1
|
Zhang Z, Zhao M, Zhou Z, Ren X, He Y, Shen T, Zeng H, Li K, Zhang Y. Identification and validation of matrix metalloproteinase hub genes as potential biomarkers for Skin Cutaneous Melanoma. Front Oncol 2024; 14:1471267. [PMID: 39493455 PMCID: PMC11527786 DOI: 10.3389/fonc.2024.1471267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 10/01/2024] [Indexed: 11/05/2024] Open
Abstract
Objectives The role of matrix metalloproteinases (MMPs) in Skin Cutaneous Melanoma (SKCM) development and progression is unclear so far. This comprehensive study delved into the intricate role of MMPs in SKCM development and progression. Methods RT-qPCR, bisulfite sequencing, and WES analyzed MMP gene expression, promoter methylation, and mutations in SKCM cell lines. TCGA datasets validated findings. DrugBank and molecular docking identified potential regulatory drugs, and cell line experiments confirmed the role of key MMP genes in tumorigenesis. Results Our findings unveiled significant up-regulation of MMP9, MMP12, MMP14, and MMP16, coupled with hypomethylation of their promoters in SKCM cell lines, implicating their involvement in disease progression. Mutational analysis highlighted a low frequency of mutations in these genes, indicating less involvement of mutations in the expression regulatory mechanisms. Prognostic assessments showcased a significant correlation between elevated expression of these genes and poor overall survival (OS) in SKCM patients. Additionally, functional experiments involving gene silencing revealed a potential impact on cellular proliferation, further emphasizing the significance of MMP9, MMP12, MMP14, and MMP16 in SKCM pathobiology. Conclusion This study identifies Estradiol and Calcitriol as potential drugs for modulating MMP expression in SKCM, highlighting MMP9, MMP12, MMP14, and MMP16 as key diagnostic and prognostic biomarkers.
Collapse
Affiliation(s)
- Zhongyi Zhang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mei Zhao
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zubing Zhou
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaodan Ren
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yunliang He
- Institute of Traditional Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, Sichuan, China
| | - Tao Shen
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hongping Zeng
- Department of Combined Chinese and Western Pulmonary Diseases, Zigong First People's Hospital, Zigong, Sichuan, China
| | - Kai Li
- Sichuan Institute of Tourism College of Great Health Industry, Chengdu, Sichuan, China
| | - Yong Zhang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Institute of Traditional Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Wang J, Zheng C, Lu J, Xu X, Xiang G, Li J, Zhang J, Mu X, Lu Q. The mechanism of MMP14-positive tumor-associated fibroblast subsets in inhibiting PD-1 immunotherapy for esophageal cancer through exosomal tsRNA-10522. Funct Integr Genomics 2024; 24:186. [PMID: 39377944 PMCID: PMC11461773 DOI: 10.1007/s10142-024-01447-3] [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: 06/28/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 10/09/2024]
Abstract
Esophageal cancer (EC) continues to pose a significant health risk. Cancer-associated fibroblasts (CAFs), an essential part of the tumor microenvironment (TME), are viewed as potential therapeutic targets. However, their role in tumor mechanisms specific to esophageal cancer remains to be elucidated. This study identified MMP14+ CAFs and MMP14- CAFs using immunofluorescence staining. The cytotoxic activity of CD8 T cells was assessed via western blot and ELISA. Using a transwell test, the migratory potential of MMP14+ CAFs was evaluated. Using flow cytometry, apoptosis was found in the esophageal squamous cell carcinoma cell line KYSE30. To determine the important tsRNAs released by MMP14+ CAFs, tsRNA-seq was used. Two subgroups of EC receiving PD-1 immunotherapy were identified by our research: MMP14+ CAFs and MMP14- CAFs. MMP14+ CAFs showed improved migratory capacity and released more inflammatory factors linked to cancer. Through exosomes, these CAFs may prevent anti-PD-1-treated CD8 T cells from being cytotoxic. Furthermore, exosomal tsRNA from MMP14+ CAFs primarily targeted signaling pathways connected with cancer. Notably, it was discovered that tsRNA-10522 plays a critical role within inhibiting CD8 T cell tumor cell death. The tumor cell killing of CD8 T cells by exosomal tsRNA-10522 is inhibited by a subgroup of cells called MMP14+ CAFs inside the EC microenvironment during PD-1 immunotherapy. This reduces the effectiveness of PD-1 immunotherapy for EC. Our findings demonstrate the inhibitory function of MMP14+ CAFs within EC receiving PD-1 immunotherapy, raising the prospect that MMP14+ CAFs might serve as predictive indicators in EC receiving PD-1 immunotherapy.
Collapse
Affiliation(s)
- Juzheng Wang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
- Department of Thoracic Surgery, The First People's Hospital of Xianyang, Xianyang, 712000, Shaanxi, China
| | - Chunlong Zheng
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, NO. 569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Jiayu Lu
- Basic Medical College, Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Xinyao Xu
- College of Life Sciences, Northwest University, 229 Taibai North Road, Beilin District, Xi'an, 710069, Shaanxi, China
| | - Guangyu Xiang
- Basic Medical College, Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Jiahe Li
- Basic Medical College, Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Jipeng Zhang
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, NO. 569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Xiaorong Mu
- Department of Pathology, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an, 710039, Shaanxi, China.
| | - Qiang Lu
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, NO. 569 Xinsi Road, Xi'an, 710038, Shaanxi, China.
| |
Collapse
|
3
|
Młynarczyk G, Tokarzewicz A, Gudowska-Sawczuk M, Mroczko B, Novák V, Novák A, Mitura P, Romanowicz L. MMP-14 Exhibits Greater Expression, Content and Activity Compared to MMP-15 in Human Renal Carcinoma. Int J Mol Sci 2024; 25:8107. [PMID: 39125675 PMCID: PMC11312297 DOI: 10.3390/ijms25158107] [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: 06/23/2024] [Revised: 07/16/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Membrane-type metalloproteinases (including MMP-14 and MMP-15) are enzymes involved in the degradation of extracellular matrix components. In cancer, they are involved in processes such as cellular invasion, angiogenesis and metastasis. Therefore, the aim of this study was to evaluate the expression, content and activity of MMP-14 and MMP-15 in human renal cell carcinoma. Samples of healthy kidney tissue (n = 20) and tissue from clear-cell kidney cancer (n = 20) were examined. The presence and contents of the MMPs were assessed using Western blot and ELISA techniques, respectively. Their activity-both actual and specific-was evaluated using fluorimetric analysis. Both control and cancer human kidney tissues contain MMP-14 and MMP-15 enzymes in the form of high-molecular-weight complexes. Moreover, these enzymes occur in both active and latent forms. Their content in cancer tissues is very similar, but with a noteworthy decrease in content with an increase in the kidney cancer grade for both membrane-type metalloproteinases. Even more notable is the highest content of the investigated enzymes represented by MMP-14 in the control tissues. Considering the actual and specific activity outcomes, MMP-14 dominates over MMP-15 in all of the investigated tissues. Nevertheless, we also noted a significant enhancement of the activity of both metalloproteinases with an increase in the grade of renal cancer. The expression and activity of both enzymes were detected in all examined renal cancer tissues. However, our findings suggest that transmembrane metalloproteinase 14 (MMP-14) plays a much more significant and essential role than MMP-15 in the studied renal carcinoma tissues. Therefore, it seems that MMP-14 could be a promising target in the diagnosis, prognosis and therapy of renal cell carcinoma.
Collapse
Affiliation(s)
- Grzegorz Młynarczyk
- Department of Urology, Medical University of Białystok, 15-276 Białystok, Poland
| | - Anna Tokarzewicz
- Department of Medical Biochemistry, Medical University of Białystok, ul. Mickiewicza 2, 15-089 Białystok, Poland; (A.T.); (L.R.)
| | - Monika Gudowska-Sawczuk
- Department of Biochemical Diagnostics, Medical University of Białystok, 15-269 Białystok, Poland; (M.G.-S.); (B.M.)
| | - Barbara Mroczko
- Department of Biochemical Diagnostics, Medical University of Białystok, 15-269 Białystok, Poland; (M.G.-S.); (B.M.)
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Białystok, Poland
| | - Vojtěch Novák
- Department of Urology, Charles University and University Hospital Motol, 150 06 Praha, Czech Republic; (V.N.); (A.N.)
| | - Adam Novák
- Department of Urology, Charles University and University Hospital Motol, 150 06 Praha, Czech Republic; (V.N.); (A.N.)
| | - Przemysław Mitura
- Department of Urology and Oncological Urology, Medical University of Lublin, 20-059 Lublin, Poland;
| | - Lech Romanowicz
- Department of Medical Biochemistry, Medical University of Białystok, ul. Mickiewicza 2, 15-089 Białystok, Poland; (A.T.); (L.R.)
| |
Collapse
|
4
|
Xia Y, Wang X, Lin J, Li Y, Dong L, Liang X, Wang HY, Ding X, Wang Q. Gastric cancer fibroblasts affect the effect of immunotherapy and patient prognosis by inducing micro-vascular production. Front Immunol 2024; 15:1375013. [PMID: 39040110 PMCID: PMC11260615 DOI: 10.3389/fimmu.2024.1375013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/28/2024] [Indexed: 07/24/2024] Open
Abstract
Introduction Immunotherapy is critical for treating many cancers, and its therapeutic success is linked to the tumor microenvironment. Although anti-angiogenic drugs are used to treat gastric cancer (GC), their efficacy remains limited. Cancer-associated fibroblast (CAF)-targeted therapies complement immunotherapy; however, the lack of CAF-specific markers poses a challenge. Therefore, we developed a CAF angiogenesis prognostic score (CAPS) system to evaluate prognosis and immunotherapy response in patients with GC, aiming to improve patient stratification and treatment efficacy. Methods We assessed patient-derived GC CAFs for promoting angiogenesis using EdU, cell cycle, apoptosis, wound healing, and angiogenesis analysis. Results We then identified CAF-angiogenesis-associated differentially-expressed genes, leading to the development of CAPS, which included THBS1, SPARC, EDNRA, and VCAN. We used RT-qPCR to conduct gene-level validation, and eight GEO datasets and the HPA database to validate the CAPS system at the gene and protein levels. Six independent GEO datasets were utilized for validation. Overall survival time was shorter in the high- than the low-CAPS group. Immune microenvironment and immunotherapy response analysis showed that the high-CAPS group had a greater tendency toward immune escape and reduced immunotherapy efficacy than the low-CAPS group. Discussion CAPS is closely associated with GC prognosis and immunotherapy outcomes. It is therefore an independent predictor of GC prognosis and immunotherapy efficacy.
Collapse
Affiliation(s)
- Yan Xia
- National Institute of Traditional Chinese Medicine (TCM) Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaolu Wang
- National Institute of Traditional Chinese Medicine (TCM) Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jie Lin
- National Institute of Traditional Chinese Medicine (TCM) Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yuan Li
- National Institute of Traditional Chinese Medicine (TCM) Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Lidan Dong
- National Institute of Traditional Chinese Medicine (TCM) Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xue Liang
- National Institute of Traditional Chinese Medicine (TCM) Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Huai-Yu Wang
- National Institute of Traditional Chinese Medicine (TCM) Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xia Ding
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qi Wang
- National Institute of Traditional Chinese Medicine (TCM) Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
5
|
Moshrefiravasjani R, Kamrani A, Nazari N, Jafari F, Nasiri H, Jahanban-Esfahlan R, Akbari M. Exosome-mediated tumor metastasis: Biology, molecular targets and immuno-therapeutic options. Pathol Res Pract 2024; 254:155083. [PMID: 38277749 DOI: 10.1016/j.prp.2023.155083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 12/26/2023] [Accepted: 12/30/2023] [Indexed: 01/28/2024]
Abstract
Small extracellular vesicles called exosomes play a crucial part in promoting intercellular communication. They act as intermediaries for the exchange of bioactive chemicals between cells, released into the extracellular milieu by a variety of cell types. Within the context of cancer progression, metastasis is a complex process that plays a significant role in the spread of malignant cells from their main site of origin to distant anatomical locations. This complex process plays a key role in the domain of cancer-related deaths. In summary, the trajectory of current research in the field of exosome-mediated metastasis is characterized by its unrelenting quest for more profound understanding of the molecular nuances, the development of innovative diagnostic tools and therapeutic approaches, and the unwavering dedication to transforming these discoveries into revolutionary clinical applications. This unrelenting pursuit represents a shared desire to improve the prognosis for individuals suffering from metastatic cancer and to nudge the treatment paradigm in the direction of more effective and customized interventions.
Collapse
Affiliation(s)
| | - Amin Kamrani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran
| | - Nazanin Nazari
- Department of Immunology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farzaneh Jafari
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hadi Nasiri
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rana Jahanban-Esfahlan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Morteza Akbari
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
6
|
Padmanabhan J, Chen K, Sivaraj D, Henn D, Kuehlmann BA, Kussie HC, Zhao ET, Kahn A, Bonham CA, Dohi T, Beck TC, Trotsyuk AA, Stern-Buchbinder ZA, Than PA, Hosseini HS, Barrera JA, Magbual NJ, Leeolou MC, Fischer KS, Tigchelaar SS, Lin JQ, Perrault DP, Borrelli MR, Kwon SH, Maan ZN, Dunn JCY, Nazerali R, Januszyk M, Prantl L, Gurtner GC. Allometrically scaling tissue forces drive pathological foreign-body responses to implants via Rac2-activated myeloid cells. Nat Biomed Eng 2023; 7:1419-1436. [PMID: 37749310 PMCID: PMC10651488 DOI: 10.1038/s41551-023-01091-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 08/02/2023] [Indexed: 09/27/2023]
Abstract
Small animals do not replicate the severity of the human foreign-body response (FBR) to implants. Here we show that the FBR can be driven by forces generated at the implant surface that, owing to allometric scaling, increase exponentially with body size. We found that the human FBR is mediated by immune-cell-specific RAC2 mechanotransduction signalling, independently of the chemistry and mechanical properties of the implant, and that a pathological FBR that is human-like at the molecular, cellular and tissue levels can be induced in mice via the application of human-tissue-scale forces through a vibrating silicone implant. FBRs to such elevated extrinsic forces in the mice were also mediated by the activation of Rac2 signalling in a subpopulation of mechanoresponsive myeloid cells, which could be substantially reduced via the pharmacological or genetic inhibition of Rac2. Our findings provide an explanation for the stark differences in FBRs observed in small animals and humans, and have implications for the design and safety of implantable devices.
