1
|
Gu Y, Taifour T, Bui T, Zuo D, Pacis A, Poirier A, Attalla S, Fortier AM, Sanguin-Gendreau V, Pan TC, Papavasiliou V, Lin NU, Hughes ME, Smith K, Park M, Tremblay ML, Chodosh LA, Jeselsohn R, Muller WJ. Osteopontin is a therapeutic target that drives breast cancer recurrence. Nat Commun 2024; 15:9174. [PMID: 39448577 PMCID: PMC11502809 DOI: 10.1038/s41467-024-53023-9] [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: 04/05/2024] [Accepted: 09/29/2024] [Indexed: 10/26/2024] Open
Abstract
Recurrent breast cancers often develop resistance to standard-of-care therapies. Identifying targetable factors contributing to cancer recurrence remains the rate-limiting step in improving long-term outcomes. In this study, we identify tumor cell-derived osteopontin as an autocrine and paracrine driver of tumor recurrence. Osteopontin promotes tumor cell proliferation, recruits macrophages, and synergizes with IL-4 to further polarize them into a pro-tumorigenic state. Macrophage depletion and osteopontin inhibition decrease recurrent tumor growth. Furthermore, targeting osteopontin in primary tumor-bearing female mice prevents metastasis, permits T cell infiltration and activation, and improves anti-PD-1 immunotherapy response. Clinically, osteopontin expression is higher in recurrent metastatic tumors versus female patient-matched primary breast tumors. Osteopontin positively correlates with macrophage infiltration, increases with higher tumor grade, and its elevated pathway activity is associated with poor prognosis and long-term recurrence. Our findings suggest clinical implications and an alternative therapeutic strategy based on osteopontin's multiaxial role in breast cancer progression and recurrence.
Collapse
Affiliation(s)
- Yu Gu
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Tarek Taifour
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Division of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Tung Bui
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Dongmei Zuo
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Alain Pacis
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Canadian Centre for Computational Genomics, McGill University Genome Center, Montreal, QC, Canada
| | - Alexandre Poirier
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Division of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Sherif Attalla
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Anne-Marie Fortier
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | | | - Tien-Chi Pan
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Vasilios Papavasiliou
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Nancy U Lin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Melissa E Hughes
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kalie Smith
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Morag Park
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Michel L Tremblay
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- Division of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Lewis A Chodosh
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rinath Jeselsohn
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - William J Muller
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada.
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada.
- Faculty of Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
2
|
Wang XR, Zhou XH, Sun XT, Shen YQ, Wu YY, Wu CD, Zhu FJ, Wei YT, Chen JP, Chen J, Zheng SY, Wang LX. Tumour cell-released autophagosomes promote lung metastasis by upregulating PD-L1 expression in pulmonary vascular endothelial cells in breast cancer. Cell Oncol (Dordr) 2024:10.1007/s13402-024-00994-y. [PMID: 39373859 DOI: 10.1007/s13402-024-00994-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2024] [Indexed: 10/08/2024] Open
Abstract
PURPOSE Establishing an immunosuppressive premetastatic niche (PMN) in distant organs is crucial for breast cancer metastasis. Vascular endothelial cells (VECs) act as barriers to transendothelial cell migration. However, the immune functions of PMNs remain unclear. Tumour cell-released autophagosomes (TRAPs) are critical modulators of antitumour immune responses. Herein, we investigated the mechanism through which TRAPs modulate the immune function of pulmonary VECs in lung PMN in breast cancer. METHODS Immortalised mouse pulmonary microvascular endothelial cells were incubated with TRAPs in vitro. RNA sequencing, flow cytometry, and western blotting were employed to assess immunosuppressive function and mechanism. In vivo, TRAP-trained and autophagy-deficient tumour mice were used to detect immunosuppression, and high-mobility group box 1 (HMGB1)-deficient TRAP-trained and TLR4 knockout mice were utilised to investigate the underlying mechanisms of pulmonary VECs. Additionally, the efficacy of anti-programmed cell death ligand-1 (PD-L1) immunotherapy was evaluated in early tumour-bearing mice. RESULTS HMGB1 on TRAPs surfaces stimulated VECs to upregulate PD-L1 via a TLR4-MyD88-p38/STAT3 signalling cascade that depended on the cytoskeletal movement of VECs. Importantly, PD-L1 on TRAP-induced VECs can inhibit T cell function, promote lung PMN immunosuppression, and result in more pronounced lung metastasis. Treatment with anti-PD-L1 reduces lung metastasis in early stage tumour-bearing mice. CONCLUSIONS These findings revealed a novel role and mechanism of TRAP-induced immunosuppression of pulmonary VECs in lung PMN. TRAPs and their surface HMGB1 are important therapeutic targets for reversing immunosuppression, providing a new theoretical basis for the treatment of early stage breast cancer using an anti-PD-L1 antibody.
Collapse
Affiliation(s)
- Xu-Ru Wang
- Department of Microbiology and Immunology, School of Medicine, Jiangsu Provincial Key Laboratory of Critical Care Medicine, Southeast University, Nanjing, 210009, China
| | - Xiao-He Zhou
- Department of Microbiology and Immunology, School of Medicine, Jiangsu Provincial Key Laboratory of Critical Care Medicine, Southeast University, Nanjing, 210009, China
| | - Xiao-Tong Sun
- Department of Laboratory Medicine, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, 264000, China
| | - Yu-Qing Shen
- Department of Microbiology and Immunology, School of Medicine, Jiangsu Provincial Key Laboratory of Critical Care Medicine, Southeast University, Nanjing, 210009, China
| | - Yu-Yang Wu
- Department of Microbiology and Immunology, School of Medicine, Jiangsu Provincial Key Laboratory of Critical Care Medicine, Southeast University, Nanjing, 210009, China
| | - Cheng-Dong Wu
- Department of Microbiology and Immunology, School of Medicine, Jiangsu Provincial Key Laboratory of Critical Care Medicine, Southeast University, Nanjing, 210009, China
| | - Feng-Jiao Zhu
- Department of Microbiology and Immunology, School of Medicine, Jiangsu Provincial Key Laboratory of Critical Care Medicine, Southeast University, Nanjing, 210009, China
| | - Yi-Ting Wei
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Jin-Peng Chen
- Department of General Surgery, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Jing Chen
- Department of Microbiology and Immunology, School of Medicine, Jiangsu Provincial Key Laboratory of Critical Care Medicine, Southeast University, Nanjing, 210009, China
| | - Shi-Ya Zheng
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Li-Xin Wang
- Department of Microbiology and Immunology, School of Medicine, Jiangsu Provincial Key Laboratory of Critical Care Medicine, Southeast University, Nanjing, 210009, China.
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China.
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, Jiangsu Province, 210009, China.
| |
Collapse
|
3
|
Naito K, Sangai T, Yamashita K. CAF-Associated Genes in Breast Cancer for Novel Therapeutic Strategies. Biomedicines 2024; 12:1964. [PMID: 39335478 PMCID: PMC11428270 DOI: 10.3390/biomedicines12091964] [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: 06/25/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/30/2024] Open
Abstract
Breast cancer (BC) is the most common cancer in women, and therapeutic strategies for it are based on the molecular subtypes of luminal BC, HER2 BC, and triple-negative BC (TNBC) because each subtype harbors different unique genetic aberrations. Recently, features of the tumor microenvironment (TME), especially cancer-associated fibroblasts (CAFs), have been demonstrated to play a critical role in BC progression, and we would like to understand the molecular features of BC CAFs for novel therapeutic strategies. In a recent study, 115 CAF-associated genes (CAFGs) were identified in a public database of microdissection and microarray data (GSE35602) from 13 colorectal cancer (CRC) tumors. Using a public database (GSE10797) of 28 BC tumors, a similar analysis was performed. In BC, 59 genes from the 115 CAFGs identified in CRC (CRC CAFGs) were also closely associated with a CAFs marker, SPARC (R = 0.6 or beyond), and POSTN was of particular interest as one of the BC CAFGs with the highest expression levels and a close association with SPARC expression (R = 0.94) in the cancer stroma of BC tumors. In BC stroma, POSTN was followed in expression levels by DKK3, MMP2, PDPN, and ACTA2. Unexpectedly, FAP and VIM were not as highly associated with SPARC expression in the cancer stroma of BC tumors and exhibited low expression. These findings suggested that ACTA2 might be the most relevant conventional CAFs marker in BC, and ACTA2 was actually correlated in expression with many CRC CAFGs, such as SPARC. Surprisingly, the SE ratio values of the BC CAFGs were much lower (average SE = 3.8) than those of the CRC CAFGs (SE = 10 or beyond). We summarized the current understanding of BC CAFs from the literature. Finally, in triple-negative BC (TNBC) (n = 5), SPARC expression uniquely showed a close association with COL11A1 and TAGLN expression, representing a myofibroblast (myCAFs) marker in the cancer stroma of the BC tumors, suggesting that myCAFs may be molecularly characterized by TNBC in contrast to other BC phenotypes. In summary, CAFs could have unique molecular characteristics in BC, and such TME uniqueness could be therapeutically targeted in BC.
Collapse
Affiliation(s)
- Kanako Naito
- Division of Advanced Surgical Oncology, Research and Development Center for New Medical Frontiers, Kitasato University School of Medicine, Sagamihara 252-0374, Japan
| | - Takafumi Sangai
- Department of Breast and Thyroid Surgery, Kitasato University School of Medicine, Sagamihara 252-0374, Japan
| | - Keishi Yamashita
- Division of Advanced Surgical Oncology, Research and Development Center for New Medical Frontiers, Kitasato University School of Medicine, Sagamihara 252-0374, Japan
| |
Collapse
|
4
|
Wang Y, Ding W, Hao W, Gong L, Peng Y, Zhang J, Qian Z, Xu K, Cai W, Gao Y. CXCL3/TGF-β-mediated crosstalk between CAFs and tumor cells augments RCC progression and sunitinib resistance. iScience 2024; 27:110224. [PMID: 39040058 PMCID: PMC11261419 DOI: 10.1016/j.isci.2024.110224] [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: 10/25/2023] [Revised: 03/17/2024] [Accepted: 06/06/2024] [Indexed: 07/24/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs) play a significant role in tumor development and treatment failure, yet the precise mechanisms underlying their contribution to renal cell carcinoma (RCC) remains underexplored. This study explored the interaction between CAFs and tumor cells, and related mechanisms. CAFs isolated from tumor tissues promoted the tumor progression and drugs resistance both in vivo and in vitro. Mechanistically, chemokine (C-X-C motif) ligand (CXCL) 3 secreted from CAFs mediated its effects. CXCL3 activated its receptor CXCR2 to active the downstream ERK1/2 signaling pathway, subsequently promoting epithelial-mesenchymal transition and cell stemness. Blocking the crosstalk between CAFs and tumor cells by CXCR2 inhibitor SB225002 attenuated the functions of CAFs. Furthermore, Renca cells facilitated the transformation of normal interstitial fibroblasts (NFs) into CAFs and the expression of CXCL3 through TGF-β-Smad2/3 signaling pathway. In turn, transformed NFs promoted the tumor progression and drug resistance of RCC. These findings may constitute potential therapeutic strategies for RCC treatment.
Collapse
Affiliation(s)
- Yunxia Wang
- School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Weihong Ding
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Wenjing Hao
- School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Luyao Gong
- School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yeheng Peng
- School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jun Zhang
- School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Zhiyu Qian
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ke Xu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Weimin Cai
- School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yuan Gao
- School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
5
|
Panda VK, Mishra B, Nath AN, Butti R, Yadav AS, Malhotra D, Khanra S, Mahapatra S, Mishra P, Swain B, Majhi S, Kumari K, Radharani NNV, Kundu GC. Osteopontin: A Key Multifaceted Regulator in Tumor Progression and Immunomodulation. Biomedicines 2024; 12:1527. [PMID: 39062100 PMCID: PMC11274826 DOI: 10.3390/biomedicines12071527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 06/22/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
The tumor microenvironment (TME) is composed of various cellular components such as tumor cells, stromal cells including fibroblasts, adipocytes, mast cells, lymphatic vascular cells and infiltrating immune cells, macrophages, dendritic cells and lymphocytes. The intricate interplay between these cells influences tumor growth, metastasis and therapy failure. Significant advancements in breast cancer therapy have resulted in a substantial decrease in mortality. However, existing cancer treatments frequently result in toxicity and nonspecific side effects. Therefore, improving targeted drug delivery and increasing the efficacy of drugs is crucial for enhancing treatment outcome and reducing the burden of toxicity. In this review, we have provided an overview of how tumor and stroma-derived osteopontin (OPN) plays a key role in regulating the oncogenic potential of various cancers including breast. Next, we dissected the signaling network by which OPN regulates tumor progression through interaction with selective integrins and CD44 receptors. This review addresses the latest advancements in the roles of splice variants of OPN in cancer progression and OPN-mediated tumor-stromal interaction, EMT, CSC enhancement, immunomodulation, metastasis, chemoresistance and metabolic reprogramming, and further suggests that OPN might be a potential therapeutic target and prognostic biomarker for the evolving landscape of cancer management.
Collapse
Affiliation(s)
- Venketesh K. Panda
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (A.N.N.); (D.M.); (S.K.); (S.M.); (P.M.); (B.S.); (S.M.); (K.K.)
| | - Barnalee Mishra
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (A.N.N.); (D.M.); (S.K.); (S.M.); (P.M.); (B.S.); (S.M.); (K.K.)
| | - Angitha N. Nath
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (A.N.N.); (D.M.); (S.K.); (S.M.); (P.M.); (B.S.); (S.M.); (K.K.)
| | - Ramesh Butti
- Division of Hematology and Oncology, Department of Internal Medicine, Southwestern Medical Center, University of Texas, Dallas, TX 75235, USA;
| | - Amit Singh Yadav
- Biomedical Centre, Faculty of Medicine, Lund University, 223 62 Lund, Sweden; (A.S.Y.); (N.N.V.R.)
| | - Diksha Malhotra
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (A.N.N.); (D.M.); (S.K.); (S.M.); (P.M.); (B.S.); (S.M.); (K.K.)
| | - Sinjan Khanra
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (A.N.N.); (D.M.); (S.K.); (S.M.); (P.M.); (B.S.); (S.M.); (K.K.)
| | - Samikshya Mahapatra
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (A.N.N.); (D.M.); (S.K.); (S.M.); (P.M.); (B.S.); (S.M.); (K.K.)
| | - Priyanka Mishra
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (A.N.N.); (D.M.); (S.K.); (S.M.); (P.M.); (B.S.); (S.M.); (K.K.)
| | - Biswajit Swain
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (A.N.N.); (D.M.); (S.K.); (S.M.); (P.M.); (B.S.); (S.M.); (K.K.)
| | - Sambhunath Majhi
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (A.N.N.); (D.M.); (S.K.); (S.M.); (P.M.); (B.S.); (S.M.); (K.K.)
| | - Kavita Kumari
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (A.N.N.); (D.M.); (S.K.); (S.M.); (P.M.); (B.S.); (S.M.); (K.K.)
| | - N. N. V. Radharani
- Biomedical Centre, Faculty of Medicine, Lund University, 223 62 Lund, Sweden; (A.S.Y.); (N.N.V.R.)
| | - Gopal C. Kundu
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (A.N.N.); (D.M.); (S.K.); (S.M.); (P.M.); (B.S.); (S.M.); (K.K.)
