28
1 Title: The Principles of Protein Targeting and Transport Across Cell Membranes Yuanyuan Chen 1 , Sri Karthika Shanmugam 1 and Ross E. Dalbey 1 * 1 Department of Chemistry and Biochemistry, Ohio State University, Columbus, Ohio, USA * Corresponding Author: [email protected], 614-292-2384 Abstract The past several decades have witnessed tremendous growth in the protein targeting, transport and translocation field. Major advances were made during this time period. Now the molecular details of the targeting factors, receptors and the membrane channels that were envisioned in Blobel’s Signal Hypothesis in the 1970s have been revealed by powerful structural methods. It is evident that there is a myriad of cytosolic and membrane associated systems that accurately sort and target newly synthesized proteins to their correct membrane translocases for membrane insertion or protein translocation. Here we will describe the common principles for protein transport in prokaryotes and eukaryotes. Keywords: Protein targeting, chaperones, translocase, insertases, energetics

Title: The Principles of Protein Targeting and Transport

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Title: The Principles of Protein Targeting and Transport

1

Title: The Principles of Protein Targeting and Transport Across Cell Membranes

Yuanyuan Chen1, Sri Karthika Shanmugam1 and Ross E. Dalbey1* 1Department of Chemistry and Biochemistry, Ohio State University, Columbus, Ohio, USA *Corresponding Author: [email protected], 614-292-2384

Abstract The past several decades have witnessed tremendous growth in the protein targeting, transport

and translocation field. Major advances were made during this time period. Now the molecular

details of the targeting factors, receptors and the membrane channels that were envisioned in

Blobel’s Signal Hypothesis in the 1970s have been revealed by powerful structural methods. It

is evident that there is a myriad of cytosolic and membrane associated systems that accurately

sort and target newly synthesized proteins to their correct membrane translocases for membrane

insertion or protein translocation. Here we will describe the common principles for protein

transport in prokaryotes and eukaryotes.

Keywords: Protein targeting, chaperones, translocase, insertases, energetics

Page 2: Title: The Principles of Protein Targeting and Transport

2

Introduction

One of the most significant events in the course of evolution is the development of cellular

compartmentalization [1-4]. It enables different physiological functions to be distributed among

different organelles. However, a great challenge arises at the same time; i.e. proteins synthesized

in the cytosol need to be sorted and transported to their destined locations.

In 1971, Blobel and Sabatini reported their signal hypothesis, which proposed that the

fundamental information for protein sorting and translocation is encoded in a signal sequence

found within the nascent chain, that can be recognized and shuttled to the endoplasmic reticulum

(ER) membrane by a “binding factor” [5-7]. Over the past half century, this hypothesis has been

confirmed and elaborated; the cellular targeting signals have been shown to direct proteins to the

mitochondria, chloroplast, peroxisome, and nucleus [8-13]. Playing center stage in protein

translocation among different organelles are the signal sequences of the respective substrates,

cytosolic chaperones, membrane receptors, membrane channels, and energy as reviewed in [14].

In this overview, we will revisit these principles, and update them with new findings from the

past two decades.

Protein translocation systems

In archaea and bacteria, proteins are synthesized by ribosomes in the cytoplasm. Over one-third

of these proteins need to be sorted, and either inserted into the plasma membrane, or exported

across one or two membranes to reach their destined location [15-17].

In contrast, this process is more sophisticated in eukaryotic cells [16], due to the variety of

compartments and endomembrane systems that they possess. The majority of proteins are

synthesized in the cytosol, and inserted into the ER, or imported into the mitochondria,

chloroplasts, peroxisomes or nucleus. Interestingly, some proteins imported into mitochondria

[18] and peroxisome [19] are done so by an ER-mediated process [18,20,21]. Lastly,

mitochondria [22,23] and chloroplasts [24] also have their own DNAs that encode a small group

of proteins that are synthesized by the organellar ribosomes, and inserted into or exported across

the inner membrane (or thylakoid membrane in chloroplasts).

Page 3: Title: The Principles of Protein Targeting and Transport

3

Interestingly, several translocon complexes have evolved in order to facilitate protein

translocation for a variety of substrates. Good examples are the Sec complex [25] and the Twin

arginine translocation (Tat) complex [26,27] for the export of bacterial proteins to the periplasm,

the Sec61 translocon [28-30] and the Get pathway [31,32] for membrane insertion of proteins

into the ER, or the different secretion systems in bacteria [33-36] for transporting proteins to the

extracellular space.

This overview will focus on the major players in protein targeting and translocation, as well as

the common principles that are shared among various systems. Briefly, membrane-targeted

proteins typically contain one or more N-terminal signal sequences that are recognized by

different cytosolic binding partners, which sort newly synthesized proteins to different transport

pathways or organelles. The binding partners include, in some cases, the signal recognition

particle (SRP) for co-translational translocation, or chaperones that help keep newly synthesized

proteins unfolded in the case of post-translational translocation. The binding partners then shuttle

the synthesized proteins to the surface of membranes via interaction with their membrane-

associated receptors, and then hand the proteins over to their respective membrane channels.

This process is most likely driven by a stepwise increment in the binding affinity. The

translocation through membrane channels is generally driven using ATP hydrolysis by motor

ATPases peripherally associated with the channels, or the membrane proton motif force. Finally,

during or after the translocation is complete, the precursor proteins containing cleavable signal

sequences are processed by signal peptidases, and subsequently fold into their mature forms.

Signal Sequence

It was exciting in the 70s and 80s to discover how precursor proteins are targeted to their correct

destinations in the cell and are able to discriminate among different translocation pathways.

Blobel first proposed that proteins destined for export from the cytoplasm are synthesized with

an N-terminal signal sequence that contains the information needed for targeting [5]. This

hypothesis has been supported during the past ~50 years. Moreover, it is clear that different

signal peptides target proteins to different membrane systems in the cell (Figure 1).

Despite the identification of a great number of signal peptides of ER destined proteins in

eukaryotic cells and bacterial signal sequences, no significant sequence conservation could be

Page 4: Title: The Principles of Protein Targeting and Transport

4

found. In fact, artificial signal sequences [37,38] and randomly generated peptides [39] could

function as signal sequences, suggesting that it is the overall properties, rather than the precise

sequence, that play the role in targeting.

ER and bacterial plasma membrane targeting signal peptides contain a three-domain structure: an

N-terminal, 1-5 residues long, positively charged region (N-domain); a central, 7-15 residues

long, hydrophobic region (H-domain); and a C-terminal, 3-7 residues long, polar region (C-

domain) which contains small aliphatic residues at the -1 and -3 position allowing the secretory

protein to bind and be cleaved by signal peptidase [40-44]. It has been proposed that the

hydrophobicity of H-domain plays a role in discriminating between co-translational and post-

translational pathways [45-49]. A hydrophobic H-domain favors SRP mediated co-translational

translocation, while proteins with a less hydrophobic H-domain tend to be transported post-

translationally.

Distinct from ER and bacterial plasma membrane targeting sequences are the N-terminal

mitochondrial presequences [50-53] and chloroplast transit peptides [54,55] that target proteins

to mitochondria and chloroplasts, respectively. They are less hydrophobic than the signal

peptides mentioned earlier. The mitochondria presequence tend to form an amphiphilic D-helix,

while the chloroplast transit peptides are largely unstructured in aqueous solution, but become D-

helical upon insertion [56,57]. Mitochondrial presequences are typically rich in arginine [58,59],

while chloroplast transit peptides are often rich in hydroxylated amino acids [60]. Notably,

proteins synthesized with only a mitochondrial presequence or a chloroplast transit peptide are

destined to the mitochondrial matrix and chloroplasts stroma compartment, respectively. In the

matrix and stroma compartments, the presequence and transit peptide are removed by the

mitochondrial processing peptidase and stroma processing peptidase, respectively [61,62].

In addition, some mitochondrial and chloroplast proteins contain additional sequences to direct

the pre-proteins to other subcellular locations within the organelle. Chloroplast thylakoid

proteins are typically synthesized with a secondary thylakoid signal responsible for thylakoid

targeting, immediately following the transit peptide [63]. The thylakoid signal peptide is cleaved

following translocation into the thylakoid lumen by the thylakoidal processing peptidase [64].

Mitochondrial intermembrane space (IMS) proteins often contain an intermembrane IMS sorting

signal following the presequence. The IMS signal resembles bacterial and ER signal peptides.

Page 5: Title: The Principles of Protein Targeting and Transport

5

Alternatively, some IMS proteins have C-Xn-C motifs in the mature region of the mitochondrial

protein that form disulfide bonds and thereby function in retaining the proteins in the IMS [51].

Although protein import to peroxisome and nucleus seem to have less in common with other

systems, signal sequences are also required for these systems. The type 1 peroxisome targeting

signal (PTS1) is located at the extreme C-terminal, consisting of a conserved tripeptide (SKL).

PTS2 is a conserved nonapeptide (RL-X5-HL) embedded in an N-terminal sequence [65-67].

Nuclear localization signals (NLS) typically consist of one or two stretches of basic amino acids

[68,69].

