13
Original Article Comparative Characterization of Vaginal Cells Derived From Premenopausal Women With and Without Severe Pelvic Organ Prolapse Hala Kufaishi, MD, MSc 1 , May Alarab, MD, MSc 2,3 , Harold Drutz, MD 2,3 , Stephen Lye, PhD 1,3 , and Oksana Shynlova, PhD 1,3 Abstract Background: This study tested a hypothesis that primary human vaginal cells derived from tissue of premenopausal women with severe pelvic organ prolapse (POP-HVCs) would display differential functional characteristics as compared to vaginal cells derived from asymptomatic women with normal pelvic floor support (control-HVCs). Methods: Vaginal tissue biopsies were collected from premenopausal patients with POP (n ¼ 8) and asymptomatic controls (n ¼ 7) during vaginal hysterectomy or repair. Primary vaginal cells were isolated by enzymatic digestion and characterized by immunocytochemistry. Cell attachment and proliferation on different matrices (collagen I, collagen II, collagen IV, fibronectin, laminin, tenascin, and vitronectin) were compared between POP-HVCs and control-HVCs. RNA was extracted, and the expression of 84 genes was screened using Human Extracellular Matrix and Adhesion Molecules RT 2 Profiler PCR array. The expression of selected genes was verified by quantitative reverse transcription-polymerase chain reaction. Results: (1) Control-HVCs attached to collagen IV more efficiently than POP-HVCs; (2) control-HVCs and POP-HVCs show a similar proliferation rate when plated on proNectin and collagen I; (3) when seeded on collagen I, resting POP-HVCs expressed significantly (P < .05) increased transcript levels of collagen VII, multiple matrix metal- loproteinases (MMP3, MMP7, MMP10, MMP12, MMP13, and MMP14), integrins (ITGA1, ITGA4, ITGA6, ITGA8, ITGB1, ITGB2, and ITGB3), and cell adhesion molecules as compared to control-HVCs. Collagen XV and tissue inhibitors of MMPs (TIMP1 and TIMP2) as well as genes involved in the biogenesis and maturation of collagen and elastin fibers (LOX, LOXL1-LOXL3, BMP1, and ADAMTS2) were significantly downregulated in POP-HVCs versus control-HVCs (P < .05). Conclusions: Resting primary POP- HVCs in vitro show altered cellular characteristics as compared to control-HVCs, which may influence their dynamic responses to external mechanical or hormonal stimuli. Keywords POP, fibroblasts, vagina, premenopausal, ECM Introduction Pelvic floor disorders (PFDs) are widely underrecognized con- ditions that have a significant impact on the quality of women’s life. They include stress urinary incontinence (SUI), anal incontinence, and pelvic organ prolapse (POP). Pelvic floor disorders have a substantial emotional impact on the affected women, which results in social isolation, psychological dis- tress, anxiety, and depression. 1,2 A recent cross-sectional study concluded that 24% of women older than 20 years have at least 1 PFD. 3 For instance, POP affects one-third of premenopausal women and nearly half of postmenopausal women. 4 There are multiple risk factors for the development of POP, with vaginal delivery being the greatest independent inciting risk factor. 5,6 Other factors include age, race, family history and genetics, and most importantly abnormalities in their connective tissue his- tology and morphology. 7-9 Caucasian women have a 5 times higher risk of developing a symptomatic prolapse in compari- son to African American women and 1.4-fold increase in the relative risk of developing severe POP. 8 Currently, treatment of POP is dominated by surgical repair of native tissue with vaginal meshes and generally results in 1 Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada 2 Division of Urogynecology and Reconstructive Pelvic Surgery, Department of Obstetrics and Gynecology, Mount Sinai Hospital, Toronto, Ontario, Canada 3 Department of Obstetrics and Gynecology, University of Toronto, Toronto, Ontario, Canada Corresponding Author: Oksana Shynlova, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 25 Orde Street, Suite 6-1019, Toronto, Ontario, Canada M5G 1X5. Email: [email protected] Reproductive Sciences 2016, Vol. 23(7) 931-943 ª The Author(s) 2016 Reprints and permission: sagepub.com/journalsPermissions.nav DOI: 10.1177/1933719115625840 rs.sagepub.com

Reproductive Sciences Comparative Characterization of Vaginal … · 2020. 7. 27. · prolapsed tissue. This study examined the ability of primary vaginal cells to attach to different

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

  • Original Article

    Comparative Characterization of VaginalCells Derived From PremenopausalWomen With and Without SeverePelvic Organ Prolapse

    Hala Kufaishi, MD, MSc1, May Alarab, MD, MSc2,3, Harold Drutz, MD2,3,Stephen Lye, PhD1,3, and Oksana Shynlova, PhD1,3

    AbstractBackground: This study tested a hypothesis that primary human vaginal cells derived from tissue of premenopausal women withsevere pelvic organ prolapse (POP-HVCs) would display differential functional characteristics as compared to vaginal cells derivedfrom asymptomatic women with normal pelvic floor support (control-HVCs). Methods: Vaginal tissue biopsies were collectedfrom premenopausal patients with POP (n¼ 8) and asymptomatic controls (n¼ 7) during vaginal hysterectomy or repair. Primaryvaginal cells were isolated by enzymatic digestion and characterized by immunocytochemistry. Cell attachment and proliferationon different matrices (collagen I, collagen II, collagen IV, fibronectin, laminin, tenascin, and vitronectin) were compared betweenPOP-HVCs and control-HVCs. RNA was extracted, and the expression of 84 genes was screened using Human ExtracellularMatrix and Adhesion Molecules RT2 Profiler PCR array. The expression of selected genes was verified by quantitative reversetranscription-polymerase chain reaction. Results: (1) Control-HVCs attached to collagen IV more efficiently than POP-HVCs; (2)control-HVCs and POP-HVCs show a similar proliferation rate when plated on proNectin and collagen I; (3) when seeded oncollagen I, resting POP-HVCs expressed significantly (P < .05) increased transcript levels of collagen VII, multiple matrix metal-loproteinases (MMP3, MMP7, MMP10, MMP12, MMP13, and MMP14), integrins (ITGA1, ITGA4, ITGA6, ITGA8, ITGB1, ITGB2,and ITGB3), and cell adhesion molecules as compared to control-HVCs. Collagen XV and tissue inhibitors of MMPs (TIMP1 andTIMP2) as well as genes involved in the biogenesis and maturation of collagen and elastin fibers (LOX, LOXL1-LOXL3, BMP1, andADAMTS2) were significantly downregulated in POP-HVCs versus control-HVCs (P < .05). Conclusions: Resting primary POP-HVCs in vitro show altered cellular characteristics as compared to control-HVCs, which may influence their dynamic responsesto external mechanical or hormonal stimuli.

    KeywordsPOP, fibroblasts, vagina, premenopausal, ECM

    Introduction

    Pelvic floor disorders (PFDs) are widely underrecognized con-

    ditions that have a significant impact on the quality of women’s

    life. They include stress urinary incontinence (SUI), anal

    incontinence, and pelvic organ prolapse (POP). Pelvic floor

    disorders have a substantial emotional impact on the affected

    women, which results in social isolation, psychological dis-

    tress, anxiety, and depression.1,2 A recent cross-sectional study

    concluded that 24% of women older than 20 years have at least1 PFD.3 For instance, POP affects one-third of premenopausal

    women and nearly half of postmenopausal women.4 There are

    multiple risk factors for the development of POP, with vaginal

    delivery being the greatest independent inciting risk factor.5,6

    Other factors include age, race, family history and genetics, and

    most importantly abnormalities in their connective tissue his-

    tology and morphology.7-9 Caucasian women have a 5 times

    higher risk of developing a symptomatic prolapse in compari-

    son to African American women and 1.4-fold increase in the

    relative risk of developing severe POP.8

    Currently, treatment of POP is dominated by surgical repair

    of native tissue with vaginal meshes and generally results in

    1 Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto,

    Ontario, Canada2 Division of Urogynecology and Reconstructive Pelvic Surgery, Department of

    Obstetrics and Gynecology, Mount Sinai Hospital, Toronto, Ontario, Canada3 Department of Obstetrics and Gynecology, University of Toronto, Toronto,

    Ontario, Canada

    Corresponding Author:

    Oksana Shynlova, Lunenfeld-Tanenbaum Research Institute, Mount Sinai

    Hospital, 25 Orde Street, Suite 6-1019, Toronto, Ontario, Canada M5G 1X5.

    Email: [email protected]

    Reproductive Sciences2016, Vol. 23(7) 931-943ª The Author(s) 2016Reprints and permission:sagepub.com/journalsPermissions.navDOI: 10.1177/1933719115625840rs.sagepub.com

    http://www.sagepub.com/journalsPermissions.navhttp://rs.sagepub.com

  • good anatomic and subjective outcomes.10 However, the use of

    meshes is associated with considerable risk of erosion, pain,

    infection, and vaginal stenosis.11 More recently, a tissue engi-

    neering approach was suggested to be a good alternative to

    POP repair surgery.12 The goal is to develop a bioresorbable

    scaffold in combination with autologous cells (fibroblasts,

    myocytes, or myofibroblasts) that will be able to contribute

    to tissue regeneration, thereby forming the functional pelvic

    tissue. It is suggested that the successful outcome of vaginal

    reconstructive surgery will depend on utilizing an optimized

    scaffold substrate, such as collagen, to promote the growth and

    distribution of cells with high proliferation potential that would

    exhibit rates of protein synthesis similar to cells from aged-

    matched controls.13 Research is urgently needed to character-

    ize the potential autologous cellular source for use in tissue

    engineering to improve surgical outcome in POP.

