1
|
Liu Y, Qi L, Li Z, Yong VW, Xue M. Crosstalk Between Matrix Metalloproteinases and Their Inducer EMMPRIN/CD147: a Promising Therapeutic Target for Intracerebral Hemorrhage. Transl Stroke Res 2025; 16:557-567. [PMID: 38100014 DOI: 10.1007/s12975-023-01225-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/22/2023] [Accepted: 12/05/2023] [Indexed: 04/08/2025]
Abstract
Intracerebral hemorrhage (ICH) is characterized by the disruption of cerebrovascular integrity, resulting in hematoma enlargement, edema formation, and physical damage in the brain parenchyma. Primary ICH also leads to secondary brain injury contributed by oxidative stress, dysregulated immune responses, and proteolysis. In this context, matrix metalloproteinases (MMPs) represent a ubiquitous superfamily of structurally related zinc-dependent endopeptidases capable of degrading all components of the extracellular matrix. They disrupt the blood-brain barrier and promote neuroinflammation. Importantly, several MMP members are upregulated following ICH, and members may have different functions at specific periods in ICH. Hence, the modulation and function of MMPs are more complex than expected. Extracellular matrix metalloproteinase inducer (EMMPRIN, CD147) is a transmembrane glycoprotein that induces the production of MMPs. In this review, we systematically discuss the biology and functions of MMPs and EMMPRIN/CD147 in ICH and the complex crosstalk between them.
Collapse
Affiliation(s)
- Yang Liu
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan International Joint Laboratory of Intracerebral Hemorrhage and Brain Injury, Zhengzhou, Henan, China
| | - Lingxiao Qi
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan International Joint Laboratory of Intracerebral Hemorrhage and Brain Injury, Zhengzhou, Henan, China
| | - Zhe Li
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan International Joint Laboratory of Intracerebral Hemorrhage and Brain Injury, Zhengzhou, Henan, China
| | - V Wee Yong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada.
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- Henan International Joint Laboratory of Intracerebral Hemorrhage and Brain Injury, Zhengzhou, Henan, China.
| |
Collapse
|
2
|
Grötter LG, Cainelli S, Peralta MB, Angeli E, Belotti EM, Ortega HH, Rey F, Velázquez MML, Gareis NC. Metalloproteases and their inhibitors in the postpartum endometrial remodeling in dairy cows: their relationship with days to conception after parturition. Vet Res Commun 2024; 49:53. [PMID: 39714564 DOI: 10.1007/s11259-024-10622-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/14/2024] [Indexed: 12/24/2024]
Abstract
In dairy herds, it is expected that cows will be cycling and the uterus will be ready for a new conception before the fourth week postpartum. However, an alteration in the endometrial remodeling can delay conception, increasing the parturition-to-conception interval, and consequently decreasing the reproductive performance. The endometrial matrix has a relevant participation in the processes of postpartum uterine remodeling. In this sense, the matrix metalloprotease (MMP) system and its inhibitors (TIMPs) are directly involved in the proteolytic degradation of the matrix and their action is related to the concentration of steroid hormones. The aim of this study was to evaluate the protein expression of MMP2, MMP14, MMP9, and their inhibitors, TIMP1 and TIMP2, in the luminal epithelium, glandular epithelium and stroma of endometrial biopsies from dairy cows, at 60 days in milk. Together, the results obtained provide evidence about the expression of MMP2, MMP14 and MMP9, and their inhibitors, TIMP1 and TIMP2, in the postpartum uterus of dairy cows, and about how the balance in their expression could be associated with the achievement of pregnancy. The high protein expression of MMP2, MMP14 and TIMP1 in dairy cows with short parturition-to-conception interval could be important for uterine remodeling and early conception in dairy cows. In addition, the imbalance observed in the MMP9/TIMP1 ratio could be generating an excess of gelatinase activity in the endometrium, causing a delayed conception.
Collapse
Affiliation(s)
- L G Grötter
- Facultad de Ciencias Veterinarias (FCV), Universidad Nacional del Litoral (UNL), Esperanza, Santa Fe, Argentina
- Laboratorio de Biología Celular y Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral) Universidad Nacional del Litoral (UNL). Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Esperanza, Santa Fe, Argentina
| | - S Cainelli
- Laboratorio de Biología Celular y Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral) Universidad Nacional del Litoral (UNL). Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Esperanza, Santa Fe, Argentina
| | - M B Peralta
- Instituto de Ecología Humana y Desarrollo Sustentable, Universidad Católica de Santa Fe. Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Santa Fe, Argentina
| | - E Angeli
- Facultad de Ciencias Veterinarias (FCV), Universidad Nacional del Litoral (UNL), Esperanza, Santa Fe, Argentina
- Laboratorio de Biología Celular y Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral) Universidad Nacional del Litoral (UNL). Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Esperanza, Santa Fe, Argentina
| | - E M Belotti
- Facultad de Ciencias Veterinarias (FCV), Universidad Nacional del Litoral (UNL), Esperanza, Santa Fe, Argentina
- Laboratorio de Biología Celular y Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral) Universidad Nacional del Litoral (UNL). Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Esperanza, Santa Fe, Argentina
| | - H H Ortega
- Facultad de Ciencias Veterinarias (FCV), Universidad Nacional del Litoral (UNL), Esperanza, Santa Fe, Argentina
- Laboratorio de Biología Celular y Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral) Universidad Nacional del Litoral (UNL). Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Esperanza, Santa Fe, Argentina
| | - F Rey
- Facultad de Ciencias Veterinarias (FCV), Universidad Nacional del Litoral (UNL), Esperanza, Santa Fe, Argentina
- Laboratorio de Biología Celular y Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral) Universidad Nacional del Litoral (UNL). Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Esperanza, Santa Fe, Argentina
| | - M M L Velázquez
- Facultad de Ciencias Veterinarias (FCV), Universidad Nacional del Litoral (UNL), Esperanza, Santa Fe, Argentina
- Instituto de Ecología Humana y Desarrollo Sustentable, Universidad Católica de Santa Fe. Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Santa Fe, Argentina
| | - N C Gareis
- Facultad de Ciencias Veterinarias (FCV), Universidad Nacional del Litoral (UNL), Esperanza, Santa Fe, Argentina.
- Laboratorio de Biología Celular y Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral) Universidad Nacional del Litoral (UNL). Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Esperanza, Santa Fe, Argentina.
- Instituto Nacional de Tecnología Agropecuaria (INTA), Rafaela, Santa Fe, Argentina.
| |
Collapse
|
3
|
Pang G, Ye L, Jiang Y, Wu Y, Zhang R, Yang H, Yang Y. Unveiling the bidirectional role of MMP9: A key player in kidney injury. Cell Signal 2024; 122:111312. [PMID: 39074714 DOI: 10.1016/j.cellsig.2024.111312] [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: 05/15/2024] [Revised: 07/04/2024] [Accepted: 07/22/2024] [Indexed: 07/31/2024]
Abstract
Matrix metalloproteinases (MMPs) are a group of zinc-dependent proteolytic metalloenzymes that are involved in numerous pathological conditions, including nephropathy. MMP9, a member of the MMPs family, is categorized as a constituent of the gelatinase B subgroup, and its involvement in extracellular matrix (ECM) remodeling and renal fibrosis highlights its importance in the development and progression of renal diseases. The exact role of MMP9 in the development of kidney diseases is still controversial. This study investigated the dual role of MMP9 in kidney injury, discussing its implications in the pathogenesis of kidney diseases and investigating the design and mechanism of MMP9 inhibitors based on previous studies. This study provides an effective basis for the development of novel and selective MMP9 inhibitors for treating renal diseases.
Collapse
Affiliation(s)
- Guiying Pang
- Anhui University of Traditional Chinese Medicine, Hefei 230000, People's Republic of China; Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd., Beijing 102609, People's Republic of China; Joint Graduate School, Yangtze Delta Drug Advanced Research Institute, Nantong 226133, People's Republic of China
| | - Ling Ye
- Anhui University of Traditional Chinese Medicine, Hefei 230000, People's Republic of China; Department of Pharmacology, Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing 102609, People's Republic of China; Joint Graduate School, Yangtze Delta Drug Advanced Research Institute, Nantong 226133, People's Republic of China
| | - Yinxiao Jiang
- Anhui University of Traditional Chinese Medicine, Hefei 230000, People's Republic of China; Joint Graduate School, Yangtze Delta Drug Advanced Research Institute, Nantong 226133, People's Republic of China
| | - Yilin Wu
- Anhui University of Traditional Chinese Medicine, Hefei 230000, People's Republic of China; Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd., Beijing 102609, People's Republic of China; Joint Graduate School, Yangtze Delta Drug Advanced Research Institute, Nantong 226133, People's Republic of China
| | - Rufeng Zhang
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd., Beijing 102609, People's Republic of China; Department of Pharmacology, Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing 102609, People's Republic of China
| | - Hongxu Yang
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd., Beijing 102609, People's Republic of China.
| | - Yi Yang
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd., Beijing 102609, People's Republic of China; Joint Graduate School, Yangtze Delta Drug Advanced Research Institute, Nantong 226133, People's Republic of China.
| |
Collapse
|
4
|
Sabeh F, Li XY, Olson AW, Botvinick E, Kurup A, Gimenez LE, Cho JS, Weiss SJ. Mmp14-dependent remodeling of the pericellular-dermal collagen interface governs fibroblast survival. J Cell Biol 2024; 223:e202312091. [PMID: 38990714 PMCID: PMC11244150 DOI: 10.1083/jcb.202312091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 05/10/2024] [Accepted: 05/30/2024] [Indexed: 07/13/2024] Open
Abstract
Dermal fibroblasts deposit type I collagen, the dominant extracellular matrix molecule found in skin, during early postnatal development. Coincident with this biosynthetic program, fibroblasts proteolytically remodel pericellular collagen fibrils by mobilizing the membrane-anchored matrix metalloproteinase, Mmp14. Unexpectedly, dermal fibroblasts in Mmp14-/- mice commit to a large-scale apoptotic program that leaves skin tissues replete with dying cells. A requirement for Mmp14 in dermal fibroblast survival is recapitulated in vitro when cells are embedded within, but not cultured atop, three-dimensional hydrogels of crosslinked type I collagen. In the absence of Mmp14-dependent pericellular proteolysis, dermal fibroblasts fail to trigger β1 integrin activation and instead actuate a TGF-β1/phospho-JNK stress response that leads to apoptotic cell death in vitro as well as in vivo. Taken together, these studies identify Mmp14 as a requisite cell survival factor that maintains dermal fibroblast viability in postnatal dermal tissues.
Collapse
Affiliation(s)
- Farideh Sabeh
- Division of Genetic Medicine, Department of Internal Medicine, Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Xiao-Yan Li
- Division of Genetic Medicine, Department of Internal Medicine, Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Adam W. Olson
- Division of Genetic Medicine, Department of Internal Medicine, Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Elliot Botvinick
- The Henry Samueli School of Engineering, University of California, Irvine, CA, USA
| | - Abhishek Kurup
- The Henry Samueli School of Engineering, University of California, Irvine, CA, USA
| | - Luis E. Gimenez
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Jung-Sun Cho
- Division of Genetic Medicine, Department of Internal Medicine, Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Stephen J. Weiss
- Division of Genetic Medicine, Department of Internal Medicine, Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
5
|
Matrix Metalloproteinases Shape the Tumor Microenvironment in Cancer Progression. Int J Mol Sci 2021; 23:ijms23010146. [PMID: 35008569 PMCID: PMC8745566 DOI: 10.3390/ijms23010146] [Citation(s) in RCA: 212] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer progression with uncontrolled tumor growth, local invasion, and metastasis depends largely on the proteolytic activity of numerous matrix metalloproteinases (MMPs), which affect tissue integrity, immune cell recruitment, and tissue turnover by degrading extracellular matrix (ECM) components and by releasing matrikines, cell surface-bound cytokines, growth factors, or their receptors. Among the MMPs, MMP-14 is the driving force behind extracellular matrix and tissue destruction during cancer invasion and metastasis. MMP-14 also influences both intercellular as well as cell-matrix communication by regulating the activity of many plasma membrane-anchored and extracellular proteins. Cancer cells and other cells of the tumor stroma, embedded in a common extracellular matrix, interact with their matrix by means of various adhesive structures, of which particularly invadopodia are capable to remodel the matrix through spatially and temporally finely tuned proteolysis. As a deeper understanding of the underlying functional mechanisms is beneficial for the development of new prognostic and predictive markers and for targeted therapies, this review examined the current knowledge of the interplay of the various MMPs in the cancer context on the protein, subcellular, and cellular level with a focus on MMP14.