Cytosolic Sorting and Membrane Receptors

One big challenge for precursor proteins is their tendency to aggregate in the cytosol prior to

translocation. Two major strategies have been developed to solve this problem: coupling

translocation with protein synthesis (co-translational translocation), or recruiting cytosolic

chaperones to prevent aggregation (post-translational translocation). The co-translational

pathway is commonly employed by bacterial inner membrane proteins and ER proteins in

eukaryotic cells that possess hydrophobic signal peptides. Examples of proteins that are co-

translationally imported have also been reported in mitochondria [70,71]. Post-translational

pathway can be found in all protein translocation systems.

Co-translational translocation to the bacterial plasma membrane or the ER requires the initial

recognition of the nascent polypeptide containing a hydrophobic signal peptide or TM segment

by SRP. In E. coli, SRP consists of a stem-loop shaped 4.5S RNA and a single polypeptide Ffh

with three conserved domains: an N-terminal N domain; a Ras-like G domain with GTPase

activity; and a C-terminal methionine-rich M domain, which possesses a hydrophobic groove for

signal peptide binding [72,46]. After substrate recognition, SRP shuttles the ribosome-nascent

chain complex (RNC) to its membrane associated receptor (SR, FtsY in E. coli). FtsY is also

composed of three domains: The N and G domains, similar to the NG domains of SRP; and an

acidic A domain involved in membrane binding [73]. The GTP-loaded NG domains of SRP and

FtsY interact with each other to form a hetero dimer, bringing the RNC to the vicinity of the Sec

translocon. The dimer formation also triggers the hydrolysis of GTPs, leading to the hand-over of

the RNC to the translocon and the release of SRP [74,75]. A snapshot of the molecular details of

Page 6: Title: The Principles of Protein Targeting and Transport

6

these interactions and how the hand-over takes place have been provided by a cryo-electron

microscopy study where a complex of the ribosome nascent chain, SRP, SR and the SecYEG

was formed [75] (Figure 2).

Post-translational translocation is arguably less understood but typically occurs when the signal

peptide is less hydrophobic. Most precursor proteins that are exported post-translationally require

cytosolic chaperones to keep them in an unfolded state. Trigger factor or SecB binds to the

preproteins that are not recognized by SRP in the cytosol, and deliver them to a SecY-associated

SecA [76-81]. In bacteria, the proteins that go post-translational are secreted proteins while most

membrane proteins go co-translational. In ER targeting, Hsp70 [82-84] or calmodulin [85] have

been reported to function as cytosolic binding factors, and Sec72 has been suggested to play the

role of a membrane receptor for Hsp70 in yeast [86]. In mitochondrial import, the presequences

are recognized by membrane receptors Tom20/Tom22 or Tom70, with the assistance of cytosolic

heat shock proteins Hsp70 and Hsp90 [87]. In the case of chloroplast import, transit peptides are

recognized by Hsp70/14-3-3 or Hsp90 in the cytosol, and the precursor is delivered to the surface

of the outer membrane via the receptors Toc34/Toc159 or Toc64, respectively [88-90].

Preproteins imported into peroxisome or nucleus also require cytosolic binding factors for

targeting, although their subsequent import mechanisms are different from other systems.

Peroxisome destined preproteins with PTS1 signals are recognized by peroxin 5 (Pex5), while

those with PTS2 signals bind to Pex7 [65,66,91-95]. In the case of Pex5, in addition to

recognizing peroxisome proteins, it also forms a part of the translocation channel for the

substrate [96]. Nuclear targeted proteins with NLS are recognized by importin-D (ImpD) and are

transported through nuclear pore complexes (NPC) with the help of carrier proteins like

importin-E (ImpE1) or E-Kap [68,69].

The common features that are shared by cytosolic binding factors and membrane receptors

among different protein translocation systems are: 1) cytosolic binding factors contain binding

sites that recognize the translocation signal of exported proteins. For example, cytosolic factors

that bind preproteins with bacterial or ER signal peptides contain hydrophobic pockets

surrounded by charged and polar amino acids, to accommodate for the H-region and positively

charged N-region of signal sequences, respectively; 2) receptor binding step is generally

accomplished by the formation of heterogenous oligomers between the chaperone and receptor; 3)

Page 7: Title: The Principles of Protein Targeting and Transport

7

Nucleotide triphosphate/nucleotide diphosphate exchange is often involved in receptor binding

and dissociation, but the energy from the hydrolysis at this step is not necessarily essential for

driving protein translocation; 4) the unidirectional hand-over of cargoes from the chaperone to

the receptor, and subsequently to the membrane channel is mostly driven by an increased affinity

chain [45,56].

Membrane channels

Once the preproteins are targeted to the cytosolic surface of the membrane, most are either

inserted into or membrane translocated through hetero-oligomeric translocases with channel-like

structures. A small group of these channels are wide enough for large, folded substrates to pass

through, like the Tat complex found in the plasma membrane in prokaryotes or the thylakoid

membrane of chloroplast in which the TatA component has been suggested to form channels of

various sizes under various conditions [26,97]. However, it is still under debate whether these

TatA-derived channels, which have been observed in detergent, are indeed formed in biological

membranes and function in protein translocation. However, the majority of membrane channels

possess highly regulated pores with relatively small diameters (ranging from 1.4 nm to 2.6 nm),

which allow the translocation of unfolded polypeptides without jeopardizing the permeability

barrier of the membrane [29,98-100].

The most well studied membrane channel is SecYEG in bacteria (Sec61 in eukaryotic cells)

[101,102]. The central component, SecY (Sec61D), is composed of 10 D-helical transmembrane

segments arranged in an hour-glass shape, with the pore ring blocked by a plug domain in the

resting state (Figure 3). The SecY (Sec61D) channel is typically stabilized and regulated by

auxiliary proteins such as SecE, SecG, SecDF, YajC, or SecA in bacteria (Sec61E, Sec61J,

Sec62/63, BiP in the ER) [103,104]. The SecY (Sec61D) channel is capable of both vertical and

lateral opening, and the binding of the signal peptide or stop transfer sequence of the inserting

protein plays an important role in its gate regulation [105,29,106-110]. Details of the lateral

opening and the plug displacement have been elucidated from structures of the Sec channel

actively involved in membrane insertion of a substrate [110].

Page 8: Title: The Principles of Protein Targeting and Transport

8

At the outer membrane of mitochondria and chloroplast, Tom40 and Toc75 form translocation

channels for protein import into mitochondria and chloroplasts [56,99,111]. Interestingly, these

E-barrel channels are capable of forming oligomers. The Tom40 complex, along with the

auxiliary proteins Tom20/Tom22, can contain two to three pore forming units [112,113], while

the Toc75 complex with the auxiliary Toc159/Toc34 components can contain three to four pore

forming channels [114-116]. The exact gating mechanisms of these channels are not clear, but it

has been suggested that the mitochondrial Tom22 and the chloroplast Toc159 proteins might be

involved in regulating their pore opening [117-119]. Once the preproteins are translocated across

the outer membrane, they can either be reinserted into the outer membrane, remain in the inter-

membrane space, or be further imported across the inner membrane to the matrix of

mitochondria or the stroma of chloroplasts. This secondary import step across the inner

membrane requires the membrane channels formed by Tim23 or Tic110, respectively [120,121].

The core architecture of TIM complex is composed of Tim23, which forms the D-helical channel;

Tim50 with a large intermembrane space domain, which functions to recruit the matrix targeting

signal of the imported protein at Tom40 channel and brings it to Tim23 [122] and Tim17 which

coordinates the pmf-dependent channel opening [123]. The TIC channel is mainly composed of

Tic110, and the IMS chaperone, Tic22, which plays a similar shuttle role as Tim50 [124,125].

Tic20 has also been reported to form channels at the inner envelop [126].

The membrane channel system is unique in peroxisome compared to other organelles. It has been

reported that after recognizing preproteins with PTS1, the import receptor Pex5, at least a

fraction of it, is integrated into the membrane, forming part of the translocation channel in

collaboration with Pex14 [96]. Following the completion of translocation, by a mechanism that is

not yet understood, Pex5 is mono- or poly-ubiquitylated, and released back to the cytosol for

recycling [127,128].

In contrast, the Oxa1/Alb3/YidC family members found in mitochondria, chloroplast and

bacteria, respectively, function as insertases without forming channels [131-133]. They contain

a 5-transmembrane-helix core domain that folds in such a way to form a hydrophilic groove

within the inner leaflet of membrane bilayer (Fig. 3). In addition to the polar groove, another

important structural element is the greasy slide where the TM segments of YidC substrates make

contact during insertion. The protein substrates of YidC family insertases are typically

Page 9: Title: The Principles of Protein Targeting and Transport

9

membrane proteins that have small hydrophilic domains that need to be translocated [134-

144,133].

Another unique channel-forming insertase is the BAM family of proteins that insert E-barrel

proteins into the outer membrane of gram-negative bacteria, mitochondria and chloroplasts [145].

