1
|
Sculthorpe DJ, Denton A, Fadhil W, Rusnita D, Ilyas M, Mukherjee A. High α-SMA expression in the tumor stroma is associated with adverse clinical parameters in mismatch repair-proficient colorectal cancers only. Am J Clin Pathol 2025; 163:464-472. [PMID: 39495028 PMCID: PMC11890275 DOI: 10.1093/ajcp/aqae145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 10/01/2024] [Indexed: 11/05/2024] Open
Abstract
OBJECTIVES As mismatch repair status confers differential prognosis in colorectal cancers, this study aimed to determine associations of α-smooth muscle actin (α-SMA) protein expression in mismatch repair-proficient (pMMR) and mismatch repair-deficient (dMMR) colorectal tumors with clinicopathologic and prognostic features. METHODS Tissue microarrays from patients with colorectal cancer, immunostained with α-SMA, were assessed through digital image analysis. Total (n = 962), pMMR (n = 782), and dMMR (n = 156) stromal H-scores were assessed for associations with clinicopathologic and survival data. RESULTS Higher α-SMA expression was correlated with pMMR status (P = 5.2223 × 10-8). In the pMMR subgroup, higher α-SMA stromal expression at the tumor periphery was correlated with higher T stage (P = .002), perineural invasion (P = .038), infiltrative tumor edge (P = .01), involved nodal status (P = .036), metastases (P = .013), synchronous metastases (P = .007), recurrence (P = .004), and both 3-year and 5-year survival (P = .018). dMMR tumors showed no significant correlations with α-SMA staining. CONCLUSIONS The findings highlight that immunostaining with α-SMA in pMMR colorectal tumors, especially at the tumor periphery, has the potential to identify patients with adverse prognostic features. Digital assessment of α-SMA may offer improved objectivity, accuracy, economy of time, and risk stratification for management.
Collapse
Affiliation(s)
- Declan J Sculthorpe
- Molecular Pathology Research, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, UK
| | - Amy Denton
- Molecular Pathology Research, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, UK
| | - Wakkas Fadhil
- Molecular Pathology Research, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, UK
| | - Dewi Rusnita
- Molecular Pathology Research, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, UK
| | - Mohammad Ilyas
- Molecular Pathology Research, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, UK
- Department of Histopathology, Nottingham University Hospitals NHS Trust, Queen’s Medical Centre, Nottingham, UK
| | - Abhik Mukherjee
- Molecular Pathology Research, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, UK
- Department of Histopathology, Nottingham University Hospitals NHS Trust, Queen’s Medical Centre, Nottingham, UK
| |
Collapse
|
2
|
Wu NC, Quevedo R, Nurse M, Hezaveh K, Liu H, Sun F, Muffat J, Sun Y, Simmons CA, McGaha TL, Prinos P, Arrowsmith CH, Ailles L, D'Arcangelo E, McGuigan AP. The use of a multi-metric readout screen to identify EHMT2/G9a-inhibition as a modulator of cancer-associated fibroblast activation state. Biomaterials 2025; 314:122879. [PMID: 39395244 DOI: 10.1016/j.biomaterials.2024.122879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 09/20/2024] [Accepted: 10/04/2024] [Indexed: 10/14/2024]
Abstract
Cancer-associated fibroblasts (CAFs) play a pivotal role in cancer progression, including mediating tumour cell invasion via their pro-invasive secretory profile and ability to remodel the extracellular matrix (ECM). Given that reduced CAF abundance in tumours correlates with improved outcomes in various cancers, we set out to identify epigenetic targets involved in CAF activation in regions of tumour-stromal mixing with the goal of reducing tumour aggressiveness. Using the GLAnCE (Gels for Live Analysis of Compartmentalized Environments) platform, we performed an image-based, phenotypic screen that enabled us to identify modulators of CAF abundance and the capacity of CAFs to induce tumour cell invasion. We identified EHMT2 (also known as G9a), an enzyme that targets the methylation of histone 3 lysine 9 (H3K9), as a potent modulator of CAF abundance and CAF-mediated tumour cell invasion. Transcriptomic and functional analysis of EHMT2-inhibited CAFs revealed EHMT2 participated in driving CAFs towards a pro-invasive phenotype and mediated CAF hyperproliferation, a feature typically associated with activated fibroblasts in tumours. Our study suggests that EHMT2 regulates CAF state within the tumour microenvironment by impacting CAF activation, as well as by magnifying the pro-invasive effects of these activated CAFs on tumour cell invasion through promoting CAF hyperproliferation.
Collapse
Affiliation(s)
- Nila C Wu
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Rene Quevedo
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Michelle Nurse
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, Toronto, ON, Canada
| | - Kebria Hezaveh
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Haijiao Liu
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada; Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada
| | - Fumao Sun
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; The Hospital for Sick Children, Toronto, ON, Canada
| | - Julien Muffat
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; The Hospital for Sick Children, Toronto, ON, Canada
| | - Yu Sun
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada
| | - Craig A Simmons
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada; Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada
| | - Tracy L McGaha
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Panagiotis Prinos
- Structural Genomics Consortium, University of Toronto, Toronto, ON, Canada
| | - Cheryl H Arrowsmith
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Structural Genomics Consortium, University of Toronto, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Laurie Ailles
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Elisa D'Arcangelo
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada.
| | - Alison P McGuigan
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Department of Chemical Engineering & Applied Chemistry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
3
|
Kraus SG, Johnson KA, Emmerich PB, Clipson L, Pasch CA, Zhang W, Matkowskyj KA, Deming DA. Micro-environmental changes indicate potential for subclinical intestinal tissue damage in early-age-onset colorectal cancer patients. Gastroenterol Rep (Oxf) 2025; 13:goaf015. [PMID: 39980836 PMCID: PMC11842056 DOI: 10.1093/gastro/goaf015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 12/17/2024] [Accepted: 01/14/2025] [Indexed: 02/22/2025] Open
Abstract
Background While improved screening rates have contributed to an overall decrease in the incidence of colorectal cancer (CRC), the incidence of early-age-onset CRC (EAO CRC; age <50 years) has increased. Here, we characterize the genetic alterations and tumor microenvironment (TME) for EAO and later-age-onset (LAO) CRCs to identify relevant biological differences that might point to etiologic factors. Methods A cohort of EAO (n = 60) and LAO (n = 93) CRC patients were evaluated for mutations by using targeted DNA sequencing and for TME differences by using immunohistochemistry and immunofluorescence. The Cancer Genome Atlas (TCGA) PanCancer Atlas colorectal adenocarcinoma cohort was evaluated for transcriptional changes between EAO (n = 82) and LAO (n = 510) patients. Results KRAS and BRAF mutations were less frequent in EAO CRCs. Gene-set enrichment analysis of TCGA data revealed the downregulation of immune-related pathways in EAO CRCs. Both age cohorts had similar numbers of CD8+ tumor-infiltrating lymphocytes (TILs), although LAO patients had more CD4+ TILs and Th1-polarized CD4s. While significant associations between immune subsets and versican (VCAN), versikine, and alpha-smooth muscle actin (αSMA) were found, none of these trends differed between age cohorts. EAO patients trended towards greater VCAN accumulation in adjacent normal tissue, lower rates of VCAN proteolysis, and decreased αSMA accumulation vs LAO patients. Conclusions Overall, established EAO cancers are similar to LAO cancers in mutational profile and key TME features. High VCAN and αSMA expression in adjacent normal colon indicates a presence of factors that are associated with increased intestinal subclinical inflammation. Future mechanistic studies will be conducted to better understand the importance of these findings and related processes should be prioritized as potential etiologic factors for EAO tumorigenesis.
Collapse
Affiliation(s)
- Sean G Kraus
- Division of Hematology, Medical Oncology, and Palliative Care, Department of Medicine, University of Wisconsin School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI, USA
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin–Madison, Madison, WI, USA
| | - Katherine A Johnson
- Division of Hematology, Medical Oncology, and Palliative Care, Department of Medicine, University of Wisconsin School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI, USA
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin–Madison, Madison, WI, USA
| | - Philip B Emmerich
- Division of Hematology, Medical Oncology, and Palliative Care, Department of Medicine, University of Wisconsin School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI, USA
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin–Madison, Madison, WI, USA
| | - Linda Clipson
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin–Madison, Madison, WI, USA
| | - Cheri A Pasch
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Wei Zhang
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin–Madison, Madison, WI, USA
| | - Kristina A Matkowskyj
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin–Madison, Madison, WI, USA
- William S. Middleton Veterans Administration Health System, Madison, WI, USA
| | - Dustin A Deming
- Division of Hematology, Medical Oncology, and Palliative Care, Department of Medicine, University of Wisconsin School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI, USA
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin–Madison, Madison, WI, USA
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| |
Collapse
|
4
|
Liang C, Wang M, Huang Y, Yam JWP, Zhang X, Zhang X. Recent Advances of Small Extracellular Vesicles for the Regulation and Function of Cancer-Associated Fibroblasts. Int J Mol Sci 2024; 25:12548. [PMID: 39684264 DOI: 10.3390/ijms252312548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/12/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs) are a heterogeneous cell population in the tumor microenvironment (TME) that critically affect cancer progression. Small extracellular vesicles (sEVs) act as information messengers by transmitting a wide spectrum of biological molecules, including proteins, nucleic acids, and metabolites, from donor cells to recipient cells. Previous studies have demonstrated that CAFs play important roles in tumor progression by regulating tumor cell proliferation, metastasis, therapeutic resistance, and metabolism via sEVs. In turn, tumor-derived sEVs can also regulate the activation and phenotype switch of CAFs. The dynamic crosstalk between CAFs and cancer cells via sEVs could ultimately determine cancer progression. In this review, we summarized the recent advance of the biological roles and underlying mechanisms of sEVs in mediating CAF-tumor cell interaction and its impact on cancer progression. We also reviewed the clinical applications of tumor- and CAF-derived sEVs, which could identify novel potential targets and biomarkers for cancer diagnosis, therapy, and prognosis.
Collapse
Affiliation(s)
- Chengdong Liang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Maoye Wang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Yongli Huang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Judy Wai Ping Yam
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Xu Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Xiaoxin Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| |
Collapse
|
5
|
Eskandari-Malayeri F, Rezeai M, Narimani T, Esmaeil N, Azizi M. Investigating the effect of Fusobacterium nucleatum on the aggressive behavior of cancer-associated fibroblasts in colorectal cancer. Discov Oncol 2024; 15:292. [PMID: 39030445 PMCID: PMC11264641 DOI: 10.1007/s12672-024-01156-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 07/13/2024] [Indexed: 07/21/2024] Open
Abstract
Fusobacterium nucleatum, (F. nucleatum) as a known factor in inducing oncogenic, invasive, and inflammatory responses, can lead to an increase in the incidence and progression of colorectal cancer (CRC). Cancer-associated fibroblasts (CAF) are also one of the key components of the tumor microenvironment (TME), which lead to resistance to treatment, metastasis, and disease recurrence with their markers, secretions, and functions. This study aimed to investigate the effect of F. nucleatum on the invasive phenotype and function of fibroblast cells isolated from normal and cancerous colorectal tissue. F. nucleatum bacteria were isolated from deep periodontal pockets and confirmed by various tests. CAF cells from tumor tissue and normal fibroblasts (NF) from a distance of 10 cm of tumor tissue were isolated from 5 patients by the explant method and were exposed to secretions and ghosts of F. nucleatum. The expression level of two markers, fibroblast activation protein (FAP), and α-smooth muscle actin (α-SMA), and the amount of production of two cytokines TGF-β and IL-6 from fibroblast cells were measured by flow cytometry and ELISA test, respectively before and after exposure to different bacterial components. The expression of the FAP marker was significantly higher in CAF cells compared to NF cells (P < 0.05). Also, the expression of IL-6 in CAF cells was higher than that of NF cells. In investigating the effect of bacterial components on the function of fibroblastic cells, after comparing the amount of IL-6 produced between the normal tissue of each patient and his tumoral tissue under 4 treated conditions, it was found that the amount of IL-6 production from the CAF cells of patients in the control group, treated with heat-killed ghosts and treated with paraformaldehyde-fixed ghosts had a significant increase compared to NF cells (P < 0.05). Due to the significant increase in FAP marker expression in fibroblast cells of tumor tissue compared to normal tissue, it seems that FAP can be used as a very good therapeutic marker, especially in patients with high levels of CAF cells. Various components of F. nucleatum could affect fibroblast cells differentially and at least part of the effect of this bacterium in the TME is mediated by CAF cells.
Collapse
Affiliation(s)
| | - Marzieh Rezeai
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Tahmineh Narimani
- Department of Microbiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nafiseh Esmaeil
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahdieh Azizi
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
6
|
Verma S, Giagnocavo SD, Curtin MC, Arumugam M, Osburn-Staker SM, Wang G, Atkinson A, Nix DA, Lum DH, Cox JE, Hilgendorf KI. Zinc-alpha-2-glycoprotein Secreted by Triple-Negative Breast Cancer Promotes Peritumoral Fibrosis. CANCER RESEARCH COMMUNICATIONS 2024; 4:1655-1666. [PMID: 38888911 PMCID: PMC11224648 DOI: 10.1158/2767-9764.crc-24-0218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
Obesity is a modifiable predisposition factor for postmenopausal breast cancer. This suggests a localized, reciprocal interaction between breast cancer cells and the surrounding mammary white adipose tissue. To investigate how breast cancer cells alter the composition and function of adipose tissue, we screened the secretomes of 10 human breast cancer cell lines for the ability to modulate the differentiation of adipocyte stem and progenitor cells. The screen identified an adipogenic modulator, zinc-alpha-2-glycoprotein (ZAG/AZGP1) that is secreted by triple-negative breast cancer (TNBC) cells. TNBC-secreted ZAG inhibits adipogenesis and instead induces the expression of fibrotic genes. Accordingly, depletion of ZAG in TNBC cells attenuates fibrosis in white adipose tissue and inhibits tumor growth. Further, high expression of ZAG is linked to poor prognosis in patients with TNBC but not in patients with other clinical subtypes of breast cancer. Our findings suggest a role of TNBC-secreted ZAG in promoting the transdifferentiation of adipocyte stem and progenitor cells into cancer-associated fibroblasts to support tumorigenesis. SIGNIFICANCE Functional screening of breast cancer secretomes revealed that triple-negative breast cancer promotes fibrosis in the adipose tissue microenvironment by secreting zinc-alpha-2-glycoprotein and promoting the transdifferentiation of adipocyte stem cells into myofibroblasts.
Collapse
Affiliation(s)
- Surbhi Verma
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, Utah.
| | | | - Meghan C. Curtin
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, Utah.
| | - Menusha Arumugam
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, Utah.
| | - Sandra M. Osburn-Staker
- Metabolomics, Proteomics and Mass Spectrometry Core, School of Medicine, University of Utah, Salt Lake City, Utah.
| | - Guoying Wang
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah.
| | - Aaron Atkinson
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah.
| | - David A. Nix
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah.
| | - David H. Lum
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah.
| | - James E. Cox
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, Utah.
- Metabolomics, Proteomics and Mass Spectrometry Core, School of Medicine, University of Utah, Salt Lake City, Utah.
| | - Keren I. Hilgendorf
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, Utah.