- Kalinga Institute of Medical Sciences (KIMS), KIIT Deemed to be University, Bhubaneswar 751024, India
| |
Collapse
|
6
|
Talia M, Cesario E, Cirillo F, Scordamaglia D, Di Dio M, Zicarelli A, Mondino AA, Occhiuzzi MA, De Francesco EM, Belfiore A, Miglietta AM, Di Dio M, Capalbo C, Maggiolini M, Lappano R. Cancer-associated fibroblasts (CAFs) gene signatures predict outcomes in breast and prostate tumor patients. J Transl Med 2024; 22:597. [PMID: 38937754 PMCID: PMC11210052 DOI: 10.1186/s12967-024-05413-2] [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: 05/29/2024] [Accepted: 06/17/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND Over the last two decades, tumor-derived RNA expression signatures have been developed for the two most commonly diagnosed tumors worldwide, namely prostate and breast tumors, in order to improve both outcome prediction and treatment decision-making. In this context, molecular signatures gained by main components of the tumor microenvironment, such as cancer-associated fibroblasts (CAFs), have been explored as prognostic and therapeutic tools. Nevertheless, a deeper understanding of the significance of CAFs-related gene signatures in breast and prostate cancers still remains to be disclosed. METHODS RNA sequencing technology (RNA-seq) was employed to profile and compare the transcriptome of CAFs isolated from patients affected by breast and prostate tumors. The differentially expressed genes (DEGs) characterizing breast and prostate CAFs were intersected with data from public datasets derived from bulk RNA-seq profiles of breast and prostate tumor patients. Pathway enrichment analyses allowed us to appreciate the biological significance of the DEGs. K-means clustering was applied to construct CAFs-related gene signatures specific for breast and prostate cancer and to stratify independent cohorts of patients into high and low gene expression clusters. Kaplan-Meier survival curves and log-rank tests were employed to predict differences in the outcome parameters of the clusters of patients. Decision-tree analysis was used to validate the clustering results and boosting calculations were then employed to improve the results obtained by the decision-tree algorithm. RESULTS Data obtained in breast CAFs allowed us to assess a signature that includes 8 genes (ITGA11, THBS1, FN1, EMP1, ITGA2, FYN, SPP1, and EMP2) belonging to pro-metastatic signaling routes, such as the focal adhesion pathway. Survival analyses indicated that the cluster of breast cancer patients showing a high expression of the aforementioned genes displays worse clinical outcomes. Next, we identified a prostate CAFs-related signature that includes 11 genes (IL13RA2, GDF7, IL33, CXCL1, TNFRSF19, CXCL6, LIFR, CXCL5, IL7, TSLP, and TNFSF15) associated with immune responses. A low expression of these genes was predictive of poor survival rates in prostate cancer patients. The results obtained were significantly validated through a two-step approach, based on unsupervised (clustering) and supervised (classification) learning techniques, showing a high prediction accuracy (≥ 90%) in independent RNA-seq cohorts. CONCLUSION We identified a huge heterogeneity in the transcriptional profile of CAFs derived from breast and prostate tumors. Of note, the two novel CAFs-related gene signatures might be considered as reliable prognostic indicators and valuable biomarkers for a better management of breast and prostate cancer patients.
Collapse
Affiliation(s)
- Marianna Talia
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, 87036, Italy
| | - Eugenio Cesario
- Department of Cultures, Education and Society, University of Calabria, Rende, 87036, Italy
| | - Francesca Cirillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, 87036, Italy
| | - Domenica Scordamaglia
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, 87036, Italy
| | - Marika Di Dio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, 87036, Italy
| | - Azzurra Zicarelli
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, 87036, Italy
| | - Adelina Assunta Mondino
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, 87036, Italy
| | | | | | - Antonino Belfiore
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, Catania, 95122, Italy
| | - Anna Maria Miglietta
- Breast and General Surgery Unit, Annunziata Hospital Cosenza, Cosenza, 87100, Italy
| | - Michele Di Dio
- Division of Urology, Department of Surgery, Annunziata Hospital, Cosenza, 87100, Italy
| | - Carlo Capalbo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, 87036, Italy
- Complex Operative Oncology Unit, Annunziata Hospital Cosenza, Cosenza, 87100, Italy
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, 87036, Italy.
| | - Rosamaria Lappano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, 87036, Italy.
| |
Collapse
|
7
|
Akinpelu A, Akinsipe T, Avila LA, Arnold RD, Mistriotis P. The impact of tumor microenvironment: unraveling the role of physical cues in breast cancer progression. Cancer Metastasis Rev 2024; 43:823-844. [PMID: 38238542 PMCID: PMC11156564 DOI: 10.1007/s10555-024-10166-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 01/02/2024] [Indexed: 01/30/2024]
Abstract
Metastasis accounts for the vast majority of breast cancer-related fatalities. Although the contribution of genetic and epigenetic modifications to breast cancer progression has been widely acknowledged, emerging evidence underscores the pivotal role of physical stimuli in driving breast cancer metastasis. In this review, we summarize the changes in the mechanics of the breast cancer microenvironment and describe the various forces that impact migrating and circulating tumor cells throughout the metastatic process. We also discuss the mechanosensing and mechanotransducing molecules responsible for promoting the malignant phenotype in breast cancer cells. Gaining a comprehensive understanding of the mechanobiology of breast cancer carries substantial potential to propel progress in prognosis, diagnosis, and patient treatment.
Collapse
Affiliation(s)
- Ayuba Akinpelu
- Department of Chemical Engineering, Samuel Ginn College of Engineering, Auburn University, Auburn, AL, 36849, USA
| | - Tosin Akinsipe
- Department of Biological Sciences, College of Science and Mathematics, Auburn University, Auburn, AL, 36849, USA
| | - L Adriana Avila
- Department of Biological Sciences, College of Science and Mathematics, Auburn University, Auburn, AL, 36849, USA
| | - Robert D Arnold
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL, 36849, USA
| | - Panagiotis Mistriotis
- Department of Chemical Engineering, Samuel Ginn College of Engineering, Auburn University, Auburn, AL, 36849, USA.
| |
Collapse
|
8
|
Lv Q, Zhang Z, Fu H, Li D, Liu Y, Sun Y, Wu M. Predictive Panel for Immunotherapy in Low-Grade Glioma. World Neurosurg 2024; 183:e825-e837. [PMID: 38216032 DOI: 10.1016/j.wneu.2024.01.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/14/2024]
Abstract
BACKGROUND The main treatment of low-grade glioma (LGG) is still surgical resection followed by radiotherapy and/or chemotherapy, which has certain limitations, including side effects and drug resistance. Immunotherapy is a promising treatment for LGG, but it is generally hindered by the tumor microenvironment with the limited expression of tumor antigens. METHODS We integrated RNA sequencing data sets and clinical information and conducted consistent cluster analysis to explore the most suitable patients for immune checkpoint therapy. Gene set enrichment analysis, UMAP analysis, mutation correlation analysis, TIMER analysis, and TIDE analysis were used to identify the immune characteristics of 3 immune subtypes and the feasibility of 5 antigens as immune checkpoint markers. RESULTS We analyzed the isolation and mutation of homologous recombination repair genes (HRR) of the 3 immune subtypes, and the HRR genes of the 3 subtypes were obviously segregated. Among them, the IS2 subtype has a large number of HRR gene mutations, which increases the immunogenicity of tumors-this is consistent with the results of tumor mutation load analysis of 3 immune subtypes. Then we evaluated the immune cell infiltration of immune subtypes and found that IS2 and IS3 subtypes were rich in immune cells. It is worth noting that there are many Treg cells and NK cells in the IS1 subtype. In addition, when analyzing the immune checkpoint gene expression of the 3 subtypes, we found that they were upregulated most in IS2 subtypes compared with other subtypes. Then when we further confirmed the role of immune-related genes in LGG; through TIDE analysis and TISIDB analysis, we obtained 5 markers that can predict the efficacy of ICB in patients with LGG. In addition, we confirmed that they were associated with poor prognosis through survival analysis. CONCLUSIONS We obtained 3 reliable immune subtypes, and patients with the IS2 subtype are suitable for immunotherapy, in which NAMPT, SLC11A1, TNC, VIM, and SPP1 are predictive panel markers for ICB in the LGG group. Our findings provide a rationale for immunotherapy selection and prediction of patient prognosis in LGG patients.
Collapse
Affiliation(s)
- Qingqing Lv
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Zhaoyu Zhang
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Haijuan Fu
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Danyang Li
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yihao Liu
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yingnan Sun
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Minghua Wu
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.
| |
Collapse
|
9
|
Ghosh A, Mitra AK. Metastasis and cancer associated fibroblasts: taking it up a NOTCH. Front Cell Dev Biol 2024; 11:1277076. [PMID: 38269089 PMCID: PMC10806909 DOI: 10.3389/fcell.2023.1277076] [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: 08/14/2023] [Accepted: 12/27/2023] [Indexed: 01/26/2024] Open
Abstract
Metastasis is the least understood aspect of cancer biology. 90% of cancer related deaths occur due extensive metastatic burden in patients. Apart from metastasizing cancer cells, the pro-tumorigenic and pro-metastatic role of the tumor stroma plays a crucial part in this complex process often leading to disease relapse and therapy resistance. Cellular signaling processes play a crucial role in the process of tumorigenesis and metastasis when aberrantly turned on, not just in the cancer cells, but also in the cells of the tumor microenvironment (TME). One of the most conserved pathways includes the Notch signaling pathway that plays a crucial role in the development and progression of many cancers. In addition to its well documented role in cancer cells, recent evidence suggests crucial involvement of Notch signaling in the stroma as well. This review aims to highlight the current findings focusing on the oncogenic role of notch signaling in cancer cells and the TME, with a specific focus on cancer associated fibroblasts (CAFs), which constitute a major part of the tumor stroma and are important for tumor progression. Recent efforts have focused on the development of anti-cancer and anti-metastatic therapies targeting TME. Understanding the importance of Notch signaling in the TME would help identify important drivers for stromal reprogramming, metastasis and importantly, drive future research in the effort to develop TME-targeted therapies utilizing Notch.
Collapse
Affiliation(s)
- Argha Ghosh
- Indiana University School of Medicine-Bloomington, Bloomington, IN, United States
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, United States
| | - Anirban K. Mitra
- Indiana University School of Medicine-Bloomington, Bloomington, IN, United States
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, United States
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
10
|
Poleboyina PK, Alagumuthu M, Pasha A, Ravinder D, Pasumarthi D, Pawar SC. Entrectinib a Plausible Inhibitor for Osteopontin (SPP1) in Cervical Cancer-Integrated Bioinformatic Approach. Appl Biochem Biotechnol 2023; 195:7766-7795. [PMID: 37086377 DOI: 10.1007/s12010-023-04541-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2023] [Indexed: 04/23/2023]
Abstract
Cervical cancer is one of the major causes of death in women, especially in developing countries bearing more than a quarter of the global burden. Secreted phosphoprotein-1, also known as OPN (osteopontin), is an integrin-binding glycophosphoprotein that is overexpressed in a variety of tumors. OPN is a chemokine-like calcified ECM-associated protein that plays a crucial role in evaluating the metastatic potential of various cancers. However, the role of SPP1 in the tumor microenvironment and associated signaling pathways in CC is still unclear. In our study, three CC microarray datasets (GSE9750, GSE46857, and GSE67522) were obtained from the GEO database to identify the differentially expressed genes. Enrichment analysis was carried out by Enrichr and ShinyGO and the PPI interaction network was created by using String and Cytoscape. GEPIA datasets were used to validate the top 10 hub genes, and virtual screening, docking, and dynamic simulation studies were used to identify a suitable inhibitor against the OPN protein using MVD, PyRx, and GROMACS respectively. Our results show that a total of 11 DEGs were common for three datasets and gene ontology pathway enrichment analysis revealed that 2 biological processes i.e. programmed cell death and animal organ development commonly affected mechanisms in all three datasets. Docking and dynamic studies revealed that Entrectinib showed excellent binding affinity against OPN protein. Based on the results, we conclude that OPN is one of the most upregulated genes in cervical cancer and Entrectinib emerges to be a promising potential OPN inhibitor to curtail cervical cancer progression. Schematic representation: The schematic representation of methodology steps is illustrated in the graphical abstract. Schematic representation of methodology.
Collapse
Affiliation(s)
- Pavan Kumar Poleboyina
- Department of Genetics & Biotechnology, University College of Science, Osmania University, -500007, Hyderabad, Telangana, India
| | - Manikandan Alagumuthu
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, -632014, Vellore, India
| | - Akbar Pasha
- Department of Genetics & Biotechnology, University College of Science, Osmania University, -500007, Hyderabad, Telangana, India
| | - Doneti Ravinder
- Department of Genetics & Biotechnology, University College of Science, Osmania University, -500007, Hyderabad, Telangana, India
| | - Deepthi Pasumarthi
- Department of Genetics & Biotechnology, University College of Science, Osmania University, -500007, Hyderabad, Telangana, India
| | - Smita C Pawar
- Department of Genetics & Biotechnology, University College of Science, Osmania University, -500007, Hyderabad, Telangana, India.
| |
Collapse
|
11
|
Chen C, Guo Q, Liu Y, Hou Q, Liao M, Guo Y, Zang Y, Wang F, Liu H, Luan X, Liang Y, Guan Z, Li Y, Liu H, Dong X, Zhang X, Liu J, Xu Q. Single-cell and spatial transcriptomics reveal POSTN + cancer-associated fibroblasts correlated with immune suppression and tumour progression in non-small cell lung cancer. Clin Transl Med 2023; 13:e1515. [PMID: 38115703 PMCID: PMC10731139 DOI: 10.1002/ctm2.1515] [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: 06/01/2023] [Revised: 11/27/2023] [Accepted: 12/01/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) are potential targets for cancer therapy. Due to the heterogeneity of CAFs, the influence of CAF subpopulations on the progression of lung cancer is still unclear, which impedes the translational advances in targeting CAFs. METHODS We performed single-cell RNA sequencing (scRNA-seq) on tumour, paired tumour-adjacent, and normal samples from 16 non-small cell lung cancer (NSCLC) patients. CAF subpopulations were analyzed after integration with published NSCLC scRNA-seq data. SpaTial enhanced resolution omics-sequencing (Stereo-seq) was applied in tumour and tumour-adjacent samples from seven NSCLC patients to map the architecture of major cell populations in tumour microenvironment (TME). Immunohistochemistry (IHC) and multiplexed IHC (mIHC) were used to validate marker gene expression and the association of CAFs with immune infiltration in TME. RESULTS A subcluster of myofibroblastic CAFs, POSTN+ CAFs, were significantly enriched in advanced tumours and presented gene expression signatures related to extracellular matrix remodeling, tumour invasion pathways and immune suppression. Stereo-seq and mIHC demonstrated that POSTN+ CAFs were in close localization with SPP1+ macrophages and were associated with the exhausted phenotype and lower infiltration of T cells. POSTN expression or the abundance of POSTN+ CAFs were associated with poor prognosis of NSCLC. CONCLUSIONS Our study identified a myofibroblastic CAF subpopulation, POSTN+ CAFs, which might associate with SPP1+ macrophages to promote the formation of desmoplastic architecture and participate in immune suppression. Furthermore, we showed that POSTN+ CAFs associated with cancer progression and poor clinical outcomes and may provide new insights on the treatment of NSCLC.