The BAM machinery contains a E-barrel domain that catalyzes insertion, and several

polypeptide-transport-associated (POTRA) domains that bind the proteins destined to insert into

the outer membrane (Fig. 3). Two mechanisms have been proposed for the E. coli BAM to insert

and fold E-barrel proteins into the outer membrane. The E-augmentation mechanism proposes

that the substrate initiates E-barrel formation, possibly using the exposed BamA E1 strand as a

template [146]. The protein would then insert into the membrane, possibly using pairs of E-

strands, to form a substrate-BamA super-barrel complex, where the existing E-strand would

function as a template for the newly forming strand. The building of the E-barrel of the substrate

most likely occurs at the exposed edge of the BamA barrel after a lateral opening between the

unstable E1 and E16 strands. When the substrate is still bound to BamA and the E-barrel is

almost completely formed, the complex collapses and the substrate barrel buds off and moves

away from BamA. The second mechanism proposes that BamA functions to thin and destabilize

the membrane region adjacent to the BamA barrel [147,148]. With these perturbations in the

membrane, the substrate is proposed to insert and fold into a E-barrel structure. It should be

noted that the chloroplast Toc75 protein is a member of the BamA family of proteins (Fig. 3e).

The Toc75 POTRA domain has been found to possess chaperone activity and function to

promote import of proteins into chloroplast [149].

Energetics of Protein Translocation

It is clear that energy is required to transport proteins across the hydrophobic barrier of

membranes. For co-translational translocation, GTP hydrolysis occurring during translation

provides the energy source for the coupled translocation process. For post-translational

translocation, a combination of membrane potential and ATP hydrolysis powers the translocation

of proteins across bacterial plasma membrane, ER and inner membrane of mitochondria and the

thylakoid membrane of chloroplasts [150-158].

Page 10: Title: The Principles of Protein Targeting and Transport

10

In several translocation systems, specific subunits function to couple membrane potential (or pmf)

with protein translocation. SecDF possesses a proton tunnel that utilizes the influx of protons to

fuel protein translocation through SecYEG [159]. In mitochondria, Tim17 has been reported to

coordinate membrane potential dependent pore opening of Tim23 [123]. Similar functioning

subunits are yet to be identified for other systems.

ATPase motors are broadly required for protein translocation in different organelles: SecA in

bacterial cytoplasm [160], BiP in ER lumen [161,105], mtHsp70 in the mitochondria matrix

[51,162,50], and cpHsp70 in the stroma of chloroplasts [163].

While SecA, the motor ATPase, performs its function on the cis-side of the membrane, other

ATPases function on the trans-side. For example, figure 4 shows a model of BiP in the ER

lumen binding to Sec63 within the Sec61-Sec63-Sec71-Sec72 complex [104]. Itskanov and Park

[104] used cryo-electron microscopy to solve the structure of the post-translational protein

translocation machinery in yeast, which determined the positioning of the Sec63, Sec71 and

Sec72 on Sec61. Due to flexibility, the Sec62 and the J domain of Sec63 could not be fitted to

the electron density. Sec63 has 3 N-terminal TM segments and the J domain that interacts with

BiP. The structure revealed that it is mainly the 3 N-terminal TM segments and a cytosolic

region of Sec63 (of the Sec71-Sec72 complex), which makes the connections with Sec61. They

showed that the structure had a wide-open lateral gate and an open pore as a result of the Sec63

interactions that would allow it to translocate bigger peptide chains compared to a partially

opened channel with SecA or ribosome bound. They used a homology model based on a

structure of the bacterial J domain bound to Hsp70 complex to provide insight into how the J

domain might bind and position BiP. Intriguingly, the homology model showed that the peptide-

binding cleft of BiP was right below the translocation pore, in an ideal location to latch onto the

polypeptide chain in the ER lumen and promote translocation. It should be noted that insight

into how the Sec61 channel is activated for post-translational translocation was also provided by

Wu et al. where they solved the post-translational Sec complex in yeast as well [164].

One popular model proposed for the ATPase motors is the power stroke mechanism. A recent

study on SecA [165] showed that the two-helix finger of SecA pushes the polypeptide to be

translocated into the SecYEG channel. During ATP hydrolysis, the finger retracts and the clamp

region of SecA binds the preprotein preventing backsliding. Another model argues that the

Page 11: Title: The Principles of Protein Targeting and Transport

11

ATPase motors are likely functioning via a Brownian Ratchet mechanism where the polypeptide

moves through the channel by Brownian diffusion, and the ATPase motors drive translocation

through the channel by repetitive binding, which prevents backsliding [166-168]. In addition, a

recent model called “entropic pulling” has also been proposed [169], suggesting that the

translocation is driven by the change of conformational freedom of both motors and incoming

preprotein.

Peroxisomal import also requires ATP as the energy source, but in a different manner [170].

Although no ATPase activity has been shown to be involved in the import process, the release of

receptor Pex5 requires AAA ATPase Pex1 and Pex6 [171,172]. Since the import of preprotein

and export of Pex5 are closely coupled, this phenomenon is called “export-driven protein import”

[173].

Finally, GTP hydrolysis is involved in nuclear trafficking by creating a RanGTP gradient across

the nuclear envelope, which leads to the unidirectional import of most nuclear proteins [68,174].

Outlook

Considerable progress has been achieved in the protein transport field in the past 40 to 45 years

since Blobel proposed the Signal Hypothesis [5-7]. The signal peptide address codes for

subcellular targeting have been elucidated. Advanced algorithms have been developed to predict

the subcellular localization of a protein based on the signal peptides [175]. We now understand a

great deal about the molecular mechanism of protein targeting. Structures of targeting factors,

chaperones, receptors, and membrane channels have now been solved at atomic resolution

[106,72,108-110,107,29,104,102,145,75]. We can now visualize the ribosome-Sec complex in

the process of inserting a substrate in its membrane environment. We are gaining understanding

about how the transmembrane segments are recognized at the molecular level by the Sec

translocon [75,110,104].

Nonetheless, there are still many intriguing questions yet to be answered. For instance, what are

the structures of the mitochondrial (TIM) and chloroplasts (TOC/TIC) transport channels? What

are the non-traditional protein translocation mechanisms used to import proteins into the

peroxisome and to translocate folded proteins by the Tat pathway? Can single molecule

Page 12: Title: The Principles of Protein Targeting and Transport

12

fluorescence techniques shed light on how proteins pass across the translocation channels in real

time? Obviously, there is much to be done in the field of protein transport and new works will

ultimately lead to deeper understanding of how cellular compartmentalization is achieved.

Acknowledgements

This work was partially supported by National Science Foundation grant MCB-1814936 (R.E.D).

References

1. Martin W, Baross J, Kelley D, Russell MJ (2008) Hydrothermal vents and the origin of life. Nat Rev Microbiol 6 (11):805-814. doi:10.1038/nrmicro1991 2. Lake JA (2009) Evidence for an early prokaryotic endosymbiosis. Nature 460 (7258):967-971. doi:10.1038/nature08183 3. Tocheva EI, Ortega DR, Jensen GJ (2016) Sporulation, bacterial cell envelopes and the origin of life. Nat Rev Microbiol 14 (8):535-542. doi:10.1038/nrmicro.2016.85 4. Gould SB, Garg SG, Martin WF (2016) Bacterial Vesicle Secretion and the Evolutionary Origin of the Eukaryotic Endomembrane System. Trends Microbiol 24 (7):525-534. doi:10.1016/j.tim.2016.03.005 5. Blobel G, Sabatini DD (1971) Ribosome-Membrane Interaction in Eukaryotic Cells. In: Manson LA (ed) Biomembranes: Volume 2. Springer US, Boston, MA, pp 193-195. doi:10.1007/978-1-4684-3330-2_16 6. Blobel G (2000) Protein targeting (Nobel lecture). Chembiochem 1 (2):86-102 7. Blobel G (2000) Protein targeting. Biosci Rep 20 (5):303-344 8. Maccecchini ML, Rudin Y, Blobel G, Schatz G (1979) Import of proteins into mitochondria: precursor forms of the extramitochondrially made F1-ATPase subunits in yeast. Proc Natl Acad Sci U S A 76 (1):343-347. doi:10.1073/pnas.76.1.343 9. Dobberstein B, Blobel G, Chua NH (1977) In vitro synthesis and processing of a putative precursor for the small subunit of ribulose-1,5-bisphosphate carboxylase of Chlamydomonas reinhardtii. Proc Natl Acad Sci U S A 74 (3):1082-1085. doi:10.1073/pnas.74.3.1082 10. Cokol M, Nair R, Rost B (2000) Finding nuclear localization signals. EMBO Rep 1 (5):411-415. doi:10.1093/embo-reports/kvd092 11. Sacksteder KA, Gould SJ (2000) The genetics of peroxisome biogenesis. Annu Rev Genet 34:623-652. doi:10.1146/annurev.genet.34.1.623 12. Gould SG, Keller GA, Subramani S (1987) Identification of a peroxisomal targeting signal at the carboxy terminus of firefly luciferase. J Cell Biol 105 (6 Pt 2):2923-2931 13. Osumi T, Tsukamoto T, Hata S, Yokota S, Miura S, Fujiki Y, Hijikata M, Miyazawa S, Hashimoto T (1991) Amino-terminal presequence of the precursor of peroxisomal 3-ketoacyl-CoA thiolase is a cleavable signal peptide for peroxisomal targeting. Biochem Biophys Res Commun 181 (3):947-954

