1
|
Yu Y, Cao WM, Cheng F, Shi Z, Han L, Yi J, da Silva EM, Dopeso H, Chen H, Yang J, Wang X, Zhang C, Zhang H. FOXK2 amplification promotes breast cancer development and chemoresistance. Cancer Lett 2024; 597:217074. [PMID: 38901667 PMCID: PMC11290987 DOI: 10.1016/j.canlet.2024.217074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 05/22/2024] [Accepted: 06/14/2024] [Indexed: 06/22/2024]
Abstract
Oncogene activation through DNA amplification or overexpression is a crucial driver of cancer initiation and progression. The FOXK2 gene, located on chromosome 17q25, encodes a transcription factor with a forkhead DNA-binding domain. Analysis of genomic datasets reveals that FOXK2 is frequently amplified and overexpressed in breast cancer, correlating with poor patient survival. Knockdown of FOXK2 significantly inhibited breast cancer cell proliferation, migration, anchorage-independent growth, and delayed tumor growth in a xenograft mouse model. Additionally, inhibiting FOXK2 sensitized breast cancer cells to chemotherapy. Co-overexpression of FOXK2 and mutant PI3KCA transformed non-tumorigenic MCF-10A cells, suggesting a role for FOXK2 in PI3KCA-driven tumorigenesis. CCNE2, PDK1, and ESR1 were identified as transcriptional targets of FOXK2 in MCF-7 cells. Small-molecule inhibitors of CCNE2/CDK2 (dinaciclib) and PDK1 (dichloroacetate) exhibited synergistic anti-tumor effects with PI3KCA inhibitor (alpelisib) in vitro. Inhibition of FOXK2 by dinaciclib synergistically enhanced the anti-tumor effects of alpelisib in a xenograft mouse model. Collectively, these findings highlight the oncogenic function of FOXK2 and suggest that FOXK2 and its downstream genes represent potential therapeutic targets in breast cancer.
Collapse
Affiliation(s)
- Yang Yu
- Center for Cancer and Immunology Research, Children's National Research Institute, Children's National Hospital, Washington, DC, 20010, USA
| | - Wen-Ming Cao
- Department of Pathology & Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Feng Cheng
- Center for Cancer and Immunology Research, Children's National Research Institute, Children's National Hospital, Washington, DC, 20010, USA
| | - Zhongcheng Shi
- Advanced Technology Cores, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Lili Han
- Department of Pathology & Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jinling Yi
- Texas Children's Hospital, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Edaise M da Silva
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Higinio Dopeso
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Hui Chen
- Department of Pathology & Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jianhua Yang
- Center for Cancer and Immunology Research, Children's National Research Institute, Children's National Hospital, Washington, DC, 20010, USA; Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20010, USA
| | - Xiaosong Wang
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Chunchao Zhang
- Center for Cancer and Immunology Research, Children's National Research Institute, Children's National Hospital, Washington, DC, 20010, USA; Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20010, USA.
| | - Hong Zhang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| |
Collapse
|
2
|
Brodesser DM, Kummer S, Eichberger JA, Schlangen K, Corteggio A, Borzacchiello G, Bertram CA, Brandt S, Pratscher B. Deregulation of Metalloproteinase Expression in Gray Horse Melanoma Ex Vivo and In Vitro. Cells 2024; 13:956. [PMID: 38891088 PMCID: PMC11172212 DOI: 10.3390/cells13110956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 05/25/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
The ability of human melanoma cells to switch from an epithelial to a mesenchymal phenotype contributes to the metastatic potential of disease. Metalloproteinases (MPs) are crucially involved in this process by promoting the detachment of tumor cells from the primary lesion and their migration to the vasculature. In gray horse melanoma, epithelial-mesenchymal transition (EMT) is poorly understood, prompting us to address MP expression in lesions versus intact skin by transcriptome analyses and the immunofluorescence staining (IF) of gray horse tumor tissue and primary melanoma cells. RNAseq revealed the deregulation of several MPs in gray horse melanoma and, notably, a 125-fold upregulation of matrix metalloproteinase 1 (MMP1) that was further confirmed by RT-qPCR from additional tumor material. The IF staining of melanoma tissue versus intact skin for MMP1 and tumor marker S100 revealed MMP1 expression in all lesions. The co-expression of S100 was observed at different extents, with some tumors scoring S100-negative. The IF staining of primary tumor cells explanted from the tumors for MMP1 showed that the metalloproteinase is uniformly expressed in the cytoplasm of 100% of tumor cells. Overall, the presented data point to MP expression being deregulated in gray horse melanoma, and suggest that MMP1 has an active role in gray horse melanoma by driving EMT-mediated tumor cell dissemination via the degradation of the extracellular matrix. Whilst S100 is considered a reliable tumor marker in human MM, gray horse melanomas do not seem to regularly express this protein.
Collapse
Affiliation(s)
- Daniela M. Brodesser
- Research Group Oncology (RGO), Centre for Equine Health and Research, Department for Small Animals and Horses, University of Veterinary Medicine, Veterinaerplatz 1, 1210 Vienna, Austria; (D.M.B.); (J.A.E.)
| | - Stefan Kummer
- VetImaging, VetCore Facility, University of Veterinary Medicine, Veterinaerplatz 1, 1210 Vienna, Austria;
| | - Julia A. Eichberger
- Research Group Oncology (RGO), Centre for Equine Health and Research, Department for Small Animals and Horses, University of Veterinary Medicine, Veterinaerplatz 1, 1210 Vienna, Austria; (D.M.B.); (J.A.E.)
| | - Karin Schlangen
- Section for Biosimulation and Bioinformatics, Centre for Medical Statistics, Informatics and Intelligent Systems, Medical University of Vienna (MUV), Waehringer Guertel 18-20, 1090 Vienna, Austria;
| | - Annunziata Corteggio
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Via Pietro Castellino 111, 80131 Naples, Italy;
| | - Giuseppe Borzacchiello
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Corso Umberto I 40, 80138 Naples, Italy;
| | - Christof A. Bertram
- Institute of Pathology, Department of Pathobiology, University of Veterinary Medicine, Veterinaerplatz 1, 1210 Vienna, Austria;
| | - Sabine Brandt
- Research Group Oncology (RGO), Centre for Equine Health and Research, Department for Small Animals and Horses, University of Veterinary Medicine, Veterinaerplatz 1, 1210 Vienna, Austria; (D.M.B.); (J.A.E.)
| | - Barbara Pratscher
- Division of Small Animal Internal Medicine, Department for Small Animals and Horses, University of Veterinary Medicine, Veterinaerplatz 1, 1210 Vienna, Austria;
| |
Collapse
|
3
|
Wang Y, Zou L, Song M, Zong J, Wang S, Meng L, Jia Z, Zhao L, Han X, Lu M. Establishment of skin cutaneous melanoma prognosis model based on vascular mimicry risk score. Medicine (Baltimore) 2024; 103:e36679. [PMID: 38363903 PMCID: PMC10869071 DOI: 10.1097/md.0000000000036679] [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: 07/02/2023] [Accepted: 11/24/2023] [Indexed: 02/18/2024] Open
Abstract
Studies have indicated that Vascular mimicry (VM) could contribute to the unfavorable prognosis of skin cutaneous melanoma (SKCM). Thus, the objective of this study was to identify therapeutic targets associated with VM in SKCM and develop a novel prognostic model. Gene expression data from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) were utilized to identify differentially expressed genes (DEGs). By intersecting these DEGs with VM genes, we acquired VM-related DEGs specific to SKCM, and then identified prognostic-related VM genes. A VM risk score system was established based on these prognosis-associated VM genes, and patients were then categorized into high- and low-score groups using the median score. Subsequently, differences in clinical characteristics, gene set enrichment analysis (GSEA), and other analyses were further presented between the 2 groups of patients. Finally, a novel prognostic model for SKCM was established using the VM score and clinical characteristics. 26 VM-related DEGs were identified in SKCM, among the identified DEGs associated with VM in SKCM, 5 genes were found to be prognostic-related. The VM risk score system, comprised of these genes, is an independent prognostic risk factor. There were significant differences between the 2 patient groups in terms of age, pathological stage, and T stage. VM risk scores are associated with epithelial biological processes, angiogenesis, regulation of the SKCM immune microenvironment, and sensitivity to targeted drugs. The novel prognostic model demonstrates excellent predictive ability. Our study identified VM-related prognostic markers and therapeutic targets for SKCM, providing novel insights for clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Yubo Wang
- Dalian Medical University, Dalian, China
- Department of Trauma and Tissue Repair Surgery, Dalian Municipal Central Hospital, Dalian, China
| | - Linxuan Zou
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Mingzhi Song
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Junwei Zong
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shouyu Wang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Lei Meng
- The First Affiliated Hospital of Nanhua Medical University, Hengyang, China
| | - Zhuqiang Jia
- The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Naqu People’s Hospital, Tibet, China
| | - Lin Zhao
- Department of Quality Management, Dalian Municipal Central Hospital, Dalian, China
| | - Xin Han
- Naqu People’s Hospital, Tibet, China
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ming Lu
- Department of Trauma and Tissue Repair Surgery, Dalian Municipal Central Hospital, Dalian, China
- Department of Trauma and Tissue Repair Surgery, Dalian Municipal Central Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
4
|
Slama Y, Ah-Pine F, Khettab M, Arcambal A, Begue M, Dutheil F, Gasque P. The Dual Role of Mesenchymal Stem Cells in Cancer Pathophysiology: Pro-Tumorigenic Effects versus Therapeutic Potential. Int J Mol Sci 2023; 24:13511. [PMID: 37686315 PMCID: PMC10488262 DOI: 10.3390/ijms241713511] [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: 08/02/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are multipotent cells involved in numerous physiological events, including organogenesis, the maintenance of tissue homeostasis, regeneration, or tissue repair. MSCs are increasingly recognized as playing a major, dual, and complex role in cancer pathophysiology through their ability to limit or promote tumor progression. Indeed, these cells are known to interact with the tumor microenvironment, modulate the behavior of tumor cells, influence their functions, and promote distant metastasis formation through the secretion of mediators, the regulation of cell-cell interactions, and the modulation of the immune response. This dynamic network can lead to the establishment of immunoprivileged tissue niches or the formation of new tumors through the proliferation/differentiation of MSCs into cancer-associated fibroblasts as well as cancer stem cells. However, MSCs exhibit also therapeutic effects including anti-tumor, anti-proliferative, anti-inflammatory, or anti-oxidative effects. The therapeutic interest in MSCs is currently growing, mainly due to their ability to selectively migrate and penetrate tumor sites, which would make them relevant as vectors for advanced therapies. Therefore, this review aims to provide an overview of the double-edged sword implications of MSCs in tumor processes. The therapeutic potential of MSCs will be reviewed in melanoma and lung cancers.
Collapse
Affiliation(s)
- Youssef Slama
- Unité de Recherche Études Pharmaco-Immunologiques (EPI), Université de La Réunion, CHU de La Réunion, Allée des Topazes, 97400 Saint-Denis, La Réunion, France; (F.A.-P.); (M.K.); (P.G.)
- Service de Radiothérapie, Clinique Sainte-Clotilde, Groupe Clinifutur, 127 Route de Bois de Nèfles, 97400 Saint-Denis, La Réunion, France; (M.B.); (F.D.)
- Laboratoire Interdisciplinaire de Recherche en Santé (LIRS), RunResearch, Clinique Sainte-Clotilde, 127 Route de Bois de Nèfles, 97400 Saint-Denis, La Réunion, France;
| | - Franck Ah-Pine
- Unité de Recherche Études Pharmaco-Immunologiques (EPI), Université de La Réunion, CHU de La Réunion, Allée des Topazes, 97400 Saint-Denis, La Réunion, France; (F.A.-P.); (M.K.); (P.G.)