    The pelvic floor comprises a highly interconnected system

    of striated muscle, smooth muscle, and connective tissue. The

    connective tissue that supports the pelvic floor is mainly com-

    posed of the fibrous elements of extracellular matrix (ECM),

    collagen and elastin, and cellular components, fibroblasts, and

    smooth muscle cells (SMCs). Collagen is the major structural

    protein in connective tissue. There are 29 members of the col-

    lagen family, including fibril-forming, microfibrillar, fibril-

    associated collagens with interrupted triple helices (FACITs),

    basement membrane, and transmembrane collagens. The bio-

    genesis of structural proteins is controlled by enzymes from the

    LOX family proteins, BMP1/PCP, and ADAMTS2/PNP.14,15

    Connective tissue matrix also contains a variety of multiadhe-

    sive glycoproteins, fibronectin, laminins, vitronectin, throm-

    bospondin, fibrinogen, and others, which allow cells to

    adhere to the ECM. Cells in the tissue bind to the ECM and

    to other cells through transmembrane integrin receptors con-

    sisting of a and b subunits.16 Integrins also transduce biochem-ical signals across the cell membrane, which influence survival,

    metabolism, and cell fate.17 Extracellular matrix proteins exist

    under a constant state of remodeling and turnover. This process

    is mediated by a family of cell-derived proteolytic enzymes

    known as matrix metalloproteinases (MMPs). A group of pro-

    teins named tissue inhibitors of MMPs (TIMPs) strictly control

    these enzymes to maintain tight balance between ECM biogen-

    esis and breakdown.18 Numerous studies using samples from

    different pelvic floor tissues (uterosacral ligaments [USL],

    pubocervical fascia, and vaginal wall) reported differences in

    tissue composition and remodeling proteins between patients

    presenting with severe POP and their asymptomatic counter-

    parts.19-21 This combined with the ease of access makes the

    vagina a good model for use as a source of cells for in vitro

    studies.

    Our aim therefore was to derive primary cells from vaginal

    biopsies of premenopausal Caucasian women with severe POP

    (POP-HVCs) and premenopausal asymptomatic controls (con-

    trol-HVCs) and to examine their biological characteristics. We

    hypothesized that POP-HVCs will display differences in their

    characteristics as compared to control-HVCs due to the intrin-

    sic genetic defects or due to the microenvironment of diseased

    prolapsed tissue. This study examined the ability of primary

    vaginal cells to attach to different protein substrates, to prolif-

    erate, and to produce/modulate the expression of native ECM

    proteins (collagens, laminins, and adhesive glycoproteins),

    their integrin receptors, MMPs/TIMPs/ADAMTSs, and LOX

    family enzymes.

    Materials and methods

    Tissue Collection

    The Research Ethics Board of Mount Sinai Hospital, Uni-

    versity of Toronto, has approved this study. Strict criteria

    were determined to select a homogenous group of preme-

    nopausal Caucasian women with severe POP and asympto-

    matic control group. Eligible patients were invited to

    participate.

    Inclusion criteria. Premenopausal adult women undergoing pel-vic surgery for POP equal or greater than stage 3 by Pelvic

    Organ Prolapse Quantification System (POP-Q) were identi-

    fied as patients. Controls were identified as premenopausal

    adult women with POP-Q at stage 0 undergoing abdominal

    hysterectomy for indications other than prolapse.

    Exclusion criteria. Exclusion criteria were women with a historyof gynecological malignancy, connective tissue disorders,

    emphysema, endometriosis, steroid therapy in the past 6

    months, and prolapse or incontinence surgery. We rationa-

    lized that stage 0 is the ‘‘gold standard’’ for normal pelvic

    support.15,22

    The principal authors (H.K. or M.A.) obtained written

    informed consent from each patient before surgery, performed

    the examination for POP staging, and collected clinical and

    demographic data and tissue samples. Premenopausal status

    was confirmed by endometrial histology report of uterine spe-

    cimens. The tissue biopsy technique was reported in previous

    publication.22 Briefly, after removal of the uterus or pelvic

    floor repair, vaginal tissue specimen (at least 1 cm2) was

    obtained by sharp dissection down to the avascular space of

    loose areolar connective tissue of the vagina. The site of tissue

    collection was standardized at the anterior middle portion of

    the vaginal vault to account for variations in stretch conditions.

    All collected tissue samples were immediately washed in ice-

    cold basic salt solution (Hank balanced salt solution [HBSS]

    without Ca2þ and Mg2þ, HBSS�/�, pH 7.4) and transferredfrom operation room (OR) to the cell culture facility for pri-

    mary cell derivation.

    Derivation of Primary Human Vaginal Cells

    The technique for the derivation of primary HVCs was modi-

    fied from Srikhajon et al.23 Briefly, human vaginal tissues were

    cut into small pieces (1� 1� 1 mm), enzymatically digested at37�C with agitation in buffer supplemented with 2 mg/mLcollagenase type IA (Sigma, Missouri), 0.15 mg/mL DNase I

    932 Reproductive Sciences 23(7)

  • (Roche Applied Science, Québec, Canada), and 1 mg/mL

    bovine serum albumin (BSA; Sigma). After 1.5-hour incuba-

    tion, the digestion mixture was mechanically disrupted by

    pipetting. A suspension of isolated primary cells was centri-

    fuged at 250g for 8 minutes at 4�C, and the cell pellet wasresuspended in phenol red-free cell culture media (Dulbecco

    modified eagle medium [DMEM]; Thermo Fisher Scientific

    Inc, Delaware) supplemented with 20% fetal bovine serum(FBS; Gibco, Burlington, Ontario, Canada), 50 mg/L Normo-

    cin (Invivogen, California), and 25 mmol/L Hepes (Invivo-

    gen, California). These cells were seeded in tissue culture

    dishes and incubated overnight (ON) at 37�C with 5% CO2.Cells were washed with HBSS�/� to remove unattached cellsand cultured in DMEM/20% FBS. Cell culture medium waschanged every 3 days. After 2 to 4 weeks, primary vaginal

    cells (passage 0) reached 90% confluence and were passaged.After 3 passages, HVCs were trypsinized, resuspended in the

    freezing medium (90% FBS; 10% dimethyl sulfoxide[DMSO]), and frozen in liquid nitrogen.

    Cell Morphology and Phenotype Identification

    To verify the cellular origin of primary vaginal cells, we used

    specific biomarkers of fibroblastic versus myogenic origin via

    immunofluorescent staining. Cells derived from the tissue (pas-

    sages 0) and passaged cells (passage 1) were cultured in 8-well

    cell chamber slides until 50% to 75% confluence, fixed with4% paraformaldehyde for 15 minutes at room temperature(RT), permeabilized with 0.1% Triton X-100 (Sigma-Aldrich,Missouri) for 30 seconds, and blocked with serum-free protein

    block (DAKO, Ontario, Canada) for 30 minutes at RT. Human

    vaginal cells were then incubated with a specific antibody for

    fibroblasts biomarker, mouse antihuman vimentin (Sc-7558;

    Santa Cruz, Texas), with antibodies for myogenic biomarker,

    mouse anti-human smooth muscle actin (SMA; M0851,

    DAKO), diluted 1:100 in phosphate-buffered saline (PBS),

    or with endothelial biomarker anti-cytokeratin (M0821, dilu-

    tion 1:200; DAKO), for 18 hours at 4�C. Slides were thenwashed in PBS and incubated with fluorochrome-linked sec-

    ondary antibody goat anti-mouse Alexa Fluor-488 (A21202,

    dilution 1:100; Invitrogen, Burlington, Ontario, Canada) or

    donkey anti-goat Alexa Fluor-594 (A11058, dilution 1:200;

    Invitrogen, Oregon) plus 4,6-diamidino-2-phenylindole dihy-

    drochloride (DAPI) nuclear staining (D-9542, Sigma-Aldrich;

    dilution 1:1000) in the dark for 40 minutes. Slides were

    mounted with media (Thermo Fisher, Ontario, Canada),

    viewed under a fluorescent microscope (Leica Microsystems,

    Richmond Hill, Ontario, Canada), and photographed with

    Sony DXC-970 MD (Sony Ltd, Toronto, Ontario, Canada)

    3CCD color video camera.

    Cell Attachment to Different Extracellular Matrices

    The attachment of HVCs was studied using the Human ECM

    Cell Adhesion Array Kit (colorimetric, ECM540; Millipore,

    Massachusetts) according to the manufacturer’s instruction.

    The kit is comprised of 8 well strips coated with 7 different

    human ECM proteins (collagen [COL] I, COL II, COL IV,

    fibronectin, laminin, tenascin, and vitronectin) and BSA (neg-

    ative control). Briefly, 13 confluent POP-HVCs and control-

    HVCs at passage 2 (7 POP-HVCs and 6 control-HVCs) were

    trypsinized (Gibco), centrifuged, washed 3 times in HBSS�/�

    counted on CASY Cell Counter (Roche Applied Science, Qué-

    bec, Canada), and diluted to a final concentration of 1 � 106cells/ mL. Hundred microliters of each cell suspension

    (100 000 cells) was added in triplicates to 7 ECM-coated wells

    and the control well. The plate was incubated for 1 hour at 37�Cin a CO2 chamber, and then cell stain solution was added to

    each well, followed by extraction buffer, and incubated in the

    dark under gentle rotation at RT for 10 minutes; the absorbance

    was determined at 560 nm on a microplate reader (mQuantBiotek, Vermont, USA).