Page 13: Title: The Principles of Protein Targeting and Transport

13

14. Schatz G, Dobberstein B (1996) Common principles of protein translocation across membranes. Science 271 (5255):1519-1526 15. Dalbey RE, Wang P, Kuhn A (2011) Assembly of bacterial inner membrane proteins. Annu Rev Biochem 80:161-187. doi:10.1146/annurev-biochem-060409-092524 16. Pohlschroder M, Prinz WA, Hartmann E, Beckwith J (1997) Protein translocation in the three domains of life: variations on a theme. Cell 91 (5):563-566 17. Pohlschroder M, Gimenez MI, Jarrell KF (2005) Protein transport in Archaea: Sec and twin arginine translocation pathways. Curr Opin Microbiol 8 (6):713-719. doi:10.1016/j.mib.2005.10.006 18. Hansen KG, Aviram N, Laborenz J, Bibi C, Meyer M, Spang A, Schuldiner M, Herrmann JM (2018) An ER surface retrieval pathway safeguards the import of mitochondrial membrane proteins in yeast. Science 361 (6407):1118-1122. doi:10.1126/science.aar8174 19. Titorenko VI, Ogrydziak DM, Rachubinski RA (1997) Four distinct secretory pathways serve protein secretion, cell surface growth, and peroxisome biogenesis in the yeast Yarrowia lipolytica. Mol Cell Biol 17 (9):5210-5226 20. Titorenko VI, Rachubinski RA (1998) Mutants of the yeast Yarrowia lipolytica defective in protein exit from the endoplasmic reticulum are also defective in peroxisome biogenesis. Mol Cell Biol 18 (5):2789-2803 21. Titorenko VI, Rachubinski RA (1998) The endoplasmic reticulum plays an essential role in peroxisome biogenesis. Trends Biochem Sci 23 (7):231-233 22. Lang BF, Burger G, O'Kelly CJ, Cedergren R, Golding GB, Lemieux C, Sankoff D, Turmel M, Gray MW (1997) An ancestral mitochondrial DNA resembling a eubacterial genome in miniature. Nature 387 (6632):493-497. doi:10.1038/387493a0 23. Pfanner N, Warscheid B, Wiedemann N (2019) Mitochondrial proteins: from biogenesis to functional networks. Nat Rev Mol Cell Biol. doi:10.1038/s41580-018-0092-0 24. Martin W, Stoebe B, Goremykin V, Hapsmann S, Hasegawa M, Kowallik KV (1998) Gene transfer to the nucleus and the evolution of chloroplasts. Nature 393 (6681):162-165. doi:10.1038/30234 25. Orfanoudaki G, Economou A (2014) Proteome-wide subcellular topologies of E. coli polypeptides database (STEPdb). Mol Cell Proteomics 13 (12):3674-3687. doi:10.1074/mcp.O114.041137 26. Palmer T, Berks BC (2012) The twin-arginine translocation (Tat) protein export pathway. Nat Rev Microbiol 10 (7):483-496. doi:10.1038/nrmicro2814 27. Berks BC (2015) The twin-arginine protein translocation pathway. Annu Rev Biochem 84:843-864. doi:10.1146/annurev-biochem-060614-034251 28. Simon SM, Blobel G (1991) A protein-conducting channel in the endoplasmic reticulum. Cell 65 (3):371-380 29. Van den Berg B, Clemons WM, Jr., Collinson I, Modis Y, Hartmann E, Harrison SC, Rapoport TA (2004) X-ray structure of a protein-conducting channel. Nature 427 (6969):36-44. doi:10.1038/nature02218 30. Osborne AR, Rapoport TA, van den Berg B (2005) Protein translocation by the Sec61/SecY channel. Annu Rev Cell Dev Biol 21:529-550. doi:10.1146/annurev.cellbio.21.012704.133214 31. Schuldiner M, Collins SR, Thompson NJ, Denic V, Bhamidipati A, Punna T, Ihmels J, Andrews B, Boone C, Greenblatt JF, Weissman JS, Krogan NJ (2005) Exploration of the function and

Page 14: Title: The Principles of Protein Targeting and Transport

14

organization of the yeast early secretory pathway through an epistatic miniarray profile. Cell 123 (3):507-519. doi:10.1016/j.cell.2005.08.031 32. Schuldiner M, Metz J, Schmid V, Denic V, Rakwalska M, Schmitt HD, Schwappach B, Weissman JS (2008) The GET complex mediates insertion of tail-anchored proteins into the ER membrane. Cell 134 (4):634-645. doi:10.1016/j.cell.2008.06.025 33. Green ER, Mecsas J (2016) Bacterial Secretion Systems: An Overview. Microbiol Spectr 4 (1). doi:10.1128/microbiolspec.VMBF-0012-2015 34. Durand E, Nguyen VS, Zoued A, Logger L, Pehau-Arnaudet G, Aschtgen MS, Spinelli S, Desmyter A, Bardiaux B, Dujeancourt A, Roussel A, Cambillau C, Cascales E, Fronzes R (2015) Biogenesis and structure of a type VI secretion membrane core complex. Nature 523 (7562):555-560. doi:10.1038/nature14667 35. Hu J, Worrall LJ, Hong C, Vuckovic M, Atkinson CE, Caveney N, Yu Z, Strynadka NCJ (2018) Cryo-EM analysis of the T3S injectisome reveals the structure of the needle and open secretin. Nat Commun 9 (1):3840. doi:10.1038/s41467-018-06298-8 36. Low HH, Gubellini F, Rivera-Calzada A, Braun N, Connery S, Dujeancourt A, Lu F, Redzej A, Fronzes R, Orlova EV, Waksman G (2014) Structure of a type IV secretion system. Nature 508 (7497):550-553. doi:10.1038/nature13081 37. Kendall DA, Bock SC, Kaiser ET (1986) Idealization of the hydrophobic segment of the alkaline phosphatase signal peptide. Nature 321 (6071):706-708. doi:10.1038/321706a0 38. Yamamoto Y, Taniyama Y, Kikuchi M, Ikehara M (1987) Engineering of the hydrophobic segment of the signal sequence for efficient secretion of human lysozyme by Saccharomyces cerevisiae. Biochem Biophys Res Commun 149 (2):431-436 39. Kaiser CA, Preuss D, Grisafi P, Botstein D (1987) Many random sequences functionally replace the secretion signal sequence of yeast invertase. Science 235 (4786):312-317 40. von Heijne G (1990) The signal peptide. J Membr Biol 115 (3):195-201 41. Dalbey RE (1995) Signal Peptidases - Intracellular Proteases in the Export Pathway. Asm News 61 (11):586-590 42. Karla A, Lively MO, Paetzel M, Dalbey R (2005) The identification of residues that control signal peptidase cleavage fidelity and substrate specificity. J Biol Chem 280 (8):6731-6741. doi:10.1074/jbc.M413019200 43. Ekici OD, Karla A, Paetzel M, Lively MO, Pei D, Dalbey RE (2007) Altered -3 substrate specificity of Escherichia coli signal peptidase 1 mutants as revealed by screening a combinatorial peptide library. J Biol Chem 282 (1):417-425. doi:10.1074/jbc.M608779200 44. Evans EA, Gilmore R, Blobel G (1986) Purification of microsomal signal peptidase as a complex. Proc Natl Acad Sci U S A 83 (3):581-585 45. Tsirigotaki A, De Geyter J, Sostaric N, Economou A, Karamanou S (2017) Protein export through the bacterial Sec pathway. Nat Rev Microbiol 15 (1):21-36. doi:10.1038/nrmicro.2016.161 46. Akopian D, Shen K, Zhang X, Shan SO (2013) Signal recognition particle: an essential protein-targeting machine. Annu Rev Biochem 82:693-721. doi:10.1146/annurev-biochem-072711-164732 47. Ng DT, Brown JD, Walter P (1996) Signal sequences specify the targeting route to the endoplasmic reticulum membrane. J Cell Biol 134 (2):269-278