- Service d’Anatomie et Cytologie Pathologiques, CHU de La Réunion sites SUD—Saint-Pierre, Avenue François Mitterrand, 97448 Saint-Pierre Cedex, La Réunion, France
| | - Mohamed Khettab
- Unité de Recherche Études Pharmaco-Immunologiques (EPI), Université de La Réunion, CHU de La Réunion, Allée des Topazes, 97400 Saint-Denis, La Réunion, France; (F.A.-P.); (M.K.); (P.G.)
- Service d’Oncologie Médicale, CHU de La Réunion sites SUD—Saint-Pierre, Avenue François Mitterrand, 97448 Saint-Pierre Cedex, La Réunion, France
| | - Angelique Arcambal
- Laboratoire Interdisciplinaire de Recherche en Santé (LIRS), RunResearch, Clinique Sainte-Clotilde, 127 Route de Bois de Nèfles, 97400 Saint-Denis, La Réunion, France;
| | - Mickael Begue
- Service de Radiothérapie, Clinique Sainte-Clotilde, Groupe Clinifutur, 127 Route de Bois de Nèfles, 97400 Saint-Denis, La Réunion, France; (M.B.); (F.D.)
- Laboratoire Interdisciplinaire de Recherche en Santé (LIRS), RunResearch, Clinique Sainte-Clotilde, 127 Route de Bois de Nèfles, 97400 Saint-Denis, La Réunion, France;
| | - Fabien Dutheil
- Service de Radiothérapie, Clinique Sainte-Clotilde, Groupe Clinifutur, 127 Route de Bois de Nèfles, 97400 Saint-Denis, La Réunion, France; (M.B.); (F.D.)
- Laboratoire Interdisciplinaire de Recherche en Santé (LIRS), RunResearch, Clinique Sainte-Clotilde, 127 Route de Bois de Nèfles, 97400 Saint-Denis, La Réunion, France;
| | - Philippe Gasque
- Unité de Recherche Études Pharmaco-Immunologiques (EPI), Université de La Réunion, CHU de La Réunion, Allée des Topazes, 97400 Saint-Denis, La Réunion, France; (F.A.-P.); (M.K.); (P.G.)
| |
Collapse
|
5
|
Chhabra Y, Weeraratna AT. Fibroblasts in cancer: Unity in heterogeneity. Cell 2023; 186:1580-1609. [PMID: 37059066 PMCID: PMC11422789 DOI: 10.1016/j.cell.2023.03.016] [Citation(s) in RCA: 90] [Impact Index Per Article: 90.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 04/16/2023]
Abstract
Tumor cells do not exist in isolation in vivo, and carcinogenesis depends on the surrounding tumor microenvironment (TME), composed of a myriad of cell types and biophysical and biochemical components. Fibroblasts are integral in maintaining tissue homeostasis. However, even before a tumor develops, pro-tumorigenic fibroblasts in close proximity can provide the fertile 'soil' to the cancer 'seed' and are known as cancer-associated fibroblasts (CAFs). In response to intrinsic and extrinsic stressors, CAFs reorganize the TME enabling metastasis, therapeutic resistance, dormancy and reactivation by secreting cellular and acellular factors. In this review, we summarize the recent discoveries on CAF-mediated cancer progression with a particular focus on fibroblast heterogeneity and plasticity.
Collapse
Affiliation(s)
- Yash Chhabra
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Department of Oncology, Sidney Kimmel Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA.
| | - Ashani T Weeraratna
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Department of Oncology, Sidney Kimmel Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
6
|
González-Zamora J, Hernandez M, Recalde S, Bezunartea J, Montoliu A, Bilbao-Malavé V, Llorente-González S, García-Layana A, Fernández-Robredo P. Matrix Metalloproteinase 13 Is Associated with Age-Related Choroidal Neovascularization. Antioxidants (Basel) 2023; 12:antiox12040884. [PMID: 37107259 PMCID: PMC10135211 DOI: 10.3390/antiox12040884] [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: 02/14/2023] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of severe vision loss in older individuals in developed countries. Despite advances in our understanding of AMD, its pathophysiology remains poorly understood. Matrix metalloproteinases (MMPs) have been proposed to play a role in AMD development. In this study, we aimed to characterize MMP-13 in AMD. We used retinal pigment epithelial cells, a murine model of laser-induced choroidal neovascularization, and plasma samples from patients with neovascular AMD to conduct our study. Our results show that MMP13 expression significantly increased under oxidative stress conditions in cultured retinal pigment epithelial cells. In the murine model, MMP13 was overexpressed in both retinal pigment epithelial cells and endothelial cells during choroidal neovascularization. Additionally, the total MMP13 levels in the plasma of patients with neovascular AMD were significantly lower than those in the control group. This suggests a reduced diffusion from the tissues or release from circulating cells in the bloodstream, given that the number and function of monocytes have been reported to be deficient in patients with AMD. Although more studies are needed to elucidate the role of MMP13 in AMD, it could be a promising therapeutic target for treating AMD.
Collapse
Affiliation(s)
- Jorge González-Zamora
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Clinica Universidad de Navarra, 31008 Pamplona, Spain
| | - María Hernandez
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Navarra Institute for Health Research, IdiSNA, Clinica Universidad de Navarra, 31008 Pamplona, Spain
| | - Sergio Recalde
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Navarra Institute for Health Research, IdiSNA, Clinica Universidad de Navarra, 31008 Pamplona, Spain
| | - Jaione Bezunartea
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Navarra Institute for Health Research, IdiSNA, Clinica Universidad de Navarra, 31008 Pamplona, Spain
| | - Ana Montoliu
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Clinica Universidad de Navarra, 31008 Pamplona, Spain
| | - Valentina Bilbao-Malavé
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Clinica Universidad de Navarra, 31008 Pamplona, Spain
| | - Sara Llorente-González
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Navarra Institute for Health Research, IdiSNA, Clinica Universidad de Navarra, 31008 Pamplona, Spain
| | - Alfredo García-Layana
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Navarra Institute for Health Research, IdiSNA, Clinica Universidad de Navarra, 31008 Pamplona, Spain
| | - Patricia Fernández-Robredo
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Navarra Institute for Health Research, IdiSNA, Clinica Universidad de Navarra, 31008 Pamplona, Spain
| |
Collapse
|
7
|
Fuerst R, Choi JY, Knapinska AM, Cameron MD, Ruiz C, Delmas A, Sundrud MS, Fields GB, Roush WR. Development of a putative Zn2+-chelating but highly selective MMP-13 inhibitor. Bioorg Med Chem Lett 2022; 76:129014. [DOI: 10.1016/j.bmcl.2022.129014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/17/2022] [Accepted: 09/28/2022] [Indexed: 11/25/2022]
|
8
|
Kümper M, Zamek J, Steinkamp J, Pach E, Mauch C, Zigrino P. Role of MMP3 and fibroblast-MMP14 in skin homeostasis and repair. Eur J Cell Biol 2022; 101:151276. [PMID: 36162272 DOI: 10.1016/j.ejcb.2022.151276] [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: 07/06/2022] [Revised: 09/09/2022] [Accepted: 09/21/2022] [Indexed: 12/14/2022] Open
Abstract
Early lethality of mice with complete deletion of the matrix metalloproteinase MMP14 emphasized the proteases' pleiotropic functions. MMP14 deletion in adult dermal fibroblasts (MMP14Sf-/-) caused collagen type I accumulation and upregulation of MMP3 expression. To identify the compensatory role of MMP3, mice were generated with MMP3 deletion in addition to MMP14 loss in fibroblasts. These double deficient mice displayed a fibrotic phenotype in skin and tendons as detected in MMP14Sf-/- mice, but no additional obvious defects were detected. However, challenging the mice with full thickness excision wounds resulted in delayed closure of early wounds in the double deficient mice compared to wildtype and MMP14 single knockout controls. Over time wounds closed and epidermal integrity was restored. Interestingly, on day seven, post-wounding myofibroblast density was lower in the wounds of all knockout than in controls, they were higher on day 14. The delayed resolution of myofibroblasts from the granulation tissue is paralleled by reduced apoptosis of these cells, although proliferation of myofibroblasts is induced in the double deficient mice. Further analysis showed comparable TGFβ1 and TGFβR1 expression among all genotypes. In addition, in vitro, fibroblasts lacking MMP3 and MMP14 retained their ability to differentiate into myofibroblasts in response to TGFβ1 treatment and mechanical stress. However, in vivo, p-Smad2 was reduced in myofibroblasts at day 5 post-wounding, in double, but most significant in single knockout, indicating their involvement in TGFβ1 activation. Thus, although MMP3 does not compensate for the lack of fibroblast-MMP14 in tissue homeostasis, simultaneous deletion of both proteases in fibroblasts delays wound closure during skin repair. Notably, single and double deficiency of these proteases modulates myofibroblast formation and resolution in wounds.
Collapse
Affiliation(s)
- Maike Kümper
- Department of Dermatology and Venereology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.
| | - Jan Zamek
- Department of Dermatology and Venereology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.
| | - Joy Steinkamp
- Department of Dermatology and Venereology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.
| | - Elke Pach
- Department of Dermatology and Venereology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.
| | - Cornelia Mauch
- Department of Dermatology and Venereology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.
| | - Paola Zigrino
- Department of Dermatology and Venereology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.
| |
Collapse
|
9
|
Fromme JE, Zigrino P. The Role of Extracellular Matrix Remodeling in Skin Tumor Progression and Therapeutic Resistance. Front Mol Biosci 2022; 9:864302. [PMID: 35558554 PMCID: PMC9086898 DOI: 10.3389/fmolb.2022.864302] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/23/2022] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix remodeling in the skin results from a delicate balance of synthesis and degradation of matrix components, ensuring tissue homeostasis. These processes are altered during tumor invasion and growth, generating a microenvironment that supports growth, invasion, and metastasis. Apart from the cellular component, the tumor microenvironment is rich in extracellular matrix components and bound factors that provide structure and signals to the tumor and stromal cells. The continuous remodeling in the tissue compartment sustains the developing tumor during the various phases providing matrices and proteolytic enzymes. These are produced by cancer cells and stromal fibroblasts. In addition to fostering tumor growth, the expression of specific extracellular matrix proteins and proteinases supports tumor invasion after the initial therapeutic response. Lately, the expression and structural modification of matrices were also associated with therapeutic resistance. This review will focus on the significant alterations in the extracellular matrix components and the function of metalloproteinases that influence skin cancer progression and support the acquisition of therapeutic resistance.
Collapse
Affiliation(s)
- Julia E. Fromme
- Department of Dermatology and Venereology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Mildred Scheel School of Oncology Aachen Bonn Cologne Düsseldorf (MSSO ABCD), Cologne, Germany
| | - Paola Zigrino
- Department of Dermatology and Venereology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- *Correspondence: Paola Zigrino,
| |
Collapse
|
10
|
Effects of Maternal Diabetes and Diet on Gene Expression in the Murine Placenta. Genes (Basel) 2022; 13:genes13010130. [PMID: 35052470 PMCID: PMC8775503 DOI: 10.3390/genes13010130] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/08/2022] [Accepted: 01/10/2022] [Indexed: 11/16/2022] Open
Abstract
Adverse exposures during pregnancy have been shown to contribute to susceptibility for chronic diseases in offspring. Maternal diabetes during pregnancy is associated with higher risk of pregnancy complications, structural birth defects, and cardiometabolic health impairments later in life. We showed previously in a mouse model that the placenta is smaller in diabetic pregnancies, with reduced size of the junctional zone and labyrinth. In addition, cell migration is impaired, resulting in ectopic accumulation of spongiotrophoblasts within the labyrinth. The present study had the goal to identify the mechanisms underlying the growth defects and trophoblast migration abnormalities. Based upon gene expression assays of 47 candidate genes, we were able to attribute the reduced growth of diabetic placenta to alterations in the Insulin growth factor and Serotonin signaling pathways, and provide evidence for Prostaglandin signaling deficiencies as the possible cause for abnormal trophoblast migration. Furthermore, our results reinforce the notion that the exposure to maternal diabetes has particularly pronounced effects on gene expression at midgestation time points. An implication of these findings is that mechanisms underlying developmental programming act early in pregnancy, during placenta morphogenesis, and before the conceptus switches from histiotrophic to hemotrophic nutrition.