Collapse
|
6
|
Yu J, He Z, He X, Luo Z, Lian L, Wu B, Lan P, Chen H. Comprehensive Analysis of the Expression and Prognosis for MMPs in Human Colorectal Cancer. Front Oncol 2021; 11:771099. [PMID: 34804973 PMCID: PMC8602079 DOI: 10.3389/fonc.2021.771099] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 10/13/2021] [Indexed: 12/22/2022] Open
Abstract
Background Previous study implicated that genes of matrix metalloproteinase (MMP) family play an important role in tumor invasion, neoangiogenesis, and metastasis. However, the diverse expression patterns and prognostic values of 24 MMPs in colorectal cancer are yet to be analyzed. Methods In this study, by integrating public database and our data, we first investigated the expression levels and protein levels of MMPs in patients with colorectal cancer. Then, by using TCGA and GEO datasets, we evaluated the association of MMPs with clinicopathological parameters and prognosis of colorectal cancer. Finally, by using the cBioPortal online tool, we analyzed the alterations of MMPs and did the network and pathway analyses for MMPs and their nearby genes. Results We found that, MMP1, MMP3, MMP7, MMP9–MMP12, and MMP14 were consistently upregulated in public dataset and our samples. Whereas, MMP28 was consistently downregulated in public dataset and our samples. In the clinicopathological analyses, upregulated MMP11, MMP14, MMP16, MMP17, MMP19, and MMP23B were significantly associated with a higher tumor stage. In the survival analyses, upregulated MMP11, MMP14, MMP17, and MMP19 were significantly associated with a shorter progression-free survival (PFS) time and a shorter relapse-free (RFS) time. Discussion This study implied that MMP11, MMP14, MMP17, and MMP19 are potential targets of precision therapy for patients with colorectal cancer.
Collapse
Affiliation(s)
- Jing Yu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Guangzhou, China
| | - Zhen He
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Guangzhou, China
| | - Xiaowen He
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Guangzhou, China
| | - Zhanhao Luo
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Guangzhou, China
| | - Lei Lian
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Guangzhou, China
| | - Baixing Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for Ribose Nucleic Acid (RNA) Medicine, Ribose Nucleic Acid (RNA) Biomedical Institute, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ping Lan
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Guangzhou, China
| | - Haitao Chen
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China.,School of Public Health, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
7
|
Luo Y, Hu J, Liu Y, Li L, Li Y, Sun B, Kong R. Invadopodia: A potential target for pancreatic cancer therapy. Crit Rev Oncol Hematol 2021; 159:103236. [PMID: 33482351 DOI: 10.1016/j.critrevonc.2021.103236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 01/05/2021] [Accepted: 01/16/2021] [Indexed: 02/08/2023] Open
Abstract
Dissemination of cancer cells is an intricate multistep process that represents the most deadly aspect of cancer. Cancer cells form F-actin-rich protrusions known as invadopodia to invade surrounding tissues, blood vessels and lymphatics. A number of studies have demonstrated the significant roles of invadopodia in cancer. Therefore, the specific cells and molecules involved in invadopodia activity can provide as therapeutic targets. In this review, we included a thorough overview of studies in invadopodia and discussed their functions in cancer metastasis. We then presented the specific cells and molecules involved in invadopodia activity in pancreatic cancer and analyzed their suitability to be effective therapeutic targets. Currently, drugs targeting invadopodia and relevant clinical trials are negligible. Here, we highlighted the significance of potential drugs and discussed future obstacles in implementing clinical trials. This review presents a new perspective on invadopodia-induced pancreatic cancer metastasis and may prosper the development of targeted therapeutics against pancreatic cancer.
Collapse
Affiliation(s)
- Yan Luo
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jisheng Hu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yong Liu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Le Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yilong Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bei Sun
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Rui Kong
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
8
|
Sarkar P, Li Z, Ren W, Wang S, Shao S, Sun J, Ren X, Perkins NG, Guo Z, Chang CEA, Song J, Xue M. Inhibiting Matrix Metalloproteinase-2 Activation by Perturbing Protein-Protein Interactions Using a Cyclic Peptide. J Med Chem 2020; 63:6979-6990. [PMID: 32491863 DOI: 10.1021/acs.jmedchem.0c00180] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We report on a cyclic peptide that inhibits matrix metalloproteinase-2 (MMP2) activation with a low-nM-level potency. This inhibitor specifically binds to the D570-A583 epitope on proMMP2 and interferes with the protein-protein interaction (PPI) between proMMP2 and tissue inhibitor of metalloproteinases-2 (TIMP2), thereby preventing the TIMP2-assisted proMMP2 activation process. We developed this cyclic peptide inhibitor through an epitope-targeted library screening process and validated its binding to proMMP2. Using a human melanoma cell line, we demonstrated the cyclic peptide's ability to modulate cellular MMP2 activities and inhibit cell migration. These results provide the first successful example of targeting the PPI between proMMP2 and TIMP2, confirming the feasibility of an MMP2 inhibition strategy that has been sought after for 2 decades.
Collapse
Affiliation(s)
- Priyanka Sarkar
- Department of Chemistry, University of California, Riverside, Riverside, California 92521, United States
| | - Zhonghan Li
- Department of Chemistry, University of California, Riverside, Riverside, California 92521, United States
| | - Wendan Ren
- Department of Biochemistry, University of California, Riverside, Riverside, California 92521, United States
| | - Siwen Wang
- Department of Chemistry, University of California, Riverside, Riverside, California 92521, United States
| | - Shiqun Shao
- Department of Chemistry, University of California, Riverside, Riverside, California 92521, United States
| | - Jianan Sun
- Department of Chemistry, University of California, Riverside, Riverside, California 92521, United States
| | - Xiaodong Ren
- Medical College, Guizhou University, Guiyang, Guizhou 550025, China
| | - Nicole G Perkins
- Department of Chemistry, University of California, Riverside, Riverside, California 92521, United States
| | - Zhili Guo
- Department of Chemistry, University of California, Riverside, Riverside, California 92521, United States
| | - Chia-En A Chang
- Department of Chemistry, University of California, Riverside, Riverside, California 92521, United States
| | - Jikui Song
- Department of Biochemistry, University of California, Riverside, Riverside, California 92521, United States
| | - Min Xue
- Department of Chemistry, University of California, Riverside, Riverside, California 92521, United States
| |
Collapse
|
9
|
Signaling Determinants of Glioma Cell Invasion. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1202:129-149. [PMID: 32034712 DOI: 10.1007/978-3-030-30651-9_7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Tumor cell invasiveness is a critical challenge in the clinical management of glioma patients. In addition, there is accumulating evidence that current therapeutic modalities, including anti-angiogenic therapy and radiotherapy, can enhance glioma invasiveness. Glioma cell invasion is stimulated by both autocrine and paracrine factors that act on a large array of cell surface-bound receptors. Key signaling elements that mediate receptor-initiated signaling in the regulation of glioblastoma invasion are Rho family GTPases, including Rac, RhoA and Cdc42. These GTPases regulate cell morphology and actin dynamics and stimulate cell squeezing through the narrow extracellular spaces that are typical of the brain parenchyma. Transient attachment of cells to the extracellular matrix is also necessary for glioblastoma cell invasion. Interactions with extracellular matrix components are mediated by integrins that initiate diverse intracellular signalling pathways. Key signaling elements stimulated by integrins include PI3K, Akt, mTOR and MAP kinases. In order to detach from the tumor mass, glioma cells secrete proteolytic enzymes that cleave cell surface adhesion molecules, including CD44 and L1. Key proteases produced by glioma cells include uPA, ADAMs and MMPs. Increased understanding of the molecular mechanisms that control glioma cell invasion has led to the identification of molecular targets for therapeutic intervention in this devastating disease.
Collapse
|
10
|
Benzing C, Lam H, Tsang CM, Rimmer A, Arroyo-Berdugo Y, Calle Y, Wells CM. TIMP-2 secreted by monocyte-like cells is a potent suppressor of invadopodia formation in pancreatic cancer cells. BMC Cancer 2019; 19:1214. [PMID: 31836008 PMCID: PMC6911299 DOI: 10.1186/s12885-019-6429-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 12/03/2019] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Monocytes are a major component of the tumor microenvironment (TME) in pancreatic ductal adenocarcinoma (PDAC). However, the complex interactions between tumor cells and monocytes and their role in tumor invasion have not been fully established. METHODS To specifically test the impact of interaction on invasive potential two PDAC cell lines PaTu8902 and CFPAC-1 were selected on their ability to form invasive adhesions, otherwise known as invadopodia and invade in a spheroid invasion assay. RESULTS Interestingly when the PDAC cells were co-cultured with undifferentiated THP1 monocyte-like cells invadopodia formation was significantly suppressed. Moreover, conditioned media of THP1 cells (CM) was also able to suppress invadopodia formation. Further investigation revealed that both tissue inhibitor of metalloproteinase (TIMP) 1 and 2 were present in the CM. However, suppression of invadopodia formation was found that was specific to TIMP2 activity. CONCLUSIONS Our findings indicate that TIMP2 levels in the tumour microenvironment may have prognostic value in patients with PDAC. Furthermore, activation of TIMP2 expressing monocytes in the primary tumour could present a potential therapeutic opportunity to suppress cell invasion in PDAC.
Collapse
Affiliation(s)
- Christian Benzing
- School of Cancer and Pharmaceutical Sciences, New Hunts House, Kings College London, London, SE1 1UL, UK.,Department of Surgery, Campus Charité-Mitte and Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Hoyin Lam
- School of Cancer and Pharmaceutical Sciences, New Hunts House, Kings College London, London, SE1 1UL, UK
| | - Chi Man Tsang
- Department of Anatomical and Cellular Pathology and State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hongkong, Hong Kong SAR
| | - Alexander Rimmer
- School of Cancer and Pharmaceutical Sciences, New Hunts House, Kings College London, London, SE1 1UL, UK
| | | | - Yolanda Calle
- Department of Life Sciences, University of Roehampton, London, UK
| | - Claire M Wells
- School of Cancer and Pharmaceutical Sciences, New Hunts House, Kings College London, London, SE1 1UL, UK.
| |
Collapse
|
11
|
McElvaney OJ, Wade P, Murphy M, Reeves EP, McElvaney NG. Targeting airway inflammation in cystic fibrosis. Expert Rev Respir Med 2019; 13:1041-1055. [PMID: 31530195 DOI: 10.1080/17476348.2019.1666715] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Introduction: The major cause of morbidity and mortality in patients with cystic fibrosis (CF) is lung disease. Inflammation in the CF airways occurs from a young age and contributes significantly to disease progression and shortened life expectancy. Areas covered: In this review, we discuss the key immune cells involved in airway inflammation in CF, the contribution of the intrinsic genetic defect to the CF inflammatory phenotype, and anti-inflammatory strategies designed to overcome what is a critical factor in the pathogenesis of CF lung disease. Review of the literature was carried out using the MEDLINE (from 1975 to 2018), Google Scholar and The Cochrane Library databases. Expert opinion: Therapeutic interventions specifically targeting the defective CF transmembrane conductance regulator (CFTR) protein have changed the clinical landscape and significantly improved the outlook for CF. As survival estimates for people with CF increase, long-term management has become an important focus, with an increased need for therapies targeted at specific elements of inflammation, to complement CFTR modulator therapies.
Collapse
Affiliation(s)
- Oliver J McElvaney
- Irish Centre for Genetic Lung Disease, Royal College of Surgeons in Ireland, Beaumont Hospital , Dublin , Ireland
| | - Patricia Wade
- Irish Centre for Genetic Lung Disease, Royal College of Surgeons in Ireland, Beaumont Hospital , Dublin , Ireland
| | - Mark Murphy
- Irish Centre for Genetic Lung Disease, Royal College of Surgeons in Ireland, Beaumont Hospital , Dublin , Ireland
| | - Emer P Reeves
- Irish Centre for Genetic Lung Disease, Royal College of Surgeons in Ireland, Beaumont Hospital , Dublin , Ireland
| | - Noel G McElvaney
- Irish Centre for Genetic Lung Disease, Royal College of Surgeons in Ireland, Beaumont Hospital , Dublin , Ireland
| |
Collapse
|
12
|
Hsu KS, Otsu W, Li Y, Wang HC, Chen S, Tsang SH, Chuang JZ, Sung CH. CLIC4 regulates late endosomal trafficking and matrix degradation activity of MMP14 at focal adhesions in RPE cells. Sci Rep 2019; 9:12247. [PMID: 31439888 PMCID: PMC6706427 DOI: 10.1038/s41598-019-48438-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 08/05/2019] [Indexed: 12/17/2022] Open
Abstract
Dysregulation in the extracellular matrix (ECM) microenvironment surrounding the retinal pigment epithelium (RPE) has been implicated in the etiology of proliferative vitreoretinopathy and age-related macular degeneration. The regulation of ECM remodeling by RPE cells is not well understood. We show that membrane-type matrix metalloproteinase 14 (MMP14) is central to ECM degradation at the focal adhesions in human ARPE19 cells. The matrix degradative activity, but not the assembly, of the focal adhesion is regulated by chloride intracellular channel 4 (CLIC4). CLIC4 is co-localized with MMP14 in the late endosome. CLIC4 regulates the proper sorting of MMP14 into the lumen of the late endosome and its proteolytic activation in lipid rafts. CLIC4 has the newly-identified “late domain” motif that binds to MMP14 and to Tsg101, a component of the endosomal sorting complex required for transport (ESCRT) complex. Unlike the late domain mutant CLIC4, wild-type CLIC4 can rescue the late endosomal sorting defect of MMP14. Finally, CLIC4 knockdown inhibits the apical secretion of MMP2 in polarized human RPE monolayers. These results, taken together, demonstrate that CLIC4 is a novel matrix microenvironment modulator and a novel regulator for late endosomal cargo sorting. Moreover, the late endosomal sorting of MMP14 actively regulates its surface activation in RPE cells.