Page 15: Title: The Principles of Protein Targeting and Transport

15

48. Ast T, Cohen G, Schuldiner M (2013) A network of cytosolic factors targets SRP-independent proteins to the endoplasmic reticulum. Cell 152 (5):1134-1145. doi:10.1016/j.cell.2013.02.003 49. Neumann-Haefelin C, Schafer U, Muller M, Koch HG (2000) SRP-dependent co-translational targeting and SecA-dependent translocation analyzed as individual steps in the export of a bacterial protein. EMBO J 19 (23):6419-6426. doi:10.1093/emboj/19.23.6419 50. Wiedemann N, Pfanner N (2017) Mitochondrial Machineries for Protein Import and Assembly. Annu Rev Biochem 86:685-714. doi:10.1146/annurev-biochem-060815-014352 51. Backes S, Herrmann JM (2017) Protein Translocation into the Intermembrane Space and Matrix of Mitochondria: Mechanisms and Driving Forces. Front Mol Biosci 4:83. doi:10.3389/fmolb.2017.00083 52. Abe Y, Shodai T, Muto T, Mihara K, Torii H, Nishikawa S, Endo T, Kohda D (2000) Structural basis of presequence recognition by the mitochondrial protein import receptor Tom20. Cell 100 (5):551-560 53. Voos W, Martin H, Krimmer T, Pfanner N (1999) Mechanisms of protein translocation into mitochondria. Biochim Biophys Acta 1422 (3):235-254 54. Bolter B (2018) En route into chloroplasts: preproteins' way home. Photosynth Res 138 (3):263-275. doi:10.1007/s11120-018-0542-8 55. Emanuelsson O, Nielsen H, von Heijne G (1999) ChloroP, a neural network-based method for predicting chloroplast transit peptides and their cleavage sites. Protein Sci 8 (5):978-984. doi:10.1110/ps.8.5.978 56. Schleiff E, Becker T (2011) Common ground for protein translocation: access control for mitochondria and chloroplasts. Nat Rev Mol Cell Biol 12 (1):48-59. doi:10.1038/nrm3027 57. Bruce BD (2000) Chloroplast transit peptides: structure, function and evolution. Trends Cell Biol 10 (10):440-447 58. Huang S, Taylor NL, Whelan J, Millar AH (2009) Refining the definition of plant mitochondrial presequences through analysis of sorting signals, N-terminal modifications, and cleavage motifs. Plant Physiol 150 (3):1272-1285. doi:10.1104/pp.109.137885 59. Vogtle FN, Wortelkamp S, Zahedi RP, Becker D, Leidhold C, Gevaert K, Kellermann J, Voos W, Sickmann A, Pfanner N, Meisinger C (2009) Global analysis of the mitochondrial N-proteome identifies a processing peptidase critical for protein stability. Cell 139 (2):428-439. doi:10.1016/j.cell.2009.07.045 60. Martin T, Sharma R, Sippel C, Waegemann K, Soll J, Vothknecht UC (2006) A protein kinase family in Arabidopsis phosphorylates chloroplast precursor proteins. J Biol Chem 281 (52):40216-40223. doi:10.1074/jbc.M606580200 61. Oblong JE, Lamppa GK (1992) Identification of two structurally related proteins involved in proteolytic processing of precursors targeted to the chloroplast. EMBO J 11 (12):4401-4409 62. Hawlitschek G, Schneider H, Schmidt B, Tropschug M, Hartl FU, Neupert W (1988) Mitochondrial protein import: identification of processing peptidase and of PEP, a processing enhancing protein. Cell 53 (5):795-806 63. Bruce BD (2001) The paradox of plastid transit peptides: conservation of function despite divergence in primary structure. Biochim Biophys Acta 1541 (1-2):2-21 64. Midorikawa T, Endow JK, Dufour J, Zhu J, Inoue K (2014) Plastidic type I signal peptidase 1 is a redox-dependent thylakoidal processing peptidase. Plant J 80 (4):592-603. doi:10.1111/tpj.12655

Page 16: Title: The Principles of Protein Targeting and Transport

16

65. Brocard C, Hartig A (2006) Peroxisome targeting signal 1: is it really a simple tripeptide? Biochim Biophys Acta 1763 (12):1565-1573. doi:10.1016/j.bbamcr.2006.08.022 66. Schliebs W, Kunau WH (2006) PTS2 co-receptors: diverse proteins with common features. Biochim Biophys Acta 1763 (12):1605-1612. doi:10.1016/j.bbamcr.2006.08.051 67. Reumann S (2004) Specification of the peroxisome targeting signals type 1 and type 2 of plant peroxisomes by bioinformatics analyses. Plant Physiol 135 (2):783-800. doi:10.1104/pp.103.035584 68. Christie M, Chang CW, Rona G, Smith KM, Stewart AG, Takeda AA, Fontes MR, Stewart M, Vertessy BG, Forwood JK, Kobe B (2016) Structural Biology and Regulation of Protein Import into the Nucleus. J Mol Biol 428 (10 Pt A):2060-2090. doi:10.1016/j.jmb.2015.10.023 69. Lange A, Mills RE, Lange CJ, Stewart M, Devine SE, Corbett AH (2007) Classical nuclear localization signals: definition, function, and interaction with importin alpha. J Biol Chem 282 (8):5101-5105. doi:10.1074/jbc.R600026200 70. Eliyahu E, Pnueli L, Melamed D, Scherrer T, Gerber AP, Pines O, Rapaport D, Arava Y (2010) Tom20 mediates localization of mRNAs to mitochondria in a translation-dependent manner. Mol Cell Biol 30 (1):284-294. doi:10.1128/MCB.00651-09 71. Lesnik C, Cohen Y, Atir-Lande A, Schuldiner M, Arava Y (2014) OM14 is a mitochondrial receptor for cytosolic ribosomes that supports co-translational import into mitochondria. Nat Commun 5:5711. doi:10.1038/ncomms6711 72. Schaffitzel C, Oswald M, Berger I, Ishikawa T, Abrahams JP, Koerten HK, Koning RI, Ban N (2006) Structure of the E. coli signal recognition particle bound to a translating ribosome. Nature 444 (7118):503-506. doi:10.1038/nature05182 73. Angelini S, Deitermann S, Koch HG (2005) FtsY, the bacterial signal-recognition particle receptor, interacts functionally and physically with the SecYEG translocon. EMBO Rep 6 (5):476-481. doi:10.1038/sj.embor.7400385 74. Shen K, Arslan S, Akopian D, Ha T, Shan SO (2012) Activated GTPase movement on an RNA scaffold drives co-translational protein targeting. Nature 492 (7428):271-275. doi:10.1038/nature11726 75. Jomaa A, Boehringer D, Leibundgut M, Ban N (2016) Structures of the E. coli translating ribosome with SRP and its receptor and with the translocon. Nat Commun 7:10471. doi:10.1038/ncomms10471 76. Castanie-Cornet MP, Bruel N, Genevaux P (2014) Chaperone networking facilitates protein targeting to the bacterial cytoplasmic membrane. Biochim Biophys Acta 1843 (8):1442-1456. doi:10.1016/j.bbamcr.2013.11.007 77. Saio T, Guan X, Rossi P, Economou A, Kalodimos CG (2014) Structural basis for protein antiaggregation activity of the trigger factor chaperone. Science 344 (6184):1250494. doi:10.1126/science.1250494 78. Oh E, Becker AH, Sandikci A, Huber D, Chaba R, Gloge F, Nichols RJ, Typas A, Gross CA, Kramer G, Weissman JS, Bukau B (2011) Selective ribosome profiling reveals the cotranslational chaperone action of trigger factor in vivo. Cell 147 (6):1295-1308. doi:10.1016/j.cell.2011.10.044 79. Chatzi KE, Sardis MF, Economou A, Karamanou S (2014) SecA-mediated targeting and translocation of secretory proteins. Biochim Biophys Acta 1843 (8):1466-1474. doi:10.1016/j.bbamcr.2014.02.014

Page 17: Title: The Principles of Protein Targeting and Transport

17

80. Xu Z, Knafels JD, Yoshino K (2000) Crystal structure of the bacterial protein export chaperone secB. Nat Struct Biol 7 (12):1172-1177. doi:10.1038/82040 81. Bechtluft P, Nouwen N, Tans SJ, Driessen AJ (2010) SecB--a chaperone dedicated to protein translocation. Mol Biosyst 6 (4):620-627. doi:10.1039/b915435c 82. Chirico WJ, Waters MG, Blobel G (1988) 70K heat shock related proteins stimulate protein translocation into microsomes. Nature 332 (6167):805-810. doi:10.1038/332805a0 83. Becker J, Walter W, Yan W, Craig EA (1996) Functional interaction of cytosolic hsp70 and a DnaJ-related protein, Ydj1p, in protein translocation in vivo. Mol Cell Biol 16 (8):4378-4386 84. Deshaies RJ, Koch BD, Werner-Washburne M, Craig EA, Schekman R (1988) A subfamily of stress proteins facilitates translocation of secretory and mitochondrial precursor polypeptides. Nature 332 (6167):800-805. doi:10.1038/332800a0 85. Shao S, Hegde RS (2011) A calmodulin-dependent translocation pathway for small secretory proteins. Cell 147 (7):1576-1588. doi:10.1016/j.cell.2011.11.048 86. Tripathi A, Mandon EC, Gilmore R, Rapoport TA (2017) Two alternative binding mechanisms connect the protein translocation Sec71-Sec72 complex with heat shock proteins. J Biol Chem 292 (19):8007-8018. doi:10.1074/jbc.M116.761122 87. Endo T, Mitsui S, Nakai M, Roise D (1996) Binding of mitochondrial presequences to yeast cytosolic heat shock protein 70 depends on the amphiphilicity of the presequence. J Biol Chem 271 (8):4161-4167 88. Waegemann K, Paulsen H, Soll J (1990) Translocation of Proteins into Isolated-Chloroplasts Requires Cytosolic Factors to Obtain Import Competence. Febs Letters 261 (1):89-92. doi:Doi 10.1016/0014-5793(90)80643-W 89. May T, Soll J (2000) 14-3-3 proteins form a guidance complex with chloroplast precursor proteins in plants. Plant Cell 12 (1):53-64 90. Qbadou S, Becker T, Mirus O, Tews I, Soll J, Schleiff E (2006) The molecular chaperone Hsp90 delivers precursor proteins to the chloroplast import receptor Toc64. EMBO J 25 (9):1836-1847. doi:10.1038/sj.emboj.7601091 91. McCollum D, Monosov E, Subramani S (1993) The pas8 mutant of Pichia pastoris exhibits the peroxisomal protein import deficiencies of Zellweger syndrome cells--the PAS8 protein binds to the COOH-terminal tripeptide peroxisomal targeting signal, and is a member of the TPR protein family. J Cell Biol 121 (4):761-774 92. Glover JR, Andrews DW, Rachubinski RA (1994) Saccharomyces cerevisiae peroxisomal thiolase is imported as a dimer. Proc Natl Acad Sci U S A 91 (22):10541-10545. doi:10.1073/pnas.91.22.10541 93. Stanley WA, Filipp FV, Kursula P, Schuller N, Erdmann R, Schliebs W, Sattler M, Wilmanns M (2006) Recognition of a functional peroxisome type 1 target by the dynamic import receptor pex5p. Mol Cell 24 (5):653-663. doi:10.1016/j.molcel.2006.10.024 94. Einwachter H, Sowinski S, Kunau WH, Schliebs W (2001) Yarrowia lipolytica Pex20p, Saccharomyces cerevisiae Pex18p/Pex21p and mammalian Pex5pL fulfil a common function in the early steps of the peroxisomal PTS2 import pathway. EMBO Rep 2 (11):1035-1039. doi:10.1093/embo-reports/kve228 95. Sichting M, Schell-Steven A, Prokisch H, Erdmann R, Rottensteiner H (2003) Pex7p and Pex20p of Neurospora crassa function together in PTS2-dependent protein import into peroxisomes. Mol Biol Cell 14 (2):810-821. doi:10.1091/mbc.e02-08-0539