Collapse
|
11
|
Kümper M, Hessenthaler S, Zamek J, Niland S, Pach E, Mauch C, Zigrino P. LOSS OF ENDOTHELIAL CELL MMP14 REDUCES MELANOMA GROWTH AND METASTASIS BY INCREASING TUMOR VESSEL STABILITY. J Invest Dermatol 2021; 142:1923-1933.e5. [DOI: 10.1016/j.jid.2021.12.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 12/03/2021] [Accepted: 12/14/2021] [Indexed: 12/13/2022]
|
12
|
Rapid orderly migration of neutrophils after traumatic brain injury depends on MMP9/13. Biochem Biophys Res Commun 2021; 579:161-167. [PMID: 34601201 DOI: 10.1016/j.bbrc.2021.09.044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 09/19/2021] [Indexed: 12/20/2022]
Abstract
Macrophages and granulocytes play an important role in various injuries and post-traumatic repair. Due to the limited number of neutrophils in the brain, their role in traumatic brain injury has rarely been mentioned. Here, neutrophils were found to take over the role of macrophages after brain injury in the absence of macrophages. Neutrophils have the characteristics of long residence time and number advantage to actively remove the apoptotic debris. The number of neutrophils recruited was effectively reduced by inhibiting IL-1β. Interestingly, neutrophils migrated regularly and rapidly to the wound during the early stages of brain injury through three paths. They first infiltrated the wound mainly through blood circulation around the eyes, then became unscrupulous and began to move directly across the brain. In addition, MMP9 and MMP13 were found to be related to the migration of neutrophils, and inhibition of MMP could significantly inhibit the number and speed of neutrophils' migration. Our study showed that neutrophils rely on MMP9 and MMP13 for a rapid and orderly response to brain injury to maintain central nervous system stability in the absence or decrease of macrophages.
Collapse
|
13
|
Treps L, Faure S, Clere N. Vasculogenic mimicry, a complex and devious process favoring tumorigenesis – Interest in making it a therapeutic target. Pharmacol Ther 2021; 223:107805. [DOI: 10.1016/j.pharmthera.2021.107805] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
14
|
Soltantoyeh T, Akbari B, Karimi A, Mahmoodi Chalbatani G, Ghahri-Saremi N, Hadjati J, Hamblin MR, Mirzaei HR. Chimeric Antigen Receptor (CAR) T Cell Therapy for Metastatic Melanoma: Challenges and Road Ahead. Cells 2021; 10:cells10061450. [PMID: 34207884 PMCID: PMC8230324 DOI: 10.3390/cells10061450] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 05/31/2021] [Accepted: 06/09/2021] [Indexed: 12/11/2022] Open
Abstract
Metastatic melanoma is the most aggressive and difficult to treat type of skin cancer, with a survival rate of less than 10%. Metastatic melanoma has conventionally been considered very difficult to treat; however, recent progress in understanding the cellular and molecular mechanisms involved in the tumorigenesis, metastasis and immune escape have led to the introduction of new therapies. These include targeted molecular therapy and novel immune-based approaches such as immune checkpoint blockade (ICB), tumor-infiltrating lymphocytes (TILs), and genetically engineered T-lymphocytes such as chimeric antigen receptor (CAR) T cells. Among these, CAR T cell therapy has recently made promising strides towards the treatment of advanced hematological and solid cancers. Although CAR T cell therapy might offer new hope for melanoma patients, it is not without its shortcomings, which include off-target toxicity, and the emergence of resistance to therapy (e.g., due to antigen loss), leading to eventual relapse. The present review will not only describe the basic steps of melanoma metastasis, but also discuss how CAR T cells could treat metastatic melanoma. We will outline specific strategies including combination approaches that could be used to overcome some limitations of CAR T cell therapy for metastatic melanoma.
Collapse
Affiliation(s)
- Tahereh Soltantoyeh
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1417613151, Iran; (T.S.); (B.A.); (G.M.C.); (N.G.-S.); (J.H.)
| | - Behnia Akbari
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1417613151, Iran; (T.S.); (B.A.); (G.M.C.); (N.G.-S.); (J.H.)
| | - Amirali Karimi
- School of Medicine, Tehran University of Medical Sciences, Tehran 1417613151, Iran;
| | - Ghanbar Mahmoodi Chalbatani
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1417613151, Iran; (T.S.); (B.A.); (G.M.C.); (N.G.-S.); (J.H.)
| | - Navid Ghahri-Saremi
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1417613151, Iran; (T.S.); (B.A.); (G.M.C.); (N.G.-S.); (J.H.)
| | - Jamshid Hadjati
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1417613151, Iran; (T.S.); (B.A.); (G.M.C.); (N.G.-S.); (J.H.)
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa;
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Hamid Reza Mirzaei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1417613151, Iran; (T.S.); (B.A.); (G.M.C.); (N.G.-S.); (J.H.)
- Correspondence: ; Tel.: +98-21-64053268; Fax: +98-21-66419536
| |
Collapse
|
15
|
Fibroblast MMP14-Dependent Collagen Processing Is Necessary for Melanoma Growth. Cancers (Basel) 2021; 13:cancers13081984. [PMID: 33924099 PMCID: PMC8074311 DOI: 10.3390/cancers13081984] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/12/2021] [Accepted: 04/16/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Matrix metalloproteinases (MMPs) were considered as targets for the treatment of various cancers. However, initial trials using broad inhibitors to MMPs have failed, partly attributed to the contrasting functions of these proteases acting as tumor promoters and suppressors, among other reasons. Our data now suggest that specific inhibition of MMP14 might represent a more specific approach, as loss of this protease in fibroblasts resulted in reduced growth of grafted melanomas. Here, we found that deletion of MMP14 in fibroblasts generates a matrix-rich environment that reduces tumor vascularization and melanoma cell proliferation. In in vitro and ex vivo assays, we showed that the latter is mediated by stiffening of the tissue due to collagen accumulation. Additionally, in vivo, we show that independently of MMP14 deletion, a collagen-rich stiff matrix inhibits the growth of melanomas. Abstract Skin homeostasis results from balanced synthesis and degradation of the extracellular matrix in the dermis. Deletion of the proteolytic enzyme MMP14 in dermal fibroblasts (MMP14Sf−/−) leads to a fibrotic skin phenotype with the accumulation of collagen type I, resulting from impaired proteolysis. Here, we show that melanoma growth in these mouse fibrotic dermal samples was decreased, paralleled by reduced tumor cell proliferation and vessel density. Using atomic force microscopy, we found increased peritumoral matrix stiffness of early but not late melanomas in the absence of fibroblast-derived MMP14. However, total collagen levels were increased at late melanoma stages in MMP14Sf−/− mice compared to controls. In ex vivo invasion assays, melanoma cells formed smaller tumor islands in MMP14Sf−/− skin, indicating that MMP14-dependent matrix accumulation regulates tumor growth. In line with these data, in vitro melanoma cell growth was inhibited in high collagen 3D spheroids or stiff substrates. Most importantly, in vivo induction of fibrosis using bleomycin reduced melanoma tumor growth. In summary, we show that MMP14 expression in stromal fibroblasts regulates melanoma tumor progression by modifying the peritumoral matrix and point to collagen accumulation as a negative regulator of melanoma.
Collapse
|
16
|
Salazar Y, Zheng X, Brunn D, Raifer H, Picard F, Zhang Y, Winter H, Guenther S, Weigert A, Weigmann B, Dumoutier L, Renauld JC, Waisman A, Schmall A, Tufman A, Fink L, Brüne B, Bopp T, Grimminger F, Seeger W, Pullamsetti SS, Huber M, Savai R. Microenvironmental Th9 and Th17 lymphocytes induce metastatic spreading in lung cancer. J Clin Invest 2021; 130:3560-3575. [PMID: 32229721 DOI: 10.1172/jci124037] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 03/24/2020] [Indexed: 01/10/2023] Open
Abstract
Immune microenvironment plays a critical role in lung cancer control versus progression and metastasis. In this investigation, we explored the effect of tumor-infiltrating lymphocyte subpopulations on lung cancer biology by studying in vitro cocultures, in vivo mouse models, and human lung cancer tissue. Lymphocyte conditioned media (CM) induced epithelial-mesenchymal transition (EMT) and migration in both primary human lung cancer cells and cell lines. Correspondingly, major accumulation of Th9 and Th17 cells was detected in human lung cancer tissue and correlated with poor survival. Coculturing lung cancer cells with Th9/Th17 cells or exposing them to the respective CM induced EMT in cancer cells and modulated the expression profile of genes implicated in EMT and metastasis. These features were reproduced by the signatory cytokines IL-9 and IL-17, with gene regulatory profiles evoked by these cytokines partly overlapping and partly complementary. Coinjection of Th9/Th17 cells with tumor cells in WT, Rag1-/-, Il9r-/-, and Il17ra-/- mice altered tumor growth and metastasis. Accordingly, inhibition of IL-9 or IL-17 cytokines by neutralizing antibodies decreased EMT and slowed lung cancer progression and metastasis. In conclusion, Th9 and Th17 lymphocytes induce lung cancer cell EMT, thereby promoting migration and metastatic spreading and offering potentially novel therapeutic strategies.
Collapse
Affiliation(s)
- Ylia Salazar
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, member of the German Center for Lung Research (DZL), member of Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Xiang Zheng
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, member of the German Center for Lung Research (DZL), member of Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - David Brunn
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, member of the German Center for Lung Research (DZL), member of Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Hartmann Raifer
- Institute for Medical Microbiology and.,CoreFacility Flow Cytometry, University of Marburg, Marburg, Germany
| | | | | | - Hauke Winter
- Translational Research Unit, Thoraxklinik at Heidelberg University, member of the DZL, Heidelberg, Germany
| | - Stefan Guenther
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Benno Weigmann
- Department of Medicine 1, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Laure Dumoutier
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | | | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Anja Schmall
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, member of the German Center for Lung Research (DZL), member of Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Amanda Tufman
- Respiratory Medicine and Thoracic Oncology, Internal Medicine V, Ludwig-Maximilians-University of Munich and Thoracic Oncology Centre, member of the DZL, Munich, Germany
| | - Ludger Fink
- Institute of Pathology and Cytology, Wetzlar, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany.,Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
| | - Tobias Bopp
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany. Research Center for Immunotherapy and University Medical Center, Johannes Gutenberg-University, Mainz, Germany. German Cancer Consortium, Heidelberg, Germany
| | - Friedrich Grimminger
- Department of Internal Medicine, member of the DZL, member of CPI, Justus Liebig University, Giessen, Germany
| | - Werner Seeger
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, member of the German Center for Lung Research (DZL), member of Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Department of Internal Medicine, member of the DZL, member of CPI, Justus Liebig University, Giessen, Germany.,Institute or Lung Health (ILH), Justus Liebig University, Giessen, Germany
| | - Soni Savai Pullamsetti
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, member of the German Center for Lung Research (DZL), member of Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Department of Internal Medicine, member of the DZL, member of CPI, Justus Liebig University, Giessen, Germany
| | | | - Rajkumar Savai
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, member of the German Center for Lung Research (DZL), member of Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany.,Department of Internal Medicine, member of the DZL, member of CPI, Justus Liebig University, Giessen, Germany.,Institute or Lung Health (ILH), Justus Liebig University, Giessen, Germany
| |
Collapse
|
17
|
Loss of ADAM9 Leads to Modifications of the Extracellular Matrix Modulating Tumor Growth. Biomolecules 2020; 10:biom10091290. [PMID: 32906814 PMCID: PMC7564588 DOI: 10.3390/biom10091290] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/01/2020] [Accepted: 09/03/2020] [Indexed: 12/23/2022] Open
Abstract
ADAM9 is a metalloproteinase strongly expressed at the tumor-stroma border by both tumor and stromal cells. We previously showed that the host deletion of ADAM9 leads to enhanced growth of grafted B16F1 melanoma cells by a mechanism mediated by TIMP1 and the TNF-α/sTNFR1 pathway. This study aimed to dissect the structural modifications in the tumor microenvironment due to the stromal expression of ADAM9 during melanoma progression. We performed proteomic analysis of peritumoral areas of ADAM9 deleted mice and identified the altered expression of several matrix proteins. These include decorin, collagen type XIV, fibronectin, and collagen type I. Analysis of these matrices in the matrix producing cells of the dermis, fibroblasts, showed that ADAM9-/- and wild type fibroblasts synthesize and secreted almost comparable amounts of decorin. Conversely, collagen type I expression was moderately, but not significantly, decreased at the transcriptional level, and the protein increased in ADAM9-/- fibroblast mono- and co-cultures with melanoma media. We show here for the first time that ADAM9 can release a collagen fragment. Still, it is not able to degrade collagen type I. However, the deletion of ADAM9 in fibroblasts resulted in reduced MMP-13 and -14 expression that may account for the reduced processing of collagen type I. Altogether, the data show that the ablation of ADAM9 in the host leads to the altered expression of peritumoral extracellular matrix proteins that generate a more favorable environment for melanoma cell growth. These data underscore the suppressive role of stromal expression of ADAM9 in tumor growth and call for a better understanding of how protease activities function in a cellular context for improved targeting.