Collapse
Affiliation(s)
- Chao Chen
- Department of Thoracic SurgeryPeking University Shenzhen HospitalShenzhen Peking University‐The Hong Kong University of Science and Technology Medical CenterShenzhenChina
| | - Qiang Guo
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Yang Liu
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Qinghua Hou
- Department of Thoracic SurgeryPeking University Shenzhen HospitalShenzhen Peking University‐The Hong Kong University of Science and Technology Medical CenterShenzhenChina
| | - Mengying Liao
- Department of PathologyPeking University Shenzhen HospitalShenzhenChina
| | - Yanying Guo
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
| | - Yupeng Zang
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | | | - Huanyu Liu
- Department of PathologyPeking University Shenzhen HospitalShenzhenChina
| | - Xinyu Luan
- Department of Thoracic SurgeryPeking University Shenzhen HospitalShenzhen Peking University‐The Hong Kong University of Science and Technology Medical CenterShenzhenChina
| | - Yanling Liang
- BGI ResearchShenzhenChina
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Zhuojue Guan
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Yanling Li
- Central Laboratory of Peking University Shenzhen HospitalShenzhenChina
| | - Haozhen Liu
- Department of Thoracic SurgeryPeking University Shenzhen HospitalShenzhen Peking University‐The Hong Kong University of Science and Technology Medical CenterShenzhenChina
| | - Xuan Dong
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
- Guangdong Provincial Key Laboratory of Human Disease Genomics, Shenzhen Key Laboratory of GenomicsBGI ResearchShenzhenChina
| | - Xiuqing Zhang
- BGI ResearchShenzhenChina
- Guangdong Provincial Key Laboratory of Human Disease Genomics, Shenzhen Key Laboratory of GenomicsBGI ResearchShenzhenChina
| | - Jixian Liu
- Department of Thoracic SurgeryPeking University Shenzhen HospitalShenzhen Peking University‐The Hong Kong University of Science and Technology Medical CenterShenzhenChina
| | - Qumiao Xu
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
- Guangdong Provincial Key Laboratory of Human Disease Genomics, Shenzhen Key Laboratory of GenomicsBGI ResearchShenzhenChina
| |
Collapse
|
12
|
Zhang Y, Li S, Cui X, Wang Y. microRNA-944 inhibits breast cancer cell proliferation and promotes cell apoptosis by reducing SPP1 through inactivating the PI3K/Akt pathway. Apoptosis 2023; 28:1546-1563. [PMID: 37486406 DOI: 10.1007/s10495-023-01870-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2023] [Indexed: 07/25/2023]
Abstract
Breast cancer is a common malignancy in women with poor prognosis. This study aimed to investigate the molecular mechanism of microRNA-944 (miR-944) mediated secreted phosphoprotein-1 (SPP1) in breast cancer progression and its regulatory effect on the phosphoinositide 3-kinase (PI3K)/Akt signaling pathway. Differential gene analysis was performed to identify key genes associated with breast cancer development by screening breast cancer-related microarray data. The expression of miR-944 and SPP1 and their relationship were determined in clinical samples and cells. sh-SPP1, oe-SPP1, LY294002 or miR-944 mimic were transfected into MCF-7 cells to investigate the role of miR-944 mediated SPP1 in breast cancer development and its regulatory effect on the PI3K/Akt pathway. Finally, the tumorigenicity of breast cancer cells was observed in nude mice. Through bioinformatics analysis, we identified SPP1 as a key gene in breast cancer, and miR-944 as an upstream miRNA of SPP1. In breast cancer tissues and cells, the expression of miR-944 was decreased while that of SPP1 was increased. miR-944 negatively regulated the expression of SPP1. In breast cancer cells, SPP1 activated the PI3K/Akt pathway to promote cell proliferation and inhibit apoptosis. In vitro cell experiments showed that the downregulation of miR-944 promoted the high expression of SPP1, which then activated the PI3K/Akt signaling pathway, promoting breast cancer cell proliferation. In vivo experiments further confirmed the anti-cancer role of miR-944 mediated SPP1 in breast cancer. Our study highlights the role of miR-944 mediated SPP1 in inhibiting breast cancer progression by blocking the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Thyroid and Breast Surgery, the People's Hospital of Liaoning Province, Shengyang, Liaoning, 110001, China
| | - Shan Li
- Department of General Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Xiangguo Cui
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital, China Medical University, Shenyang, Liaoning, 110022, China.
| | - Yiliang Wang
- Department of Anesthesiology, the First Hospital of China Medical University, No.155, Nanjing North Street, Heping District, Shengyang, Liaoning, 110001, China.
| |
Collapse
|
13
|
Yan Z, Hu X, Tang B, Deng F. Role of osteopontin in cancer development and treatment. Heliyon 2023; 9:e21055. [PMID: 37867833 PMCID: PMC10587537 DOI: 10.1016/j.heliyon.2023.e21055] [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: 07/19/2023] [Revised: 10/06/2023] [Accepted: 10/13/2023] [Indexed: 10/24/2023] Open
Abstract
Osteopontin (OPN) is a multifunctional protein secreted intracellularly and extracellularly by various cell types, including NK cells, macrophages, osteoblasts, T cells, and cancer cells. Owing to its diverse distribution, OPN plays a role in cell proliferation, stem-cell-like properties, epithelial-mesenchymal transformation, glycolysis, angiogenesis, fibrosis, invasion, and metastasis. In this review, we discuss recent findings, interpret representative studies on OPN expression in cancer, clarify that elevated OPN levels are observed in multiple cancer types (including colorectal, breast, lung, and liver cancer), and explore how OPN-macrophage interactions shape the tumor microenvironment. We also summarize progress in OPN research with regard to tumor therapy, which can facilitate the development of novel anti-tumor treatment strategies.
Collapse
Affiliation(s)
- Zhihua Yan
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610500, China
| | - Xue Hu
- School of Basic Medical Science, Chengdu Medical College, Chengdu, 610500, China
| | - Bin Tang
- Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Fengmei Deng
- School of Basic Medical Science, Chengdu Medical College, Chengdu, 610500, China
| |
Collapse
|
14
|
Knipper K, Lyu SI, Quaas A, Bruns CJ, Schmidt T. Cancer-Associated Fibroblast Heterogeneity and Its Influence on the Extracellular Matrix and the Tumor Microenvironment. Int J Mol Sci 2023; 24:13482. [PMID: 37686288 PMCID: PMC10487587 DOI: 10.3390/ijms241713482] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
The tumor microenvironment comprises multiple cell types, like cancer cells, endothelial cells, fibroblasts, and immune cells. In recent years, there have been massive research efforts focusing not only on cancer cells, but also on other cell types of the tumor microenvironment, thereby aiming to expand and determine novel treatment options. Fibroblasts represent a heterogenous cell family consisting of numerous subtypes, which can alter immune cell fractions, facilitate or inhibit tumor growth, build pre-metastatic niches, or stabilize vessels. These effects can be achieved through cell-cell interactions, which form the extracellular matrix, or via the secretion of cytokines or chemokines. The pro- or antitumorigenic fibroblast phenotypes show variability not only among different cancer entities, but also among intraindividual sites, including primary tumors or metastatic lesions. Commonly prescribed for arterial hypertension, the inhibitors of the renin-angiotensin system have recently been described as having an inhibitory effect on fibroblasts. This inhibition leads to modified immune cell fractions and increased tissue stiffness, thereby contributing to overcoming therapy resistance and ultimately inhibiting tumor growth. However, it is important to note that the inhibition of fibroblasts can also have the opposite effect, potentially resulting in increased tumor growth. We aim to summarize the latest state of research regarding fibroblast heterogeneity and its intricate impact on the tumor microenvironment and extracellular matrix. Specifically, we focus on highlighting recent advancements in the comprehension of intraindividual heterogeneity and therapy options within this context.
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; (K.K.); (C.J.B.)
| | - Su Ir Lyu
- Institute of Pathology, University Hospital of Cologne, Faculty of Medicine, University of Cologne, 50937 Cologne, Germany; (S.I.L.); (A.Q.)
| | - Alexander Quaas
- Institute of Pathology, University Hospital of Cologne, Faculty of Medicine, University of Cologne, 50937 Cologne, Germany; (S.I.L.); (A.Q.)
| | - Christiane J. Bruns
- Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, Faculty of Medicine, University of Cologne, 50937 Cologne, Germany; (K.K.); (C.J.B.)
| | - Thomas Schmidt
- Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, Faculty of Medicine, University of Cologne, 50937 Cologne, Germany; (K.K.); (C.J.B.)
| |
Collapse
|
15
|
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: 7] [Impact Index Per Article: 3.5] [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
|
16
|
Wang T, Guo K, Zhang D, Wang H, Yin J, Cui H, Wu W. Disulfidptosis classification of hepatocellular carcinoma reveals correlation with clinical prognosis and immune profile. Int Immunopharmacol 2023; 120:110368. [PMID: 37247499 DOI: 10.1016/j.intimp.2023.110368] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/26/2023] [Accepted: 05/18/2023] [Indexed: 05/31/2023]
Abstract
A new mode of cell death, disulfidptosis, has been discovered. Clinical prognostic significance of disulfidptosis related pattern in hepatocellular carcinoma(HCC). In this study, a risk score model was established based on disulfidptosis model to analyze the role of risk score in clinical prognosis, immune cell infiltration, drug sensitivity and immunotherapy response. Disulfidptosis subtype were constructed based on the transcriptional profiles of 15 disulfidptosis-related genes(DRGs). All 601 samples were defined as high risk group(HRG) and low risk group(LRG) based on the disulfidptosis risk score. Drug sensitivity and response to immunotherapy were calculated by immunophenotypic score(IPS), tumor prediction, tumor immune dysfunction and rejection(TIDE). RT-qPCR was used to determine the mRNA level of disulfidptosis prognostic gene. Risk groups was identified as potential predictors of immune cell infiltration, drug sensitivity, and immunotherapy responsiveness. HRG may benefit from immunotherapy. Classification is very effective in predicting the prognosis and therapeutic effect of patients, and provides a reference for accurate individualized treatment. This study suggests that new biomarkers related to Disulfidptosis can be used in clinical diagnosis of liver cancer to predict prognosis and treatment targets.
Collapse
Affiliation(s)
- Tianbing Wang
- Department of General Surgery, Anhui No.2 Provincial People's Hospital, Hefei 230000, China
- Anhui Medical University, Hefei 230000, China
- Anhui No.2 Provincial People's Hospital affiliated to Anhui Medical University, Hefei 230000, China
- The Fifth Clinical Medical College of Anhui Medical University, Hefei 230000, China
| | - Kai Guo
- Department of General Surgery, Anhui No.2 Provincial People's Hospital, Hefei 230000, China
- Anhui Medical University, Hefei 230000, China
- Anhui No.2 Provincial People's Hospital affiliated to Anhui Medical University, Hefei 230000, China
- The Fifth Clinical Medical College of Anhui Medical University, Hefei 230000, China
| | - Di Zhang
- Department of General Surgery, Anhui No.2 Provincial People's Hospital, Hefei 230000, China
| | - Haibo Wang
- Anhui Medical University, Hefei 230000, China
- Department of General Surgery, First affiliated Hospital of Anhui Medical University, Hefei 230000, China
| | - Jimin Yin
- Anhui No.2 Provincial People's Hospital affiliated to Anhui Medical University, Hefei 230000, China
- The Fifth Clinical Medical College of Anhui Medical University, Hefei 230000, China
| | - Haodong Cui
- Anhui No.2 Provincial People's Hospital affiliated to Anhui Medical University, Hefei 230000, China
- The Fifth Clinical Medical College of Anhui Medical University, Hefei 230000, China
| | - Wenyong Wu
- Department of General Surgery, Anhui No.2 Provincial People's Hospital, Hefei 230000, China
- Anhui No.2 Provincial People's Hospital affiliated to Anhui Medical University, Hefei 230000, China
- The Fifth Clinical Medical College of Anhui Medical University, Hefei 230000, China
| |
Collapse
|
17
|
Saup R, Nair N, Shen J, Schmaus A, Thiele W, Garvalov BK, Sleeman JP. Increased Circulating Osteopontin Levels Promote Primary Tumour Growth, but Do Not Induce Metastasis in Melanoma. Biomedicines 2023; 11:biomedicines11041038. [PMID: 37189656 DOI: 10.3390/biomedicines11041038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/21/2023] [Accepted: 03/25/2023] [Indexed: 03/30/2023] Open
Abstract
Osteopontin (OPN) is a phosphoprotein with diverse functions in various physiological and pathological processes. OPN expression is increased in multiple cancers, and OPN within tumour tissue has been shown to promote key stages of cancer development. OPN levels are also elevated in the circulation of cancer patients, which in some cases has been correlated with enhanced metastatic propensity and poor prognosis. However, the precise impact of circulating OPN (cOPN) on tumour growth and progression remains insufficiently understood. To examine the role of cOPN, we used a melanoma model, in which we stably increased the levels of cOPN through adeno-associated virus-mediated transduction. We found that increased cOPN promoted the growth of primary tumours, but did not significantly alter the spontaneous metastasis of melanoma cells to the lymph nodes or lungs, despite an increase in the expression of multiple factors linked to tumour progression. To assess whether cOPN has a role at later stages of metastasis formation, we employed an experimental metastasis model, but again could not detect any increase in pulmonary metastasis in animals with elevated levels of cOPN. These results demonstrate that increased levels of OPN in the circulation play distinct roles during different stages of melanoma progression.