Page 18: Title: The Principles of Protein Targeting and Transport

18

96. Meinecke M, Cizmowski C, Schliebs W, Kruger V, Beck S, Wagner R, Erdmann R (2010) The peroxisomal importomer constitutes a large and highly dynamic pore. Nat Cell Biol 12 (3):273-277. doi:10.1038/ncb2027 97. Gohlke U, Pullan L, McDevitt CA, Porcelli I, de Leeuw E, Palmer T, Saibil HR, Berks BC (2005) The TatA component of the twin-arginine protein transport system forms channel complexes of variable diameter. Proc Natl Acad Sci U S A 102 (30):10482-10486. doi:10.1073/pnas.0503558102 98. Hinnah SC, Wagner R, Sveshnikova N, Harrer R, Soll J (2002) The chloroplast protein import channel Toc75: pore properties and interaction with transit peptides. Biophys J 83 (2):899-911. doi:10.1016/S0006-3495(02)75216-8 99. Hill K, Model K, Ryan MT, Dietmeier K, Martin F, Wagner R, Pfanner N (1998) Tom40 forms the hydrophilic channel of the mitochondrial import pore for preproteins [see comment]. Nature 395 (6701):516-521. doi:10.1038/26780 100. Ganesan I, Shi LX, Labs M, Theg SM (2018) Evaluating the Functional Pore Size of Chloroplast TOC and TIC Protein Translocons: Import of Folded Proteins. Plant Cell 30 (9):2161-2173. doi:10.1105/tpc.18.00427 101. Spiess M (2014) Protein translocation: the Sec61/SecYEG translocon caught in the act. Curr Biol 24 (8):R317-319. doi:10.1016/j.cub.2014.02.051 102. Voorhees RM, Fernandez IS, Scheres SH, Hegde RS (2014) Structure of the mammalian ribosome-Sec61 complex to 3.4 A resolution. Cell 157 (7):1632-1643. doi:10.1016/j.cell.2014.05.024 103. Schulze RJ, Komar J, Botte M, Allen WJ, Whitehouse S, Gold VA, Lycklama ANJA, Huard K, Berger I, Schaffitzel C, Collinson I (2014) Membrane protein insertion and proton-motive-force-dependent secretion through the bacterial holo-translocon SecYEG-SecDF-YajC-YidC. Proc Natl Acad Sci U S A 111 (13):4844-4849. doi:10.1073/pnas.1315901111 104. Itskanov S, Park E (2019) Structure of the posttranslational Sec protein-translocation channel complex from yeast. Science 363 (6422):84-87. doi:10.1126/science.aav6740 105. Rapoport TA, Li L, Park E (2017) Structural and Mechanistic Insights into Protein Translocation. Annu Rev Cell Dev Biol 33:369-390. doi:10.1146/annurev-cellbio-100616-060439 106. Tanaka Y, Sugano Y, Takemoto M, Mori T, Furukawa A, Kusakizako T, Kumazaki K, Kashima A, Ishitani R, Sugita Y, Nureki O, Tsukazaki T (2015) Crystal Structures of SecYEG in Lipidic Cubic Phase Elucidate a Precise Resting and a Peptide-Bound State. Cell Rep 13 (8):1561-1568. doi:10.1016/j.celrep.2015.10.025 107. Breyton C, Haase W, Rapoport TA, Kuhlbrandt W, Collinson I (2002) Three-dimensional structure of the bacterial protein-translocation complex SecYEG. Nature 418 (6898):662-665. doi:10.1038/nature00827 108. Voorhees RM, Hegde RS (2016) Structure of the Sec61 channel opened by a signal sequence. Science 351 (6268):88-91. doi:10.1126/science.aad4992 109. Park E, Menetret JF, Gumbart JC, Ludtke SJ, Li W, Whynot A, Rapoport TA, Akey CW (2014) Structure of the SecY channel during initiation of protein translocation. Nature 506 (7486):102-106. doi:10.1038/nature12720 110. Gogala M, Becker T, Beatrix B, Armache JP, Barrio-Garcia C, Berninghausen O, Beckmann R (2014) Structures of the Sec61 complex engaged in nascent peptide translocation or membrane insertion. Nature 506 (7486):107-110. doi:10.1038/nature12950

Page 19: Title: The Principles of Protein Targeting and Transport

19

111. Bolter B, Soll J, Schulz A, Hinnah S, Wagner R (1998) Origin of a chloroplast protein importer. Proc Natl Acad Sci U S A 95 (26):15831-15836. doi:10.1073/pnas.95.26.15831 112. Model K, Meisinger C, Kuhlbrandt W (2008) Cryo-electron microscopy structure of a yeast mitochondrial preprotein translocase. J Mol Biol 383 (5):1049-1057. doi:10.1016/j.jmb.2008.07.087 113. Kunkele KP, Heins S, Dembowski M, Nargang FE, Benz R, Thieffry M, Walz J, Lill R, Nussberger S, Neupert W (1998) The preprotein translocation channel of the outer membrane of mitochondria. Cell 93 (6):1009-1019 114. Chen KY, Li HM (2007) Precursor binding to an 880-kDa Toc complex as an early step during active import of protein into chloroplasts. Plant J 49 (1):149-158. doi:10.1111/j.1365-313X.2006.02944.x 115. Kikuchi S, Hirohashi T, Nakai M (2006) Characterization of the preprotein translocon at the outer envelope membrane of chloroplasts by blue native PAGE. Plant Cell Physiol 47 (3):363-371. doi:10.1093/pcp/pcj002 116. Schleiff E, Soll J, Kuchler M, Kuhlbrandt W, Harrer R (2003) Characterization of the translocon of the outer envelope of chloroplasts. J Cell Biol 160 (4):541-551. doi:10.1083/jcb.200210060 117. van Wilpe S, Ryan MT, Hill K, Maarse AC, Meisinger C, Brix J, Dekker PJ, Moczko M, Wagner R, Meijer M, Guiard B, Honlinger A, Pfanner N (1999) Tom22 is a multifunctional organizer of the mitochondrial preprotein translocase. Nature 401 (6752):485-489. doi:10.1038/46802 118. Poynor M, Eckert R, Nussberger S (2008) Dynamics of the preprotein translocation channel of the outer membrane of mitochondria. Biophys J 95 (3):1511-1522. doi:10.1529/biophysj.108.131003 119. Schleiff E, Jelic M, Soll J (2003) A GTP-driven motor moves proteins across the outer envelope of chloroplasts. Proc Natl Acad Sci U S A 100 (8):4604-4609. doi:10.1073/pnas.0730860100 120. Truscott KN, Kovermann P, Geissler A, Merlin A, Meijer M, Driessen AJ, Rassow J, Pfanner N, Wagner R (2001) A presequence- and voltage-sensitive channel of the mitochondrial preprotein translocase formed by Tim23. Nat Struct Biol 8 (12):1074-1082. doi:10.1038/nsb726 121. Balsera M, Goetze TA, Kovacs-Bogdan E, Schurmann P, Wagner R, Buchanan BB, Soll J, Bolter B (2009) Characterization of Tic110, a channel-forming protein at the inner envelope membrane of chloroplasts, unveils a response to Ca(2+) and a stromal regulatory disulfide bridge. J Biol Chem 284 (5):2603-2616. doi:10.1074/jbc.M807134200 122. Mokranjac D, Sichting M, Popov-Celeketic D, Mapa K, Gevorkyan-Airapetov L, Zohary K, Hell K, Azem A, Neupert W (2009) Role of Tim50 in the transfer of precursor proteins from the outer to the inner membrane of mitochondria. Mol Biol Cell 20 (5):1400-1407. doi:10.1091/mbc.E08-09-0934 123. Demishtein-Zohary K, Gunsel U, Marom M, Banerjee R, Neupert W, Azem A, Mokranjac D (2017) Role of Tim17 in coupling the import motor to the translocation channel of the mitochondrial presequence translocase. Elife 6. doi:10.7554/eLife.22696 124. Glaser S, van Dooren GG, Agrawal S, Brooks CF, McFadden GI, Striepen B, Higgins MK (2012) Tic22 is an essential chaperone required for protein import into the apicoplast. J Biol Chem 287 (47):39505-39512. doi:10.1074/jbc.M112.405100