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah.
| |
Collapse
|
7
|
Varveri A, Papadopoulou M, Papadovasilakis Z, Compeer EB, Legaki AI, Delis A, Damaskou V, Boon L, Papadogiorgaki S, Samiotaki M, Foukas PG, Eliopoulos AG, Hatzioannou A, Alissafi T, Dustin ML, Verginis P. Immunological synapse formation between T regulatory cells and cancer-associated fibroblasts promotes tumour development. Nat Commun 2024; 15:4988. [PMID: 38862534 PMCID: PMC11167033 DOI: 10.1038/s41467-024-49282-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/23/2024] [Indexed: 06/13/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs) have emerged as a dominant non-hematopoietic cell population in the tumour microenvironment, serving diverse functions in tumour progression. However, the mechanisms via which CAFs influence the anti-tumour immunity remain poorly understood. Here, using multiple tumour models and biopsies from cancer patients, we report that α-SMA+ CAFs can form immunological synapses with Foxp3+ regulatory T cells (Tregs) in tumours. Notably, α-SMA+ CAFs can phagocytose and process tumour antigens and exhibit a tolerogenic phenotype which instructs movement arrest, activation and proliferation in Tregs in an antigen-specific manner. Moreover, α-SMA+ CAFs display double-membrane structures resembling autophagosomes in their cytoplasm. Single-cell transcriptomic data showed an enrichment in autophagy and antigen processing/presentation pathways in α-SMA-expressing CAF clusters. Conditional knockout of Atg5 in α-SMA+ CAFs promoted inflammatory re-programming in CAFs, reduced Treg cell infiltration and attenuated tumour development. Overall, our findings reveal an immunosuppressive mechanism entailing the formation of synapses between α-SMA+ CAFs and Tregs in an autophagy-dependent manner.
Collapse
Affiliation(s)
- Athina Varveri
- Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
- Laboratory of Immune Regulation and Tolerance, Division of Basic Sciences, Medical School, University of Crete, Heraklion, Greece
| | - Miranta Papadopoulou
- Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
- Laboratory of Immune Regulation and Tolerance, Division of Basic Sciences, Medical School, University of Crete, Heraklion, Greece
| | - Zacharias Papadovasilakis
- Laboratory of Immune Regulation and Tolerance, Division of Basic Sciences, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Heraklion, Greece
| | - Ewoud B Compeer
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Aigli-Ioanna Legaki
- Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Anastasios Delis
- Center of Basic Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Vasileia Damaskou
- 2nd Department of Pathology, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | | | | | - Martina Samiotaki
- Institute for Bioinnovation, Biomedical Sciences Research Centre Alexander Fleming, Vari, Athens, 166 72, Greece
| | - Periklis G Foukas
- 2nd Department of Pathology, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - Aristides G Eliopoulos
- Laboratory of Biology, School of Medicine, Medical School National and Kapodistrian University of Athens, Athens, Greece
| | - Aikaterini Hatzioannou
- Laboratory of Biology, School of Medicine, Medical School National and Kapodistrian University of Athens, Athens, Greece
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden, Dresden, Germany
| | - Themis Alissafi
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
- Laboratory of Biology, School of Medicine, Medical School National and Kapodistrian University of Athens, Athens, Greece
| | - Michael L Dustin
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Panayotis Verginis
- Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece.
- Laboratory of Immune Regulation and Tolerance, Division of Basic Sciences, Medical School, University of Crete, Heraklion, Greece.
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Heraklion, Greece.
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden, Dresden, Germany.
| |
Collapse
|
8
|
Wang Y, Li X, Gang Q, Huang Y, Liu M, Zhang H, Shen S, Qi Y, Zhang J. Pathomics and single-cell analysis of papillary thyroid carcinoma reveal the pro-metastatic influence of cancer-associated fibroblasts. BMC Cancer 2024; 24:710. [PMID: 38858612 PMCID: PMC11163752 DOI: 10.1186/s12885-024-12459-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/31/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND Papillary thyroid carcinoma (PTC) is globally prevalent and associated with an increased risk of lymph node metastasis (LNM). The role of cancer-associated fibroblasts (CAFs) in PTC remains unclear. METHODS We collected postoperative pathological hematoxylin-eosin (HE) slides from 984 included patients with PTC to analyze the density of CAF infiltration at the invasive front of the tumor using QuPath software. The relationship between CAF density and LNM was assessed. Single-cell RNA sequencing (scRNA-seq) data from GSE193581 and GSE184362 datasets were integrated to analyze CAF infiltration in PTC. A comprehensive suite of in vitro experiments, encompassing EdU labeling, wound scratch assays, Transwell assays, and flow cytometry, were conducted to elucidate the regulatory role of CD36+CAF in two PTC cell lines, TPC1 and K1. RESULTS A significant correlation was observed between high fibrosis density at the invasive front of the tumor and LNM. Analysis of scRNA-seq data revealed metastasis-associated myoCAFs with robust intercellular interactions. A diagnostic model based on metastasis-associated myoCAF genes was established and refined through deep learning methods. CD36 positive expression in CAFs can significantly promote the proliferation, migration, and invasion abilities of PTC cells, while inhibiting the apoptosis of PTC cells. CONCLUSION This study addresses the significant issue of LNM risk in PTC. Analysis of postoperative HE pathological slides from a substantial patient cohort reveals a notable association between high fibrosis density at the invasive front of the tumor and LNM. Integration of scRNA-seq data comprehensively analyzes CAF infiltration in PTC, identifying metastasis-associated myoCAFs with strong intercellular interactions. In vitro experimental results indicate that CD36 positive expression in CAFs plays a promoting role in the progression of PTC. Overall, these findings provide crucial insights into the function of CAF subset in PTC metastasis.
Collapse
Affiliation(s)
- Yixian Wang
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, Liaoning, 110001, China
| | - Xin Li
- Department of Head and Neck Surgery, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, 110042, China
| | - Qingwei Gang
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, Liaoning, 110001, China
| | - Yinde Huang
- Department of Breast and Thyroid Surgery, Chongqing General Hospital, Chongqing, 401147, China
| | - Mingyu Liu
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, Liaoning, 110001, China
| | - Han Zhang
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, Liaoning, 110001, China
| | - Shikai Shen
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, Liaoning, 110001, China
| | - Yao Qi
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, Liaoning, 110001, China
| | - Jian Zhang
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, Liaoning, 110001, China.
| |
Collapse
|
9
|
Sánchez-Ramírez D, Mendoza-Rodríguez MG, Alemán OR, Candanedo-González FA, Rodríguez-Sosa M, Montesinos-Montesinos JJ, Salcedo M, Brito-Toledo I, Vaca-Paniagua F, Terrazas LI. Impact of STAT-signaling pathway on cancer-associated fibroblasts in colorectal cancer and its role in immunosuppression. World J Gastrointest Oncol 2024; 16:1705-1724. [PMID: 38764833 PMCID: PMC11099434 DOI: 10.4251/wjgo.v16.i5.1705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/28/2024] [Accepted: 04/01/2024] [Indexed: 05/09/2024] Open
Abstract
Colorectal cancer (CRC) remains one of the most commonly diagnosed and deadliest types of cancer worldwide. CRC displays a desmoplastic reaction (DR) that has been inversely associated with poor prognosis; less DR is associated with a better prognosis. This reaction generates excessive connective tissue, in which cancer-associated fibroblasts (CAFs) are critical cells that form a part of the tumor microenvironment. CAFs are directly involved in tumorigenesis through different mechanisms. However, their role in immunosuppression in CRC is not well understood, and the precise role of signal transducers and activators of transcription (STATs) in mediating CAF activity in CRC remains unclear. Among the myriad chemical and biological factors that affect CAFs, different cytokines mediate their function by activating STAT signaling pathways. Thus, the harmful effects of CAFs in favoring tumor growth and invasion may be modulated using STAT inhibitors. Here, we analyze the impact of different STATs on CAF activity and their immunoregulatory role.
Collapse
Affiliation(s)
- Damián Sánchez-Ramírez
- Unidad de Investigacion en Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autonoma de Mexico, Tlalnepantla 54090, Estado de Mexico, Mexico
| | - Mónica G Mendoza-Rodríguez
- Unidad de Investigacion en Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autonoma de Mexico, Tlalnepantla 54090, Estado de Mexico, Mexico
| | - Omar R Alemán
- Department of Biology, Facultad de Quimica, Universidad Nacional Autonoma de Mexico, Ciudad Universitaria, Mexico City 04510, Mexico
| | - Fernando A Candanedo-González
- Department of Pathology, National Medical Center Century XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| | - Miriam Rodríguez-Sosa
- Unidad de Investigacion en Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autonoma de Mexico, Tlalnepantla 54090, Estado de Mexico, Mexico
| | - Juan José Montesinos-Montesinos
- Laboratorio de Células Troncales Mesenquimales, Unidad de Investigación Médica en Enfermedades Oncológicas, Hospital de Oncología Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| | - Mauricio Salcedo
- Unidad de Investigacion en Biomedicina y Oncologia Genomica, Instituto Mexciano del Seguro Social, Mexico City 07300, Mexico
| | - Ismael Brito-Toledo
- Servicio de Colon y Recto, Hospital de Oncología Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| | - Felipe Vaca-Paniagua
- Unidad de Investigacion en Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autonoma de Mexico, Tlalnepantla 54090, Estado de Mexico, Mexico
- Laboratorio Nacional en Salud, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Estado de Mexico, Mexico
| | - Luis I Terrazas
- Unidad de Investigacion en Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autonoma de Mexico, Tlalnepantla 54090, Estado de Mexico, Mexico
- Laboratorio Nacional en Salud, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Estado de Mexico, Mexico
| |
Collapse
|
10
|
Takahashi K, Shoda K, Takiguchi K, Higuchi Y, Matsuoka K, Nakayama T, Saito R, Maruyama S, Nakata Y, Furuya S, Shiraishi K, Akaike H, Kawaguchi Y, Amemiya H, Kawaida H, Ichikawa D. Prognostic Impact of Stromal Profiles Educated by Gastric Cancer. Ann Surg Oncol 2024; 31:2309-2318. [PMID: 37919449 DOI: 10.1245/s10434-023-14522-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 10/16/2023] [Indexed: 11/04/2023]
Abstract
BACKGROUND Cancer-associated fibroblasts exhibit diversity and have several subtypes. The underlying relationship between the diversity of cancer-associated fibroblasts and their effect on gastric cancer progression remains unclear. In this study, mesenchymal stem cells were differentiated into cancer-associated fibroblasts with gastric cancer cell lines; clinical specimens were used to further investigate the impact of cancer-associated fibroblast diversity on cancer progression. METHODS Nine gastric cancer cell lines (NUGC3, NUGC4, MKN7, MKN45, MKN74, FU97, OCUM1, NCI-N87, and KATOIII) were used to induce mesenchymal stem cell differentiation into cancer-associated fibroblasts. The cancer-associated fibroblasts were classified based on ACTA2 and PDPN expression. Cell function analysis was used to examine the impact of cancer-associated fibroblast subtypes on cancer cell phenotype. Tissue samples from 97gastric patients who underwent gastrectomy were used to examine the clinical significance of each subtype classified according to cancer-associated fibroblast expression. RESULTS Co-culture of mesenchymal stem cells with nine gastric cancer cell lines revealed different subtypes of ACTA2 and PDPN expression in differentiated cancer-associated fibroblasts. Cancer-associated fibroblast subtypes with high ACTA2 plus PDPN expression levels significantly increased gastric cancer cell migration, invasion, and proliferation. The cancer-associated fibroblast subtype with ACTA2 plus PDPN expression was an independent prognostic factor along with lymph node metastasis for patients who had gastric cancer and were undergoing surgery. CONCLUSIONS Cancer-associated fibroblasts are educated by gastric cancer cells during the development of cancer-associated fibroblast diversity. Differentiated cancer-associated fibroblasts with distinct expression patterns could affect gastric cancer progression and enable prognostic stratification for gastric cancer.
Collapse
Affiliation(s)
- Kazunori Takahashi
- First Department of Surgery, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Katsutoshi Shoda
- First Department of Surgery, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan.
| | - Koichi Takiguchi
- First Department of Surgery, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Yudai Higuchi
- First Department of Surgery, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Koichi Matsuoka
- First Department of Surgery, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Takashi Nakayama
- First Department of Surgery, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Ryo Saito
- First Department of Surgery, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Suguru Maruyama
- First Department of Surgery, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Yuki Nakata
- First Department of Surgery, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Shinji Furuya
- First Department of Surgery, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Kensuke Shiraishi
- First Department of Surgery, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Hidenori Akaike
- First Department of Surgery, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Yoshihiko Kawaguchi
- First Department of Surgery, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Hidetake Amemiya
- First Department of Surgery, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Hiromichi Kawaida
- First Department of Surgery, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Daisuke Ichikawa
- First Department of Surgery, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| |
Collapse
|
11
|
Lyu SI, Johannsen J, Simon AG, Knipper K, Wuerdemann N, Sharma SJ, Thelen M, Hansen KK, Fretter C, Klasen C, Esser J, Suchan MC, Abing H, Zimmermann PH, Schultheis AM, Schloesser HA, Klussmann JP, Quaas A, Eckel HNC. Co-expression patterns of cancer associated fibroblast markers reveal distinct subgroups related to patient survival in oropharyngeal squamous cell carcinoma. Front Cell Dev Biol 2024; 12:1337361. [PMID: 38328551 PMCID: PMC10847231 DOI: 10.3389/fcell.2024.1337361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/05/2024] [Indexed: 02/09/2024] Open
Abstract
Background: The incidence of oropharyngeal squamous cell carcinoma (OPSCC) is rapidly increasing in high income countries due to its association with persistent high-risk human papilloma virus (HPV) infection. Recent scientific advances have highlighted the importance of the tumor microenvironment in OPSCC. In this study, including 216 OPSCC patients, we analyze the composition of four established markers of cancer associated fibroblasts (CAFs) in the context of intratumoral CD8 T-cell infiltration. Methods: Immunohistochemical staining for fibroblast activation protein (FAP), platelet-derived growth factor receptor beta (PDGFRb), periostin, alpha smooth muscle actin (α-SMA) and CD8 were analyzed digitally and their association with survival, tumor- and patient characteristics was assessed. Results: Co-expression of CAF markers was frequent but not associated with HPV status. FAPhigh and PDGFRbhigh expression were associated with increased CD8 T-cell infiltration. Low expression of PDGFRb improved patient survival in female patients but not in male patients. We identified PDGFRblow periostinlow α-SMAlow status as an independent predictor of improved survival (hazard ratio 0.377, p = 0.006). Conclusion: These findings elucidate the co-expression of four established CAF markers in OPSCC and underscore their association with T-cell infiltration and patient survival. Future analyses of CAF subgroups in OPSCC may enable the development of individualized therapies.
Collapse
Affiliation(s)
- Su Ir Lyu
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, Cologne, Germany
| | - Jannik Johannsen
- Faculty of Medicine and University Hospital of Cologne, Department of Otorhinolaryngology, Head and Neck Surgery, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Adrian Georg Simon
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, Cologne, Germany
| | - Karl Knipper
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Nora Wuerdemann
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Shachi Jenny Sharma
- Faculty of Medicine and University Hospital of Cologne, Department of Otorhinolaryngology, Head and Neck Surgery, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Martin Thelen
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Kevin Karl Hansen
- Faculty of Medicine and University Hospital of Cologne, Department of Otorhinolaryngology, Head and Neck Surgery, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Caroline Fretter
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, Cologne, Germany
| | - Charlotte Klasen
- Faculty of Medicine and University Hospital of Cologne, Department of Otorhinolaryngology, Head and Neck Surgery, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Julia Esser
- Faculty of Medicine and University Hospital of Cologne, Department of Otorhinolaryngology, Head and Neck Surgery, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Malte Christian Suchan
- Faculty of Medicine and University Hospital of Cologne, Department of Otorhinolaryngology, Head and Neck Surgery, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Helen Abing
- Faculty of Medicine and University Hospital of Cologne, Department of Otorhinolaryngology, Head and Neck Surgery, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Philipp Heinrich Zimmermann
- Faculty of Medicine and University Hospital of Cologne, Department of Otorhinolaryngology, Head and Neck Surgery, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Anne Maria Schultheis
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, Cologne, Germany
| | - Hans Anton Schloesser
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Jens Peter Klussmann
- Faculty of Medicine and University Hospital of Cologne, Department of Otorhinolaryngology, Head and Neck Surgery, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Alexander Quaas
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, Cologne, Germany
| | - Hans Nikolaus Caspar Eckel
- Faculty of Medicine and University Hospital of Cologne, Department of Otorhinolaryngology, Head and Neck Surgery, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
12
|
Domínguez-Cejudo MA, Gil-Torralvo A, Cejuela M, Molina-Pinelo S, Salvador Bofill J. Targeting the Tumor Microenvironment in Breast Cancer: Prognostic and Predictive Significance and Therapeutic Opportunities. Int J Mol Sci 2023; 24:16771. [PMID: 38069096 PMCID: PMC10706312 DOI: 10.3390/ijms242316771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
Breast cancer is one of the most prevalent tumors among women. Its prognosis and treatment outcomes depend on factors related to tumor cell biology. However, recent studies have revealed the critical role of the tumor microenvironment (TME) in the development, progression, and treatment response of breast cancer. In this review, we explore the different components of the TME and their relevance as prognostic and predictive biomarkers in breast cancer. In addition, techniques for assessing the tumor microenvironment, such as immunohistochemistry or gene expression profiling, and their clinical utility in therapeutic decision-making are examined. Finally, therapeutic strategies targeting the TME are reviewed, highlighting their potential clinical benefits. Overall, this review emphasizes the importance of the TME in breast cancer and its potential as a clinical tool for better patient stratification and the design of personalized therapies.
