Next Article in Journal
Conventional and Unconventional Mechanisms by which Exocytosis Proteins Oversee β-cell Function and Protection
Next Article in Special Issue
Novel Molecular Insights into Human Lipid-Mediated T Cell Immunity
Previous Article in Journal
Atorvastatin Inhibits Endothelial PAI-1-Mediated Monocyte Migration and Alleviates Radiation-Induced Enteropathy
Previous Article in Special Issue
Genetic Bias, Diversity Indices, Physiochemical Properties and CDR3 Motifs Divide Auto-Reactive from Allo-Reactive T-Cell Repertoires
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

In Vitro Suppression of T Cell Proliferation Is a Conserved Function of Primary and Immortalized Human Cancer-Associated Fibroblasts

1
Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton 3800, Australia
2
Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
3
Australian Regenerative Medicine Institute, Monash University, Clayton 3800, Australia
4
Institute of Immunology and Immunotherapy, University of Birmingham, Edgbaston B15 2TT, UK
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(4), 1827; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22041827
Submission received: 5 November 2020 / Revised: 3 February 2021 / Accepted: 4 February 2021 / Published: 12 February 2021
(This article belongs to the Special Issue Recent Advances in T Cell Immunity)

Abstract

:
T cell immunotherapy is now a mainstay therapy for several blood-borne cancers as well as metastatic melanoma. Unfortunately, many epithelial tumors respond poorly to immunotherapy, and the reasons for this are not well understood. Cancer-associated fibroblasts (CAFs) are the most frequent non-neoplastic cell type in most solid tumors, and they are emerging as a key player in immunotherapy resistance. A range of immortalized CAF lines will be essential tools that will allow us to understand immune responses against cancer and develop novel strategies for cancer immunotherapy. To study the effect of CAFs on T cell proliferation, we created and characterized a number of novel immortalized human CAFs lines (Im-CAFs) from human breast, colon, and pancreatic carcinomas. Im-CAFs shared similar phenotypes, matrix remodeling and contraction capabilities, and growth and migration rates compared to the primary CAFs. Using primary isolates from breast carcinoma, colorectal carcinoma, and pancreatic ductal adenocarcinoma, we report that CAFs across major tumor types are able to potently suppress T cell proliferation in vitro. Im-CAFs retained this property. Im-CAFs are a key tool that will provide important insights into the mechanisms of CAF-mediated T cell suppression through techniques such as CRISPR-Cas9 modification, molecular screens, and pipeline drug testing.

1. Introduction

Immune surveillance is a monitoring process in which immune cells target transformed or malignant cells for recognition and elimination [1]. Tumors overcome immune surveillance by acquiring mechanisms to evade or suppress the immune system. T cell cancer immunotherapies have shown promising results across multiple types of malignancies, by improving the host immune response against tumors and overcoming immune evasion. Examples of these therapies are immune checkpoint blockade and chimeric antigen receptor (CAR) T cells, which respectively seek to reawaken or infuse anti-tumor T cells [2,3].
However, despite the clinical success of immune checkpoint inhibitors and CAR-T cells in treating several circulating hematologic cancers, they have not been as effective in solid tumors [4]. Reasons for this are still an active area of study; however, one key difference between these tumor types is the presence of the tumor microenvironment, also termed the tumor stroma. Emerging evidence suggests that this could be a major obstacle for immunotherapies [5].
The tumor microenvironment is comprised of cells that substantially influence tumor growth, metastasis, and therapeutic resistance, including fibroblasts, adipocytes, endothelial cells, pericytes, macrophages, lymphocytes, and other innate and adaptive immune cells as well as extracellular matrix (ECM) and basement membrane [6,7]. Immune cells such as lymphocytes and macrophages influence pro-tumorigenic and anti-tumorigenic functions through the production of an array of cytokines, chemokines, polypeptide growth factors, hormones, matrix remodeling proteases, and metabolites [8,9]. Adipocytes support tumors predominantly via the production of growth factors and cytokines, and they contribute to resistance to chemotherapies and radiotherapies [10]. Endothelial cells form new vessels via sprouting from locally pre-existing vessels to provide nutritional support and gas exchange to the growing tumor, and they have also been reported as playing important functions in resistance to radiation and chemotherapies [11,12]. Pericytes also play roles in promoting angiogenesis and contributing to resistance to antiangiogenic treatments [13].
Cancer-associated fibroblasts (CAFs) are a dominant cell type found in solid tumors, and their abundance is often associated with poor prognosis [14]. Tumors with a high proportion of CAFs associate with poor patient outcomes in breast, colorectal, pancreatic, urothelial, and oral cancers [15,16,17,18,19,20]. For example, high stroma burden in patients with colorectal carcinoma displayed the worst overall survival and relapse-free survival of any patient group [16]. Moreover, a high expression of epithelial-mesenchymal transition (EMT)/stroma-related gene led to a significantly decreased overall survival in urothelial cancer patients in comparison to low EMT patients [15]. Similarly, lung squamous cell carcinoma patients with expression of vimentin, a mesenchymal marker, have significantly reduced overall survival than patients whose tumors lack vimentin [17].
CAFs are themselves a group of heterogeneous cells that are likely mostly derived from chronically inflamed tissue-resident fibroblasts [21]. Several important roles have been ascribed to CAFs, including extracellular matrix remodeling and secretion of growth factors that regulate tumor survival, angiogenesis, tissue invasion, and metastasis [22]. Murine and human studies have shown that CAFs also play essential roles in promoting immunosuppression and evasion from immune surveillance by inhibiting effector T cell differentiation, promoting regulatory T cells (Treg) and type 2-immunity, sequestering tumor infiltrating T cells away from the tumor epithelium and reducing their migration into the TME [14,23,24,25].
CAFs are often characterized by elevated expression of markers such as alpha-Smooth Muscle Actin (αSMA), fibroblast activation protein (FAP), fibroblast-specific protein 1 (FSP1 or S100A4), Vimentin, CD90 (Thy-1), podoplanin, platelet-derived growth factor receptor-α (PDGFRα), and/or PDGFRβ, which are present in various combinations across different types of tumor and play different pro-tumorgenic functions [26,27,28]. For instance, αSMA expression is responsible for CAF contractility and formation of altered collagen structure in tumors [29]. FAP belongs to the membrane-bound serine protease family and is involved in ECM remodeling [30,31]. PDGFRs are tyrosine kinase receptors, and their expression has been shown to be associated with CAF infiltration into the tumor and tumor development [32,33]. These markers are not unique to CAFs, as they are also expressed in other cell types and in healthy tissue, and as CAFs are heterogeneous, these markers can be differentially expressed in vivo. CAFs lack expression of lineage markers for epithelium and tumor cells, e.g., epithelial cell adhesion molecule (EpCAM), endothelium and lymphatics (e.g., CD31, LYVE1), and hematopoietic cells (e.g., CD45) [18,34,35].
Primary CAFs without immortalization can only be sub-cultured for a limited number of passages. Batch differences are an important consideration as this field progresses and are likely to affect data reproducibility as well as slow down progress in the field by requiring researchers to source primary clinical tissues. Additionally, long-term culture of primary cells may impair the cell morphology, proliferation, and expression of surface markers [36,37]. Moreover, senescent cells are thought to have an altered secretory phenotype that could affect their immunosuppressive abilities [38].
In this study, we generated four novel human immortalized CAF (Im-CAFs) lines from primary human breast carcinoma, colorectal carcinoma, and pancreatic ductal adenocarcinoma tumors. We investigated their phenotypic and functional properties in comparison to their parental counterparts. The aim of this work was to create tools to dissect the mechanisms and pathways associated with CAF-driven T cell suppression, by generating CAF lines that maintain long-term cell proliferation and T cell modulating properties without compromising central CAF characteristics.