Page 20: Title: The Principles of Protein Targeting and Transport

20

125. Rudolf M, Machettira AB, Gross LE, Weber KL, Bolte K, Bionda T, Sommer MS, Maier UG, Weber AP, Schleiff E, Tripp J (2013) In vivo function of Tic22, a protein import component of the intermembrane space of chloroplasts. Mol Plant 6 (3):817-829. doi:10.1093/mp/sss114 126. Kovacs-Bogdan E, Benz JP, Soll J, Bolter B (2011) Tic20 forms a channel independent of Tic110 in chloroplasts. BMC Plant Biol 11:133. doi:10.1186/1471-2229-11-133 127. Platta HW, El Magraoui F, Schlee D, Grunau S, Girzalsky W, Erdmann R (2007) Ubiquitination of the peroxisomal import receptor Pex5p is required for its recycling. J Cell Biol 177 (2):197-204. doi:10.1083/jcb.200611012 128. Carvalho AF, Pinto MP, Grou CP, Alencastre IS, Fransen M, Sa-Miranda C, Azevedo JE (2007) Ubiquitination of mammalian Pex5p, the peroxisomal import receptor. J Biol Chem 282 (43):31267-31272. doi:10.1074/jbc.M706325200 129. Paine PL, Moore LC, Horowitz SB (1975) Nuclear envelope permeability. Nature 254 (5496):109-114 130. Stewart M, Baker RP, Bayliss R, Clayton L, Grant RP, Littlewood T, Matsuura Y (2001) Molecular mechanism of translocation through nuclear pore complexes during nuclear protein import. FEBS Lett 498 (2-3):145-149 131. Kumazaki K, Chiba S, Takemoto M, Furukawa A, Nishiyama K, Sugano Y, Mori T, Dohmae N, Hirata K, Nakada-Nakura Y, Maturana AD, Tanaka Y, Mori H, Sugita Y, Arisaka F, Ito K, Ishitani R, Tsukazaki T, Nureki O (2014) Structural basis of Sec-independent membrane protein insertion by YidC. Nature 509 (7501):516-520. doi:10.1038/nature13167 132. Kumazaki K, Kishimoto T, Furukawa A, Mori H, Tanaka Y, Dohmae N, Ishitani R, Tsukazaki T, Nureki O (2014) Crystal structure of Escherichia coli YidC, a membrane protein chaperone and insertase. Sci Rep 4:7299. doi:10.1038/srep07299 133. Chen Y, Dalbey RE (2018) Oxa1 Superfamily: New Members Found in the ER. Trends Biochem Sci 43 (3):151-153. doi:10.1016/j.tibs.2017.12.005 134. Aschtgen MS, Zoued A, Lloubes R, Journet L, Cascales E (2012) The C-tail anchored TssL subunit, an essential protein of the enteroaggregative Escherichia coli Sci-1 Type VI secretion system, is inserted by YidC. Microbiologyopen 1 (1):71-82. doi:10.1002/mbo3.9 135. Chen M, Samuelson JC, Jiang F, Muller M, Kuhn A, Dalbey RE (2002) Direct interaction of YidC with the Sec-independent Pf3 coat protein during its membrane protein insertion. J Biol Chem 277 (10):7670-7675. doi:10.1074/jbc.M110644200 136. Samuelson JC, Jiang F, Yi L, Chen M, de Gier JW, Kuhn A, Dalbey RE (2001) Function of YidC for the insertion of M13 procoat protein in Escherichia coli: translocation of mutants that show differences in their membrane potential dependence and Sec requirement. J Biol Chem 276 (37):34847-34852. doi:10.1074/jbc.M105793200 137. Pross E, Soussoula L, Seitl I, Lupo D, Kuhn A (2016) Membrane Targeting and Insertion of the C-Tail Protein SciP. J Mol Biol 428 (20):4218-4227. doi:10.1016/j.jmb.2016.09.001 138. Neugebauer SA, Baulig A, Kuhn A, Facey SJ (2012) Membrane protein insertion of variant MscL proteins occurs at YidC and SecYEG of Escherichia coli. J Mol Biol 417 (4):375-386. doi:10.1016/j.jmb.2012.01.046 139. Yi L, Celebi N, Chen M, Dalbey RE (2004) Sec/SRP requirements and energetics of membrane insertion of subunits a, b, and c of the Escherichia coli F1F0 ATP synthase. J Biol Chem 279 (38):39260-39267. doi:10.1074/jbc.M405490200

Page 21: Title: The Principles of Protein Targeting and Transport

21

140. van Bloois E, Jan Haan G, de Gier JW, Oudega B, Luirink J (2004) F(1)F(0) ATP synthase subunit c is targeted by the SRP to YidC in the E. coli inner membrane. FEBS Lett 576 (1-2):97-100. doi:10.1016/j.febslet.2004.08.069 141. Yi L, Jiang F, Chen M, Cain B, Bolhuis A, Dalbey RE (2003) YidC is strictly required for membrane insertion of subunits a and c of the F(1)F(0)ATP synthase and SecE of the SecYEG translocase. Biochemistry 42 (35):10537-10544. doi:10.1021/bi034309h 142. van der Laan M, Bechtluft P, Kol S, Nouwen N, Driessen AJ (2004) F1F0 ATP synthase subunit c is a substrate of the novel YidC pathway for membrane protein biogenesis. J Cell Biol 165 (2):213-222. doi:10.1083/jcb.200402100 143. Kuhn A, Koch HG, Dalbey RE (2017) Targeting and Insertion of Membrane Proteins. EcoSal Plus 7 (2). doi:10.1128/ecosalplus.ESP-0012-2016 144. Hennon SW, Soman R, Zhu L, Dalbey RE (2015) YidC/Alb3/Oxa1 Family of Insertases. J Biol Chem 290 (24):14866-14874. doi:10.1074/jbc.R115.638171 145. Noinaj N, Gumbart JC, Buchanan SK (2017) The beta-barrel assembly machinery in motion. Nat Rev Microbiol 15 (4):197-204. doi:10.1038/nrmicro.2016.191 146. Estrada Mallarino L, Fan E, Odermatt M, Muller M, Lin M, Liang J, Heinzelmann M, Fritsche F, Apell HJ, Welte W (2015) TtOmp85, a beta-barrel assembly protein, functions by barrel augmentation. Biochemistry 54 (3):844-852. doi:10.1021/bi5011305 147. Danoff EJ, Fleming KG (2015) Membrane defects accelerate outer membrane beta-barrel protein folding. Biochemistry 54 (2):97-99. doi:10.1021/bi501443p 148. Noinaj N, Kuszak AJ, Gumbart JC, Lukacik P, Chang H, Easley NC, Lithgow T, Buchanan SK (2013) Structural insight into the biogenesis of beta-barrel membrane proteins. Nature 501 (7467):385-390. doi:10.1038/nature12521 149. O'Neil PK, Richardson LGL, Paila YD, Piszczek G, Chakravarthy S, Noinaj N, Schnell D (2017) The POTRA domains of Toc75 exhibit chaperone-like function to facilitate import into chloroplasts. Proc Natl Acad Sci U S A 114 (24):E4868-E4876. doi:10.1073/pnas.1621179114 150. Geller BL (1991) Energy requirements for protein translocation across the Escherichia coli inner membrane. Mol Microbiol 5 (9):2093-2098 151. Zimmermann R, Wickner W (1983) Energetics and intermediates of the assembly of Protein OmpA into the outer membrane of Escherichia coli. J Biol Chem 258 (6):3920-3925 152. Geller BL, Movva NR, Wickner W (1986) Both ATP and the electrochemical potential are required for optimal assembly of pro-OmpA into Escherichia coli inner membrane vesicles. Proc Natl Acad Sci U S A 83 (12):4219-4222. doi:10.1073/pnas.83.12.4219 153. Bolender N, Sickmann A, Wagner R, Meisinger C, Pfanner N (2008) Multiple pathways for sorting mitochondrial precursor proteins. EMBO Rep 9 (1):42-49. doi:10.1038/sj.embor.7401126 154. Mokranjac D, Neupert W (2008) Energetics of protein translocation into mitochondria. Biochim Biophys Acta 1777 (7-8):758-762. doi:10.1016/j.bbabio.2008.04.009 155. Flugge UI, Hinz G (1986) Energy dependence of protein translocation into chloroplasts. Eur J Biochem 160 (3):563-570 156. Shi LX, Theg SM (2013) Energetic cost of protein import across the envelope membranes of chloroplasts. Proc Natl Acad Sci U S A 110 (3):930-935. doi:10.1073/pnas.1115886110