Collapse
Affiliation(s)
- Jagannath Padmanabhan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Kellen Chen
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Surgery, University of Arizona College of Medicine, Tucson, AZ, USA.
| | - Dharshan Sivaraj
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Surgery, University of Arizona College of Medicine, Tucson, AZ, USA.
| | - Dominic Henn
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Plastic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Britta A Kuehlmann
- Department of Plastic and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Hudson C Kussie
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Eric T Zhao
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Anum Kahn
- Cell Sciences Imaging Facility (CSIF), Beckman Center, Stanford University, Stanford, CA, USA
| | - Clark A Bonham
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Teruyuki Dohi
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Thomas C Beck
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Artem A Trotsyuk
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Zachary A Stern-Buchbinder
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Peter A Than
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Hadi S Hosseini
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Janos A Barrera
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Noah J Magbual
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Melissa C Leeolou
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Katharina S Fischer
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Seth S Tigchelaar
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - John Q Lin
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - David P Perrault
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Mimi R Borrelli
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Sun Hyung Kwon
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Zeshaan N Maan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - James C Y Dunn
- Division of Pediatric Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Rahim Nazerali
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael Januszyk
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Lukas Prantl
- Department of Plastic and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Geoffrey C Gurtner
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Surgery, University of Arizona College of Medicine, Tucson, AZ, USA.
| |
Collapse
|
7
|
Tufail M. Unlocking the potential of the tumor microenvironment for cancer therapy. Pathol Res Pract 2023; 251:154846. [PMID: 37837860 DOI: 10.1016/j.prp.2023.154846] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/15/2023] [Accepted: 10/02/2023] [Indexed: 10/16/2023]
Abstract
The tumor microenvironment (TME) holds a crucial role in the progression of cancer. Epithelial-derived tumors share common traits in shaping the TME. The Warburg effect is a notable phenomenon wherein tumor cells exhibit resistance to apoptosis and an increased reliance on anaerobic glycolysis for energy production. Recognizing the pivotal role of the TME in controlling tumor growth and influencing responses to chemotherapy, researchers have focused on developing potential cancer treatment strategies. A wide array of therapies, including immunotherapies, antiangiogenic agents, interventions targeting cancer-associated fibroblasts (CAF), and therapies directed at the extracellular matrix, have been under investigation and have demonstrated efficacy. Additionally, innovative techniques such as tumor tissue explants, "tumor-on-a-chip" models, and multicellular tumor spheres have been explored in laboratory research. This comprehensive review aims to provide insights into the intricate cross-talk between cancer-associated signaling pathways and the TME in cancer progression, current therapeutic approaches targeting the TME, the immune landscape within solid tumors, the role of the viral TME, and cancer cell metabolism.
Collapse
Affiliation(s)
- Muhammad Tufail
- Institute of Biomedical Sciences, Shanxi University, Taiyuan 030006, China.
| |
Collapse
|
8
|
Maoga JB, Riaz MA, Mwaura AN, Mecha E, Omwandho COA, Scheiner-Bobis G, Meinhold-Heerlein I, Konrad L. Analysis of Membrane Type-1 Matrix Metalloproteinase (MT1-MMP, MMP14) in Eutopic and Ectopic Endometrium and in Serum and Endocervical Mucus of Endometriosis. Biomedicines 2023; 11:2730. [PMID: 37893104 PMCID: PMC10604514 DOI: 10.3390/biomedicines11102730] [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: 08/22/2023] [Revised: 09/26/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Membrane type-matrix metalloproteinases (MT-MMPs) are a subgroup of the matrix metalloproteinases (MMPs) family and are key molecules in the degradation of the extracellular matrix. Membrane type-1 matrix metalloproteinase (MT1-MMP, MMP14) is often deregulated in different cancer tissues and body fluids of human cancer patients; however, MT1-MMP levels in endometriosis and adenomyosis patients are currently unknown. MATERIALS AND METHODS Tissue samples from patients with and without endometriosis or adenomyosis were analyzed with immunohistochemistry for the localization of MT1-MMP. Serum and endocervical mucus samples from patients with and without endometriosis or adenomyosis were investigated with MT1-MMP ELISAs. RESULTS MT1-MMP was localized preferentially in the glands of eutopic and ectopic endometrium. MT1-MMP protein levels are significantly reduced in ovarian endometriosis (HSCORE = 31) versus eutopic endometrium (HSCORE = 91) and adenomyosis (HSCORE = 149), but significantly increased in adenomyosis (HSCORE = 149) compared to eutopic endometrium (HSCORE = 91). Similarly, analysis of the levels of MT1-MMP using enzyme-linked immune assays (ELISAs) demonstrated a significant increase in the concentrations of MT1-MMP in the serum of endometriosis patients (1.3 ± 0.8) versus controls (0.7 ± 0.2), but not in the endocervical mucus. Furthermore, MT1-MMP levels in the endocervical mucus of patients with endometriosis were notably reduced in patients using contraception (3.2 ± 0.4) versus those without contraception (3.8 ± 0.2). CONCLUSIONS Taken together, our findings showed an opposite regulation of MT1-MMP in the tissue of ovarian endometriosis and adenomyosis compared to eutopic endometrium without endometriosis but increased serum levels in patients with endometriosis.
Collapse
Affiliation(s)
- Jane B. Maoga
- Center of Gynecology and Obstetrics, Faculty of Medicine, Justus Liebig University Giessen, 35392 Giessen, Germany; (J.B.M.); (M.A.R.); (A.N.M.); (I.M.-H.)
| | - Muhammad A. Riaz
- Center of Gynecology and Obstetrics, Faculty of Medicine, Justus Liebig University Giessen, 35392 Giessen, Germany; (J.B.M.); (M.A.R.); (A.N.M.); (I.M.-H.)
| | - Agnes N. Mwaura
- Center of Gynecology and Obstetrics, Faculty of Medicine, Justus Liebig University Giessen, 35392 Giessen, Germany; (J.B.M.); (M.A.R.); (A.N.M.); (I.M.-H.)
| | - Ezekiel Mecha
- Department of Biochemistry, University of Nairobi, Nairobi P.O. Box 30197-00100, Kenya;
| | - Charles O. A. Omwandho
- Department of Health Sciences, Kirinyaga University, Kerugoya P.O. Box 143-10300, Kenya;
| | - Georgios Scheiner-Bobis
- Institute for Veterinary Physiology and Biochemistry, School of Veterinary Medicine, Justus Liebig University Giessen, 35392 Giessen, Germany;
| | - Ivo Meinhold-Heerlein
- Center of Gynecology and Obstetrics, Faculty of Medicine, Justus Liebig University Giessen, 35392 Giessen, Germany; (J.B.M.); (M.A.R.); (A.N.M.); (I.M.-H.)
| | - Lutz Konrad
- Center of Gynecology and Obstetrics, Faculty of Medicine, Justus Liebig University Giessen, 35392 Giessen, Germany; (J.B.M.); (M.A.R.); (A.N.M.); (I.M.-H.)
| |
Collapse
|
9
|
Closset L, Gultekin O, Salehi S, Sarhan D, Lehti K, Gonzalez-Molina J. The extracellular matrix - immune microenvironment crosstalk in cancer therapy: Challenges and opportunities. Matrix Biol 2023; 121:217-228. [PMID: 37524251 DOI: 10.1016/j.matbio.2023.07.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 08/02/2023]
Abstract
Targeting the tumour immune microenvironment (TIME) by cancer immunotherapy has led to improved patient outcomes. However, response to these treatments is heterogeneous and cancer-type dependant. The therapeutic activity of classical cancer therapies such as chemotherapy, radiotherapy, and surgical oncology is modulated by alterations of the TIME. A major regulator of immune cell function and resistance to both immune and classical therapies is the extracellular matrix (ECM). Concurrently, cancer therapies reshape the TIME as well as the ECM, causing both pro- and anti-tumour responses. Accordingly, the TIME-ECM crosstalk presents attractive opportunities to improve therapy outcomes. Here, we review the molecular crosstalk between the TIME and the ECM in cancer and its implications in cancer progression and clinical intervention. Additionally, we discuss examples and future directions of ECM and TIME co-targeting in combination with oncological therapies including surgery, chemotherapy, and radiotherapy.
Collapse
Affiliation(s)
- Lara Closset
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solnavägen 9, Stockholm 171 65, Sweden; Saint-Antoine Research center (CRSA), UMR_S 938, INSERM, Sorbonne Université, Paris F-75012, France
| | - Okan Gultekin
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solnavägen 9, Stockholm 171 65, Sweden
| | - Sahar Salehi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solnavägen 9, Stockholm 171 65, Sweden; Department of Women's and Children's Health, Division of Obstetrics and Gynecology, Karolinska Institutet, Stockholm, Sweden; Department of Pelvic Cancer, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Dhifaf Sarhan
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Kaisa Lehti
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solnavägen 9, Stockholm 171 65, Sweden; Department of Biomedical Laboratory Science, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jordi Gonzalez-Molina
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solnavägen 9, Stockholm 171 65, Sweden.
| |
Collapse
|
10
|
Molière S, Jaulin A, Tomasetto CL, Dali-Youcef N. Roles of Matrix Metalloproteinases and Their Natural Inhibitors in Metabolism: Insights into Health and Disease. Int J Mol Sci 2023; 24:10649. [PMID: 37445827 DOI: 10.3390/ijms241310649] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/21/2023] [Accepted: 06/24/2023] [Indexed: 07/15/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc-activated peptidases that can be classified into six major classes, including gelatinases, collagenases, stromelysins, matrilysins, membrane type metalloproteinases, and other unclassified MMPs. The activity of MMPs is regulated by natural inhibitors called tissue inhibitors of metalloproteinases (TIMPs). MMPs are involved in a wide range of biological processes, both in normal physiological conditions and pathological states. While some of these functions occur during development, others occur in postnatal life. Although the roles of several MMPs have been extensively studied in cancer and inflammation, their function in metabolism and metabolic diseases have only recently begun to be uncovered, particularly over the last two decades. This review aims to summarize the current knowledge regarding the metabolic roles of metalloproteinases in physiology, with a strong emphasis on adipose tissue homeostasis, and to highlight the consequences of impaired or exacerbated MMP actions in the development of metabolic disorders such as obesity, fatty liver disease, and type 2 diabetes.
Collapse
Affiliation(s)
- Sébastien Molière
- Institut de Génétique et de Biologie Moléculaire et Cellulaire Illkirch, 67400 Illkirch-Graffenstaden, France
- Centre National de la Recherche Scientifique, UMR 7104, 67400 Illkirch-Graffenstaden, France
- Institut National de la Santé et de la Recherche Médicale, U1258, 67400 Illkirch-Graffenstaden, France
- Faculté de Médecine, Université de Strasbourg, 67000 Strasbourg, France
- Department of Radiology, Strasbourg University Hospital, Hôpital de Hautepierre, Avenue Molière, 67200 Strasbourg, France
- Breast and Thyroid Imaging Unit, ICANS-Institut de Cancérologie Strasbourg Europe, 67200 Strasbourg, France
| | - Amélie Jaulin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire Illkirch, 67400 Illkirch-Graffenstaden, France
- Centre National de la Recherche Scientifique, UMR 7104, 67400 Illkirch-Graffenstaden, France
- Institut National de la Santé et de la Recherche Médicale, U1258, 67400 Illkirch-Graffenstaden, France
- Faculté de Médecine, Université de Strasbourg, 67000 Strasbourg, France
| | - Catherine-Laure Tomasetto
- Institut de Génétique et de Biologie Moléculaire et Cellulaire Illkirch, 67400 Illkirch-Graffenstaden, France
- Centre National de la Recherche Scientifique, UMR 7104, 67400 Illkirch-Graffenstaden, France
- Institut National de la Santé et de la Recherche Médicale, U1258, 67400 Illkirch-Graffenstaden, France
| | - Nassim Dali-Youcef
- Institut de Génétique et de Biologie Moléculaire et Cellulaire Illkirch, 67400 Illkirch-Graffenstaden, France
- Centre National de la Recherche Scientifique, UMR 7104, 67400 Illkirch-Graffenstaden, France
- Institut National de la Santé et de la Recherche Médicale, U1258, 67400 Illkirch-Graffenstaden, France
- Faculté de Médecine, Université de Strasbourg, 67000 Strasbourg, France
- Laboratoire de Biochimie et Biologie Moléculaire, Pôle de Biologie, Hôpitaux Universitaires de Strasbourg, Nouvel Hôpital Civil, 67000 Strasbourg, France
| |
Collapse
|
11
|
Carminati L, Carlessi E, Longhi E, Taraboletti G. Controlled extracellular proteolysis of thrombospondins. Matrix Biol 2023; 119:82-100. [PMID: 37003348 DOI: 10.1016/j.matbio.2023.03.011] [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: 12/22/2022] [Revised: 03/17/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023]
Abstract
Limited proteolysis of thrombospondins is a powerful mechanism to ensure dynamic tuning of their activities in the extracellular space. Thrombospondins are multifunctional matricellular proteins composed of multiple domains, each with a specific pattern of interactions with cell receptors, matrix components and soluble factors (growth factors, cytokines and proteases), thus with different effects on cell behavior and responses to changes in the microenvironment. Therefore, the proteolytic degradation of thrombospondins has multiple functional consequences, reflecting the local release of active fragments and isolated domains, exposure or disruption of active sequences, altered protein location, and changes in the composition and function of TSP-based pericellular interaction networks. In this review current data from the literature and databases is employed to provide an overview of cleavage of mammalian thrombospondins by different proteases. The roles of the fragments generated in specific pathological settings, with particular focus on cancer and the tumor microenvironment, are discussed.
Collapse
Affiliation(s)
- Laura Carminati
- Laboratory of Tumor Microenvironment, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 24126 Bergamo, Italy
| | - Elena Carlessi
- Laboratory of Tumor Microenvironment, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 24126 Bergamo, Italy
| | - Elisa Longhi
- Laboratory of Tumor Microenvironment, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 24126 Bergamo, Italy
| | - Giulia Taraboletti
- Laboratory of Tumor Microenvironment, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 24126 Bergamo, Italy.
| |
Collapse
|
12
|
Wiśniowski T, Bryda J, Wątroba S. The role of matrix metalloproteinases in pathogenesis, diagnostics, and treatment of human prostate cancer. POSTEP HIG MED DOSW 2023. [DOI: 10.2478/ahem-2023-0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2023] Open
Abstract
Abstract
The prostate gland is highly susceptible to oncogenic transformation, many times more than other sex tissues, such as seminal vesicles. In fact, prostate cancer (PCa) will be diagnosed in one in seven lifetime patients, making PCa the subject of intense research aimed at clarifying its biology and providing adequate treatment. PCa is the fourth most common cancer in the world in terms of the overall population and the second most common cancer for the male population. It is postulated that the development of PCa may be influenced by dietary factors, physical and sexual activity, androgens, obesity, and inflammation, but their role in the development of prostate cancer still remains unclear. Extracellular matrix metalloproteinases (MMPs) and tissue metalloproteinase inhibitors (TIMPs) play an important role in many physiological and pathological processes, including proliferation, migration, invasion, cell differentiation, participation in inflammatory processes and angiogenesis. Numerous studies point to a direct relationship between MMPs and both local tumor invasion and the formation of distant metastases. High activity of MMPs is observed in solid tumors of various origins, which positively correlates with a poor overall survival rate. Although biochemical diagnostic markers of PCa are currently available, from the point of view of clinical practice, it seems particularly important to develop new and more sensitive markers allowing for early diagnosis and long-term monitoring of patients after PCa treatment, and the assessment of MMP activity in urine and serum of patients are potential factors that could play such a role.