Collapse
Affiliation(s)
- María A. Domínguez-Cejudo
- Institute of Biomedicine of Seville (IBiS), HUVR, CSIC, Universidad de Sevilla, 41013 Seville, Spain (S.M.-P.)
- Andalusian—Roche Network Mixed Alliance in Precision Medical Oncology, 41092 Sevilla, Spain
| | - Ana Gil-Torralvo
- Institute of Biomedicine of Seville (IBiS), HUVR, CSIC, Universidad de Sevilla, 41013 Seville, Spain (S.M.-P.)
- Andalusian—Roche Network Mixed Alliance in Precision Medical Oncology, 41092 Sevilla, Spain
- Medical Oncology Department, Virgen del Rocio Hospital, 41013 Seville, Spain
| | - Mónica Cejuela
- Medical Oncology Department, Virgen del Rocio Hospital, 41013 Seville, Spain
| | - Sonia Molina-Pinelo
- Institute of Biomedicine of Seville (IBiS), HUVR, CSIC, Universidad de Sevilla, 41013 Seville, Spain (S.M.-P.)
- Andalusian—Roche Network Mixed Alliance in Precision Medical Oncology, 41092 Sevilla, Spain
| | - Javier Salvador Bofill
- Institute of Biomedicine of Seville (IBiS), HUVR, CSIC, Universidad de Sevilla, 41013 Seville, Spain (S.M.-P.)
- Andalusian—Roche Network Mixed Alliance in Precision Medical Oncology, 41092 Sevilla, Spain
- Medical Oncology Department, Virgen del Rocio Hospital, 41013 Seville, Spain
| |
Collapse
|
13
|
Liu Z, Hou P, Fang J, Zhu J, Zha J, Liu R, Ding Y, Zuo M, Li P, Cao L, Feng C, Melino G, Shao C, Shi Y. Mesenchymal stromal cells confer breast cancer doxorubicin resistance by producing hyaluronan. Oncogene 2023; 42:3221-3235. [PMID: 37704784 DOI: 10.1038/s41388-023-02837-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/31/2023] [Accepted: 09/05/2023] [Indexed: 09/15/2023]
Abstract
Chemotherapy resistance represents a major cause of therapeutic failure and mortality in cancer patients. Mesenchymal stromal cells (MSCs), an integral component of tumor microenvironment, are known to promote drug resistance. However, the detailed mechanisms remain to be elucidated. Here, we found that MSCs confer breast cancer resistance to doxorubicin by diminishing its intratumoral accumulation. Hyaluronan (HA), a major extracellular matrix (ECM) product of MSCs, was found to mediate the chemoresistant effect. The chemoresistant effect of MSCs was abrogated when hyaluronic acid synthase 2 (HAS2) was depleted or inhibited. Exogenous HA also protected tumor grafts from doxorubicin. Molecular dynamics simulation analysis indicates that HA can bind with doxorubicin, mainly via hydrophobic and hydrogen bonds, and thus reduce its entry into breast cancer cells. This mechanism is distinct from the reported chemoresistant effect of HA via its receptor on cell surface. High HA serum levels were also found to be positively associated with chemoresistance in breast cancer patients. Our findings indicate that the HA-doxorubicin binding dynamics can confer cancer cells chemoresistance. Reducing HA may enhance chemotherapy efficacy.
Collapse
Affiliation(s)
- Zhanhong Liu
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Department of Experimental Medicine and Biochemical Sciences, University of Rome Tor Vergata, Rome, Italy
| | - Pengbo Hou
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Department of Experimental Medicine and Biochemical Sciences, University of Rome Tor Vergata, Rome, Italy
| | - Jiankai Fang
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Jingyu Zhu
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu, China
| | - Juanmin Zha
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Rui Liu
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Department of Experimental Medicine and Biochemical Sciences, University of Rome Tor Vergata, Rome, Italy
| | - Yayun Ding
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Muqiu Zuo
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Peishan Li
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Lijuan Cao
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Chao Feng
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Gerry Melino
- Department of Experimental Medicine and Biochemical Sciences, University of Rome Tor Vergata, Rome, Italy
| | - Changshun Shao
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China.
| | - Yufang Shi
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
14
|
Kok SY, Nakayama M, Morita A, Oshima H, Oshima M. Genetic and nongenetic mechanisms for colorectal cancer evolution. Cancer Sci 2023; 114:3478-3486. [PMID: 37357016 PMCID: PMC10475778 DOI: 10.1111/cas.15891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 06/10/2023] [Indexed: 06/27/2023] Open
Abstract
The stepwise accumulation of key driver mutations is responsible for the development and malignant progression of colorectal cancer in primary sites. Genetic mouse model studies have revealed combinations of driver gene mutations that induce phenotypic changes in tumors toward malignancy. However, cancer evolution is regulated by not only genetic alterations but also nongenetic mechanisms. For example, certain populations of metastatic cancer cells show a loss of malignant characteristics even after the accumulation of driver mutations, and such cells are eliminated in a negative selection manner. Furthermore, a polyclonal metastasis model has recently been proposed, in which cell clusters consisting of genetically heterogeneous cells break off from the primary site, disseminate to distant organs, and develop into heterogenous metastatic tumors. Such nongenetic mechanisms for malignant progression have been elucidated using genetically engineered mouse models as well as organoid transplantation experiments. In this review article, we discuss the role of genetic alterations in the malignant progression of primary intestinal tumors and nongenetic mechanisms for negative selection and polyclonal metastasis, which we learned from model studies.
Collapse
Affiliation(s)
- Sau Yee Kok
- Division of GeneticsCancer Research Institute, Kanazawa UniversityKanazawaJapan
| | - Mizuho Nakayama
- Division of GeneticsCancer Research Institute, Kanazawa UniversityKanazawaJapan
- WPI Nano Life Science Institute (NanoLSI), Kanazawa UniversityKanazawaJapan
| | - Atsuya Morita
- Division of GeneticsCancer Research Institute, Kanazawa UniversityKanazawaJapan
| | - Hiroko Oshima
- Division of GeneticsCancer Research Institute, Kanazawa UniversityKanazawaJapan
- WPI Nano Life Science Institute (NanoLSI), Kanazawa UniversityKanazawaJapan
| | - Masanobu Oshima
- Division of GeneticsCancer Research Institute, Kanazawa UniversityKanazawaJapan
- WPI Nano Life Science Institute (NanoLSI), Kanazawa UniversityKanazawaJapan
| |
Collapse
|
15
|
Liu Y, Ji Y, Jiang R, Fang C, Shi G, Cheng L, Zuo Y, Ye Y, Su X, Li J, Wang H, Wang Y, Lin Y, Dai L, Zhang S, Deng H. Reduced smooth muscle-fibroblasts transformation potentially decreases intestinal wound healing and colitis-associated cancer in ageing mice. Signal Transduct Target Ther 2023; 8:294. [PMID: 37553378 PMCID: PMC10409725 DOI: 10.1038/s41392-023-01554-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 05/24/2023] [Accepted: 06/24/2023] [Indexed: 08/10/2023] Open
Abstract
Cancer and impaired tissue wound healing with ageing are closely related to the quality of life of the elderly population. Given the increased incidence of cancer and the population ageing trend globally, it is very important to explore how ageing impairs tissue wound healing and spontaneous cancer. In a murine model of DSS-induced acute colitis and AOM/DSS-induced colitis-associated cancer (CAC), we found ageing significantly decreases intestinal wound healing and simultaneous CAC initiation, although ageing does not affect the incidence of AOM-induced, sporadic non-inflammatory CRC. Mechanistically, reduced fibroblasts were observed in the colitis microenvironment of ageing mice. Through conditional lineage tracing, an important source of fibroblasts potentially derived from intestinal smooth muscle cells (ISMCs) was identified orchestrating intestinal wound healing and CAC initiation in young mice. However, the number of transformed fibroblasts from ISMCs significantly decreased in ageing mice, accompanied by decreased intestinal wound healing and decreased CAC initiation. ISMCs-fibroblasts transformation in young mice and reduction of this transformation in ageing mice were also confirmed by ex-vivo intestinal muscular layer culture experiments. We further found that activation of YAP/TAZ in ISMCs is required for the transformation of ISMCs into fibroblasts. Meanwhile, the reduction of YAP/TAZ activation in ISMCs during intestinal wound healing was observed in ageing mice. Conditional knockdown of YAP/TAZ in ISMCs of young mice results in reduced fibroblasts in the colitis microenvironment, decreased intestinal wound healing and decreased CAC initiation, similar to the phenotype of ageing mice. In addition, the data from intestine samples derived from inflammatory bowel disease (IBD) patients show that activation of YAP/TAZ also occurs in ISMCs from these patients. Collectively, our work reveals an important role of the ageing stromal microenvironment in intestinal wound healing and CAC initiation. Furthermore, our work also identified a potential source of fibroblasts involved in colitis and CAC.
Collapse
Affiliation(s)
- Yi Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, The People's Republic of China
| | - Yanhong Ji
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, The People's Republic of China
| | - Ruiyi Jiang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, The People's Republic of China
| | - Chao Fang
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, 610041, Chengdu, Sichuan, The People's Republic of China
| | - Gang Shi
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, The People's Republic of China
| | - Lin Cheng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, The People's Republic of China
| | - Yinan Zuo
- Respiratory Microbiome Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, The People's Republic of China
| | - Yixin Ye
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, The People's Republic of China
| | - Xiaolan Su
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, The People's Republic of China
| | - Junshu Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, The People's Republic of China
| | - Huiling Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, The People's Republic of China
| | - Yuan Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, The People's Republic of China
| | - Yi Lin
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, The People's Republic of China
| | - Lei Dai
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, The People's Republic of China
| | - Shuang Zhang
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, Sichuan, The People's Republic of China
| | - Hongxin Deng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, The People's Republic of China.
| |
Collapse
|
16
|
Huang J, Tsang WY, Li ZH, Guan XY. The Origin, Differentiation, and Functions of Cancer-Associated Fibroblasts in Gastrointestinal Cancer. Cell Mol Gastroenterol Hepatol 2023; 16:503-511. [PMID: 37451403 PMCID: PMC10462789 DOI: 10.1016/j.jcmgh.2023.07.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 07/06/2023] [Accepted: 07/06/2023] [Indexed: 07/18/2023]
Abstract
Emerging evidence has shown the importance of the tumor microenvironment in tumorigenesis and progression. Cancer-associated fibroblasts (CAFs) are one of the most infiltrated stroma cells of the tumor microenvironment in gastrointestinal tumors. CAFs play crucial roles in tumor development and therapeutic response by biologically secreting soluble factors or structurally remodeling the extracellular matrix. Conceivably, CAFs may become excellent targets for tumor prevention and treatment. However, the limited knowledge of the heterogeneity of CAFs represents a huge challenge for clinically targeting CAFs. In this review, we summarize the newest understanding of gastrointestinal CAFs, with a special focus on their origin, differentiation, and function. We also discuss the current understanding of CAF subpopulations as shown by single-cell technologies.
Collapse
Affiliation(s)
- Jiao Huang
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; State Key Laboratory for Liver Research, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Wai-Ying Tsang
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Zhi-Hong Li
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xin-Yuan Guan
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; State Key Laboratory for Liver Research, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Department of Clinical Oncology, The University of Hong Kong, Shenzhen Hospital, Shenzhen, China; Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, China.
| |
Collapse
|
17
|
Miyata-Morita K, Morita S, Dejima H, Saito K, Sakao Y, Mochizuki M, Sasajima Y. Cytological features of stromal spindle cells and their prognostic significance in lung adenocarcinoma. Cytopathology 2023; 34:337-345. [PMID: 36946097 DOI: 10.1111/cyt.13232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 12/21/2022] [Accepted: 03/06/2023] [Indexed: 03/23/2023]
Abstract
INTRODUCTION Cancer-associated fibroblasts (CAFs) in the tumour microenvironment play a key role in tumour development, proliferation, invasion, and metastasis. The cytological features of spindle cells including CAFs-defined as stromal spindle cells (SSCs) adjacent to cancer cells-are frequently encountered in pulmonary adenocarcinomas. This study aimed to investigate the association between the presence of SSCs in cytological specimens and the clinicopathological features. METHODS We evaluated 211 patients with pulmonary adenocarcinoma who underwent surgical resection. All participants had cytological specimens corresponding to the histological specimens available for review. RESULTS Of the 211 cases examined, 89 were SSC-positive (SSC+ ) and 122 were SSC-negative (SSC- ). SSC+ cases were more frequently associated with higher pathological stage (P < 0.001), lymph node metastasis (P = 0.002), anaplastic lymphoma kinase (ALK) gene rearrangement (P = 0.04), high tumour grade (P < 0.001), solid and micropapillary predominant pattern (P = 0.02), and lymphatic vessel (P = 0.003), blood vessel (P < 0.001), and pleural invasion (P = 0.03) as compared to SSC- cases. Patients with SSC+ adenocarcinoma had a significantly shorter recurrence-free survival than those with SSC- adenocarcinoma (P = 0.009). Cytologically, necrotic background (P = 0.002), mucinous cancer cells (P = 0.02), pleomorphic cells (P < 0.001), and mutual cell inclusions (P = 0.01) were observed more frequently in SSC+ adenocarcinomas. CONCLUSIONS The presence of SSCs could be an important cytological feature for predicting poor prognosis in lung adenocarcinomas.