Page 22: Title: The Principles of Protein Targeting and Transport

22

157. Cline K, Ettinger WF, Theg SM (1992) Protein-specific energy requirements for protein transport across or into thylakoid membranes. Two lumenal proteins are transported in the absence of ATP. J Biol Chem 267 (4):2688-2696 158. Berks BC, Sargent F, Palmer T (2000) The Tat protein export pathway. Mol Microbiol 35 (2):260-274 159. Furukawa A, Yoshikaie K, Mori T, Mori H, Morimoto YV, Sugano Y, Iwaki S, Minamino T, Sugita Y, Tanaka Y, Tsukazaki T (2017) Tunnel Formation Inferred from the I-Form Structures of the Proton-Driven Protein Secretion Motor SecDF. Cell Rep 19 (5):895-901. doi:10.1016/j.celrep.2017.04.030 160. Gouridis G, Karamanou S, Sardis MF, Scharer MA, Capitani G, Economou A (2013) Quaternary dynamics of the SecA motor drive translocase catalysis. Mol Cell 52 (5):655-666. doi:10.1016/j.molcel.2013.10.036 161. Hassdenteufel S, Johnson N, Paton AW, Paton JC, High S, Zimmermann R (2018) Chaperone-Mediated Sec61 Channel Gating during ER Import of Small Precursor Proteins Overcomes Sec61 Inhibitor-Reinforced Energy Barrier. Cell Rep 23 (5):1373-1386. doi:10.1016/j.celrep.2018.03.122 162. Craig EA (2018) Hsp70 at the membrane: driving protein translocation. BMC Biol 16 (1):11. doi:10.1186/s12915-017-0474-3 163. Su PH, Li HM (2010) Stromal Hsp70 is important for protein translocation into pea and Arabidopsis chloroplasts. Plant Cell 22 (5):1516-1531. doi:10.1105/tpc.109.071415 164. Wu X, Cabanos C, Rapoport TA (2019) Structure of the post-translational protein translocation machinery of the ER membrane. Nature 566 (7742):136-139. doi:10.1038/s41586-018-0856-x 165. Catipovic MA, Bauer BW, Loparo JJ, Rapoport TA (2019) Protein translocation by the SecA ATPase occurs by a power-stroke mechanism. EMBO J 38 (9). doi:10.15252/embj.2018101140 166. Allen WJ, Corey RA, Oatley P, Sessions RB, Baldwin SA, Radford SE, Tuma R, Collinson I (2016) Two-way communication between SecY and SecA suggests a Brownian ratchet mechanism for protein translocation. Elife 5. doi:10.7554/eLife.15598 167. Yamano K, Kuroyanagi-Hasegawa M, Esaki M, Yokota M, Endo T (2008) Step-size analyses of the mitochondrial Hsp70 import motor reveal the Brownian ratchet in operation. J Biol Chem 283 (40):27325-27332. doi:10.1074/jbc.M805249200 168. Corey RA, Ahdash Z, Shah A, Pyle E, Allen WJ, Fessl T, Lovett JE, Politis A, Collinson I (2019) ATP-induced asymmetric pre-protein folding as a driver of protein translocation through the Sec machinery. Elife 8. doi:10.7554/eLife.41803 169. De Los Rios P, Ben-Zvi A, Slutsky O, Azem A, Goloubinoff P (2006) Hsp70 chaperones accelerate protein translocation and the unfolding of stable protein aggregates by entropic pulling. Proc Natl Acad Sci U S A 103 (16):6166-6171. doi:10.1073/pnas.0510496103 170. Schwerter DP, Grimm I, Platta HW, Erdmann R (2017) ATP-driven processes of peroxisomal matrix protein import. Biol Chem 398 (5-6):607-624. doi:10.1515/hsz-2016-0293 171. Platta HW, Grunau S, Rosenkranz K, Girzalsky W, Erdmann R (2005) Functional role of the AAA peroxins in dislocation of the cycling PTS1 receptor back to the cytosol. Nat Cell Biol 7 (8):817-822. doi:10.1038/ncb1281

Page 23: Title: The Principles of Protein Targeting and Transport

23

172. Miyata N, Fujiki Y (2005) Shuttling mechanism of peroxisome targeting signal type 1 receptor Pex5: ATP-independent import and ATP-dependent export. Mol Cell Biol 25 (24):10822-10832. doi:10.1128/MCB.25.24.10822-10832.2005 173. Schliebs W, Girzalsky W, Erdmann R (2010) Peroxisomal protein import and ERAD: variations on a common theme. Nat Rev Mol Cell Biol 11 (12):885-890. doi:10.1038/nrm3008 174. Koepp DM, Silver PA (1996) A GTPase controlling nuclear trafficking: running the right way or walking RANdomly? Cell 87 (1):1-4 175. Emanuelsson O, Brunak S, von Heijne G, Nielsen H (2007) Locating proteins in the cell using TargetP, SignalP and related tools. Nat Protoc 2 (4):953-971. doi:10.1038/nprot.2007.131 176. Clantin B, Delattre AS, Rucktooa P, Saint N, Meli AC, Locht C, Jacob-Dubuisson F, Villeret V (2007) Structure of the membrane protein FhaC: a member of the Omp85-TpsB transporter superfamily. Science 317 (5840):957-961. doi:10.1126/science.1143860

Page 24: Title: The Principles of Protein Targeting and Transport

24

Figure 1 Targeting signals. Targeting signals for proteins destined to the ER, mitochondria, chloroplasts, peroxisome and nucleus are shown in red and blue. Blank boxes represent hydrophobic helices; boxes with stripes represent amphiphilic helices; chloroplast transit peptides tend to be unstructured in solution, as shown by red line; black arrows represent signal peptidase (processing peptidase) cleavage sites. Figure 2 Structure of the ribosome-SRP-SR-SecYEG quaternary complex. One example of cytosolic sorting and the stepwise delivery of the ribosome-bound SRP protein to the membrane receptor/translocation machinery is shown in the cryo-EM structure of the ribosome-SRP-SR-SecYEG quaternary complex (pdb: 5NCO) (Jomaa et al. [75]). The ribosome had synthesized the FtsQ nascent chain containing the first transmembrane segment. Each of the components are color-coded as follows: ribosome in grey; SRP RNA in orange; SRP M domain in cyan; SRP NG domain in blue; SR NG domain in green; SR A domain (which is not resolved) in light green; SecYEG in red; signal sequence in magenta. Figure 3 Structures of common translocation devices. A. X-ray structure of SecYEG from Thermus thermophiles (pdb: 5AWW). SecY, SecE and SecG subunits are shown in light blue, yellow and magenta, respectively. The lateral gate is shown in red and dark blue. The plug domain is shown in green. B. X-ray structure of YidC from Escherichia coli (pdb: 3WVF). The greasy slide comprised of TM3 and TM5 is shown in dark blue and red, respectively. The conserved positively charged residue in the hydrophilic groove is shown in green. C. Structure of BamABCDE complex. It was modeled by combining X-ray structure of BamACDE (pdb: 5EKQ) and X-ray structure of BamB fused to BamA POTRA domain (pdb: 4PK1) from Escherichia coli. BamA in light blue; BamB in orange; BamC in magenta; BamD in cyan; BamE in green. D. Dimer of Tom40. The model is based on cryo-EM structure from Neurospora crassa (pdb: 5O8O). E. Model of full-length Toc75 from Arabidopsis thaliana adapted from O’Neil et al [149]. X-ray structures of FhaC (pdb: 2QDZ) [176] and Toc75 POTRA1-3 (pdb: 5UAY) are superpositioned by aligning POTRA2 of FhaC (not shown) and POTRA3 (yellow) of Toc75. The barrel domain (blue) is from FhaC, and POTRA domains 1, 2 and 3 (green, purple and yellow respectively) are from Toc75. POTRA2 contains an extra D-helix (P2h), as shown in cyan. Figure 4 Model of the post-translational Sec61 translocase pathway with the BiP ATPase

promoting translocation. The model is constructed by combining the cryo-EM structure of the yeast Sec post-translational complex (pdb: 6N3Q) and the bacterial J domain bound to DnaK Hsp70 (pdb: 5NRO) as a model for J-domain-BiP (see Itskanov et al. [104]). Each of the components are color-coded as follows: Sec61 channel (Sec61D) in light blue; SSS1 (Sec61J) in cyan; SBH1 (Sec61E) in magenta; Sec63 in orange; Sec66 in yellow; Sec72 in green; the homology model of the J-domain of Sec63 in red; the hypothetical model of BiP ATPase in pink. The blue line represents a model of the substrate.

Page 25: Title: The Principles of Protein Targeting and Transport

25

Figure 1

Page 26: Title: The Principles of Protein Targeting and Transport

26

Figure 2

Page 27: Title: The Principles of Protein Targeting and Transport

27

Figure 3

Page 28: Title: The Principles of Protein Targeting and Transport

28

Figure 4