Collapse
|
18
|
Bourn MD, Mohajerani SZ, Mavria G, Ingram N, Coletta PL, Evans SD, Peyman SA. Tumour associated vasculature-on-a-chip for the evaluation of microbubble-mediated delivery of targeted liposomes. LAB ON A CHIP 2023; 23:1674-1693. [PMID: 36779251 PMCID: PMC10013341 DOI: 10.1039/d2lc00963c] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 02/06/2023] [Indexed: 06/18/2023]
Abstract
The vascular system is the primary route for the delivery of therapeutic drugs throughout the body and is an important barrier at the region of disease interest, such as a solid tumour. The development of complex 3D tumour cultures has progressed significantly in recent years however, the generation of perfusable vascularised tumour models still presents many challenges. This study presents a microfluidic-based vasculature system that can be induced to display properties of tumour-associated blood vessels without direct incorporation of tumour cells. Conditioning healthy endothelial-fibroblast cell vasculature co-cultures with media taken from tumour cell cultures was found to result in the formation of disorganised, tortuous networks which display characteristics consistent with those of tumour-associated vasculature. Integrin αvβ3, a cell adhesion receptor associated with angiogenesis, was found to be upregulated in vasculature co-cultures conditioned with tumour cell media (TCM) - consistent with the reported αvβ3 expression pattern in angiogenic tumour vasculature in vivo. Increased accumulation of liposomes (LSs) conjugated to antibodies against αvβ3 was observed in TCM networks compared to non-conditioned networks, indicating αvβ3 may be a potential target for the delivery of drugs specifically to tumour vasculature. Furthermore, the use of microbubbles (MBs) and ultrasound (US) to further enhance the delivery of LSs to TCM-conditioned vasculature was investigated. Quantification of fluorescent LS accumulation post-perfusion of the vascular network showed 3-fold increased accumulation with the use of MBs and US, suggesting that targeted LS delivery could be further improved with the use of locally administered MBs and US.
Collapse
Affiliation(s)
- Matthew D Bourn
- School of Physics and Astronomy, University of Leeds, Leeds, LS2 9JT, UK.
- Leeds Institute for Medical Research, Wellcome Trust Brenner Building, St James' University Hospital, Leeds, LS9 7TF, UK
| | - Safoura Zahed Mohajerani
- Leeds Institute for Medical Research, Wellcome Trust Brenner Building, St James' University Hospital, Leeds, LS9 7TF, UK
| | - Georgia Mavria
- Leeds Institute for Medical Research, Wellcome Trust Brenner Building, St James' University Hospital, Leeds, LS9 7TF, UK
| | - Nicola Ingram
- Leeds Institute for Medical Research, Wellcome Trust Brenner Building, St James' University Hospital, Leeds, LS9 7TF, UK
| | - P Louise Coletta
- Leeds Institute for Medical Research, Wellcome Trust Brenner Building, St James' University Hospital, Leeds, LS9 7TF, UK
| | - Stephen D Evans
- School of Physics and Astronomy, University of Leeds, Leeds, LS2 9JT, UK.
| | - Sally A Peyman
- School of Physics and Astronomy, University of Leeds, Leeds, LS2 9JT, UK.
- Leeds Institute for Medical Research, Wellcome Trust Brenner Building, St James' University Hospital, Leeds, LS9 7TF, UK
| |
Collapse
|
19
|
Zhang J, Lu S, Lu T, Han D, Zhang K, Gan L, Wu X, Li Y, Zhao X, Li Z, Shen Y, Hu S, Yang F, Wen W, Qin W. Single-cell analysis reveals the COL11A1 + fibroblasts are cancer-specific fibroblasts that promote tumor progression. Front Pharmacol 2023; 14:1121586. [PMID: 36744260 PMCID: PMC9894880 DOI: 10.3389/fphar.2023.1121586] [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: 12/12/2022] [Accepted: 01/13/2023] [Indexed: 01/21/2023] Open
Abstract
Background: Cancer-associated fibroblasts (CAFs) promote tumor progression through extracellular matrix (ECM) remodeling and extensive communication with other cells in tumor microenvironment. However, most CAF-targeting strategies failed in clinical trials due to the heterogeneity of CAFs. Hence, we aimed to identify the cluster of tumor-promoting CAFs, elucidate their function and determine their specific membrane markers to ensure precise targeting. Methods: We integrated multiple single-cell RNA sequencing (scRNA-seq) datasets across different tumors and adjacent normal tissues to identify the tumor-promoting CAF cluster. We analyzed the origin of these CAFs by pseudotime analysis, and tried to elucidate the function of these CAFs by gene regulatory network analysis and cell-cell communication analysis. We also performed cell-type deconvolution analysis to examine the association between the proportion of these CAFs and patients' prognosis in TCGA cancer cohorts, and validated that through IHC staining in clinical tumor tissues. In addition, we analyzed the membrane molecules in different fibroblast clusters, trying to identify the membrane molecules that were specifically expressed on these CAFs. Results: We found that COL11A1+ fibroblasts specifically exist in tumor tissues but not in normal tissues and named them cancer-specific fibroblasts (CSFs). We revealed that these CSFs were transformed from normal fibroblasts. CSFs represented a more activated CAF cluster and may promote tumor progression through the regulation on ECM remodeling and antitumor immune responses. High CSF proportion was associated with poor prognosis in bladder cancer (BCa) and lung adenocarcinoma (LUAD), and IHC staining of COL11A1 confirmed their specific expression in tumor stroma in clinical BCa samples. We also identified that CSFs specifically express the membrane molecules LRRC15, ITGA11, SPHK1 and FAP, which could distinguish CSFs from other fibroblasts. Conclusion: We identified that CSFs is a tumor specific cluster of fibroblasts, which are in active state, may promote tumor progression through the regulation on ECM remodeling and antitumor immune responses. Membrane molecules LRRC15, ITGA11, SPHK1 and FAP could be used as therapeutic targets for CSF-targeting cancer treatment.
Collapse
Affiliation(s)
- Jiayu Zhang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Shiqi Lu
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
| | - Tong Lu
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Donghui Han
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Keying Zhang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Lunbiao Gan
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
| | - Xinjie Wu
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
| | - Yu Li
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Xiaolong Zhao
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Zhengxuan Li
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yajie Shen
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Sijun Hu
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
| | - Fa Yang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China,*Correspondence: Weijun Qin, ; Weihong Wen, ; Fa Yang,
| | - Weihong Wen
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China,*Correspondence: Weijun Qin, ; Weihong Wen, ; Fa Yang,
| | - Weijun Qin
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China,*Correspondence: Weijun Qin, ; Weihong Wen, ; Fa Yang,
| |
Collapse
|
20
|
Ma YB, Qiao JW, Hu X. Transmembrane serine protease 2 cleaves nidogen 1 and inhibits extrahepatic liver cancer cell migration and invasion. Exp Biol Med (Maywood) 2023; 248:91-105. [PMID: 36408877 PMCID: PMC10041054 DOI: 10.1177/15353702221134111] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We aimed to confirm whether transmembrane serine protease 2 (TMPRSS2) regulates nidogen 1 (NID1) expression in extracellular vesicles (EVs) and metastatic hepatocellular carcinoma (HCC) cells. HCC cells, HUVEC cells, MRC-5 cells, HLE cells, MHCCLM3 cells, MHCC97L cells, H2P cells, H2M cells, as well as LO2 cells were cultured according to providers' instruction and EV models were established by using BALB/cAnN-nu mice to facilitate the verifications. We found that TMPRSS2 expression was inversely correlated with the metastatic potential of HCC cell lines. The expression of TMPRSS2 decreased in a time-dependent manner in tumor-bearing model mice implanted with MHCCLM3 cells compared with uninoculated mice. TMPRSS2 overexpression in MHCCLM3 and MHCC97L cells led to the significant downregulation of NID1 expression in total cell lysates and isolated EVs. In contrast, TMPRSS2 silencing resulted in the elevation of NID1 expression in cells and EVs. Administration of EVs from MHCCLM3 and MHCC97L cells with overexpressed or silenced TMPRSS2 inhibited or strengthened, respectively, the invasion, proliferation, and migration of LO2 tumor cells. EVs derived from MHCCLM3 and MHCC97L cells with overexpressed or depleted TMPRSS2 also deactivated or activated fibroblasts, respectively. These EVs secrete inflammatory cytokines and phosphorylated p65, facilitate the colonization of fibroblasts, and augment fibroblast growth and motility. These findings provide evidence for a new candidate drug targeting tumorigenic EV-NID1 to treat HCC.
Collapse
Affiliation(s)
- Yong-Biao Ma
- Department of Hepatobiliary Surgery, Weifang People's Hospital, Weifang 261041, China
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital, Shandong University, Jinan 250021, China
| | - Jian-Wen Qiao
- Department of Hepatobiliary Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai 264200, China
| | - Xiao Hu
- Department of Hepatobiliary Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| |
Collapse
|
21
|
Toledo B, Picon-Ruiz M, Marchal JA, Perán M. Dual Role of Fibroblasts Educated by Tumour in Cancer Behavior and Therapeutic Perspectives. Int J Mol Sci 2022; 23:15576. [PMID: 36555218 PMCID: PMC9778751 DOI: 10.3390/ijms232415576] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/25/2022] [Accepted: 12/03/2022] [Indexed: 12/13/2022] Open
Abstract
Tumours are complex systems with dynamic interactions between tumour cells, non-tumour cells, and extracellular components that comprise the tumour microenvironment (TME). The majority of TME's cells are cancer-associated fibroblasts (CAFs), which are crucial in extracellular matrix (ECM) construction, tumour metabolism, immunology, adaptive chemoresistance, and tumour cell motility. CAF subtypes have been identified based on the expression of protein markers. CAFs may act as promoters or suppressors in tumour cells depending on a variety of factors, including cancer stage. Indeed, CAFs have been shown to promote tumour growth, survival and spread, and secretome changes, but they can also slow tumourigenesis at an early stage through mechanisms that are still poorly understood. Stromal-cancer interactions are governed by a variety of soluble factors that determine the outcome of the tumourigenic process. Cancer cells release factors that enhance the ability of fibroblasts to secrete multiple tumour-promoting chemokines, acting on malignant cells to promote proliferation, migration, and invasion. This crosstalk between CAFs and tumour cells has given new prominence to the stromal cells, from being considered as mere physical support to becoming key players in the tumour process. Here, we focus on the concept of cancer as a non-healing wound and the relevance of chronic inflammation to tumour initiation. In addition, we review CAFs heterogeneous origins and markers together with the potential therapeutic implications of CAFs "re-education" and/or targeting tumour progression inhibition.
Collapse
Affiliation(s)
- Belén Toledo
- Department of Health Sciences, University of Jaén, E-23071 Jaén, Spain
| | - Manuel Picon-Ruiz
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, E-18100 Granada, Spain
- Instituto de Investigación Sanitaria ibs. GRANADA, Hospitales Universitarios de Granada-Universidad de Granada, E-18071 Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, E-18016 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, E-18016 Granada, Spain
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, E-18100 Granada, Spain
- Instituto de Investigación Sanitaria ibs. GRANADA, Hospitales Universitarios de Granada-Universidad de Granada, E-18071 Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, E-18016 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, E-18016 Granada, Spain
| | - Macarena Perán
- Department of Health Sciences, University of Jaén, E-23071 Jaén, Spain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, E-18100 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, E-18016 Granada, Spain
| |
Collapse
|
22
|
Zeltz C, Navab R, Heljasvaara R, Kusche-Gullberg M, Lu N, Tsao MS, Gullberg D. Integrin α11β1 in tumor fibrosis: more than just another cancer-associated fibroblast biomarker? J Cell Commun Signal 2022; 16:649-660. [PMID: 35378690 PMCID: PMC8978763 DOI: 10.1007/s12079-022-00673-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/04/2022] [Indexed: 12/13/2022] Open
Abstract
There is currently an increased interest in understanding the role of the tumor microenvironment (TME) in tumor growth and progression. In this context the role of integrins in cancer-associated fibroblasts (CAFs) will need to be carefully re-evaluated. Fibroblast-derived cells are not only in the focus in tumors, but also in tissue fibrosis as well as in inflammatory conditions. The recent transcriptional profiling of what has been called "the pan-fibroblast cell lineage" in mouse and human tissues has identified novel transcriptional biomarker mRNAs encoding the secreted ECM proteins dermatopontin and collagen XV as well as the phosphatidylinositol-anchored membrane protein Pi16. Some of the genes identified in these fibroblasts scRNA-seq datasets will be useful for rigorous comparative characterizations of fibroblast-derived cell subpopulations. At the same time, it will be a challenge in the coming years to validate these transcriptional mRNA datasets at the protein-(expression) and at tissue-(distribution) levels and to find useful protein biomarker reagents that will facilitate fibroblast profiling at the cell level. In the current review we will focus on the role of the collagen-binding integrin α11β1 in CAFs, summarizing our own work as well as published datasets with information on α11 mRNA expression in selected tumors. Our experimental data suggest that α11β1 is more than just another biomarker and that it as a functional collagen receptor in the TME is playing a central role in regulating collagen assembly and matrix remodeling, which in turn impact tumor growth and metastasis.
Collapse
Affiliation(s)
- Cédric Zeltz
- Department of Biomedicine, Matrix Biology Group, Centre for Cancer Biomarkers, University of Bergen, Jonas Lies vei 91, 5009, Bergen, Norway
| | - Roya Navab
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, M5G 1L7, Canada
| | - Ritva Heljasvaara
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Marion Kusche-Gullberg
- Department of Biomedicine, Matrix Biology Group, Centre for Cancer Biomarkers, University of Bergen, Jonas Lies vei 91, 5009, Bergen, Norway
| | - Ning Lu
- Department of Biomedicine, Matrix Biology Group, Centre for Cancer Biomarkers, University of Bergen, Jonas Lies vei 91, 5009, Bergen, Norway
| | - Ming-Sound Tsao
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, M5G 1L7, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5G 1X8, Canada
| | - Donald Gullberg
- Department of Biomedicine, Matrix Biology Group, Centre for Cancer Biomarkers, University of Bergen, Jonas Lies vei 91, 5009, Bergen, Norway.
| |
Collapse
|
23
|
Hao B, Zhang Z, Lu Z, Xiong J, Fan T, Song C, He R, Zhang L, Pan S, Li D, Meng H, Lin W, Luo B, Yang J, Li N, Geng Q. Single-cell RNA sequencing analysis revealed cellular and molecular immune profiles in lung squamous cell carcinoma. Transl Oncol 2022; 27:101568. [PMID: 36270103 PMCID: PMC9586982 DOI: 10.1016/j.tranon.2022.101568] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 07/05/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022] Open
Abstract
Although breakthroughs have been made in the treatment of non-small cell lung cancer, there are only a few choices for advanced-stage or recurrent lung squamous cell carcinoma (LUSC) patients. In our study, we identified 7 major cell types in thedepicted the immunolandscape of LUSC microenvironment using single-cell RNA sequencing. We found that an immunosuppressive receptor, T cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain (TIGIT), was highly expressed by regulatory T cells (Tregs) and exhausted CD8+T cells, suggesting that upregulation of TIGIT might promote an immunosuppressive microenvironment and inhibit the cytotoxic ability of CD8+T cells. We also identified tumor-associated neutrophil (TAN), characterized by CXCR2, CSF3R and CXCL8, in the tumor region, and TANs upregulated the expression of interleukin 1 receptor antagonist (IL1RN) which suggested that TAN might exert an immunosuppressive role via expressing IL1RN. Furthermore, the number of SPP1+ macrophages(SPP1+M) significantly increased in tumor microenvirnment, which was correlated with the poor survival of patients. Additionally, regulatory networks based on SPP1+M revealed that the disparities of several ligand-receptor pairs existed between tumor and normal tissues. Among these pairs, SPP1-CD44 showed the most interactions between SPP1+M and other cell types. Our results provided deep insight into the immune landscape of LUSC and an essential resource for drug discovery in the future.