    Cell Proliferation on Different Extracellular Matrices

    Thiazolyl blue tetrazolium bromide (MTT) assay was used to

    assess the proliferation ability of 10 HVCs lines (5 POP-HVCs

    and 5 control-HVCs) as was described previously.24 All HVC

    lines used for the proliferation study were at passage 2. Briefly,

    24-well black high-throughput (HT) BioFlex culture plates

    coated with COL I (HTPB-3001C; Flexcell Inc, Burlington,

    NC, USA) or proNectin (HTPB-3001P; Flexcell Inc) were

    used. Human vaginal cells were trypsinized, pelleted, and

    washed in HBSS�/� as described previously. The final pelletswere resuspended in DMEM/20% FBS to a final concentrationof 5000 cells/mL. Human vaginal cells (5000 cells/well) were

    plated in quadruplicates on the HT BioFlex culture plate, and

    the control well contained only media. Identical plates for both

    ECM substrates were prepared for each cell line to study the

    proliferation of POP-HVCs and control-HVCs over the span of

    5 days. After 1, 2, 3, 4, or 5 days in culture, 500 mL of MTTsolution (5 mg/mL MTT in DMEM/20% FBS; Sigma-Aldrich)was added to each well including the control (media only)

    wells. The plate was incubated at 37�C for 3.5 hours in aCO2 chamber, then the media was carefully aspirated, and 1

    mL of DMSO/well (Sigma) was added and incubated in the

    dark at RT for 10 minutes. The absorbance was determined at

    590 nm on a microplate reader (mQuant) at the same time every24 hours for the duration of experiment.

    RNA Extraction

    Human vaginal cells were washed twice with ice-cold

    HBSS�/�, scraped, and lysed by TRIzol (Gibco). Total RNAwas extracted according to the manufacturer’s protocol,

    treated with 2.5 mL DNase I (2.73 Kunitz unit/mL; Qiagen,Mississauga, Ontario, Canada) to remove genomic DNA con-

    tamination, and column purified using RNeasy MiniElute

    Cleanup kit (Qiagen). RNA concentration was then measured

    by the NanoDrop 1000 spectrophotometer (Thermo Fisher).

    Stock complementary DNA (cDNA) solutions (50 ng/mL)were generated with iScript Reverse Transcription (RT)

    Kufaishi et al 933

  • Supermix (Bio-Rad Laboratories Inc, California) from 1 mg ofRNA following the manufacturer’s recommended protocol.

    All polymerase chain reactions (PCRs) were carried out on

    the CFX96 or CFX384 Touch Real-Time PCR Detection Sys-

    tems (Bio-Rad).

    Gene Expression Analysis: Quantitative Profiler PCR Array

    We screened for the expression of 84 genes (see Table 1) in

    HVCs using the Human ECM and Adhesion Molecules RT2

    Profiler PCR array (SABiosciences Corp, Frederick, Maryland)

    according to the manufacturer’s instructions. Two pooled sam-

    ples were prepared with equal amounts of RNA from (1) POP-

    HVCs (n ¼ 8) and (2) Control-HVCs (n ¼ 7). ComplementaryDNA was prepared from 1 mg total pooled RNA using iScriptRT Supermix (Bio-Rad). The PCR amplification was con-

    ducted with an initial 10-minute step at 95�C followed by 40cycles of 95�C for 15 seconds and 60�C for 1 minute. Theexperiment was repeated 3 times. Data were imported into the

    integrated Web-based software package (http://pcrdataanaly-

    sis.sabiosciences.com/pcr/arrayanalysis.php; Qiagen GmbH).

    Quantitative analysis was based on the DDCt method, withnormalization of the raw data and the geometric mean of 5

    housekeeping genes (GAPDH, HPRT1, ACTB, B2M, and

    RPLP0). The Web portal automatically performs all DDCt-based calculations from the uploaded raw threshold cycle

    (Ct) data.

    Real-Time RT-PCR

    To confirm the results of the RT2 Profiler PCR array, we

    conducted quantitative reverse transcription-polymerase chain

    reaction (qRT-PCR) on individual cDNA samples produced

    from 8 individual cell lines derived from patients with POP

    and 7 individual cell lines derived from asymptomatic controls

    using CFX384 Touch Real-Time PCR Detection Systems (Bio-

    Rad). Specific sequences of oligonucleotide primers for indi-

    vidual genes were obtained from Gene Bank Database of the

    National Centre for Biotechnology Information (NCBI) of the

    National Institutes of Health using Primer-BLAST (Supple-

    mental Table 1). Gene expression levels in all studies were

    normalized against the geometric mean of the following 3

    housekeeping genes: YWHAZ, TBP, and ACTB. Reverse

    transcription-PCR reactions were carried out in triplicates,

    where each reaction contained 10 ng of cDNA, LuminoCt

    SYBR Green qPCR ReadyMix (Sigma), and forward and

    reverse primers at a final concentration of 300 nmol/L in a total

    of 10 ml per well reaction mix. The cycling protocol startedwith an initial denaturation 95�C for 30 seconds, then 40 cyclesof denaturation 95�C for 5 seconds, and annealing/extension60�C for 20 seconds. Each qRT-PCR run was followed by amelting curve analysis to confirm the specificity of the primers

    used. Gene expression values were analyzed using the DDCqmode on the CFX Manager software 2.0 (Bio-Rad). Sample

    replicates with a Cq > 35 were excluded from data analysis,

    and the cutoff quantification cycle standard deviation (DCq)

    was set at �0.2. A no-template control of each primer master-mix was also loaded on every plate to ensure the absence of

    contaminations.

    Statistical Analysis

    Patient’s demographics were subjected to Fisher exact test for

    categorical data and Student t test for continuous data. To

    determine the differences in the attachment to different ECM

    substrates between POP-HVCs and control-HVCs, 2-way

    analysis of variance (ANOVA) was used. Results of prolifera-

    tion assay were subjected to 2-way ANOVA to determine

    levels of significant difference. Real-time PCR results were

    subjected to Student t test. Experimental error was reported as

    standard error of the mean (SEM). The statistical program

    Prism (version 4.0; Graph Pad Software Inc, San Diego, Cali-

    fornia) was used, with the level of significance for comparison

    set at P < .05.

    Results

    Patients’ Demographics

    Vaginal vault biopsy samples were obtained from 15 Cau-

    casian premenopausal women (8 in the ‘‘patient’’ group 1

    and 7 in the ‘‘control’’ group 2) matched for age and body

    mass index. The majority of patients with POP (88%) andcontrols (57%) reported symptoms of SUI, and the differ-ence between the 2 groups was not significant. There was

    however a statistical difference between patients with POP

    and asymptomatic controls with respect to the family history

    of POP (P ¼ .019). The mean parity and the number ofvaginal deliveries were significantly higher in patients with

    POP (P < .05; Table 2).

    Isolation and Characterization of Primary Vaginal Cells

    We derived cells from 8 patients with POP and 7 women with

    clinically normal pelvic floor support using collagen digestion

    and then used specific biomarkers of fibroblastic versus myo-

    genic origin via immunoflourescent staining to identify the

    phenotype of these cells. Our results show that immediately

    after derivation (passage 0-2), the majority of DAPI-stained

    cells were positive for vimentin (fibroblastic marker) and myo-

    genic marker a-SMA in both POP and control groups (Figure1A). To verify these results, in passage 2, we used antibodies

    against desmin, a specific marker of differentiated SMCs, and

    found only 5% to 6% desmin-positive cells in both POP andcontrol groups (Figure 1B). Furthermore, we noted no cells

    stained for the antiepithelial cell marker, cytokeratin, in pas-

    sages 0 to 2. We concluded that the majority of primary vaginal

    cells in culture were myofibroblasts, with a minor presence of

    myocytes and no epithelial contamination.