Collapse
Affiliation(s)
- Kuo-Shun Hsu
- Department of Ophthalmology, Weill Medical College of Cornell University, New York, NY, USA.,Department of Surgery, Colorectal Service and Laboratory of Signal Transduction, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wataru Otsu
- Department of Ophthalmology, Weill Medical College of Cornell University, New York, NY, USA.,Department of Biomedical Research Laboratory, Gifu Pharmaceutical University, Gifu, Japan
| | - Yao Li
- Department of Ophthalmology, Columbia University, New York, NY, USA
| | - Heuy-Ching Wang
- Ocular Trauma Task Area, US Army Institute of Surgical Research, Joint Base San Antonio-Fort Sam Houston, TX, San Antonio, USA
| | - Shuibing Chen
- Department of Surgery and Department of Biochemistry, Weill Medical College of Cornell University, New York, NY, USA
| | - Stephen H Tsang
- Department of Ophthalmology, Columbia University, New York, NY, USA.,Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA.,Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA.,Department of Pathology & Cell Biology, and Columbia Stem Cell Initiative, Columbia University Medical Center, New York, NY, USA
| | - Jen-Zen Chuang
- Department of Ophthalmology, Weill Medical College of Cornell University, New York, NY, USA
| | - Ching-Hwa Sung
- Department of Ophthalmology, Weill Medical College of Cornell University, New York, NY, USA. .,Department of Cell and Developmental Biology, Weill Medical College of Cornell University, New York, NY, USA.
| |
Collapse
|
13
|
Campbell WA, Deshmukh A, Blum S, Todd L, Mendonca N, Weist J, Zent J, Hoang TV, Blackshaw S, Leight J, Fischer AJ. Matrix-metalloproteinase expression and gelatinase activity in the avian retina and their influence on Müller glia proliferation. Exp Neurol 2019; 320:112984. [PMID: 31251936 DOI: 10.1016/j.expneurol.2019.112984] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 06/15/2019] [Accepted: 06/24/2019] [Indexed: 12/16/2022]
Abstract
Gelatinases are a class of matrix metalloproteinases (MMPs) that degrade the extracellular matrix (ECM) to regulate intercellular signaling and cell migration. Gelatinase activity is tightly regulated via proteolytic activation and through the expression of tissue inhibitors of matrix metalloproteinases (TIMPs). Gelatinase activity has been implicated in retinal pathophysiology in different animal models and human disease. However, the role of gelatinases in retinal regeneration remains uncertain. In this study we investigated the dynamic changes in gelatinase activity in response to excitotoxic damage and how this enzymatic activity influenced the formation of Müller glia progenitor cells (MGPCs) in the avian retina. This study used hydrogels containing a gelatinase-degradable fluorescent peptide to measure gelatinase activity in vitro and dye quenched gelatin to localize enzymatic activity in situ. These data were corroborated by using single cell RNA sequencing (scRNA-seq). Gelatinase mRNA, specifically MMP2, was detected in oligodendrocytes and Non-Astrocytic Inner Retinal Glia (NIRG). Total retinal gelatinase activity was reduced following NMDA-treatment, and sustained inhibition of MMP2 prior to damage or growth factor treatment increased the formation of proliferating MGPCs and c-fos signaling. We observed that microglia, Müller glia (MG), and NIRG cells were involved in regulating changes in gelatinase activity through TIMP2 and TIMP3. Collectively, these findings implicate MMP2 in reprogramming of Muller glia into MGPCs.
Collapse
Affiliation(s)
- Warren A Campbell
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, United States of America
| | - Ameya Deshmukh
- Department of Biomedical Engineering, College of Engineering, The comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States of America
| | - Sydney Blum
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, United States of America
| | - Levi Todd
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, United States of America
| | - Ninoshka Mendonca
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, United States of America
| | - Jessica Weist
- Department of Biomedical Engineering, College of Engineering, The comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States of America
| | - Joshua Zent
- Department of Biomedical Engineering, College of Engineering, The comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States of America
| | - Thanh V Hoang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Jennifer Leight
- Department of Biomedical Engineering, College of Engineering, The comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States of America
| | - Andy J Fischer
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, United States of America.
| |
Collapse
|
14
|
Post-Translational Modification-Dependent Activity of Matrix Metalloproteinases. Int J Mol Sci 2019; 20:ijms20123077. [PMID: 31238509 PMCID: PMC6627178 DOI: 10.3390/ijms20123077] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/16/2019] [Accepted: 06/18/2019] [Indexed: 12/18/2022] Open
Abstract
Due to their capacity to process different proteins of the extracellular matrix (ECM), matrix metalloproteinases (MMPs) were initially described as a family of secreted proteases, functioning as main ECM regulators. However, through proteolytic processing of various biomolecules, MMPs also modulate intra- and extracellular pathways and networks. Thereby, they are functionally implicated in the regulation of multiple physiological and pathological processes. Consequently, MMP activity is tightly regulated through a combination of epigenetic, transcriptional, and post-transcriptional control of gene expression, proteolytic activation, post-translational modifications (PTMs), and extracellular inhibition. In addition, MMPs, their substrates and ECM binding partners are frequently modified by PTMs, which suggests an important role of PTMs in modulating the pleiotropic activities of these proteases. This review summarizes the recent progress towards understanding the role of PTMs (glycosylation, phosphorylation, glycosaminoglycans) on the activity of several members of the MMP family.
Collapse
|
15
|
Pahwa S, Bhowmick M, Amar S, Cao J, Strongin AY, Fridman R, Weiss SJ, Fields GB. Characterization and regulation of MT1-MMP cell surface-associated activity. Chem Biol Drug Des 2018; 93:1251-1264. [PMID: 30480376 DOI: 10.1111/cbdd.13450] [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: 09/01/2018] [Revised: 10/18/2018] [Accepted: 11/11/2018] [Indexed: 12/19/2022]
Abstract
Quantitative assessment of MT1-MMP cell surface-associated proteolytic activity remains undefined. Presently, MT1-MMP was stably expressed and a cell-based FRET assay developed to quantify activity toward synthetic collagen-model triple-helices. To estimate the importance of cell surface localization and specific structural domains on MT1-MMP proteolysis, activity measurements were performed using a series of membrane-anchored MT1-MMP mutants and compared directly with those of soluble MT1-MMP. MT1-MMP activity (kcat /KM ) on the cell surface was 4.8-fold lower compared with soluble MT1-MMP, with the effect largely manifested in kcat . Deletion of the MT1-MMP cytoplasmic tail enhanced cell surface activity, with both kcat and KM values affected, while deletion of the hemopexin-like domain negatively impacted KM and increased kcat . Overall, cell surface localization of MT1-MMP restricts substrate binding and protein-coupled motions (based on changes in both kcat and KM ) for catalysis. Comparison of soluble and cell surface-bound MT2-MMP revealed 12.9-fold lower activity on the cell surface. The cell-based assay was utilized for small molecule and triple-helical transition state analog MMP inhibitors, which were found to function similarly in solution and at the cell surface. These studies provide the first quantitative assessments of MT1-MMP activity and inhibition in the native cellular environment of the enzyme.
Collapse
Affiliation(s)
- Sonia Pahwa
- Departments of Chemistry and Biology, Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida
| | - Manishabrata Bhowmick
- Departments of Chemistry and Biology, Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida
| | - Sabrina Amar
- Departments of Chemistry and Biology, Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida.,Department of Chemistry & Biochemistry, Florida Atlantic University, Jupiter, Florida
| | - Jian Cao
- Departments of Medicine/Cancer Prevention and Pathology, Stony Brook University, Stony Brook, New York
| | - Alex Y Strongin
- Cancer Research Center, Sanford Burnham Prebys Medical Research Institute, La Jolla, California
| | - Rafael Fridman
- Department of Pathology and the Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Stephen J Weiss
- Division of Molecular Medicine & Genetics, Department of Internal Medicine, Life Sciences Institute, University of Michigan, Ann Arbor, Michigan
| | - Gregg B Fields
- Departments of Chemistry and Biology, Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida.,Department of Chemistry & Biochemistry, Florida Atlantic University, Jupiter, Florida.,The Scripps Research Institute/Scripps Florida, Jupiter, Florida
| |
Collapse
|
16
|
Amara N, Tholen M, Bogyo M. Chemical Tools for Selective Activity Profiling of Endogenously Expressed MMP-14 in Multicellular Models. ACS Chem Biol 2018; 13:2645-2654. [PMID: 30160940 DOI: 10.1021/acschembio.8b00562] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Matrix metalloproteases (MMPs) are a large family of zinc-dependent endopeptidases involved in a diverse set of physiological and pathological processes, most notably in cancer. Current methods for imaging and quantifying MMP activity lack sufficient selectivity and spatiotemporal resolution to allow studies of specific MMP function in vivo. Previously, we reported a strategy for selective targeting of MMPs by engineering a functionally silent cysteine mutation that enables highly specific covalent modification by a designed activity-based probe. Here, we describe the translation of that technology into a mouse model of breast cancer and subsequent demonstration of the utility of the approach for studies of MMP-14 activation in the tumor microenvironment. Using this approach, we find that MMP-14 is active in late stage tumors and is predominantly associated with stromal cell populations that have been activated by specific signaling molecules (e.g., TGFβ) produced by tumor cells. Our data demonstrate the applicability of this approach for studies of MMP function in whole organisms and identify important regulatory mechanisms for MMP-14 activity in the tumor microenvironment.
Collapse
|
17
|
Gillette KM, Forbes K, Sehgal I. Detection of Matrix Metalloproteinases (MMP), Tissue Inhibitor of Metalloproteinase-2, Urokinase and Plasminogen Activator Inhibitor-1 within Matrigel and Growth Factor-Reduced Matrigel Basement Membrane. TUMORI JOURNAL 2018; 89:421-5. [PMID: 14606648 DOI: 10.1177/030089160308900415] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Aims and background Matrigel (MG) basement membrane is commonly used for in vitro studies of cellular migration, invasion and angiogenesis. It contains structural molecules such as collagen IV and laminin plus several growth factors which are diminished in growth factor-reduced Matrigel (GFR-MG). A less appreciated but important aspect of MG is the presence of matrix enzymes and their inhibitors. For relevant interpretation of data using MG/GFR-MG models, it may be necessary to know the enzymes or inhibitors contributed by these basement membranes themselves. Methods Immunoblot and zymography were used to detect the presence or absence of MMP-1 and 7, tissue inhibitor of metalloproteinase 1 and 2 (TIMP-1, TIMP-2), plasminogen activator activity and plasminogen activator inhibitor-1 (PAI-1). Growth and invasion assays using prostate cancer cells were used to assess the effects of TIMP-2 presence or absence. Results We detected MMP-7, urokinase plasminogen activator (uPA) and PAI-1 in both Matrigels, TIMP-2 was detected only in regular Matrigel and no MMP-1 or TIMP-1 was detected in either matrix. Invasion assays comparing regular MG and GFR-MG indicated cell line variability with regard to invasion efficiency as two tested prostate cancer lines were unaffected by the MG type while one was significantly more invasive in regular MG. Growth experiments suggest that the presence of TIMP-2 in regular MG may retard growth but overall proliferation is still greater in regular MG than in GFR-MG. Conclusions These data provide a useful reference for interpretation of in vitro Matrigel assays.
Collapse
Affiliation(s)
- Karin M Gillette
- Center for Protease Research, North Dakota State University College of Pharmacy, Fargo, North Dakota, USA
| | | | | |
Collapse
|
18
|
Mimura I, Hirakawa Y, Kanki Y, Nakaki R, Suzuki Y, Tanaka T, Aburatani H, Nangaku M. Genome-wide analysis revealed that DZNep reduces tubulointerstitial fibrosis via down-regulation of pro-fibrotic genes. Sci Rep 2018; 8:3779. [PMID: 29491489 PMCID: PMC5830881 DOI: 10.1038/s41598-018-22180-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 01/29/2018] [Indexed: 12/14/2022] Open
Abstract
Tubulointerstitial fibrosis has been recently reported to be caused by the collapse of the epigenetic regulation of kidney diseases. We examined whether pharmacological inhibition of histone modification is effective against renal fibrosis. DZNep (3-deazaneplanocin A) was originally developed as an anti-cancer drug to inhibit the repressive histone mark, H3K27me3. We used a model of chronic tubulointerstitial fibrosis induced by unilateral ischaemia/reperfusion and administered DZNep intravenously to the mice for 8 weeks. We found DZNep contributes to the reduction of tubulointerstitial fibrosis. We selected only tubular cells from in vivo samples using laser-capture microdissection because epigenetic regulation is specific to the cell types, and we focused on the changes in the tubular cells. We performed a genome-wide analysis of tubular cells using high-throughput sequencing (RNA-seq) to identify novel epigenetic factors associated with renal fibrosis. We found that pro-fibrotic genes such as COL3A1 (collagen type 3a1) and TIMP2 (tissue inhibitor of metalloproteinase 2) were suppressed by DZNep in vivo. In addition, pro-fibrotic genes such as COL4A1 (collagen type 4a1), TIMP2 and MMP14 were down-regulated by DZNep in vitro. In conclusion, we found that pharmacological epigenetic modification by DZNep decreased the expression levels of fibrogenic genes in tubular cells and inhibited tubulointerstitial fibrosis.