Collapse
|
18
|
Crenigacestat, a selective NOTCH1 inhibitor, reduces intrahepatic cholangiocarcinoma progression by blocking VEGFA/DLL4/MMP13 axis. Cell Death Differ 2020; 27:2330-2343. [PMID: 32042099 PMCID: PMC7370218 DOI: 10.1038/s41418-020-0505-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 01/16/2020] [Accepted: 01/21/2020] [Indexed: 02/06/2023] Open
Abstract
Intrahepatic cholangiocarcinoma (iCCA) is a deadly disease with rising incidence and few treatment options. An altered expression and/or activation of NOTCH1–3 receptors has been shown to play a role in iCCA development and progression. In this study, we established a new CCA patient-derived xenograft model, which was validated by immunohistochemistry and transcriptomic analysis. The effects of Notch pathway suppression by the Crenigacestat (LY3039478)-specific inhibitor were evaluated in human iCCA cell lines and the PDX model. In vitro, LY3039478 significantly reduced Notch pathway components, including NICD1 and HES1, but not the other Notch receptors, in a panel of five different iCCA cell lines. In the PDX model, LY3039478 significantly inhibited the Notch pathway and tumor growth to the same extent as gemcitabine. Furthermore, gene expression analysis of iCCA mouse tissues treated with LY3039478 revealed a downregulation of VEGFA, HES1, and MMP13 genes. In the same tissues, DLL4 and CD31 co-localized, and their expression was significantly inhibited in the treated mice, as it happened in the case of MMP13. In an in vitro angiogenesis model, LY3039478 inhibited vessel formation, which was restored by the addition of MMP13. Finally, RNA-sequencing expression data of iCCA patients and matched surrounding normal liver tissues downloaded from the GEO database demonstrated that NOTCH1, HES1, MMP13, DLL4, and VEGFA genes were significantly upregulated in tumors compared with adjacent nontumorous tissues. These data were confirmed by our group, using an independent cohort of iCCA specimens. Conclusion: We have developed and validated a new iCCA PDX model to test in vivo the activity of LY3039478, demonstrating its inhibitory role in Notch-dependent angiogenesis. Thus, the present data provide new knowledge on Notch signaling in iCCA, and support the inhibition of the Notch cascade as a promising strategy for the treatment of this disease.
Collapse
|
19
|
Zamolo G, Grahovac M, Žauhar G, Vučinić D, Kovač L, Brajenić N, Grahovac B. Matrix metalloproteinases MMP-1, MMP-2, and MMP-13 are overexpressed in primary nodular melanoma. J Cutan Pathol 2020; 47:139-145. [PMID: 31677173 DOI: 10.1111/cup.13603] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 10/03/2019] [Accepted: 10/26/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND The spread and invasion of malignant melanoma cells involve degradation and reorganization of the extracellular matrix by the activation of several matrix metalloproteinases (MMPs). This study analyzed the expression of MMP-1, MMP-2, and MMP-13 proteins in primary nodular melanoma (NM) and dysplastic nevi (DN) as a significant risk factor for melanoma development. The secondary goal was to analyze the correlation of MMPs protein expression in NM with tumor invasion, BRAF V600 mutation status, and overall survival. METHODS Immunohistochemistry for MMP-1, MMP-2, and MMP-13 was performed on nodular melanoma (n = 52) and dysplastic nevi (n = 28) on tissue microarray (TMA). BRAF V600 mutation analysis on NM samples was performed by the Sanger sequencing method. RESULTS A high level of MMPs expression in NM samples (>30%) compared with DN (<8%) was statistically significant (P < 0.001). BRAF V600 mutations were detected in 15 of 39 (38.5%) NM samples. This study revealed an interesting finding that MMP-1 and MMP-13 protein expression in the BRAF V600 mutated melanomas were significantly lower than in the BRAF V600 wild type (P < 0.05). CONCLUSION Cox analysis revealed that Clark categories, Breslow thickness, and MMP-1 high protein expression are predictive factors for shorter overall survival (P < 0.05).
Collapse
Affiliation(s)
- Gordana Zamolo
- Department of Pathology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
- Department of Pathology, Clinical Hospital Centre Rijeka, Rijeka, Croatia
| | - Maja Grahovac
- Polyclinic of Dermatology, Gutenbergstr. 8, 87600, Kaufbeuren, Germany
| | - Gordana Žauhar
- Department of Physics, University of Rijeka, Rijeka, Croatia
- Department of Medical Physics and Biophysics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Damir Vučinić
- Department of Radiotherapy and Oncology, Clinical Hospital Centre Rijeka, Rijeka, Croatia
| | - Leo Kovač
- Department of Pathology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
- Department of Pathology, Clinical Hospital Centre Rijeka, Rijeka, Croatia
| | - Nika Brajenić
- The University of Rijeka, Faculty of Medicine, Rijeka, Croatia
| | | |
Collapse
|
20
|
Cho WC, Jour G, Aung PP. Role of angiogenesis in melanoma progression: Update on key angiogenic mechanisms and other associated components. Semin Cancer Biol 2019; 59:175-186. [PMID: 31255774 DOI: 10.1016/j.semcancer.2019.06.015] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 06/05/2019] [Accepted: 06/26/2019] [Indexed: 01/09/2023]
Abstract
Angiogenesis, the formation of new blood vessels from existing blood vessels, is a complex and highly regulated process that plays a role in a wide variety of physiological and pathological processes. In malignancy, angiogenesis is essential for neoplastic cells to acquire the nutrients and oxygen critical for their continued proliferation. Angiogenesis requires a sequence of well-coordinated events mediated by a number of tightly regulated interactions between pro-angiogenic factors and their corresponding receptors expressed on various vascular components (e.g., endothelial cells and pericytes) and stromal components forming the extracellular matrix. In this review, we discuss the functional roles of key growth factors and cytokines known to promote angiogenesis in cutaneous melanoma and key factors implicated in the extracellular matrix remodeling that acts synergistically with angiogenesis to promote tumor progression in melanoma, incorporating some of the most up-to-date basic science knowledge from recently published in vivo and in vitro experimental studies.
Collapse
Affiliation(s)
- Woo Cheal Cho
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - George Jour
- Department of Pathology and Dermatology, NYU Langone Medical Center, New York, NY, USA
| | - Phyu P Aung
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
21
|
Wang FT, Sun W, Zhang JT, Fan YZ. Cancer-associated fibroblast regulation of tumor neo-angiogenesis as a therapeutic target in cancer. Oncol Lett 2019; 17:3055-3065. [PMID: 30867734 PMCID: PMC6396119 DOI: 10.3892/ol.2019.9973] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 12/21/2018] [Indexed: 12/20/2022] Open
Abstract
Adequate blood supply is essential for tumor survival, growth and metastasis. The tumor microenvironment (TME) is dynamic and complex, comprising cancer cells, cancer-associated stromal cells and their extracellular products. The TME serves an important role in tumor progression. Cancer-associated fibroblasts (CAFs) are the principal component of stromal cells within the TME, and contribute to tumor neo-angiogenesis by altering the proteome and degradome. The present paper reviews previous studies of the molecular signaling pathways by which CAFs promote tumor neo-angiogenesis and highlights therapeutic response targets. Also discussed are potential strategies for antitumor neo-angiogenesis to improve tumor treatment efficacy.
Collapse
Affiliation(s)
- Fang-Tao Wang
- Department of Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Wei Sun
- Department of Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Jing-Tao Zhang
- Department of Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Yue-Zu Fan
- Department of Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| |
Collapse
|
22
|
Abstract
Structure-based virtual screening (SBVS) is a common method for the fast identification of hit structures at the beginning of a medicinal chemistry program in drug discovery. The SBVS, described in this manuscript, is focused on finding small molecule hits that can be further utilized as a starting point for the development of inhibitors of matrix metalloproteinase 13 (MMP-13) via structure-based molecular design. We intended to identify a set of structurally diverse hits, which occupy all subsites (S1'-S3', S2, and S3) centering the zinc containing binding site of MMP-13, by the virtual screening of a chemical library comprising more than ten million commercially available compounds. In total, 23 compounds were found as potential MMP-13 inhibitors using Glide docking followed by the analysis of the structural interaction fingerprints (SIFt) of the docked structures.
Collapse
|
23
|
Drug repurposing screening identifies bortezomib and panobinostat as drugs targeting cancer associated fibroblasts (CAFs) by synergistic induction of apoptosis. Invest New Drugs 2018; 36:545-560. [DOI: 10.1007/s10637-017-0547-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 11/22/2017] [Indexed: 02/04/2023]
|
24
|
MMP16 is a marker of poor prognosis in gastric cancer promoting proliferation and invasion. Oncotarget 2018; 7:51865-51874. [PMID: 27340864 PMCID: PMC5239520 DOI: 10.18632/oncotarget.10177] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 06/06/2016] [Indexed: 11/25/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are closely associated with tumor proliferation, invasion and metastasis. In this study, we determined the MMPs expression and their clinical significances in gastric cancer (GC). We first extensive studied MMPs expression in GC in The Cancer Genome Atlas (TCGA) RNA sequence database and found MMP16 was candidate biomarker in GC. Then we validated clinical significance of MMP16 mRNA expression in 167 GC by RT-PCR. Survival analysis showed that high expression of MMP16 indicated poor overall and disease free survival (P<0.001). The proliferation and invasion potential of GC cells were determined by CCK8, colony formation and Transwell assays. Silencing of MMP16 expression significantly decreased the invasion and proliferation capacity of GC cells (P<0.05). In conclusion, MMP16 was highly expressed and correlated with poor prognosis in GC patients by promoting proliferation and invasion of GC cells. MMP16 could be a novel molecular target and prognostic marker for GC.