Collapse
Affiliation(s)
- Kana Miyata-Morita
- Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Tokyo, Japan
| | - Shigeki Morita
- Department of Pathology, Mitsui Memorial Hospital, Tokyo, Japan
- Department of Pathology, Teikyo University Hospital, Tokyo, Japan
| | - Hitoshi Dejima
- Department of Surgery, Teikyo University School of Medicine, Tokyo, Japan
| | - Koji Saito
- Department of Pathology, Teikyo University Hospital, Tokyo, Japan
| | - Yukinori Sakao
- Department of Surgery, Teikyo University School of Medicine, Tokyo, Japan
| | - Makoto Mochizuki
- Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Tokyo, Japan
| | - Yuko Sasajima
- Department of Pathology, Teikyo University Hospital, Tokyo, Japan
| |
Collapse
|
18
|
Dzobo K, Dandara C. The Extracellular Matrix: Its Composition, Function, Remodeling, and Role in Tumorigenesis. Biomimetics (Basel) 2023; 8:146. [PMID: 37092398 PMCID: PMC10123695 DOI: 10.3390/biomimetics8020146] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/31/2023] [Accepted: 04/03/2023] [Indexed: 04/25/2023] Open
Abstract
The extracellular matrix (ECM) is a ubiquitous member of the body and is key to the maintenance of tissue and organ integrity. Initially thought to be a bystander in many cellular processes, the extracellular matrix has been shown to have diverse components that regulate and activate many cellular processes and ultimately influence cell phenotype. Importantly, the ECM's composition, architecture, and stiffness/elasticity influence cellular phenotypes. Under normal conditions and during development, the synthesized ECM constantly undergoes degradation and remodeling processes via the action of matrix proteases that maintain tissue homeostasis. In many pathological conditions including fibrosis and cancer, ECM synthesis, remodeling, and degradation is dysregulated, causing its integrity to be altered. Both physical and chemical cues from the ECM are sensed via receptors including integrins and play key roles in driving cellular proliferation and differentiation and in the progression of various diseases such as cancers. Advances in 'omics' technologies have seen an increase in studies focusing on bidirectional cell-matrix interactions, and here, we highlight the emerging knowledge on the role played by the ECM during normal development and in pathological conditions. This review summarizes current ECM-targeted therapies that can modify ECM tumors to overcome drug resistance and better cancer treatment.
Collapse
Affiliation(s)
- Kevin Dzobo
- Medical Research Council, SA Wound Healing Unit, Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Groote Schuur Hospital, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa
| | - Collet Dandara
- Division of Human Genetics and Institute of Infectious Disease and Molecular Medicine, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa
- The South African Medical Research Council-UCT Platform for Pharmacogenomics Research and Translation, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa
| |
Collapse
|
19
|
Ahuja S, Zaheer S, Ranga S. Histomorphological Evaluation of Desmoplastic Tumor Stroma in Malignant Ovarian Surface Epithelial Tumors. J Midlife Health 2023; 14:107-111. [PMID: 38029034 PMCID: PMC10664060 DOI: 10.4103/jmh.jmh_31_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/15/2023] [Indexed: 12/01/2023] Open
Abstract
Background Ovarian cancer is the 8th most common cancer in women worldwide. Tumor budding is defined as a type of invasive growth in carcinomas with either a single tumor cell or a cluster of up to four cells at the invasive tumor front and is associated with epithelial-mesenchymal transition. A reactive stroma rich in cancer-associated fibroblasts is associated with higher tumor grade and poorer prognosis in breast, colorectal, and oral cancers. Aims and Objectives The present study was conducted to highlight the prognostic significance of tumor budding and fibrotic cancer stroma in malignant ovarian surface epithelial tumors with known prognostic parameters. Materials and Methods This was a retrospective cross-sectional study conducted over a 2-year period, in which all histologically diagnosed cases of malignant ovarian surface epithelial tumors who underwent surgery were included. The fibrotic stroma was classified into three distinct categories - mature, intermediate, and immature. The number of tumor buds was counted at the invasive front of the tumor and graded based on the number of buds - 0-5, 5-9, and ≥10 buds. Results Among the 50 cases, 32% (16 cases) had mature stroma, whereas 30% (15 cases) and 38% (19 cases) had intermediate and immature stroma, respectively. Although a significant association could not be established between tumor budding and stroma grade, a fair agreement was established between them. A significant association could be established between histological grade with both tumor budding (P = 0.03) and fibrotic stroma grade (P = 0.02). Conclusion The study highlighted the role of stromal response in malignant surface epithelial tumors of the ovary since a higher-grade tumor was associated with an immature stroma, whereas a lower-grade tumor was associated with a mature stroma.
Collapse
Affiliation(s)
- Sana Ahuja
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Sunil Ranga
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
20
|
Knipper K, Damanakis AI, Zhao Y, Bruns CJ, Schmidt T, Popp FC, Quaas A, Lyu SI. Specific Subtypes of Carcinoma-Associated Fibroblasts Are Correlated with Worse Survival in Resectable Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2023; 15:cancers15072049. [PMID: 37046710 PMCID: PMC10093167 DOI: 10.3390/cancers15072049] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/17/2023] [Accepted: 03/28/2023] [Indexed: 04/14/2023] Open
Abstract
PURPOSE The pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancer entities. Effective therapy options are still lacking. The tumor microenvironment possibly bears further treatment possibilities. This study aimed to describe the expression patterns of four established carcinoma-associated fibroblast (CAFs) markers and their correlation in PDAC tissue samples. METHODS This project included 321 patients with PDAC who underwent surgery with a curative intent in one of the PANCALYZE study centers. Immunohistochemical stainings for FAP, PDGFR, periostin, and SMA were performed. The expression patterns of each marker were divided into low- and high-expressing CAFs and correlated with patients' survival. RESULTS Tumors showing SMAhigh-, PeriostinhighSMAhigh-, or PeriostinhighSMAlowPDGFRlowFAPhigh-positive CAFs demonstrated significantly worse survival. Additionally, a high expression of SMA in PDAC tissue samples was shown to be an independent risk factor for worse survival. CONCLUSION This project identified three subgroups of PDAC with different expression patterns of CAF markers which showed significantly worse survival. This could be the base for the further characterization of the fibroblast subgroups in PDAC and contribute to the development of new targeted therapy options against CAFs.
Collapse
Affiliation(s)
- Karl Knipper
- Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, Faculty of Medicine, University of Cologne, 50937 Cologne, Germany
| | - Alexander I Damanakis
- Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, Faculty of Medicine, University of Cologne, 50937 Cologne, Germany
| | - Yue Zhao
- Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, Faculty of Medicine, University of Cologne, 50937 Cologne, Germany
| | - Christiane J Bruns
- Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, Faculty of Medicine, University of Cologne, 50937 Cologne, Germany
| | - Thomas Schmidt
- Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, Faculty of Medicine, University of Cologne, 50937 Cologne, Germany
| | - Felix C Popp
- Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, Faculty of Medicine, University of Cologne, 50937 Cologne, Germany
| | - Alexander Quaas
- Institute of Pathology, University Hospital of Cologne, Faculty of Medicine, University of Cologne, 50937 Cologne, Germany
| | - Su Ir Lyu
- Institute of Pathology, University Hospital of Cologne, Faculty of Medicine, University of Cologne, 50937 Cologne, Germany
| |
Collapse
|
21
|
Hu J, Li J, Dai C, Ren J, Yang W, He C, Meng F, Dai C, Zeng S. HES1 deficiency impairs development of human intestinal mesenchyme by suppressing WNT5A expression. Biochem Biophys Res Commun 2023; 655:50-58. [PMID: 36933307 DOI: 10.1016/j.bbrc.2023.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 02/14/2023] [Accepted: 03/07/2023] [Indexed: 03/09/2023]
Abstract
Serious intestinal side-effects that target the NOTCH-HES1 pathway in human cancer differentiation therapy make it necessary to understand the pathway at the human organ level. Herein, we endogenously introduced HES1-/- mutations into human embryonic stem cells (hESCs) and differentiated them into human intestinal organoids (HIO). The HES1-/- hESCs retained ES cell properties and showed gene expression patterns similar to those of wild-type hESCs when they differentiated into definitive endoderm and hindgut. During the formation of the HES1-/- lumen we noted an impaired development of mesenchymal cells in addition to the increased differentiation of secretory epithelium. RNA-Seq revealed that inhibited development of the mesenchymal cells may have been due to a downregulation of WNT5A signaling. Overexpression of HES1 and silencing of WNT5A in the intestinal fibroblast cell line CCD-18Co indicated that HES1 was involved in the activation of WNT5A-induced fibroblast growth and migration, suggesting the likelihood of the Notch pathway in epithelial-mesenchymal crosstalk. Our results facilitated the identification of more precise underlying molecular mechanisms displaying distinct roles in HES1 signaling in stromal and epithelial development in human intestinal mucosa.
Collapse
Affiliation(s)
- Jianmin Hu
- Hunan Guangxiu Hospital, School of Medicine, Hunan Normal University, Changsha, 410001, Hunan, China; Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of Citic-Xiangya, Changsha, 410001, Hunan, China
| | - Jin Li
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, PR China; Hunan International Scientific and Technological Cooperation Base of Development and Carcinogenesis, National Engineering and Research Center of Human Stem Cells, Changsha, 410001, Hunan, China
| | - Can Dai
- Hunan Guangxiu Hospital, School of Medicine, Hunan Normal University, Changsha, 410001, Hunan, China; Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of Citic-Xiangya, Changsha, 410001, Hunan, China
| | - Jinlin Ren
- Hunan Guangxiu Hospital, School of Medicine, Hunan Normal University, Changsha, 410001, Hunan, China; Hunan International Scientific and Technological Cooperation Base of Development and Carcinogenesis, National Engineering and Research Center of Human Stem Cells, Changsha, 410001, Hunan, China
| | - Wenru Yang
- Hunan Guangxiu Hospital, School of Medicine, Hunan Normal University, Changsha, 410001, Hunan, China
| | - Caixia He
- Hunan International Scientific and Technological Cooperation Base of Development and Carcinogenesis, National Engineering and Research Center of Human Stem Cells, Changsha, 410001, Hunan, China
| | - Fei Meng
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of Citic-Xiangya, Changsha, 410001, Hunan, China
| | - Congling Dai
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, PR China; Hunan International Scientific and Technological Cooperation Base of Development and Carcinogenesis, National Engineering and Research Center of Human Stem Cells, Changsha, 410001, Hunan, China
| | - Sicong Zeng
- Hunan Guangxiu Hospital, School of Medicine, Hunan Normal University, Changsha, 410001, Hunan, China; Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, PR China; Hunan International Scientific and Technological Cooperation Base of Development and Carcinogenesis, National Engineering and Research Center of Human Stem Cells, Changsha, 410001, Hunan, China; Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of Citic-Xiangya, Changsha, 410001, Hunan, China.
| |
Collapse
|
22
|
Zhou Z, Guo S, Lai S, Wang T, Du Y, Deng J, Zhang S, Gao G, Zhang J. Integrated single-cell and bulk RNA sequencing analysis identifies a cancer-associated fibroblast-related gene signature for predicting survival and therapy in gastric cancer. BMC Cancer 2023; 23:108. [PMID: 36717783 PMCID: PMC9887891 DOI: 10.1186/s12885-022-10332-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 11/18/2022] [Indexed: 02/01/2023] Open
Abstract
As the dominant component of the tumor microenvironment, cancer-associated fibroblasts (CAFs), play a vital role in tumor progression. An increasing number of studies have confirmed that CAFs are involved in almost every aspect of tumors including tumorigenesis, metabolism, invasion, metastasis and drug resistance, and CAFs provide an attractive therapeutic target. This study aimed to explore the feature genes of CAFs for potential therapeutic targets and reliable prediction of prognosis in patients with gastric cancer (GC). Bioinformatic analysis was utilized to identify the feature genes of CAFs in GC by performing an integrated analysis of single-cell and transcriptome RNA sequencing using R software. Based on these feature genes, a CAF-related gene signature was constructed for prognostic prediction by LASSO. Simultaneously, survival analysis and nomogram were performed to validate the prognostic predictive value of this gene signature, and qRT-PCR and immunohistochemical staining verified the expression of the feature genes of CAFs. In addition, small molecular drugs for gene therapy of CAF-related gene signatures in GC patients were identified using the connectivity map (CMAP) database. A combination of nine CAF-related genes was constructed to characterize the prognosis of GC, and the prognostic potential and differential expression of the gene signature were initially validated. Additionally, three small molecular drugs were deduced to have anticancer properties on GC progression. By integrating single-cell and bulk RNA sequencing analyses, a novel gene signature of CAFs was constructed. The results provide a positive impact on future research and clinical studies involving CAFs for GC.
Collapse
Affiliation(s)
- Zhiyang Zhou
- grid.412604.50000 0004 1758 4073Department of General Surgery, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province China
| | - Sixuan Guo
- grid.260463.50000 0001 2182 8825Nanchang University, Nanchang, Jiangxi Province China
| | - Shuhui Lai
- grid.260463.50000 0001 2182 8825Nanchang University, Nanchang, Jiangxi Province China
| | - Tao Wang
- grid.412604.50000 0004 1758 4073Department of Day Ward, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province China
| | - Yao Du
- grid.412604.50000 0004 1758 4073Department of General Surgery, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province China
| | - Junping Deng
- grid.412604.50000 0004 1758 4073Department of General Surgery, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province China
| | - Shun Zhang
- grid.412604.50000 0004 1758 4073Department of General Surgery, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province China
| | - Ge Gao
- grid.412604.50000 0004 1758 4073Department of General Surgery, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province China
| | - Jiangnan Zhang
- grid.412604.50000 0004 1758 4073Department of General Surgery, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province China
| |
Collapse
|
23
|
Arai J, Otoyama Y, Nozawa H, Kato N, Yoshida H. The immunological role of ADAMs in the field of gastroenterological chronic inflammatory diseases and cancers: a review. Oncogene 2023; 42:549-558. [PMID: 36572816 PMCID: PMC9937921 DOI: 10.1038/s41388-022-02583-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 12/27/2022]
Abstract
Metalloproteinases cleave transmembrane proteins that play critical roles in inflammation and cancers. Metalloproteinases include a disintegrin and metalloprotease (ADAM), which we previously examined using a fluorescence assay system, and described their association with resistance to systemic therapy in cancer patients. There are also many reports on the relation between ADAM expression and the prognosis of patients with gastroenterological chronic inflammatory diseases and cancers. Inhibiting their immunomodulating activity in chronic inflammation restores innate immunity and potentially prevents the development of various cancers. Among the numerous critical immune system-related molecules, we focus on major histocompatibility complex class I polypeptide-related sequence A (MICA), MICB, intracellular adhesion molecule (ICAM)-1, TNF-α, IL-6 receptor (IL-6R), and Notch. This review summarizes our current understanding of the role of ADAMs in gastroenterological diseases with regard to the immune system. Several Food and Drug Administration (FDA)-approved inhibitors of ADAMs have been identified, and potential therapies for targeting ADAMs in the treatment of chronic inflammatory diseases and cancers are discussed. Some ongoing clinical trials for cancers targeting ADAMs are also introduced.
Collapse
Affiliation(s)
- Jun Arai
- Division of Gastroenterology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan.
| | - Yumi Otoyama
- grid.410714.70000 0000 8864 3422Division of Gastroenterology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Hisako Nozawa
- grid.410714.70000 0000 8864 3422Division of Gastroenterology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Naoya Kato
- grid.136304.30000 0004 0370 1101Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hitoshi Yoshida
- grid.410714.70000 0000 8864 3422Division of Gastroenterology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
24
|
Talayero VC, Vicente-Manzanares M. A primer on cancer-associated fibroblast mechanics and immunosuppressive ability. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:17-27. [PMID: 36937319 PMCID: PMC10017186 DOI: 10.37349/etat.2023.00120] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 10/11/2022] [Indexed: 02/25/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs) are a major point of interest in modern oncology. Their interest resides in their ability to favor tumor growth without carrying genetic mutations. From a translational standpoint, they are potential therapeutic targets, particularly for hard-to-treat solid cancers. CAFs can be defined as non-tumor cells within the tumor microenvironment that have the morphological traits of fibroblasts, are negative for lineage-specific markers (e.g., leukocyte, endothelium), and enhance tumor progression in a multi-pronged manner. Two often-mentioned aspects of CAF biology are their ability to alter the mechanics and architecture of the tumor microenvironment, and also to drive local immunosuppression. These two aspects are the specific focus of this work, which also contains a brief summary of novel therapeutic interventions under study to normalize or eliminate CAFs from the tumor microenvironment.