Collapse
Affiliation(s)
- Imari Mimura
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Bunkyō, Japan
| | - Yosuke Hirakawa
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Bunkyō, Japan
| | - Yasuharu Kanki
- Isotope Science Center, The University of Tokyo, Bunkyō, Japan
| | - Ryo Nakaki
- Division of Genome Science, Research Center for Advanced Science and Technology, The University of Tokyo, Bunkyō, Japan
| | - Yutaka Suzuki
- Graduate School of Frontier Sciences, The University of Tokyo, Bunkyō, Japan
| | - Tetsuhiro Tanaka
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Bunkyō, Japan
| | - Hiroyuki Aburatani
- Division of Genome Science, Research Center for Advanced Science and Technology, The University of Tokyo, Bunkyō, Japan
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Bunkyō, Japan.
| |
Collapse
|
19
|
Mapping diet-induced alternative polyadenylation of hypothalamic transcripts in the obese rat. Physiol Behav 2018; 188:173-180. [PMID: 29391168 DOI: 10.1016/j.physbeh.2018.01.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 01/25/2018] [Accepted: 01/27/2018] [Indexed: 12/17/2022]
Abstract
RNA biogenesis has emerged as a powerful biological event that regulates energy homeostasis. In this context insertion of alternative polyadenylation sites (APSs) dictate the fate of newly synthesized RNA molecules and direct alternative splicing of nascent transcripts. Thus APSs serve a mechanistic function by regulating transcriptome expression and function. In this study we employed a novel RNA-Seq Next Generation Sequencing (NGS) approach that utilized the power of Whole Transcriptome Termini Site Sequencing (WTTS-Seq) to simultaneously measure APS events on multiple RNA biotypes. We used this technique to measure APS events in the hypothalamus of adult male Long Evans rats exposed to a palatable high fat diet (HFD) or chow. Rats maintained on HFD displayed typical hyperphagic feeding and ensuing body weight gain over the one-month manipulation period. Our WTTS-Seq analysis mapped approximately 89,000 unique hypothalamic APSs induced by HFD relative to chow fed controls. HFD exposure produced APSs on multiple RNA biotypes in the hypothalamus. The majority of detected APSs occur on mRNA transcripts that encode functional proteins. Notably we find APSs on micro (miRNA) and long non-coding RNAs (lncRNA), newly recognized transcription factors that regulate body weight in rodents. In addition we detect APSs on protein encoding mRNAs that control neuron projection development and synapse organization and glutamate signaling, key events hypothesized to maintain excess food intake. Importantly, quantitative real time PCR indicated that APS insertion led to increased hypothalamic expression of multiple RNA biotypes. Collectively these data highlight APS events as a novel genetic mechanism that directs hypothalamic RNA biogenesis stimulated by diet-induced obesity.
Collapse
|
20
|
Delgado-Calle J, Hancock B, Likine EF, Sato AY, McAndrews K, Sanudo C, Bruzzaniti A, Riancho JA, Tonra JR, Bellido T. MMP14 is a novel target of PTH signaling in osteocytes that controls resorption by regulating soluble RANKL production. FASEB J 2018; 32:2878-2890. [PMID: 29401593 DOI: 10.1096/fj.201700919rrr] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Parathyroid hormone (PTH) affects the skeleton by acting on osteocytes (Ots) in bone through yet unclear mechanisms. We report that matrix metalloproteinase 14 (MMP14) expression/activity are increased in bones from mice with genetic constitutive activation (ca) of the PTH receptor 1 (PTH1R) in Ots (caPTH1ROt) and in bones from mice exposed to elevated PTH levels but not in mice lacking [conditional knockout (cKO)] the PTH1R in Ots (cKOPTH1ROt). Furthermore, PTH upregulates MMP14 in human bone cultures and in Ot-enriched bones from floxed control mice but not from cKOPTH1ROt mice. MMP14 activity increases soluble receptor activator of NF-κΒ ligand production, which in turn, stimulates osteoclast differentiation and resorption. Pharmacologic inhibition of MMP14 activity reduced the high bone remodeling exhibited by caPTH1ROt mice or induced by chronic PTH elevation and decreased bone resorption but allowed full stimulation of bone formation induced by PTH injections, thereby potentiating bone gain. Thus, MMP14 is a new member of the intricate gene network activated in Ots by PTH1R signaling that can be targeted to adjust the skeletal responses to PTH in favor of bone preservation.-Delgado-Calle, J., Hancock, B., Likine, E. F., Sato, A. Y., McAndrews, K., Sanudo, C., Bruzzaniti, A., Riancho, J. A., Tonra, J. R., Bellido, T. MMP14 is a novel target of PTH signaling in osteocytes that controls resorption by regulating soluble RANKL production.
Collapse
Affiliation(s)
- Jesus Delgado-Calle
- Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Indiana Center for Musculoskeletal Health, Indianapolis, Indiana, USA.,Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana, USA
| | - Benjamin Hancock
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Elive F Likine
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Amy Y Sato
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Kevin McAndrews
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana, USA
| | - Carolina Sanudo
- Department of Internal Medicine, Marqués de Valdecilla University Hospital-Instituto de Investigación Marqués de Valdecilla (IDIVAL), University of Cantabria, Santander, Spain
| | - Angela Bruzzaniti
- Indiana Center for Musculoskeletal Health, Indianapolis, Indiana, USA.,Department of Biomedical and Applied Sciences, Indiana University School of Dentistry, Indianapolis, Indiana, USA
| | - Jose A Riancho
- Department of Internal Medicine, Marqués de Valdecilla University Hospital-Instituto de Investigación Marqués de Valdecilla (IDIVAL), University of Cantabria, Santander, Spain
| | | | - Teresita Bellido
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Indiana Center for Musculoskeletal Health, Indianapolis, Indiana, USA.,Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana, USA.,Division of Endocrinology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
21
|
Wang X, Khalil RA. Matrix Metalloproteinases, Vascular Remodeling, and Vascular Disease. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 81:241-330. [PMID: 29310800 DOI: 10.1016/bs.apha.2017.08.002] [Citation(s) in RCA: 421] [Impact Index Per Article: 52.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc-dependent endopeptidases that degrade various proteins in the extracellular matrix (ECM). Typically, MMPs have a propeptide sequence, a catalytic metalloproteinase domain with catalytic zinc, a hinge region or linker peptide, and a hemopexin domain. MMPs are commonly classified on the basis of their substrates and the organization of their structural domains into collagenases, gelatinases, stromelysins, matrilysins, membrane-type (MT)-MMPs, and other MMPs. MMPs are secreted by many cells including fibroblasts, vascular smooth muscle (VSM), and leukocytes. MMPs are regulated at the level of mRNA expression and by activation through removal of the propeptide domain from their latent zymogen form. MMPs are often secreted in an inactive proMMP form, which is cleaved to the active form by various proteinases including other MMPs. MMPs degrade various protein substrates in ECM including collagen and elastin. MMPs could also influence endothelial cell function as well as VSM cell migration, proliferation, Ca2+ signaling, and contraction. MMPs play a role in vascular tissue remodeling during various biological processes such as angiogenesis, embryogenesis, morphogenesis, and wound repair. Alterations in specific MMPs could influence arterial remodeling and lead to various pathological disorders such as hypertension, preeclampsia, atherosclerosis, aneurysm formation, as well as excessive venous dilation and lower extremity venous disease. MMPs are often regulated by endogenous tissue inhibitors of metalloproteinases (TIMPs), and the MMP/TIMP ratio often determines the extent of ECM protein degradation and tissue remodeling. MMPs may serve as biomarkers and potential therapeutic targets for certain vascular disorders.
Collapse
Affiliation(s)
- Xi Wang
- Vascular Surgery Research Laboratories, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
22
|
Liu M, An J, Huang M, Wang L, Tu B, Song Y, Ma K, Wang Y, Wang S, Zhu H, Xu N, Wu L. MicroRNA-492 overexpression involves in cell proliferation, migration, and radiotherapy response of cervical squamous cell carcinomas. Mol Carcinog 2017; 57:32-43. [PMID: 28802022 DOI: 10.1002/mc.22717] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 08/01/2017] [Accepted: 08/08/2017] [Indexed: 02/05/2023]
Affiliation(s)
- Mei Liu
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing P. R. China
| | - Jusheng An
- Department of Gynecological Oncology; National Cancer Center/Cancer Hospital; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing P. R. China
| | - Manni Huang
- Department of Gynecological Oncology; National Cancer Center/Cancer Hospital; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing P. R. China
| | - Liming Wang
- Department of Abdominal Surgery; National Cancer Center/Cancer Hospital; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing P. R. China
| | - Binbin Tu
- Department of Gynecological Oncology; National Cancer Center/Cancer Hospital; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing P. R. China
| | - Yan Song
- Department of Pathology; National Cancer Center/Cancer Hospital; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing P. R. China
| | - Kai Ma
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing P. R. China
| | - Yu Wang
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing P. R. China
| | - Shuren Wang
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing P. R. China
| | - Hongxia Zhu
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing P. R. China
| | - Ningzhi Xu
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing P. R. China
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University, Collaborative Innovation Center for Biotherapy; Chengdu P. R. China
| | - Lingying Wu
- Department of Gynecological Oncology; National Cancer Center/Cancer Hospital; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing P. R. China
| |
Collapse
|
23
|
Fujii M, Yoneda A, Takei N, Sakai-Sawada K, Kosaka M, Minomi K, Yokoyama A, Tamura Y. Endoplasmic reticulum oxidase 1α is critical for collagen secretion from and membrane type 1-matrix metalloproteinase levels in hepatic stellate cells. J Biol Chem 2017; 292:15649-15660. [PMID: 28774960 DOI: 10.1074/jbc.m117.783126] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 07/21/2017] [Indexed: 01/31/2023] Open
Abstract
Upon liver injury, excessive deposition of collagen from activated hepatic stellate cells (HSCs) is a leading cause of liver fibrosis. An understanding of the mechanism by which collagen biosynthesis is regulated in HSCs will provide important clues for practical anti-fibrotic therapy. Endoplasmic reticulum oxidase 1α (ERO1α) functions as an oxidative enzyme of protein disulfide isomerase, which forms intramolecular disulfide bonds of membrane and secreted proteins. However, the role of ERO1α in HSCs remains unclear. Here, we show that ERO1α is expressed and mainly localized in the endoplasmic reticulum in human HSCs. When HSCs were transfected with ERO1α siRNA or an ERO1α shRNA-expressing plasmid, expression of ERO1α was completely silenced. Silencing of ERO1α expression in HSCs markedly suppressed their proliferation but did not induce apoptosis, which was accompanied by impaired secretion of collagen type 1. Silencing of ERO1α expression induced impaired disulfide bond formation and inhibited autophagy via activation of the Akt/mammalian target of rapamycin signaling pathway, resulting in intracellular accumulation of collagen type 1 in HSCs. Furthermore, silencing of ERO1α expression also promoted proteasome-dependent degradation of membrane type 1-matrix metalloproteinase (MT1-MMP), which stimulates cell proliferation through cleavage of secreted collagens. The inhibition of HSC proliferation was reversed by treatment with MT1-MMP-cleaved collagen type 1. The results suggest that ERO1α plays a crucial role in HSC proliferation via posttranslational modification of collagen and MT1-MMP and, therefore, may be a suitable therapeutic target for managing liver fibrosis.