Collapse
|
25
|
Antsiferova M, Piwko-Czuchra A, Cangkrama M, Wietecha M, Sahin D, Birkner K, Amann VC, Levesque M, Hohl D, Dummer R, Werner S. Activin promotes skin carcinogenesis by attraction and reprogramming of macrophages. EMBO Mol Med 2017; 9:27-45. [PMID: 27932444 PMCID: PMC5210090 DOI: 10.15252/emmm.201606493] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Activin has emerged as an important player in different types of cancer, but the underlying mechanisms are largely unknown. We show here that activin overexpression is an early event in murine and human skin tumorigenesis. This is functionally important, since activin promoted skin tumorigenesis in mice induced by the human papillomavirus 8 oncogenes. This was accompanied by depletion of epidermal γδ T cells and accumulation of regulatory T cells. Most importantly, activin increased the number of skin macrophages via attraction of blood monocytes, which was prevented by depletion of CCR2‐positive monocytes. Gene expression profiling of macrophages from pre‐tumorigenic skin and bioinformatics analysis demonstrated that activin induces a gene expression pattern in skin macrophages that resembles the phenotype of tumor‐associated macrophages in different malignancies, thereby promoting angiogenesis, cell migration and proteolysis. The functional relevance of this finding was demonstrated by antibody‐mediated depletion of macrophages, which strongly suppressed activin‐induced skin tumor formation. These results demonstrate that activin induces skin carcinogenesis via attraction and reprogramming of macrophages and identify novel activin targets involved in tumor formation.
Collapse
Affiliation(s)
- Maria Antsiferova
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | | | - Michael Cangkrama
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Mateusz Wietecha
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Dilara Sahin
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Katharina Birkner
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Valerie C Amann
- Department of Dermatology, University Hospital, Zurich, Switzerland
| | | | - Daniel Hohl
- Department of Dermatology, University of Lausanne, Lausanne, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital, Zurich, Switzerland
| | - Sabine Werner
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
26
|
Karimi L, Mansoori B, shanebandi D, Mohammadi A, Aghapour M, Baradaran B. Function of microRNA-143 in different signal pathways in cancer: New insights into cancer therapy. Biomed Pharmacother 2017; 91:121-131. [DOI: 10.1016/j.biopha.2017.04.060] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/05/2017] [Accepted: 04/13/2017] [Indexed: 01/05/2023] Open
|
27
|
Choi JY, Fuerst R, Knapinska AM, Taylor AB, Smith L, Cao X, Hart PJ, Fields GB, Roush WR. Structure-Based Design and Synthesis of Potent and Selective Matrix Metalloproteinase 13 Inhibitors. J Med Chem 2017; 60:5816-5825. [PMID: 28653849 DOI: 10.1021/acs.jmedchem.7b00514] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We describe the use of comparative structural analysis and structure-guided molecular design to develop potent and selective inhibitors (10d and (S)-17b) of matrix metalloproteinase 13 (MMP-13). We applied a three-step process, starting with a comparative analysis of the X-ray crystallographic structure of compound 5 in complex with MMP-13 with published structures of known MMP-13·inhibitor complexes followed by molecular design and synthesis of potent but nonselective zinc-chelating MMP inhibitors (e.g., 10a and 10b). After demonstrating that the pharmacophores of the chelating inhibitors (S)-10a, (R)-10a, and 10b were binding within the MMP-13 active site, the Zn2+ chelating unit was replaced with nonchelating polar residues that bridged over the Zn2+ binding site and reached into a solvent accessible area. After two rounds of structural optimization, these design approaches led to small molecule MMP-13 inhibitors 10d and (S)-17b, which bind within the substrate-binding site of MMP-13 and surround the catalytically active Zn2+ ion without chelating to the metal. These compounds exhibit at least 500-fold selectivity versus other MMPs.
Collapse
Affiliation(s)
- Jun Yong Choi
- Department of Chemistry, Scripps Florida , 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Rita Fuerst
- Department of Chemistry, Scripps Florida , 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Anna M Knapinska
- Department of Chemistry & Biochemistry, Florida Atlantic University , Jupiter, Florida 33458, United States
| | - Alexander B Taylor
- Department of Biochemistry and Structural Biology and the X-ray Crystallography Core Laboratory, University of Texas Health Science Center at San Antonio , San Antonio, Texas 78229, United States
| | - Lyndsay Smith
- Department of Chemistry & Biochemistry, Florida Atlantic University , Jupiter, Florida 33458, United States
| | - Xiaohang Cao
- Department of Biochemistry and Structural Biology and the X-ray Crystallography Core Laboratory, University of Texas Health Science Center at San Antonio , San Antonio, Texas 78229, United States
| | - P John Hart
- Department of Biochemistry and Structural Biology and the X-ray Crystallography Core Laboratory, University of Texas Health Science Center at San Antonio , San Antonio, Texas 78229, United States
| | - Gregg B Fields
- Department of Chemistry & Biochemistry, Florida Atlantic University , Jupiter, Florida 33458, United States
| | - William R Roush
- Department of Chemistry, Scripps Florida , 130 Scripps Way, Jupiter, Florida 33458, United States
| |
Collapse
|
28
|
Muhsin-Sharafaldine MR, Saunderson SC, Dunn AC, McLellan AD. Melanoma growth and lymph node metastasis is independent of host CD169 expression. Biochem Biophys Res Commun 2017; 486:965-970. [PMID: 28359758 DOI: 10.1016/j.bbrc.2017.03.138] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 03/25/2017] [Indexed: 01/04/2023]
Abstract
Metastasis to the lymph node is a frequent and early event in tumour dissemination. Tumour soluble factors, including extracellular vesicles, condition host organs for metastatic tumour spread, thereby facilitating tumour cell migration and survival. In the peripheral lymphatics, extracellular vesicles are captured via their sialic acids by lymph node macrophages expressing the CD169 (sialoadhesin) molecule, thereby suppressing the immune response. We hypothesised that the CD169 molecule could modulate primary tumour growth and invasion into the regional lymph node by altering the immune response to tumour extracellular vesicles, or by directly interacting with invading tumour cells. No significant difference was noted in primary tumour growth between wild-type and CD169-/- mice, and protection against tumour challenge with tumour extracellular vesicle immunisation was similar between the strains. Subcutaneous implantation of B16 (F1 or F10) into the ventral-carpal aspect of forelimb resulted in melanoma infiltration into the axillary and brachial lymph nodes. CD169-/- mice displayed a lower level of metastatic lymph node lesions, however this failed to reach statistical significance. Although CD169 participates in the immune response to tumour antigen and appears to be a positive prognostic marker for human cancers, its role in modulating melanoma growth and metastasis is less clear.
Collapse
Affiliation(s)
| | - Sarah C Saunderson
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Amy C Dunn
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Alexander D McLellan
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
29
|
Abstract
Metastasis is the main cause of cancer patient mortality. Local tumor invasion is a key step in metastatic dissemination whereby cancer cells dislodge from primary tumors, migrate through the peritumoral stroma and reach the circulation. This is a highly dynamic process occurring in three dimensions that involves interactions between tumor, stromal cells, and the extracellular matrix. Here we describe the organotypic culture system and its utility to study breast cancer cell invasion induced by cancer-associated fibroblasts. This is a three-dimensional model that reproduces the biochemical and physiological properties of real tissue and allows for investigating the molecular and cellular mechanisms involving tumor and its microenvironment, and their contribution to cancer cell invasion . This system provides a robust, accurate, and reproducible method for measuring cancer cell invasion and represents a valuable tool to improve the mechanistic understanding of the initial steps in metastasis .
Collapse
Affiliation(s)
- Romana E. Ranftl
- 0000 0001 1271 4623grid.18886.3fTumour Microenvironment Team, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London, SW2 6JB UK
| | - Fernando Calvo
- 0000 0001 1271 4623grid.18886.3fTumour Microenvironment Team, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London, SW2 6JB UK
| |
Collapse
|
30
|
Kim YH, Jung JC, Gum SI, Park SB, Ma JY, Kim YI, Lee KW, Park YJ. Inhibition of Pterygium Fibroblast Migration and Outgrowth by Bevacizumab and Cyclosporine A Involves Down-Regulation of Matrix Metalloproteinases-3 and -13. PLoS One 2017; 12:e0169675. [PMID: 28068383 PMCID: PMC5221804 DOI: 10.1371/journal.pone.0169675] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Accepted: 12/20/2016] [Indexed: 02/05/2023] Open
Abstract
We examined the connection between matrix metalloproteinase (MMP) expression/activity and pterygium fibroblast migration, and how these were affected by bevacizumab and/or cyclosporine A (CsA). Fibroblasts were obtained from 20 pterygia and 6 normal conjunctival specimens. Expression levels of MMP-3 and MMP-13 were examined after bevacizumab administration. Immunofluorescence staining was used to examine expression of both MMPs in fibroblasts migrating out from explanted pterygium tissues. Rates of cell migration from explant-cultured pterygia tissues and scratch-wounded confluent pterygium fibroblasts were examined in the presence of MMP-3 or MMP-13 inhibitors, as well as bevacizumab and/or CsA. A scratch wound healing migration assay was performed to determine the effects of bevacizumab and/or CsA. Protein expression of both MMPs in pterygium tissues and in cells migrating from organ-cultured pterygium tissues was greater than that observed in normal cells. Inhibition of the activities of both MMPs decreased their expression levels; these were also significantly reduced in bevacizumab-injected pterygium tissues. Bevacizumab significantly reduced the expression of both MMPs and cell migration. Pretreatment with CsA prior to bevacizumab exposure markedly inhibited cell migration and the expression of both MMPs. CsA enhanced the inhibitory effects of bevacizumab on pterygium fibroblast migration in vitro, possibly by inhibiting expression of both MMPs. These findings suggest that combined CsA and bevacizumab treatment may provide a potential therapeutic strategy for reducing the rate of pterygium recurrence.
Collapse
Affiliation(s)
- Yeoun-Hee Kim
- Cheil Eye Research Institute, Cheil Eye Hospital, 1 Ayang-Ro, Dong-Gu, Daegu, Republic of Korea
- Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine (KIOM), Cheomdan-ro Dong-gu, Daegu, Republic of Korea
| | - Jae-Chang Jung
- Department of Biology, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Sang Il Gum
- Cheil Eye Research Institute, Cheil Eye Hospital, 1 Ayang-Ro, Dong-Gu, Daegu, Republic of Korea
| | - Su-Bin Park
- Department of Biology, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Jin Yeul Ma
- Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine (KIOM), Cheomdan-ro Dong-gu, Daegu, Republic of Korea
| | - Yong Il Kim
- Cheil Eye Research Institute, Cheil Eye Hospital, 1 Ayang-Ro, Dong-Gu, Daegu, Republic of Korea
| | - Kyoo Won Lee
- Cheil Eye Research Institute, Cheil Eye Hospital, 1 Ayang-Ro, Dong-Gu, Daegu, Republic of Korea
| | - Young Jeung Park
- Cheil Eye Research Institute, Cheil Eye Hospital, 1 Ayang-Ro, Dong-Gu, Daegu, Republic of Korea
- * E-mail:
| |
Collapse
|
31
|
Ghatak S, Niland S, Schulz JN, Wang F, Eble JA, Leitges M, Mauch C, Krieg T, Zigrino P, Eckes B. Role of Integrins α1β1 and α2β1 in Wound and Tumor Angiogenesis in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:3011-3027. [DOI: 10.1016/j.ajpath.2016.06.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 06/01/2016] [Accepted: 06/30/2016] [Indexed: 12/11/2022]
|
32
|
Donizy P, Kaczorowski M, Biecek P, Halon A, Szkudlarek T, Matkowski R. Golgi-Related Proteins GOLPH2 (GP73/GOLM1) and GOLPH3 (GOPP1/MIDAS) in Cutaneous Melanoma: Patterns of Expression and Prognostic Significance. Int J Mol Sci 2016; 17:E1619. [PMID: 27706081 PMCID: PMC5085652 DOI: 10.3390/ijms17101619] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Revised: 09/05/2016] [Accepted: 09/12/2016] [Indexed: 02/06/2023] Open
Abstract
GOLPH2 and GOLPH3 are Golgi-related proteins associated with aggressiveness and progression of a number of cancers. Their prognostic significance in melanoma has not yet been analyzed. We performed immunohistochemical analysis for GOLPH2 and GOLPH3 in 20 normal skin, 30 benign nevi and 100 primary melanoma tissue samples and evaluated their expression in three compartments: cancer cells, tumor-associated macrophages (TAMs) and cancer-associated fibroblasts (CAFs). High levels of both proteins in melanoma cells were associated with characteristics of aggressive disease, and shorter disease-free survival (DFS) and cancer-specific overall survival (CSOS). On the contrary, increased numbers of GOLPH2-positive and GOLPH3-positive TAMs were observed in thinner, non-ulcerated tumors, with brisk lymphocytic reaction and absent lymphangioinvasion. Distant metastases were not observed among patients with high numbers of GOLPH2-positive TAMs. Increased expression of either protein in TAMs was related to prolonged CSOS and DFS. Similarly, GOLPH3-expressing CAFs were more frequent in thin melanomas with low mitotic rate, without ulceration and lymphangioinvasion. Moreover, increased GOLPH3-positive CAFs correlated with the absence of regional or distant metastases, and with longer CSOS and DFS. GOLPH2 expression was not observed in CAFs. Our results suggest that GOLPH2 and GOLPH3 play a role in melanoma progression and are potential targets for molecular-based therapies.