2. Results

We obtained primary human CAFs from breast carcinoma (bCAFs), colorectal carcinoma (cCAFs), and pancreatic adenocarcinoma (pCAFs) (patient information shown in Supplementary Table S1). To generate immortalized CAFs, we introduced the simian virus 40 large T (SV40LT) antigen and a fluorescent selection marker (either mCherry or green fluorescent protein (GFP)) via the piggyBac transposon system [39,40]. The gene encoding SV40LT was first cloned into pDONR221 and then subcloned into a piggyBac expression plasmid upstream of an internal ribosome entry site (IRES)-GFP or -mCherry site (Figure 1A). Transfected CAFs were then sorted by fluorescent marker expression using Fluorescence Activated Cell Sorting (FACS) (Figure 1B). Immortalized breast CAFs expressing GFP were termed Im-bCAF; immortalized pancreatic and colorectal CAF expressing mCherry were termed Im-pCAF, Im-cCAF1, and Im-cCAF3 (established from separate cCAF donors), respectively, to differentiate them from parental cell isolates. The immortalized lines showed stable expression of the fluorescent markers over time, outlasting parental primary cells with stable growth out to passage 10 (Figure 1C,D).
Next, we examined the phenotype of immortalized cell lines compared to parental primary CAFs to ensure that expression of major markers was retained. A range of characteristic surface and intracellular markers were used to phenotype the primary and immortalized CAFs including Podoplanin (PDPN), fibroblast activation protein (FAP), CD105 (Endoglin), CD146 (Melanoma cell adhesion molecule (MCAM), Ecto-5′-nucleotidase (CD73), α-smooth muscle actin (αSMA), CD56 (Neural-cell adhesion molecule, NCAM), Platelet-derived growth factor receptor- α and β (CD140α and CD140β), and CD90 (Thy1). Immortalized cells maintained a similar surface phenotype of characteristic CAF markers compared to parental lines (Figure 2), including past the point of primary cell senescence (bCAF1, Figure 2; cCAF1, Supplementary Figure S1). The study was not constructed to robustly compare surface expression levels; however, we did observe a conserved reduction in both CD140a and CD140b expression in the line cCAF1, including at later passages (Supplementary Figure S1). In this line, it is likely to denote an overall reduction in expression of the platelet derived growth factor receptor, which is made up of the CD140a and b subunits (homo- or hetero-dimeric), but it was not observed for other lines. Immortalized cells broadly maintained a CAF phenotype.
Moving forward with an assessment of CAF function, a potential concern with immortalization using SV40 LT is the possibility of genomic changes, which may alter the growth rate of the cell lines; therefore, we next compared the growth rate of the immortalized lines versus the primary CAFs. We found the growth rate was similar between Im-cCAF1, Im-cCAF3, and Im-pCAF lines to compare expression with their parental CAFs. The doubling time for primary bCAFs, which appeared to be reaching senescence, was strongly outpaced by Im-bCAF (Figure 3). In this case, the rapid growth rate of the immortalized sample permitted work with these cells that was not possible with the primary isolate.
In order to show that the immortalization process did not result in functional impairment in motility, we investigated the migration rate between the immortalized CAFs and primary CAFs; wound healing/scratch assays were performed [41]. Primary and Im-CAFs were seeded in 24-well plates until they reached confluency, then they were serum-starved for one hour to ensure cell cycle synchronization before a scratch was performed. Scratches were monitored for 48 h, which demonstrated that immortalized CAFs closed the scratch at a similar migration rate to primary CAFs (Figure 4).
Mechanical remodeling of the ECM by CAFs is a crucial contributor to tumor cell migration and invasion [42]. To assess whether immortalized CAFs are capable of matrix remodeling, we performed collagen gel contraction assays. Primary and immortalized CAFs were seeded in type I collagen gels, and contraction was observed using cells treated with Rho-associated, coiled-coil containing protein kinase (ROCK) inhibitor as a negative control for contraction (Figure 5). Contraction of the gels was then monitored over 48 h, which revealed that Im-bCAF, Im-cCAF1, Im-cCAF3, and Im-pCAF were able contract the gels to an equal extent as primary cells.
Immortalized lines were then compared with primary cells for the ability to suppress T cell activation and proliferation, which is an immunomodulatory function attributed to primary CAFs [43]. This was tested using Carboxyfluorescein succinimidyl ester (CFSE)-labelled T-cells from healthy donors incubated with or without human primary or immortalized CAFs for 96 h, followed by flow cytometric analysis to determine the proliferation index of T cells. Results showed that the immortalized CAFs were able to potently suppress T cell proliferation at a level comparable with primary parental cells (Figure 6). This was conserved past senescence for all cell lines, and out to passage 13 for at least one line (Supplementary Figure S2). Immortalized CAFs are therefore likely to represent a useful tool to dissect the mechanisms and pathways associated with CAF-driven T cell suppression.

3. Discussion

Within a tumor, fibroblasts and myofibroblasts are commonly referred to as cancer-associated fibroblasts (CAFs), which are one of the most abundant cell types found in the tumor stroma [14]. CAFs simultaneously construct and remodel the ECM by expressing collagens and other fibrous proteins, alongside proteolytic enzymes, such as matrix metalloproteases, which degrade the ECM [22]. CAFs have crucial functions in recruiting and regulating leukocyte infiltration and inflammation through production of growth factors, cytokines, and chemokines [14,44].
CAFs create an immunosuppressive TME that indirectly promotes cancer growth [44] and may directly contribute to immunotherapy resistance [27]. CAFs can modulate both innate and adaptive immune systems by direct recruitment of immune cells via release of chemokines and cytokines including CCL2 [4]. Moreover, ECM remodeling by CAFs may impede trafficking of T cells leading to more restrained tumor immunity [45]. CAFs also suppress the function of T cells and inhibit their tumor cell killing capacity by limiting T cell infiltration into the tumor and inhibition of the T cells cytotoxic activity within the TME [46,47,48,49].
In our hands, primary human CAFs were sub-cultured for 3–8 passages before they stopped proliferating, reaching replicative senescence. CAFs undergoing senescence may undergo transcriptional and functional changes that may affect the phenotypes under study. Thus, for experiments that require long-term culture, such as genome editing, immortalization is imperative. In the present study, we successfully immortalized CAFs from different patients by introducing SV40LT via the efficient virus-free piggyBac transposon system [50,51]. To date, several immortalized genes have been used to immortalize cell lines, amongst which SV40LT is one of the most investigated genes. The SV40LT inhibits tumor suppressor p53 and retinoblastoma suppressor gene (RB) to prolong the cell cycle and promote cell immortalization [52,53].
Phenotypic changes are possible after immortalization with the SV40LT gene. The Im-CAFs we created expressed all of the CAF markers used in this study, indicating that the immortalization process with SV40LT did not affect their major phenotype, as evaluated by flow cytometry.
Another key function of CAFs is ECM remodeling through contractility of CAF [42,54]. This is a hallmark of the myofibroblast phenotype [22,55], which can be assessed via collagen gel contraction capability in vitro [56,57]. Our results clearly demonstrate that Im-CAFs are capable of remodeling ECM to contract a collagen gel, again confirming that Im-CAFs shared key phenotypes with the primary CAFs. Im-CAFs also showed a similar migratory capability compared to the parental CAFs. To investigate the growth rate of the immortalized lines, CAFs and Im-CAFs were cultured and monitored for 4 days. Im-CAFs expanded as fast as the primary CAFs, while the primary bCAFs grew slower than Im-bCAFs and had likely reached senescence, highlighting the value of immortalization in permitting continued study beyond the lifespan of the parental primary cell line. Importantly, immortalized cell growth, migration, and T cell suppression were maintained at late passages 9–13, representing 10,000-fold expansion after primary cells senesced.
Data shown here and from others illustrates that CAFs derived from breast, pancreatic, and colorectal tumors inhibit proliferation of human T lymphocytes in vitro [43,58,59]. Our results show that both Im-CAFs and their parental CAFs are capable of inhibiting T cell proliferation, and for the first time that CAFs exerted a conserved T cell suppressive effect across various solid tumor types, at least in vitro. However, T cell suppression is not unique to CAFs; rather, it appears to be a conserved property of activated fibroblasts [60,61,62,63,64], supporting the possibility that this observation is likely to be relevant to human biology and the development of new therapeutics. These early proof-of-concept findings indicate that the Im-CAFs are a key tool that can provide further insights into the underlying mechanisms of CAF-mediated T cell suppression, and to explore further effects on T cell function, including changes to methylation state, differentiation, cytokine production, and tumor cell killing. Further exploration of immortalized CAF phenotypes including transcriptomics and secretory profiles would also be insightful.
Identifying molecules that affect T cell activation, differentiation, and function in cancer is extremely important to the development of effective immunotherapies. A substantial body of literature has shown that a high number of tumor-infiltrating lymphocytes correlates with an increased overall survival in several cancers [65,66,67,68]. Additionally, a durable clinical response to CAR-T therapy correlates with extensive proliferation of CAR-T cells [69]. Moreover, the presence of CAFs is known to create an immunosuppressive TME that suppresses the function and proliferation of T cells and skews T helper differentiation towards Th2 and Treg subtypes [25,27,70,71,72,73]. Our finding that the deregulation of T cell proliferation in the presence of CAFs is a conserved function of CAFs from a variety of tumor types may be highly relevant to cancer immunotherapy. Preclinical data has reported that several growth factors and cytokines released by CAFs are involved in poor T cell outcomes, including prostaglandin E2, indoleamine-2,3-dioxygenase, and transforming growth factor beta [23,74,75,76]. The use of immortalized CAFs will further our understanding of these mechanisms involved in CAF-mediated T cell regulation, including effector function, T helper differentiation, and T cell proliferation, through techniques such as CRISPR-Cas9 modification, molecular screens, and drug testing.
This early proof-of-concept study shows that our immortalized CAFs from multiple tumor types share important characteristics with their parental cells, including expression of CAF markers, migration and contractility rates, and the suppression of T cell proliferation. These properties indicate that the immortalized CAFs could overcome several of the limitations of primary cells by providing a reproducible and robust model. These immortalized CAFs represent tools for future studies exploring genes controlling CAF-T cell interactions, using genetic engineering technologies. Our data further show that the suppression of T cell proliferation is a conserved function of CAFs from multiple tumor types including breast, colorectal, and pancreatic cancers.