Collapse
Affiliation(s)
- Mizuki Fujii
- From the Department of Oral Functional Prosthodontics, Division of Oral Functional Science, Graduate School of Dental Medicine, Hokkaido University, Nishi-7, Kita-13, Kita-ku, Sapporo 060-8486, Japan.,the Department of Molecular Therapeutics, Center for Food and Medical Innovation, Institute for the Promotion of Business-Regional Collaboration, Hokkaido University, Nishi-11, Kita-21, Kita-ku, Sapporo 001-0021, Japan, and
| | - Akihiro Yoneda
- the Department of Molecular Therapeutics, Center for Food and Medical Innovation, Institute for the Promotion of Business-Regional Collaboration, Hokkaido University, Nishi-11, Kita-21, Kita-ku, Sapporo 001-0021, Japan, and
| | - Norio Takei
- the Department of Molecular Therapeutics, Center for Food and Medical Innovation, Institute for the Promotion of Business-Regional Collaboration, Hokkaido University, Nishi-11, Kita-21, Kita-ku, Sapporo 001-0021, Japan, and
| | - Kaori Sakai-Sawada
- the Department of Molecular Therapeutics, Center for Food and Medical Innovation, Institute for the Promotion of Business-Regional Collaboration, Hokkaido University, Nishi-11, Kita-21, Kita-ku, Sapporo 001-0021, Japan, and
| | - Marina Kosaka
- the Department of Molecular Therapeutics, Center for Food and Medical Innovation, Institute for the Promotion of Business-Regional Collaboration, Hokkaido University, Nishi-11, Kita-21, Kita-ku, Sapporo 001-0021, Japan, and.,the Research and Development Department, Nucleic Acid Medicine Business Division, Nitto Denko Corporation, Nishi-11, Kita-21, Kita-ku, Sapporo 001-0021, Japan
| | - Kenjiro Minomi
- the Department of Molecular Therapeutics, Center for Food and Medical Innovation, Institute for the Promotion of Business-Regional Collaboration, Hokkaido University, Nishi-11, Kita-21, Kita-ku, Sapporo 001-0021, Japan, and.,the Research and Development Department, Nucleic Acid Medicine Business Division, Nitto Denko Corporation, Nishi-11, Kita-21, Kita-ku, Sapporo 001-0021, Japan
| | - Atsuro Yokoyama
- From the Department of Oral Functional Prosthodontics, Division of Oral Functional Science, Graduate School of Dental Medicine, Hokkaido University, Nishi-7, Kita-13, Kita-ku, Sapporo 060-8486, Japan
| | - Yasuaki Tamura
- the Department of Molecular Therapeutics, Center for Food and Medical Innovation, Institute for the Promotion of Business-Regional Collaboration, Hokkaido University, Nishi-11, Kita-21, Kita-ku, Sapporo 001-0021, Japan, and
| |
Collapse
|
24
|
Hussain H, Fisher DI, Abbott WM, Roth RG, Dickson AJ. Use of a protein engineering strategy to overcome limitations in the production of “Difficult to Express” recombinant proteins. Biotechnol Bioeng 2017. [DOI: 10.1002/bit.26358] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Hirra Hussain
- Manchester Institute of Biotechnology; Faculty of Science and Engineering; University of Manchester; M1 7DN Manchester United Kingdom
| | - David I. Fisher
- AstraZeneca, Cambridge Science Park; Milton Cambridge United Kingdom
| | - W. Mark Abbott
- AstraZeneca, Cambridge Science Park; Milton Cambridge United Kingdom
| | | | - Alan J. Dickson
- Manchester Institute of Biotechnology; Faculty of Science and Engineering; University of Manchester; M1 7DN Manchester United Kingdom
| |
Collapse
|
25
|
Chen J, Ren Z, Zhu M, Khalil RA. Decreased homodimerization and increased TIMP-1 complexation of uteroplacental and uterine arterial matrix metalloproteinase-9 during hypertension-in-pregnancy. Biochem Pharmacol 2017; 138:81-95. [PMID: 28506758 DOI: 10.1016/j.bcp.2017.05.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 05/10/2017] [Indexed: 12/11/2022]
Abstract
Preeclampsia is a complication of pregnancy manifested as hypertension-in-pregnancy (HTN-Preg) and often intrauterine growth restriction (IUGR). Placental ischemia could be an initiating event, but the molecular mechanisms are unclear. To test the hypothesis that dimerization of matrix metalloproteinases (MMPs) plays a role in HTN-Preg and IUGR, the levels/activity of MMP-9, tissue inhibitor of metalloproteinase (TIMP-1), and their dimerization forms were measured in the placenta, uterus, and uterine artery of normal pregnant (Preg) rats and a rat model of reduced uteroplacental perfusion pressure (RUPP). Consistent with our previous report, blood pressure (BP) was higher, pup weight was lower, and gelatin zymography showed different gelatinolytic activity for pro-MMP-9, MMP-9, pro-MMP-2 and MMP-2 in RUPP vs Preg rats. Careful examination of the zymograms showed additional bands at 200 and 135kDa. Western blots with MMP-9 antibody suggested that the 200kDa band was a MMP-9 homodimer. Western blots with TIMP-1 antibody as well as reverse zymography suggested that the 135kDa band was a MMP-9/TIMP-1 complex. The protein levels and gelatinase activity of MMP-9 homodimer were decreased while MMP-9/TIMP-1 complex was increased in placenta, uterus and uterine artery of RUPP vs Preg rats. The epidermal growth factor (EGF) receptor blocker erlotinib and protein kinase C (PKC) inhibitor bisindolylmaleimide decreased MMP-9 homodimer and increased MMP-9/TIMP-1 complex in placenta, uterus and uterine artery of Preg rats. EGF and the PKC activator phorbol-12,13-dibutyrate (PDBu) reversed the decreases in MMP-9 homodimer and the increases in MMP-9/TIMP-1 complex in tissues of RUPP rats. Thus, the increased BP and decreased pup weight in placental ischemia model of HTN-Preg are associated with a decrease in MMP-9 homodimer and an increase in MMP-9/TIMP-1 complex in placenta, uterus, and uterine artery, which together would cause a net decrease in MMP-9 activity and reduce uteroplacental and vascular remodeling in the setting of HTN-Preg and IUGR. Enhancing EGFR/PKC signaling may reverse the MMP-9 unfavorable dimerization patterns and thereby promote uteroplacental and vascular remodeling in preeclampsia.
Collapse
Affiliation(s)
- Juanjuan Chen
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Zongli Ren
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Minglin Zhu
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
26
|
Biochemical and Biological Attributes of Matrix Metalloproteinases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 147:1-73. [PMID: 28413025 DOI: 10.1016/bs.pmbts.2017.02.005] [Citation(s) in RCA: 823] [Impact Index Per Article: 102.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc-dependent endopeptidases that are involved in the degradation of various proteins in the extracellular matrix (ECM). Typically, MMPs have a propeptide sequence, a catalytic metalloproteinase domain with catalytic zinc, a hinge region or linker peptide, and a hemopexin domain. MMPs are commonly classified on the basis of their substrates and the organization of their structural domains into collagenases, gelatinases, stromelysins, matrilysins, membrane-type (MT)-MMPs, and other MMPs. MMPs are secreted by many cells including fibroblasts, vascular smooth muscle (VSM), and leukocytes. MMPs are regulated at the level of mRNA expression and by activation of their latent zymogen form. MMPs are often secreted as inactive pro-MMP form which is cleaved to the active form by various proteinases including other MMPs. MMPs cause degradation of ECM proteins such as collagen and elastin, but could influence endothelial cell function as well as VSM cell migration, proliferation, Ca2+ signaling, and contraction. MMPs play a role in tissue remodeling during various physiological processes such as angiogenesis, embryogenesis, morphogenesis, and wound repair, as well as in pathological conditions such as myocardial infarction, fibrotic disorders, osteoarthritis, and cancer. Increases in specific MMPs could play a role in arterial remodeling, aneurysm formation, venous dilation, and lower extremity venous disorders. MMPs also play a major role in leukocyte infiltration and tissue inflammation. MMPs have been detected in cancer, and elevated MMP levels have been associated with tumor progression and invasiveness. MMPs can be regulated by endogenous tissue inhibitors of metalloproteinases (TIMPs), and the MMP/TIMP ratio often determines the extent of ECM protein degradation and tissue remodeling. MMPs have been proposed as biomarkers for numerous pathological conditions and are being examined as potential therapeutic targets in various cardiovascular and musculoskeletal disorders as well as cancer.
Collapse
|
27
|
Kim HI, Lee HS, Kim TH, Lee JS, Lee ST, Lee SJ. Growth-stimulatory activity of TIMP-2 is mediated through c-Src activation followed by activation of FAK, PI3-kinase/AKT, and ERK1/2 independent of MMP inhibition in lung adenocarcinoma cells. Oncotarget 2016; 6:42905-22. [PMID: 26556867 PMCID: PMC4767480 DOI: 10.18632/oncotarget.5466] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 10/26/2015] [Indexed: 12/24/2022] Open
Abstract
Tissue inhibitors of metalloproteinases (TIMPs) control extracellular matrix (ECM) homeostasis by inhibiting the activity of matrix metalloproteinases (MMPs), which are associated with ECM turnover. Recent studies have revealed that TIMPs are implicated in tumorigenesis in both MMP-dependent and MMP-independent manners. We examined a mechanism by which TIMP-2 stimulated lung adenocarcinoma cell proliferation, independent of MMP inhibition. The stimulation of growth by TIMP-2 in A549 cells required c-Src kinase activation. c-Src kinase activity, induced by TIMP-2, concomitantly increased FAK, phosphoinositide 3-kinase (PI3-kinase)/AKT, and ERK1/2 activation. Selective knockdown of integrin α3β1, known as a TIMP-2 receptor, did not significantly change TIMP-2 growth promoting activity. Furthermore, we showed that high TIMP-2 expression in lung adenocarcinomas is associated with a worse prognosis from multiple cohorts, especially for stage I lung adenocarcinoma. Through integrated analysis of The Cancer Genome Atlas data, TIMP-2 expression was significantly associated with the alteration of driving genes, c-Src activation, and PI3-kinase/AKT pathway activation. Taken together, our results demonstrate that TIMP-2 stimulates lung adenocarcinoma cell proliferation through c-Src, FAK, PI3-kinase/AKT, and ERK1/2 pathway activation in an MMP-independent manner.
Collapse
Affiliation(s)
- Han Ie Kim
- Department of Life Science & Biotechnology, Shingyeong University, Gyeonggi-do, 445-741, Republic of Korea
| | - Hyun-Sung Lee
- Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, 77030, U.S.A
| | - Tae Hyun Kim
- Department of Life Science & Biotechnology, Shingyeong University, Gyeonggi-do, 445-741, Republic of Korea
| | - Ju-Seog Lee
- Department of Systems Biology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, U.S.A
| | - Seung-Taek Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 120-749, Republic of Korea
| | - Seo-Jin Lee
- Department of Life Science & Biotechnology, Shingyeong University, Gyeonggi-do, 445-741, Republic of Korea
| |
Collapse
|
28
|
Effects of induction and inhibition of matrix cross-linking on remodeling of the aqueous outflow resistance by ocular trabecular meshwork cells. Sci Rep 2016; 6:30505. [PMID: 27465745 PMCID: PMC4964656 DOI: 10.1038/srep30505] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 07/06/2016] [Indexed: 12/16/2022] Open
Abstract
The trabecular meshwork (TM) tissue controls drainage of aqueous humor from the anterior chamber of the eye primarily by regulating extracellular matrix (ECM) remodeling by matrix metalloproteinases (MMPs). Glaucomatous TM tissue is stiffer than age-matched controls, which may be due to alterations in ECM cross-linking. In this study, we used genipin or beta-aminopropionitrile (BAPN) agents to induce or inhibit matrix cross-linking, respectively, to investigate the effects on outflow resistance and ECM remodeling. Treatment with BAPN increased outflow rates in perfused human and porcine anterior segments, whereas genipin reduced outflow. Using a fluorogenic peptide assay, MMP activity was increased with BAPN treatment, but reduced with genipin treatment. In genipin-treated TM cells, Western immunoblotting showed a reduction of active MMP2 and MMP14 species and the presence of TIMP2-MMP14 higher molecular weight complexes. BAPN treatment increased collagen type I mRNA and protein levels, but genipin reduced the levels of collagen type I, tenascin C, elastin and versican. CD44 and fibronectin levels were unaffected by either treatment. Collectively, our results show that matrix cross-linking has profound effects on outflow resistance and ECM composition and are consistent with the emerging paradigm that the stiffer the ECM, the lower the aqueous outflow facility through the TM.
Collapse
|
29
|
Zhao L, Liu L, Wang S, Wang H, Jiang J. Transcriptome profiles of metamorphosis in the ornamented pygmy frog Microhyla fissipes clarify the functions of thyroid hormone receptors in metamorphosis. Sci Rep 2016; 6:27310. [PMID: 27254593 PMCID: PMC4890586 DOI: 10.1038/srep27310] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 05/16/2016] [Indexed: 11/09/2022] Open
Abstract
Anuran metamorphosis is an excellent system in which to study postembryonic development. Studies on Xenopus (Mesobatrachia) show that thyroid hormone receptors (TRs) regulate metamorphosis in a ligand-dependent manner by coordinating the action of hundreds of genes. However, whether this mechanism is conserved among amphibians is still unknown. To understand the molecular mechanism of this universal phenomenon, we report the transcriptional profiles of the three key developmental stages in Microhyla fissipes (Neobatrachia): premetamorphosis (PM), metamorphic climax (MC) and completion of metamorphosis (CM). In total, 2,293 differentially expressed genes were identified from comparisons of transcriptomes, and these genes showed stage-specific expression patterns. Unexpectedly, we found that TRα was highly expressed in Xenopus laevis and Bufo gargarizans at premetamorphosis but showed low expression in M. fissipes. In contrast, TRβ was highly expressed during metamorphosis in M. fissipes and X. laevis. This result may imply that TRβ is essential for initiating metamorphosis, at least in M. fissipes. Thus, our work not only identifies genes that are likely to be involved in Neobatrachia metamorphosis but also clarifies the roles of unliganded TRα in regulating tadpole growth and timing of metamorphosis, which may be conserved in anurans, and the role of liganded TRβ in launching metamorphosis.
Collapse
Affiliation(s)
- Lanying Zhao
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lusha Liu
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| | - Shouhong Wang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hongyuan Wang
- College of Life Sciences, Shaanxi Normal University, Xi'an 710062, China
| | - Jianping Jiang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| |
Collapse
|
30
|
Song YH, Shon SH, Shan M, Stroock A, Fischbach C. Adipose-derived stem cells increase angiogenesis through matrix metalloproteinase-dependent collagen remodeling. Integr Biol (Camb) 2016; 8:205-15. [PMID: 26758423 PMCID: PMC4755818 DOI: 10.1039/c5ib00277j] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Adipose-derived stem cells (ASCs) are key regulators of new blood vessel formation and widely investigated for their role in tissue regeneration and tumorigenesis. However, the cellular and molecular mechanisms through which ASCs regulate angiogenesis are not well understood. Here, it was our goal to test the functional contribution of ASC-mediated extracellular matrix (ECM) remodeling on endothelial cell invasion. To isolate the effect of ECM-remodeling, ASCs were embedded within 3-D collagen type I hydrogels and pre-cultured for 7 days; controls were not pre-cultured. A confluent monolayer of human umbilical vein endothelial cells (HUVECs) was seeded on top and its invasion into the underlying hydrogel was analyzed. Without pre-culture, ASCs inhibited vascular sprouting by stabilizing the endothelium. In contrast, 7 day pre-culture of ASCs drastically increased invasion by HUVECs. This effect was largely mediated by proteolytic ECM degradation by ASC-derived matrix metalloproteinases (MMPs) rather than vascular endothelial growth factor (VEGF), as our results indicated that blockade of MMPs, but not VEGF, inhibited endothelial sprouting. Collectively, these data suggest that the angiogenic capability of ASCs is modulated by their proteolytic remodeling of the ECM, opening new avenues for pro- and anti-angiogenic therapies.