    To determine whether POP-HVCs and control-HVCs pre-

    ferentially attach to different ECM proteins, we used the com-

    mercially available Human Cell Adhesion Array Kit. Our

    results indicate no differential attachment of HVCs on COL

    934 Reproductive Sciences 23(7)

    http://pcrdataanalysis.sabiosciences.com/pcr/arrayanalysis.phphttp://pcrdataanalysis.sabiosciences.com/pcr/arrayanalysis.php

  • Table 1. List of 84 ECM and Cell Adhesion Genes Per Functional Group.a

    Reference Sequences Symbol Description

    CollagensNM_000088 COL1A1 Collagen, type I, a1NM_001846 COL4A2 Collagen, type IV, a2NM_000093 COL5A1 Collagen, type V, a1NM_001848 COL6A1 Collagen, type VI, a1NM_001849 COL6A2 Collagen, type VI, a2NM_000094 COL7A1 Collagen, type VII, a1NM_001850 COL8A1 Collagen, type VIII, a1NM_080629 COL11A1 Collagen, type XI, a1NM_004370 COL12A1 Collagen, type XII, a1NM_021110 COL14A1 Collagen, type XIV, a1NM_001855 COL15A1 Collagen, type XV, a1NM_001856 COL16A1 Collagen, type XVI, a1

    ECM proteases and protease inhibitorsNM_002421 MMP1 Matrix metallopeptidase 1 (interstitial collagenase)NM_004530 MMP2 Matrix metallopeptidase 2 (gelatinase A, 72 kDa gelatinase, 72 kDa type IV collagenase)NM_002422 MMP3 Matrix metallopeptidase 3 (stromelysin 1, progelatinase)NM_002423 MMP7 Matrix metallopeptidase 7 (matrilysin, uterine)NM_002424 MMP8 Matrix metallopeptidase 8 (neutrophil collagenase)NM_004994 MMP9 Matrix metallopeptidase 9 (gelatinase B, 92 kDa gelatinase, 92 kDa type IV collagenase)NM_002425 MMP10 Matrix metallopeptidase 10 (stromelysin 2)NM_005940 MMP11 Matrix metallopeptidase 11 (stromelysin 3)NM_002426 MMP12 Matrix metallopeptidase 12 (macrophage elastase)NM_002427 MMP13 Matrix metallopeptidase 13 (collagenase 3)NM_004995 MMP14 Matrix metallopeptidase 14 (membrane-inserted)NM_002428 MMP15 Matrix metallopeptidase 15 (membrane inserted)NM_005941 MMP16 Matrix metallopeptidase 16 (membrane inserted)NM_006988 ADAMTS1 ADAM metallopeptidase with thrombospondin type 1 motif, 1NM_139025 ADAMTS13 ADAM metallopeptidase with thrombospondin type 1 motif, 13NM_007037 ADAMTS8 ADAM metallopeptidase with thrombospondin type 1 motif, 8NM_003254 TIMP1 TIMP metallopeptidase inhibitor 1NM_003255 TIMP2 TIMP metallopeptidase inhibitor 2NM_000362 TIMP3 TIMP metallopeptidase inhibitor 3

    Transmembrane and cell–matrix adhesion moleculesNM_001843 CNTN1 Contactin 1NM_001903 CTNNA1 Catenin (cadherin-associated protein), a1, 102 kDaNM_001904 CTNNB1 Catenin (cadherin-associated protein), b1, 88 kDaNM_001331 CTNND1 Catenin (cadherin-associated protein), d1NM_001332 CTNND2 Catenin (cadherin-associated protein), d2 (neural plakophilin-related arm-repeat protein)NM_181501 ITGA1 Integrin, a1NM_002203 ITGA2 Integrin, a2 (CD49B, alpha 2 subunit of VLA-2 receptor)NM_002204 ITGA3 Integrin, a3 (antigen CD49C, alpha 3 subunit of VLA-3 receptor)NM_000885 ITGA4 Integrin, a4 (antigen CD49D, alpha 4 subunit of VLA-4 receptor)NM_002205 ITGA5 Integrin, a5 (fibronectin receptor, alpha polypeptide)NM_000210 ITGA6 Integrin, a6NM_002206 ITGA7 Integrin, a7NM_003638 ITGA8 Integrin, a8NM_002209 ITGAL Integrin, a L (antigen CD11A [p180], lymphocyte function-associated antigen 1; a polypeptide)NM_000632 ITGAM Integrin, a M (complement component 3 receptor 3 subunit)NM_002210 ITGAV Integrin, a V (vitronectin receptor, a polypeptide, antigen CD51)NM_002211 ITGB1 Integrin, b 1 (fibronectin receptor, b polypeptide, antigen CD29 includes MDF2, MSK12)NM_000211 ITGB2 Integrin, b 2 (complement component 3 receptor 3 and 4 subunit)NM_000212 ITGB3 Integrin, b 3 (platelet glycoprotein IIIa, antigen CD61)NM_000213 ITGB4 Integrin, b 4NM_002213 ITGB5 Integrin, b 5NM_000201 ICAM1 Intercellular adhesion molecule 1NM_000615 NCAM1 Neural cell adhesion molecule 1NM_000442 PECAM1 Platelet/endothelial cell adhesion moleculeNM_001078 VCAM1 Vascular cell adhesion molecule 1

    (continued)

    Kufaishi et al 935

  • I, COL II, fibronectin, laminin, tenascin, or vitronectin pro-

    teins. A significant decrease in attachment ability of POP-

    HVCs compared to control-HVCs was observed on basement

    membrane protein COL IV (Figure 2A, P < .05). Since we did

    not detect differential attachment on COL I, the most abundant

    fibrillar collagen in the ECM, or on the major linker protein

    fibronectin, we selected these proteins as substrates for the

    proliferation study. The ability of primary HVCs derived from

    asymptomatic controls and patients with POP to proliferate was

    assessed using COL I-coated or proNectin-coated plates over a

    period of 5 days. The MTT assay was carried out to assess a

    daily proliferation rate of 5 cell lines derived from prolapsed

    tissue and 5 cell lines derived from normal vaginal biopsies.

    Our results indicate no significant difference in the prolifera-

    tion rate of POP-HVCs plated on COL I or proNectin (a syn-

    thetic analogue of fibronectin) in comparison to the control-

    HVCs (Figure 2B).

    Extracellular Matrix and Adhesion MoleculeQuantitative PCR Arrays

    This PCR array detects the expression of 84 genes involved

    in cell adhesion and communication, ECM remodeling, ECM

    ligands, and their transmembrane receptors (see Table 1). In

    particular, we compared the gene expression of multiple col-

    lagen molecules (Figure 3A), ECM proteases and protease

    inhibitors (Figure 3B), transmembrane integrin receptors and

    cell–matrix adhesion molecules (Figure 3C), cell–cell adhe-

    sion molecules, and basement membrane constituents (Figure

    3D) in resting primary HVCs derived from patients with POP

    and controls. The summary of the PCR results presented in

    the Supplemental Table 2 indicate that 36 genes were

    Table 1. (continued)

    Reference Sequences Symbol Description

    Cell–cell adhesion molecules and basement membrane constituentsNM_004425 ECM1 Extracellular matrix protein 1NM_002026 FN1 Fibronectin 1NM_001523 HAS1 Hyaluronan synthase 1NM_000216 KAL1 Kallmann syndrome 1 sequenceNM_005559 LAMA1 Laminin, a 1NM_000426 LAMA2 Laminin, a 2NM_000227 LAMA3 Laminin, a 3NM_002291 LAMB1 Laminin, b 1NM_000228 LAMB3 Laminin, b 3NM_002293 LAMC1 Laminin, g 1 (formerly LAMB2)NM_001901 CTGF Connective tissue growth factorNM_003919 SGCE Sarcoglycan, epsilonNM_000582 SPP1 Secreted phosphoprotein 1NM_003118 SPARC Secreted protein, acidic, cysteine-rich (osteonectin)NM_000450 SELE Selectin ENM_000655 SELL Selectin LNM_003005 SELP Selectin P (granule membrane protein 140 kDa, antigen CD62)NM_003119 SPG7 Spastic paraplegia 7 (pure and complicated autosomal recessive)NM_002160 TNC Tenascin CNM_003246 THBS1 Thrombospondin 1NM_003247 THBS2 Thrombospondin 2NM_007112 THBS3 Thrombospondin 3NM_000358 TGFBI Transforming growth factor, beta-induced, 68kDaNM_004385 VCAN VersicanNM_000638 VTN VitronectinNM_003278 CLEC3B C-type lectin domain family 3, member BNM_004360 CDH1 Cadherin 1, type 1, E-cadherin (epithelial)NM_000610 CD44 CD44 molecule (Indian blood group)

    aAdapted from http://www.sabiosciences.com/rt_pcr_product/HTML/PAHS-013Z.html.

    Table 2. Summary of Patient Demographics.a

    Study GroupsGroup 1 (Patient

    With POP)Group 2

    (Non-POP Control)

    n 8 7Ageb 43.5 (37-50) 45.0 (42-49)BMIb 27.8 (20.6-41.6) 27.5 (21.6-32.8)Parityb 3 (1-6)c 1 (0-3)Vaginal deliveriesb 3 (1-3)c 1 (0-1)SUI 88% 57%Family history of POP (%) 63%c 0%Stage III-IV of POP (n) 8 0No POP (n) 0 7

    Abbreviations: BMI, body mass index; POP, pelvic organ prolapse; SUI, stressurinary incontinence.aFisher exact test; level of significance: P < .05.bData are presented as median (range).cStatistical difference between groups 1 and 2.