Collapse
Affiliation(s)
- Vanessa C. Talayero
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, 37007 Salamanca, Spain
| | - Miguel Vicente-Manzanares
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, 37007 Salamanca, Spain
- Correspondence: Miguel Vicente-Manzanares, Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, 37007 Salamanca, Spain.
| |
Collapse
|
25
|
Li S, Lu R, Shu L, Chen Y, Zhao J, Dai J, Huang Q, Li X, Meng W, Long F, Li Y, Fan C, Zhou Z, Mo X. An integrated map of fibroblastic populations in human colon mucosa and cancer tissues. Commun Biol 2022; 5:1326. [PMID: 36463319 PMCID: PMC9719516 DOI: 10.1038/s42003-022-04298-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022] Open
Abstract
Fibroblasts and myofibroblasts are major mesenchymal cells in the lamina propria of colon mucosa and in colon cancer tissues. Detailed insight into the highly specific populations of fibroblasts and myofibroblasts is required to understand the integrity and homeostasis of human colon mucosa and colon cancer. Based on gene expression profiles of single cells, we identified fibroblast populations that produce extracellular matrix components, Wnt ligand- and BMP-secreting fibroblasts, chemokine- and chemokine ligand-generating fibroblasts, highly activated fibroblasts, immune-modulating fibroblasts, epithelial cell-modulating myofibroblasts, stimuli-responsive myofibroblasts, proliferating myofibroblasts, fibroblast-like myofibroblasts, matrix producing myofibroblasts, and contractile myofibroblasts in human colon mucosa. In colon cancer tissue, the compositions of fibroblasts and myofibroblasts were highly altered, as were the expressing patterns of genes including BMPs, Wnt ligands, chemokines, chemokine ligands, growth factors and extracellular matrix components in fibroblasts and myofibroblasts. Our work expands the working atlas of fibroblasts and myofibroblasts and provides a framework for interrogating the complexity of stromal cells in human healthy colon mucosa and colon cancer tissues.
Collapse
Affiliation(s)
- Siying Li
- grid.13291.380000 0001 0807 1581Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ran Lu
- grid.13291.380000 0001 0807 1581Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China ,grid.13291.380000 0001 0807 1581Department of Public Health Laboratory Sciences, West China Fourth Hospital, West China School of Public Health, Sichuan University, Chengdu, China ,grid.13291.380000 0001 0807 1581Department of Urology and Pelvic Surgery, West China Fourth Hospital, West China School of Public Health, Sichuan University, Chengdu, China
| | - Linjuan Shu
- grid.13291.380000 0001 0807 1581Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yulin Chen
- grid.13291.380000 0001 0807 1581Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jin Zhao
- grid.13291.380000 0001 0807 1581Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Junlong Dai
- grid.13291.380000 0001 0807 1581Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Qiaorong Huang
- grid.13291.380000 0001 0807 1581Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xue Li
- grid.13291.380000 0001 0807 1581Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Wentong Meng
- grid.13291.380000 0001 0807 1581Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Feiwu Long
- grid.13291.380000 0001 0807 1581Department of Gastrointestinal, Bariatric and Metabolic Surgery, Research Center for Nutrition, Metabolism & Food Safety, West China-PUMC C.C. Chen Institute of Health, West China Fourth Hospital, West China School of Public Health, Sichuan University, Chengdu, China
| | - Yuan Li
- grid.13291.380000 0001 0807 1581Institute of Digestive Surgery and Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Chuanwen Fan
- grid.13291.380000 0001 0807 1581Department of Gastrointestinal, Bariatric and Metabolic Surgery, Research Center for Nutrition, Metabolism & Food Safety, West China-PUMC C.C. Chen Institute of Health, West China Fourth Hospital, West China School of Public Health, Sichuan University, Chengdu, China ,grid.13291.380000 0001 0807 1581Institute of Digestive Surgery and Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Zongguang Zhou
- grid.13291.380000 0001 0807 1581Institute of Digestive Surgery and Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xianming Mo
- grid.13291.380000 0001 0807 1581Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
26
|
Liu J, Yu F, Liu Z, Wang X, Li J. A Robust Prognostic Signature of Tumor Microenvironment in Colorectal Cancer. Cancer Biother Radiopharm 2022; 37:963-975. [PMID: 34551265 DOI: 10.1089/cbr.2021.0171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background: Colorectal cancer (CRC) has been a major public health problem. Tumor microenvironment (TME) greatly contributes to the heterogeneity of CRC and is crucial for the regulation of CRC progression. The authors' study aimed to develop a robust prognostic signature for CRC patients based on TME-related genes. Materials and Methods: Gene expression data and clinicopathologic information of CRC patients were collected from Gene Expression Omnibus and The Cancer Genome Atlas databases. TME-related genes with prognostic value were identified by Cox regression and bootstrap method. The authors used the prognostic genes to construct a robust prognostic model using the least absolute shrinkage and selection operator (LASSO) regression method. The immune and stromal cell abundance of CRC samples were estimated by a microenvironment cell populations-counter method. Results: Based on a training set that comprised 893 CRC samples and 4775 TME-related genes, they established a prognostic model consisting of 25 TME-related genes. With specific risk score formulae, the prognostic model divided CRC patients into high-risk and low-risk subgroups with significantly different survival, which were further confirmed in validation cohorts consisting of other 473 CRC cases or subpopulation of specific stages. The result of time-dependent receiver operating characteristic analysis demonstrated strong predictive accuracy of the prognostic model both in training and validation cohorts. Multivariate Cox regression analysis showed that the 25-gene signature was an independent prognostic factor for overall survival, which was validated through clinical subgroups analysis. Further analysis revealed that CRC samples of high-risk group was abundant of stromal-relevant processes and had a significantly higher proportion of fibroblasts and endothelial cells infiltration. Conclusion: The authors established a robust prognostic signature of 25 TME-related genes which may be an effective tool for prognostic prediction and CRC patient stratification to assist in making treatment decisions.
Collapse
Affiliation(s)
- Jingwen Liu
- Department of Pathology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, P.R. China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, P.R. China.,RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Fei Yu
- Emergency Department, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, P.R. China
| | - Zhao Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Xiaojing Wang
- Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China.,Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Jianming Li
- Department of Pathology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, P.R. China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, P.R. China.,RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| |
Collapse
|
27
|
Liu JW, Yu F, Tan YF, Huo JP, Liu Z, Wang XJ, Li JM. Profiling of Tumor Microenvironment Components Identifies Five Stroma-Related Genes with Prognostic Implications in Colorectal Cancer. Cancer Biother Radiopharm 2022; 37:882-892. [PMID: 33085921 DOI: 10.1089/cbr.2020.4118] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background: Tumor microenvironment (TME) significantly affects colorectal cancer (CRC) progression and therapeutic efficacy, particularly the infiltrating stromal components. This study profiled the TME composition of tumor tissue and identify TME-related, especially stroma-related genes having prognosis value in CRC patients. Materials and Methods: We used the ESTIMATE algorithm to assess stromal/immune component and divided 524 CRC cases of public dataset into high- and low-score groups. We analyzed the effect of the score on prognosis and extracted the differential expression genes (DEGs) between groups, which were stromal- and/or immune-related genes, and performed a prognostic investigation of the DEGs. Results: Higher stromal score correlated with poor survival, whereas the immune score was the inverse. By comparing global gene expression of cases with high vs. low stromal/immune scores, we extracted 474 stroma-related genes, 76 immune-related genes, and 498 intersection genes, which were explored by function enrichment and survival analysis. We identified the expression of five stroma-related genes (including ITGA7, PTPN14, SCG2, TNS1, and GRP) significantly associated with poorer survival, which were validated in the other two independent CRC cohorts. Conclusion: These results presented a comprehensive understanding of TME components and identified five stroma-related genes that predict poor outcomes in CRC patients.
Collapse
Affiliation(s)
- Jing-Wen Liu
- Department of Pathology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China.,RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Fei Yu
- Emergency Department, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Yuan-Fei Tan
- Emergency Department, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Jian-Ping Huo
- Department of Pathology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China.,RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zhao Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xiao-Jing Wang
- Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Jian-Ming Li
- Department of Pathology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China.,RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
28
|
Nanomodulation and nanotherapeutics of tumor-microenvironment. OPENNANO 2022. [DOI: 10.1016/j.onano.2022.100099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
29
|
Modulation of Fibroblast Phenotype by Colorectal Cancer Cell-Secreted Factors Is Mostly Independent of Oncogenic KRAS. Cells 2022; 11:cells11162490. [PMID: 36010567 PMCID: PMC9406506 DOI: 10.3390/cells11162490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 11/23/2022] Open
Abstract
KRAS mutations have been shown to extend their oncogenic effects beyond the cancer cell, influencing the tumor microenvironment. Herein, we studied the impact of mutant KRAS on the modulation of the pro-tumorigenic properties of cancer-associated fibroblasts (CAFs), including α-SMA expression, TGFβ1 and HGF production, extracellular matrix components and metalloproteinases expression as well as collagen contraction and migration capacities. To do so, CCD-18Co normal-like colon fibroblasts were challenged with conditioned media from control and KRAS silenced colorectal cancer (CRC) cells. Our results showed that the mutant KRAS CRC cell-secreted factors were capable of turning normal-like fibroblasts into CAF-like by modulating the α-SMA expression, TGFβ1 and HGF production and migration capacity. Oncogenic KRAS played a secondary role as its silencing did not completely impair the capacity of CRC cells to modulate most of the fibroblast properties analyzed. In summary, our work suggests that mutant KRAS does not play a major role in controlling the CRC cell-secreted factors that modulate the behavior of fibroblasts. The fact that CRC cells retain the capacity to modulate the pro-tumorigenic features of fibroblasts independently of KRAS silencing is likely to negatively impact their response to KRAS inhibitors, thus standing as a putative mechanism of resistance to KRAS inhibition with potential therapeutical relevance.
Collapse
|
30
|
Papait A, Romoli J, Stefani FR, Chiodelli P, Montresor MC, Agoni L, Silini AR, Parolini O. Fight the Cancer, Hit the CAF! Cancers (Basel) 2022; 14:cancers14153570. [PMID: 35892828 PMCID: PMC9330284 DOI: 10.3390/cancers14153570] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/19/2022] [Accepted: 07/19/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary In the last 20 years, the tumor microenvironment (TME) has raised an increasing interest from the therapeutic point of view. Indeed, different strategies targeting either the endothelial or the immune component have been implemented. Furthermore, cancer-associated fibroblasts (CAF) have attracted even more interest due to their ability to prime the TME in order to favor tumor progression and metastasis. This current review provides a comprehensive overview on the latest discoveries regarding CAF, more specifically on their complex characterization and on preclinical studies and clinical trials that target CAF within the TME. Abstract The tumor microenvironment (TME) is comprised of different cellular components, such as immune and stromal cells, which co-operate in unison to promote tumor progression and metastasis. In the last decade, there has been an increasing focus on one specific component of the TME, the stromal component, often referred to as Cancer-Associated Fibroblasts (CAF). CAF modulate the immune response and alter the composition of the extracellular matrix with a decisive impact on the response to immunotherapies and conventional chemotherapy. The most recent publications based on single-cell analysis have underlined CAF heterogeneity and the unique plasticity that strongly impact the TME. In this review, we focus not only on the characterization of CAF based on the most recent findings, but also on their impact on the immune system. We also discuss clinical trials and preclinical studies where targeting CAF revealed controversial results. Therefore, future efforts should focus on understanding the functional properties of individual subtypes of CAF, taking into consideration the peculiarities of each pathological context.
Collapse
Affiliation(s)
- Andrea Papait
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.P.); (J.R.)
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy
| | - Jacopo Romoli
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.P.); (J.R.)
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy; (F.R.S.); (P.C.); (A.R.S.)
| | - Francesca Romana Stefani
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy; (F.R.S.); (P.C.); (A.R.S.)
| | - Paola Chiodelli
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy; (F.R.S.); (P.C.); (A.R.S.)
| | | | - Lorenzo Agoni
- Obstetrics and Gynecology Unit, Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy;
| | - Antonietta Rosa Silini
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy; (F.R.S.); (P.C.); (A.R.S.)
| | - Ornella Parolini
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.P.); (J.R.)
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy
- Correspondence: ; Tel.: +39-0630154464
| |
Collapse
|
31
|
Liu B, Yin X, Jiang G, Li Y, Jiang Z, Qiang L, Chen N, Fan Y, Shen C, Dai L, Yin Y, Zhang B. Identification of Endoplasmic Reticulum Stress-Related Subtypes, Infiltration Analysis of Tumor Microenvironment, and Construction of a Prognostic Model in Colorectal Cancer. Cancers (Basel) 2022; 14:3326. [PMID: 35884393 PMCID: PMC9322646 DOI: 10.3390/cancers14143326] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/29/2022] [Accepted: 07/05/2022] [Indexed: 02/04/2023] Open
Abstract
Recently, endoplasmic reticulum (ER) stress has been shown to influence tumor progression and immune cell function in the tumor microenvironment (TME). However, the underlying role of ER stress-related gene patterns in colorectal cancer (CRC) development remains unclear. We analyzed the ER stress-related gene patterns in 884 patients with CRC from the Gene Expression Omnibus database and evaluated the cell-infiltrating patterns in the TME. Two ER stress-related patterns were identified in patients with CRC that had distinct cell-infiltrating patterns in the TME and clinical characteristics. A risk score and nomogram based on 14 screened prognosis-correlated genes was built and validated to predict patient survival. Patients with a higher risk score were shown to have an unfavorable prognosis, and the risk score was associated with cell infiltration and drug sensitivity. Furthermore, spatial transcriptomics data were utilized to explore ER stress-related gene patterns in CRC tissues, and it was shown that ER stress phenotype involves in the formation of the immunosuppressive TME. This study demonstrated that ER stress-related gene patterns play a role in influencing the TME and predicting prognosis. These analyses of ER stress in the TME of CRC might deepen our understanding of CRC progression and immune escape and provide novel insights into therapeutic strategies.
Collapse
Affiliation(s)
- Baike Liu
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, China; (B.L.); (X.Y.); (Z.J.); (C.S.)
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China; (N.C.); (Y.F.); (L.D.)
| | - Xiaonan Yin
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, China; (B.L.); (X.Y.); (Z.J.); (C.S.)
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China; (N.C.); (Y.F.); (L.D.)
| | - Guangfu Jiang
- Department of Gastrointestinal Surgery, Guang’an People’s Hospital, Guang’an 638500, China; (G.J.); (Y.L.)
| | - Yang Li
- Department of Gastrointestinal Surgery, Guang’an People’s Hospital, Guang’an 638500, China; (G.J.); (Y.L.)
| | - Zhiyuan Jiang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, China; (B.L.); (X.Y.); (Z.J.); (C.S.)
| | - Liming Qiang
- Department of Gastroenterology Ward, Guang’an People’s Hospital, Guang’an 638500, China;
| | - Na Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China; (N.C.); (Y.F.); (L.D.)