Collapse
Affiliation(s)
- Piotr Donizy
- Department of Pathomorphology and Oncological Cytology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland.
| | - Maciej Kaczorowski
- Department of Pathomorphology and Oncological Cytology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland.
| | - Przemyslaw Biecek
- Faculty of Mathematics and Information Science, Warsaw University of Technology, Koszykowa 75, 00-662 Warsaw, Poland.
| | - Agnieszka Halon
- Department of Pathomorphology and Oncological Cytology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland.
| | - Teresa Szkudlarek
- Department of Pathomorphology and Oncological Cytology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland.
| | - Rafal Matkowski
- Department of Oncology, Wroclaw Medical University; pl. Hirszfelda 12, 53-413 Wroclaw, Poland.
- Lower Silesian Cancer Center, Hirszfelda 12, 53-413 Wroclaw, Poland.
| |
Collapse
|
33
|
Kamenisch Y, Baban TSA, Schuller W, von Thaler AK, Sinnberg T, Metzler G, Bauer J, Schittek B, Garbe C, Rocken M, Berneburg M. UVA-Irradiation Induces Melanoma Invasion via the Enhanced Warburg Effect. J Invest Dermatol 2016; 136:1866-1875. [PMID: 27185340 DOI: 10.1016/j.jid.2016.02.815] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 01/20/2016] [Accepted: 02/14/2016] [Indexed: 10/21/2022]
Abstract
Melanoma is a malignant tumor in which UVA (320-400 nm) radiation is considered to be an important risk factor. But the role of UVA in melanoma progression toward an invasive phenotype is still not adequately investigated. For most proliferating tumor cells the preference of aerobic glycolysis has been described as the Warburg effect. Here we investigate the effect of UVA irradiation on changes in the Warburg effect and tumor progression toward invasive potential. On UVA irradiation, melanoma cell lines from initial tumors show an induction of the Warburg effect with increased glucose consumption and lactate production, which is at least partially mediated by reactive oxygen species. Associated with UVA treatment and enhanced lactic acid production, tumor-relevant proteases and in situ invasion is upregulated. Simultaneously, UVA increases intracellular concentrations of progression marker transketolase and activated protein kinase Akt, both involved in metabolic changes that increase with proliferation. Using invasion assays we show that lactic acid, resulting from the UVA enhanced and partially reactive oxygen species-mediated Warburg effect, increases the invasive potential of all melanoma cell lines investigated. Therefore, we demonstrate in melanoma cells that production of lactic acid, induced by UVA irradiation, increases invasiveness of melanoma cells via expression of tumor-relevant proteases.
Collapse
Affiliation(s)
- York Kamenisch
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany; Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Tarza S A Baban
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Winfried Schuller
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | | | - Tobias Sinnberg
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Gisela Metzler
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Jürgen Bauer
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Birgit Schittek
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Claus Garbe
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Martin Rocken
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Mark Berneburg
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany.
| |
Collapse
|
34
|
Fu J, Li S, Feng R, Ma H, Sabeh F, Roodman GD, Wang J, Robinson S, Guo XE, Lund T, Normolle D, Mapara MY, Weiss SJ, Lentzsch S. Multiple myeloma-derived MMP-13 mediates osteoclast fusogenesis and osteolytic disease. J Clin Invest 2016; 126:1759-72. [PMID: 27043283 DOI: 10.1172/jci80276] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 02/18/2016] [Indexed: 12/27/2022] Open
Abstract
Multiple myeloma (MM) cells secrete osteoclastogenic factors that promote osteolytic lesions; however, the identity of these factors is largely unknown. Here, we performed a screen of human myeloma cells to identify pro-osteoclastogenic agents that could potentially serve as therapeutic targets for ameliorating MM-associated bone disease. We found that myeloma cells express high levels of the matrix metalloproteinase MMP-13 and determined that MMP-13 directly enhances osteoclast multinucleation and bone-resorptive activity by triggering upregulation of the cell fusogen DC-STAMP. Moreover, this effect was independent of the proteolytic activity of the enzyme. Further, in mouse xenograft models, silencing MMP-13 expression in myeloma cells inhibited the development of osteolytic lesions. In patient cohorts, MMP-13 expression was localized to BM-associated myeloma cells, while elevated MMP-13 serum levels were able to correctly predict the presence of active bone disease. Together, these data demonstrate that MMP-13 is critical for the development of osteolytic lesions in MM and that targeting the MMP-13 protein - rather than its catalytic activity - constitutes a potential approach to mitigating bone disease in affected patients.
Collapse
|
35
|
Zhao X, Sun B, Li Y, Liu Y, Zhang D, Wang X, Gu Q, Zhao J, Dong X, Liu Z, Che N. Dual effects of collagenase-3 on melanoma: metastasis promotion and disruption of vasculogenic mimicry. Oncotarget 2016; 6:8890-9. [PMID: 25749207 PMCID: PMC4496190 DOI: 10.18632/oncotarget.3189] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Accepted: 01/23/2015] [Indexed: 12/18/2022] Open
Abstract
Vasculogenic mimicry (VM) is a functional microcirculation formed by tumor cells. Matrix metalloproteinases (MMPs), especially MMP-2 and MMP-9, promote VM formation. Another specific MMP, collagenase-3 (MMP-13), has broad substrate specificity and potentially affects tumor metastasis and invasion. Here we found that MMP-13 was associated with metastasis and poor survival in 79 patients with melanoma. MMP-13 expression was inversely correlated with VM. These results were confirmed in human and mouse melanoma cell lines. We found that MMP-13 cleaves laminin-5 (Ln-5) into small fragments to accelerate tumor metastasis. Degradation of Ln-5 and VE-cadherin by MMP-13 inhibited VM formation. In conclusion, MMP-13 has a dual effect in melanoma, as it promotes invasion and metastasis but disrupts VM formation.
Collapse
Affiliation(s)
- Xiulan Zhao
- Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China.,Department of Pathology, Tianjin Medical University, Tianjin, China
| | - Baocun Sun
- Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China.,Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, Tianjin Cancer Hospital, Tianjin Medical University, Tianjin, China
| | - Yanlei Li
- Department of Pathology, Tianjin Medical University, Tianjin, China
| | - Yanrong Liu
- Department of Pathology, Tianjin Medical University, Tianjin, China
| | - Danfang Zhang
- Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China.,Department of Pathology, Tianjin Medical University, Tianjin, China
| | - Xudong Wang
- Department of Pathology, Tianjin Cancer Hospital, Tianjin Medical University, Tianjin, China
| | - Qiang Gu
- Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China.,Department of Pathology, Tianjin Medical University, Tianjin, China
| | - Jianmin Zhao
- Department of Pathology, Tianjin Medical University, Tianjin, China
| | - Xueyi Dong
- Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China.,Department of Pathology, Tianjin Medical University, Tianjin, China
| | - Zhiyong Liu
- Department of Pathology, Tianjin Cancer Hospital, Tianjin Medical University, Tianjin, China
| | - Na Che
- Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China.,Department of Pathology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
36
|
Huang SH, Law CH, Kuo PH, Hu RY, Yang CC, Chung TW, Li JM, Lin LH, Liu YC, Liao EC, Tsai YT, Wei YS, Lin CC, Chang CW, Chou HC, Wang WC, Chang MDT, Wang LH, Kung HJ, Chan HL, Lyu PC. MMP-13 is involved in oral cancer cell metastasis. Oncotarget 2016; 7:17144-61. [PMID: 26958809 PMCID: PMC4941377 DOI: 10.18632/oncotarget.7942] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 12/31/1969] [Indexed: 12/20/2022] Open
Abstract
The oral cancer cell line OC3-I5 with a highly invasive ability was selected and derived from an established OSCC line OC3. In this study, we demonstrated that matrix metalloproteinases protein MMP-13 was up-regulated in OC3-I5 than in OC3 cells. We also observed that expression of epithelial-mesenchymal transition (EMT) markers including Twist, p-Src, Snail1, SIP1, JAM-A, and vinculin were increased in OC3-I5 compared to OC3 cells, whereas E-cadherin expression was decreased in the OC3-I5 cells. Using siMMP-13 knockdown techniques, we showed that siMMP-13 not only reduced the invasion and migration, but also the adhesion abilities of oral cancer cells. In support of the role of MMP-13 in metastasis, we used MMP-13 expressing plasmid-transfected 293T cells to enhance MMP-13 expression in the OC3 cells, transplanting the MMP-13 over expressing OC3 cells into nude mice led to enhanced lung metastasis. In summary, our findings show that MMP-13 promotes invasion and metastasis in oral cancer cells, suggesting altered expression of MMP-13 may be utilized to impede the process of metastasis.