    936 Reproductive Sciences 23(7)

    http://www.sabiosciences.com/rt_pcr_product/HTML/PAHS-013Z.html

  • differentially expressed between the 2 groups (marked red,

    P < .05). These results demonstrate that under resting culture

    conditions and in comparison to control-HVCs, (1) POP-

    HVCs expressed significantly higher levels of COL7A1,

    numerous ECM integrin receptors (ITGA1, ITGA4, ITGA6,

    ITGA8, ITGAAV, ITGB1, ITGB2, and ITGB3), catenins

    (CTNNA1, CTNNB1, and CTNND1), cadherin 1 (CDH1),

    contactin 1 (CNTN1), hyaluronic acid receptor (CD44), cell

    adhesion molecules (NCAM1), thrombospondin-2 (THBS2)

    and laminins (LAMB1 and LAMB13), and ECM remodeling

    genes (MMP3, MMP7, MMP10, MMP12, MMP13, MMP14,

    and TIMP2; P < .05), (2) whereas messenger RNA (mRNA)

    0 1 2 (passage)

    VIM VIM VIM

    ASMA ASMA DESMIN

    NEG CONTROL NEG CONTROL

    CK CK CK

    SM Ac�n

    0

    20

    40

    60

    80

    100

    Perc

    enta

    ge o

    f (+)

    cel

    ls

    Desmin

    CONTROL

    Vimen�n

    POP

    B

    A

    Figure 1. Characterization of primary human cells derived from vaginal biopsies. A, Representative immunofluorescence images of primary cellsderived from premenopausal woman at passage 0, 1, and 2. Cells were fixed in paraformaldehyde (PFA) and incubated with antibodies againstvimentin (red), a-smooth muscle actin (a-SMA), desmin (only passage 2), and cytokeratin (CK; all green). Nuclei were stained with4,6-diamidino-2-phenylindole dihydrochloride (DAPI; blue). Negative controls show staining with secondary antibodies only. Original magnifica-tions: �100. B, Quantitative analysis of human vaginal cells (HVCs) from passage 1 derived from control (black bars) and patients with pelvicorgan prolapse (POP; white bars), which expressed vimentin, desmin, and a-SMA, respectively (percentage of positive cells versus all cells). (Thecolor version of this figure is available in the online version at http://rs.sagepub.com/.)

    Kufaishi et al 937

    http://rs.sagepub.com/

  • levels of COL15A1, TIMP3, VCAM1, and ICAM1 were sig-

    nificantly (P < .05) downregulated (Supplemental Table 2;

    Figure 4).

    The results of the PCR array for selected ECM proteins, their

    integrin receptors, proteases, and protease inhibitors that showed

    significant difference were chosen for verification by quantita-

    tive RT-PCR using individual RNA samples isolated from POP-

    HVCs (n ¼ 8) and control-HVCs (n ¼ 7). Overall, the RT-PCRanalysis confirmed the PCR array results, in particular, the basal

    expression of multiple integrins (ITGA1, ITGA4, ITGA8, and

    ITGB1), COL7A1, and MMP3 was significantly higher,

    whereas COL15A1, MMP2, TIMP2, and TIMP3 genes were

    significantly lower in cells isolated from patients with POP than

    in control vaginal cells (Figure 5A and B, P < .05).

    Our previous in vivo study found differential mRNA expres-

    sion of proteins involved in the processing and maturation of

    collagen and elastin polymers, namely LOX, LOXL1 to

    LOXL4, BMP1/PCP, and ADAMTS2/PNP, in vaginal biopsy

    samples from premenopausal patients with POP and asympto-

    matic controls.14,15,22 Unfortunately, these genes were not

    included in the PCR array. Hence, to understand whether these

    in vivo changes occur due to the differences in the cellular

    expression, we examined mRNA levels of those genes by

    qRT-PCR in the unstimulated (resting) HVC cultures derived

    from patients with POP and asymptomatic controls. Similar to

    the earlier in vivo data, a significant decrease in LOX, LOXL1

    to LOXL3, ADAMTS2, and BMP1 mRNA expression was

    noted in vitro in cultured POP-HVCs compared to resting

    control-HVCs (Figure 5C)

    Discussion

    In an attempt to improve the outcome of POP surgery, urogy-

    necologists have extensively used surgical implants to rein-

    force the weakened pelvic floor tissue. The efficacy of this

    approach is questionable, as many women report serious com-

    plications, such as erosion, pain, vaginal stenosis, and infec-

    tion.11,25 Simultaneously, the need for improved long-lasting

    repair therapies for POP is growing due to the increased life

    expectancy and activity levels of modern women. Because of

    their accessibility, muscle-derived stem cells were the first

    explored in cell-based therapy for the treatment of POP in the

    animal model.26,27 The use of human fibroblasts derived from

    the endometrium, vagina, foreskin, and buccal membranes in

    conjunction with synthetic scaffolds has also been suggested as

    a treatment for POP.12,13,28,29 To understand the reason for

    unsuccessful outcomes after POP repair surgery and to deter-

    mine whether the cause of failure might be a defective pro-

    lapsed tissue, we designed this study in which we compare

    characteristics of vaginal cells originated from POP and non-

    POP tissue.

    Our current data indicate that human vaginal cells derived

    from patients with POP and asymptomatic controls were

    mostly myofibroblasts with a small presence of SMCs. Cells

    from diseased and healthy tissue showed a similarity in the

    attachment to different ECM components, except for COL

    IV, the protein that present ubiquitously in basement mem-

    branes. COL IV provides a scaffold for cellular assembly and

    mechanical stability and plays an important role in cell adhe-

    sion, growth, migration, proliferation, and differentiation. It is

    the main ECM protein excreted from fibroblasts following

    trauma and is induced during scar formation.30 Attachment of

    cells to COL IV is mediated via multiple binding sites and

    involves several integrins and nonintegrin receptors.31 There-

    fore, we speculate that decreased attachment of POP-HVCs to

    COL IV may contribute to the weakening of the pelvic floor in

    patients with POP. On the other hand, cellular proliferation of

    HVCs derived from patients with POP in comparison to control

    HVCs was similar on major fibrillar COL I and proNectin

    (which contains the Arg-Gly-Asp attachment domain of

    fibronectin).

    0

    0.05

    0.1

    0.15

    0.2

    0 1 2 3 4 5

    OD

    Day

    Control Collagen 1

    Control proNec�nPOP Collagen 1

    0

    0.1

    0.2

    0.3

    0.4

    0.5

    Col I Col II Col IV FN LN TN VN

    OD

    ECM Substrate

    ControlA

    B

    POP

    POP proNec�n

    Figure 2. Cellular characteristics of primary human vaginal cells(HVCs) derived from premenopausal women. A, Attachment of HVCsfrom asymptomatic controls (n ¼ 6, black bars) and patients withsevere pelvic organ prolapse (POP; n ¼ 7, white bars) on 7 differenthuman extracellular matrix (ECM) proteins: collagen (Col) I, Col II,Col IV, fibronectin (FN), laminin (LN), tenascin (TN), and vitronectin(VN). The results shown are the mean + standard error of the mean(SEM) of absorbance relative to the control protein, bovine serumalbumin (BSA). A significant difference is indicated by *P < .05. B,Proliferation of HVCs derived from premenopausal asymptomaticcontrols (solid line) and patients with severe POP (dashed line) oncollagen I (black) and proNectin (gray)-coated plates, respectively(n ¼ 5/group). The results shown are the optical density per well,mean + SEM for each patient group. No significant difference wasdetected by analysis of variance (ANOVA).

    938 Reproductive Sciences 23(7)

  • Our previous in vivo studies demonstrated impairment in the

    expression and activity of ECM homeostasis-related proteins

    (enzymes regulating ECM assembly, synthesis, and degrada-

    tion) in vaginal tissue biopsies derived from patients with POP

    in comparison to asymptomatic controls.14,15,32 Other research

    groups assessed collagen content in pelvic floor tissue of

    women with and without POP and concluded that the overall

    collagen content is decreased in the pelvic floor tissue of

    women with POP in comparison to asymptomatic controls.33,34

    However, there are limited data on the metabolism of collagen

    and elastin and the lack of detailed analysis of different col-

    lagen subtypes in HVCs. Makinen et al found that the rate of

    collagen synthesis and procollagen mRNA levels in fibroblasts

    derived from patients with POP was similar or slightly higher

    than those from age-matched asymptomatic controls.35

    Recently, Kerkhof et al compared tissue sample from non-

    POP sites and POP sites in the same patient and found that

    there was a tendency toward an increase in the COL III and

    elastin content and a significantly increased number of collagen

    cross-links in the POP site.36 We report here key differences in

    mRNA expression levels of multiple collagens and integrins

    between POP-HVCs and control-HVCs. In particular, under

    resting culture conditions, POP-HVCs in comparison with

    control-HVCs expressed lower mRNA levels of FACIT COL

    XV that codistributes with COL I. Interestingly, COL XV has

    the strongest expression in basement membrane zones and

    functions to adhere basement membranes to underlying con-

    nective tissue stroma. It was reported in mouse that COL XV

    deficiency is associated with muscle and microvessel deteriora-

    tion.37 In contrast, COL VII, which functions to anchor fibril

    between the external stratified epithelia and the underlying

    stroma, was upregulated by POP-HVF. The expression of COL

    VII was shown to be restricted to the basement zone beneath

    stratified squamous epithelia, and its mutations were associated

    Figure 3. Gene expression heat map from polymerase chain reaction (PCR) array analysis. In the resting human vaginal cells (HVCs) derivedfrom asymptomatic controls (CONTROL) and patients with severe pelvic organ prolapse (POP), gene expression levels of (A) collagens andextracellular matrix (ECM) structural constituents; (B) ECM proteases and protease inhibitors; (C) cell–cell adhesion molecules and basementmembrane constituents; and (D) transmembrane and cell–matrix adhesion molecules were different between groups. Pooled RNA samples ofCONTROL (n ¼ 7) and patients with POP (n ¼ 8) were used. Shown are results of 3 separate PCR arrays per group. A significant difference isindicated in red (see Supplemental Table 2, P < .05).