Collapse
Affiliation(s)
- Bo Hao
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ziyao Zhang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zilong Lu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Juan Xiong
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Tao Fan
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Congkuan Song
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ruyuan He
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Lin Zhang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Shize Pan
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Donghang Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Heng Meng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Weichen Lin
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Bin Luo
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jinfeng Yang
- Department of Pathology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China,Corresponding author.
| |
Collapse
|
24
|
Abstract
Understanding how macrophages promote myocardial repair can help create new therapies for infarct repair. We aimed to determine what mechanisms underlie the reparative properties of macrophages. Cytokine arrays revealed that neonatal cardiac macrophages from the injured neonatal heart secreted high amounts of osteopontin (OPN). In vitro, recombinant OPN stimulated cardiac cell outgrowth, cardiomyocyte (CM) cell-cycle re-entry, and CM migration. In addition, OPN induced nuclear translocation of the cytoplasmatic yes-associated protein 1 (YAP1) and upregulated transcriptional factors and cell-cycle genes. Significantly, by blocking the OPN receptor CD44, we eliminated the effects of OPN on CMs. OPN also activated the proliferation and migration of non-CM cells: endothelial cells and cardiac mesenchymal stromal cells in vitro. Notably, the significant role of OPN in myocardial healing was demonstrated by impaired healing in OPN-deficient neonatal hearts. Finally, in the adult mice, a single injection of OPN into the border of the ischemic zone induced CM cell-cycle re-entry, improved scar formation, local and global cardiac function, and LV remodelling 30 days after MI. In summary, we have shown, for the first time, that recombinant OPN activates cell-cycle re-entry in CMs. In addition, recombinant OPN stimulates multiple cardiac cells and improves scar formation, LV remodelling, and regional and global function after MI. Therefore, we propose OPN as a new cell-free therapy to optimize infarct repair.
Collapse
|
25
|
Zhang X, Zhang Y, Qiu X, Cai J, Yang Z, Song F. Extracellular Vesicles Derived from Lung Cancer Cells Induce Transformation of Normal Fibroblasts into Lung Cancer-Associated Fibroblasts and Promote Metastasis of Lung Cancer by Delivering lncRNA HOTAIR. Stem Cells Int 2022; 2022:3805013. [PMID: 36267242 PMCID: PMC9578906 DOI: 10.1155/2022/3805013] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/11/2022] [Accepted: 09/02/2022] [Indexed: 11/29/2022] Open
Abstract
Human lung cancer (LC) cells A549/H358, normal lung epithelial cells BEAS-2B, and lung normal fibroblasts (NFs) were cultured, followed by transfection of H358 cells with HOTAIR shRNA. Extracellular vesicles (EVs) extracted from H358 cells were identified. The internalization of Dil-labeled-EVs by NFs was tested, and protein levels of cancer-associated fibroblast (CAF) surface markers, inflammatory cytokines, cell proliferation, invasion, and migration, and lncRNA HOTAIR levels were determined. A549 cells were cultured in an H358-EVs-treated conditioned medium of NFs (NFCM), followed by intravenous injection of A549 cells into nude mice. The lesions and Ki-67-positive cells in lung tissues were measured. The results showed that tumor cell-derived EVs (T-EVs) motivated the transformation of NFs into CAFs. Specifically, EVs can be internalized by NFs, and the protein levels of CAF surface markers and inflammation levels were elevated in H358-EVs-treated NFs. The proliferation, invasion, and migration of A549 cells cultured in T-EVs-treated NFCM were increased. H358-EVs carried HOTAIR into NFs and promoted the transformation of NFs into CAFs. Inhibition of HOTAIR partially reversed the promoting effect of H358-EVs on the transformation of NFs into CAFs and invasion and migration of LC cells. T-EVs promoted metastasis of LC in vivo by transforming NFs into CAFs.
Collapse
Affiliation(s)
- Xiaoxuan Zhang
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
- Department of Cancer Center, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Yan Zhang
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
| | - Xin Qiu
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
| | - Jing Cai
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
| | - Zhenzhou Yang
- Department of Cancer Center, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Fangzhou Song
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
26
|
Ferguson AM, Rubin MA. Lineage plasticity in prostate cancer: Looking beyond intrinsic alterations. Cancer Lett 2022; 548:215901. [PMID: 36075486 DOI: 10.1016/j.canlet.2022.215901] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/12/2022] [Accepted: 08/29/2022] [Indexed: 01/22/2023]
Abstract
Emergence of small cell prostate cancer is linked to the plasticity of tumour cells and avoidance of environmental pressures. This process is thought to be reversable, however to-date evidence of this has been demonstrated in small-cell prostate cancer. To study the plasticity of prostate tumours, we look to clinical cohorts of patients covering the spectra of malignancy subtypes and utilise in vitro and in vivo models of disease progression. Current models have assisted in the understanding of the extremities of this plasticity, elucidating internal mechanisms and adaptations to stressors through transition to altered cell states. By interrogating the tumour microenvironment and earlier time points, we are beginning to form a deeper understanding of the full spectra of tumour plasticity. It could be proffered that this deeper understanding will lead to better patient outcome, with earlier interventions more likely to reverse plasticity and prevent trans-differentiation to the aggressive, small cell phenotype.
Collapse
Affiliation(s)
- Alison M Ferguson
- Department for BioMedical Research, University of Bern, Bern, 3008, Switzerland
| | - Mark A Rubin
- Department for BioMedical Research, University of Bern, Bern, 3008, Switzerland; Bern Center for Precision Medicine, Inselspital, University Hospital of Bern, Bern, 3008, Switzerland.
| |
Collapse
|
27
|
Ruan H, Tang Q, Zhao X, Zhang Y, Zhao X, Xiang Y, Geng W, Feng Y, Cai W. The levels of osteopontin in human milk of Chinese mothers and its associations with maternal body composition. FOOD SCIENCE AND HUMAN WELLNESS 2022. [DOI: 10.1016/j.fshw.2022.04.033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
28
|
Shi X, Wu H, Liu Y, Huang H, Liu L, Yang Y, Jiang T, Zhou M, Dai M. Inhibiting vascular smooth muscle cell proliferation mediated by osteopontin via regulating gut microbial lipopolysaccharide: A novel mechanism for paeonol in atherosclerosis treatment. Front Pharmacol 2022; 13:936677. [PMID: 36034838 PMCID: PMC9403310 DOI: 10.3389/fphar.2022.936677] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/05/2022] [Indexed: 11/15/2022] Open
Abstract
Background: Although the gut microbiota is involved in metabolic disease such as atherosclerosis, the underlying mechanism remains elusive. Paeonol (Pae) is a natural phenolic compound isolated from Cortex Moutan, which exhibits anti-atherosclerotic effects. Our previous research demonstrated gut microbiota as a site of Pae action. However, the mechanism by which Pae exerts its anti-atherosclerotic effect by the regulation of gut microbiota remains unclear. Objective: To investigate a potential mechanistic link between the gut microbial lipopolysaccharide (LPS) and vascular smooth muscle cell (VSMC) proliferation in atherosclerosis progression and explore the possible role of Pae. Methods: Experimental atherosclerosis was established in ApoE−/− mice, and the atherosclerosis mice were treated with Pae for 4 weeks before being sacrificed for analyses while conducting fecal microbiota transplantation (FMT). The plaque area, levels of serum LPS, expressions of inflammatory factors in serum or aorta, and intestinal barrier permeability were determined. VSMCs were co-cultured with THP-1 cells. CCK-8 assay and EdU staining were performed to assess the proliferative capacity of VSMCs. Immunofluorescence staining was performed to observe the nuclear transfer of p65. Western blotting was used to detect the candidate protein expression level, and quantitative real-time PCR (qRT-PCR) was used to detect the mRNA expression level in tissues or cells of each group. Results: During atherosclerosis progression, gut dysbiosis leads to the peripheral accumulation of gut microbial LPS, which acts as a trigger to stimulate osteopontin (OPN) production from circulating monocytes, inducing cell-to-cell crosstalk to promote VSMC proliferation in the aorta. Importantly, the elevation of LPS and OPN concentrations in the blood was also observed in patients with atherosclerosis. Pae could significantly improve atherosclerosis, suppress gut microbial LPS accumulation, and inhibit monocyte/macrophage activation and VSMC proliferation. Conclusions: The present study provides a mechanistic scenario for how long-term stimulation of gut microbial LPS in circulating blood generates a pathological secondary response that leads to abnormal proliferation of VSMCs using high OPN expression in circulating monocytes and suggests a novel strategy for atherosclerosis therapy by remodeling the gut microbiota.
Collapse
Affiliation(s)
- Xiaoyan Shi
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Hongfei Wu
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Key Laboratory for Research and Development of Traditional Chinese Medicine, Hefei, China
| | - Yarong Liu
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Key Laboratory for Research and Development of Traditional Chinese Medicine, Hefei, China
| | - Hanwen Huang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Ling Liu
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Yulong Yang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Tingting Jiang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Min Zhou
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Min Dai
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Key Laboratory for Research and Development of Traditional Chinese Medicine, Hefei, China
- *Correspondence: Min Dai,
| |
Collapse
|
29
|
Immunotherapy and immunoengineering for breast cancer; a comprehensive insight into CAR-T cell therapy advancements, challenges and prospects. Cell Oncol (Dordr) 2022; 45:755-777. [PMID: 35943716 DOI: 10.1007/s13402-022-00700-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2022] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Breast cancer (BC) is a highly prevalent solid cancer with a high-rise infiltration of immune cells, turning it into a significant candidate for tumor-specific immunotherapies. Chimeric antigen receptor (CAR)-T cells are emerging as immunotherapeutic tools with genetically engineered receptors to efficiently recognize and attack tumor cells that express specific target antigens. Technological advancements in CAR design have provided five generations of CAR-T cells applicable to a wide range of cancer patients while boosting CAR-T cell therapy safety. However, CAR-T cell therapy is ineffective against breast cancer because of the loss of specified antigens, the immunosuppressive nature of the tumor and CAR-T cell-induced toxicities. Next-generation CAR-T cells actively pass through the tumor vascular barriers, persist for extended periods and disrupt the tumor microenvironment (TME) to block immune escape. CONCLUSION CAR-T cell therapy embodies advanced immunotherapy for BC, but further pre-clinical and clinical assessments are recommended to achieve maximized efficiency and safety.
Collapse
|
30
|
Papanicolaou M, Parker AL, Yam M, Filipe EC, Wu SZ, Chitty JL, Wyllie K, Tran E, Mok E, Nadalini A, Skhinas JN, Lucas MC, Herrmann D, Nobis M, Pereira BA, Law AMK, Castillo L, Murphy KJ, Zaratzian A, Hastings JF, Croucher DR, Lim E, Oliver BG, Mora FV, Parker BL, Gallego-Ortega D, Swarbrick A, O'Toole S, Timpson P, Cox TR. Temporal profiling of the breast tumour microenvironment reveals collagen XII as a driver of metastasis. Nat Commun 2022; 13:4587. [PMID: 35933466 PMCID: PMC9357007 DOI: 10.1038/s41467-022-32255-7] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/22/2022] [Indexed: 01/21/2023] Open
Abstract
The tumour stroma, and in particular the extracellular matrix (ECM), is a salient feature of solid tumours that plays a crucial role in shaping their progression. Many desmoplastic tumours including breast cancer involve the significant accumulation of type I collagen. However, recently it has become clear that the precise distribution and organisation of matrix molecules such as collagen I is equally as important in the tumour as their abundance. Cancer-associated fibroblasts (CAFs) coexist within breast cancer tissues and play both pro- and anti-tumourigenic roles through remodelling the ECM. Here, using temporal proteomic profiling of decellularized tumours, we interrogate the evolving matrisome during breast cancer progression. We identify 4 key matrisomal clusters, and pinpoint collagen type XII as a critical component that regulates collagen type I organisation. Through combining our proteomics with single-cell transcriptomics, and genetic manipulation models, we show how CAF-secreted collagen XII alters collagen I organisation to create a pro-invasive microenvironment supporting metastatic dissemination. Finally, we show in patient cohorts that collagen XII may represent an indicator of breast cancer patients at high risk of metastatic relapse. The distribution and organisation of matrix molecules in the tumour stroma help shape solid tumour progression. Here they perform temporal proteomic profiling of the matrisome during breast cancer progression and show that collagen XII secreted from CAFs provides a pro-invasive microenvironment.
Collapse
Affiliation(s)
- Michael Papanicolaou
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Amelia L Parker
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Michelle Yam
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Elysse C Filipe
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Sunny Z Wu
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Jessica L Chitty
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Kaitlin Wyllie
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Emmi Tran
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Ellie Mok
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Audrey Nadalini
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Joanna N Skhinas
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Morghan C Lucas
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia
| | - David Herrmann
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Max Nobis
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Brooke A Pereira
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Andrew M K Law
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia
| | - Lesley Castillo
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia
| | - Kendelle J Murphy
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Anaiis Zaratzian
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia
| | - Jordan F Hastings
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia
| | - David R Croucher
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Elgene Lim
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Brian G Oliver
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia.,Woolcock Institute of Medical Research, Respiratory Cellular and Molecular Biology, The University of Sydney, Sydney, NSW, Australia
| | - Fatima Valdes Mora
- Cancer Epigenetic Biology and Therapeutics, Personalised Medicine, Children's Cancer Institute, Sydney, NSW, 2031, Australia.,School of Women's and Children's Health, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - Benjamin L Parker
- Metabolic Systems Biology Laboratory, Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, Australia
| | - David Gallego-Ortega
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia.,School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, NSW, Australia
| | - Alexander Swarbrick
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Sandra O'Toole
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia.,Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia
| | - Paul Timpson
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia. .,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia. .,School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia.
| | - Thomas R Cox
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia. .,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia. .,School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia.
| |
Collapse
|
31
|
Sun X, Wang X, Yan C, Zheng S, Gao R, Huang F, Wei Y, Wen Z, Chen Y, Zhou X, Liu X, Chen B, Shen Y, Cai Y, Pan N, Wang L. Tumor cell-released LC3-positive EVs promote lung metastasis of breast cancer through enhancing premetastatic niche formation. Cancer Sci 2022; 113:3405-3416. [PMID: 35879596 PMCID: PMC9530874 DOI: 10.1111/cas.15507] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/07/2022] [Accepted: 07/14/2022] [Indexed: 12/24/2022] Open
Abstract
Most breast cancer–related deaths are caused by metastasis in vital organs including the lungs. Development of supportive metastatic microenvironments, referred to as premetastatic niches (PMNs), in certain distant organs before arrival of metastatic cells, is critical in metastasis. However, the mechanisms of PMN formation are not fully clear. Here, we demonstrated that chemoattractant C–C motif chemokine ligand 2 (CCL2) could be stimulated by heat shock protein 60 (HSP60) on the surface of murine 4 T1 breast cancer cell–released LC3+ extracellular vesicles (LC3+ EVs) via the TLR2‐MyD88‐NF‐κB signal cascade in lung fibroblasts, which subsequently promoted lung PMN formation through recruiting monocytes and suppressing T cell function. Consistently, reduction of LC3+ EV release or HSP60 level or neutralization of CCL2 markedly attenuated PMN formation and lung metastasis. Furthermore, the number of circulating LC3+ EVs and HSP60 level on LC3+ EVs in the plasma of breast cancer patients were positively correlated with disease progression and lung metastasis, which might have potential value as biomarkers of lung metastasis in breast cancer patients (AUC = 0.898, 0.694, respectively). These findings illuminate a novel mechanism of PMN formation and might provide therapeutic targets for anti‐metastasis therapy for patients with breast cancer.