4. Materials and Methods

4.1. Primary Cells

Primary human cells from breast (bCAFs), colorectal (cCAFs), and pancreatic CAFs (pCAFs) were purchased from Asterand Bioscience (now BioIVT) and used to generate immortalized cells. All cells were maintained in Alpha Modified Eagle Medium (αMEM) (Sigma-Aldrich) with 10% fetal bovine serum (FBS). Blood from healthy donors was obtained from LifeBlood Australia under material supply deed 18-06VIC-10. All work was conducted with the approval of Monash Human Research Ethics Committee, where CAFs were immortalized under project number 18,025, and blood was obtained and used under project number 11,939. All work was conducted in accordance with institutional guidelines and according to the principles expressed in the Declaration of Helsinki.

4.2. Construction of PiggyBac Transposon Vectors

A cassette containing a cytomegalovirus early enhancer/chicken β-actin (CAG) promoter and downstream gateway recombination site (Addgene #20960) was ligated into a backbone containing a 5′ and 3′ piggybac long-terminal repeats and an SV40 PA terminator (Addgene #80758) at SgfI/EcoRV. Then, using extension PCR, loxP-flanked IRES-GFP and IRES-mCherry fragments were generated and ligated into the piggyBac-CAG-Gateway vector at Bsu361/SphI. Finally, SV40LT was cloned into these plasmids by gateway recombination.

4.3. Establishment of Immortalized CAFs

Early passage primary CAFs (<3 passages) were seeded at a density of 5 × 104 cells/well in 6-well plates in complete media (αMEM/10% FBS). Using Lipofectamine 3000, CAFs were co-transfected with a piggyBac expression vector, which expresses SV40LT with GFP or mCherry, and the piggyBac transposase expression vector, hyPBase (The Sanger Center, pCMV-hyPBase). Four days after transfection, and after refreshing media, the cells were cultured until confluency. Transfected CAFs were then cell-sorted by GFP or mCherry expression using FACSAria machines and FACSDiva software (all from BD Bioscience, San Jose, CA, USA) and further expanded in complete media, testing for maintained fluorescent marker expression at regular intervals.

4.4. Scratch Assay

The migratory behavior of CAFs was measured using a common in vitro wound healing assay [77]. CAFs were seeded at a density of 5 × 104 cells/well in 24-well plates and cultured until confluent. Media was replaced with serum-free αMEM for 2 h to synchronize cell cycling. A scratch was made through the cell monolayer with a 1000 µL pipette tip. The wounded monolayers were gently washed twice with the phosphate-buffered saline (PBS) 1× to remove the debris or the floating cells. The cells were then cultured in 1 mL/well complete media. Scratches and cells were imaged at 0, 24, and 48 h with a Leica DMi8 microscope, and the images were analyzed via ImageJ software, using the “MRI Wound Healing Tool” [78]. Migration distance was measured using the following equation: (area of gap (cm2) at 24 or 48 h)/(area of gap (cm2) at 0 h).

4.5. T Cell Suppression Assay

CAFs (2 × 104) were plated in a 96-well flat-bottom plate in complete media and allowed to adhere for 4 h. Human peripheral blood mononuclear cells (PBMCs) from healthy donors were isolated from whole blood using a density gradient Lymphoprep (Stem Cell Technologies, Canada), according to the manufacturer’s instructions, and then counted using a hemocytometer and Trypan Blue viability dye. PBMCs were then labelled with 5 μM CFSE, as previously described [79]. PBMCs (5 × 105) were co-cultured with CAFs and T cell stimulating beads conjugated to anti-CD2, anti-CD3, and anti-CD28 antibodies (1 bead/4 T cells) (Miltenyi Biotec, NSW, Australia). The final volume per well was 200 μL, and cells were incubated for 96 h in complete media with 1% penicillin-streptomycin (Sigma-Aldrich, NSW, Australia).

4.6. Collagen Gel Contraction Assay

CAFs at a final concentration of 1 × 104 cells/mL were cultured in collagen lattice constructed from 330 μL of a neutralized rat tail Collagen I (final concentration 1 mg/mL and 33% v/v) (Merck Millipore, Victoria, Australia), 33 μL of 10× αMEM stock (i.e., a 1:10 dilution with the volume of collagen added), made in house from αMEM powder (Invitrogen), and 637 μL complete media containing cells to be plated, for a final volume of 1ml. Gel was mixed on ice, and 100 μL was plated in 96-well plates. The resultant mixture was incubated for 2 h at 37 °C to induce gelation, followed by addition of 100 μL of complete media per well with or without 100 μM ROCK inhibitor (Y-27632, Stem Cell). Gels were dislodged from the walls of the well and photographed after 72 h of incubation. Contraction was measured using the following equation: area contracted gel (cm2)/area well (cm2) * 100 = % contraction.

4.7. Cell Proliferation

CAFs were seeded into 12-well plates at a density of 15 × 104 cells per well and supplemented in complete media with 1% penicillin-streptomycin. Cell proliferation was assessed at 24, 48, 72, and 96 h by counting the number of cells after trypan blue staining (Invitrogen, Victoria, Australia).

4.8. Flow Cytometry

Cells obtained from suppression or phenotyping assays were stained at a density of 0.5–5 × 105 cells per sample in 50 μL volume of FACS buffer (PBS containing 0.5% Bovine Serum Albumin (BSA), 2mM EDTA, and 0.1% sodium azide). Cells were stained with 30 μL of Zombie Aqua viability dye (Biolegend, San Diego, CA, USA); prepared at a dilution of 1:1000 in PBS) for 10 min in the dark. Cells were washed using 150 μL of FACS buffer, centrifuged, and decanted. For surface marker staining, the pellets were resuspended in 30 μL of appropriate antibody cocktail in FACS buffer and incubated for 15 min on ice in the dark to stain for surface markers, followed by a washing step. Cells were fixed with 100 μL of fixative (BD Cytofix, BD Biosciences, San Jose, CA, USA) and incubated for 15 min on ice in the dark, and either washed with 100 μL of FACS buffer or, for intracellular staining, permeabilization buffer (prepared at 1/10 with Milli Q water; BD Cytoperm), followed by 30 μL of antibody in permeabilization buffer for 30 min on ice. Cells were washed, resuspended in FACS buffer, and filtered through 70 μm nylon mesh. Samples were acquired on a BD Fortessa X20 (BD Biosciences, San Jose, CA, USA) and analyzed using FlowJo (v10) (BD Biosciences, San Jose, CA, USA). The CAF antibody panel included mouse anti-human CD146 (BV650, P1H12), mouse anti-human CD73 (BV785, AD2), rat anti-human PDPN (Alexa Fluor 647, NC-08), mouse anti-human CD105/Endoglin (PerCP/Cy5.5, 43A3), mouse anti-human PDGFRα (PE/Cy7, 16A1), mouse anti-human PDGFRβ (PE, 18A2), mouse anti-human CD56/NCAM (APC/Cy7, 5.1H11), mouse anti-human CD90/Thy1 (PE/Cy5, 5E10), mouse anti-human CD3 (Pacific Blue, HIB19), rat anti-human CD4 (Alexa Fluor 647, NC-08), and mouse anti-human CD8α (Biotinylated, UCHT1), which were all obtained from Biolegend, San Diego, CA, USA. Mouse anti-human FAP (Alexa Fluor 488, 427819) and mouse anti-human α-SMA (Alexa Fluor 700, 1A4) were both obtained from R&D Systems, Minneapolis, MN, USA.