Collapse
Affiliation(s)
- Shun-Hong Huang
- Department of Medical Sciences and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Ching-Hsuan Law
- Department of Medical Sciences and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Ping-Hsueh Kuo
- Department of Medical Sciences and Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Ren-Yu Hu
- Department of Medical Sciences and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Ching-Chieh Yang
- Department of Radiation Oncology, Chi-Mei Medical Center, Tainan, Taiwan.,Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Ting-Wen Chung
- Department of Medical Sciences and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Ji-Min Li
- Department of Medical Sciences and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Li-Hsun Lin
- Department of Medical Sciences and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Chung Liu
- Department of Medical Sciences and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan.,Institute of Population Sciences, National Health Research Institutes, Miaoli County, Taiwan
| | - En-Chi Liao
- Department of Medical Sciences and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Ting Tsai
- Department of Medical Sciences and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Yu-Shan Wei
- Department of Medical Sciences and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Chi-Chen Lin
- Institute of Biomedical Science, National Chung Hsing University, Taichung, Taiwan.,Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan.,Department of Medical Research and Education, Taichung Veterans General Hospital, Taichung, Taiwan.,Division of Chest Medicine. Department of Internal Medicine, Changhua Christian Hospital, Changhua, Taiwan
| | - Chien-Wen Chang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Taiwan
| | - Hsiu-Chuan Chou
- Department of Applied Science, National Hsinchu University of Education, Hsinchu, Taiwan
| | - Wen-Ching Wang
- Department of Medical Sciences and Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Margaret Dah-Tsyr Chang
- Department of Medical Sciences and Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Lu-Hai Wang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli County, Taiwan
| | - Hsing-Jien Kung
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli County, Taiwan
| | - Hong-Lin Chan
- Department of Medical Sciences and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan.,Department of Medical Sciences and Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Ping-Chiang Lyu
- Department of Medical Sciences and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan.,Department of Medical Sciences and Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
37
|
Fischer T, Riedl R. Molecular Recognition of the Catalytic Zinc(II) Ion in MMP-13: Structure-Based Evolution of an Allosteric Inhibitor to Dual Binding Mode Inhibitors with Improved Lipophilic Ligand Efficiencies. Int J Mol Sci 2016; 17:314. [PMID: 26938528 PMCID: PMC4813177 DOI: 10.3390/ijms17030314] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 02/02/2016] [Accepted: 02/14/2016] [Indexed: 11/21/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are a class of zinc dependent endopeptidases which play a crucial role in a multitude of severe diseases such as cancer and osteoarthritis. We employed MMP-13 as the target enzyme for the structure-based design and synthesis of inhibitors able to recognize the catalytic zinc ion in addition to an allosteric binding site in order to increase the affinity of the ligand. Guided by molecular modeling, we optimized an initial allosteric inhibitor by addition of linker fragments and weak zinc binders for recognition of the catalytic center. Furthermore we improved the lipophilic ligand efficiency (LLE) of the initial inhibitor by adding appropriate zinc binding fragments to lower the clogP values of the inhibitors, while maintaining their potency. All synthesized inhibitors showed elevated affinity compared to the initial hit, also most of the novel inhibitors displayed better LLE. Derivatives with carboxylic acids as the zinc binding fragments turned out to be the most potent inhibitors (compound 3 (ZHAWOC5077): IC50 = 134 nM) whereas acyl sulfonamides showed the best lipophilic ligand efficiencies (compound 18 (ZHAWOC5135): LLE = 2.91).
Collapse
Affiliation(s)
- Thomas Fischer
- Center for Organic and Medicinal Chemistry, Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences ZHAW, Einsiedlerstrasse 31, 8820 Wädenswil, Switzerland.
| | - Rainer Riedl
- Center for Organic and Medicinal Chemistry, Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences ZHAW, Einsiedlerstrasse 31, 8820 Wädenswil, Switzerland.
| |
Collapse
|
38
|
Matrix metalloproteinases: new directions toward inhibition in the fight against cancers. Future Med Chem 2016; 8:297-309. [PMID: 26910530 DOI: 10.4155/fmc.15.184] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Matrix metalloproteinases are zinc-dependent enzymes whose main function is to cleave the components of the extracellular matrix. Their overexpression is evident in all cancers but to date there is no satisfactory way to inhibit their actions. Here, we look at their types, their structures, their functions and the developing understanding we have of them in the search for ways to drug them and inhibit their actions selectively. We investigate their subtle but exploitable differences in order that we can develop drugs to target them and even to target specific substrates and functions that they carry out. To date there are no new matrix metalloproteinase inhibitors developed to treat cancer, but we are progressing in our understanding of them, which is leading us ever closer to our goal.
Collapse
|
39
|
Chen L, Yang H, Xiao Y, Tang X, Li Y, Han Q, Fu J, Yang Y, Zhu Y. Lentiviral-mediated overexpression of long non-coding RNA GAS5 reduces invasion by mediating MMP2 expression and activity in human melanoma cells. Int J Oncol 2016; 48:1509-18. [PMID: 26846479 DOI: 10.3892/ijo.2016.3377] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 01/07/2016] [Indexed: 11/05/2022] Open
Abstract
The present study evaluated the effects of long non-coding RNA GAS5 on the migration and invasion of melanoma cells. Using the SK-Mel‑110 melanoma cell line, we stably expressed GAS5, visualized the distribution of GAS5 by RNA fluorescence in situ hybridization (FISH) and examined changes in cell migration and invasion with Transwell assays. In GAS5 overexpressed SK-Mel‑110 cells, migrated and invaded cells decreased by 65.3 and 55.6%, respectively. Moreover, the MMP2 protein level, and its activity was downregulated by 67.9 and 15.8%, respectively. Overexpressing lncRNA GAS5 inhibited the migration and invasion ability of melanoma SK-Mel‑110 cells, partially by decreasing the MMP2 expression and its activity. This study is the first to reveal a potential relationship between lncRNA GAS5 and the migration and invasion of melanoma.
Collapse
Affiliation(s)
- Long Chen
- Department of Biochemistry and Molecular Biology of Kunming Medical University, Yunnan, P.R. China
| | - Huixin Yang
- Department of Biochemistry and Molecular Biology of Kunming Medical University, Yunnan, P.R. China
| | - Yanbin Xiao
- Department of Orthopaedic Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan, P.R. China
| | - Xiaoxia Tang
- Department of Pharmacy, The Second Affiliated Hospital of Kunming Medical University, Yunnan, P.R. China
| | - Yuqian Li
- Department of Biochemistry and Molecular Biology of Kunming Medical University, Yunnan, P.R. China
| | - Qiaoqiao Han
- Department of Biochemistry and Molecular Biology of Kunming Medical University, Yunnan, P.R. China
| | - Junping Fu
- Department of Orthopaedic Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan, P.R. China
| | - Yuye Yang
- Department of Biochemistry and Molecular Biology of Kunming Medical University, Yunnan, P.R. China
| | - Yuechun Zhu
- Department of Biochemistry and Molecular Biology of Kunming Medical University, Yunnan, P.R. China
| |
Collapse
|
40
|
Disruption of Anti-tumor T Cell Responses by Cancer-Associated Fibroblasts. RESISTANCE TO TARGETED ANTI-CANCER THERAPEUTICS 2016. [DOI: 10.1007/978-3-319-42223-7_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
41
|
Jiang YN, Yan HQ, Huang XB, Wang YN, Li Q, Gao FG. Interleukin 6 trigged ataxia-telangiectasia mutated activation facilitates lung cancer metastasis via MMP-3/MMP-13 up-regulation. Oncotarget 2015; 6:40719-33. [PMID: 26528698 PMCID: PMC4747364 DOI: 10.18632/oncotarget.5825] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 09/23/2015] [Indexed: 12/24/2022] Open
Abstract
Our previous studies show that the phosphorylation of ataxia-telangiectasia mutated (ATM) induced by interleukin 6 (IL-6) treatment contributes to multidrug resistance formation in lung cancer cells, but the exact role of ATM activation in IL-6 increased metastasis is still elusive. In the present study, matrix metalloproteinase-3 (MMP-3) and MMP-13 were firstly demonstrated to be involved in IL-6 correlated cell migration. Secondly, IL-6 treatment not only increased MMP-3/MMP-13 expression but also augmented its activities. Thirdly, the inhibition of ATM phosphorylation efficiently abolished IL-6 up-regulating MMP-3/MMP-13 expression and increasing abilities of cell migration. Most importantly, the in vivo test showed that the inhibition of ATM abrogate the effect of IL-6 on lung cancer metastasis via MMP-3/MMP-13 down-regulation. Taken together, these findings demonstrate that IL-6 inducing ATM phosphorylation increases the expression of MMP-3/MMP-13, augments the abilities of cell migration, and promotes lung cancer metastasis, indicating that ATM is a potential target molecule to overcome IL-6 correlated lung cancer metastasis.
Collapse
Affiliation(s)
- Yi Na Jiang
- Department of Immunology, Basic Medicine Science, Medical College, Xiamen University, Xiamen 361005, People's Republic of China
| | - Hong Qiong Yan
- Department of Immunology, Basic Medicine Science, Medical College, Xiamen University, Xiamen 361005, People's Republic of China
| | - Xiao Bo Huang
- Department of Immunology, Basic Medicine Science, Medical College, Xiamen University, Xiamen 361005, People's Republic of China
| | - Yi Nan Wang
- Department of Immunology, Basic Medicine Science, Medical College, Xiamen University, Xiamen 361005, People's Republic of China
| | - Qing Li
- Department of Immunology, Basic Medicine Science, Medical College, Xiamen University, Xiamen 361005, People's Republic of China
| | - Feng Guang Gao
- Department of Immunology, Basic Medicine Science, Medical College, Xiamen University, Xiamen 361005, People's Republic of China
- State Key Laboratory of Oncogenes and Related Genes, Shang Hai Jiao Tong University, Shanghai 200032, People's Republic of China
| |
Collapse
|
42
|
Chowdhury R, Webber JP, Gurney M, Mason MD, Tabi Z, Clayton A. Cancer exosomes trigger mesenchymal stem cell differentiation into pro-angiogenic and pro-invasive myofibroblasts. Oncotarget 2015; 6:715-31. [PMID: 25596732 PMCID: PMC4359250 DOI: 10.18632/oncotarget.2711] [Citation(s) in RCA: 200] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 11/11/2014] [Indexed: 12/25/2022] Open
Abstract
Stromal fibroblasts become altered in response to solid cancers, to exhibit myofibroblastic characteristics, with disease promoting influence. Infiltrating mesenchymal stem cells (MSC) may contribute towards these changes, but the factors secreted by cancer cells that impact MSC differentiation are poorly understood. We investigated the role of nano-metre sized vesicles (exosomes), secreted by prostate cancer cells, on the differentiation of bone-marrow MSC (BM-MSC), and the subsequent functional consequences of such changes. Purified exosomes impaired classical adipogenic differentiation, skewing differentiation towards alpha-smooth muscle actin (αSMA) positive myofibroblastic cells. A single exosomes treatment generated myofibroblasts secreting high levels of VEGF-A, HGF and matrix regulating factors (MMP-1, −3 and −13). Differentiated MSC had pro-angiogenic functions and enhanced tumour proliferation and invasivity assessed in a 3D co-culture model. Differentiation was dependent on exosomal-TGFβ, but soluble TGFβ at matched dose could not generate the same phenotype. Exosomes present in the cancer cell secretome were the principal factors driving this phenotype. Prostate cancer exosomes dominantly dictate a programme of MSC differentiation generating myofibroblasts with functional properties consistent with disease promotion.
