] “pKDsg-ack plasmid.” [Online]. Available: https://www.addgene.org/62654/. [Accessed: 18-

Aug-2016].

[67] “pKDsg-p15 plasmid.” [Online]. Available: https://www.addgene.org/62656/. [Accessed: 18-

Aug-2016].

[68] “Plasmids 101: Origin of Replication.” [Online]. Available: http://blog.addgene.org/plasmid-101-

origin-of-replication. [Accessed: 12-Sep-2016].

[69] “noSCAR Protocol.” [Online]. Available: https://www.addgene.org/62654/. [Accessed: 27-Sep-

2016].

[70] “DpnI.” [Online]. Available: https://www.neb.com/products/r0176-dpni. [Accessed: 18-Nov-

2016].

[71] I. Plasmid, D. Digest, and M. Primers, “Site Directed Mutagenesis Protocol,” Program, vol. 3, pp.

Page 78: Genomic deletions in Escherichia coli K-12 MG1655 for ...repositorio.ul.pt/bitstream/10451/27675/1/ulfc120782_tm_Diana... · Genomic deletions in Escherichia coli K-12 MG1655 for

78

1–2, 2005.

[72] J. Q. and J. Tian and J. Quan, “Circular Polymerase Extension Cloning of Complex Gene

Libraries and Pathways,” PLoS One, vol. 4, no. 7, p. e6441, 2009.

[73] Qiagen, “QIAquick ® Gel Extraction Kit - Manual,” no. July, pp. 6–7, 2015.

[74] Roche, “High Pure Plasmid Isolation Kit, Version 8 (Handbuch),” Mannheim, Deutschl., 2011.

[75] “BamHI restriction enzymes.” [Online]. Available:

https://worldwide.promega.com/products/cloning-and-dna-markers/restriction-enzymes/bamhi/.

[Accessed: 31-Aug-2016].

[76] “HindIII restriction enzyme.” [Online]. Available:

https://worldwide.promega.com/products/cloning-and-dna-markers/restriction-enzymes/hindiii/.

[Accessed: 31-Aug-2016].

[77] “EcoRI restriction enzyme.” [Online]. Available:

https://worldwide.promega.com/products/cloning-and-dna-markers/restriction-enzymes/ecori/.

[Accessed: 31-Aug-2016].

[78] “KpnI restriction enzyme.” [Online]. Available:

https://worldwide.promega.com/products/cloning-and-dna-markers/restriction-enzymes/kpni/.

[Accessed: 31-Aug-2016].

[79] “BglII restriction enzymes.” [Online]. Available:

https://worldwide.promega.com/products/cloning-and-dna-markers/restriction-enzymes/bglii/.

[Accessed: 31-Aug-2016].

[80] “Composition of Promega Restriction Enzyme Reaction Buffers (1X).” [Online]. Available:

https://www.promega.com/-/media/files/resources/technical-references/restriction-enzyme-1x-

buffer-composition.pdf. [Accessed: 16-Dec-2016].

[81] J. L. Hobman, M. D. Patel, G. A. Hidalgo-Arroyo, S. J. L. Cariss, M. B. Avison, C. W. Penn, and

C. Constantinidou, “Comparative genomic hybridization detects secondary chromosomal

deletions in Escherichia coli K-12 MG1655 mutants and highlights instability in the flhDC

region,” J. Bacteriol., vol. 189, no. 24, pp. 8786–8792, 2007.

[82] “TopicPage for Rac of Escherichia coli K-12.” [Online]. Available:

http://www.ecogene.org/old/topic.php?topic_id=100. [Accessed: 17-Jan-2017].

[83] “Datasheet_Miseq.Pdf.” .

[84] D. S. Cunningham, R. R. Koepsel, M. M. Ataai, and M. M. Domach, “Factors affecting plasmid

production in Escherichia coli from a resource allocation standpoint.,” Microb. Cell Fact., vol. 8,

no. 1, p. 27, 2009.

[85] E. Noor, E. Eden, R. Milo, and U. Alon, “Central Carbon Metabolism as a Minimal Biochemical

Walk between Precursors for Biomass and Energy,” Mol. Cell, vol. 39, no. 5, pp. 809–820, 2010.

[86] S. Z. Geng, X. A. Jiao, Z. M. Pan, Q. Fang, Z. F. Wen, and X. Chen, “A simpler method for the

efficient and precise deletion of genes in Salmonella sp.,” J. Anim. Vet. Adv., vol. 10, no. 16, pp.

2090–2094, 2011.

[87] “CPEC reaction.” [Online]. Available: https://j5.jbei.org/j5manual/pages/22.html. [Accessed: 18-

Nov-2016].

[88] K. Reddy, M. Tam, R. P. Bowater, M. Barber, M. Tomlinson, K. Nichol Edamura, Y. H. Wang,

and C. E. Pearson, “Determinants of R-loop formation at convergent bidirectionally transcribed

trinucleotide repeats,” Nucleic Acids Res., vol. 39, no. 5, pp. 1749–1762, 2011.