    Kufaishi et al 939

  • with chronic subepidermal blistering disease.38 We also noted

    differential expression of basement membrane proteins

    (LAMB1 and LAMB3) between POP-HVCs and control-

    HVCs. Basement membranes separate cell from the underlying

    connective tissue, stabilize and maintain the tissue architecture.

    Laminins are very large cross-shaped molecules constituted by

    the association of a, b, and g chains. They have the ability toself-associate into polymers forming a high-equimolecular

    complex, which in combination with COL IV creates a stable

    scaffold anchoring cells and other components of the basement

    membrane (ie, fibronectin). Changes in the expression of ECM

    constituents (in particular, basement membranes) show a gen-

    eralized dysregulation in the prolapsed vaginal tissue also man-

    ifested by the decreased attachment capability of POP-HVCs

    probably indicating misbalance of pelvic floor tissue morphol-

    ogy on a micromolecular level.

    Specific interactions between cells and the ECM are

    mediated by integrin molecules. Our results also demonstrate

    that POP-HVCs expressed higher mRNA levels of numerous

    integrins (a1, a4, a6, a8, av, b1, b2, and b3) and cell–celladhesion molecules (contactin 1 and catenins a, b, d). Integrinsare transmembrane a/b noncovalently-associated heterodimersignal transduction molecules, sensing physical forces and

    transducing mechanical signals (stretch) into a biochemical

    response. It is possible that the expression of integrins is upre-

    gulated in POP-HVCs due to an increased mechanical stimula-

    tion of the prolapsed tissue and may reflect the secondary

    effects of POP. Signaling through integrins involves the assem-

    bly of a complex of multiple proteins inside the cell, which may

    implicate the altered cell–matrix interaction and potential dif-

    ference in mechanotransduction mechanism, especially when

    external stimuli are applied. For instance, integrin duplex a1b1is a cell surface receptor for collagen and laminin, and avb3 is acell surface receptor for vitronectin. Integrin b1 in combinationwith integrin a4 forms integrin VLA-4 that act as a receptor forfibronectin,39 with integrin a6–VLA-6 and integrin a8 receptorfor tenascin, fibronectin, and vitronectin. These heterodimeric

    receptors are involved in cell–cell adhesion and may play a role

    in inflammation and fibrosis. It was reported that the integrin

    a8 gene may be a major effector of HOXA11,40 a homeoboxtranscription factor that orchestrates embryonic development

    C

    A

    0

    0.5

    1

    1.5

    LOX LOXL1 LOXL2 LOXL3 LOXL4 BMP1 ADAMTS2

    * * * *** **

    Gen

    e Ex

    pres

    sion

    (Rel

    a�ve

    Fol

    d Ch

    ange

    )

    0

    0.5

    1

    1.5

    2

    2.5

    3

    3.5

    COL7A1 COL15A1 MMP2 MMP3 TIMP 2 TIMP 3

    CONTROLPOP

    ** ** **

    *

    *

    B

    0

    2

    4

    6

    8

    CNTN1 ITGA1 ITGA4 ITGA8 ITGB1 ITGB2

    ***

    **

    **

    Figure 5. Expression of selected genes by human vaginal cells (HVCs)from asymptomatic controls (n ¼ 7, black bars) and patients withpelvic organ prolapse (POP; n ¼ 8, white bars) validated by quantita-tive reverse transcription-polymerase chain reaction (qRT-PCR) andnormalized to 3 housekeeping genes YWHAZ, TBP and ACTB: (A)extracellular matrix (ECM) and ECM-degrading genes; (B) ECM integ-rin receptors and cell adhesion genes; and (C) ECM maturation pro-teins. The results shown are fold change relative to asymptomaticCONTROL (mean + standard error of the mean [SEM]). A sig-nificant difference is indicated by *P < .05 and **P < .01.

    Gen

    e Ex

    pres

    sion

    (Rel

    a�ve

    Fol

    d Ch

    ange

    )

    0

    0.5

    1

    1.5

    2

    2.5

    3

    3.5

    CNTN1 ITGA1 ITGA4 ITGA8 ITGB1 ITGB2

    A

    B

    0

    0.5

    1

    1.5

    2

    2.5

    COL7A1 COL15A1 MMP2 MMP3 TIMP2 TIMP3

    CONTROL

    POP

    Figure 4. Relative expression of extracellular matrix (ECM) genes byresting human vaginal cells (HVCs) derived from asymptomatic con-trols (CONTROL) and patients with severe pelvic organ prolapse(POP) determined using a reverse transcription-polymerase chainreaction (RT-PCR)-based mRNA expression array. The resultsshown are fold change relative to asymptomatic controls (mean +standard error of the mean [SEM]). A significant difference is indicatedby *P < .05, **P < .01 and ***P < 0.001.

    940 Reproductive Sciences 23(7)

  • of the urogenital tract, in particular uterosacral ligaments

    (USL).41 It has been suggested that this signaling pathway

    involving HOXA11, COL III, and MMP2 contributes to the

    weakened connective tissue of women with POP.41 Altogether,

    these results confirm that fibroblasts derived from patients with

    POP show cellular characteristics that are already modified by

    their environment as compared to the fibroblasts derived from

    non-POP asymptomatic controls. We suggest that when

    exposed to pathologic external stimuli42,43 or surgical

    trauma,44 POP-HVCs may not be able to properly adhere to

    ECM, resulting in loss of tissue strength and subsequent devel-

    opment or reoccurrence of prolapse.

    Little is known about the active interactions between the cell

    and the ECM and how this affects the pathophysiology of POP.

    The understanding of the cell–matrix interaction in tissue of

    women with POP is necessary to cultivate new therapeutic

    approaches, especially in the development of new bioscaffolds

    for prolapse repair. There is an evidence that the vaginal con-

    nective tissue from patients with POP is phenotypically differ-

    ent than the connective tissue derived from asymptomatic

    controls, showing diminished COL I and COL III levels,45 and

    ECM that is disordered, less dense, and loosely arranged.46

    Unfortunately, we cannot comment on the expression levels

    of COL III in the HVCs-derived POP and control tissues as

    this gene was not included in the analysis. Importantly, here we

    demonstrated that under static conditions, cultured HVCs from

    patients with POP expressed lower transcript levels of the LOX

    family proteins, which parallels with our earlier in vivo

    results.14 The key known function of LOX enzymes is to con-

    tribute to collagen and elastin fiber cross-linking, however,

    they also play a role in fibrotic disorders.47 In contrast to our

    in vivo results, we detected a decrease in the expression of PCP

    and PNP genes in POP-HVCs in vitro in comparison to asymp-

    tomatic controls.15 It has been shown before that mechanical

    tension can decrease the transcript levels of LOX, pro-COL I

    and III, PNP, and PCP.48 Thus, it is possible that HVCs derived

    from severely prolapsed (and therefore stretched) tissues would

    express lower levels of LOX, PNP, and PCP. During wound

    healing, resting fibroblasts differentiate to a migratory myofi-

    broblasts characterized by suppressed expression of MMP2 and

    increased expression of the contractile protein, a-SMA.49

    Importantly, our RT-PCR results indicate that MMP2 transcript

    levels were downregulated in the POP-HVCs. It is in a good

    agreement with Ruiz-Zapata and colleagues who reported that

    vaginal fibroblasts derived from women with mild POP showed

    lower gene expression levels of MMP2 mRNA50 and secreted

    less MMP2 protein51 when compared with healthy controls. It

    is known that factors that increase myofibroblast contractility,

    such as transforming growth factor b and serum, decreaseMMP2 expression. Since in pathological wound healing envir-

    onments, such as that of the pelvic floor of patients with POP,

    the myofibroblast phenotype persists, this could explain why

    MMP2 expression was lower in POP-HVCs in comparison to

    control-HVCs. In addition, we detected an increased expres-

    sion of MMP3, an enzyme involved in tissue repair and crucial

    in connective tissue remodeling, as well as MMP13

    (collagenase 3) and membrane-type MMP14 in POP-HVCs;

    however, there was no change in the expression of collagenases

    MMP1 and MMP8, which was in accordance with our recently

    published in vivo data.32 Matrix metalloproteinase 14 is able to

    cleave pro-MMP2 to active MMP2 in a trimolecular complex

    involving TIMP2.52 The MMP14–mediated MMP2 activation

    is upregulated by certain ECM molecules in order to regulate

    COL I synthesis and prevent fibrosis53; however, it could be

    selectively suppressed by ITGB3,54 which colocalizes with

    MMP2 on the surface of different cells.55 Importantly, ITGB3

    transcript was significantly upregulated in the POP-HVCs (see

    supplemental Table 2). We speculate that overexpression of

    ITGB3 on the surface of HVCs may serve as a protective

    mechanism to decrease the activity of MMP2 in patients with

    POP. We previously reported a reduction in TIMP2 and TIMP3

    mRNA in POP vaginal biopsies in comparison to asympto-

    matic controls.32 In accordance, current in vitro results showed

    that cultured HVCs derived from patients with POP expressed

    lower levels of TIMP2 and TIMP3 mRNA, in comparison to

    HVCs derived from asymptomatic controls. These studies con-

    firm the differential expression of the ECM-degrading proteins

    and their inhibitors in POP-HVCs versus non-POP-HVCs,

    showing genotypic and phenotypic differences in these cells,

    which potentially may predispose them to different modes of

    reaction to remodeling of the ECM. Altogether, our results

    indicate that phenotypic changes that we and others detected

    in vivo on the macro level in the prolapsed tissue are actually a

    reflection of changes in a cellular level possibly due to

    increased mechanical (stretch) or decreased hormonal (meno-

    pause) stimulation.