Collapse
|
13
|
Guo X, Cao J, Cai JP, Wu J, Huang J, Asthana P, Wong SKK, Ye ZW, Gurung S, Zhang Y, Wang S, Wang Z, Ge X, Kwan HY, Lyu A, Chan KM, Wong N, Huang J, Zhou Z, Bian ZX, Yuan S, Wong HLX. Control of SARS-CoV-2 infection by MT1-MMP-mediated shedding of ACE2. Nat Commun 2022; 13:7907. [PMID: 36564389 PMCID: PMC9780620 DOI: 10.1038/s41467-022-35590-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a global pandemic. Angiotensin-converting enzyme 2 (ACE2) is an entry receptor for SARS-CoV-2. The full-length membrane form of ACE2 (memACE2) undergoes ectodomain shedding to generate a shed soluble form (solACE2) that mediates SARS-CoV-2 entry via receptor-mediated endocytosis. Currently, it is not known how the physiological regulation of ACE2 shedding contributes to the etiology of COVID-19 in vivo. The present study identifies Membrane-type 1 Matrix Metalloproteinase (MT1-MMP) as a critical host protease for solACE2-mediated SARS-CoV-2 infection. SARS-CoV-2 infection leads to increased activation of MT1-MMP that is colocalized with ACE2 in human lung epithelium. Mechanistically, MT1-MMP directly cleaves memACE2 at M706-S to release solACE218-706 that binds to the SARS-CoV-2 spike proteins (S), thus facilitating cell entry of SARS-CoV-2. Human solACE218-706 enables SARS-CoV-2 infection in both non-permissive cells and naturally insusceptible C57BL/6 mice. Inhibition of MT1-MMP activities suppresses solACE2-directed entry of SARS-CoV-2 in human organoids and aged mice. Both solACE2 and circulating MT1-MMP are positively correlated in plasma of aged mice and humans. Our findings provide in vivo evidence demonstrating the contribution of ACE2 shedding to the etiology of COVID-19.
Collapse
Affiliation(s)
- Xuanming Guo
- grid.221309.b0000 0004 1764 5980School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Jianli Cao
- grid.194645.b0000000121742757Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Jian-Piao Cai
- grid.194645.b0000000121742757State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Jiayan Wu
- grid.221309.b0000 0004 1764 5980School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Jiangang Huang
- grid.12955.3a0000 0001 2264 7233Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Pallavi Asthana
- grid.221309.b0000 0004 1764 5980School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Sheung Kin Ken Wong
- grid.194645.b0000000121742757School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Zi-Wei Ye
- grid.194645.b0000000121742757Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Susma Gurung
- grid.221309.b0000 0004 1764 5980School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Yijing Zhang
- grid.221309.b0000 0004 1764 5980School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Sheng Wang
- grid.470187.dRespiratory Department, Jinhua Guangfu Hospital, Jinhua, China
| | - Zening Wang
- grid.267308.80000 0000 9206 2401Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX USA
| | - Xin Ge
- grid.267308.80000 0000 9206 2401Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX USA
| | - Hiu Yee Kwan
- grid.221309.b0000 0004 1764 5980School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Aiping Lyu
- grid.221309.b0000 0004 1764 5980School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Kui Ming Chan
- grid.35030.350000 0004 1792 6846Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Nathalie Wong
- grid.415197.f0000 0004 1764 7206Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, N.T., Hong Kong SAR, China
| | - Jiandong Huang
- grid.194645.b0000000121742757School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Zhongjun Zhou
- grid.194645.b0000000121742757School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Zhao-Xiang Bian
- grid.221309.b0000 0004 1764 5980Centre for Chinese Herbal Medicine Drug Development Limited, Hong Kong Baptist University, Hong Kong SAR, China
| | - Shuofeng Yuan
- grid.194645.b0000000121742757State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Hoi Leong Xavier Wong
- grid.221309.b0000 0004 1764 5980School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| |
Collapse
|
14
|
Ikeda K, Kaneko R, Tsukamoto E, Funahashi N, Koshikawa N. Proteolytic cleavage of membrane proteins by membrane type-1 MMP regulates cancer malignant progression. Cancer Sci 2022; 114:348-356. [PMID: 36336966 PMCID: PMC9899627 DOI: 10.1111/cas.15638] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/24/2022] [Accepted: 10/27/2022] [Indexed: 11/09/2022] Open
Abstract
Strategies to develop cancer therapies using inhibitors that target matrix metalloproteinases (MMPs), particularly membrane type-1 MMP (MT1-MMP), have failed. This is predominantly attributed to the specificity of MMP inhibitors and numerous functions of MMPs; therefore, targeting substrates with such broad specificity can lead to off-target effects. Thus, new drug development for cancer therapeutics should focus on the ability of MT1-MMP to break down substrates, such as functional cell membrane proteins, to regulate the functions of these proteins that promote tumor malignancy. In this review, we discuss the mechanism by which proteolysis of cell surface proteins by MT1-MMP promotes progression of malignant tumor cells. In addition, we discuss the two protein fragments generated by limited cleavage of erythropoietin-producing hepatoma receptor tyrosine kinase A2 (EphA2-NF, -CF), which represent a promising basis for developing new cancer therapies and diagnostic techniques.
Collapse
Affiliation(s)
- Kazuki Ikeda
- Department of Life Science and TechnologyTokyo Institute of TechnologyYokohamaJapan
| | - Ryo Kaneko
- Department of Life Science and TechnologyTokyo Institute of TechnologyYokohamaJapan
| | - Eiki Tsukamoto
- Department of Life Science and TechnologyTokyo Institute of TechnologyYokohamaJapan
| | - Nobuaki Funahashi
- Department of Life Science and TechnologyTokyo Institute of TechnologyYokohamaJapan
| | - Naohiko Koshikawa
- Department of Life Science and TechnologyTokyo Institute of TechnologyYokohamaJapan,Clinical Proteomics LaboratoryKanagawa Cancer Center Research InstituteYokohamaJapan
| |
Collapse
|
15
|
Cayetano-Salazar L, de la Cruz-Concepción B, Navarro-Tito N, Álvarez-Fitz P, Leyva-Vázquez MA, Acevedo-Quiroz M, Zacapala-Gómez AE, Ortuño-Pineda C, Martinez-Carrillo DN, Castañeda-Saucedo E, García-Hernández AP, Mendoza-Catalán MA. Ficus crocata leaf extracts decrease the proliferation and invasiveness of breast cancer cells. Heliyon 2022; 8:e11405. [DOI: 10.1016/j.heliyon.2022.e11405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 10/28/2022] [Accepted: 10/31/2022] [Indexed: 11/08/2022] Open
|
16
|
Schoutrop E, Moyano-Galceran L, Lheureux S, Mattsson J, Lehti K, Dahlstrand H, Magalhaes I. Molecular, cellular and systemic aspects of epithelial ovarian cancer and its tumor microenvironment. Semin Cancer Biol 2022; 86:207-223. [PMID: 35395389 DOI: 10.1016/j.semcancer.2022.03.027] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 03/11/2022] [Accepted: 03/30/2022] [Indexed: 02/07/2023]
Abstract
Ovarian cancer encompasses a heterogeneous group of malignancies that involve the ovaries, fallopian tubes and the peritoneal cavity. Despite major advances made within the field of cancer, the majority of patients with ovarian cancer are still being diagnosed at an advanced stage of the disease due to lack of effective screening tools. The overall survival of these patients has, therefore, not substantially improved over the past decades. Most patients undergo debulking surgery and treatment with chemotherapy, but often micrometastases remain and acquire resistance to the therapy, eventually leading to disease recurrence. Here, we summarize the current knowledge in epithelial ovarian cancer development and metastatic progression. For the most common subtypes, we focus further on the properties and functions of the immunosuppressive tumor microenvironment, including the extracellular matrix. Current and future treatment modalities are discussed and finally we provide an overview of the different experimental models used to develop novel therapies.
Collapse
Affiliation(s)
- Esther Schoutrop
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Lidia Moyano-Galceran
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Stephanie Lheureux
- University of Toronto, Toronto, Ontario, Canada; Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Jonas Mattsson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; University of Toronto, Toronto, Ontario, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; Gloria and Seymour Epstein Chair in Cell Therapy and Transplantation, Toronto, Ontario, Canada
| | - Kaisa Lehti
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden; Department of Biomedical Laboratory Science, Norwegian University of Science and Technology, Trondheim, Norway
| | - Hanna Dahlstrand
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Medical unit Pelvic Cancer, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden.
| | - Isabelle Magalhaes
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
17
|
Gonzalez‐Molina J, Kirchhof KM, Rathod B, Moyano‐Galceran L, Calvo‐Noriega M, Kokaraki G, Bjørkøy A, Ehnman M, Carlson JW, Lehti K. Mechanical Confinement and DDR1 Signaling Synergize to Regulate Collagen-Induced Apoptosis in Rhabdomyosarcoma Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2202552. [PMID: 35957513 PMCID: PMC9534977 DOI: 10.1002/advs.202202552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/29/2022] [Indexed: 06/15/2023]
Abstract
Fibrillar collagens promote cell proliferation, migration, and survival in various epithelial cancers and are generally associated with tumor aggressiveness. However, the impact of fibrillar collagens on soft tissue sarcoma behavior remains poorly understood. Unexpectedly, this study finds that fibrillar collagen-related gene expression is associated with favorable patient prognosis in rhabdomyosarcoma. By developing and using collagen matrices with distinct stiffness and in vivo-like microarchitectures, this study uncovers that the activation of DDR1 has pro-apoptotic and of integrin β1 pro-survival function, specifically in 3D rhabdomyosarcoma cell cultures. It demonstrates that rhabdomyosarcoma cell-intrinsic or extrinsic matrix remodeling promotes cell survival. Mechanistically, the 3D-specific collagen-induced apoptosis results from a dual DDR1-independent and a synergistic DDR1-dependent TRPV4-mediated response to mechanical confinement. Altogether, these results indicate that dense microfibrillar collagen-rich microenvironments are detrimental to rhabdomyosarcoma cells through an apoptotic response orchestrated by the induction of DDR1 signaling and mechanical confinement. This mechanism helps to explain the preference of rhabdomyosarcoma cells to grow in and metastasize to low fibrillar collagen microenvironments such as the lung.
Collapse
Affiliation(s)
- Jordi Gonzalez‐Molina
- Department of MicrobiologyTumor and Cell BiologyKarolinska InstitutetSolnavägen 9Solna17165Sweden
- Department of Oncology‐PathologyKarolinska InstitutetKarolinskavägenSolna17164Sweden
| | - Katharina Miria Kirchhof
- Department of MicrobiologyTumor and Cell BiologyKarolinska InstitutetSolnavägen 9Solna17165Sweden
| | - Bhavik Rathod
- Department of MicrobiologyTumor and Cell BiologyKarolinska InstitutetSolnavägen 9Solna17165Sweden
- Department of Laboratory MedicineDivision of PathologyKarolinska InstitutetAlfred Nobels Allé 8Stockholm14152Sweden
| | - Lidia Moyano‐Galceran
- Department of MicrobiologyTumor and Cell BiologyKarolinska InstitutetSolnavägen 9Solna17165Sweden
| | - Maria Calvo‐Noriega
- Department of MicrobiologyTumor and Cell BiologyKarolinska InstitutetSolnavägen 9Solna17165Sweden
| | - Georgia Kokaraki
- Department of Oncology‐PathologyKarolinska InstitutetKarolinskavägenSolna17164Sweden
- Keck School of MedicineUniversity of Southern California1975 Zonal AveLos AngelesCA90033USA
| | - Astrid Bjørkøy
- Department of PhysicsNorwegian University of Science and TechnologyHøgskoleringen 5TrondheimNO‐7491Norway
| | - Monika Ehnman
- Department of Oncology‐PathologyKarolinska InstitutetKarolinskavägenSolna17164Sweden
| | - Joseph W. Carlson
- Department of Oncology‐PathologyKarolinska InstitutetKarolinskavägenSolna17164Sweden
- Keck School of MedicineUniversity of Southern California1975 Zonal AveLos AngelesCA90033USA
| | - Kaisa Lehti
- Department of MicrobiologyTumor and Cell BiologyKarolinska InstitutetSolnavägen 9Solna17165Sweden
- Department of Biomedical Laboratory ScienceNorwegian University of Science and TechnologyErling Skjalgssons gate 1TrondheimNO‐7491Norway
| |
Collapse
|
18
|
Epithelial-to-Mesenchymal Transition in Metastasis: Focus on Laryngeal Carcinoma. Biomedicines 2022; 10:biomedicines10092148. [PMID: 36140250 PMCID: PMC9496235 DOI: 10.3390/biomedicines10092148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/27/2022] [Accepted: 08/30/2022] [Indexed: 11/18/2022] Open
Abstract
In epithelial neoplasms, such as laryngeal carcinoma, the survival indexes deteriorate abruptly when the tumor becomes metastatic. A molecular phenomenon that normally appears during embryogenesis, epithelial-to-mesenchymal transition (EMT), is reactivated at the initial stage of metastasis when tumor cells invade the adjacent stroma. The hallmarks of this phenomenon are the abolishment of the epithelial and acquisition of mesenchymal traits by tumor cells which enhance their migratory capacity. EMT signaling is mediated by complex molecular pathways that regulate the expression of crucial molecules contributing to the tumor’s metastatic potential. Effectors of EMT include loss of adhesion, cytoskeleton remodeling, evasion of apoptosis and immune surveillance, upregulation of metalloproteinases, neovascularization, acquisition of stem-cell properties, and the activation of tumor stroma. However, the current approach to EMT involves a holistic model that incorporates the acquisition of potentials beyond mesenchymal transition. As EMT is inevitably associated with a reverse mesenchymal-to-epithelial transition (MET), a model of partial EMT is currently accepted, signifying the cell plasticity associated with invasion and metastasis. In this review, we identify the cumulative evidence which suggests that various aspects of EMT theory apply to laryngeal carcinoma, a tumor of significant morbidity and mortality, introducing novel molecular targets with prognostic and therapeutic potential.
Collapse
|
19
|
Jin Y, Liang ZY, Zhou WX, Zhou L. An MMP-based risk score strongly distinguishes prognosis in hepatocellular carcinoma after resection. Future Oncol 2022; 18:2903-2917. [PMID: 35861053 DOI: 10.2217/fon-2021-1558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To first explore the prognostic value of MMP11 and MMP15 in hepatocellular carcinoma. Methods: MMP11/MMP15 expression was immunohistochemically detected and correlated with clinicopathologic variables and survival and confirmed in publicly available databases. An MMP-based risk score (MMPRS) was established. Results: Tumoral MMP11/MMP15 expression was higher and univariately associated with crucial clinicopathologic parameters, overall survival and disease-free survival in all patients and/or many subsets. Multivariately, MMP11/MMP15 expression remained significant. Their overexpression and prognostic value were confirmed in the Ualcan and Kaplan-Meier plotter databases. Critically, the novel MMPRS integrating MMP11, MMP15 and tumor-node-metastasis stage identified subgroups with the best and worst prognoses, with much higher predictive power. Conclusion: MMP11 and MMP15 served as prognosticators in hepatocellular carcinoma. MMPRS might work more accurately.