Collapse
Affiliation(s)
- Ridwana Chowdhury
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Velindre Cancer Centre, Whitchurch, Cardiff, United Kingdom, CF14 2TL
| | - Jason P Webber
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Velindre Cancer Centre, Whitchurch, Cardiff, United Kingdom, CF14 2TL.,Cardiff Institute for Tissue Engineering and Repair, Cardiff University
| | - Mark Gurney
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Velindre Cancer Centre, Whitchurch, Cardiff, United Kingdom, CF14 2TL
| | - Malcolm D Mason
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Velindre Cancer Centre, Whitchurch, Cardiff, United Kingdom, CF14 2TL
| | - Zsuzsanna Tabi
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Velindre Cancer Centre, Whitchurch, Cardiff, United Kingdom, CF14 2TL
| | - Aled Clayton
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Velindre Cancer Centre, Whitchurch, Cardiff, United Kingdom, CF14 2TL.Cardiff Institute for Tissue Engineering and Repair, Cardiff University.,Institute of Cancer and Genetics, School of Medicine, Cardiff University, Velindre Cancer Centre, Whitchurch, Cardiff, United Kingdom, CF14 2TL.Cardiff Institute for Tissue Engineering and Repair, Cardiff University
| |
Collapse
|
43
|
Sun Z, Cao B, Wu J. Protease-activated receptor 2 enhances renal cell carcinoma cell invasion and migration via PI3K/AKT signaling pathway. Exp Mol Pathol 2015; 98:382-9. [DOI: 10.1016/j.yexmp.2015.03.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Revised: 03/10/2015] [Accepted: 03/11/2015] [Indexed: 01/22/2023]
|
44
|
Jenkins MH, Croteau W, Mullins DW, Brinckerhoff CE. The BRAF(V600E) inhibitor, PLX4032, increases type I collagen synthesis in melanoma cells. Matrix Biol 2015; 48:66-77. [PMID: 25989506 PMCID: PMC5048745 DOI: 10.1016/j.matbio.2015.05.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 05/11/2015] [Accepted: 05/12/2015] [Indexed: 01/08/2023]
Abstract
Vertical growth phase (VGP) melanoma is frequently metastatic, a process mediated by changes in gene expression, which are directed by signal transduction pathways in the tumor cells. A prominent signaling pathway is the Ras-Raf-Mek-Erk MAPK pathway, which increases expression of genes that promote melanoma progression. Many melanomas harbor a mutation in this pathway, BRAF(V600E), which constitutively activates MAPK signaling and expression of downstream target genes that facilitate tumor progression. In BRAF(V600E) melanoma, the small molecule inhibitor, vemurafenib (PLX4032), has revolutionized therapy for melanoma by inducing rapid tumor regression. This compound down-regulates the expression of many genes. However, in this study, we document that blocking the Ras-Raf-Mek-Erk MAPK pathway, either with an ERK (PLX4032) or a MEK (U1026) signaling inhibitor, in BRAF(V600E) human and murine melanoma cell lines increases collagen synthesis in vitro and collagen deposition in vivo. Since TGFß signaling is a major mediator of collagen synthesis, we examined whether blocking TGFß signaling with a small molecule inhibitor would block this increase in collagen. However, there was minimal reduction in collagen synthesis in response to blocking TGFß signaling, suggesting additional mechanism(s), which may include activation of the p38 MAPK pathway. Presently, it is unclear whether this increased collagen synthesis and deposition in melanomas represent a therapeutic benefit or an unwanted "off target" effect of inhibiting the Ras-Raf-Erk-Mek pathway.
Collapse
Affiliation(s)
- Molly H Jenkins
- Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, United States; Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, United States.
| | - Walburga Croteau
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, United States
| | - David W Mullins
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, United States
| | - Constance E Brinckerhoff
- Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, United States; Department of Biochemistry, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, United States
| |
Collapse
|
45
|
Mendonsa AM, VanSaun MN, Ustione A, Piston DW, Fingleton BM, Gorden DL. Host and tumor derived MMP13 regulate extravasation and establishment of colorectal metastases in the liver. Mol Cancer 2015; 14:49. [PMID: 25880591 PMCID: PMC4351934 DOI: 10.1186/s12943-014-0282-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 12/22/2014] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Non alcoholic fatty liver disease (NAFLD) is one of the most common liver diseases in the United States and worldwide. Our studies have previously shown an increase in metastatic burden in steatotic vs. normal livers using a mouse model of diet induced steatosis. In the present study we aim to identify and evaluate the molecular factors responsible for this increase in tumor burden. METHODS We assessed changes in expression of a panel of matrix metalloproteinases (MMPs) using qRT-PCR between normal and steatotic livers and validated them with western blot analysis of protein levels. To evaluate the role of MMP13 on tumor development, we utilized a splenic injection model of liver metastasis in Wildtype and Mmp13 deficient mice, using either parental or stable Mmp13 knockdown cell lines. Further, to evaluate changes in the ability of tumor cells to extravasate we utilized whole organ confocal microscopy to identify individual tumor cells relative to the vasculature. MTT, migration and invasion assays were performed to evaluate the role of tumor derived MMP13 on hallmarks of cancer in vitro. RESULTS We found that MMP13 was significantly upregulated in the steatotic liver both in mice as well as human patients with NAFLD. We showed a decrease in metastatic tumor burden in Mmp13-/- mice compared to wildtype mice, explained in part by a reduction in the number of tumor cells extravasating from the hepatic vasculature in the Mmp13-/- mice compared to wildtype mice. Additionally, loss of tumor derived MMP13 through stable knockdown in tumor cell lines lead to decreased migratory and invasive properties in vitro and metastatic burden in vivo. CONCLUSIONS This study demonstrates that stromal as well as tumor derived MMP13 contribute to tumor cell extravasation and establishment of metastases in the liver microenvironment.
Collapse
Affiliation(s)
- Alisha M Mendonsa
- Department of Cancer Biology, Vanderbilt University, 2220 Pierce Ave S, Nashville, TN, 37232, USA.
| | - Michael N VanSaun
- Department of Cancer Biology, Vanderbilt University, 2220 Pierce Ave S, Nashville, TN, 37232, USA. .,Department of Surgery, Vanderbilt University, 801 Oxford House, 1313 21st Ave. S, Nashville, TN, 37212, USA.
| | - Alessandro Ustione
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 702 Light Hall 21st Avenue South, Nashville, TN, 37232, USA.
| | - David W Piston
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 702 Light Hall 21st Avenue South, Nashville, TN, 37232, USA.
| | - Barbara M Fingleton
- Department of Cancer Biology, Vanderbilt University, 2220 Pierce Ave S, Nashville, TN, 37232, USA.
| | - David Lee Gorden
- Department of Cancer Biology, Vanderbilt University, 2220 Pierce Ave S, Nashville, TN, 37232, USA. .,Department of Surgery, Vanderbilt University, 801 Oxford House, 1313 21st Ave. S, Nashville, TN, 37212, USA.
| |
Collapse
|
46
|
Yang SW, Lim L, Ju S, Choi DH, Song H. Effects of matrix metalloproteinase 13 on vascular smooth muscle cells migration via Akt–ERK dependent pathway. Tissue Cell 2015; 47:115-21. [DOI: 10.1016/j.tice.2014.12.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 12/12/2014] [Accepted: 12/13/2014] [Indexed: 02/08/2023]
|
47
|
Burghoff S, Gong X, Viethen C, Jacoby C, Flögel U, Bongardt S, Schorr A, Hippe A, Homey B, Schrader J. Growth and metastasis of B16-F10 melanoma cells is not critically dependent on host CD73 expression in mice. BMC Cancer 2014; 14:898. [PMID: 25465225 PMCID: PMC4265456 DOI: 10.1186/1471-2407-14-898] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 11/27/2014] [Indexed: 12/03/2022] Open
Abstract
Background Recent studies have suggested that adenosine generated by ecto-5′-nucleotidase (CD73) in the tumor microenvironment plays a major role in promoting tumor growth by suppressing the immune response and stimulating angiogenesis via A2A and A2B receptors. However, adenosine has also been reported to inhibit tumor growth acting via A1 and A3 receptors. Therefore the aim of this study was to clarify the role of host CD73, which catalyzes the extracellular hydrolysis of AMP to adenosine, on tumor growth and metastasis of B16-F10 melanoma cells. Methods CD73 and alkaline phosphatase (AP) activity of B16-F10 melanoma cells were measured by HPLC. Tumor cells were injected either subcutaneously or intradermally in WT and CD73−/− mice and tumor growth was monitored by MRI at 9.4 T. Immune cell subpopulations within tumors were assessed by FACS after enzymatic digestion. An endothelium specific CD73−/− was created using Tie2-Cre+ mice and CD73flox/flox (loxP) mice. Chimeric mice lacking CD73−/− on hematopoietic cells was generated by bone marrow transplantation. Lung metastatic spread was measured after intravenous B16-F10 application. Results B16-F10 cells showed very little CD73 and negligible AP activity. Neither complete loss of host CD73 nor specific knockout of CD73 on endothelial cells or hematopoietic cells affected tumor growth after subcutaneous or intradermal tumor cell application. Only peritumoral edema formation was significantly attenuated in global CD73−/− mice in the intradermal model. Immune cell composition revealed no differences in the different transgenic mice models. Also lung metastasis after intravenous B16-F10 injection was not altered in CD73−/− mice. Conclusions CD73 expression on host cells, particularly on endothelial and hematopoietic cells, does not modulate tumor growth and metastatic spread of B16-F10 melanoma cells most likely because of insufficient adenosine formation by the tumor itself. Electronic supplementary material The online version of this article (doi:10.1186/1471-2407-14-898) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Jürgen Schrader
- Institute of Molecular Cardiology, Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany.
| |
Collapse
|
48
|
Foley CJ, Kuliopulos A. Mouse matrix metalloprotease-1a (Mmp1a) gives new insight into MMP function. J Cell Physiol 2014; 229:1875-80. [PMID: 24737602 DOI: 10.1002/jcp.24650] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 04/14/2014] [Indexed: 01/18/2023]
Abstract
Matrix metalloprotease-1 (MMP1) has been implicated in many human disease processes, however the lack of a well characterized murine homologue has significantly limited the study of MMP1 and the development of MMP-targeted therapeutics. The discovery of murine Mmp1a in 2001, the functional mouse homologue of MMP1, offers a valuable tool for modeling MMP1-mediated processes in mice. Variation in physiologic expression levels of Mmp1a in mice as compared to MMP1 in humans highlights the importance of understanding the similarities and differences between the homologues. Recent studies have demonstrated tumor growth-, invasion-, and angiogenesis-promoting functions of Mmp1a in lung cancer models, consistent with the analogous functions observed for human MMP1. Biochemical investigations have shown that point mutations in the pro-domain of mouse Mmp1a weaken docking between the pro- and catalytic domains, generating an unstable zymogen primed for activation. The difficulty to effectively maintain Mmp1a in the zymogen form may account for the tight control of Mmp1a expression and reduced expression in normal tissue as compared to inflammatory states or cancer. This discovery raises important questions about the activation mechanisms and regulation of the MMP family in general.
Collapse
Affiliation(s)
- Caitlin J Foley
- Molecular Oncology Research Institute, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts; Program in Genetics, Tufts University School of Medicine, Boston, Massachusetts
| | | |
Collapse
|
49
|
Abstract
Dr. Tuveson and colleagues provide a comprehensive review on the fundamental role of cancer-associated fibroblasts in shaping the tumor microenvironment and promoting tumor initiation and progression. Fibroblasts regulate the structure and function of healthy tissues, participate transiently in tissue repair after acute inflammation, and assume an aberrant stimulatory role during chronic inflammatory states including cancer. Such cancer-associated fibroblasts (CAFs) modulate the tumor microenvironment and influence the behavior of neoplastic cells in either a tumor-promoting or tumor-inhibiting manner. These pleiotropic functions highlight the inherent plasticity of fibroblasts and may provide new avenues to understand and therapeutically intervene in malignancies. We discuss the emerging themes of CAF biology in the context of tumorigenesis and therapy.
Collapse
Affiliation(s)
- Daniel Öhlund
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - Ela Elyada
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - David Tuveson
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| |
Collapse
|
50
|
Pastushenko I, Vermeulen PB, Van den Eynden GG, Rutten A, Carapeto FJ, Dirix LY, Van Laere S. Mechanisms of tumour vascularization in cutaneous malignant melanoma: clinical implications. Br J Dermatol 2014; 171:220-33. [PMID: 24641095 DOI: 10.1111/bjd.12973] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2014] [Indexed: 01/02/2023]
Abstract
Malignant melanoma represents < 10% of all skin cancers but is responsible for the majority of skin-cancer-related deaths. Metastatic melanoma has historically been considered as one of the most therapeutically challenging malignancies. Fortunately, for the first time after decades of basic research and clinical investigation, new drugs have produced major clinical responses. Angiogenesis has been considered an important target for cancer treatment. Initial efforts have focused primarily on targeting endothelial and tumour-related vascular endothelial growth factor signalling. Here, we review different mechanisms of tumour vascularization described in melanoma and discuss the potential clinical implications.
Collapse
Affiliation(s)
- I Pastushenko
- Department of Dermatology, Hospital Clínico Universitario 'Lozano Blesa', Zaragoza, 50009, Spain
| | | | | | | | | | | | | |
Collapse
|