Collapse
Affiliation(s)
- Xiaotong Sun
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Immunology, Medical School of Southeast University, Nanjing, China
| | - Xuru Wang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Immunology, Medical School of Southeast University, Nanjing, China
| | - Chunguang Yan
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Immunology, Medical School of Southeast University, Nanjing, China
| | - Shiya Zheng
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Rong Gao
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Immunology, Medical School of Southeast University, Nanjing, China
| | - Fang Huang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Immunology, Medical School of Southeast University, Nanjing, China
| | - Yiting Wei
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Immunology, Medical School of Southeast University, Nanjing, China
| | - Zhifa Wen
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Immunology, Medical School of Southeast University, Nanjing, China
| | - Yongqiang Chen
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Immunology, Medical School of Southeast University, Nanjing, China
| | - Xiaohe Zhou
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Immunology, Medical School of Southeast University, Nanjing, China
| | - Xueming Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Immunology, Medical School of Southeast University, Nanjing, China
| | - Bohao Chen
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Immunology, Medical School of Southeast University, Nanjing, China
| | - Yuqing Shen
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Immunology, Medical School of Southeast University, Nanjing, China
| | - Yunlang Cai
- Department of Obstetrics and Gynecology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Ning Pan
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Immunology, Medical School of Southeast University, Nanjing, China
| | - Lixin Wang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Immunology, Medical School of Southeast University, Nanjing, China
| |
Collapse
|
32
|
Tan Y, Zhao L, Yang YG, Liu W. The Role of Osteopontin in Tumor Progression Through Tumor-Associated Macrophages. Front Oncol 2022; 12:953283. [PMID: 35898884 PMCID: PMC9309262 DOI: 10.3389/fonc.2022.953283] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 06/15/2022] [Indexed: 11/18/2022] Open
Abstract
Osteopontin (OPN) is a multifunctional phosphorylated protein. It is widely involved in solid tumor progression, such as intensification of macrophage recruitment, inhibition of T-cell activity, aggravation of tumor interstitial fibrosis, promotion of tumor metastasis, chemotherapy resistance, and angiogenesis. Most of these pathologies are affected by tumor-associated macrophages (TAMs), an important component of the tumor microenvironment (TME). TAMs have been extensively characterized, including their subsets, phenotypes, activation status, and functions, and are considered a promising therapeutic target for cancer treatment. This review focuses on the interaction between OPN and TAMs in mediating tumor progression. We discuss the strategies for targeting OPN and TAMs to treat cancer and factors that may affect the therapeutic outcomes of blocking OPN or depleting TAMs. We also discuss the role of cancer cell- vs. TAM-derived OPN in tumorigenesis, the mechanisms of how OPN affects TAM recruitment and polarization, and why OPN could mediate anti-tumor and pro-tumor effects, as well as previously reported discrepancies.
Collapse
Affiliation(s)
- Yuying Tan
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
- National–Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, China
| | - Lei Zhao
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
- National–Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
- National–Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, China
- International Center of Future Science, Jilin University, Changchun, China
- *Correspondence: Yong-Guang Yang, ; Wentao Liu,
| | - Wentao Liu
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
- National–Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, China
- *Correspondence: Yong-Guang Yang, ; Wentao Liu,
| |
Collapse
|
33
|
Zhang Z, Jia G, Pan G, Cao K, Yang Q, Meng H, Yang J, Zhang L, Wang T, Cheng C, Zuo C. Comparison of the diagnostic efficacy of 68 Ga-FAPI-04 PET/MR and 18F-FDG PET/CT in patients with pancreatic cancer. Eur J Nucl Med Mol Imaging 2022; 49:2877-2888. [PMID: 35243518 DOI: 10.1007/s00259-022-05729-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 02/13/2022] [Indexed: 12/26/2022]
Abstract
PURPOSE We sought to assess the performance of 68 Ga-FAPI-04 PET/MR for the diagnosis of primary tumours as well as metastatic lesions in patients with pancreatic cancer and to compare the results with those of 18F-FDG PET/CT. METHODS Prospectively, we evaluated 33 patients suspected to have pancreatic adenocarcinoma, of whom thirty-two were confirmed by histopathology, and one had autoimmune pancreatitis confirmed by needle biopsy and glucocorticoid treatment. Within 1 week, each patient underwent both 68 Ga-FAPI-04 PET/MR and 18F-FDG PET/CT. Comparisons of the detection abilities for primary tumours, lymph nodes, and metastases were conducted for the two imaging approaches. The original maximum standard uptake values (SUVmax) and normalised SUVmax (SUVmax/SUVbkgd) of paired lesions on 68 Ga-FAPI-04 PET/MR and 18F-FDG PET/CT were measured and compared. RESULTS Thirty pancreatic cancer patients and three pancreatitis patients were enrolled. 68 Ga-FAPI-04 PET/MR and 18F-FDG PET/CT exhibited equivalent (100%) detection rates for primary tumours. The original/normalised SUVmax of primary tumours on 68 Ga-FAPI-04 PET was markedly higher than that on 18F-FDG (p < 0.05). Sixteen pancreatic cancer patients had pancreatic parenchymal uptake, whereas 18F-FDG PET images showed parenchymal uptake in only four patients (53.33% vs. 13.33%, p < 0.001). 68 Ga-FAPI-04 PET detected more positive lymph nodes than 18F-FDG PET (42 vs. 30, p < 0.001), while 18F-FDG PET was able to detect more liver metastases than 68 Ga-FAPI-04 (181 vs. 104, p < 0.001). In addition, multisequence MR imaging helped explain ten pancreatic cancers that could not be definitively revealed due to 68 Ga-FAPI-04 inflammatory uptake and identified more liver metastases than 18F-FDG (256 vs. 181, p < 0.001). CONCLUSION 68 Ga-FAPI-04 PET might be better than 18F-FDG PET in the detection of suspicious lymph node metastases. MR multiple sequence imaging of 68 Ga-FAPI-04 PET/MR was helpful for explaining pancreatic lesions in patients with obstructive inflammation and detecting tiny liver metastases.
Collapse
Affiliation(s)
- Zeyu Zhang
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Guorong Jia
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Guixia Pan
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Kai Cao
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Qinqin Yang
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Hongyu Meng
- Department of Radiology, Shanghai Fourth People's Hospital Affiliated to Tongji University, School of Medicine, Shanghai, 200434, China
| | - Jian Yang
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Lu Zhang
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Tao Wang
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Chao Cheng
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| | - Changjing Zuo
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| |
Collapse
|
34
|
Lavie D, Ben-Shmuel A, Erez N, Scherz-Shouval R. Cancer-associated fibroblasts in the single-cell era. NATURE CANCER 2022; 3:793-807. [PMID: 35883004 PMCID: PMC7613625 DOI: 10.1038/s43018-022-00411-z] [Citation(s) in RCA: 222] [Impact Index Per Article: 74.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 06/14/2022] [Indexed: 01/28/2023]
Abstract
Cancer-associated fibroblasts (CAFs) are central players in the microenvironment of solid tumors, affecting cancer progression and metastasis. CAFs have diverse phenotypes, origins and functions and consist of distinct subpopulations. Recent progress in single-cell RNA-sequencing technologies has enabled detailed characterization of the complexity and heterogeneity of CAF subpopulations in multiple tumor types. In this Review, we discuss the current understanding of CAF subsets and functions as elucidated by single-cell technologies, their functional plasticity, and their emergent shared and organ-specific features that could potentially be harnessed to design better therapeutic strategies for cancer.
Collapse
Affiliation(s)
- Dor Lavie
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Aviad Ben-Shmuel
- Department of Biomolecular Sciences, the Weizmann Institute of Science, Rehovot, Israel
| | - Neta Erez
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Ruth Scherz-Shouval
- Department of Biomolecular Sciences, the Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
35
|
Muchlińska A, Nagel A, Popęda M, Szade J, Niemira M, Zieliński J, Skokowski J, Bednarz-Knoll N, Żaczek AJ. Alpha-smooth muscle actin-positive cancer-associated fibroblasts secreting osteopontin promote growth of luminal breast cancer. Cell Mol Biol Lett 2022; 27:45. [PMID: 35690734 PMCID: PMC9188043 DOI: 10.1186/s11658-022-00351-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 05/31/2022] [Indexed: 12/27/2022] Open
Abstract
Background Cancer-associated fibroblasts (CAFs) have been shown to support tumor development in a variety of cancers. Different markers were applied to classify CAFs in order to elucidate their impact on tumor progression. However, the exact mechanism by which CAFs enhance cancer development and metastasis is yet unknown.
Methods Alpha-smooth muscle actin (α-SMA) was examined immunohistochemically in intratumoral CAFs of nonmetastatic breast cancers and correlated with clinicopathological data. Four CAF cell lines were isolated from patients with luminal breast cancer (lumBC) and classified according to the presence of α-SMA protein. Conditioned medium (CM) from CAF cultures was used to assess the influence of CAFs on lumBC cell lines: MCF7 and T47D cells using Matrigel 3D culture assay. To identify potential factors accounting for promotion of tumor growth by α-SMAhigh CAFs, nCounter PanCancer Immune Profiling Panel (NanoString) was used. Results In luminal breast cancer, presence of intratumoral CAFs expressing high level of α-SMA (13% of lumBC group) correlated with poor prognosis (p = 0.019). In in vitro conditions, conditioned medium obtained from primary cultures of α-SMA-positive CAFs isolated from luminal tumors was observed to enhance growth of lumBC cell line colonies in 3D Matrigel, in contrast to CM derived from α-SMA-negative CAFs. Multigene expression analysis indicated that osteopontin (OPN) was overexpressed in α-SMA-positive CAFs in both clinical samples and in vitro models. OPN expression was associated with higher percentage of Ki67-positive cells in clinical material (p = 0.012), while OPN blocking in α-SMA-positive CAF-derived CM attenuated growth of lumBC cell line colonies in 3D Matrigel. Conclusions Our findings demonstrate that α-SMA-positive CAFs might enhance tumor growth via secretion of OPN. Supplementary Information The online version contains supplementary material available at 10.1186/s11658-022-00351-7.
Collapse
Affiliation(s)
- Anna Muchlińska
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, Medical University of Gdansk, 80-211, Gdansk, Poland
| | - Anna Nagel
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, Medical University of Gdansk, 80-211, Gdansk, Poland
| | - Marta Popęda
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, Medical University of Gdansk, 80-211, Gdansk, Poland
| | - Jolanta Szade
- Department of Pathomorphology, Medical University of Gdansk, 80-214, Gdansk, Poland
| | - Magdalena Niemira
- Clinical Research Centre, Medical University of Bialystok, 15-276, Bialystok, Poland
| | - Jacek Zieliński
- Department of Surgical Oncology, Medical University of Gdansk, 80-214, Gdansk, Poland
| | - Jarosław Skokowski
- Department of Surgical Oncology, Medical University of Gdansk, 80-214, Gdansk, Poland.,Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.PL), 80-211, Gdansk, Poland
| | - Natalia Bednarz-Knoll
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, Medical University of Gdansk, 80-211, Gdansk, Poland
| | - Anna J Żaczek
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, Medical University of Gdansk, 80-211, Gdansk, Poland.
| |
Collapse
|
36
|
Zhang M, Chen Z, Wang Y, Zhao H, Du Y. The Role of Cancer-Associated Fibroblasts in Ovarian Cancer. Cancers (Basel) 2022; 14:2637. [PMID: 35681617 PMCID: PMC9179444 DOI: 10.3390/cancers14112637] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/20/2022] [Accepted: 05/24/2022] [Indexed: 12/30/2022] Open
Abstract
Ovarian cancer is a lethal gynecologic tumor and is generally resistant to conventional treatments. Stable cancer-associated fibroblasts (CAFs) are important cellular components in the ovarian cancer tumor microenvironment and may provide novel resources for future treatment strategies. Different subtypes of CAFs display specific functions in tumor pathogenesis and various CAF markers suggest potential treatment targets, such as FAP and GPR77. Both autocrine and paracrine cytokines play important roles in the CAF activation process and regulate tumor progression. Downstream mediators and pathways, including IL-6, TGF-β, NF-κB, mitogen-activated protein kinase (MAPK), and AKT/mTOR/(p70S6K), play important roles in the initiation, proliferation, invasiveness, and metastasis of ovarian cancer cells and also participate in angiogenesis, therapeutic resistance, and other biological processes. Several clinical or preclinical trials have targeted stromal fibroblasts and focused on the properties of CAFs to enhance ovarian cancer treatment outcomes. This review concentrates on the origins, subtypes, and activation of CAFs, as well as specific roles of CAFs in regulating tumor development and drug resistance, and aims to provide potential and prospective targets for improving the therapeutic efficacy of ovarian cancer treatment.
Collapse
Affiliation(s)
- Mo Zhang
- Clinical Research Unit, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (M.Z.); (Z.C.); (Y.W.)
- Department of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Zhixian Chen
- Clinical Research Unit, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (M.Z.); (Z.C.); (Y.W.)
- Department of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Yan Wang
- Clinical Research Unit, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (M.Z.); (Z.C.); (Y.W.)
- Department of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Hongbo Zhao
- Clinical Research Unit, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (M.Z.); (Z.C.); (Y.W.)
- Department of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Yan Du
- Clinical Research Unit, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (M.Z.); (Z.C.); (Y.W.)