Collapse
Affiliation(s)
- Ye Jin
- Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Zhi-Yong Liang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Wei-Xun Zhou
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Li Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| |
Collapse
|
20
|
Di Trani CA, Cirella A, Arrizabalaga L, Fernandez-Sendin M, Bella A, Aranda F, Melero I, Berraondo P. Overcoming the limitations of cytokines to improve cancer therapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 369:107-141. [PMID: 35777862 DOI: 10.1016/bs.ircmb.2022.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Cytokines are pleiotropic soluble proteins used by immune cells to orchestrate a coordinated response against pathogens and malignancies. In cancer immunotherapy, cytokine-based drugs can be developed potentiating pro-inflammatory cytokines or blocking immunosuppressive cytokines. However, the complexity of the mechanisms of action of cytokines requires the use of biotechnological strategies to minimize systemic toxicity, while potentiating the antitumor response. Sequence mutagenesis, fusion proteins and gene therapy strategies are employed to enhance the half-life in circulation, target the desired bioactivity to the tumor microenvironment, and to optimize the therapeutic window of cytokines. In this review, we provide an overview of the different strategies currently being pursued in pre-clinical and clinical studies to make the most of cytokines for cancer immunotherapy.
Collapse
Affiliation(s)
- Claudia Augusta Di Trani
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Assunta Cirella
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Leire Arrizabalaga
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Myriam Fernandez-Sendin
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Angela Bella
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Fernando Aranda
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Ignacio Melero
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
| |
Collapse
|
21
|
Goïta AA, Guenot D. Colorectal Cancer: The Contribution of CXCL12 and Its Receptors CXCR4 and CXCR7. Cancers (Basel) 2022; 14:1810. [PMID: 35406582 PMCID: PMC8997717 DOI: 10.3390/cancers14071810] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/22/2022] [Accepted: 03/29/2022] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer is one of the most common cancers, and diagnosis at late metastatic stages is the main cause of death related to this cancer. This progression to metastasis is complex and involves different molecules such as the chemokine CXCL12 and its two receptors CXCR4 and CXCR7. The high expression of receptors in CRC is often associated with a poor prognosis and aggressiveness of the tumor. The interaction of CXCL12 and its receptors activates signaling pathways that induce chemotaxis, proliferation, migration, and cell invasion. To this end, receptor inhibitors were developed, and their use in preclinical and clinical studies is ongoing. This review provides an overview of studies involving CXCR4 and CXCR7 in CRC with an update on their targeting in anti-cancer therapies.
Collapse
Affiliation(s)
| | - Dominique Guenot
- INSERM U1113/Unistra, IRFAC—Interface de Recherche Fondamentale et Appliquée en Cancérologie, 67200 Strasbourg, France;
| |
Collapse
|
22
|
Li X, Li K, Li M, Lin X, Mei Y, Huang X, Yang H. Chemoresistance Transmission via Exosome-Transferred MMP14 in Pancreatic Cancer. Front Oncol 2022; 12:844648. [PMID: 35223528 PMCID: PMC8865617 DOI: 10.3389/fonc.2022.844648] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 01/17/2022] [Indexed: 12/19/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest malignancies. Gemcitabine is the most commonly used chemotherapy for the treatment of PDAC, but the development of drug resistance still remains challenging. Recently, exosomes have emerged as important mediators for intercellular communication. Exosomes affect recipient cells’ behavior through the engulfed cargos, however the specific cargos responsible for gemcitabine resistance in PDAC are poorly understood. Here, we reported that exosomes could transfer gemcitabine resistance via a metalloproteinase 14 (MMP14)-dependent mechanism. MMP14 was identified as a major differentially secreted protein from the gemcitabine-resistant PDAC cells by comparative secretome. It was packaged into the exosomes and transmitted from the chemoresistant cells to the sensitive ones. The exosome-transferred MMP14 could enhance drug resistance and promotes the sphere-formation and migration abilities of the recipient sensitive PDAC cells. Mechanically, exosome-transferred MMP14 promotes the stability of CD44, the cancer stem cell marker in the recipient cells. Our results indicate that MMP14 is a key player for exosome-mediated transfer of gemcitabine resistance, thus targeting MMP14 in exosomes may represent a novel strategy to limit gemcitabine resistance in PDAC.
Collapse
Affiliation(s)
- Xinyuan Li
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Kai Li
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Mengmeng Li
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Xiaoyu Lin
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Yu Mei
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Xuemei Huang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Huanjie Yang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| |
Collapse
|
23
|
Liang Z, Yu J, Gu D, Liu X, Liu J, Wu M, Xu M, Shen M, Duan W, Li W. M2-phenotype tumour-associated macrophages upregulate the expression of prognostic predictors MMP14 and INHBA in pancreatic cancer. J Cell Mol Med 2022; 26:1540-1555. [PMID: 35150061 PMCID: PMC8899166 DOI: 10.1111/jcmm.17191] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 12/11/2021] [Accepted: 12/17/2021] [Indexed: 12/25/2022] Open
Abstract
Pancreatic cancer is one of the most lethal gastrointestinal tumours, the most common pathological type is pancreatic adenocarcinoma (PAAD). In recent year, immune imbalanced in tumour microenvironment has been shown to play an important role in the evolution of tumours progression, and the efficacy of immunotherapy has been gradually demonstrated in clinical practice. In this study, we propose to construct an immune-related prognostic risk model based on immune-related genes MMP14 and INHBA expression that can assess the prognosis of pancreatic cancer patients and identify potential therapeutic targets for pancreatic cancer, to provide new ideas for the treatment of pancreatic cancer. We also investigate the correlation between macrophage infiltration and MMP14 and INHBA expression. First, the gene expression data of pancreatic cancer and normal pancreatic tissue were obtained from The Cancer Genome Atlas Program (TCGA) and The Genotype-Tissue Expression public database (GTEx). The differentially expressed immune-related genes between pancreatic cancer samples and normal sample were screened by R software. Secondly, univariate Cox regression analysis were used to evaluate the relationship between immune-related genes and the prognosis of pancreatic cancer patients. A polygenic risk score model was constructed by Cox regression analysis. The prognostic nomogram was constructed, and its performance was evaluated comprehensively by internal calibration curve and C-index. Using the risk model, each patient gets a risk score, and was divided into high- or low- risk groups. The proportion of 22 types of immune cells infiltration in pancreatic cancer samples was inferred by CIBERSOFT algorithm, correlation analysis (Pearson method) was used to analyse the correlation between the immune-related genes and immunes cells. Then, we applied macrophage conditioned medium to culture pancreatic cancer cell line PANC1, detected the expression of MMP14 and INHBA by qRT-PCR and Western blot methods. Knock-down MMP14 and INHBA in PANC1 cells by transfected with shRNA lentiviruses. Detection of migration ability of pancreatic cells was done by trans-well cell migration assay. A subcutaneous xenograft tumour model of human pancreatic cancer in nude mice was constructed. In conclusion, an immune-related gene prognostic model was constructed, patients with high-risk scores have poorer survival status, M2-phenotype tumour-associated macrophages (TAMs) up-regulate two immune-related genes, MMP14 and INHBA, which were used to establish the prognostic model. Knock-down of MMP14 and INHBA inhibited invasion of pancreatic cancer.
Collapse
Affiliation(s)
- Zhan‐Wen Liang
- Department of OncologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Jie Yu
- Department of PathologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Dong‐Mei Gu
- Department of PathologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Xiao‐Meng Liu
- Department of OncologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Jin Liu
- Department of OncologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Meng‐Yao Wu
- Department of OncologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Meng‐Dan Xu
- Department of OncologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Meng Shen
- Department of OncologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Weiming Duan
- Department of OncologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Wei Li
- Department of OncologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
24
|
Xue D, Moon B, Liao J, Guo J, Zou Z, Han Y, Cao S, Wang Y, Fu YX, Peng H. A tumor-specific pro-IL-12 activates preexisting cytotoxic T cells to control established tumors. Sci Immunol 2022; 7:eabi6899. [PMID: 34995098 PMCID: PMC9009736 DOI: 10.1126/sciimmunol.abi6899] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
It is a challenge to effectively reactivate preexisting tumor-infiltrating lymphocytes (TILs) without causing severe toxicity. Interleukin-12 (IL-12) can potently activate lymphocytes, but its clinical use is limited by its short half-life and dose-related toxicity. In this study, we developed a tumor-conditional IL-12 (pro-IL-12), which masked IL-12 with selective extracellular receptor–binding domains of the IL-12 receptor while preferentially and persistently activating TILs after being unmasked by matrix metalloproteinases expressed by tumors. Systemic delivery of pro-IL-12 demonstrated reduced toxicity but better control of established tumors compared with IL-12-Fc. Mechanistically, antitumor responses induced by pro-IL-12 were dependent on TILs and IFNγ. Furthermore, direct binding of IL-12 to IL-12R on CD8+, not CD4+, T cells was essential for maximal effectiveness. Pro-IL-12 improved the efficacy of both immune checkpoint blockade and targeted therapy when used in combination. Therefore, our study demonstrated that pro-IL-12 could rejuvenate TILs, which then combined with current treatment modalities while limiting adverse effects for treating established tumors.
Collapse
Affiliation(s)
- Diyuan Xue
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Benjamin Moon
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Jing Liao
- G uangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Jingya Guo
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhuangzhi Zou
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanfei Han
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuaishuai Cao
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Wang
- Immune Targeting Inc, Dallas, Texas 75247
| | - Yang-Xin Fu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Hua Peng
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
25
|
Matrix Metalloproteinases Shape the Tumor Microenvironment in Cancer Progression. Int J Mol Sci 2021; 23:ijms23010146. [PMID: 35008569 PMCID: PMC8745566 DOI: 10.3390/ijms23010146] [Citation(s) in RCA: 162] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer progression with uncontrolled tumor growth, local invasion, and metastasis depends largely on the proteolytic activity of numerous matrix metalloproteinases (MMPs), which affect tissue integrity, immune cell recruitment, and tissue turnover by degrading extracellular matrix (ECM) components and by releasing matrikines, cell surface-bound cytokines, growth factors, or their receptors. Among the MMPs, MMP-14 is the driving force behind extracellular matrix and tissue destruction during cancer invasion and metastasis. MMP-14 also influences both intercellular as well as cell-matrix communication by regulating the activity of many plasma membrane-anchored and extracellular proteins. Cancer cells and other cells of the tumor stroma, embedded in a common extracellular matrix, interact with their matrix by means of various adhesive structures, of which particularly invadopodia are capable to remodel the matrix through spatially and temporally finely tuned proteolysis. As a deeper understanding of the underlying functional mechanisms is beneficial for the development of new prognostic and predictive markers and for targeted therapies, this review examined the current knowledge of the interplay of the various MMPs in the cancer context on the protein, subcellular, and cellular level with a focus on MMP14.
Collapse
|
26
|
Wu PS, Wang CY, Chen PS, Hung JH, Yen JH, Wu MJ. 8-Hydroxydaidzein Downregulates JAK/STAT, MMP, Oxidative Phosphorylation, and PI3K/AKT Pathways in K562 Cells. Biomedicines 2021; 9:biomedicines9121907. [PMID: 34944720 PMCID: PMC8698423 DOI: 10.3390/biomedicines9121907] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/05/2021] [Accepted: 12/10/2021] [Indexed: 12/12/2022] Open
Abstract
A metabolite isolated from fermented soybean, 8-hydroxydaidzein (8-OHD, 7,8,4′-trihydroxyisoflavone, NSC-678112), is widely used in ethnopharmacological research due to its anti-proliferative and anti-inflammatory effects. We reported previously that 8-OHD provoked reactive oxygen species (ROS) overproduction, and induced autophagy, apoptosis, breakpoint cluster region-Abelson murine leukemia viral oncogene (BCR-ABL) degradation, and differentiation in K562 human chronic myeloid leukemia (CML) cells. However, how 8-OHD regulates metabolism, the extracellular matrix during invasion and metastasis, and survival signaling pathways in CML remains largely unexplored. High-throughput technologies have been widely used to discover the therapeutic targets and pathways of drugs. Bioinformatics analysis of 8-OHD-downregulated differentially expressed genes (DEGs) revealed that Janus kinase/signal transducer and activator of transcription (JAK/STAT), matrix metalloproteinases (MMPs), c-Myc, phosphoinositide 3-kinase (PI3K)/AKT, and oxidative phosphorylation (OXPHOS) metabolic pathways were significantly altered by 8-OHD treatment. Western blot analyses validated that 8-OHD significantly downregulated cytosolic JAK2 and the expression and phosphorylation of STAT3 dose- and time-dependently in K562 cells. Zymography and transwell assays also confirmed that K562-secreted MMP9 and invasion activities were dose-dependently inhibited by 8-OHD after 24 h of treatment. RT-qPCR analyses verified that 8-OHD repressed metastasis and OXPHOS-related genes. In combination with DisGeNET, it was found that 8-OHD’s downregulation of PI3K/AKT is crucial for controlling CML development. A STRING protein–protein interaction analysis further revealed that AKT and MYC are hub proteins for cancer progression. Western blotting revealed that AKT phosphorylation and nuclear MYC expression were significantly inhibited by 8-OHD. Collectively, this systematic investigation revealed that 8-OHD exerts anti-CML effects by downregulating JAK/STAT, PI3K/AKT, MMP, and OXPHOS pathways, and MYC expression. These results could shed new light on the development of 8-OHD for CML therapy.
Collapse
Affiliation(s)
- Pei-Shan Wu
- Department of Applied Life Science and Health, Chia Nan University of Pharmacy and Science, Tainan 717, Taiwan; (P.-S.W.); (P.-S.C.)
| | - Chih-Yang Wang
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, Taipei Medical University, Taipei 11031, Taiwan;
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei 11031, Taiwan
| | - Pin-Shern Chen
- Department of Applied Life Science and Health, Chia Nan University of Pharmacy and Science, Tainan 717, Taiwan; (P.-S.W.); (P.-S.C.)
- Department of Biotechnology, Chia Nan University of Pharmacy and Science, Tainan 717, Taiwan;
| | - Jui-Hsiang Hung
- Department of Biotechnology, Chia Nan University of Pharmacy and Science, Tainan 717, Taiwan;
| | - Jui-Hung Yen
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien 970, Taiwan;
- Institute of Medical Sciences, Tzu Chi University, Hualien 970, Taiwan
| | - Ming-Jiuan Wu
- Department of Applied Life Science and Health, Chia Nan University of Pharmacy and Science, Tainan 717, Taiwan; (P.-S.W.); (P.-S.C.)
- Department of Biotechnology, Chia Nan University of Pharmacy and Science, Tainan 717, Taiwan;
- Correspondence: or ; Tel.: +886-6-2664911 (ext. 2520)
| |
Collapse
|
27
|
Muntz I, Fenu M, van Osch GJVM, Koenderink G. The role of cell-matrix interactions in connective tissue mechanics. Phys Biol 2021; 19. [PMID: 34902848 DOI: 10.1088/1478-3975/ac42b8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 12/13/2021] [Indexed: 11/12/2022]
Abstract
Living tissue is able to withstand large stresses in everyday life, yet it also actively adapts to dynamic loads. This remarkable mechanical behaviour emerges from the interplay between living cells and their non-living extracellular environment. Here we review recent insights into the biophysical mechanisms involved in the reciprocal interplay between cells and the extracellular matrix and how this interplay determines tissue mechanics, with a focus on connective tissues. We first describe the roles of the main macromolecular components of the extracellular matrix in regards to tissue mechanics. We then proceed to highlight the main routes via which cells sense and respond to their biochemical and mechanical extracellular environment. Next we introduce the three main routes via which cells can modify their extracellular environment: exertion of contractile forces, secretion and deposition of matrix components, and matrix degradation. Finally we discuss how recent insights in the mechanobiology of cell-matrix interactions are furthering our understanding of the pathophysiology of connective tissue diseases and cancer, and facilitating the design of novel strategies for tissue engineering.