4.9. Statistical Analysi

Data are presented as mean ± standard deviation. Data analysis was performed using Graph Stat Prizm software. For all analyses, a p-value of <0.05 was considered statistically significant. One-way ANOVA was used to compare three or more groups of normally distributed data, and a one sample t-test was used where data were normalized to a comparator group.

Supplementary Materials

The following are available online at https://0-www-mdpi-com.brum.beds.ac.uk/1422-0067/22/4/1827/s1, Figure S1: Phenotype of immortalized cCAF1 at early and late passages, Figure S2: Immortalized cCAF1 maintain suppression of T-cell proliferation at late passages, Table S1: Patient Information.

Author Contributions

Conceptualization, M.H.A., K.K., and A.L.F.; formal analysis, M.H.A.; investigation, M.H.A., A.T.B., and A.L.F.; methodology, M.H.A., A.T.B., J.H., N.L.P., A.N., and A.L.F.; resources, J.H., N.L.P., and A.N.; supervision, A.L.F.; visualization, M.H.A.; writing—original draft, M.H.A.; writing—review and editing, M.H.A., A.T.B., J.H., N.L.P., A.N., K.K., and A.L.F. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by a Monash Fellowship (A.L.F., K.K.); the Saudi Arabian Cultural Mission, affiliate to the Royal Embassy of Saudi Arabia in Australia, and the Ministry of Education, Saudi Arabia (M.H.A.), and a Monash University Graduate Scholarship (A.T.B.).

Institutional Review Board Statement

Blood from healthy donors was obtained from LifeBlood Australia under material supply deed 18-06VIC-10 and work was conducted with the approval of Monash Human Research Ethics Committee, where CAFs were immortalized under project number 18,025 (approved 12 December 2018), and blood was obtained and used under project number 11,939 (approved 24 August 2018). All work was conducted in accordance with institutional guidelines and according to the principles expressed in the Declaration of Helsinki.

Informed Consent Statement

Informed consent was obtained by LifeBlood Australia from all blood donors involved in this study. All fibroblast samples were purchased from a commercial source.

Data Availability Statement

The data presented in this study are available on request from the corresponding authors.

Conflicts of Interest

A.L.F. is a consultant and Scientific Advisory Board member for Phenomic AI. No commercial funding was used for this work. Funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

Abbreviations

CAFCancer-associated fibroblast
Im-CAFsImmortalized CAFs
CARChimeric antigen receptor
bCAFsBreast carcinoma CAFs
cCAFsColorectal carcinoma CAFs
pCAFsPancreatic adenocarcinoma CAFs
Im-bCAFsImmortalized breast carcinoma CAFs
Im-cCAFsImmortalized colorectal carcinoma CAFs
Im-pCAFsImmortalized pancreatic adenocarcinoma CAFs
SV40LT Simian virus 40 large T
IRES Internal ribosome entry site
ITRInverted terminal repeat
FACSFluorescence Activated Cell Sorting
PDPNPodoplanin
FAPFibroblast activation protein
αSMAα-smooth muscle actin
CD146/MCAMMelanoma cell adhesion molecule
CD105Endoglin
CD73Ecto-5′-nucleotidase
CD56/NCAMNeural-cell adhesion molecule
CD140αPlatelet-derived growth factor receptor- α
CD140βPlatelet-derived growth factor receptor- β
CD90/Thy1Thymus cell antigen 1
ROCKRho-associated, coiled-coil containing protein kinase
CFSECarboxyfluorescein succinimidyl ester
RBretinoblastoma suppressor gene