- School of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| | - Yating Fan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China; (N.C.); (Y.F.); (L.D.)
| | - Chaoyong Shen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, China; (B.L.); (X.Y.); (Z.J.); (C.S.)
| | - Lei Dai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China; (N.C.); (Y.F.); (L.D.)
| | - Yuan Yin
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, China; (B.L.); (X.Y.); (Z.J.); (C.S.)
| | - Bo Zhang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, China; (B.L.); (X.Y.); (Z.J.); (C.S.)
| |
Collapse
|
32
|
Xiang X, Niu YR, Wang ZH, Ye LL, Peng WB, Zhou Q. Cancer-associated fibroblasts: Vital suppressors of the immune response in the tumor microenvironment. Cytokine Growth Factor Rev 2022; 67:35-48. [DOI: 10.1016/j.cytogfr.2022.07.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 12/17/2022]
|
33
|
Chen B, Mu C, Zhang Z, He X, Liu X. The Love-Hate Relationship Between TGF-β Signaling and the Immune System During Development and Tumorigenesis. Front Immunol 2022; 13:891268. [PMID: 35720407 PMCID: PMC9204485 DOI: 10.3389/fimmu.2022.891268] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/25/2022] [Indexed: 11/20/2022] Open
Abstract
Since TGF-β was recognized as an essential secreted cytokine in embryogenesis and adult tissue homeostasis a decade ago, our knowledge of the role of TGF-β in mammalian development and disease, particularly cancer, has constantly been updated. Mounting evidence has confirmed that TGF-β is the principal regulator of the immune system, as deprivation of TGF-β signaling completely abrogates adaptive immunity. However, enhancing TGF-β signaling constrains the immune response through multiple mechanisms, including boosting Treg cell differentiation and inducing CD8+ T-cell apoptosis in the disease context. The love-hate relationship between TGF-β signaling and the immune system makes it challenging to develop effective monotherapies targeting TGF-β, especially for cancer treatment. Nonetheless, recent work on combination therapies of TGF-β inhibition and immunotherapy have provide insights into the development of TGF-β-targeted therapies, with favorable outcomes in patients with advanced cancer. Hence, we summarize the entanglement between TGF-β and the immune system in the developmental and tumor contexts and recent progress on hijacking crucial TGF-β signaling pathways as an emerging area of cancer therapy.
Collapse
Affiliation(s)
- Baode Chen
- Department of Laboratory Medicine, Key Laboratory of Clinical In Vitro Diagnostic Techniques of Zhejiang Province, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Chenglin Mu
- Institute for Intelligent Bio/Chem Manufacturing (iBCM), Zhejiang University (ZJU)-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China
| | - Zhiwei Zhang
- Institute for Intelligent Bio/Chem Manufacturing (iBCM), Zhejiang University (ZJU)-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China
| | - Xuelin He
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xia Liu
- Institute for Intelligent Bio/Chem Manufacturing (iBCM), Zhejiang University (ZJU)-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China
| |
Collapse
|
34
|
Bhome R, Emaduddin M, James V, House LM, Thirdborough SM, Mellone M, Tulkens J, Primrose JN, Thomas GJ, De Wever O, Mirnezami AH, Sayan AE. Epithelial to mesenchymal transition influences fibroblast phenotype in colorectal cancer by altering miR-200 levels in extracellular vesicles. J Extracell Vesicles 2022; 11:e12226. [PMID: 35595718 PMCID: PMC9122835 DOI: 10.1002/jev2.12226] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 02/14/2022] [Accepted: 04/26/2022] [Indexed: 12/28/2022] Open
Abstract
Colorectal cancer (CRC) with a mesenchymal gene expression signature has the greatest propensity for distant metastasis and is characterised by the accumulation of cancer‐associated fibroblasts in the stroma. We investigated whether the epithelial to mesenchymal transition status of CRC cells influences fibroblast phenotype, with a focus on the transfer of extracellular vesicles (EVs), as a controlled means of cell–cell communication. Epithelial CRC EVs suppressed TGF‐β‐driven myofibroblast differentiation, whereas mesenchymal CRC EVs did not. This was driven by miR‐200 (miR‐200a/b/c, ‐141), which was enriched in epithelial CRC EVs and transferred to recipient fibroblasts. Ectopic miR‐200 expression or ZEB1 knockdown, in fibroblasts, similarly suppressed myofibroblast differentiation. Supporting these findings, there was a strong negative correlation between miR‐200 and myofibroblastic markers in a cohort of CRC patients in the TCGA dataset. This was replicated in mice, by co‐injecting epithelial or mesenchymal CRC cells with fibroblasts and analysing stromal markers of myofibroblastic phenotype. Fibroblasts from epithelial tumours contained more miR‐200 and expressed less ACTA2 and FN1 than those from mesenchymal tumours. As such, these data provide a new mechanism for the development of fibroblast heterogeneity in CRC, through EV‐mediated transfer of miRNAs, and provide an explanation as to why CRC tumours with greater metastatic potential are CAF rich.
Collapse
Affiliation(s)
- Rahul Bhome
- Cancer Sciences Unit, University of Southampton, Southampton, UK.,University Surgery, University of Southampton, Southampton, UK
| | | | - Victoria James
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, UK
| | - Louise M House
- Cancer Sciences Unit, University of Southampton, Southampton, UK
| | | | | | - Joeri Tulkens
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - John N Primrose
- University Surgery, University of Southampton, Southampton, UK
| | - Gareth J Thomas
- Cancer Sciences Unit, University of Southampton, Southampton, UK
| | - Olivier De Wever
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Alex H Mirnezami
- Cancer Sciences Unit, University of Southampton, Southampton, UK.,University Surgery, University of Southampton, Southampton, UK
| | - A Emre Sayan
- Cancer Sciences Unit, University of Southampton, Southampton, UK
| |
Collapse
|
35
|
Vega PN, Nilsson A, Kumar MP, Niitsu H, Simmons AJ, Ro J, Wang J, Chen Z, Joughin BA, Li W, McKinley ET, Liu Q, Roland JT, Washington MK, Coffey RJ, Lauffenburger DA, Lau KS. Cancer-Associated Fibroblasts and Squamous Epithelial Cells Constitute a Unique Microenvironment in a Mouse Model of Inflammation-Induced Colon Cancer. Front Oncol 2022; 12:878920. [PMID: 35600339 PMCID: PMC9114773 DOI: 10.3389/fonc.2022.878920] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/06/2022] [Indexed: 11/13/2022] Open
Abstract
The tumor microenvironment plays a key role in the pathogenesis of colorectal tumors and contains various cell types including epithelial, immune, and mesenchymal cells. Characterization of the interactions between these cell types is necessary for revealing the complex nature of tumors. In this study, we used single-cell RNA-seq (scRNA-seq) to compare the tumor microenvironments between a mouse model of sporadic colorectal adenoma (Lrig1CreERT2/+;Apc2lox14/+) and a mouse model of inflammation-driven colorectal cancer induced by azoxymethane and dextran sodium sulfate (AOM/DSS). While both models develop tumors in the distal colon, we found that the two tumor types have distinct microenvironments. AOM/DSS tumors have an increased abundance of two populations of cancer-associated fibroblasts (CAFs) compared with APC tumors, and we revealed their divergent spatial association with tumor cells using multiplex immunofluorescence (MxIF) imaging. We also identified a unique squamous cell population in AOM/DSS tumors, whose origins were distinct from anal squamous epithelial cells. These cells were in higher proportions upon administration of a chemotherapy regimen of 5-Fluorouracil/Irinotecan. We used computational inference algorithms to predict cell-cell communication mediated by ligand-receptor interactions and downstream pathway activation, and identified potential mechanistic connections between CAFs and tumor cells, as well as CAFs and squamous epithelial cells. This study provides important preclinical insight into the microenvironment of two distinct models of colorectal tumors and reveals unique roles for CAFs and squamous epithelial cells in the AOM/DSS model of inflammation-driven cancer.
Collapse
Affiliation(s)
- Paige N. Vega
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University, Nashville, TN, United States
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Avlant Nilsson
- Department of Biological Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Manu P. Kumar
- Department of Biological Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Hiroaki Niitsu
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Alan J. Simmons
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University, Nashville, TN, United States
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | - James Ro
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University, Nashville, TN, United States
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jiawei Wang
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University, Nashville, TN, United States
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Zhengyi Chen
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University, Nashville, TN, United States
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Brian A. Joughin
- Department of Biological Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Wei Li
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Eliot T. McKinley
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University, Nashville, TN, United States
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Qi Liu
- Department of Biostatistics and Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Joseph T. Roland
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, United States
| | - M. Kay Washington
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Robert J. Coffey
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University, Nashville, TN, United States
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Douglas A. Lauffenburger
- Department of Biological Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Ken S. Lau
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University, Nashville, TN, United States
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
36
|
Hu D, Li Z, Zheng B, Lin X, Pan Y, Gong P, Zhuo W, Hu Y, Chen C, Chen L, Zhou J, Wang L. Cancer-associated fibroblasts in breast cancer: Challenges and opportunities. Cancer Commun (Lond) 2022; 42:401-434. [PMID: 35481621 PMCID: PMC9118050 DOI: 10.1002/cac2.12291] [Citation(s) in RCA: 127] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 02/06/2022] [Accepted: 04/07/2022] [Indexed: 12/13/2022] Open
Abstract
The tumor microenvironment is proposed to contribute substantially to the progression of cancers, including breast cancer. Cancer-associated fibroblasts (CAFs) are the most abundant components of the tumor microenvironment. Studies have revealed that CAFs in breast cancer originate from several types of cells and promote breast cancer malignancy by secreting factors, generating exosomes, releasing nutrients, reshaping the extracellular matrix, and suppressing the function of immune cells. CAFs are also becoming therapeutic targets for breast cancer due to their specific distribution in tumors and their unique biomarkers. Agents interrupting the effect of CAFs on surrounding cells have been developed and applied in clinical trials. Here, we reviewed studies examining the heterogeneity of CAFs in breast cancer and expression patterns of CAF markers in different subtypes of breast cancer. We hope that summarizing CAF-related studies from a historical perspective will help to accelerate the development of CAF-targeted therapeutic strategies for breast cancer.
Collapse
Affiliation(s)
- Dengdi Hu
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, P. R. China
| | - Zhaoqing Li
- Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), Hangzhou, Zhejiang, 310016, P. R. China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang, 310016, P. R. China
| | - Bin Zheng
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, P. R. China
| | - Xixi Lin
- Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), Hangzhou, Zhejiang, 310016, P. R. China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang, 310016, P. R. China
| | - Yuehong Pan
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, P. R. China
| | - Peirong Gong
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, P. R. China
| | - Wenying Zhuo
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, P. R. China.,Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), Hangzhou, Zhejiang, 310016, P. R. China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang, 310016, P. R. China
| | - Yujie Hu
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, P. R. China
| | - Cong Chen
- Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), Hangzhou, Zhejiang, 310016, P. R. China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang, 310016, P. R. China
| | - Lini Chen
- Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), Hangzhou, Zhejiang, 310016, P. R. China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang, 310016, P. R. China
| | - Jichun Zhou
- Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), Hangzhou, Zhejiang, 310016, P. R. China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang, 310016, P. R. China
| | - Linbo Wang
- Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), Hangzhou, Zhejiang, 310016, P. R. China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang, 310016, P. R. China
| |
Collapse
|
37
|
Mele V, Basso C, Governa V, Glaus Garzon JF, Muraro MG, Däster S, Nebiker CA, Mechera R, Bolli M, Schmidt A, Geiger R, Spagnoli GC, Christoforidis D, Majno PE, Borsig L, Iezzi G. Identification of TPM2 and CNN1 as Novel Prognostic Markers in Functionally Characterized Human Colon Cancer-Associated Stromal Cells. Cancers (Basel) 2022; 14:cancers14082024. [PMID: 35454931 PMCID: PMC9025001 DOI: 10.3390/cancers14082024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/28/2022] [Accepted: 04/12/2022] [Indexed: 01/27/2023] Open
Abstract
Simple Summary Non-transformed cells of tumor microenvironment also impact on cancer outgrowth and progression. In colon cancer, a leading cause of cancer-related death worldwide, a high abundance of a heterogeneous cell population generally referred to as cancer-associated fibroblasts (CAFs) or tumor-associated stromal cells (TASCs) is associated with poor prognosis. The identification of TASC-specific markers could help to select patients for additional treatments and may provide novel targets for innovative therapies. Some markers have been proposed, but their prognostic significance is modest. We successfully expanded TASCs from human colon cancers and demonstrated their capacity to promote tumor growth and metastatic spread in vitro and in in vivo models. By comparing TASC whole protein expression, the so-called “proteome”, with that of stromal cells derived from matched healthy colon tissues, we identified two novel markers highly significantly associated with severe prognosis. Our results might help to identify patients at risk and might suggest new treatment options. Abstract Stromal infiltration is associated with poor prognosis in human colon cancers. However, the high heterogeneity of human tumor-associated stromal cells (TASCs) hampers a clear identification of specific markers of prognostic relevance. To address these issues, we established short-term cultures of TASCs and matched healthy mucosa-associated stromal cells (MASCs) from human primary colon cancers and, upon characterization of their phenotypic and functional profiles in vitro and in vivo, we identified differentially expressed markers by proteomic analysis and evaluated their prognostic significance. TASCs were characterized by higher proliferation and differentiation potential, and enhanced expression of mesenchymal stem cell markers, as compared to MASCs. TASC triggered epithelial–mesenchymal transition (EMT) in tumor cells in vitro and promoted their metastatic spread in vivo, as assessed in an orthotopic mouse model. Proteomic analysis of matched TASCs and MASCs identified a panel of markers preferentially expressed in TASCs. The expression of genes encoding two of them, calponin 1 (CNN1) and tropomyosin beta chain isoform 2 (TPM2), was significantly associated with poor outcome in independent databases and outperformed the prognostic significance of currently proposed TASC markers. The newly identified markers may improve prognostication of primary colon cancers and identification of patients at risk.
Collapse
Affiliation(s)
- Valentina Mele
- Department of Biomedicine, University Hospital Basel and University of Basel, 4031 Basel, Switzerland; (V.M.); (M.G.M.)
| | - Camilla Basso
- Laboratory for Surgical Research, Ente Ospedaliero Cantonale, 6500 Bellinzona, Switzerland;
- Faculty of Biomedical Sciences, Università della Svizzera italiana, 6900 Lugano, Switzerland; (D.C.); (P.E.M.)
| | - Valeria Governa
- Department of Clinical Sciences Lund, Section of Oncology, Lund University, 221 85 Lund, Sweden;
| | - Jesus F. Glaus Garzon
- Institute of Physiology, University of Zürich, 8006 Zürich, Switzerland; (J.F.G.G.); (L.B.)
| | - Manuele G. Muraro
- Department of Biomedicine, University Hospital Basel and University of Basel, 4031 Basel, Switzerland; (V.M.); (M.G.M.)
| | - Silvio Däster
- Department of General Surgery, University Hospital Basel, 4031 Basel, Switzerland; (S.D.); (C.A.N.); (R.M.)
| | - Christian A. Nebiker
- Department of General Surgery, University Hospital Basel, 4031 Basel, Switzerland; (S.D.); (C.A.N.); (R.M.)
| | - Robert Mechera
- Department of General Surgery, University Hospital Basel, 4031 Basel, Switzerland; (S.D.); (C.A.N.); (R.M.)
| | - Martin Bolli
- Department of Visceral Surgery, Clarunis-University Center for Gastrointestinal and Liver Diseases, St. Claraspital and University Hospital Basel, 4002 Basel, Switzerland;
| | - Alexander Schmidt
- Proteomics Core Facility, Biozentrum, University of Basel, 4056 Basel, Switzerland;
| | - Roger Geiger
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland;
- Institute of Oncology Research, Università della Svizzera italiana, 6900 Lugano, Switzerland
| | - Giulio C. Spagnoli
- Institute of Translational Pharmacology, National Research Council, 00133 Rome, Italy;
| | - Dimitri Christoforidis
- Faculty of Biomedical Sciences, Università della Svizzera italiana, 6900 Lugano, Switzerland; (D.C.); (P.E.M.)
- Department of Surgery, Ente Ospedaliero Cantonale, 6500 Bellinzona, Switzerland
| | - Pietro E. Majno
- Faculty of Biomedical Sciences, Università della Svizzera italiana, 6900 Lugano, Switzerland; (D.C.); (P.E.M.)
- Department of Surgery, Ente Ospedaliero Cantonale, 6500 Bellinzona, Switzerland
| | - Lubor Borsig
- Institute of Physiology, University of Zürich, 8006 Zürich, Switzerland; (J.F.G.G.); (L.B.)
| | - Giandomenica Iezzi
- Laboratory for Surgical Research, Ente Ospedaliero Cantonale, 6500 Bellinzona, Switzerland;
- Faculty of Biomedical Sciences, Università della Svizzera italiana, 6900 Lugano, Switzerland; (D.C.); (P.E.M.)