- Department of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| |
Collapse
|
37
|
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: 93] [Impact Index Per Article: 31.0] [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
|
38
|
Han L, Seward C, Leone G, Ostrowski MC. Origin, activation and heterogeneity of fibroblasts associated with pancreas and breast cancers. Adv Cancer Res 2022; 154:169-201. [PMID: 35459469 DOI: 10.1016/bs.acr.2022.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Pancreas and breast cancers both contain abundant stromal components within the tumor tissues. A prominent cell type within the stroma is cancer-associated fibroblasts (CAFs). CAFs play critical and complex roles establishing the tumor microenvironment to either promote or prevent tumor progression. Recently, complex genetic models and single cell-based techniques have provided emerging insights on the precise functions and cellular heterogeneity of CAFs. The transformation of normal fibroblasts into CAFs is a key event during tumor initiation and progression. Such coordination between tumor cells and fibroblasts plays an important role in cancer development. Reprograming fibroblasts is currently being explored for therapeutic benefits. In this review, we will discuss recent literature shedding light on the tissues of origin, activation mechanisms, and heterogeneity of CAFs comparing pancreas and breast cancers.
Collapse
Affiliation(s)
- Lu Han
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, United States.
| | - Cara Seward
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, United States
| | - Gustavo Leone
- Department of Biochemistry, Medical College of Wisconsin Cancer Center, Medical college of Wisconsin, Milwaukee, WI, United States
| | - Michael C Ostrowski
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, United States.
| |
Collapse
|
39
|
Ma X, Dighe A, Maziarz J, Neumann E, Erkenbrack E, Hei YY, Liu Y, Suhail Y, Pak I, Levchenko A, Wagner GP. Evolution of higher mesenchymal CD44 expression in the human lineage. Evol Med Public Health 2022; 10:447-462. [PMID: 36148042 PMCID: PMC9487634 DOI: 10.1093/emph/eoac036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 08/21/2022] [Indexed: 12/24/2022] Open
Abstract
CD44 is an extracellular matrix receptor implicated in cancer progression. CD44 increases the invasibility of skin (SF) and endometrial stromal fibroblasts (ESF) by cancer and trophoblast cells. We reasoned that the evolution of CD44 expression can affect both, the fetal–maternal interaction through CD44 in ESF as well as vulnerability to malignant cancer through expression in SF. We studied the evolution of CD44 expression in mammalian SF and ESF and demonstrate that in the human lineage evolved higher CD44 expression. Isoform expression in cattle and human is very similar suggesting that differences in invasibility are not due to the nature of expressed isoforms. We then asked whether the concerted gene expression increase in both cell types is due to shared regulatory mechanisms or due to cell type-specific factors. Reporter gene experiments with cells and cis-regulatory elements from human and cattle show that the difference of CD44 expression is due to cis effects as well as cell type-specific trans effects. These results suggest that the concerted expression increase is likely due to selection acting on both cell types because the evolutionary change in cell type-specific factors requires selection on cell type-specific functions. This scenario implies that the malignancy enhancing effects of elevated CD44 expression in humans likely evolved as a side-effect of positive selection on a yet unidentified other function of CD44. A possible candidate is the anti-fibrotic effect of CD44 but there are no reliable data showing that humans and primates are less fibrotic than other mammals.
Collapse
Affiliation(s)
- Xinghong Ma
- Systems Biology Institute, Yale University , West Haven, CT 06516, USA
- Department of Ecology and Evolutionary Biology, Yale University , New Haven, CT 06520, USA
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Sciences, Northeast Agricultural University , Harbin, China
| | - Anasuya Dighe
- Systems Biology Institute, Yale University , West Haven, CT 06516, USA
- Department of Ecology and Evolutionary Biology, Yale University , New Haven, CT 06520, USA
| | - Jamie Maziarz
- Systems Biology Institute, Yale University , West Haven, CT 06516, USA
- Department of Ecology and Evolutionary Biology, Yale University , New Haven, CT 06520, USA
| | - Edwin Neumann
- Systems Biology Institute, Yale University , West Haven, CT 06516, USA
| | - Eric Erkenbrack
- Systems Biology Institute, Yale University , West Haven, CT 06516, USA
- Department of Ecology and Evolutionary Biology, Yale University , New Haven, CT 06520, USA
| | - Yuan-Yuan Hei
- Cancer Biology Institute, Yale University , West Haven, CT 06516, USA
- Department of Pharmacology, Yale Medical School , New Haven, CT 06510, USA
| | - Yansheng Liu
- Cancer Biology Institute, Yale University , West Haven, CT 06516, USA
- Department of Pharmacology, Yale Medical School , New Haven, CT 06510, USA
| | - Yasir Suhail
- Department of Biomedical Engineering, University of Connecticut Health Center , Farmington, CT 06030, USA
| | - Irene Pak
- Systems Biology Institute, Yale University , West Haven, CT 06516, USA
- Department of Ecology and Evolutionary Biology, Yale University , New Haven, CT 06520, USA
| | - Andre Levchenko
- Systems Biology Institute, Yale University , West Haven, CT 06516, USA
- Department of Biomedical Engineering, Yale University , New Haven, CT 06520, USA
| | - Günter P Wagner
- Systems Biology Institute, Yale University , West Haven, CT 06516, USA
- Department of Ecology and Evolutionary Biology, Yale University , New Haven, CT 06520, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale Medical School , New Haven, CT 06510, USA
- Department of Obstetrics and Gynecology, Wayne State University , Detroit, MI 48202, USA
| | | |
Collapse
|
40
|
Williams MM, Hafeez SA, Christenson JL, O’Neill KI, Hammond NG, Richer JK. Reversing an Oncogenic Epithelial-to-Mesenchymal Transition Program in Breast Cancer Reveals Actionable Immune Suppressive Pathways. Pharmaceuticals (Basel) 2021; 14:ph14111122. [PMID: 34832904 PMCID: PMC8622696 DOI: 10.3390/ph14111122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 12/16/2022] Open
Abstract
Approval of checkpoint inhibitors for treatment of metastatic triple negative breast cancer (mTNBC) has opened the door for the use of immunotherapies against this disease. However, not all patients with mTNBC respond to current immunotherapy approaches such as checkpoint inhibitors. Recent evidence demonstrates that TNBC metastases are more immune suppressed than primary tumors, suggesting that combination or additional immunotherapy strategies may be required to activate an anti-tumor immune attack at metastatic sites. To identify other immune suppressive mechanisms utilized by mTNBC, our group and others manipulated oncogenic epithelial-to-mesenchymal transition (EMT) programs in TNBC models to reveal differences between this breast cancer subtype and its more epithelial counterpart. This review will discuss how EMT modulation revealed several mechanisms, including tumor cell metabolism, cytokine milieu and secretion of additional immune modulators, by which mTNBC cells may suppress both the innate and adaptive anti-tumor immune responses. Many of these pathways/proteins are under preclinical or clinical investigation as therapeutic targets in mTNBC and other advanced cancers to enhance their response to chemotherapy and/or checkpoint inhibitors.
Collapse
|
41
|
Elwakeel E, Weigert A. Breast Cancer CAFs: Spectrum of Phenotypes and Promising Targeting Avenues. Int J Mol Sci 2021; 22:11636. [PMID: 34769066 PMCID: PMC8583860 DOI: 10.3390/ijms222111636] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/22/2021] [Accepted: 10/22/2021] [Indexed: 01/03/2023] Open
Abstract
Activationof the tumor-associated stroma to support tumor growth is a common feature observed in different cancer entities. This principle is exemplified by cancer-associated fibroblasts (CAFs), which are educated by the tumor to shape its development across all stages. CAFs can alter the extracellular matrix (ECM) and secrete a variety of different molecules. In that manner they have the capability to affect activation, survival, proliferation, and migration of other stromal cells and cancer cell themselves. Alteration of the ECM, desmoplasia, is a common feature of breast cancer, indicating a prominent role for CAFs in shaping tumor development in the mammary gland. In this review, we summarize the multiple roles CAFs play in mammary carcinoma. We discuss experimental and clinical strategies to interfere with CAFs function in breast cancer. Moreover, we highlight the issues arising from CAFs heterogeneity and the need for further research to identify CAFs subpopulation(s) that can be targeted to improve breast cancer therapy.
Collapse
Affiliation(s)
- Eiman Elwakeel
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany;
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany;
- Frankfurt Cancer Institute, Goethe-University Frankfurt, 60596 Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, 60590 Frankfurt, Germany
| |
Collapse
|
42
|
Deasy SK, Erez N. A glitch in the matrix: organ-specific matrisomes in metastatic niches. Trends Cell Biol 2021; 32:110-123. [PMID: 34479765 DOI: 10.1016/j.tcb.2021.08.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/02/2021] [Accepted: 08/09/2021] [Indexed: 02/07/2023]
Abstract
Modification of the extracellular matrix (ECM) is a critical aspect of developing a metastasis-supportive organ niche. Recent work investigating ECM changes that facilitate metastasis has revealed ways in which different metastatic organ niches are similar as well as the distinct characteristics that make them unique. In this review, we present recent findings regarding how ECM modifications support metastasis in four frequent metastatic sites: the lung, liver, bone, and brain. We discuss ways in which these modifications are shared between metastatic organs as well as features specific to each location. We also discuss areas of technical innovation that could be advantageous to future research and areas of inquiry that merit further investigation.
Collapse
Affiliation(s)
- Sarah K Deasy
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Neta Erez
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
43
|
Shani O, Raz Y, Monteran L, Scharff Y, Levi-Galibov O, Megides O, Shacham H, Cohen N, Silverbush D, Avivi C, Sharan R, Madi A, Scherz-Shouval R, Barshack I, Tsarfaty I, Erez N. Evolution of fibroblasts in the lung metastatic microenvironment is driven by stage-specific transcriptional plasticity. eLife 2021; 10:e60745. [PMID: 34169837 PMCID: PMC8257251 DOI: 10.7554/elife.60745] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 06/24/2021] [Indexed: 12/21/2022] Open
Abstract
Mortality from breast cancer is almost exclusively a result of tumor metastasis, and lungs are one of the main metastatic sites. Cancer-associated fibroblasts are prominent players in the microenvironment of breast cancer. However, their role in the metastatic niche is largely unknown. In this study, we profiled the transcriptional co-evolution of lung fibroblasts isolated from transgenic mice at defined stage-specific time points of metastases formation. Employing multiple knowledge-based platforms of data analysis provided powerful insights on functional and temporal regulation of the transcriptome of fibroblasts. We demonstrate that fibroblasts in lung metastases are transcriptionally dynamic and plastic, and reveal stage-specific gene signatures that imply functional tasks, including extracellular matrix remodeling, stress response, and shaping the inflammatory microenvironment. Furthermore, we identified Myc as a central regulator of fibroblast rewiring and found that stromal upregulation of Myc transcriptional networks is associated with disease progression in human breast cancer.
Collapse
Affiliation(s)
- Ophir Shani
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Yael Raz
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv UniversityTel AvivIsrael
- Department of Obstetrics and Gynecology, Tel Aviv Sourasky Medical CenterTel AvivIsrael
| | - Lea Monteran
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Ye'ela Scharff
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Oshrat Levi-Galibov
- Department of Biomolecular Sciences, The Weizmann Institute of ScienceRehovotIsrael
| | - Or Megides
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Hila Shacham
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Noam Cohen
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Dana Silverbush
- Blavatnik School of Computer Sciences, Faculty of Exact Sciences, Tel Aviv UniversityTel AvivIsrael
| | - Camilla Avivi
- Department of Pathology, Sheba Medical Center, Tel Hashomer, affiliated with Sackler Faculty of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Roded Sharan
- Blavatnik School of Computer Sciences, Faculty of Exact Sciences, Tel Aviv UniversityTel AvivIsrael
| | - Asaf Madi
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Ruth Scherz-Shouval
- Department of Biomolecular Sciences, The Weizmann Institute of ScienceRehovotIsrael
| | - Iris Barshack
- Department of Pathology, Sheba Medical Center, Tel Hashomer, affiliated with Sackler Faculty of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Ilan Tsarfaty
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Neta Erez
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv UniversityTel AvivIsrael
| |
Collapse
|
44
|
Wu F, Yang J, Liu J, Wang Y, Mu J, Zeng Q, Deng S, Zhou H. Signaling pathways in cancer-associated fibroblasts and targeted therapy for cancer. Signal Transduct Target Ther 2021; 6:218. [PMID: 34108441 PMCID: PMC8190181 DOI: 10.1038/s41392-021-00641-0] [Citation(s) in RCA: 310] [Impact Index Per Article: 77.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/20/2021] [Accepted: 05/06/2021] [Indexed: 02/05/2023] Open
Abstract
To flourish, cancers greatly depend on their surrounding tumor microenvironment (TME), and cancer-associated fibroblasts (CAFs) in TME are critical for cancer occurrence and progression because of their versatile roles in extracellular matrix remodeling, maintenance of stemness, blood vessel formation, modulation of tumor metabolism, immune response, and promotion of cancer cell proliferation, migration, invasion, and therapeutic resistance. CAFs are highly heterogeneous stromal cells and their crosstalk with cancer cells is mediated by a complex and intricate signaling network consisting of transforming growth factor-beta, phosphoinositide 3-kinase/AKT/mammalian target of rapamycin, mitogen-activated protein kinase, Wnt, Janus kinase/signal transducers and activators of transcription, epidermal growth factor receptor, Hippo, and nuclear factor kappa-light-chain-enhancer of activated B cells, etc., signaling pathways. These signals in CAFs exhibit their own special characteristics during the cancer progression and have the potential to be targeted for anticancer therapy. Therefore, a comprehensive understanding of these signaling cascades in interactions between cancer cells and CAFs is necessary to fully realize the pivotal roles of CAFs in cancers. Herein, in this review, we will summarize the enormous amounts of findings on the signals mediating crosstalk of CAFs with cancer cells and its related targets or trials. Further, we hypothesize three potential targeting strategies, including, namely, epithelial-mesenchymal common targets, sequential target perturbation, and crosstalk-directed signaling targets, paving the way for CAF-directed or host cell-directed antitumor therapy.
Collapse
Affiliation(s)
- Fanglong Wu
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Jin Yang
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Junjiang Liu
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Ye Wang
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Jingtian Mu
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Qingxiang Zeng
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Shuzhi Deng
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Hongmei Zhou
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
45
|
Giorello MB, Borzone FR, Labovsky V, Piccioni FV, Chasseing NA. Cancer-Associated Fibroblasts in the Breast Tumor Microenvironment. J Mammary Gland Biol Neoplasia 2021; 26:135-155. [PMID: 33398516 DOI: 10.1007/s10911-020-09475-y] [Citation(s) in RCA: 24] [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: 05/29/2020] [Accepted: 11/30/2020] [Indexed: 12/11/2022] Open
Abstract
Years of investigation have shed light on a theory in which breast tumor epithelial cells are under the effect of the stromal microenvironment. This review aims to discuss recent findings concerning the phenotypic and functional characteristics of cancer associated fibroblasts (CAFs) and their involvement in tumor evolution, as well as their potential implications for anti-cancer therapy. In this manuscript, we reviewed that CAFs play a fundamental role in initiation, growth, invasion, and metastasis of breast cancer, and also serve as biomarkers in the clinical diagnosis, therapy, and prognosis of this disease.