Collapse
Affiliation(s)
- Young Hye Song
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY
| | - Seung Hee Shon
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY
| | - Mengrou Shan
- School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY
| | - Abraham Stroock
- School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY
- Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY
- Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY
| |
Collapse
|
31
|
Amin M, Pushpakumar S, Muradashvili N, Kundu S, Tyagi SC, Sen U. Regulation and involvement of matrix metalloproteinases in vascular diseases. FRONT BIOSCI-LANDMRK 2016; 21:89-118. [PMID: 26709763 PMCID: PMC5462461 DOI: 10.2741/4378] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc dependent endopeptidases whose main function is to degrade and deposit structural proteins within the extracellular matrix (ECM). A dysregulation of MMPs is linked to vascular diseases. MMPs are classified into collagenases, gelatinases, membrane-type, metalloelastase, stromelysins, matrilysins, enamelysins, and unclassified subgroups. The production of MMPs is stimulated by factors such as oxidative stress, growth factors and inflammation which lead to its up- or down-regulation with subsequent ECM remodeling. Normally, excess activation of MMPs is controlled by their endogenous inhibitors, tissue inhibitors of metalloproteinases (TIMPs). An imbalance of MMPs and TIMPs has been implicated in hypertension, atherosclerotic plaque formation and instability, aortic aneurysms and varicose vein wall remodeling. Also, recent evidence suggests epigenetic regulation of some MMPs in angiogenesis and atherosclerosis. Over the years, pharmacological inhibitors of MMPs have been used to modify or prevent the development of the disease with some success. In this review, we discuss recent advances in MMP biology, and their involvement in the manifestation of vascular disease.
Collapse
Affiliation(s)
- Matthew Amin
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY-40202
| | - Sathnur Pushpakumar
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY-40202
| | - Nino Muradashvili
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY-40202
| | - Sourav Kundu
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY-40202
| | - Suresh C Tyagi
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY-40202
| | - Utpal Sen
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY-40202,
| |
Collapse
|
32
|
Shin JU, Lee WJ, Tran TN, Jung I, Lee JH. Hsp70 Knockdown by siRNA Decreased Collagen Production in Keloid Fibroblasts. Yonsei Med J 2015; 56:1619-26. [PMID: 26446645 PMCID: PMC4630051 DOI: 10.3349/ymj.2015.56.6.1619] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 12/30/2014] [Accepted: 01/03/2015] [Indexed: 11/27/2022] Open
Abstract
PURPOSE There are currently no consistently effective treatments for the excessive collagen produced by keloid fibroblasts. Previously, we reported that heat shock protein 70 (Hsp70) is up-regulated in keloid fibroblasts and keloid tissue. We, therefore, investigated whether Hsp70 is related to excessive collagen production in keloid fibroblasts. MATERIALS AND METHODS We inhibited Hsp70 in keloid fibroblasts by RNA interference and examined the resulting collagen expression. Thus, we selected small interfering RNAs (siRNAs) specific for human Hsp70, transfected them into keloid fibroblasts, and evaluated the resulting phenotypes and protein production using real-time polymerase chain reaction (PCR), Western blot, and a collagen assay. RESULTS The siRNAs dramatically suppressed Hsp70 mRNA expression, resulting in a decrease in collagen production in the keloid fibroblasts compared with controls. The siRNAs did not influence the viability of the keloid fibroblasts. CONCLUSION Hsp70 overexpression likely plays an important role in the excessive collagen production by keloid fibroblasts. RNA interference has therapeutic potential for the treatment of keloids.
Collapse
Affiliation(s)
- Jung U Shin
- Department of Dermatology, Severance Hospital, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Won Jai Lee
- Department of Plastic and Reconstructive Surgery, Severance Hospital, Institute for Human Tissue Restoration, Yonsei University College of Medicine, Seoul, Korea
| | - Thanh-Nga Tran
- Department of Dermatology and Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Inhee Jung
- Department of Dermatology, Severance Hospital, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Ju Hee Lee
- Department of Dermatology, Severance Hospital, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
33
|
Ohkawara H, Ikeda K, Ogawa K, Takeishi Y. MEMBRANE TYPE 1-MATRIX METALLOPROTEINASE (MT1-MMP) IDENTIFIED AS A MULTIFUNCTIONAL REGULATOR OF VASCULAR RESPONSES. Fukushima J Med Sci 2015; 61:91-100. [PMID: 26370683 DOI: 10.5387/fms.2015-15] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Membrane type 1-matrix metalloproteinase (MT1-MMP) functions as a signaling molecules in addition to a transmembrane metalloprotease, which degrades interstitial collagens and extracellular matrix components. This review focuses on the multifunctional roles of MT1-MMP as a signaling molecule in vascular responses to pro-atherosclerotic stimuli in the pathogenesis of cardiovascular diseases. First, the lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1)-MT1-MMP signaling axis contributes to endothelial dysfunction, which is mediated via small GTP-binding protein RhoA and Rac1 activation. Second, MT1-MMP plays a crucial role in reactive oxygen species (ROS) generation through the activation of receptor for advanced glycation end products (AGEs) in smooth muscle cells, indicating that MT1-MMP may be a therapeutic target for diabetic vascular complications. Third, MT1-MMP is involved in RhoA/Rac1 activation and Ca(2+) signaling in the mechanism of thrombin-stimulated endothelial dysfunction and oxidant stress. Fourth, the inhibition of the MT1-MMP/Akt signaling pathway may be an attractive strategy for treating endothelial disordered hemostasis in the development of vascular diseases linked to TNF-α-induced inflammation. Fifth, MT1-MMP through RAGE induced RhoA/Rac1 activation and tissue factor protein upregulation through NF-κB phosphorylation in endothelial cells stimulated by high-mobility group box-1, which plays a key role in the systemic inflammation. These findings suggest that the MT1-MMP-mediated signaling axis may be a promising target for treating atherosclerosis and subsequent cardiovascular diseases.
Collapse
Affiliation(s)
- Hiroshi Ohkawara
- Department of Cardiology and Hematology, Fukushima Medical University
| | | | | | | |
Collapse
|
34
|
Valacca C, Tassone E, Mignatti P. TIMP-2 Interaction with MT1-MMP Activates the AKT Pathway and Protects Tumor Cells from Apoptosis. PLoS One 2015; 10:e0136797. [PMID: 26331622 PMCID: PMC4558019 DOI: 10.1371/journal.pone.0136797] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 08/07/2015] [Indexed: 02/07/2023] Open
Abstract
Membrane-type 1 matrix metalloproteinase (MT1-MMP), a transmembrane proteinase with an extracellular catalytic domain and a short cytoplasmic tail, degrades a variety of extracellular matrix (ECM) components. In addition, MT1-MMP activates intracellular signaling through proteolysis-dependent and independent mechanisms. We have previously shown that binding of tissue inhibitor of metalloproteinases-2 (TIMP-2) to MT1-MMP controls cell proliferation and migration, as well as tumor growth in vivo by activating the Ras—extracellular signal regulated kinase-1 and -2 (ERK1/2) pathway through a mechanism that requires the cytoplasmic but not the proteolytic domain of MT1-MMP. Here we show that in MT1-MMP expressing cells TIMP-2 also induces rapid and sustained activation of AKT in a dose- and time-dependent manner and by a mechanism independent of the proteolytic activity of MT1-MMP. Fibroblast growth factor receptor-1 mediates TIMP-2 induction of ERK1/2 but not of AKT activation; however, Ras activation is necessary to transduce the TIMP-2-activated signal to both the ERK1/2 and AKT pathways. ERK1/2 and AKT activation by TIMP-2 binding to MT1-MMP protects tumor cells from apoptosis induced by serum starvation. Conversely, TIMP-2 upregulates apoptosis induced by three-dimensional type I collagen in epithelial cancer cells. Thus, TIMP-2 interaction with MT1-MMP provides tumor cells with either pro- or anti-apoptotic signaling depending on the extracellular environment and apoptotic stimulus.
Collapse
Affiliation(s)
- Cristina Valacca
- Department of Cardiothoracic Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Evelyne Tassone
- Department of Cardiothoracic Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Paolo Mignatti
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
- Department of Cell Biology, New York University School of Medicine, New York University School of Medicine, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
35
|
Al-Alem L, Curry TE. Ovarian cancer: involvement of the matrix metalloproteinases. Reproduction 2015; 150:R55-64. [PMID: 25918438 DOI: 10.1530/rep-14-0546] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 04/24/2015] [Indexed: 12/12/2022]
Abstract
Ovarian cancer is the leading cause of death from gynecologic malignancies. One of the reasons for the high mortality rate associated with ovarian cancer is its late diagnosis, which often occurs after the cancer has metastasized throughout the peritoneal cavity. Cancer metastasis is facilitated by the remodeling of the extracellular tumor matrix by a family of proteolytic enzymes known as the matrix metalloproteinases (MMPs). There are 23 members of the MMP family, many of which have been reported to be associated with ovarian cancer. In the current paradigm, ovarian tumor cells and the surrounding stromal cells stimulate the synthesis and/or activation of various MMPs to aid in tumor growth, invasion, and eventual metastasis. The present review sheds light on the different MMPs in the various types of ovarian cancer and on their impact on the progression of this gynecologic malignancy.
Collapse
Affiliation(s)
- Linah Al-Alem
- Department of Obstetrics and GynecologyUniversity of Kentucky Medical Center, 800 Rose Street, Room C355, Lexington, Kentucky 40536-0293, USA
| | - Thomas E Curry
- Department of Obstetrics and GynecologyUniversity of Kentucky Medical Center, 800 Rose Street, Room C355, Lexington, Kentucky 40536-0293, USA
| |
Collapse
|
36
|
Inflammatory mediators in vascular disease: identifying promising targets for intracranial aneurysm research. Mediators Inflamm 2015; 2015:896283. [PMID: 25922566 PMCID: PMC4397479 DOI: 10.1155/2015/896283] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 03/18/2015] [Accepted: 03/19/2015] [Indexed: 12/21/2022] Open
Abstract
Inflammatory processes are implicated in many diseases of the vasculature and have been shown to play a key role in the formation of intracranial aneurysms (IAs). Although the specific mechanisms underlying these processes have been thoroughly investigated in related pathologies, such as atherosclerosis, there remains a paucity of information regarding the immunopathology of IA. Cells such as macrophages and lymphocytes and their effector molecules have been suggested to be players in IA, but their specific interactions and the role of other components of the inflammatory response have yet to be determined. Drawing parallels between the pathogenesis of IA and other vascular disorders could provide a roadmap for developing a mechanistic understanding of the immunopathology of IA and uncovering useful targets for therapeutic intervention. Future research should address the presence and function of leukocyte subsets, mechanisms of leukocyte recruitment and activation, and the role of damage-associated molecular patterns in IA.
Collapse
|
37
|
Wiechmann AF, Ceresa BP, Howard EW. Diurnal variation of tight junction integrity associates inversely with matrix metalloproteinase expression in Xenopus laevis corneal epithelium: implications for circadian regulation of homeostatic surface cell desquamation. PLoS One 2014; 9:e113810. [PMID: 25412440 PMCID: PMC4239109 DOI: 10.1371/journal.pone.0113810] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 10/31/2014] [Indexed: 01/08/2023] Open
Abstract
Background and Objectives The corneal epithelium provides a protective barrier against pathogen entrance and abrasive forces, largely due to the intercellular junctional complexes between neighboring cells. After a prescribed duration at the corneal surface, tight junctions between squamous surface cells must be disrupted to enable them to desquamate as a component of the tissue homeostatic renewal. We hypothesize that matrix metalloproteinase (MMPs) are secreted by corneal epithelial cells and cleave intercellular junctional proteins extracellularly at the epithelial surface. The purpose of this study was to examine the expression of specific MMPs and tight junction proteins during both the light and dark phases of the circadian cycle, and to assess their temporal and spatial relationships in the Xenopus laevis corneal epithelium. Methodology/Principal Findings Expression of MMP-2, tissue inhibitor of MMP-2 (TIMP-2), membrane type 1-MMP (MT1-MMP) and the tight junction proteins occludin and claudin-4 were examined by confocal double-label immunohistochemistry on corneas obtained from Xenopus frogs at different circadian times. Occludin and claudin-4 expression was generally uniformly intact on the surface corneal epithelial cell lateral membranes during the daytime, but was frequently disrupted in small clusters of cells at night. Concomitantly, MMP-2 expression was often elevated in a mosaic pattern at nighttime and associated with clusters of desquamating surface cells. The MMP-2 binding partners, TIMP-2 and MT1-MMP were also localized to surface corneal epithelial cells during both the light and dark phases, with TIMP-2 tending to be elevated during the daytime. Conclusions/Significance MMP-2 protein expression is elevated in a mosaic pattern in surface corneal epithelial cells during the nighttime in Xenopus laevis, and may play a role in homeostatic surface cell desquamation by disrupting intercellular junctional proteins. The sequence of MMP secretion and activation, tight junction protein cleavage, and subsequent surface cell desquamation and renewal may be orchestrated by nocturnal circadian signals.