References

  1. Finn, O.J. A Believer’s Overview of Cancer Immunosurveillance and Immunotherapy. J. Immunol. 2018, 200, 385–391. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Darvin, P.; Toor, S.M.; Nair, V.S.; Elkord, E. Immune checkpoint inhibitors: Recent progress and potential biomarkers. Exp. Mol. Med. 2018, 50, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Brown, C.E.; Mackall, C.L. CAR T cell therapy: Inroads to response and resistance. Nat. Rev. Immunol. 2019, 19, 73–74. [Google Scholar] [CrossRef]
  4. Chen, L.; Qiu, X.; Wang, X.; He, J. FAP positive fibroblasts induce immune checkpoint blockade resistance in colorectal cancer via promoting immunosuppression. Biochem. Biophys. Res. Commun. 2017, 487, 8–14. [Google Scholar] [CrossRef]
  5. Su, S.; Chen, J.; Yao, H.; Liu, J.; Yu, S.; Lao, L.; Wang, M.; Luo, M.; Xing, Y.; Chen, F.; et al. CD10+GPR77+ Cancer-Associated Fibroblasts Promote Cancer Formation and Chemoresistance by Sustaining Cancer Stemness. Cell 2018, 172, 841–856.e16. [Google Scholar] [CrossRef]
  6. Quail, D.F.; Joyce, J.A. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 2013, 19, 1423–1437. [Google Scholar] [CrossRef] [PubMed]
  7. Balkwill, F.R.; Capasso, M.; Hagemann, T. The tumor microenvironment at a glance. J. Cell Sci. 2012, 125, 5591–5596. [Google Scholar] [CrossRef] [Green Version]
  8. Ruffell, B.; DeNardo, D.G.; Affara, N.I.; Coussens, L.M. Lymphocytes in cancer development: Polarization towards pro-tumor immunity. Cytokine Growth Factor Rev. 2010, 21, 3–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Ruffell, B.; Coussens, L.M. Macrophages and Therapeutic Resistance in Cancer. Cancer Cell 2015, 27, 462–472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Choi, J.; Cha, Y.J.; Koo, J.S. Adipocyte biology in breast cancer: From silent bystander to active facilitator. Prog. Lipid Res. 2018, 69, 11–20. [Google Scholar] [CrossRef] [PubMed]
  11. Schaaf, M.B.; Garg, A.D.; Agostinis, P. Defining the role of the tumor vasculature in antitumor immunity and immunotherapy. Cell Death Dis. 2018, 9, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Hida, K.; Maishi, N.; Annan, D.A.; Hida, Y. Contribution of Tumor Endothelial Cells in Cancer Progression. Int. J. Mol. Sci. 2018, 19, 1272. [Google Scholar] [CrossRef] [Green Version]
  13. Ribeiro, A.L.; Okamoto, O.K. Combined Effects of Pericytes in the Tumor Microenvironment. Stem Cells Int. 2015, 2015, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Sahai, E.; Astsaturov, I.; Cukierman, E.; DeNardo, D.G.; Egeblad, M.; Evans, R.M.; Fearon, D.; Greten, F.R.; Hingorani, S.R.; Hunter, T.; et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nat. Rev. Cancer 2020, 20, 174–186. [Google Scholar] [CrossRef] [Green Version]
  15. Wang, L.; Saci, A.; Szabo, P.M.; Chasalow, S.D.; Castillo-Martin, M.; Domingo-Domenech, J.; Siefker-Radtke, A.; Sharma, P.; Sfakianos, J.P.; Gong, Y.; et al. EMT- and stroma-related gene expression and resistance to PD-1 blockade in urothelial cancer. Nat. Commun. 2018, 9, 1–12. [Google Scholar] [CrossRef]
  16. Guinney, J.; Dienstmann, R.; Wang, X.; De Reyniès, A.; Schlicker, A.; Soneson, C.; Marisa, L.; Roepman, P.; Nyamundanda, G.; Angelino, P.; et al. The consensus molecular subtypes of colorectal cancer. Nat. Med. 2015, 21, 1350–1356. [Google Scholar] [CrossRef] [PubMed]
  17. Aruga, N.; Kijima, H.; Masuda, R.; Onozawa, H.; Yoshizawa, T.; Tanaka, M.; Inokuchi, S.; Iwazaki, M. Epithelial-mesenchymal Transition (EMT) is Correlated with Patient’s Prognosis of Lung Squamous Cell Carcinoma. Tokai J. Exp. Clin. Med. 2018, 43, 5–13. [Google Scholar]
  18. Calon, A.; Lonardo, E.; Berenguer-Llergo, A.; Espinet, E.; Hernando-Momblona, X.; Iglesias, M.; Sevillano, M.; Palomo-Ponce, S.; Tauriello, D.V.F.; Byrom, D.; et al. Stromal gene expression defines poor-prognosis subtypes in colorectal cancer. Nat. Genet. 2015, 47, 320–329. [Google Scholar] [CrossRef] [Green Version]
  19. Surowiak, P.; Murawa, D.; Materna, V.; Maciejczyk, A.; Pudelko, M.; Ciesla, S.; Breborowicz, J.; Murawa, P.; Zabel, M.; Dietel, M.; et al. Occurence of stromal myofibroblasts in the invasive ductal breast cancer tissue is an unfavourable prognostic factor. Anticancer Res. 2007, 27, 2917–2924. [Google Scholar]
  20. Marsh, D.; Suchak, K.; Moutasim, K.A.; Vallath, S.; Hopper, C.; Jerjes, W.; Upile, T.; Kalavrezos, N.; Violette, S.M.; Weinreb, P.H.; et al. Stromal features are predictive of disease mortality in oral cancer patients. J. Pathol. 2011, 223, 470–481. [Google Scholar] [CrossRef] [PubMed]
  21. LeBleu, V.S.; Kalluri, R. A peek into cancer-associated fibroblasts: Origins, functions and translational impact. Dis. Model. Mech. 2018, 11, dmm029447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Alexander, J.; Cukierman, E. Stromal dynamic reciprocity in cancer: Intricacies of fibroblastic-ECM interactions. Curr. Opin. Cell Biol. 2016, 42, 80–93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Mariathasan, S.; Turley, S.J.; Nickles, D.; Castiglioni, A.; Yuen, K.; Wang, Y.; Kadel III, E.E.; Koeppen, H.; Astarita, J.L.; Cubas, R.; et al. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 2018, 554, 544–548. [Google Scholar] [CrossRef]
  24. Wu, S.Z.; Roden, D.L.; Wang, C.; Holliday, H.; Harvey, K.; Cazet, A.S.; Murphy, K.J.; Pereira, B.; Al-Eryani, G.; Bartonicek, N.; et al. Stromal cell diversity associated with immune evasion in human triple-negative breast cancer. EMBO J. 2020, 39, 104063. [Google Scholar] [CrossRef]
  25. Ene-Obong, A.; Clear, A.J.; Watt, J.; Wang, J.; Fatah, R.; Riches, J.C.; Marshall, J.F.; Chin-Aleong, J.; Chelala, C.; Gribben, J.G.; et al. Activated Pancreatic Stellate Cells Sequester CD8+ T Cells to Reduce Their Infiltration of the Juxtatumoral Compartment of Pancreatic Ductal Adenocarcinoma. Gastroenterology 2013, 145, 1121–1132. [Google Scholar] [CrossRef] [Green Version]
  26. Neuzillet, C.; Tijeras-Raballand, A.; Ragulan, C.; Cros, J.; Patil, Y.; Martinet, M.; Erkan, M.; Kleeff, J.; Wilson, J.; Apte, M.; et al. Inter- and intra-tumoural heterogeneity in cancer-associated fibroblasts of human pancreatic ductal adenocarcinoma. J. Pathol. 2019, 248, 51–65. [Google Scholar] [CrossRef] [Green Version]
  27. Costa, A.; Kieffer, Y.; Scholer-Dahirel, A.; Pelon, F.; Bourachot, B.; Cardon, M.; Sirven, P.; Magagna, I.; Fuhrmann, L.; Bernard, C.; et al. Fibroblast heterogeneity and immunosuppressive environment in human breast cancer. Cancer Cell 2018, 33, 463–479. [Google Scholar] [CrossRef] [Green Version]
  28. Gascard, P.; Tlsty, T.D. Carcinoma-associated fibroblasts: Orchestrating the composition of malignancy. Genes Dev. 2016, 30, 1002–1019. [Google Scholar] [CrossRef]
  29. Hanley, C.J.; Noble, F.; Ward, M.; Bullock, M.; Drifka, C.; Mellone, M.; Manousopoulou, A.; Johnston, H.E.; Hayden, A.; Thirdborough, S.; et al. A subset of myofibroblastic cancer-associated fibroblasts regulate collagen fiber elongation, which is prognostic in multiple cancers. Oncotarget 2015, 7, 6159–6174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Shi, M.; Yu, D.-H.; Chen, Y.; Zhao, C.-Y.; Zhang, J.; Liu, Q.-H.; Ni, C.-R.; Zhu, M.-H. Expression of fibroblast activation protein in human pancreatic adenocarcinoma and its clinicopathological significance. World J. Gastroenterol. 2012, 18, 840–846. [Google Scholar] [CrossRef] [PubMed]