Collapse
Affiliation(s)
- Iain Muntz
- Bionanoscience, TU Delft, Kavli Institute of Nanoscience Delft, Delft University of Technology, Van der Maasweg 9, Delft, Zuid-Holland, 2629 HC, NETHERLANDS
| | - Michele Fenu
- Otorhinolaryngology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Zuid-Holland, 3000 CA, NETHERLANDS
| | - Gerjo J V M van Osch
- Orthopaedics; Otorhinolaryngology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Zuid-Holland, 3000 CA, NETHERLANDS
| | - Gijsje Koenderink
- Bionanoscience, TU Delft, Kavli Institute of Nanoscience Delft, Delft University of Technology, Van der Maasweg 9, Delft, Zuid-Holland, 2629 HZ, NETHERLANDS
| |
Collapse
|
28
|
Tang R, Dang M, Zhang X, Tao J, Shi W, Lu W, Yu R, Su X, Tang Y, Teng Z. Disrupting stromal barriers to enhance photothermal-chemo therapy using a halofuginone-loaded Janus mesoporous nanoplatform. J Colloid Interface Sci 2021; 610:313-320. [PMID: 34923269 DOI: 10.1016/j.jcis.2021.11.190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/29/2021] [Accepted: 11/29/2021] [Indexed: 12/23/2022]
Abstract
Dense tumor stroma is the physiological barrier in drug delivery that prevents anticancer drugs from entering the tumor, thereby seriously limiting the drugs' therapeutic effect. In this study, a Janus nanoplatform consisting of periodic mesoporous organosilica-coated platinum nanoplatforms (JPMO-Pt) and anti-stroma drug halofuginone (HF) (denoted as JPMO-Pt-HF), was developed to deplete the tumor stroma and synergistically treat breast cancer in BALB/c mice. The prepared JPMO-Pt had a uniform size of 245 nm, a good dispersion, an excellent in vitro and in vivo biocompatibility, and a high loading capacity for HF (up to 50 μg/mg). The antitumor experiments showed that the survival rate of 4 T1 cells exhibited an obvious downward trend when the cells were incubated with the JPMO-Pt-HF and irradiated with 808 nm laser. Moreover, the cell survival rate was only about 10% at 48 h when the HF concentration was 2.0 μg/mL. Notably, JPMO-Pt-HF under irradiation had an excellent synergistic therapeutic effect on tumor cells. In vivo antitumor experiment further showed that the JPMO-Pt-HF, in combination with laser irradiation, could minimize tumor growth, showing significantly better effects than those observed for the case of monotherapy involving photothermal therapy (PTT) (152 vs. 670 mm3, p < 0.0001) and HF (152 vs. 419 mm3, p = 0.0208). In addition, immunohistochemistry of tumor tissues indicated that JPMO-Pt-HF obviously reduced the relative collagen and α-smooth muscle actin (α-SMA) area fraction. Taken together, this research designs a new platform that not only possesses the ability to degrade the tumor matrix but also combines PTT and chemotherapeutic effects, and holds promise for effective tumor treatment.
Collapse
Affiliation(s)
- Rui Tang
- Key Laboratory for Organic Electronics and Information Displays and Institute of Advanced Materials, Jiangsu Key Laboratory for Biosensors, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210046 Jiangsu, PR China
| | - Meng Dang
- Key Laboratory for Organic Electronics and Information Displays and Institute of Advanced Materials, Jiangsu Key Laboratory for Biosensors, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210046 Jiangsu, PR China
| | - Xiaojun Zhang
- Department of Medical Imaging, Children's hospital of Nanjing Medical University, 210008 Jiangsu, PR China
| | - Jun Tao
- Key Laboratory for Organic Electronics and Information Displays and Institute of Advanced Materials, Jiangsu Key Laboratory for Biosensors, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210046 Jiangsu, PR China
| | - Wenhui Shi
- Key Laboratory for Organic Electronics and Information Displays and Institute of Advanced Materials, Jiangsu Key Laboratory for Biosensors, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210046 Jiangsu, PR China
| | - Wei Lu
- Key Laboratory for Organic Electronics and Information Displays and Institute of Advanced Materials, Jiangsu Key Laboratory for Biosensors, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210046 Jiangsu, PR China
| | - Ruifa Yu
- Key Laboratory for Organic Electronics and Information Displays and Institute of Advanced Materials, Jiangsu Key Laboratory for Biosensors, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210046 Jiangsu, PR China
| | - Xiaodan Su
- Key Laboratory for Organic Electronics and Information Displays and Institute of Advanced Materials, Jiangsu Key Laboratory for Biosensors, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210046 Jiangsu, PR China
| | - Yuxia Tang
- Department of Medical Imaging, Jinling Hospital, School of Medicine, Nanjing University, 210002 Jiangsu, PR China.
| | - Zhaogang Teng
- Key Laboratory for Organic Electronics and Information Displays and Institute of Advanced Materials, Jiangsu Key Laboratory for Biosensors, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210046 Jiangsu, PR China; State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210093, PR China.
| |
Collapse
|
29
|
Djediai S, Gonzalez Suarez N, El Cheikh-Hussein L, Rodriguez Torres S, Gresseau L, Dhayne S, Joly-Lopez Z, Annabi B. MT1-MMP Cooperates with TGF-β Receptor-Mediated Signaling to Trigger SNAIL and Induce Epithelial-to-Mesenchymal-like Transition in U87 Glioblastoma Cells. Int J Mol Sci 2021; 22:13006. [PMID: 34884812 PMCID: PMC8657819 DOI: 10.3390/ijms222313006] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/25/2021] [Accepted: 11/28/2021] [Indexed: 12/27/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) recapitulates metastasis and can be induced in vitro through transforming growth factor (TGF)-β signaling. A role for MMP activity in glioblastoma multiforme has been ascribed to EMT, but the molecular crosstalk between TGF-β signaling and membrane type 1 MMP (MT1-MMP) remains poorly understood. Here, the expression of common EMT biomarkers, induced through TGF-β and the MT1-MMP inducer concanavalin A (ConA), was explored using RNA-seq analysis and differential gene arrays in human U87 glioblastoma cells. TGF-β triggered SNAIL and fibronectin expressions in 2D-adherent and 3D-spheroid U87 glioblastoma cell models. Those inductions were antagonized by the TGF-β receptor kinase inhibitor galunisertib, the JAK/STAT inhibitors AG490 and tofacitinib, and by the diet-derived epigallocatechin gallate (EGCG). Transient gene silencing of MT1-MMP prevented the induction of SNAIL by ConA and abrogated TGF-β-induced cell chemotaxis. Moreover, ConA induced STAT3 and Src phosphorylation, suggesting these pathways to be involved in the MT1-MMP-mediated signaling axis that led to SNAIL induction. Our findings highlight a new signaling axis linking MT1-MMP to TGF-β-mediated EMT-like induction in glioblastoma cells, the process of which can be prevented by the diet-derived EGCG.
Collapse
Affiliation(s)
- Souad Djediai
- Laboratoire d’Oncologie Moléculaire, Université du Québec à Montréal, C.P. 8888, Succ. Centre-ville, Montreal, QC H3C 3P8, Canada; (S.D.); (N.G.S.); (L.E.C.-H.); (S.R.T.); (L.G.)
- Département de Chimie, and CERMO-FC, Université du Québec à Montréal, Montreal, QC H3C 3P8, Canada; (S.D.); (Z.J.-L.)
| | - Narjara Gonzalez Suarez
- Laboratoire d’Oncologie Moléculaire, Université du Québec à Montréal, C.P. 8888, Succ. Centre-ville, Montreal, QC H3C 3P8, Canada; (S.D.); (N.G.S.); (L.E.C.-H.); (S.R.T.); (L.G.)
- Département de Chimie, and CERMO-FC, Université du Québec à Montréal, Montreal, QC H3C 3P8, Canada; (S.D.); (Z.J.-L.)
| | - Layal El Cheikh-Hussein
- Laboratoire d’Oncologie Moléculaire, Université du Québec à Montréal, C.P. 8888, Succ. Centre-ville, Montreal, QC H3C 3P8, Canada; (S.D.); (N.G.S.); (L.E.C.-H.); (S.R.T.); (L.G.)
- Département de Chimie, and CERMO-FC, Université du Québec à Montréal, Montreal, QC H3C 3P8, Canada; (S.D.); (Z.J.-L.)
| | - Sahily Rodriguez Torres
- Laboratoire d’Oncologie Moléculaire, Université du Québec à Montréal, C.P. 8888, Succ. Centre-ville, Montreal, QC H3C 3P8, Canada; (S.D.); (N.G.S.); (L.E.C.-H.); (S.R.T.); (L.G.)
- Département de Chimie, and CERMO-FC, Université du Québec à Montréal, Montreal, QC H3C 3P8, Canada; (S.D.); (Z.J.-L.)
| | - Loraine Gresseau
- Laboratoire d’Oncologie Moléculaire, Université du Québec à Montréal, C.P. 8888, Succ. Centre-ville, Montreal, QC H3C 3P8, Canada; (S.D.); (N.G.S.); (L.E.C.-H.); (S.R.T.); (L.G.)
- Département de Chimie, and CERMO-FC, Université du Québec à Montréal, Montreal, QC H3C 3P8, Canada; (S.D.); (Z.J.-L.)
| | - Sheraz Dhayne
- Département de Chimie, and CERMO-FC, Université du Québec à Montréal, Montreal, QC H3C 3P8, Canada; (S.D.); (Z.J.-L.)
| | - Zoé Joly-Lopez
- Département de Chimie, and CERMO-FC, Université du Québec à Montréal, Montreal, QC H3C 3P8, Canada; (S.D.); (Z.J.-L.)
| | - Borhane Annabi
- Laboratoire d’Oncologie Moléculaire, Université du Québec à Montréal, C.P. 8888, Succ. Centre-ville, Montreal, QC H3C 3P8, Canada; (S.D.); (N.G.S.); (L.E.C.-H.); (S.R.T.); (L.G.)
- Département de Chimie, and CERMO-FC, Université du Québec à Montréal, Montreal, QC H3C 3P8, Canada; (S.D.); (Z.J.-L.)
| |
Collapse
|
30
|
Modulation of peritumoral fibroblasts with a membrane-tethered tissue inhibitor of metalloproteinase (TIMP) for the elimination of cancer cells. Invest New Drugs 2021; 40:198-208. [PMID: 34519970 DOI: 10.1007/s10637-021-01177-4] [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: 06/27/2021] [Accepted: 09/02/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Peritumoral fibroblasts are key components of the tumor microenvironment. Through remodeling of the extracellular matrix (ECM) and secretion of pro-tumorigenic cytokines, peritumoral fibroblasts foster an immunosuppressive milieu conducive to tumor cell proliferation. In this study, we investigated if peritumoral fibroblasts could be therapeutically engineered to elicit an anti-cancer response by abolishing the proteolytic activities of membrane-bound metalloproteinases involved in ECM modulation. METHODS A high affinity, glycosylphosphatidylinositol (GPI)-anchored Tissue Inhibitor of Metalloproteinase (TIMP) named "T1PrαTACE" was created for dual inhibition of MT1-MMP and TACE. T1PrαTACE was expressed in fibroblasts and its effects on cancer cell proliferation investigated in 3D co-culture models. RESULTS T1PrαTACE abrogated the activities of MT1-MMP and TACE in host fibroblasts. As a GPI protein, T1PrαTACE could spontaneously detach from the plasma membrane of the fibroblast to co-localize with MT1-MMP and TACE on neighboring cancer cells. In a 3D co-culture model, T1PrαTACE promoted adherence between the cancer cells and surrounding fibroblasts, which led to an attenuation in tumor development. CONCLUSION Peritumoral fibroblasts can be modulated with the TIMP for the elimination of cancer cells. As a novel anti-tumor strategy, our approach could potentially be used in combination with conventional chemo- and immunotherapies for a more effective cancer therapy.
Collapse
|
31
|
Guo J, Liang Y, Xue D, Shen J, Cai Y, Zhu J, Fu YX, Peng H. Tumor-conditional IL-15 pro-cytokine reactivates anti-tumor immunity with limited toxicity. Cell Res 2021; 31:1190-1198. [PMID: 34376814 DOI: 10.1038/s41422-021-00543-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 07/08/2021] [Indexed: 12/20/2022] Open
Abstract
IL-15 is a promising cytokine to expand NK and CD8+ T cells for cancer immunotherapy, but its application is limited by dose-limiting, on-target off-tumor toxicity. Here, we have developed a next-generation IL-15 that is activated inside the tumor microenvironment (TME). This pro-IL-15 has the extracellular domain of IL-15Rβ fused to the N-terminus of sIL-15-Fc through a tumor-enriched Matrix Metalloproteinase (MMP) cleavable peptide linker to block its activity. Unlike sIL-15-Fc, pro-IL-15 does not activate the peripheral expansion of NK cells and T cells, thus reducing systemic toxicity, but it still preserves efficient anti-tumor abilities. In various mouse tumors, the anti-tumor effect of pro-IL-15 depends on intratumoral CD8+ T cells and IFN-γ. Pro-IL-15 increases the stem-like TCF1+Tim-3-CD8+ T cells within tumor tissue and helps overcome immune checkpoint blockade (ICB) resistance. Moreover, pro-IL-15 synergizes with current tyrosine kinase inhibitor (TKI) targeted-therapy in a poorly inflamed TUBO tumor model, suggesting that pro-IL-15 helps overcome targeted-therapy resistance. Our results demonstrate a next-generation IL-15 cytokine that can stimulate potent anti-tumor activity without severe toxicity.
Collapse
Affiliation(s)
- Jingya Guo
- Chinese Academy of Sciences Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yong Liang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Diyuan Xue
- Chinese Academy of Sciences Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jiao Shen
- Chinese Academy of Sciences Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yueqi Cai
- Chinese Academy of Sciences Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jiankun Zhu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yang-Xin Fu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Hua Peng
- Chinese Academy of Sciences Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
32
|
Vos MC, van der Wurff AAM, van Kuppevelt TH, Massuger LFAG. The role of MMP-14 in ovarian cancer: a systematic review. J Ovarian Res 2021; 14:101. [PMID: 34344453 PMCID: PMC8336022 DOI: 10.1186/s13048-021-00852-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 07/23/2021] [Indexed: 12/18/2022] Open
Abstract
AIM In order to evaluate the role of MMP-14 in ovarian cancer, a systematic review was conducted. METHODS In March 2020, a search in Pubmed was performed with MMP-14 and ovarian cancer as search terms. After exclusion of the references not on MMP-14 or ovarian cancer or not in English, the studies found were classified into two categories: basic research and clinicopathological research. RESULTS In total, 94 references were found of which 33 were excluded. Two additional articles were found in the reference lists of the included studies. Based on the full texts, another 4 were excluded. Eventually, 59 studies were included in the review, 32 on basic research and 19 on clinicopathological research. 8 studies fell in both categories. The basic research studies show that MMP-14 plays an important role in ovarian cancer in the processes of proliferation, invasion, angiogenesis and metastasis. In clinocopathological research, MMP-14 expression is found in most tumours with characteristics of poor prognosis but this immunohistochemical MMP-14 determination does not seem to be an independent predictor of prognosis. CONCLUSIONS From this systematic review of the literature concerning MMP-14 in ovarian cancer it becomes clear that MMP-14 plays various important roles in the pathophysiology of ovarian cancer. The exact translation of these roles in the pathophysiology to the importance of MMP-14 in clinicopathological research in ovarian cancer and possible therapeutic role of anti-MMP-14 agents needs further elucidation.
Collapse
Affiliation(s)
- M. Caroline Vos
- Department of Obstetrics and Gynaecology, Elisabeth-Tweesteden Hospital, PO Box 90151, 5000 LC Tilburg, the Netherlands
| | | | - Toin H. van Kuppevelt
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, PO Box 9101, 6500 HB Nijmegen, the Netherlands
| | - Leon F. A. G. Massuger
- Department of Obstetrics and Gynaecology, Radboud University Nijmegen Medical Centre, PO Box 9101, 6500 HB Nijmegen, the Netherlands
| |
Collapse
|
33
|
Habič A, Novak M, Majc B, Lah Turnšek T, Breznik B. Proteases Regulate Cancer Stem Cell Properties and Remodel Their Microenvironment. J Histochem Cytochem 2021; 69:775-794. [PMID: 34310223 DOI: 10.1369/00221554211035192] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Proteolytic activity is perturbed in tumors and their microenvironment, and proteases also affect cancer stem cells (CSCs). CSCs are the therapy-resistant subpopulation of cancer cells with tumor-initiating capacity that reside in specialized tumor microenvironment niches. In this review, we briefly summarize the significance of proteases in regulating CSC activities with a focus on brain tumor glioblastoma. A plethora of proteases and their inhibitors participate in CSC invasiveness and affect intercellular interactions, enhancing CSC immune, irradiation, and chemotherapy resilience. Apart from their role in degrading the extracellular matrix enabling CSC migration in and out of their niches, we review the ability of proteases to modulate CSC properties, which prevents their elimination. When designing protease-oriented therapies, the multifaceted roles of proteases should be thoroughly investigated.
Collapse
Affiliation(s)
- Anamarija Habič
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia.,The Jožef Stefan International Postgraduate School, Ljubljana, Slovenia
| | - Metka Novak
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Bernarda Majc
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia.,The Jožef Stefan International Postgraduate School, Ljubljana, Slovenia
| | - Tamara Lah Turnšek
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia.,The Jožef Stefan International Postgraduate School, Ljubljana, Slovenia.,Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Barbara Breznik
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| |
Collapse
|
34
|
Moracho N, Learte AIR, Muñoz-Sáez E, Marchena MA, Cid MA, Arroyo AG, Sánchez-Camacho C. Emerging roles of MT-MMPs in embryonic development. Dev Dyn 2021; 251:240-275. [PMID: 34241926 DOI: 10.1002/dvdy.398] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 06/17/2021] [Accepted: 06/30/2021] [Indexed: 12/19/2022] Open
Abstract
Membrane-type matrix metalloproteinases (MT-MMPs) are cell membrane-tethered proteinases that belong to the family of the MMPs. Apart from their roles in degradation of the extracellular milieu, MT-MMPs are able to activate through proteolytic processing at the cell surface distinct molecules such as receptors, growth factors, cytokines, adhesion molecules, and other pericellular proteins. Although most of the information regarding these enzymes comes from cancer studies, our current knowledge about their contribution in distinct developmental processes occurring in the embryo is limited. In this review, we want to summarize the involvement of MT-MMPs in distinct processes during embryonic morphogenesis, including cell migration and proliferation, epithelial-mesenchymal transition, cell polarity and branching, axon growth and navigation, synapse formation, and angiogenesis. We also considered information about MT-MMP functions from studies assessed in pathological conditions and compared these data with those relevant for embryonic development.
Collapse
Affiliation(s)
- Natalia Moracho
- Department of Medicine, School of Biomedical Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, Madrid, Spain
| | - Ana I R Learte
- Department of Dentistry, School of Biomedical Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, Madrid, Spain
| | - Emma Muñoz-Sáez
- Department of Health Science, School of Biomedical Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, Madrid, Spain
| | - Miguel A Marchena
- Department of Medicine, School of Biomedical Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, Madrid, Spain
| | - María A Cid
- Department of Dentistry, School of Biomedical Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, Madrid, Spain
| | - Alicia G Arroyo
- Vascular Pathophysiology Department, Centro Nacional de Investigaciones Cardiovasculares (CNIC-CSIC), Madrid, Spain.,Molecular Biomedicine Department, Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
| | - Cristina Sánchez-Camacho
- Department of Medicine, School of Biomedical Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, Madrid, Spain.,Vascular Pathophysiology Department, Centro Nacional de Investigaciones Cardiovasculares (CNIC-CSIC), Madrid, Spain
| |
Collapse
|
35
|
Li Y, Su Z, Wei B, Qin M, Liang Z. Bioinformatics analysis identified MMP14 and COL12A1 as immune-related biomarkers associated with pancreatic adenocarcinoma prognosis. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2021; 18:5921-5942. [PMID: 34517516 DOI: 10.3934/mbe.2021296] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
BACKGROUND Pancreatic adenocarcinoma (PAAD) is one of the most common malignant tumors with high mortality rates and a poor prognosis. There is an urgent need to determine the molecular mechanism of PAAD tumorigenesis and identify promising biomarkers for the diagnosis and targeted therapy of the disease. METHODS Three GEO datasets (GSE62165, GSE15471 and GSE62452) were analyzed to obtain differentially expressed genes (DEGs). The PPI networks and hub genes were identified through the STRING database and MCODE plugin in Cytoscape software. GO and KEGG enrichment pathways were analyzed by the DAVID database. The GEPIA database was utilized to estimate the prognostic value of hub genes. Furthermore, the roles of MMP14 and COL12A1 in immune infiltration and tumor-immune interaction and their biological functions in PAAD were explored by TIMER, TISIDB, GeneMANIA, Metascape and GSEA. RESULTS A total of 209 common DEGs in the three datasets were obtained. GO function analysis showed that the 209 DEGs were significantly enriched in calcium ion binding, serine-type endopeptidase activity, integrin binding, extracellular matrix structural constituent and collagen binding. KEGG pathway analysis showed that DEGs were mainly enriched in focal adhesion, protein digestion and absorption and ECM-receptor interaction. The 14 genes with the highest degree of connectivity were defined as the hub genes of PAAD development. GEPIA revealed that PAAD patients with upregulated MMP14 and COL12A1 expression had poor prognoses. In addition, TIMER analysis revealed that MMP14 and COL12A1 were closely associated with the infiltration levels of macrophages, neutrophils and dendritic cells in PAAD. TISIDB revealed that MMP14 was strongly positively correlated with CD276, TNFSF4, CD70 and TNFSF9, while COL12A1 was strongly positively correlated with TNFSF4, CD276, ENTPD1 and CD70. GSEA revealed that MMP14 and COL12A1 were significantly enriched in epithelial mesenchymal transition, extracellular matrix receptor interaction, apical junction, and focal adhesion in PAAD development. CONCLUSIONS Our study revealed that overexpression of MMP14 and COL12A1 is significantly correlated with PAAD patient poor prognosis. MMP14 and COL12A1 participate in regulating tumor immune interactions and might become promising biomarkers for PAAD.
Collapse
Affiliation(s)
- Yuexian Li
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Zhou Su
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Biwei Wei
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Mengbin Qin
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, China
| | - Zhihai Liang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
36
|
Nørregaard KS, Jürgensen HJ, Gårdsvoll H, Engelholm LH, Behrendt N, Søe K. Osteosarcoma and Metastasis Associated Bone Degradation-A Tale of Osteoclast and Malignant Cell Cooperativity. Int J Mol Sci 2021; 22:ijms22136865. [PMID: 34202300 PMCID: PMC8269025 DOI: 10.3390/ijms22136865] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 12/28/2022] Open
Abstract
Cancer-induced bone degradation is part of the pathological process associated with both primary bone cancers, such as osteosarcoma, and bone metastases originating from, e.g., breast, prostate, and colon carcinomas. Typically, this includes a cancer-dependent hijacking of processes also occurring during physiological bone remodeling, including osteoclast-mediated disruption of the inorganic bone component and collagenolysis. Extensive research has revealed the significance of osteoclast-mediated bone resorption throughout the course of disease for both primary and secondary bone cancer. Nevertheless, cancer cells representing both primary bone cancer and bone metastasis have also been implicated directly in bone degradation. We will present and discuss observations on the contribution of osteoclasts and cancer cells in cancer-associated bone degradation and reciprocal modulatory actions between these cells. The focus of this review is osteosarcoma, but we will also include relevant observations from studies of bone metastasis. Additionally, we propose a model for cancer-associated bone degradation that involves a collaboration between osteoclasts and cancer cells and in which both cell types may directly participate in the degradation process.
Collapse
Affiliation(s)
- Kirstine Sandal Nørregaard
- Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Center (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark; (H.J.J.); (H.G.); (L.H.E.); (N.B.)
- Correspondence: ; Tel.: +45-3545-6030
| | - Henrik Jessen Jürgensen
- Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Center (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark; (H.J.J.); (H.G.); (L.H.E.); (N.B.)
| | - Henrik Gårdsvoll
- Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Center (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark; (H.J.J.); (H.G.); (L.H.E.); (N.B.)
| | - Lars Henning Engelholm
- Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Center (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark; (H.J.J.); (H.G.); (L.H.E.); (N.B.)
| | - Niels Behrendt
- Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Center (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark; (H.J.J.); (H.G.); (L.H.E.); (N.B.)
| | - Kent Søe
- Clinical Cell Biology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, 5230 Odense, Denmark;
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, 5000 Odense, Denmark
- Department of Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark
| |
Collapse
|
37
|
Xue D, Hsu E, Fu YX, Peng H. Next-generation cytokines for cancer immunotherapy. Antib Ther 2021; 4:123-133. [PMID: 34263141 PMCID: PMC8271143 DOI: 10.1093/abt/tbab014] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/09/2021] [Accepted: 06/22/2021] [Indexed: 12/12/2022] Open
Abstract
Most studies focus on the first and second signals of T cell activation. However, the roles of cytokines in immunotherapy are not fully understood, and cytokines have not been widely used in patient care. Clinical application of cytokines is limited due to their short half-life in vivo, severe toxicity at therapeutic doses, and overall lack of efficacy. Several modifications have been engineered to extend their half-life and increase tumor targeting, including polyethylene glycol conjugation, fusion to tumor-targeting antibodies, and alteration of cytokine/cell receptor-binding affinity. These modifications demonstrate an improvement in either increased antitumor efficacy or reduced toxicity. However, these cytokine engineering strategies may still be improved further, as each strategy poses advantages and disadvantages in the delicate balance of targeting tumor cells, tumor-infiltrating lymphocytes, and peripheral immune cells. This review focuses on selected cytokines, including interferon-α, interleukin (IL)-2, IL-15, IL-21, and IL-12, in both preclinical studies and clinical applications. We review next-generation designs of these cytokines that improve half-life, tumor targeting, and antitumor efficacy. We also present our perspectives on the development of new strategies to potentiate cytokine-based immunotherapy.
Collapse
Affiliation(s)
- Diyuan Xue
- Key laboratory of Infection and Immunity Institute of Biophysics, Chinese Academy of Sciences, 15 Da Tun Rd, Chaoyang District, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Eric Hsu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Yang-Xin Fu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Hua Peng
- Key laboratory of Infection and Immunity Institute of Biophysics, Chinese Academy of Sciences, 15 Da Tun Rd, Chaoyang District, Beijing 100101, China
| |
Collapse
|
38
|
Mitre GP, Balbinot KM, Ribeiro ALR, da Silva Kataoka MS, de Melo Alves Júnior S, de Jesus Viana Pinheiro J. Key proteins of invadopodia are overexpressed in oral squamous cell carcinoma suggesting an important role of MT1-MMP in the tumoral progression. Diagn Pathol 2021; 16:33. [PMID: 33879222 PMCID: PMC8059181 DOI: 10.1186/s13000-021-01090-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 03/22/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is the most relevant malignant neoplasm among all head and neck tumours due to its high prevalence and unfavourable prognosis. Tumour invasion and metastasis that affect prognosis are result of a set of complex events that cells with invasive potential use to spread to other regions. These cells use several mechanisms to invade tissues, including a type of finger-like membrane protrusion called invadopodia. This study aims to investigate the immunoexpression of invaopodia related-proteins TKs5, cortactin, TKs4 and MT1-MMP in OSCC and correlate it to clinicopathological data. METHODS An immunohistochemical evaluation of fifty cases of OSCCs and 20 cases of oral mucosa (OM) were assessed. The expression of invadopodia proteins were analysed in comparison to normal tissue (OM) and correlated to different clinical-stage and histological grade of OSCC. RESULTS TKs5, cortactin, TKs4 and MT1-MMP were significantly overexpressed in OSCC when compared to OM (p < 0.0001). Among tumour stages, TKs5 showed a statistical difference in immunolabelling between stage I and III (p = 0.026). Cortactin immunolabelling was statistically higher in grade I than in grade II and III. No differences were seen on TKs4 expression based on tumour staging or grading. MT1-MMP was higher expressed and showed statistical difference between stages I and III and grades I compared to II and III. CONCLUSIONS The invadopodia related-proteins were found to be overexpressed in OSCC when compared to OM, suggesting invadopodia formation and activity. Besides overexpressed in OSCC, cortactin, TKs4 and TKs5 showed no or ambiguous differences in protein expression when compared among clinical-stages or histological grades groups. Conversely, the expression of MT1-MMP increased in advanced stages and less differentiated tumours, suggesting MT1-MMP expression as a promising prognostic marker in OSCC.
Collapse
Affiliation(s)
- Geovanni Pereira Mitre
- Laboratory of Histopathology and Immunohistochemistry, School of Dentistry, Cell Culture Laboratory, Federal University of Pará, Rua Augusto Corrêa, 01, Guamá, PA, 66075110, Belém, Brazil
| | - Karolyny Martins Balbinot
- Laboratory of Histopathology and Immunohistochemistry, School of Dentistry, Cell Culture Laboratory, Federal University of Pará, Rua Augusto Corrêa, 01, Guamá, PA, 66075110, Belém, Brazil
| | - André Luis Ribeiro Ribeiro
- Laboratory of Histopathology and Immunohistochemistry, School of Dentistry, Cell Culture Laboratory, Federal University of Pará, Rua Augusto Corrêa, 01, Guamá, PA, 66075110, Belém, Brazil
| | - Maria Sueli da Silva Kataoka
- Laboratory of Histopathology and Immunohistochemistry, School of Dentistry, Cell Culture Laboratory, Federal University of Pará, Rua Augusto Corrêa, 01, Guamá, PA, 66075110, Belém, Brazil
| | - Sérgio de Melo Alves Júnior
- Laboratory of Histopathology and Immunohistochemistry, School of Dentistry, Cell Culture Laboratory, Federal University of Pará, Rua Augusto Corrêa, 01, Guamá, PA, 66075110, Belém, Brazil
| | - João de Jesus Viana Pinheiro
- Laboratory of Histopathology and Immunohistochemistry, School of Dentistry, Cell Culture Laboratory, Federal University of Pará, Rua Augusto Corrêa, 01, Guamá, PA, 66075110, Belém, Brazil.
| |
Collapse
|
39
|
The Role of the Metzincin Superfamily in Prostate Cancer Progression: A Systematic-Like Review. Int J Mol Sci 2021; 22:ijms22073608. [PMID: 33808504 PMCID: PMC8036576 DOI: 10.3390/ijms22073608] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/21/2021] [Accepted: 03/26/2021] [Indexed: 02/07/2023] Open
Abstract
Prostate cancer remains a leading cause of cancer-related morbidity in men. Potentially important regulators of prostate cancer progression are members of the metzincin superfamily of proteases, principally through their regulation of the extracellular matrix. It is therefore timely to review the role of the metzincin superfamily in prostate cancer and its progression to better understand their involvement in this disease. A systematic-like search strategy was conducted. Articles that investigated the roles of members of the metzincin superfamily and their key regulators in prostate cancer were included. The extracted articles were synthesized and data presented in tabular and narrative forms. Two hundred and five studies met the inclusion criteria. Of these, 138 investigated the role of the Matrix Metalloproteinase (MMP) subgroup, 34 the Membrane-Tethered Matrix Metalloproteinase (MT-MMP) subgroup, 22 the A Disintegrin and Metalloproteinase (ADAM) subgroup, 8 the A Disintegrin and Metalloproteinase with Thrombospondin Motifs (ADAMTS) subgroup and 53 the Tissue Inhibitor of Metalloproteinases (TIMP) family of regulators, noting that several studies investigated multiple family members. There was clear evidence that specific members of the metzincin superfamily are involved in prostate cancer progression, which can be either in a positive or negative manner. However, further understanding of their mechanisms of action and how they may be used as prognostic indicators or molecular targets is required.
Collapse
|
40
|
Vizovisek M, Ristanovic D, Menghini S, Christiansen MG, Schuerle S. The Tumor Proteolytic Landscape: A Challenging Frontier in Cancer Diagnosis and Therapy. Int J Mol Sci 2021; 22:ijms22052514. [PMID: 33802262 PMCID: PMC7958950 DOI: 10.3390/ijms22052514] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/24/2021] [Accepted: 02/25/2021] [Indexed: 02/06/2023] Open
Abstract
In recent decades, dysregulation of proteases and atypical proteolysis have become increasingly recognized as important hallmarks of cancer, driving community-wide efforts to explore the proteolytic landscape of oncologic disease. With more than 100 proteases currently associated with different aspects of cancer development and progression, there is a clear impetus to harness their potential in the context of oncology. Advances in the protease field have yielded technologies enabling sensitive protease detection in various settings, paving the way towards diagnostic profiling of disease-related protease activity patterns. Methods including activity-based probes and substrates, antibodies, and various nanosystems that generate reporter signals, i.e., for PET or MRI, after interaction with the target protease have shown potential for clinical translation. Nevertheless, these technologies are costly, not easily multiplexed, and require advanced imaging technologies. While the current clinical applications of protease-responsive technologies in oncologic settings are still limited, emerging technologies and protease sensors are poised to enable comprehensive exploration of the tumor proteolytic landscape as a diagnostic and therapeutic frontier. This review aims to give an overview of the most relevant classes of proteases as indicators for tumor diagnosis, current approaches to detect and monitor their activity in vivo, and associated therapeutic applications.
Collapse
|
41
|
Cai Z, Zhai T, Muhanhali D, Ling Y. TNRC6C Functions as a Tumor Suppressor and Is Frequently Downregulated in Papillary Thyroid Cancer. Int J Endocrinol 2021; 2021:6686998. [PMID: 33564303 PMCID: PMC7867448 DOI: 10.1155/2021/6686998] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/29/2020] [Accepted: 01/22/2021] [Indexed: 12/21/2022] Open
Abstract
Our previous study found that trinucleotide repeat containing adaptor 6C (TNRC6C) may act as a tumor suppressor in papillary thyroid cancer (PTC). In this study, we aimed to confirm the effect of TNRC6C on PTC and investigate the underlying molecular mechanism. The difference of mRNA level of TNRC6C between PTC tissue and noncancerous thyroid tissue and the association of expression level of TNRC6C with clinicopathological features of PTC were analyzed using TCGA data. Immunohistochemical assay was performed to detect the protein expression of TNRC6C in PTC and its adjacent noncancerous tissue. Cell proliferation, migration, invasion, and apoptosis were analyzed after knockdown or overexpression of TNRC6C in BCPAP cells. RNA-sequencing was performed to find the target genes of TNRC6C, and potential targets were validated in BCPAP and TPC1 cells. Our results showed that TNRC6C was downregulated in PTC, and lower expression level of TNRC6C was associated with worse clinicopathological features. Overexpression of TNRC6C significantly inhibited proliferation, migration, and invasion of BCPAP cells and promoted its apoptosis, while knockdown of TNRC6C acted the opposite role. By analyzing RNA-sequencing data and TCGA data, 12 genes (SCD, CRLF1, APCDD1L, CTHRC1, PTPRU, ALDH1A3, VCAN, TNC, ECE1, COL1A1, CAMK2N2, and MMP14) were considered as potential target genes of TNRC6C, and most of them were associated with clinicopathological features of PTC in TCGA. All of them except CAMK2N2 were significantly downregulated after overexpressing TNRC6C. Our study demonstrated that TNRC6C functions as a tumor suppressor in PTC and may serve as a useful therapeutic target and prognostic marker for PTC patients.
Collapse
Affiliation(s)
- Zhenqin Cai
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai 200032, China
| | - Tianyu Zhai
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai 200032, China
| | - Dilidaer Muhanhali
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai 200032, China
| | - Yan Ling
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai 200032, China
| |
Collapse
|
42
|
Marusak C, Thakur V, Li Y, Freitas JT, Zmina PM, Thakur VS, Chang M, Gao M, Tan J, Xiao M, Lu Y, Mills GB, Flaherty K, Frederick DT, Miao B, Sullivan RJ, Moll T, Boland GM, Herlyn M, Zhang G, Bedogni B. Targeting Extracellular Matrix Remodeling Restores BRAF Inhibitor Sensitivity in BRAFi-resistant Melanoma. Clin Cancer Res 2020; 26:6039-6050. [PMID: 32820016 PMCID: PMC7669662 DOI: 10.1158/1078-0432.ccr-19-2773] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 07/07/2020] [Accepted: 08/17/2020] [Indexed: 12/16/2022]
Abstract
PURPOSE The extracellular matrix (ECM) is an intriguing, yet understudied component of therapy resistance. Here, we investigated the role of ECM remodeling by the collagenase, MT1-MMP, in conferring resistance of v-Raf murine sarcoma viral oncogene homolog B1 (BRAF)-mutant melanoma to BRAF inhibitor (BRAFi) therapy. EXPERIMENTAL DESIGN Publicly available RNA-sequencing data and reverse phase protein array were used to determine the relevance of MT1-MMP upregulation in BRAFi-resistant melanoma in patients, patient-derived xenografts, and cell line-derived tumors. Short hairpin RNA (shRNA)-mediated knockdown of MT1-MMP, inhibition via the selective MT1-MMP/MMP2 inhibitor, ND322, or overexpression of MT1-MMP was used to assess the role of MT1-MMP in mediating resistance to BRAFi. RESULTS MT1-MMP was consistently upregulated in posttreatment tumor samples derived from patients upon disease progression and in melanoma xenografts and cell lines that acquired resistance to BRAFi. shRNA- or ND322-mediated inhibition of MT1-MMP synergized with BRAFi leading to resensitization of resistant cells and tumors to BRAFi. The resistant phenotype depends on the ability of cells to cleave the ECM. Resistant cells seeded in MT1-MMP uncleavable matrixes were resensitized to BRAFi similarly to MT1-MMP inhibition. This is due to the inability of cells to activate integrinβ1 (ITGB1)/FAK signaling, as restoration of ITGB1 activity is sufficient to maintain resistance to BRAFi in the context of MT1-MMP inhibition. Finally, the increase in MT1-MMP in BRAFi-resistant cells is TGFβ dependent, as inhibition of TGFβ receptors I/II dampens MT1-MMP overexpression and restores sensitivity to BRAF inhibition. CONCLUSIONS BRAF inhibition results in a selective pressure toward higher expression of MT1-MMP. MT1-MMP is pivotal to an ECM-based signaling pathway that confers resistance to BRAFi therapy.
Collapse
Affiliation(s)
- Charles Marusak
- Department of Dermatology, University of Miami Miller School of Medicine, Miami, Florida
| | - Varsha Thakur
- Department of Dermatology, University of Miami Miller School of Medicine, Miami, Florida
| | - Yuan Li
- Department of Dermatology, University of Miami Miller School of Medicine, Miami, Florida
| | - Juliano T Freitas
- Department of Dermatology, University of Miami Miller School of Medicine, Miami, Florida
| | - Patrick M Zmina
- Department of Dermatology, University of Miami Miller School of Medicine, Miami, Florida
| | - Vijay S Thakur
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, Florida
| | - Mayland Chang
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana
| | - Ming Gao
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana
| | - Jiufeng Tan
- The Wistar Institute, Philadelphia, Pennsylvania
| | - Min Xiao
- The Wistar Institute, Philadelphia, Pennsylvania
| | - Yiling Lu
- Department of Genomic Medicine, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, Texas
| | - Gordon B Mills
- The Knight Cancer Institute, Oregon Health Sciences University, Portland, Oregon
| | - Keith Flaherty
- Department of Medicine, Harvard Medical School, Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | | | - Benchun Miao
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Ryan J Sullivan
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Tabea Moll
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts
| | - Genevieve M Boland
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts
| | | | - Gao Zhang
- Department of Neurosurgery, Duke University, Durham, North Carolina
| | - Barbara Bedogni
- Department of Dermatology, University of Miami Miller School of Medicine, Miami, Florida.
| |
Collapse
|
43
|
Tumor cell MT1-MMP is dispensable for osteosarcoma tumor growth, bone degradation and lung metastasis. Sci Rep 2020; 10:19138. [PMID: 33154487 PMCID: PMC7645741 DOI: 10.1038/s41598-020-75995-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 10/16/2020] [Indexed: 02/03/2023] Open
Abstract
The membrane-anchored matrix metalloprotease MT1-MMP is a potent collagenolytic enzyme with a well-established role in extracellular matrix turnover and cellular invasion into collagen-rich tissues. MT1-MMP is highly expressed in various types of cancer and has been demonstrated to be directly involved in several stages of tumor progression, including primary tumor growth, angiogenesis, invasion and metastasis. Osteosarcoma is the most common type of primary bone cancer. This disease is characterized by invasive tumor growth, leading to extensive bone destruction, and metastasis to the lungs. The tumor cells in human osteosarcoma display a strong expression of MT1-MMP, but the role of MT1-MMP in osteosarcoma progression is currently unknown. In this study, we investigated the role of MT1-MMP during various stages of osteosarcoma development. We utilized an optimized orthotopic murine osteosarcoma model and human osteosarcoma cells in which the MT1-MMP gene was knocked out using CRISPR/Cas9. We observed a strong expression of MT1-MMP in wildtype cells of both primary tumors and lung metastases, but, surprisingly, MT1-MMP deficiency did not affect primary tumor growth, bone degradation or the formation and growth of lung metastases. We therefore propose that, unlike findings reported in other cancers, tumor-expressed MT1-MMP is dispensable for all stages of osteosarcoma progression.
Collapse
|
44
|
Ragusa S, Prat-Luri B, González-Loyola A, Nassiri S, Squadrito ML, Guichard A, Cavin S, Gjorevski N, Barras D, Marra G, Lutolf MP, Perentes J, Corse E, Bianchi R, Wetterwald L, Kim J, Oliver G, Delorenzi M, De Palma M, Petrova TV. Antiangiogenic immunotherapy suppresses desmoplastic and chemoresistant intestinal tumors in mice. J Clin Invest 2020; 130:1199-1216. [PMID: 32015230 DOI: 10.1172/jci129558] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 11/20/2019] [Indexed: 12/24/2022] Open
Abstract
Mutations in APC promote colorectal cancer (CRC) progression through uncontrolled WNT signaling. Patients with desmoplastic CRC have a significantly worse prognosis and do not benefit from chemotherapy, but the mechanisms underlying the differential responses of APC-mutant CRCs to chemotherapy are not well understood. We report that expression of the transcription factor prospero homeobox 1 (PROX1) was reduced in desmoplastic APC-mutant human CRCs. In genetic Apc-mutant mouse models, loss of Prox1 promoted the growth of desmoplastic, angiogenic, and immunologically silent tumors through derepression of Mmp14. Although chemotherapy inhibited Prox1-proficient tumors, it promoted further stromal activation, angiogenesis, and invasion in Prox1-deficient tumors. Blockade of vascular endothelial growth factor A (VEGFA) and angiopoietin-2 (ANGPT2) combined with CD40 agonistic antibodies promoted antiangiogenic and immunostimulatory reprogramming of Prox1-deficient tumors, destroyed tumor fibrosis, and unleashed T cell-mediated killing of cancer cells. These results pinpoint the mechanistic basis of chemotherapy-induced hyperprogression and illustrate a therapeutic strategy for chemoresistant and desmoplastic CRCs.
Collapse
Affiliation(s)
- Simone Ragusa
- Department of Oncology, University of Lausanne and CHUV, Epalinges, Switzerland.,Ludwig Institute for Cancer Research Lausanne, Epalinges, Switzerland
| | - Borja Prat-Luri
- Department of Oncology, University of Lausanne and CHUV, Epalinges, Switzerland.,Ludwig Institute for Cancer Research Lausanne, Epalinges, Switzerland
| | - Alejandra González-Loyola
- Department of Oncology, University of Lausanne and CHUV, Epalinges, Switzerland.,Ludwig Institute for Cancer Research Lausanne, Epalinges, Switzerland
| | - Sina Nassiri
- Bioinformatics Core Facility, Swiss Institute of Bioinformatics, Lausanne, Switzerland.,Swiss Institute for Experimental Cancer Research, Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne, Switzerland
| | - Mario Leonardo Squadrito
- Swiss Institute for Experimental Cancer Research, Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne, Switzerland
| | - Alan Guichard
- Swiss Institute for Experimental Cancer Research, Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne, Switzerland
| | - Sabrina Cavin
- Division of Thoracic Surgery, CHUV, Lausanne, Switzerland
| | - Nikolce Gjorevski
- Institute of Bioengineering, School of Life Sciences, EPFL, Lausanne, Switzerland
| | - David Barras
- Bioinformatics Core Facility, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Giancarlo Marra
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Matthias P Lutolf
- Institute of Bioengineering, School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Jean Perentes
- Division of Thoracic Surgery, CHUV, Lausanne, Switzerland
| | - Emily Corse
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, (pRED), Schlieren, Switzerland
| | - Roberta Bianchi
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, (pRED), Schlieren, Switzerland
| | - Laureline Wetterwald
- Department of Oncology, University of Lausanne and CHUV, Epalinges, Switzerland.,Ludwig Institute for Cancer Research Lausanne, Epalinges, Switzerland
| | - Jaeryung Kim
- Department of Oncology, University of Lausanne and CHUV, Epalinges, Switzerland.,Ludwig Institute for Cancer Research Lausanne, Epalinges, Switzerland
| | - Guillermo Oliver
- Center for Vascular and Developmental Biology, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Mauro Delorenzi
- Department of Oncology, University of Lausanne and CHUV, Epalinges, Switzerland.,Ludwig Institute for Cancer Research Lausanne, Epalinges, Switzerland.,Bioinformatics Core Facility, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Michele De Palma
- Swiss Institute for Experimental Cancer Research, Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne, Switzerland
| | - Tatiana V Petrova
- Department of Oncology, University of Lausanne and CHUV, Epalinges, Switzerland.,Ludwig Institute for Cancer Research Lausanne, Epalinges, Switzerland
| |
Collapse
|
45
|
Shoucair I, Weber Mello F, Jabalee J, Maleki S, Garnis C. The Role of Cancer-Associated Fibroblasts and Extracellular Vesicles in Tumorigenesis. Int J Mol Sci 2020; 21:ijms21186837. [PMID: 32957712 PMCID: PMC7555043 DOI: 10.3390/ijms21186837] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/10/2020] [Accepted: 09/13/2020] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) play a key role in the communication between cancer cells and stromal components of the tumor microenvironment (TME). In this context, cancer cell-derived EVs can regulate the activation of a CAF phenotype in TME cells, which can be mediated by several EV cargos (e.g., miRNA, proteins, mRNA and lncRNAs). On the other hand, CAF-derived EVs can mediate several processes during tumorigenesis, including tumor growth, invasion, metastasis, and therapy resistance. This review aimed to discuss the molecular aspects of EV-based cross-talk between CAFs and cancer cells during tumorigenesis, in addition to assessing the roles of EV cargo in therapy resistance and pre-metastatic niche formation.
Collapse
Affiliation(s)
- Issraa Shoucair
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada; (I.S.); (F.W.M.); (J.J.); (S.M.)
| | - Fernanda Weber Mello
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada; (I.S.); (F.W.M.); (J.J.); (S.M.)
- Postgraduate Program in Dentistry, Federal University of Santa Catarina, Florianópolis 88.040-370, Brazil
| | - James Jabalee
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada; (I.S.); (F.W.M.); (J.J.); (S.M.)
| | - Saeideh Maleki
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada; (I.S.); (F.W.M.); (J.J.); (S.M.)
| | - Cathie Garnis
- Department of Surgery, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
- Correspondence:
| |
Collapse
|
46
|
Armistead J, Hatzold J, van Roye A, Fahle E, Hammerschmidt M. Entosis and apical cell extrusion constitute a tumor-suppressive mechanism downstream of Matriptase. J Cell Biol 2020; 219:132730. [PMID: 31819976 PMCID: PMC7041680 DOI: 10.1083/jcb.201905190] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 10/02/2019] [Accepted: 11/04/2019] [Indexed: 12/21/2022] Open
Abstract
Armistead et al. show that in a bilayered epithelium in vivo, apical cell extrusion of basal cells is achieved via their engulfment by surface cells. In zebrafish hai1a mutants, this constitutes a tumor-suppressive mechanism, revealing a double face of Matriptase. The type II transmembrane serine protease Matriptase 1 (ST14) is commonly known as an oncogene, yet it also plays an understudied role in suppressing carcinogenesis. This double face is evident in the embryonic epidermis of zebrafish loss-of-function mutants in the cognate Matriptase inhibitor Hai1a (Spint1a). Mutant embryos display epidermal hyperplasia, but also apical cell extrusions, during which extruding outer keratinocytes carry out an entosis-like engulfment and entrainment of underlying basal cells, constituting a tumor-suppressive effect. These counteracting Matriptase effects depend on EGFR and the newly identified mediator phospholipase D (PLD), which promotes both mTORC1-dependent cell proliferation and sphingosine-1-phosphate (S1P)–dependent entosis and apical cell extrusion. Accordingly, hypomorphic hai1a mutants heal spontaneously, while otherwise lethal hai1a amorphs are efficiently rescued upon cotreatment with PLD inhibitors and S1P. Together, our data elucidate the mechanisms underlying the double face of Matriptase function in vivo and reveal the potential use of combinatorial carcinoma treatments when such double-face mechanisms are involved.
Collapse
Affiliation(s)
- Joy Armistead
- Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Julia Hatzold
- Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany
| | - Anna van Roye
- Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany
| | - Evelin Fahle
- Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany
| | - Matthias Hammerschmidt
- Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| |
Collapse
|
47
|
Thome I, Lacle R, Voß A, Bortolussi G, Pantazis G, Schmidt A, Conrad C, Jacob R, Timmesfeld N, Bartsch JW, Pagenstecher A. Neoplastic Cells are the Major Source of MT-MMPs in IDH1-Mutant Glioma, Thus Enhancing Tumor-Cell Intrinsic Brain Infiltration. Cancers (Basel) 2020; 12:E2456. [PMID: 32872536 PMCID: PMC7565296 DOI: 10.3390/cancers12092456] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/07/2020] [Accepted: 08/24/2020] [Indexed: 11/16/2022] Open
Abstract
Tumor-cell infiltration is a major obstacle to successful therapy for brain tumors. Membrane-type matrix metalloproteinases (MT-MMPs), a metzincin subfamily of six proteases, are important mediators of infiltration. The cellular source of MT-MMPs and their role in glioma biology, however, remain controversial. Thus, we comprehensively analyzed the expression of MT-MMPs in primary brain tumors. All MT-MMPs were differentially expressed in primary brain tumors. In diffuse gliomas, MT-MMP1, -3, and -4 were predominantly expressed by IDH1mutated tumor cells, while macrophages/microglia contributed significantly less to MT-MMP expression. For functional analyses, individual MT-MMPs were expressed in primary mouse p53-/- astrocytes. Invasion and migration potential of MT-MMP-transduced astrocytes was determined via scratch, matrigel invasion, and novel organotypic porcine spinal slice migration (OPoSSM) and invasion assays. Overall, MT-MMP-transduced astrocytes showed enhanced migration compared to controls. MMP14 was the strongest mediator of migration in scratch assays. However, in the OPoSSM assays, the glycosylphosphatidylinositol (GPI)-anchored MT-MMPs MMP17 and MMP25, not MMP14, mediated the highest infiltration rates of astrocytes. Our data unequivocally demonstrate for the first time that glioma cells, not microglia, are the predominant producers of MT-MMPs in glioma and can act as potent mediators of tumor-cell infiltration into CNS tissue. These proteases are therefore promising targets for therapeutic interventions.
Collapse
Affiliation(s)
- Ina Thome
- Departments of Neuropathology, Philipps University Marburg, 35043 Marburg, Germany; (I.T.); (R.L.); (A.V.); (G.B.); (G.P.); (C.C.)
| | - Raphael Lacle
- Departments of Neuropathology, Philipps University Marburg, 35043 Marburg, Germany; (I.T.); (R.L.); (A.V.); (G.B.); (G.P.); (C.C.)
| | - Andreas Voß
- Departments of Neuropathology, Philipps University Marburg, 35043 Marburg, Germany; (I.T.); (R.L.); (A.V.); (G.B.); (G.P.); (C.C.)
| | - Ginette Bortolussi
- Departments of Neuropathology, Philipps University Marburg, 35043 Marburg, Germany; (I.T.); (R.L.); (A.V.); (G.B.); (G.P.); (C.C.)
| | - Georgios Pantazis
- Departments of Neuropathology, Philipps University Marburg, 35043 Marburg, Germany; (I.T.); (R.L.); (A.V.); (G.B.); (G.P.); (C.C.)
| | - Ansgar Schmidt
- Departments of Pathology, Philipps University Marburg, 35043 Marburg, Germany;
| | - Catharina Conrad
- Departments of Neuropathology, Philipps University Marburg, 35043 Marburg, Germany; (I.T.); (R.L.); (A.V.); (G.B.); (G.P.); (C.C.)
| | - Ralf Jacob
- Departments of Clinical Cytobiology and Cytopathology, Philipps University Marburg, 35037 Marburg, Germany;
| | - Nina Timmesfeld
- Department of Medical Informatics, Biometry and Epidemiology, Ruhr University Bochum, 44780 Bochum, Germany;
| | - Jörg W. Bartsch
- Departments of Neurosurgery, Philipps University Marburg, 35043 Marburg, Germany;
- Centre for Mind, Brain, and Behaviour, 35032 Marburg, Germany
| | - Axel Pagenstecher
- Departments of Neuropathology, Philipps University Marburg, 35043 Marburg, Germany; (I.T.); (R.L.); (A.V.); (G.B.); (G.P.); (C.C.)
- Centre for Mind, Brain, and Behaviour, 35032 Marburg, Germany
| |
Collapse
|
48
|
Kallikrein-Related Peptidase 14 Activates Zymogens of Membrane Type Matrix Metalloproteinases (MT-MMPs)-A CleavEx Based Analysis. Int J Mol Sci 2020; 21:ijms21124383. [PMID: 32575583 PMCID: PMC7352328 DOI: 10.3390/ijms21124383] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/09/2020] [Accepted: 06/17/2020] [Indexed: 01/02/2023] Open
Abstract
Kallikrein-related peptidases (KLKs) and matrix metalloproteinases (MMPs) are secretory proteinases known to proteolytically process components of the extracellular matrix, modulating the pericellular environment in physiology and in pathologies. The interconnection between these families remains elusive. To assess the cross-activation of these families, we developed a peptide, fusion protein-based exposition system (Cleavage of exposed amino acid sequences, CleavEx) aiming at investigating the potential of KLK14 to recognize and hydrolyze proMMP sequences. Initial assessment identified ten MMP activation domain sequences which were validated by Edman degradation. The analysis revealed that membrane-type MMPs (MT-MMPs) are targeted by KLK14 for activation. Correspondingly, proMMP14-17 were investigated in vitro and found to be effectively processed by KLK14. Again, the expected neo-N-termini of the activated MT-MMPs was confirmed by Edman degradation. The effectiveness of proMMP activation was analyzed by gelatin zymography, confirming the release of fully active, mature MT-MMPs upon KLK14 treatment. Lastly, MMP14 was shown to be processed on the cell surface by KLK14 using murine fibroblasts overexpressing human MMP14. Herein, we propose KLK14-mediated selective activation of cell-membrane located MT-MMPs as an additional layer of their regulation. As both, KLKs and MT-MMPs, are implicated in cancer, their cross-activation may constitute an important factor in tumor progression and metastasis.
Collapse
|
49
|
Napoli S, Scuderi C, Gattuso G, Di Bella V, Candido S, Basile MS, Libra M, Falzone L. Functional Roles of Matrix Metalloproteinases and Their Inhibitors in Melanoma. Cells 2020; 9:cells9051151. [PMID: 32392801 PMCID: PMC7291303 DOI: 10.3390/cells9051151] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/01/2020] [Accepted: 05/06/2020] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix (ECM) plays an important role in the regulation of the tissue microenvironment and in the maintenance of cellular homeostasis. Several proteins with a proteolytic activity toward several ECM components are involved in the regulation and remodeling of the ECM. Among these, Matrix Metalloproteinases (MMPs) are a class of peptidase able to remodel the ECM by favoring the tumor invasive processes. Of these peptidases, MMP-9 is the most involved in the development of cancer, including that of melanoma. Dysregulations of the MAPKs and PI3K/Akt signaling pathways can lead to an aberrant overexpression of MMP-9. Even ncRNAs are implicated in the aberrant production of MMP-9 protein, as well as other proteins responsible for the activation or inhibition of MMP-9, such as Osteopontin and Tissue Inhibitors of Metalloproteinases. Currently, there are different therapeutic approaches for melanoma, including targeted therapies and immunotherapies. However, no biomarkers are available for the prediction of the therapeutic response. In this context, several studies have tried to understand the diagnostic, prognostic and therapeutic potential of MMP-9 in melanoma patients by performing clinical trials with synthetic MMPs inhibitors. Therefore, MMP-9 may be considered a promising molecule for the management of melanoma patients due to its role as a biomarker and therapeutic target.
Collapse
Affiliation(s)
- Salvatore Napoli
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (S.N.); (C.S.); (G.G.); (V.D.B.); (S.C.); (M.S.B.)
| | - Chiara Scuderi
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (S.N.); (C.S.); (G.G.); (V.D.B.); (S.C.); (M.S.B.)
| | - Giuseppe Gattuso
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (S.N.); (C.S.); (G.G.); (V.D.B.); (S.C.); (M.S.B.)
| | - Virginia Di Bella
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (S.N.); (C.S.); (G.G.); (V.D.B.); (S.C.); (M.S.B.)
| | - Saverio Candido
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (S.N.); (C.S.); (G.G.); (V.D.B.); (S.C.); (M.S.B.)
- Research Center for Prevention, Diagnosis and Treatment of Cancer, University of Catania, 95123 Catania, Italy
| | - Maria Sofia Basile
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (S.N.); (C.S.); (G.G.); (V.D.B.); (S.C.); (M.S.B.)
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (S.N.); (C.S.); (G.G.); (V.D.B.); (S.C.); (M.S.B.)
- Research Center for Prevention, Diagnosis and Treatment of Cancer, University of Catania, 95123 Catania, Italy
- Correspondence: (M.L.); or (L.F.); Tel.: +39-095-478-1271 (M.L.); +39-094-478-1278 (L.F.)
| | - Luca Falzone
- Epidemiology Unit, IRCCS Istituto Nazionale Tumori “Fondazione G. Pascale”, 80131 Naples, Italy
- Correspondence: (M.L.); or (L.F.); Tel.: +39-095-478-1271 (M.L.); +39-094-478-1278 (L.F.)
| |
Collapse
|
50
|
Andrieu C, Montigny A, Bibonne A, Despin-Guitard E, Alfandari D, Théveneau E. MMP14 is required for delamination of chick neural crest cells independently of its catalytic activity. Development 2020; 147:dev.183954. [PMID: 32280063 DOI: 10.1242/dev.183954] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 02/24/2020] [Indexed: 12/13/2022]
Abstract
Matrix metalloproteinases have a broad spectrum of substrates ranging from extracellular matrix components and adhesion molecules to chemokines and growth factors. Despite being mostly secreted, MMPs have been detected in the cytosol, the mitochondria or the nucleus. Although most of the attention is focused on their role in matrix remodeling, the diversity of their substrates and their complex trafficking open the possibility for non-canonical functions. Yet in vivo examples and experimental demonstration of the physiological relevance of such activities are rare. Here, we have used chick neural crest (NC) cells, a highly migratory stem cell population likened to invasive cancer cells, as a model for physiological epithelial-mesenchymal transition (EMT). We demonstrate that MMP14 is required for NC delamination. Interestingly, this role is independent of its cytoplasmic tail and of its catalytic activity. Our in vivo data indicate that, in addition to being a late pro-invasive factor, MMP14 is also likely to be an early player, owing to its role in EMT.
Collapse
Affiliation(s)
- Cyril Andrieu
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, 31062, France
| | - Audrey Montigny
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, 31062, France
| | - Anne Bibonne
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, 31062, France
| | - Evangeline Despin-Guitard
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, 31062, France
| | - Dominique Alfandari
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Eric Théveneau
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, 31062, France
| |
Collapse
|