Collapse
Affiliation(s)
- Allan F. Wiechmann
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- * E-mail:
| | - Brian P. Ceresa
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Eric W. Howard
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| |
Collapse
|
38
|
Shin JU, Gantsetseg D, Jung JY, Jung I, Shin S, Lee JH. Comparison of non-ablative and ablative fractional laser treatments in a postoperative scar study. Lasers Surg Med 2014; 46:741-9. [DOI: 10.1002/lsm.22297] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Jung U Shin
- Department of Dermatology and Cutaneous Biology Research Institute; Yonsei University College of Medicine; Seoul South Korea
| | - Dorjsuren Gantsetseg
- Department of Dermatology and Cutaneous Biology Research Institute; Yonsei University College of Medicine; Seoul South Korea
| | - Jin Young Jung
- Yeouido Oracle Cosmetic & Dermatologic Surgery Clinic; Seoul South Korea
| | - Inhee Jung
- Department of Dermatology and Cutaneous Biology Research Institute; Yonsei University College of Medicine; Seoul South Korea
| | - Sungsik Shin
- Department of Dermatology and Cutaneous Biology Research Institute; Yonsei University College of Medicine; Seoul South Korea
| | - Ju Hee Lee
- Department of Dermatology and Cutaneous Biology Research Institute; Yonsei University College of Medicine; Seoul South Korea
| |
Collapse
|
39
|
Zaleska M, Olszewski WL, Durlik M, Miller NE. Signaling proteins are represented in tissue fluid/lymph from soft tissues of normal human legs at concentrations different from serum. Lymphat Res Biol 2014; 11:203-10. [PMID: 24364843 DOI: 10.1089/lrb.2013.0014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The mobile intercellular fluid flowing to and in the lymphatics contains filtered plasma products and substances synthesized and excreted by tissue cells. Among them are signaling proteins such as cytokines, chemokines, enzymes, and growth factors. They act locally in autocrine and paracrine systems regulating cell metabolism, proliferation, and formation of the ground matrix. They play an immunoregulatory role in infections, wound healing, and tumor cell growth. METHODS AND RESULTS In this study we measured the concentration of selected cytokines, chemokines, tissue enzymes, and growth factors in tissue fluid/lymph drained from normal human leg soft tissues. Legs exposed to infections and trauma often result in development of lymphedema. Lymph was drained from superficial calf lymphatics using microsurgical techniques. Our studies showed generally higher concentrations of cytokines, chemokines, enzymes, and growth factors in lymph than in serum. The total protein L/S ratio was 0.22, whereas that of various lymph signaling proteins ranged between 1 and 10. CONCLUSIONS This indicates that in addition to proteins filtered from blood, local cells contribute to lymph concentration by own production, depending on the actual cell requirement. Moreover, there were major individual differences of lymph levels with simultaneous stable serum levels. This suggests existence of a local autonomous regulatory humoral mechanism in tissues, not reflected in serum.
Collapse
Affiliation(s)
- Marzanna Zaleska
- 1 Department of Surgical Research, Transplantation and Epigenetics, Medical Research Center , Polish Academy of Sciences, Warsaw, Poland
| | | | | | | |
Collapse
|
40
|
Kwok HF, Botkjaer KA, Tape CJ, Huang Y, McCafferty J, Murphy G. Development of a 'mouse and human cross-reactive' affinity-matured exosite inhibitory human antibody specific to TACE (ADAM17) for cancer immunotherapy. Protein Eng Des Sel 2014; 27:179-90. [PMID: 24769623 DOI: 10.1093/protein/gzu010] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
We previously showed that a human anti-TACE antibody, D1(A12), is a potent inhibitor of TNF-α converting enzyme (TACE) ectodomain proteolysis and has pharmacokinetic properties suitable for studies of the inhibition of TACE-dependent growth factor shedding in relation to possible therapeutic applications. However, the lack of murine TACE immunoreactivity limits pre-clinical in vivo studies to human xenograft models which are poor analogies to in situ pathology and are not considered clinically predictive. Here, to overcome these limitations, we set out to develop a 'mouse and human cross-reactive' specific anti-TACE antibody. We first re-investigated the originally selected anti-TACE ectodomain phage-display clones, and isolated a lead 'mouse-human cross-reactive' anti-TACE scFv, clone A9. We reformatted scFv-A9 into an IgG2 framework for comprehensive biochemical and cellular characterization and further demonstrated that A9 is an exosite TACE inhibitor. However, surface plasmon resonance analysis and quenched-fluorescent (QF) peptide assay indicated that IgG reformatting of A9 caused low binding affinity and an 80-fold reduction in TACE ectodomain inhibition, severely limiting its efficacy. To address this, we constructed second generation phage-display randomization libraries focused on the complementarity-determining region 3, and carried out affinity selections shuffling between human and mouse TACE ectodomain as antigen in addition to an off-rate selection to increase the chance of affinity improvement. The bespoke 'three-step' selections enabled a 100-fold affinity enhancement of A9 IgG, and also improved its IC50 in a QF peptide assay to 0.2 nM. In human and mouse cancer cell assays, matured A9 IgG showed significant cell-surface TACE inhibition as a monotherapy or combination therapy with chemotherapeutic agent. Collectively, these data suggest that we successfully developed an exosite inhibitor of TACE with sub-nanomolar affinity, which possesses both murine and human immunoreactive properties that can be used for in vivo application in murine pre-clinical cancer models.
Collapse
Affiliation(s)
- Hang Fai Kwok
- Proteases and Tumour Microenvironment Group, Department of Oncology, Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UK Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Kenneth A Botkjaer
- Proteases and Tumour Microenvironment Group, Department of Oncology, Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UK Department of Molecular Biology and Genetics, University of Aarhus, Denmark
| | - Christopher J Tape
- Proteases and Tumour Microenvironment Group, Department of Oncology, Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UK Present address: Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Yanchao Huang
- Proteases and Tumour Microenvironment Group, Department of Oncology, Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UK
| | - John McCafferty
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1QW, UK
| | - Gillian Murphy
- Proteases and Tumour Microenvironment Group, Department of Oncology, Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UK
| |
Collapse
|
41
|
Sudhakar YA, Verma RK, Pawar SC. Type IV collagen α1-chain noncollagenous domain blocks MMP-2 activation both in-vitro and in-vivo. Sci Rep 2014; 4:4136. [PMID: 24670518 PMCID: PMC3966261 DOI: 10.1038/srep04136] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 01/09/2014] [Indexed: 12/11/2022] Open
Abstract
α1(IV)NC1 inhibits angiogenesis by regulating MAPK activation, this biological function was partly attributed α1(IV)NC1 binding to α1β1-integrin. However, its potent antiangiogenic activity and the molecular targets of α1(IV)NC1 has not been investigated. In the present study, the regulation of MMP-2 activation by α1(IV)NC1 was evaluated. α1β1-integrin which is required for inhibition of angiogenesis is not playing a role in cellular invasion and inhibition of MMP-2 activation by α1(IV)NC1. We found that α1(IV)NC1 binds the CBD of MMP-2 and forming a stable complex that prevents activation of MMP-2. The antiangiogenic activity of α1(IV)NC1 is mediated, in part, by this binding activity. In addition, up-regulation of TIMP-2 by α1(IV)NC1 led to saturation of MT1-MMP binding sites, which in turn led to inhibition of MMP-2 activation. In-vivo studies using α1-integrin null-mice treated with higher doses of α1(IV)NC1 showed integrin independent inhibition of tumor growth and active-MMP-2, without affecting MMP-9, MMP-7 and angiostatin.
Collapse
Affiliation(s)
- Yakkanti Akul Sudhakar
- 1] Cell Signaling Laboratory, Bioscience Division, Center for Cancer and Metabolism, SRI International, Menlo Park, CA 94025, USA [2] Cell Signaling and Tumor Angiogenesis Laboratory, Department of Genetics, Boys Town National Research Hospital, Omaha, NE 68131, USA
| | - Raj Kumar Verma
- Irma Lerma Rangel College of Pharmacy, Texas A&M Health Science Center, Kingsville, Texas 78363, USA
| | - Smita C Pawar
- Department of Genetics, Osmania University, Hyderabad, AP 500007, India
| |
Collapse
|
42
|
Shiryaev SA, Remacle AG, Golubkov VS, Ingvarsen S, Porse A, Behrendt N, Cieplak P, Strongin AY. A monoclonal antibody interferes with TIMP-2 binding and incapacitates the MMP-2-activating function of multifunctional, pro-tumorigenic MMP-14/MT1-MMP. Oncogenesis 2013; 2:e80. [PMID: 24296749 PMCID: PMC3940861 DOI: 10.1038/oncsis.2013.44] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 10/29/2013] [Indexed: 01/01/2023] Open
Abstract
Matrix metalloproteinases (MMPs) and, especially membrane type 1 (MT1)-MMP/MMP-14, are promising drug targets in malignancies. In contrast with multiple small-molecule and protein pan-inhibitors of MT1–MMP cleavage activity, the murine 9E8 monoclonal antibody targets the MMP-2-activating function of cellular MT1–MMP alone, rather than the general proteolytic activity and the pro-migratory function of MT1–MMP. Furthermore, the antibody does not interact in any detectable manner with other members of the membrane type (MT)-MMP family. The mechanism of this selectivity remained unknown. Using mutagenesis, binding and activity assays, and modeling in silico, we have demonstrated that the 9E8 antibody recognizes the MT-loop structure, an eight residue insertion that is specific for MT–MMPs and that is distant from the MT1–MMP active site. The binding of the 9E8 antibody to the MT-loop, however, prevents tissue inhibitor of metalloproteinases-2 (TIMP-2) association with MT1–MMP. As a result, the 9E8 antibody incapacitates the TIMP-2-dependent MMP-2-activating function alone rather than the general enzymatic activity of human MT1–MMP. The specific function of the 9E8 antibody we determined directly supports an essential, albeit paradoxical, role of the protein inhibitor (TIMP-2) in MMP-2 activation via a unique membrane-tethered mechanism. In this mechanism, the formation of a tri-molecular MT1–MMPTIMP-2MMP-2 complex is required for both the capture of the soluble MMP-2 proenzyme by cells and then its well-controlled conversion into the mature MMP-2 enzyme. In sum, understanding of the structural requirements for the 9E8 antibody specificity may pave the way for the focused design of the inhibitory antibodies against other individual MMPs.
Collapse
Affiliation(s)
- S A Shiryaev
- Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Niarakis A, Giannopoulou E, Ravazoula P, Panagiotopoulos E, Zarkadis IK, Aletras AJ. Detection of a latent soluble form of membrane type 1 matrix metalloprotease bound with tissue inhibitor of matrix metalloproteinases-2 in periprosthetic tissues and fluids from loose arthroplasty endoprostheses. FEBS J 2013; 280:6541-55. [PMID: 24112707 DOI: 10.1111/febs.12555] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Revised: 09/20/2013] [Accepted: 09/23/2013] [Indexed: 01/22/2023]
Abstract
Membrane type 1 matrix metalloproteinase (MT1-MMP) is implicated in pericellular proteolysis, and, together with tissue inhibitor of matrix metalloproteinases-2 (TIMP-2), in the activation of pro-matrix metalloproteinase-2 on the cell surface. It is expressed on the cell surface either activated or as a proenzyme. A soluble form of MT1-MMP (sMT1-MMP) has been previously identified in periprosthetic tissues and fluid of patients with loose arthroplasty endoprostheses. The aim of this study was to examine periprosthetic tissues and fluids from patients with loose arthroplasty endoprostheses, as well as tissues and fluids from patients with other disorders, for the presence of sMT1-MMP, and to investigate its activation state and possible role. With antibody against MT1-MMP, a protein with molecular mass of ~ 57 kDa was detected by western blotting in all samples tested, representing a soluble form of MT1-MMP, which cannot be ascribed to alternative splicing, as northern blotting showed only one transcript. With various biochemical methods, it was shown that this species occurs in a latent form bearing the N-terminal prodomain, and, additionally, it is bound to TIMP-2, which appeared to be bound via its C-terminal domain to a site different from the active site. Cell ELISA and immunohistochemical analysis revealed that, besides fibroblasts, all other cells, such as inflammatory, epithelial, endothelial, giant and cancer cells, express MT1-MMP on their plasma membrane as a proenzyme. Taking into account the proteolytic abilities of MT1-MMP, the latent sMT1-MMP-TIMP-2 complex could be considered as a new interstitial collagenase. However, the exact role, the production mechanism and the cell origin of this complex remain to be elucidated.