    Our study has limitations. It would be useful to compare our

    in vitro results with in vivo studies; however, the size of the

    tissue specimen limited us to use it only for primary cell deri-

    vation. We also acknowledge that the premenopausal study

    groups were not matched for parity and vaginal delivery, a

    known factor in POP development. We speculate now that

    repetitive trauma from delivery may affect the basement mem-

    brane assembly/formation influencing their scaffolding charac-

    teristics, which appears critical for the pelvic tissue strength. In

    conclusion, we were able to derive a homogenous human vagi-

    nal cell population, with similar attachment and proliferative

    abilities on COL I, from patients with POP and from asympto-

    matic control women. However, we detected key differences in

    the biological characteristics of these cells, particularly in their

    ability to produce ECM. Thus, we suggest that utilization of

    cells from an anatomical site not related to pelvic tissue and not

    affected by the prolapse may be a better alternative to mesh-

    augmented reconstructive surgery of the pelvic floor in order to

    generate sufficiently strong new tissue by replacing defective

    cells with healthy autologous cells. Further research is neces-

    sary to evaluate the effect of external stimuli mimicking risk

    factors for POP on the cellular responses of HVCs.

    Acknowledgements

    The authors gratefully thank Ms Yaryna Rybak for her assistance in

    the RT-PCR validation analysis. The authors would like to thank

    Kufaishi et al 941

  • patients of the Urogynecology Division, Mount Sinai Hospital,

    Toronto, for helping in tissues collection.

    Declaration of Conflicting Interests

    The author(s) declared no potential conflicts of interest with respect to

    the research, authorship, and/or publication of this article.

    Funding

    The author(s) disclosed receipt of the following financial support for

    the research, authorship, and/or publication of this article: This study

    was supported by a grant from the Department of Obstetrics and

    Gynecology, Mount Sinai Hospital, Toronto, and Dean’s fund, Faculty

    of Medicine, University of Toronto, Canada.

    Supplemental Material

    The online supplemental tables are available at http://rs.sagepub.com/

    supplemental.

    References

    1. Barber MD, Visco AG, Wyman JF, Fantl JA, Bump RC. Sexual

    function in women with urinary incontinence and pelvic organ

    prolapse. Obstet Gynecol. 2002;99(2):281-289.

    2. Heit M, Rosenquist C, Culligan P, Graham C, Murphy M, Shott S.

    Predicting treatment choice for patients with pelvic organ pro-

    lapse. Obstet Gynecol. 2003;101(6):1279-1284.

    3. Nygaard I, Barber MD, Burgio KL, et al. Prevalence of sympto-

    matic pelvic floor disorders in US women. JAMA. 2008;300(11):

    1311-1316.

    4. Olsen AL, Smith VJ, Bergstrom JO, Colling JC, Clark AL. Epi-

    demiology of surgically managed pelvic organ prolapse and urin-

    ary incontinence. Obstet Gynecol. 1997;89(4):501-506.

    5. Lukacz ES, Lawrence JM, Contreras R, Nager CW, Luber KM.

    Parity, mode of delivery, and pelvic floor disorders. Obstet Gyne-

    col. 2006;107(6):1253-1260.

    6. Moalli PA, Jones IS, Meyn LA, Zyczynski HM. Risk factors

    associated with pelvic floor disorders in women undergoing sur-

    gical repair. Obstet Gynecol. 2003;101(5 pt 1):869-874.

    7. Buchsbaum GM, Duecy EE, Kerr LA, Huang LS, Perevich M,

    Guzick DS. Pelvic organ prolapse in nulliparous women and their

    parous sisters. Obstet Gynecol. 2006;108(6):1388-1393.

    8. Whitcomb EL, Rortveit G, Brown JS, et al. Racial differences in

    pelvic organ prolapse. Obstet Gynecol. 2009;114(6):1271-1277.

    9. Norton PA, Allen-Brady K, Wu J, Egger M, Cannon-Albright L.

    Clinical characteristics of women with familial pelvic floor dis-

    orders. Int Urogynecol J. 2015;26(3):401-406.

    10. Carey M, Slack M, Higgs P, Wynn-Williams M, Cornish A.

    Vaginal surgery for pelvic organ prolapse using mesh and a vagi-

    nal support device. BJOG. 2008;115(3):391-397.

    11. Abed H, Rahn DD, Lowenstein L, Balk EM, Clemons JL, Rogers

    RG. Incidence and management of graft erosion, wound granula-

    tion, and dyspareunia following vaginal prolapse repair with graft

    materials: a systematic review. Int Urogynecol J. 2011;22(7):

    789-798.

    12. Skala CE, Petry IB, Gebhard S, et al. Isolation of fibroblasts for

    coating of meshes for reconstructive surgery: differences between

    mesh types. Regen Med. 2009;4(2):197-204.

    13. Chen G, Sato T, Ohgushi H, Ushida T, Tateishi T, Tanaka J.

    Culturing of skin fibroblasts in a thin PLGA-collagen hybrid

    mesh. Biomaterials. 2005;26(15):2559-2566.

    14. Alarab M, Bortolini MA, Drutz H, Lye S, Shynlova O. LOX

    family enzymes expression in vaginal tissue of premenopausal

    women with severe pelvic organ prolapse. Int Urogynecol J.

    2010;21(11):1397-1404.

    15. Bortolini MA, Shynlova O, Drutz HP, et al. Expression of bone

    morphogenetic protein-1 in vaginal tissue of women with severe

    pelvic organ prolapse. Am J Obstet Gynecol. 2011;204(6):

    544-548.

    16. Reddy KV, Mangale SS. Integrin receptors: the dynamic modu-

    lators of endometrial function. Tissue Cell. 2003;35(4):260-273.

    17. Martinez-Lemus LA, Wu X, Wilson E, et al. Integrins as unique

    receptors for vascular control. J Vasc Res. 2003;40(3):211-233.

    18. Verma RP, Hansch C. Matrix metalloproteinases (MMPs):

    chemical-biological functions and (Q)SARs. Bioorg Med Chem.

    2007;15(6):2223-2268.

    19. Liang CC, Huang HY, Tseng LH, Chang SD, Lo TS, Lee CL.

    Expression of matrix metalloproteinase-2 and tissue inhibitors of

    metalloproteinase-1 (TIMP1, TIMP2 and TIMP3) in women with

    uterine prolapse but without urinary incontinence. Eur J Obstet

    Gynecol Reprod Biol. 2010;153(1):94-98.

    20. Moalli PA, Shand SH, Zyczynski HM, Gordy SC, Meyn LA.

    Remodeling of vaginal connective tissue in patients with pro-

    lapse. Obstet Gynecol. 2005;106(5 pt 1):953-963.

    21. Phillips CH, Anthony F, Benyon C, Monga AK. Collagen meta-

    bolism in the uterosacral ligaments and vaginal skin of women

    with uterine prolapse. BJOG. 2006;113(1):39-46.

    22. Shynlova O, Bortolini MA, Alarab M. Genes responsible for

    vaginal extracellular matrix metabolism are modulated by

    women’s reproductive cycle and menopause. Int Braz J Urol.

    2013;39(2):257-267.

    23. Srikhajon K, Shynlova O, Preechapornprasert A, Chanrachakul

    B, Lye S. A new role for monocytes in modulating myometrial

    inflammation during human labor. Biol Reprod. 2014;91(1):10.

    24. Medel S, Alarab M, Kufaishi H, Drutz H, Shynlova O. Attach-

    ment of primary vaginal fibroblasts to absorbable and nonabsorb-

    able implant materials coated with platelet-rich plasma: potential

    application in pelvic organ prolapse surgery. Female Pelvic Med

    Reconstr Surg. 2015;21(4):190-197.

    25. Maher CM, Feiner B, Baessler K, Glazener CM. Surgical

    management of pelvic organ prolapse in women: the updated

    summary version Cochrane review. Int Urogynecol J. 2011;22:

    1445-1457.

    26. Boennelycke M, Christensen L, Nielsen LF, Gras S, Lose G.

    Fresh muscle fiber fragments on a scaffold in rats—a new concept

    in urogynecology? Am J Obstet Gynecol. 2011;205(3):235-234.

    27. Ho MH, Heydarkhan S, Vernet D, et al. Stimulating vaginal repair

    in rats through skeletal muscle-derived stem cells seeded on small

    intestinal submucosal scaffolds. Obstet Gynecol. 2009;114(2 pt

    1):300-309.

    28. Hung MJ, Wen MC, Hung CN, Ho ES, Chen GD, Yang VC.

    Tissue-engineered fascia from vaginal fibroblasts for patients

    needing reconstructive pelvic surgery. Int Urogynecol J. 2010;

    21(9):1085-1093.

    942 Reproductive Sciences 23(7)

    http://rs.sagepub.com/supplementalhttp://rs.sagepub.com/supplemental

  • 29. Ulrich D, Edwards SL, Su K, et al. Human endometrial mesench-

    ymal stem cells modulate the tissue response and mechanical

    behavior of polyamide mesh implants for pelvic organ prolapse

    repair. Tissue Eng Part A. 2014;20(3-4):785-798.