- Correspondence:
| |
Collapse
|
38
|
Qin X, Zhao M, Deng W, Huang Y, Cheng Z, Chung JPW, Chen X, Yang K, Chan DYL, Wang H. Development and Validation of a Novel Prognostic Nomogram Combined With Desmoplastic Reaction for Synchronous Colorectal Peritoneal Metastasis. Front Oncol 2022; 12:826830. [PMID: 35359399 PMCID: PMC8963183 DOI: 10.3389/fonc.2022.826830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/15/2022] [Indexed: 11/13/2022] Open
Abstract
PurposeThe prognostic value of desmoplastic reaction (DR) has not been investigated in colorectal cancer (CRC) patients with synchronous peritoneal metastasis (SPM). The present study aimed to identify whether DR can predict overall survival (OS) and develop a novel prognostic nomogram.MethodsCRC patients with SPM were enrolled from a single center between July 2007 and July 2019. DR patterns in primary tumors were classified as mature, intermediate, or immature according to the existence and absence of keloid-like collagen or myxoid stroma. Cox regression analysis was used to identify independent factors associated with OS and a nomogram was developed subsequently.ResultsOne hundred ninety-eight and 99 patients were randomly allocated into the training and validation groups. The median OS in the training group was 36, 25, and 12 months in mature, intermediate, and immature DR categories, respectively. Age, T stage, extraperitoneal metastasis, differentiation, cytoreductive surgery (CRS), hyperthermic intraperitoneal chemotherapy (HIPEC), and DR categorization were independent variables for OS, based on which the nomogram was developed. The C-index of the nomogram in the training and validation groups was 0.773 (95% CI 0.734–0.812) and 0.767 (95% CI 0.708–0.826). The calibration plots showed satisfactory agreement between the actual outcome and nomogram-predicted OS probabilities in the training and validation cohorts.ConclusionsDR classification in the primary tumor is a potential prognostic index for CRC patients with SPM. The novel prognostic nomogram combined with DR classification has good discrimination and accuracy in predicting the OS for CRC patients with SPM.
Collapse
Affiliation(s)
- Xiusen Qin
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by the National Key Clinical Discipline, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Mingpeng Zhao
- Assisted Reproductive Technology Unit, Department of Obstetrics and Gynaecology, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Weihao Deng
- Department of Pathology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yan Huang
- Department of Pathology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhiqiang Cheng
- Department of Pathology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jacqueline Pui Wah Chung
- Assisted Reproductive Technology Unit, Department of Obstetrics and Gynaecology, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Xufei Chen
- Department of Obstetrics and Gynaecology, Songshan Lake Central Hospital, Affiliated Dongguan Shilong People’s Hospital of Southern Medical University, Dongguan, China
| | - Keli Yang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by the National Key Clinical Discipline, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- *Correspondence: Keli Yang, ; David Yiu Leung Chan, ; Hui Wang,
| | - David Yiu Leung Chan
- Assisted Reproductive Technology Unit, Department of Obstetrics and Gynaecology, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- *Correspondence: Keli Yang, ; David Yiu Leung Chan, ; Hui Wang,
| | - Hui Wang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by the National Key Clinical Discipline, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- *Correspondence: Keli Yang, ; David Yiu Leung Chan, ; Hui Wang,
| |
Collapse
|
39
|
Tinsley SL, Allen-Petersen BL. PP2A and cancer epigenetics: a therapeutic opportunity waiting to happen. NAR Cancer 2022; 4:zcac002. [PMID: 35118387 PMCID: PMC8807117 DOI: 10.1093/narcan/zcac002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 12/08/2021] [Accepted: 01/10/2022] [Indexed: 12/13/2022] Open
Abstract
The epigenetic state of chromatin is altered by regulators which influence gene expression in response to environmental stimuli. While several post-translational modifications contribute to chromatin accessibility and transcriptional programs, our understanding of the role that specific phosphorylation sites play is limited. In cancer, kinases and phosphatases are commonly deregulated resulting in increased oncogenic signaling and loss of epigenetic regulation. Aberrant epigenetic states are known to promote cellular plasticity and the development of therapeutic resistance in many cancer types, highlighting the importance of these mechanisms to cancer cell phenotypes. Protein Phosphatase 2A (PP2A) is a heterotrimeric holoenzyme that targets a diverse array of cellular proteins. The composition of the PP2A complex influences its cellular targets and activity. For this reason, PP2A can be tumor suppressive or oncogenic depending on cellular context. Understanding the nuances of PP2A regulation and its effect on epigenetic alterations can lead to new therapeutic avenues that afford more specificity and contribute to the growth of personalized medicine in the oncology field. In this review, we summarize the known PP2A-regulated substrates and potential phosphorylation sites that contribute to cancer cell epigenetics and possible strategies to therapeutically leverage this phosphatase to suppress tumor growth.
Collapse
Affiliation(s)
- Samantha L Tinsley
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | | |
Collapse
|
40
|
Gommes CJ, Louis T, Bourgot I, Noël A, Blacher S, Maquoi E. Remodelling of the fibre-aggregate structure of collagen gels by cancer-associated fibroblasts: A time-resolved grey-tone image analysis based on stochastic modelling. Front Immunol 2022; 13:988502. [PMID: 36818478 PMCID: PMC9936192 DOI: 10.3389/fimmu.2022.988502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 12/19/2022] [Indexed: 02/05/2023] Open
Abstract
Introduction Solid tumors consist of tumor cells associated with stromal and immune cells, secreted factors and extracellular matrix (ECM), which together constitute the tumor microenvironment. Among stromal cells, activated fibroblasts, known as cancer-associated fibroblasts (CAFs) are of particular interest. CAFs secrete a plethora of ECM components including collagen and modulate the architecture of the ECM, thereby influencing cancer cell migration. The characterization of the collagen fibre network and its space and time-dependent microstructural modifications is key to investigating the interactions between cells and the ECM. Developing image analysis tools for that purpose is still a challenge because the structural complexity of the collagen network calls for specific statistical descriptors. Moreover, the low signal-to-noise ratio of imaging techniques available for time-resolved studies rules out standard methods based on image segmentation. Methods In this work, we develop a novel approach based on the stochastic modelling of the gel structure and on grey-tone image analysis. The method is then used to study the remodelling of a collagen matrix by migrating breast cancer-derived CAFs in a three-dimensional spheroid model of cellular invasion imaged by time-lapse confocal microscopy. Results The structure of the collagen at the scale of a few microns consists in regions with high fibre density separated by depleted regions, which can be thought of as aggregates and pores. The approach developped captures this two-scale structure with a clipped Gaussian field model to describe the aggregates-and-pores large-scale structure, and a homogeneous Boolean model to describe the small-scale fibre network within the aggregates. The model parameters are identified by fitting the grey-tone histograms and correlation functions of the images. The method applies to unprocessed grey-tone images, and it can therefore be used with low magnification, noisy time-lapse reflectance images. When applied to the CAF spheroid time-resolved images, the method reveals different matrix densification mechanisms for the matrix in direct contact or far from the cells. Conclusion We developed a novel and multidisciplinary image analysis approach to investigate the remodelling of fibrillar collagen in a 3D spheroid model of cellular invasion. The specificity of the method is that it applies to the unprocessed grey-tone images, and it can therefore be used with noisy time-lapse reflectance images of non-fluorescent collagen. When applied to the CAF spheroid time-resolved images, the method reveals different matrix densification mechanisms for the matrix in direct contact or far from the cells.
Collapse
Affiliation(s)
- Cedric J Gommes
- Department of Chemical Engineering, School of Engineering, University of Liège, Liège, Belgium
| | - Thomas Louis
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Isabelle Bourgot
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Agnès Noël
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Silvia Blacher
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Erik Maquoi
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège, Liège, Belgium
| |
Collapse
|
41
|
Loh JJ, Ma S. The Role of Cancer-Associated Fibroblast as a Dynamic Player in Mediating Cancer Stemness in the Tumor Microenvironment. Front Cell Dev Biol 2021; 9:727640. [PMID: 34760886 PMCID: PMC8573407 DOI: 10.3389/fcell.2021.727640] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 09/24/2021] [Indexed: 01/15/2023] Open
Abstract
The enrichment of cancer-associated fibroblast (CAFs) in a tumor microenvironment (TME) cultivates a pro-tumorigenic niche via aberrant paracrine signaling and matrix remodeling. A favorable niche is critical to the maintenance of cancer stem cells (CSCs), a population of cells that are characterized by their enhanced ability to self-renew, metastasis, and develop therapy resistance. Mounting evidence illustrates the interplay between CAF and cancer cells expedites malignant progression. Therefore, targeting the key cellular components and factors in the niche may promote a more efficacious treatment. In this study, we discuss how CAF orchestrates a niche that enhances CSC features and the potential therapeutic implication.
Collapse
Affiliation(s)
- Jia Jian Loh
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Stephanie Ma
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong.,State Key Laboratory of Liver Research, The University of Hong Kong, Pokfulam, Hong Kong, SAR China
| |
Collapse
|
42
|
Luo T, von der Ohe J, Hass R. MSC-Derived Extracellular Vesicles in Tumors and Therapy. Cancers (Basel) 2021; 13:cancers13205212. [PMID: 34680359 PMCID: PMC8533755 DOI: 10.3390/cancers13205212] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 10/15/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Therapeutic functions of mesenchymal stroma-/stem-like cells (MSCs) are mediated predominantly through paracrine effects by the release of various different components. Upon recruitment of MSCs to damaged tissue sites or tumors, several bioactive molecules and organelles that are secreted by MSCs among others are cytokines, chemokines, metabolites, and extracellular vesicles including exosomes. The MSC-mediated cargo of released exosomes contains specific proteins and nucleic acids with varying regulatory microRNAs according to the tissue origin and the MSC microenvironment. These MSC-released exosomes are taken up by different target cells in damaged tissues to promote a regulatory network of tissue repair, including immune modulation and induction of angiogenesis. Conversely, in tumors, MSC-derived exosomes can confer predominant signals to suppress neovascularization and to relay further tumor-inhibitory effects. However, MSCs that adapted to the tumor tissue by mutual interaction with cancer cells progressively alter to an aberrant phenotype with the release of exosomes carrying tumor-supportive material. Abstract Exosomes derived from mesenchymal stroma-/stem-like cells (MSCs) as part of extracellular vesicles are considered cell-free biocompatible nanovesicles that promote repair activities of damaged tissues or organs by exhibiting low immunogenic and cytotoxic effects. Contributions to regenerative activities include wound healing, maintenance of stem cell niches, beneficial regenerative effects in various diseases, and reduction of senescence. However, the mode of action in MSC-derived exosomes strongly depends on the biological content like different regulatory microRNAs that are determined by the tissue origin of MSCs. In tumors, MSCs use indirect and direct pathways in a communication network to interact with cancer cells. This leads to mutual functional changes with the acquisition of an aberrant tumor-associated MSC phenotype accompanied by altered cargo in the exosomes. Consequently, MSC-derived exosomes either from normal tissue-originating MSCs or from aberrant tumor-associated MSCs can confer different actions on tumor development. These processes exhibiting tumor-inhibitory and tumor-supportive effects with a focus on exosome microRNA content will be discriminated and discussed within this review.
Collapse
Affiliation(s)
| | | | - Ralf Hass
- Correspondence: ; Tel.: +49-511-532-6070
| |
Collapse
|
43
|
Paulsen JD, Polydorides AD. Pathology and Prognosis of Colonic Adenocarcinomas With Intermediate Primary Tumor Stage Between pT2 and pT3. Arch Pathol Lab Med 2021; 146:591-602. [PMID: 34473229 DOI: 10.5858/arpa.2021-0109-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2021] [Indexed: 11/06/2022]
Abstract
CONTEXT.— Primary tumor stage (pT) is an important prognostic indicator in colonic adenocarcinomas; however, cases that have no muscle fibers beyond the advancing tumor edge but also show no extension beyond the apparent outer border of muscularis propria (termed pT2int), have not been previously studied. OBJECTIVE.— To address the clinicopathologic characteristics and prognosis of pT2int tumors. DESIGN.— We recharacterized 168 colon carcinomas and compared pT2int cases to bona fide pT2 and pT3 tumors. RESULTS.— In side-by-side analysis, 21 pT2int cases diverged from 29 pT2 tumors only in terms of larger size (P = .03), but they were less likely to show high-grade (P = .03), lymphovascular (P < .001), and extramural venous invasion (P = .04); discontinuous tumor deposits (P = .02); lymph node involvement (P = .001); and advanced stage (P = .001), compared with 118 pT3 tumors. Combining pT2int with pT2 cases (versus pT3) was a better independent predictor of negative lymph nodes in multivariate analysis (P = .04; odds ratio [OR], 3.96; CI, 1.09-14.42) and absent distant metastasis in univariate analysis (P = .04), compared with sorting pT2int with pT3 cases (versus pT2). Proportional hazards regression showed that pT2 and pT2int cases together were associated with better disease-free survival compared with pT3 tumors (P = .04; OR, 3.65; CI, 1.05-12.70). Kaplan-Meier analysis demonstrated that when pT2int were grouped with pT2 tumors, they were significantly less likely to show disease progression compared with pT3 (P = .002; log-rank test) and showed a trend toward better disease-specific survival (P = .06), during a mean patient follow-up of 44.9 months. CONCLUSIONS.— These data support the conclusion that pT2int carcinomas have clinicopathologic characteristics and are associated with patient outcomes more closely aligned with pT2 rather than pT3 tumors.
Collapse
Affiliation(s)
- John D Paulsen
- From the Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alexandros D Polydorides
- From the Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
44
|
TGF-β Signaling: From Tissue Fibrosis to Tumor Microenvironment. Int J Mol Sci 2021; 22:ijms22147575. [PMID: 34299192 PMCID: PMC8303588 DOI: 10.3390/ijms22147575] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/09/2021] [Accepted: 07/09/2021] [Indexed: 02/06/2023] Open
Abstract
Transforming growth factor-β (TGF-β) signaling triggers diverse biological actions in inflammatory diseases. In tissue fibrosis, it acts as a key pathogenic regulator for promoting immunoregulation via controlling the activation, proliferation, and apoptosis of immunocytes. In cancer, it plays a critical role in tumor microenvironment (TME) for accelerating invasion, metastasis, angiogenesis, and immunosuppression. Increasing evidence suggest a pleiotropic nature of TGF-β signaling as a critical pathway for generating fibrotic TME, which contains numerous cancer-associated fibroblasts (CAFs), extracellular matrix proteins, and remodeling enzymes. Its pathogenic roles and working mechanisms in tumorigenesis are still largely unclear. Importantly, recent studies successfully demonstrated the clinical implications of fibrotic TME in cancer. This review systematically summarized the latest updates and discoveries of TGF-β signaling in the fibrotic TME.