Collapse
Affiliation(s)
- María Belén Giorello
- Laboratorio de Inmunohematología (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biología y Medicina Experimental, Vuelta de Obligado 2490, CP, 1428, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina.
| | - Francisco Raúl Borzone
- Laboratorio de Inmunohematología (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biología y Medicina Experimental, Vuelta de Obligado 2490, CP, 1428, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Vivian Labovsky
- Laboratorio de Inmunohematología (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biología y Medicina Experimental, Vuelta de Obligado 2490, CP, 1428, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Flavia Valeria Piccioni
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos (IBYME) y Laboratorio de Inmunohematología (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biología y Medicina Experimental, Vuelta de Obligado 2490, CP, 1428, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Norma Alejandra Chasseing
- Laboratorio de Inmunohematología (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biología y Medicina Experimental, Vuelta de Obligado 2490, CP, 1428, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
46
|
Desbois M, Wang Y. Cancer-associated fibroblasts: Key players in shaping the tumor immune microenvironment. Immunol Rev 2021; 302:241-258. [PMID: 34075584 DOI: 10.1111/imr.12982] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/14/2021] [Accepted: 04/30/2021] [Indexed: 12/18/2022]
Abstract
Cancer immunotherapies have rapidly changed the therapeutic landscape for cancer. Nevertheless, most of the patients show innate or acquired resistance to these therapies. Studies conducted in recent years have highlighted an emerging role of cancer-associated fibroblasts (CAFs) in immune regulation that shapes the tumor immune microenvironment (TIME) and influences response to cancer immunotherapies. In this review, we outline recent advances in the understanding of phenotypic and functional heterogeneity of CAFs. We will focus on emerging roles of CAFs in shaping the TIME, especially under a framework of tumor immunity continuum, and discuss current and future CAF-targeting therapeutic strategies in particular in the context of optimizing the success of immunotherapies.
Collapse
Affiliation(s)
- Mélanie Desbois
- Department of Oncology Biomarker Development, Genentech, Inc., South San Francisco, CA, USA
| | - Yulei Wang
- Department of Oncology Biomarker Development, Genentech, Inc., South San Francisco, CA, USA
| |
Collapse
|
47
|
Liu L, Zhang R, Deng J, Dai X, Zhu X, Fu Q, Zhang H, Tong Z, Zhao P, Fang W, Zheng Y, Bao X. Construction of TME and Identification of crosstalk between malignant cells and macrophages by SPP1 in hepatocellular carcinoma. Cancer Immunol Immunother 2021; 71:121-136. [PMID: 34028567 DOI: 10.1007/s00262-021-02967-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 05/11/2021] [Indexed: 12/19/2022]
Abstract
Liver cancer accounts for 6% of all malignancies causing death worldwide, and hepatocellular carcinoma (HCC) is the most common histological type. HCC is a heterogeneous cancer, but how the tumour microenvironment (TME) of HCC contributes to the progression of HCC remains unclear. In this study, we investigated the immune microenvironment by multiomics analysis. The tumour immune infiltration characteristics of HCC were determined at the genomic, epigenetic, bulk transcriptome and single-cell levels by data from The Cancer Genome Atlas portal and the Gene Expression Omnibus (GEO). An epigenetic immune-related scoring system (EIRS) was developed to stratify patients with poor prognosis. SPP1, one gene in the EIRS system, was identified as an immune-related predictor of poor survival in HCC patients. Through receptor-ligand pair analysis in single-cell RNA-seq, SPP1 was indicated to mediate the crosstalk between HCC cells and macrophages via SPP1-CD44 and SPP1-PTGER4 association. In vitro experiments further validate SPP1 can trigger the polarization of macrophages to M2-phenotype tumour-associated macrophages (TAMs).
Collapse
Affiliation(s)
- Lulu Liu
- Department of Medical Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Qingchun Road No.79, Shangcheng District, Hangzhou, 310003, China
| | - Ruyi Zhang
- Department of Medical Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Qingchun Road No.79, Shangcheng District, Hangzhou, 310003, China
| | - Jingwen Deng
- Department of Pathology, Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Xiaomeng Dai
- Department of Medical Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Qingchun Road No.79, Shangcheng District, Hangzhou, 310003, China
| | - Xudong Zhu
- Department of Medical Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Qingchun Road No.79, Shangcheng District, Hangzhou, 310003, China
| | - Qihan Fu
- Department of Medical Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Qingchun Road No.79, Shangcheng District, Hangzhou, 310003, China
| | - Hangyu Zhang
- Department of Medical Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Qingchun Road No.79, Shangcheng District, Hangzhou, 310003, China
| | - Zhou Tong
- Department of Medical Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Qingchun Road No.79, Shangcheng District, Hangzhou, 310003, China
| | - Peng Zhao
- Department of Medical Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Qingchun Road No.79, Shangcheng District, Hangzhou, 310003, China
| | - Weijia Fang
- Department of Medical Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Qingchun Road No.79, Shangcheng District, Hangzhou, 310003, China
| | - Yi Zheng
- Department of Medical Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Qingchun Road No.79, Shangcheng District, Hangzhou, 310003, China.
- Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, Hangzhou, China.
| | - Xuanwen Bao
- Department of Medical Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Qingchun Road No.79, Shangcheng District, Hangzhou, 310003, China.
| |
Collapse
|
48
|
Godugu K, Sudha T, Davis PJ, Mousa SA. Nano Diaminopropane tetrac and integrin αvβ3 expression in different cancer types: Anti-cancer efficacy and Safety. Cancer Treat Res Commun 2021; 28:100395. [PMID: 34034044 DOI: 10.1016/j.ctarc.2021.100395] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/27/2021] [Accepted: 05/06/2021] [Indexed: 01/01/2023]
Abstract
Integrins are a family of heterodimeric plasma membrane glycoproteins, which regulate tumor growth, angiogenesis, migration, and metastasis. Integrin αvβ3 has been recognized as a putative target for the treatment of several cancers. Thus, the characterization of αvβ3 distribution in different human tumors is of substantial interest in tumor targeting and its suppression. In this study we evaluated the expression of integrin αvβ3 in different cancer types to define the expression pattern in cancer model. Furthermore, we investigated the effect of novel αvβ3 antagonist Diaminopropane Tetraiodothyroacetic acid conjugated to poly (lactic-co-glycolic acid) polymer and its nanoformulated form (NDAT), on different cancer cell lines both in vitro and in xenografts. In vitro, NDAT downregulated αv and β3 monomer expression. In vivo in tumor xenografts, similarly, NDAT downregulated αv and β3. Distinct reduction in tumor weight and viability was observed in glioblastoma xenografts treated with NDAT. Furthermore, NDAT was safe and tolerable in mice treated with high doses. In conclusion, NDAT is an effective and safe inhibitor of integrin αvβ3 expression in various cancer types, which indicates its impact on the targetability and suppression of αvβ3-associated tumor functions.
Collapse
Affiliation(s)
- Kavitha Godugu
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, , 1 Discovery Drive, Rensselaer, NY, USA
| | - Thangirala Sudha
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, , 1 Discovery Drive, Rensselaer, NY, USA
| | - Paul J Davis
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, , 1 Discovery Drive, Rensselaer, NY, USA
| | - Shaker A Mousa
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, , 1 Discovery Drive, Rensselaer, NY, USA.
| |
Collapse
|
49
|
Association of the Collagen Signature in the Tumor Microenvironment With Recurrence and Survival of Patients With T4N0M0 Colon Cancer. Dis Colon Rectum 2021; 64:563-575. [PMID: 33538520 DOI: 10.1097/dcr.0000000000001907] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND The current clinicopathological risk factors do not accurately predict disease recurrence in patients with T4N0M0 colon cancer. We hypothesized that the collagen signature combined with clinicopathological risk factors (new model) had a better prognostic value than clinicopathological risk factors alone (clinicopathological model). OBJECTIVE This study aimed to establish a collagen signature in the tumor microenvironment and to validate its role in predicting the recurrence of T4N0M0 colon cancer. DESIGN This was a retrospective study. SETTINGS This study took place at a tertiary medical center. PATIENTS Patients with T4N0M0 colon cancer who underwent surgery at our center between 2009 and 2015 (n = 416) were included. INTERVENTION A total of 142 collagen features were analyzed in the tumor microenvironment in specimens of colon cancer by using second-harmonic generation imaging. A collagen signature was constructed using a least-absolute shrinkage and selection operator Cox regression model. MAIN OUTCOME MEASURES The primary outcomes measured were disease-free survival and overall survival. RESULTS The training and testing cohorts consisted of 291 and 125 randomly assigned samples, with recurrence rates of 19.9% and 22.4%. A 3-feature-based collagen signature predicted the recurrence risk at 1, 3, and 5 years, with the area under the receiver-operating characteristic curves of 0.808, 0.832, and 0.791 in the training cohort and 0.836, 0.807, and 0.794 in the testing cohort. Multivariate analysis revealed that the collagen signature could independently predict the disease-free survival (HR = 7.17, p < 0.001) and overall survival rates (HR = 5.03, p < 0.001). The new model had a better prognostic value than the clinicopathological model, which included 4 clinicopathological risk factors: obstruction or perforation, lymphovascular invasion, tumor budding, and no chemotherapy. LIMITATIONS This study was limited by its retrospective design. CONCLUSIONS The collagen signature in the tumor microenvironment may be a new prognostic marker that can effectively predict the recurrence and survival of patients with T4N0M0 colon cancer. See Video Abstract at http://links.lww.com/DCR/B503. ASOCIACIÓN DE LA RÚBRICA DE COLÁGENO EN EL MICROAMBIENTE TUMORAL CON LA RECIDIVA Y LA SOBREVIDA DE PACIENTES CON CÁNCER DE COLON T4N0M0: Los factores de riesgo clínico-patológicos actuales no predicen con precisión la recurrencia de la enfermedad en pacientes con cáncer de colon estadío T4N0M0. Presumimos que la rúbrica de colágeno combinada con factores de riesgo clínico-patológicos (nuevo modelo) tendrían un mejor valor pronóstico que los factores de riesgo clínico-patológicos solos (modelo clínico-patológico).El establecer una rúbrica de colágeno en el microambiente tumoral y validar su papel en la predicción de la recidiva del cáncer de colon T4N0M0.Estudio retrospectivo.Investigación llevada a cabo en un centro médico terciario.Se incluyeron pacientes con cáncer de colon T4N0M0 operados en nuestro centro entre 2009 y 2015 (n = 416).Se analizaron un total de 142 características de colágeno en el microambiente tumoral en muestras de cáncer de colon utilizando imágenes de segunda generación armónica. Se construyó una rúbrica de colágeno utilizando un modelo de regresión LASSO Cox.Sobrevida libre de enfermedad y sobrevida global.Las cohortes de entrenamiento y prueba consistieron en 291 y 125 muestras asignadas al azar, con tasas de recurrencia del 19,9% y 22,4%, respectivamente. La rúbrica del colágeno basada en 3 características predijo el riesgo de recurrencia a 1, 3 y 5 años, con el área bajo las curvas características operativas del receptor de 0,808, 0,832 y 0,791 en la cohorte de entrenamiento y 0,836, 0,807 y 0,794 en la cohorte de prueba, respectivamente. El análisis multivariado reveló que la firma de colágeno podría predecir de forma independiente la supervivencia libre de enfermedad (HR = 7,17, p <0,001) y las tasas de sobrevida general (HR = 5,03, p <0,001). El nuevo modelo tuvo un mejor valor pronóstico que el modelo clínico-patológico, que incluyó cuatro factores de riesgo clínico-patológicos: obstrucción o perforación, invasión linfovascular, gemación tumoral y ausencia de quimioterapia.Este estudio estuvo limitado por su diseño retrospectivo.La rúbrica de colágeno en el microambiente tumoral puede ser un nuevo marcador pronóstico para predecir eficazmente la recurrencia y la subrevida de los pacientes con cáncer de colon T4N0M0. Consulte Video Resumen en http://links.lww.com/DCR/B503. (Traducción-Dr. Xavier Delgadillo).
Collapse
|
50
|
Khongsti K, Pasupuleti BG, Das B, Bez G. 1,2,3-Triazole tethered 1,2,4‑trioxane trimer induces apoptosis in metastatic cancer cells and inhibits their proliferation, migration and invasion. Bioorg Chem 2021; 112:104952. [PMID: 33971565 DOI: 10.1016/j.bioorg.2021.104952] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 04/20/2021] [Accepted: 04/24/2021] [Indexed: 12/24/2022]
Abstract
Artemisinin (ART) has been in use against different cancer cells and its derivatives and conjugates are more cytotoxic to iron-rich cancer cells. It is desirable to develop easily achievable synthetic 1,2,4-trioxanes having the same pharmacophore as that of ART. To explore more efficient compounds, a 1,2,3-triazole tethered 1,2,4‑trioxane trimer (4T) was synthesized and the anti-cancer effects of ART and 4T on MDA-MB-435 and MDA-MB-231 cells were investigated concerning regulation of osteopontin (OPN) expression, which is associated with cancer progression and malignancy. 1H NMR and 13C NMR, oxidative stress analysis, flow cytometry, western blot, Real-Time PCR, transfections, luciferase assay, cell viability, proliferation, migration and chemotactic invasion assays were used in this study. It was observed that the 4T induced apoptosis by inhibiting Bcl-2 (~0.6-fold) and cleavage of caspase-3 (intrinsic pathway) in these metastatic cancer cells, and also reduced colony formation, migration and invasion of these cancer cells. The treatment of 4T decreased the reduced glutathione level and increased the activities of glucose-6-phosphate dehydrogenase and glutathione reductase in the 4T treated cancer cells as compared to their respective controls. Further, the expression of OPN was diminished (~0.5-fold) by the 4T in these cell lines. It was also observed that the key mitogen-activated protein kinase pathway proteins, mitogen-activated protein kinase kinase1/2 (~1.8-fold) and extracellular signal-regulated kinase1/2 (~16-fold), were also activated following the treatment of the 4T. However, the phosphorylated c-Jun level, a component of activator protein-1, was significantly reduced in these cancer cells upon 4T treatment. Taken together, we hypothesize that 4T may be useful for controlling cancer progression and malignancy.
Collapse
Affiliation(s)
- Kitboklang Khongsti
- Department of Zoology, North-Eastern Hill University, Shillong 793022, India
| | | | - Bidyadhar Das
- Department of Zoology, North-Eastern Hill University, Shillong 793022, India.
| | - Ghanashyam Bez
- Department of Chemistry, North-Eastern Hill University, Shillong 793022, India.
| |
Collapse
|