  31. Lee, H.-O.; Mullins, S.R.; Franco-Barraza, J.; Valianou, M.; Cukierman, E.; Cheng, J.D. FAP-overexpressing fibroblasts produce an extracellular matrix that enhances invasive velocity and directionality of pancreatic cancer cells. BMC Cancer 2011, 11, 1–13. [Google Scholar] [CrossRef] [Green Version]
  32. Tejada, M.L.; Yu, L.; Dong, J.; Jung, K.; Meng, G.; Peale, F.V.; Frantz, G.D.; Hall, L.; Liang, X.; Gerber, H.-P.; et al. Tumor-Driven Paracrine Platelet-Derived Growth Factor Receptor α Signaling Is a Key Determinant of Stromal Cell Recruitment in a Model of Human Lung Carcinoma. Clin. Cancer Res. 2006, 12, 2676–2688. [Google Scholar] [CrossRef] [Green Version]
  33. Forsberg, K.; Valyi-Nagy, I.; Heldin, C.H.; Herlyn, M.; Westermark, B. Platelet-derived growth factor (PDGF) in oncogenesis: Development of a vascular connective tissue stroma in xenotransplanted human melanoma producing PDGF-BB. Proc. Natl. Acad. Sci. USA 1993, 90, 393–397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Öhlund, D.; Handly-Santana, A.; Biffi, G.; Elyada, E.; Almeida, A.S.; Ponz-Sarvise, M.; Corbo, V.; Oni, T.E.; Hearn, S.A.; Lee, E.J.; et al. Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. J. Exp. Med. 2017, 214, 579–596. [Google Scholar] [CrossRef] [PubMed]
  35. Kobayashi, H.; Enomoto, A.; Woods, S.L.; Burt, A.D.; Takahashi, M.; Worthley, D.L. Cancer-associated fibroblasts in gastrointestinal cancer. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 282–295. [Google Scholar] [CrossRef]
  36. Bork, S.; Pfister, S.; Witt, H.; Horn, P.; Korn, B.; Ho, A.D.; Wagner, W. DNA methylation pattern changes upon long-term culture and aging of human mesenchymal stromal cells. Aging Cell 2010, 9, 54–63. [Google Scholar] [CrossRef] [Green Version]
  37. Wagner, W. Implications of long-term culture for mesenchymal stem cells: Genetic defects or epigenetic regulation? Stem Cell Res. Ther. 2012, 3, 1–3. [Google Scholar] [CrossRef] [Green Version]
  38. Davalos, A.R.; Coppe, J.-P.; Campisi, J.; Desprez, P.-Y. Senescent cells as a source of inflammatory factors for tumor progression. Cancer Metastasis Rev. 2010, 29, 273–283. [Google Scholar] [CrossRef] [Green Version]
  39. Ding, S.; Wu, X.; Li, G.; Han, M.; Zhuang, Y.; Xu, T. Efficient Transposition of the piggyBac (PB) Transposon in Mammalian Cells and Mice. Cell 2005, 122, 473–483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Zhao, S.; Jiang, E.; Chen, S.; Gu, Y.; Shangguan, A.J.; Lv, T.; Luo, L.; Yu, Z. PiggyBac transposon vectors: The tools of the human gene encoding. Transl. Lung Cancer Res. 2016, 5, 120–125. [Google Scholar] [PubMed]
  41. Henke, A.; Franco, O.E.; Stewart, G.D.; Riddick, A.C.; Katz, E.; Hayward, S.W.; Thomson, A.A. Reduced Contractility and Motility of Prostatic Cancer-Associated Fibroblasts after Inhibition of Heat Shock Protein 90. Cancers 2016, 8, 77. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Gaggioli, C.; Hooper, S.; Hidalgo-Carcedo, C.; Grosse, R.; Marshall, J.F.; Harrington, K.; Sahai, E. Fibroblast-led collective invasion of carcinoma cells with differing roles for RhoGTPases in leading and following cells. Nat. Cell Biol. 2007, 9, 1392–1400. [Google Scholar] [CrossRef]
  43. Cremasco, V.; Astarita, J.L.; Grauel, A.L.; Keerthivasan, S.; MacIsaac, K.D.; Woodruff, M.C.; Wu, M.; Spel, L.; Santoro, S.; Amoozgar, Z.; et al. FAP Delineates Heterogeneous and Functionally Divergent Stromal Cells in Immune-Excluded Breast Tumors. Cancer Immunol. Res. 2018, 6, 1472–1485. [Google Scholar] [CrossRef] [Green Version]
  44. Kalluri, R. The biology and function of fibroblasts in cancer. Nat. Rev. Cancer 2016, 16, 582–598. [Google Scholar] [CrossRef]
  45. Salmon, H.; Franciszkiewicz, K.; Damotte, D.; Dieu-Nosjean, M.-C.; Validire, P.; Trautmann, A.; Mami-Chouaib, F.; Donnadieu, E. Matrix architecture defines the preferential localization and migration of T cells into the stroma of human lung tumors. J. Clin. Investig. 2012, 122, 899–910. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Joyce, J.A.; Fearon, D.T. T cell exclusion, immune privilege, and the tumor microenvironment. Science 2015, 348, 74–80. [Google Scholar] [CrossRef] [Green Version]
  47. Jiang, H.; Hegde, S.; Knolhoff, B.L.; Zhu, Y.; Herndon, J.M.; Meyer, M.A.; Nywening, T.M.; Hawkins, T.M.N.W.G.; Shapiro, I.M.; Weaver, D.T.; et al. Targeting focal adhesion kinase renders pancreatic cancers responsive to checkpoint immunotherapy. Nat. Med. 2016, 22, 851–860. [Google Scholar] [CrossRef]
  48. Érsek, B.; Silló, P.; Cakir, U.; Molnár, V.; Bencsik, A.; Mayer, B.; Mezey, E.; Kárpáti, S.; Pós, Z.; Németh, K. Melanoma-associated fibroblasts impair CD8+ T cell function and modify expression of immune checkpoint regulators via increased arginase activity. Cell. Mol. Life Sci. 2020, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Goehrig, D.; Nigri, J.; Samain, R.; Wu, Z.; Cappello, P.; Gabiane, G.; Zhang, X.; Zhao, Y.; Kim, I.-S.; Chanal, M.; et al. Stromal protein βig-h3 reprogrammes tumour microenvironment in pancreatic cancer. Gut 2019, 68, 693–707. [Google Scholar] [CrossRef] [Green Version]
  50. Wilson, M.H.; Coates, C.J.; George, A.L. PiggyBac Transposon-mediated Gene Transfer in Human Cells. Mol. Ther. 2007, 15, 139–145. [Google Scholar] [CrossRef] [PubMed]
  51. Sato, M.; Inada, E.; Saitoh, I.; Matsumoto, Y.; Ohtsuka, M.; Miura, H.; Nakamura, S.; Sakurai, T.; Watanabe, S. A combination of targeted toxin technology and the piggyBac-mediated gene transfer system enables efficient isolation of stable transfectants in nonhuman mammalian cells. Biotechnol. J. 2014, 10, 143–153. [Google Scholar] [CrossRef]
  52. Sullivan, C.S.; Grundhoff, A.T.; Tevethia, S.; Pipas, J.M.; Ganem, D. SV40-encoded microRNAs regulate viral gene expression and reduce susceptibility to cytotoxic T cells. Nat. Cell Biol. 2005, 435, 682–686. [Google Scholar] [CrossRef] [PubMed]
  53. Umehara, K.; Sun, Y.; Hiura, S.; Hamada, K.; Itoh, M.; Kitamura, K.; Oshima, M.; Iwama, A.; Saito, K.; Anzai, N.; et al. A New Conditionally Immortalized Human Fetal Brain Pericyte Cell Line: Establishment and Functional Characterization as a Promising Tool for Human Brain Pericyte Studies. Mol. Neurobiol. 2017, 55, 5993–6006. [Google Scholar] [CrossRef] [PubMed]
  54. Yamaguchi, H.; Yoshida, N.; Takanashi, M.; Ito, Y.; Fukami, K.; Yanagihara, K.; Yashiro, M.; Sakai, R. Stromal Fibroblasts Mediate Extracellular Matrix Remodeling and Invasion of Scirrhous Gastric Carcinoma Cells. PLoS ONE 2014, 9, e85485. [Google Scholar] [CrossRef] [Green Version]
  55. Winkler, J.; Abisoye-Ogunniyan, A.; Metcalf, K.J.; Werb, Z. Concepts of extracellular matrix remodelling in tumour progression and metastasis. Nat. Commun. 2020, 11, 1–19. [Google Scholar] [CrossRef]
  56. Patel, A.K.; Vipparthi, K.; Thatikonda, V.; Arun, I.; Bhattacharjee, S.; Sharan, R.; Arun, P.; Singh, S. A subtype of cancer-associated fibroblasts with lower expression of alpha-smooth muscle actin suppresses stemness through BMP4 in oral carcinoma. Oncogenesis 2018, 7, 1–15. [Google Scholar] [CrossRef] [PubMed]
  57. Ridge, K.M.; Shumaker, D.; Robert, A.; Hookway, C.; Gelfand, V.I.; Janmey, P.A.; Lowery, J.; Guo, M.; Weitz, D.A.; Kuczmarski, E.; et al. Methods for Determining the Cellular Functions of Vimentin Intermediate Filaments. Methods Enzymol. 2016, 568, 389–426. [Google Scholar]