    30. Kawano H, Kimura-Kuroda J, Komuta Y, et al. Role of the lesion

    scar in the response to damage and repair of the central nervous

    system. Cell Tissue Res. 2012;349(1):169-180.

    31. Chelberg MK, Tsilibary EC, Hauser AR, McCarthy JB. Type IV

    collagen-mediated melanoma cell adhesion and migration: invol-

    vement of multiple, distinct domains of the collagen molecule.

    Cancer Res. 1989;49(17):4796-4802.

    32. Alarab M, Kufaishi H, Lye S, Drutz H, Shynlova O. Expression of

    extracellular matrix-remodeling proteins is altered in vaginal tis-

    sue of premenopausal women with severe pelvic organ prolapse.

    Reprod Sci. 2014;21(6):704-715.

    33. Chen BH, Wen Y, Li H, Polan ML. Collagen metabolism and

    turnover in women with stress urinary incontinence and pelvic

    prolapse. Int Urogynecol J Pelvic Floor Dysfunct. 2002;13(2):

    80-87.

    34. Kannan K, McConnell A, McLeod M, Rane A. Microscopic

    alterations of vaginal tissue in women with pelvic organ prolapse.

    J Obstet Gynaecol. 2011;31(3):250-253.

    35. Makinen J, Kahari VM, Soderstrom KO, Vuorio E, Hirvonen T.

    Collagen synthesis in the vaginal connective tissue of patients

    with and without uterine prolapse. Eur J Obstet Gynecol Reprod

    Biol. 1987;24(4):319-325.

    36. Kerkhof MH, Ruiz-Zapata AM, Bril H, et al. Changes in tissue

    composition of the vaginal wall of premenopausal women with

    prolapse. Am J Obstet Gynecol. 2014;210(2):168-169.

    37. Muona A, Eklund L, Vaisanen T, Pihlajaniemi T. Developmentally

    regulated expression of type XV collagen correlates with abnorm-

    alities in Col15a1(-/-) mice. Matrix Biol. 2002;21(1):89-102.

    38. Nishie W, Natsuga K, Nakamura H, et al. A recurrent ‘hot spot’

    glycine substitution mutation, G2043 R in COL7A1, induces

    dominant dystrophic epidermolysis bullosa associated with intra-

    cytoplasmic accumulation of pro-collagen VII. J Dermatol Sci.

    2014;75(1):69-71.

    39. Gailit J, Pierschbacher M, Clark RA. Expression of functional

    alpha 4 beta 1 integrin by human dermal fibroblasts. J Invest

    Dermatol. 1993;100(3):323-328.

    40. Valerius MT, Patterson LT, Feng Y, Potter SS. Hoxa 11 is

    upstream of Integrin alpha8 expression in the developing kidney.

    Proc Natl Acad Sci U S A. 2002;99(12):8090-8095.

    41. Ma Y, Guess M, Datar A, et al. Knockdown of Hoxa11 in vivo in

    the uterosacral ligament and uterus of mice results in altered

    collagen and matrix metalloproteinase activity. Biol Reprod.

    2012;86(4):100.

    42. Bump RC, Norton PA. Epidemiology and natural history of pelvic

    floor dysfunction. Obstet Gynecol Clin North Am. 1998;25(4):

    723-746.

    43. Swift S, Woodman P, O’Boyle A, et al. Pelvic Organ Support

    Study (POSST): the distribution, clinical definition, and epide-

    miologic condition of pelvic organ support defects. Am J Obstet

    Gynecol. 2005;192(3):795-806.

    44. Dallenbach P, Kaelin-Gambirasio I, Dubuisson JB, Boulvain M.

    Risk factors for pelvic organ prolapse repair after hysterectomy.

    Obstet Gynecol. 2007;110(3):625-632.

    45. Lin SY, Tee YT, Ng SC, Chang H, Lin P, Chen GD. Changes in

    the extracellular matrix in the anterior vagina of women with or

    without prolapse. Int Urogynecol J Pelvic Floor Dysfunct. 2007;

    18(1):43-48.

    46. Salman MC, Ozyuncu O, Sargon MF, Kucukali T, Durukan T.

    Light and electron microscopic evaluation of cardinal ligaments

    in women with or without uterine prolapse. Int Urogynecol J.

    2010;21(12):235-239.

    47. Szauter KM, Cao T, Boyd CD, Csiszar K. Lysyl oxidase in devel-

    opment, aging and pathologies of the skin. Pathol Biol (Paris).

    2005;53(7):448-456.

    48. Lambert CA, Colige AC, Lapiere CM, Nusgens BV. Coordinated

    regulation of procollagens I and III and their post-translational

    enzymes by dissipation of mechanical tension in human dermal

    fibroblasts. Eur J Cell Biol. 2001;80(7):479-485.

    49. Howard EW, Crider BJ, Updike DL, et al. MMP-2 expression by

    fibroblasts is suppressed by the myofibroblast phenotype. Exp

    Cell Res. 2012;318(13):1542-1553.

    50. Ruiz-Zapata AM, Kerkhof MH, Zandieh-Doulabi B, Brolmann

    HA, Smit TH, Helder MN. Fibroblasts from women with pel-

    vic organ prolapse show differential mechanoresponses

    depending on surface substrates. Int Urogynecol J. 2013;

    24(9):1567-1575.

    51. Ruiz-Zapata AM, Kerkhof MH, Zandieh-Doulabi B, Brolmann

    HA, Smit TH, Helder MN. Functional characteristics of vaginal

    fibroblastic cells from premenopausal women with pelvic organ

    prolapse. Mol Hum Reprod. 2014;20(11):1135-1143.

    52. Lafleur MA, Tester AM, Thompson EW. Selective involvement

    of TIMP2 in the second activational cleavage of pro-MMP-2:

    refinement of the pro-MMP-2 activation mechanism. FEBS Lett.

    2003;553(3):457-463.

    53. Guo C, Piacentini L. Type I collagen-induced MMP-2 activation

    coincides with up-regulation of membrane type 1-matrix metal-

    loproteinase and TIMP2 in cardiac fibroblasts. J Biol Chem. 2003;

    278(47):46699-46708.

    54. Borrirukwanit K, Lafleur MA, Mercuri FA, et al. The type I

    collagen induction of MT1-MMP-mediated MMP-2 activation

    is repressed by alphaVbeta3 integrin in human breast cancer cells.

    Matrix Biol. 2007;26(4):291-305.

    55. Brooks PC, Stromblad S, Sanders LC, et al. Localization of

    matrix metalloproteinase MMP-2 to the surface of invasive cells

    by interaction with integrin alpha v beta 3. Cell. 1996;85(5):

    683-693.

    Kufaishi et al 943

    /ColorImageDict > /JPEG2000ColorACSImageDict > /JPEG2000ColorImageDict > /AntiAliasGrayImages false /CropGrayImages false /GrayImageMinResolution 266 /GrayImageMinResolutionPolicy /OK /DownsampleGrayImages true /GrayImageDownsampleType /Average /GrayImageResolution 175 /GrayImageDepth -1 /GrayImageMinDownsampleDepth 2 /GrayImageDownsampleThreshold 1.50286 /EncodeGrayImages true /GrayImageFilter /DCTEncode /AutoFilterGrayImages true /GrayImageAutoFilterStrategy /JPEG /GrayACSImageDict > /GrayImageDict > /JPEG2000GrayACSImageDict > /JPEG2000GrayImageDict > /AntiAliasMonoImages false /CropMonoImages false /MonoImageMinResolution 900 /MonoImageMinResolutionPolicy /OK /DownsampleMonoImages true /MonoImageDownsampleType /Average /MonoImageResolution 175 /MonoImageDepth -1 /MonoImageDownsampleThreshold 1.50286 /EncodeMonoImages true /MonoImageFilter /CCITTFaxEncode /MonoImageDict > /AllowPSXObjects false /CheckCompliance [ /None ] /PDFX1aCheck false /PDFX3Check false /PDFXCompliantPDFOnly false /PDFXNoTrimBoxError true /PDFXTrimBoxToMediaBoxOffset [ 0.00000 0.00000 0.00000 0.00000 ] /PDFXSetBleedBoxToMediaBox false /PDFXBleedBoxToTrimBoxOffset [ 0.00000 0.00000 0.00000 0.00000 ] /PDFXOutputIntentProfile (U.S. Web Coated \050SWOP\051 v2) /PDFXOutputConditionIdentifier (CGATS TR 001) /PDFXOutputCondition () /PDFXRegistryName (http://www.color.org) /PDFXTrapped /Unknown

    /Description > /Namespace [ (Adobe) (Common) (1.0) ] /OtherNamespaces [ > > /FormElements true /GenerateStructure false /IncludeBookmarks false /IncludeHyperlinks false /IncludeInteractive false /IncludeLayers false /IncludeProfiles true /MarksOffset 9 /MarksWeight 0.125000 /MultimediaHandling /UseObjectSettings /Namespace [ (Adobe) (CreativeSuite) (2.0) ] /PDFXOutputIntentProfileSelector /DocumentCMYK /PageMarksFile /RomanDefault /PreserveEditing true /UntaggedCMYKHandling /UseDocumentProfile /UntaggedRGBHandling /UseDocumentProfile /UseDocumentBleed false >> ] /SyntheticBoldness 1.000000>> setdistillerparams> setpagedevice