Collapse
|
45
|
Jiang Y, Liang X, Han Z, Wang W, Xi S, Li T, Chen C, Yuan Q, Li N, Yu J, Xie Y, Xu Y, Zhou Z, Poultsides GA, Li G, Li R. Radiographical assessment of tumour stroma and treatment outcomes using deep learning: a retrospective, multicohort study. LANCET DIGITAL HEALTH 2021; 3:e371-e382. [PMID: 34045003 DOI: 10.1016/s2589-7500(21)00065-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/16/2021] [Accepted: 04/07/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND The tumour stroma microenvironment plays an important part in disease progression and its composition can influence treatment response and outcomes. Histological evaluation of tumour stroma is limited by access to tissue, spatial heterogeneity, and temporal evolution. We aimed to develop a radiological signature for non-invasive assessment of tumour stroma and treatment outcomes. METHODS In this multicentre, retrospective study, we analysed CT images and outcome data of 2209 patients with resected gastric cancer from five independent cohorts recruited from two centres (Nanfang Hospital of Southern Medical University [Guangzhou, China] and Sun Yat-sen University Cancer Center [Guangzhou, China]). Patients with histologically confirmed gastric cancer, at least 15 lymph nodes harvested, preoperative abdominal CT available, and complete clinicopathological and follow-up data were eligible for inclusion. Tumour tissue was collected for patients in the training cohort (321 patients), internal validation cohort one (246 patients), and external validation cohort one (128 patients). Four stroma classes were defined according to the protein expression of α-smooth muscle actin and periostin assessed by immunohistochemistry. The primary objective was to predict the histologically based stroma classes by using preoperative CT images. We trained a deep convolutional neural network model using the training cohort and tested the model in the internal and external validation cohort one. We evaluated the model's association with prognosis in the training cohort, two internal, and two external validation cohorts and compared outcomes of patients who received or did not receive adjuvant chemotherapy. FINDINGS The deep-learning model achieved a high diagnostic accuracy for assessing tumour stroma in both internal validation cohort one (area under the receiver operating characteristic curve [AUC] 0·96-0·98]) and external validation cohort one (AUC 0·89-0·94). The stromal imaging signature was significantly associated with disease-free survival and overall survival in all cohorts (p<0·0001). The predicted stroma classes remained an independent prognostic factor adjusting for clinicopathological variables including tumour size, stage, differentiation, and Lauren histology. In patients with stage II or III disease in predicted stroma classes one and two subgroups, patients who received adjuvant chemotherapy had improved survival compared with those who did not (in those with stage II disease hazard ratio [HR] 0·48 [95% CI 0·29-0·77], p=0·0021; and in those with stage III disease HR 0·70 [0·57-0·85], p=0·00042). However, in the other two subgroups adjuvant chemotherapy was not associated with survival and might even be detrimental in the predicted stroma class 4 subgroup (HR 1·48 [1·08-2·03], p=0·013). INTERPRETATION The deep-learning model could allow for accurate and non-invasive evaluation of tumour stroma from CT images in gastric cancer. The radiographical model predicted chemotherapy outcomes and could be used in combination with clinicopathological criteria to refine prognosis and inform treatment decisions of patients with gastric cancer. FUNDING None.
Collapse
Affiliation(s)
- Yuming Jiang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Xiaokun Liang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA; Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Shenzhen Colleges of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen, China
| | - Zhen Han
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Guangzhou, China
| | - Wei Wang
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Sujuan Xi
- The Reproductive Medical Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Tuanjie Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Guangzhou, China
| | - Chuanli Chen
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qingyu Yuan
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Na Li
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Shenzhen Colleges of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen, China
| | - Jiang Yu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Guangzhou, China
| | - Yaoqin Xie
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Shenzhen Colleges of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen, China
| | - Yikai Xu
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiwei Zhou
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - George A Poultsides
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Guoxin Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ruijiang Li
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
46
|
Dzobo K, Dandara C. Architecture of Cancer-Associated Fibroblasts in Tumor Microenvironment: Mapping Their Origins, Heterogeneity, and Role in Cancer Therapy Resistance. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2021; 24:314-339. [PMID: 32496970 DOI: 10.1089/omi.2020.0023] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The tumor stroma, a key component of the tumor microenvironment (TME), is a key determinant of response and resistance to cancer treatment. The stromal cells, extracellular matrix (ECM), and blood vessels influence cancer cell response to therapy and play key roles in tumor relapse and therapeutic outcomes. Of the stromal cells present in the TME, much attention has been given to cancer-associated fibroblasts (CAFs) as they are the most abundant and important in cancer initiation, progression, and therapy resistance. Besides releasing several factors, CAFs also synthesize the ECM, a key component of the tumor stroma. In this expert review, we examine the role of CAFs in the regulation of tumor cell behavior and reveal how CAF-derived factors and signaling influence tumor cell heterogeneity and development of novel strategies to combat cancer. Importantly, CAFs display both phenotypic and functional heterogeneity, with significant ramifications on CAF-directed therapies. Principal anti-cancer therapies targeting CAFs take the form of: (1) CAFs' ablation through use of immunotherapies, (2) re-education of CAFs to normalize the cells, (3) cellular therapies involving CAFs delivering drugs such as oncolytic adenoviruses, and (4) stromal depletion via targeting the ECM and its related signaling. The CAFs' heterogeneity could be a result of different cellular origins and the cancer-specific tumor microenvironmental effects, underscoring the need for further multiomics and biochemical studies on CAFs and the subsets. Lastly, we present recent advances in therapeutic targeting of CAFs and the success of such endeavors or their lack thereof. We recommend that to advance global public health and personalized medicine, treatments in the oncology clinic should be combinatorial in nature, strategically targeting both cancer cells and stromal cells, and their interactions.
Collapse
Affiliation(s)
- Kevin Dzobo
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa.,Division of Medical Biochemistry, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Collet Dandara
- Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
47
|
Baker AT, Abuwarwar MH, Poly L, Wilkins S, Fletcher AL. Cancer-Associated Fibroblasts and T Cells: From Mechanisms to Outcomes. THE JOURNAL OF IMMUNOLOGY 2021; 206:310-320. [PMID: 33397745 DOI: 10.4049/jimmunol.2001203] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 11/06/2020] [Indexed: 12/20/2022]
Abstract
Over the past decade, T cell immunotherapy has changed the face of cancer treatment, providing robust treatment options for several previously intractable cancers. Unfortunately, many epithelial tumors with high mortality rates respond poorly to immunotherapy, and an understanding of the key impediments is urgently required. Cancer-associated fibroblasts (CAFs) comprise the most frequent nonneoplastic cellular component in most solid tumors. Far from an inert scaffold, CAFs significantly influence tumor neogenesis, persistence, and metastasis and are emerging as a key player in immunotherapy resistance. In this review, we discuss the physical and chemical barriers that CAFs place between effector T cells and their tumor cell targets, and the therapies poised to target them.
Collapse
Affiliation(s)
- Alfie T Baker
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
| | - Mohammed H Abuwarwar
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
| | - Lylarath Poly
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
| | - Simon Wilkins
- Cabrini Monash University Department of Surgery, Cabrini Hospital, Malvern 3144, Victoria, Australia.,Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Melbourne 3004, Victoria, Australia; and
| | - Anne L Fletcher
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia; .,Institute of Immunology and Immunotherapy, University of Birmingham, Edgbaston B15 2TT, United Kingdom
| |
Collapse
|
48
|
Mehdi SJ, Moerman-Herzog A, Wong HK. Normal and cancer fibroblasts differentially regulate TWIST1, TOX and cytokine gene expression in cutaneous T-cell lymphoma. BMC Cancer 2021; 21:492. [PMID: 33941102 PMCID: PMC8091512 DOI: 10.1186/s12885-021-08142-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 04/02/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Mycosis fungoides (MF) is a primary cutaneous T-cell lymphoma (CTCL) that transforms from mature, skin-homing T cells and progresses during the early stages in the skin. The role of the skin microenvironment in MF development is unclear, but recent findings in a variety of cancers have highlighted the role of stromal fibroblasts in promoting or inhibiting tumorigenesis. Stromal fibroblasts are an important part of the cutaneous tumor microenvironment (TME) in MF. Here we describe studies into the interaction of TME-fibroblasts and malignant T cells to gain insight into their role in CTCL. METHODS Skin from normal (n = 3) and MF patients (n = 3) were analyzed for FAPα by immunohistochemistry. MyLa is a CTCL cell line that retains expression of biomarkers TWIST1 and TOX that are frequently detected in CTCL patients. MyLa cells were cultured in the presence or absence of normal or MF skin derived fibroblasts for 5 days, trypsinized to detached MyL a cells, and gene expression analyzed by RT-PCR for MF biomarkers (TWIST1 and TOX), Th1 markers (IFNG, TBX21), Th2 markers (GATA3, IL16), and proliferation marker (MKI67). Purified fibroblasts were assayed for VIM and ACTA2 gene expression. Cellular senescence assay was performed to assess senescence. RESULTS MF skin fibroblast showed increased expression of FAP-α with increasing stage compared to normal. Normal fibroblasts co-cultured with MyLa cells suppressed expression of TWIST1 (p < 0.0006), and TOX (p < 0.03), GATA3 (p < 0.02) and IL16 (p < 0.03), and increased expression of IFNG (p < 0.03) and TBX21 (p < 0.03) in MyLa cells. In contrast, MyLa cells cultured with MF fibroblasts retained high expression of TWIST1, TOX and GATA3. MF fibroblasts co-culture with MyLa cells increased expression of IL16 (p < 0.01) and IL4 (p < 0.02), and suppressed IFNG and TBX21 in MyLa cells. Furthermore, expression of MKI67 in MyLa cells was suppressed by normal fibroblasts compared to MF fibroblasts. CONCLUSION Skin fibroblasts represent important components of the TME in MF. In co-culture model, normal and MF fibroblasts have differential influence on T-cell phenotype in modulating expression of Th1 cytokine and CTCL biomarker genes to reveal distinct roles with implications in MF progression.
Collapse
Affiliation(s)
- Syed Jafar Mehdi
- Department of Dermatology, University of Arkansas for Medical Sciences, 4301 West Markham St, #576, Little Rock, AR, 72205, USA
| | - Andrea Moerman-Herzog
- Department of Dermatology, University of Arkansas for Medical Sciences, 4301 West Markham St, #576, Little Rock, AR, 72205, USA
| | - Henry K Wong
- Department of Dermatology, University of Arkansas for Medical Sciences, 4301 West Markham St, #576, Little Rock, AR, 72205, USA.
| |
Collapse
|
49
|
Zheng H, Bai Y, Wang J, Chen S, Zhang J, Zhu J, Liu Y, Wang X. Weighted Gene Co-expression Network Analysis Identifies CALD1 as a Biomarker Related to M2 Macrophages Infiltration in Stage III and IV Mismatch Repair-Proficient Colorectal Carcinoma. Front Mol Biosci 2021; 8:649363. [PMID: 33996905 PMCID: PMC8116739 DOI: 10.3389/fmolb.2021.649363] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 02/23/2021] [Indexed: 12/24/2022] Open
Abstract
Immunotherapy has achieved efficacy for advanced colorectal cancer (CRC) patients with a mismatch-repair-deficient (dMMR) subtype. However, little immunotherapy efficacy was observed in patients with the mismatch repair-proficient (pMMR) subtype, and hence, identifying new immune therapeutic targets is imperative for those patients. In this study, transcriptome data of stage III/IV CRC patients were retrieved from the Gene Expression Omnibus database. The CIBERSORT algorithm was used to quantify immune cellular compositions, and the results revealed that M2 macrophage fractions were higher in pMMR patients as compared with those with the dMMR subtype; moreover, pMMR patients with higher M2 macrophage fractions experienced shorter overall survival (OS). Subsequently, weighted gene co-expression network analysis and protein–protein interaction network analysis identified six hub genes related to M2 macrophage infiltrations in pMMR CRC patients: CALD1, COL6A1, COL1A2, TIMP3, DCN, and SPARC. Univariate and multivariate Cox regression analyses then determined CALD1 as the independent prognostic biomarker for OS. CALD1 was upregulated specifically the in CMS4 CRC subtype, and single-sample Gene Set Enrichment Analysis (ssGSEA) revealed that CALD1 was significantly correlated with angiogenesis and TGF-β signaling gene sets enrichment scores in stage III/IV pMMR CRC samples. The Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data (ESTIMATE) algorithm and correlation analysis revealed that CALD1 was significantly associated with multiple immune and stromal components in a tumor microenvironment. In addition, GSEA demonstrated that high expression of CALD1 was significantly correlated with antigen processing and presentation, chemokine signaling, leukocyte transendothelial migration, vascular smooth muscle contraction, cytokine–cytokine receptor interaction, cell adhesion molecules, focal adhesion, MAPK, and TGF-beta signaling pathways. Furthermore, the proliferation, invasion, and migration abilities of cancer cells were suppressed after reducing CALD1 expression in CRC cell lines. Taken together, multiple bioinformatics analyses and cell-level assays demonstrated that CALD1 could serve as a prognostic biomarker and a prospective therapeutic target for stage III/IV pMMR CRCs.
Collapse
Affiliation(s)
- Hang Zheng
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Yuge Bai
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Jingui Wang
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Shanwen Chen
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Junling Zhang
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Jing Zhu
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Yucun Liu
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Xin Wang
- Department of General Surgery, Peking University First Hospital, Beijing, China
| |
Collapse
|
50
|
Wilkinson K, Ng W, Roberts TL, Becker TM, Lim SHS, Chua W, Lee CS. Tumour immune microenvironment biomarkers predicting cytotoxic chemotherapy efficacy in colorectal cancer. J Clin Pathol 2021; 74:625-634. [PMID: 33753562 PMCID: PMC8461409 DOI: 10.1136/jclinpath-2020-207309] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 12/22/2020] [Indexed: 12/22/2022]
Abstract
The role of the local tumour and stromal immune landscape is increasingly recognised to be important in cancer development, progression and response to therapy. The composition, function, spatial orientation and gene expression profile of the infiltrate of the innate and adaptive immune system at the tumour and surrounding tissue has an established prognostic role in colorectal cancer (CRC). Multiple studies have confirmed that a tumour immune microenvironment (TIME) reflective of a type 1 adaptive immune response is associated with improved prognosis. There have been significant efforts to evolve these observations into validated, histopathology-based prognostic biomarkers, such as the Immunoscore. However, the clinical need lies much more in the development of predictive, not prognostic, biomarkers which have the potential to improve patient outcomes. This is particularly pertinent to help guide cytotoxic chemotherapy use in CRC, which remains the standard of care. Cytotoxic chemotherapy has recognised immunomodulatory activity distinct from its antimitotic effects, including mechanisms such as immunogenic cell death (ICD) and induction/inhibition of key immune players. Response to chemotherapy may differ with regard to molecular subtype of CRC, which are strongly associated with immune phenotypes. Thus, immune markers are potentially useful, though under-reported, predictive biomarkers. In this review, we discuss the impact of the TIME on response to cytotoxic chemotherapy in CRC, with a focus on baseline immune markers, and associated genomic and transcriptomic signatures.
Collapse
Affiliation(s)
- Kate Wilkinson
- Liverpool Cancer Therapy Centre, Liverpool Hospital, Liverpool, New South Wales, Australia .,School of Medicine, Western Sydney University, Liverpool, New South Wales, Australia
| | - Weng Ng
- Liverpool Cancer Therapy Centre, Liverpool Hospital, Liverpool, New South Wales, Australia.,School of Medicine, Western Sydney University, Liverpool, New South Wales, Australia
| | - Tara Laurine Roberts
- School of Medicine, Western Sydney University, Liverpool, New South Wales, Australia.,Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
| | - Therese M Becker
- School of Medicine, Western Sydney University, Liverpool, New South Wales, Australia.,Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
| | - Stephanie Hui-Su Lim
- School of Medicine, Western Sydney University, Liverpool, New South Wales, Australia.,Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia.,Macarthur Cancer Therapy Centre, Campbelltown Hospital, Campbelltown, New South Wales, Australia
| | - Wei Chua
- Liverpool Cancer Therapy Centre, Liverpool Hospital, Liverpool, New South Wales, Australia.,School of Medicine, Western Sydney University, Liverpool, New South Wales, Australia.,Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
| | - Cheok Soon Lee
- School of Medicine, Western Sydney University, Liverpool, New South Wales, Australia.,Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia.,Department of Anatomical Pathology, Liverpool Hospital, Liverpool, New South Wales, Australia
| |
Collapse
|