  58. O’Malley, G.; Treacy, O.; Lynch, K.; Naicker, S.D.; Leonard, N.A.; Lohan, P.; Dunne, P.D.; Ritter, T.; Egan, L.J.; Ryan, A.E. Stromal Cell PD-L1 Inhibits CD8(+) T-cell Antitumor Immune Responses and Promotes Colon Cancer. Cancer Immunol. Res. 2018, 6, 1426–1441. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Gorchs, L.; Moro, C.F.; Bankhead, P.; Kern, K.P.; Sadeak, I.; Meng, Q.; Rangelova, E.; Kaipe, H. Human Pancreatic Carcinoma-Associated Fibroblasts Promote Expression of Co-inhibitory Markers on CD4+ and CD8+ T-Cells. Front. Immunol. 2019, 10, 847. [Google Scholar] [CrossRef]
  60. Fletcher, A.L.; Lukacs-Kornek, V.; Reynoso, E.D.; Pinner, S.E.; Bellemare-Pelletier, A.; Curry, M.S.; Collier, A.-R.; Boyd, R.L.; Turley, S.J. Lymph node fibroblastic reticular cells directly present peripheral tissue antigen under steady-state and inflammatory conditions. J. Exp. Med. 2010, 207, 689–697. [Google Scholar] [CrossRef]
  61. Dubrot, J.; Duraes, F.D.V.; Potin, L.; Capotosti, F.; Brighouse, D.; Suter, T.; LeibundGut-Landmann, S.; Garbi, N.; Reith, W.; Swartz, M.A.; et al. Lymph node stromal cells acquire peptide-MHCII complexes from dendritic cells and induce antigen-specific CD4+ T cell tolerance. J. Exp. Med. 2014, 211, 1153–1166. [Google Scholar] [CrossRef] [PubMed]
  62. Knoblich, K.; Migoni, S.C.; Siew, S.M.; Jinks, E.; Kaul, B.; Jeffery, H.C.; Baker, A.T.; Suliman, M.; Vrzalikova, K.; Mehenna, H.; et al. The human lymph node microenvironment unilaterally regulates T-cell activation and differentiation. PLoS Biol. 2018, 16, e2005046. [Google Scholar] [CrossRef]
  63. Schaeuble, K.; Cannelle, H.; Favre, S.; Huang, H.-Y.; Oberle, S.G.; Speiser, D.E.; Zehn, D.; Luther, S.A. Attenuation of chronic antiviral T-cell responses through constitutive COX2-dependent prostanoid synthesis by lymph node fibroblasts. PLoS Biol. 2019, 17, e3000072. [Google Scholar] [CrossRef] [Green Version]
  64. Fletcher, A.L.; Acton, S.E.; Knoblich, K. Lymph node fibroblastic reticular cells in health and disease. Nat. Rev. Immunol. 2015, 15, 350–361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Lohr, J.; Ratliff, T.; Huppertz, A.; Ge, Y.; Dictus, C.; Ahmadi, R.; Grau, S.; Hiraoka, N.; Eckstein, V.; Ecker, R.C.; et al. Effector T-Cell Infiltration Positively Impacts Survival of Glioblastoma Patients and Is Impaired by Tumor-Derived TGF-β. Clin. Cancer Res. 2011, 17, 4296–4308. [Google Scholar] [CrossRef] [Green Version]
  66. Fukunaga, A.; Miyamoto, M.; Cho, Y.; Murakami, S.; Kawarada, Y.; Oshikiri, T.; Kato, K.; Kurokawa, T.; Suzuoki, M.; Nakakubo, Y.; et al. CD8+ Tumor-Infiltrating Lymphocytes Together with CD4+ Tumor-Infiltrating Lymphocytes and Dendritic Cells Improve the Prognosis of Patients with Pancreatic Adenocarcinoma. Pancreas 2004, 28, e26–e31. [Google Scholar] [CrossRef]
  67. Narayanan, S.; Kawaguchi, T.; Yan, L.; Peng, X.; Qi, Q.; Takabe, K. Cytolytic Activity Score to Assess Anticancer Immunity in Colorectal Cancer. Ann. Surg. Oncol. 2018, 25, 2323–2331. [Google Scholar] [CrossRef]
  68. Carstens, J.L.; De Sampaio, P.C.; Yang, D.; Barua, S.; Wang, H.; Rao, A.; Allison, J.P.; LeBleu, V.S.; Kalluri, R. Spatial computation of intratumoral T cells correlates with survival of patients with pancreatic cancer. Nat. Commun. 2017, 8, 1–13. [Google Scholar] [CrossRef]
  69. Sheih, A.; Voillet, V.; Hanafi, L.-A.; DeBerg, H.A.; Yajima, M.; Hawkins, R.; Gersuk, V.; Riddell, S.R.; Maloney, D.G.; Wohlfahrt, M.E.; et al. Clonal kinetics and single-cell transcriptional profiling of CAR-T cells in patients undergoing CD19 CAR-T immunotherapy. Nat. Commun. 2020, 11, 1–13. [Google Scholar] [CrossRef] [Green Version]
  70. Lakins, M.A.; Ghorani, E.; Munir, H.; Martins, C.P.; Shields, J.D. Cancer-associated fibroblasts induce antigen-specific deletion of CD8+ T Cells to protect tumour cells. Nat. Commun. 2018, 9, 1–9. [Google Scholar] [CrossRef]
  71. Fox, A.; Harland, K.L.; Kedzierska, K.; Kelso, A. Exposure of Human CD8+ T Cells to Type-2 Cytokines Impairs Division and Differentiation and Induces Limited Polarization. Front. Immunol. 2018, 9, 1141. [Google Scholar] [CrossRef] [PubMed]
  72. Liao, D.; Luo, Y.; Markowitz, D.; Xiang, R.; Reisfeld, R.A. Cancer Associated Fibroblasts Promote Tumor Growth and Metastasis by Modulating the Tumor Immune Microenvironment in a 4T1 Murine Breast Cancer Model. PLoS ONE 2009, 4, e7965. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Kato, T.; Noma, K.; Ohara, T.; Kashima, H.; Katsura, Y.; Sato, H.; Komoto, S.; Katsube, R.; Ninomiya, T.; Tazawa, H.; et al. Cancer-Associated Fibroblasts Affect Intratumoral CD8+ and FoxP3+ T Cells Via IL6 in the Tumor Microenvironment. Clin. Cancer Res. 2018, 24, 4820–4833. [Google Scholar] [CrossRef] [Green Version]
  74. Zelenay, S.; Van Der Veen, A.G.; Böttcher, J.P.; Snelgrove, K.J.; Rogers, N.; Acton, S.E.; Chakravarty, P.; Girotti, M.R.; Marais, R.; Quezada, S.A.; et al. Cyclooxygenase-Dependent Tumor Growth through Evasion of Immunity. Cell 2015, 162, 1257–1270. [Google Scholar] [CrossRef] [Green Version]
  75. Uyttenhove, C.; Pilotte, L.; Théate, I.; Stroobant, V.; Colau, D.; Parmentier, N.; Boon, T.; Van den Eynde, B.J. Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase. Nat. Med. 2003, 9, 1269–1274. [Google Scholar] [CrossRef]
  76. Tauriello, D.V.F.; Palomo-Ponce, S.; Stork, D.; Berenguer-Llergo, A.; Badia-Ramentol, J.; Iglesias, M.; Sevillano, M.; Ibiza, S.; Cañellas, A.; Hernando-Momblona, X.; et al. TGF drives immune evasion in genetically reconstituted colon cancer metastasis. Nature 2018, 554, 538–543. [Google Scholar] [CrossRef] [Green Version]
  77. Liang, C.-C.; Park, A.Y.; Guan, J.-L. In vitro scratch assay: A convenient and inexpensive method for analysis of cell migration in vitro. Nat. Protoc. 2007, 2, 329–333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Bäcker, V. ImageJ Macro Toolsets for Biological Image Analysis. In Proceedings of the ImageJ User and Developer Conference 2012, Luxembourg, 24–26 October 2012. [Google Scholar]
  79. Quah, B.J.C.; Warren, H.S.; Parish, C.R. Monitoring lymphocyte proliferation in vitro and in vivo with the intracellular fluorescent dye carboxyfluorescein diacetate succinimidyl ester. Nat. Protoc. 2007, 2, 2049–2056. [Google Scholar] [CrossRef]
Figure 1. Generation of immortalized cancer associated fibroblasts (Im-CAFs). (A). The simian virus 40 large T (SV40LT) antigen + green fluorescent protein (GFP)/or mCherry expression plasmid was co-transfected into primary CAFs with the transposase expression plasmid. Once inside the cell, the transposase (Tnp, blue) is expressed and recognizes the inverted terminal repeat (ITR, red) sequences flanking the SV40LT and fluorescent marker sequences. The transposase excises the gene of interest, fluorescent marker and flanking ITR sequences from the expression plasmid and integrates it randomly into TTAA recognition sites throughout the genome (yellow). (B). Transfected CAFs were sorted by fluorescent selection marker using fluorescence activated cell sorting (FACS). bCAF sample shown; representative of 4 cell lines. (C). Brightfield and immunofluorescence of primary and immortalized cCAF1 cell lines. Scale bar is 100 μm. (D). Expression of mCherry and GFP in immortalized CAFs at different passages. P = passage number. Red = transfected, grey = untransfected.