Collapse
Affiliation(s)
- Anna Niarakis
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | | | | | | | | | | |
Collapse
|
44
|
Membrane localization of membrane type 1 matrix metalloproteinase by CD44 regulates the activation of pro-matrix metalloproteinase 9 in osteoclasts. BIOMED RESEARCH INTERNATIONAL 2013; 2013:302392. [PMID: 23984338 PMCID: PMC3745902 DOI: 10.1155/2013/302392] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2013] [Revised: 06/22/2013] [Accepted: 06/22/2013] [Indexed: 12/02/2022]
Abstract
CD44, MT1-MMP, and MMP9 are implicated in the migration of osteoclast and bone resorption. This study was designed to determine the functional relationship between CD44 and MT1-MMP in the activation of pro-MMP9. We used osteoclasts isolated from wild-type and CD44-null mice. Results showed that MT1-MMP is present in multiple forms with a molecular mass ~63, 55, and 45 kDa in the membrane of wild-type osteoclasts. CD44-null osteoclasts demonstrated a 55 kDa active MT1-MMP form in the membrane and conditioned medium. It failed to activate pro-MMP9 because TIMP2 binds and inhibits this MT1-MMP (~55 kDa) in CD44-null osteoclasts. The role of MT1-MMP in the activation of pro-MMP9, CD44 expression, and migration was confirmed by knockdown of MT1-MMP in wild-type osteoclasts. Although knockdown of MMP9 suppressed osteoclast migration, it had no effects on MT1-MMP activity or CD44 expression. These results suggest that CD44 and MT1-MMP are directly or indirectly involved in the regulation of pro-MMP9 activation. Surface expression of CD44, membrane localization of MT1-MMP, and activation of pro-MMP9 are the necessary sequence of events in osteoclast migration.
Collapse
|
45
|
Li H, Daculsi R, Bareille R, Bourget C, Amedee J. uPA and MMP-2 were involved in self-assembled network formation in a two dimensional co-culture model of bone marrow stromal cells and endothelial cells. J Cell Biochem 2013; 114:650-7. [DOI: 10.1002/jcb.24407] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 09/21/2012] [Indexed: 11/11/2022]
|
46
|
Signaling determinants of glioma cell invasion. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 986:121-41. [PMID: 22879067 DOI: 10.1007/978-94-007-4719-7_7] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Tumor cell invasiveness is a critical challenge in the clinical management of glioma patients. In addition, there is accumulating evidence that current therapeutic modalities, including anti-angiogenic therapy and radiotherapy, can enhance glioma invasiveness. Glioma cell invasion is stimulated by both autocrine and paracrine factors that act on a large array of cell surface-bound receptors. Key signaling elements that mediate receptor-initiated signaling in the regulation of glioblastoma invasion are Rho family GTPases, including Rac, RhoA and Cdc42. These GTPases regulate cell morphology and actin dynamics and stimulate cell squeezing through the narrow extracellular spaces that are typical of the brain parenchyma. Transient attachment of cells to the extracellular matrix is also necessary for glioblastoma cell invasion. Interactions with extracellular matrix components are mediated by integrins that initiate diverse intracellular signalling pathways. Key signaling elements stimulated by integrins include PI3K, Akt, mTOR and MAP kinases. In order to detach from the tumor mass, glioma cells secrete proteolytic enzymes that cleave cell surface adhesion molecules, including CD44 and L1. Key proteases produced by glioma cells include uPA, ADAMs and MMPs. Increased understanding of the molecular mechanisms that control glioma cell invasion has led to the identification of molecular targets for therapeutic intervention in this devastating disease.
Collapse
|
47
|
HUA DONG, SHEN LI, XU LAN, JIANG ZHI, ZHOU YINGHUI, YUE AIHUAN, ZOU SHITAO, CHENG ZHIHONG, WU SHILIANG. Polypeptide N-acetylgalactosaminyltransferase 2 regulates cellular metastasis-associated behavior in gastric cancer. Int J Mol Med 2012; 30:1267-74. [PMID: 22992780 PMCID: PMC4042861 DOI: 10.3892/ijmm.2012.1130] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Accepted: 07/27/2012] [Indexed: 01/12/2023] Open
Abstract
Aberrant glycosylation of cell surface glycoprotein due to specific alterations of glycosyltransferase activity is usually associated with invasion and metastasis of cancer, particularly of gastric carcinomas. Polypeptide N-acetylgalactosaminyltransferase 2 (ppGalNAc-T2), which catalyzes initiation of mucin-type O-glycosylation, is also involved in tumor migration and invasion. However, a comprehensive understanding of how ppGalNAc-T2 correlates with the metastasic potential of human gastric cancer is not currently available. In the present study, ppGalNAc-T2 was detected in a variety of human poorly differentiated tumor cells, and expression appeared to be higher in SGC7901 gastric cancer cells. In addition, we investigated the potential effects of ppGalNAc-T2 on growth and metastasis-associated behavior in SGC7901 cells after stable transfection with ppGalNAc-T2 sense and antisense vectors. We found that cell proliferation, adhesion and invasion were decreased in ppGalNAc-T2 overexpressed cells but increased in ppGalNAc-T2 downregulated cells. Therefore, we attempted to clarify the mechanisms underlying the anti-metastatic activities of ppGalNAc-T2. Further investigation indicated that overexpression of ppGalNAc-T2 is involved in the inhibition of matrix metalloproteinase (MMP)-2 expression at both the protein and mRNA levels, which may be associated with ppGalNAc-T2 suppressing the expression of transforming growth factor (TGF)-β1. However, it did not exhibit any apparent correlation with MMP-14 expression levels. Our data show the effect of ppGalNAc-T2 on proliferation, adhesion or invasion of SGC7901 gastric cancer cells, suggesting that ppGalNAc-T2 may exert anti-proliferative and anti-metastatic activity through the decrease of MMP-2 and TGF-β1. These results indicate that ppGalNAc‑T2 may be used as a novel therapeutic target for human gastric cancer treatment.
Collapse
Affiliation(s)
- DONG HUA
- The Fourth Affiliated Hospital of Soochow University, Wuxi, Jiangsu
214062
| | - LI SHEN
- Department of Biochemistry and Mollecular Biology, School of Medicine,
Soochow University, Suzhou, Jiangsu 215123
- Department of Biochemistry and Molecular Biology, Hubei University of
Medicine, Shiyan, Hubei 442000, P.R. China
| | - LAN XU
- Department of Biochemistry and Mollecular Biology, School of Medicine,
Soochow University, Suzhou, Jiangsu 215123
| | - ZHI JIANG
- Department of Biochemistry and Mollecular Biology, School of Medicine,
Soochow University, Suzhou, Jiangsu 215123
| | - YINGHUI ZHOU
- Department of Biochemistry and Mollecular Biology, School of Medicine,
Soochow University, Suzhou, Jiangsu 215123
| | - AIHUAN YUE
- Department of Biochemistry and Mollecular Biology, School of Medicine,
Soochow University, Suzhou, Jiangsu 215123
| | - SHITAO ZOU
- Department of Biochemistry and Mollecular Biology, School of Medicine,
Soochow University, Suzhou, Jiangsu 215123
| | - ZHIHONG CHENG
- The Fourth Affiliated Hospital of Soochow University, Wuxi, Jiangsu
214062
| | - SHILIANG WU
- Department of Biochemistry and Mollecular Biology, School of Medicine,
Soochow University, Suzhou, Jiangsu 215123
| |
Collapse
|
48
|
Survivin, MMP-2, MT1-MMP, and TIMP-2: their impact on survival, implantation, and proliferation of endometriotic tissues. Virchows Arch 2012; 461:589-99. [DOI: 10.1007/s00428-012-1301-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Revised: 08/09/2012] [Accepted: 08/09/2012] [Indexed: 02/03/2023]
|
49
|
Diaz PS, Solar PA, Juica NE, Orihuela PA, Cardenas H, Christodoulides M, Vargas R, Velasquez LA. Differential expression of extracellular matrix components in the Fallopian tubes throughout the menstrual cycle. Reprod Biol Endocrinol 2012; 10:56. [PMID: 22897899 PMCID: PMC3489778 DOI: 10.1186/1477-7827-10-56] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Accepted: 08/08/2012] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND One of the unique characteristics of the female genital tract is the extensive tissue remodeling observed throughout the menstrual cycle. Multiple components of the extracellular matrix take part in this tissue rebuilding; however, the individual components involved have not been identified. METHODS In the present study, the expression of extracellular matrix proteins and selected matrix metalloproteinase (MMP) activities in Fallopian tubes (FT) throughout the menstrual cycle were examined by PCR array, immunocytochemistry, zymography and bioinformatics. RESULTS Of the eighty-four genes analyzed, eighty-three were expressed in the FT during at least one stage of the menstrual cycle. We observed a significant increase (>/=2-fold) in ADAMTS1, ADAMTS13, COL7A1, MMP3, MMP9, PECAM1, and THBS3 in the periovulatory phase compared to the follicular phase. Meanwhile, we observed a significant decrease (>/= 2-fold) in COL7A1, ICAM1, ITGA8, MMP16, MMP9, CLEC3B, SELE and TIMP2 in the lutheal phase compared to the periovulatory phase. Immunocytochemistry showed that MMP-3 and MMP-9 were localized in the endosalpinx during all phases of the menstrual cycle. Gelatin zymograms detected non-cycle-dependent protease activity. CONCLUSIONS Several extracellular matrix components were regulated throughout the menstrual cycle in a cyclic pattern, suggesting a possible steroid regulation and a role in tissue remodeling and FT functions.
Collapse
Affiliation(s)
- Patricia S Diaz
- Centro para el Desarrollo de la Nanociencia y Nanotecnología, Universidad de Santiago, Santiago, Chile
- Laboratorio de Inmunología de la Reproducción, Facultad de Química y Biología, Universidad de Santiago, Santiago, Chile
| | - Paula A Solar
- Centro para el Desarrollo de la Nanociencia y Nanotecnología, Universidad de Santiago, Santiago, Chile
- Center for integrative medicine and innovative sciences (CIMIS), Facultad de Medicina, Universidad Andrés Bello, Santiago, Chile
| | - Natalia E Juica
- Centro para el Desarrollo de la Nanociencia y Nanotecnología, Universidad de Santiago, Santiago, Chile
- Center for integrative medicine and innovative sciences (CIMIS), Facultad de Medicina, Universidad Andrés Bello, Santiago, Chile
| | - Pedro A Orihuela
- Centro para el Desarrollo de la Nanociencia y Nanotecnología, Universidad de Santiago, Santiago, Chile
- Laboratorio de Inmunología de la Reproducción, Facultad de Química y Biología, Universidad de Santiago, Santiago, Chile
| | - Hugo Cardenas
- Centro para el Desarrollo de la Nanociencia y Nanotecnología, Universidad de Santiago, Santiago, Chile
- Laboratorio de Inmunología de la Reproducción, Facultad de Química y Biología, Universidad de Santiago, Santiago, Chile
| | - Myron Christodoulides
- Neisseria Research Group, Sir Henry Wellcome Laboratories, Division of Infection, Inflammation and Immunity, University of Southampton Medical School, Southampton, SO16 6YD, England, UK
| | - Renato Vargas
- Servicio de Ginecología y Obstetricia, Hospital San José, Santiago, Chile
| | - Luis A Velasquez
- Centro para el Desarrollo de la Nanociencia y Nanotecnología, Universidad de Santiago, Santiago, Chile
- Center for integrative medicine and innovative sciences (CIMIS), Facultad de Medicina, Universidad Andrés Bello, Santiago, Chile
| |
Collapse
|
50
|
Dao Thi MU, Trocmé C, Montmasson MP, Fanchon E, Toussaint B, Tracqui P. Investigating metalloproteinases MMP-2 and MMP-9 mechanosensitivity to feedback loops involved in the regulation of in vitro angiogenesis by endogenous mechanical stresses. Acta Biotheor 2012; 60:21-40. [PMID: 22271286 DOI: 10.1007/s10441-012-9147-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Accepted: 01/11/2012] [Indexed: 12/31/2022]
Abstract
Angiogenesis is a complex morphogenetic process regulated by growth factors, but also by the force balance between endothelial cells (EC) traction stresses and extracellular matrix (ECM) viscoelastic resistance. Studies conducted with in vitro angiogenesis assays demonstrated that decreasing ECM stiffness triggers an angiogenic switch that promotes organization of EC into tubular cords or pseudo-capillaries. Thus, mechano-sensitivity of EC with regard to proteases secretion, and notably matrix metalloproteinases (MMPs), should likely play a pivotal role in this switching mechanism. While most studies analysing strain regulation of MMPs used cell cultured on stretched membranes, this work focuses on MMP expression during self-assembly of EC into capillary-like structures within fibrin gels, i.e. on conditions that mimics more closely the in vivo cellular mechanical microenvironment. The activity of MMP-2 and MMP-9, two MMPs that have a pivotal role in capillaries formation, has been monitored in pace with the progressive elongation of EAhy926 cells that takes place during the emergence of cellular cords. We found an increase of the zymogen proMMP-2 that correlates with the initial stages of EC cords formation. However, MMP-2 was not detected. ProMMP-9 secretion decreased, with levels of MMP-9 kept at a rather low value. In order to analyse more precisely the observed differences of EAhy926 response on fibrin and plastic substrates, we proposed a theoretical model of the mechano-regulation of proMMP-2 activation in the presence of type 2 tissue inhibitor of MMPs (TIMP-2). Using association/dissociation rates experimentally reported for this enzymatic network, the model adequately describes the synergism of proMMP-2 and TIMP-2 strain activation during pseudo-capillary morphogenesis. All together, these results provide a first step toward a systems biology approach of angiogenesis mechano-regulation by cell-generated extracellular stresses and strains.
Collapse
Affiliation(s)
- Minh-Uyen Dao Thi
- Faculté de Médecine de Grenoble, DyCTiM team, UJF-Grenoble, CNRS, Laboratoire TIMC-IMAG UMR, France
| | | | | | | | | | | |
Collapse
|