Figure 1. Generation of immortalized cancer associated fibroblasts (Im-CAFs). (A). The simian virus 40 large T (SV40LT) antigen + green fluorescent protein (GFP)/or mCherry expression plasmid was co-transfected into primary CAFs with the transposase expression plasmid. Once inside the cell, the transposase (Tnp, blue) is expressed and recognizes the inverted terminal repeat (ITR, red) sequences flanking the SV40LT and fluorescent marker sequences. The transposase excises the gene of interest, fluorescent marker and flanking ITR sequences from the expression plasmid and integrates it randomly into TTAA recognition sites throughout the genome (yellow). (B). Transfected CAFs were sorted by fluorescent selection marker using fluorescence activated cell sorting (FACS). bCAF sample shown; representative of 4 cell lines. (C). Brightfield and immunofluorescence of primary and immortalized cCAF1 cell lines. Scale bar is 100 μm. (D). Expression of mCherry and GFP in immortalized CAFs at different passages. P = passage number. Red = transfected, grey = untransfected.
Ijms 22 01827 g001
Figure 2. Phenotype of immortalized CAFs. Primary CAFs at passage 3 and immortalized (Im-)CAFs at passage 4–5 were phenotyped for a range of characteristic CAF markers. Histograms shown are gated on non-debris, viable cells, and mCherry+ for Im-cCAF1, Im-cCAF3, and Im-pCAF, or GFP+ for Im-bCAF, with no fluorescent selection for primary cells. Data represent 2–3 independent experiments.
Figure 2. Phenotype of immortalized CAFs. Primary CAFs at passage 3 and immortalized (Im-)CAFs at passage 4–5 were phenotyped for a range of characteristic CAF markers. Histograms shown are gated on non-debris, viable cells, and mCherry+ for Im-cCAF1, Im-cCAF3, and Im-pCAF, or GFP+ for Im-bCAF, with no fluorescent selection for primary cells. Data represent 2–3 independent experiments.
Ijms 22 01827 g002
Figure 3. Growth assay of primary CAFs and immortalized (Im-)CAFs. 15.0 × 103 cells of the indicated Im-CAF (passage 4) or primary (passage 3) cell lines were plated and then harvested at the indicated time points. The line graph depicts the mean ± SD of 6 replicates from 2 independent experiments.
Figure 3. Growth assay of primary CAFs and immortalized (Im-)CAFs. 15.0 × 103 cells of the indicated Im-CAF (passage 4) or primary (passage 3) cell lines were plated and then harvested at the indicated time points. The line graph depicts the mean ± SD of 6 replicates from 2 independent experiments.
Ijms 22 01827 g003
Figure 4. Immortalized CAFs are migratory. (A) Representative images of migration of CAFs across a scratch at 0, 24, and 48 h after the scratch was made. Dotted lines depict where the scratch was made. (B) The proportion of the original scratch area closed after 24 and 48 hrs. Data depicts evaluations of primary cells at P3 and immortalized cells from P4–P9. The line graph represents the mean value ±SD of 10–16 replicates per cell line, from 2 independent experiments.
Figure 4. Immortalized CAFs are migratory. (A) Representative images of migration of CAFs across a scratch at 0, 24, and 48 h after the scratch was made. Dotted lines depict where the scratch was made. (B) The proportion of the original scratch area closed after 24 and 48 hrs. Data depicts evaluations of primary cells at P3 and immortalized cells from P4–P9. The line graph represents the mean value ±SD of 10–16 replicates per cell line, from 2 independent experiments.
Ijms 22 01827 g004
Figure 5. Immortalized CAFs are contractile. Primary CAFs and immortalized CAFs were seeded into a collagen gel with or without Rho-associated protein kinase (ROCK) inhibitor (100 mM) for 48 h. (A). Schematic representation of gel remodeling. Gels were detached, the gel contraction was digitally photo-documented, and % contraction calculated as shown. (B). Contraction was measured as a % reduction of gel surface after 48 h. (C). Pooled data representing mean values ± SD of 6 replicates from 2 independent experiments. Primary cells were evaluated at passage 3; immortalized cells at passage 4–5. One-way ANOVA, ns: non-significant, *** p < 0.001, **** p < 0.0001.
Figure 5. Immortalized CAFs are contractile. Primary CAFs and immortalized CAFs were seeded into a collagen gel with or without Rho-associated protein kinase (ROCK) inhibitor (100 mM) for 48 h. (A). Schematic representation of gel remodeling. Gels were detached, the gel contraction was digitally photo-documented, and % contraction calculated as shown. (B). Contraction was measured as a % reduction of gel surface after 48 h. (C). Pooled data representing mean values ± SD of 6 replicates from 2 independent experiments. Primary cells were evaluated at passage 3; immortalized cells at passage 4–5. One-way ANOVA, ns: non-significant, *** p < 0.001, **** p < 0.0001.
Ijms 22 01827 g005
Figure 6. Human primary CAFs and immortalized CAFs inhibit T-cell proliferation. (A). Carboxyfluorescein succinimidyl ester (CFSE)-labelled peripheral blood mononuclear cells (PBMCs) (5 × 105) were cultured with or without CAFs grown to 90% confluency in 96-well plates, with or without anti-CD3/CD28/CD2-coated activation beads. After 96 h, cells were harvested and analyzed using flow cytometry. Histograms depict T-cell CFSE staining on plots gated for CD3 and CD4 or CD8. (B). Normalized division index of gated CFSE+ T cells, which were cultured for 96 h unstimulated or stimulated with anti-CD3/CD28/CD2-coated beads, with or without primary or Im-CAFs. Data are normalized to stimulated T cell controls and represent mean values ± SD of 6 replicates from 2 independent experiments. Data represent primary cells evaluated at passage 3 and immortalized cells at passages 4–13. Statistics: one sample t test, *** p < 0.001, **** p < 0.0001).
Figure 6. Human primary CAFs and immortalized CAFs inhibit T-cell proliferation. (A). Carboxyfluorescein succinimidyl ester (CFSE)-labelled peripheral blood mononuclear cells (PBMCs) (5 × 105) were cultured with or without CAFs grown to 90% confluency in 96-well plates, with or without anti-CD3/CD28/CD2-coated activation beads. After 96 h, cells were harvested and analyzed using flow cytometry. Histograms depict T-cell CFSE staining on plots gated for CD3 and CD4 or CD8. (B). Normalized division index of gated CFSE+ T cells, which were cultured for 96 h unstimulated or stimulated with anti-CD3/CD28/CD2-coated beads, with or without primary or Im-CAFs. Data are normalized to stimulated T cell controls and represent mean values ± SD of 6 replicates from 2 independent experiments. Data represent primary cells evaluated at passage 3 and immortalized cells at passages 4–13. Statistics: one sample t test, *** p < 0.001, **** p < 0.0001).
Ijms 22 01827 g006
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Abuwarwar, M.H.; Baker, A.T.; Harding, J.; Payne, N.L.; Nagy, A.; Knoblich, K.; Fletcher, A.L. In Vitro Suppression of T Cell Proliferation Is a Conserved Function of Primary and Immortalized Human Cancer-Associated Fibroblasts. Int. J. Mol. Sci. 2021, 22, 1827. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22041827

AMA Style

Abuwarwar MH, Baker AT, Harding J, Payne NL, Nagy A, Knoblich K, Fletcher AL. In Vitro Suppression of T Cell Proliferation Is a Conserved Function of Primary and Immortalized Human Cancer-Associated Fibroblasts. International Journal of Molecular Sciences. 2021; 22(4):1827. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22041827

Chicago/Turabian Style

Abuwarwar, Mohammed H., Alfie T. Baker, Jeffrey Harding, Natalie L. Payne, Andras Nagy, Konstantin Knoblich, and Anne L. Fletcher. 2021. "In Vitro Suppression of T Cell Proliferation Is a Conserved Function of Primary and Immortalized Human Cancer-Associated Fibroblasts" International Journal of Molecular Sciences 22, no. 4: 1827. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22041827

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop