1
|
Ghareeb H, Yi Li C, Shenoy A, Rotenberg N, Shifman JM, Katoh T, Sagi I, Suga H, Metanis N. Mirror-Image Random Nonstandard Peptides Integrated Discovery (MI-RaPID) Technology Yields Highly Stable and Selective Macrocyclic Peptide Inhibitors for Matrix Metallopeptidase 7. Angew Chem Int Ed Engl 2025; 64:e202414256. [PMID: 39215490 PMCID: PMC11833282 DOI: 10.1002/anie.202414256] [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] [Academic Contribution Register] [Received: 07/28/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/04/2024]
Abstract
Matrix metallopeptidase 7 (MMP7) plays a crucial role in cancer metastasis and progression, making it an attractive target for therapeutic development. However, the development of selective MMP7 inhibitors is challenging due to the conservation of active sites across various matrix metalloproteinases (MMPs). Here, we have developed mirror-image random nonstandard peptides integrated discovery (MI-RaPID) technology to discover innate protease-resistant macrocyclic peptides that specifically bind to and inhibit human MMP7. One identified macrocyclic peptide against D-MMP7, termed D20, was synthesized in its mirror-image form, D'20, consisting of 12 D-amino acids, one cyclic β-amino acid, and a thioether bond. Notably, it potently inhibited MMP7 with an IC50 value of 90 nM, and showed excellent selectivity over other MMPs with similar substrate specificity. Moreover, D'20 inhibited the migration of pancreatic cell line CFPAC-1, but had no effect on the cell proliferation and viability. D'20 exhibited excellent stability in human serum, as well as in simulated gastric and intestinal fluids. This study highlights that MI-RaPID technology can serve as a powerful tool to develop in vivo stable macrocyclic peptides for therapeutic applications.
Collapse
Affiliation(s)
- Hiba Ghareeb
- Institute of ChemistryThe Center for Nanoscience and NanotechnologyCasali Center of Applied ChemistryThe Hebrew University of JerusalemJerusalem9190401Israel
| | - Choi Yi Li
- Department of ChemistryGraduate School of ScienceThe University of TokyoTokyo113-0033 Japan
| | - Anjana Shenoy
- Department of Immunology and Regenerative BiologyWeizmann Institute of ScienceRehovotIL76100Israel
| | - Naama Rotenberg
- Department of Biological ChemistryThe Alexander Silverman Institute of Life ScienceThe Hebrew University of JerusalemJerusalem9190401Israel
| | - Julia M. Shifman
- Department of Biological ChemistryThe Alexander Silverman Institute of Life ScienceThe Hebrew University of JerusalemJerusalem9190401Israel
| | - Takayuki Katoh
- Department of ChemistryGraduate School of ScienceThe University of TokyoTokyo113-0033 Japan
| | - Irit Sagi
- Department of Immunology and Regenerative BiologyWeizmann Institute of ScienceRehovotIL76100Israel
| | - Hiroaki Suga
- Department of ChemistryGraduate School of ScienceThe University of TokyoTokyo113-0033 Japan
| | - Norman Metanis
- Institute of ChemistryThe Center for Nanoscience and NanotechnologyCasali Center of Applied ChemistryThe Hebrew University of JerusalemJerusalem9190401Israel
| |
Collapse
|
2
|
Deng M, Tang C, Yin L, Jiang Y, Huang Y, Feng Y, Chen C. Clinical and omics biomarkers in osteoarthritis diagnosis and treatment. J Orthop Translat 2025; 50:295-305. [PMID: 39911590 PMCID: PMC11795539 DOI: 10.1016/j.jot.2024.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 08/20/2024] [Revised: 11/03/2024] [Accepted: 12/09/2024] [Indexed: 02/07/2025] Open
Abstract
Osteoarthritis (OA) is a prevalent degenerative joint disease that significantly impacts the quality of life for hundreds of millions, and is a major cause of disability. Despite this, diagnostic and therapeutic options for OA are still limited. With advances in molecular biology, an increasing number of OA biomarkers have been identified, which not only enhances our understanding of OA pathogenesis, but also offers new approaches for OA diagnosis and treatment. This review discussed the research progress on traditional OA biomarkers, and analyzed the application of various omics, including genomics, transcriptomics, proteomics, and metabolomics, in the diagnosis and treatment of OA. Furthermore, we explored how integrating multi-omics methods can reveal interactions among different biomolecules and their roles in the development of OA. This emerging interdisciplinary approach not only provides a more comprehensive understanding of the fundamental biological characteristics of OA, but also aids in identifying new integrated biomarkers, thereby allowing for more accurate predictions of disease progression and treatment responses. The identification and development of biomarkers offer new perspectives in understanding OA, enhancing the specificity and sensitivity of biological diagnostic markers, providing a basis for the design of targeted drugs, and ultimately advancing the development of precision diagnosis and treatment strategies in clinical OA. This study provides an overview of both commonly used and emerging biomarkers of OA which is beneficial for a more accurate, timely, effective clinical diagnosis and treatment for OA.
Collapse
Affiliation(s)
- Muhai Deng
- College of Medical Informatics, Chongqing Medical University, Chongqing, 400016, China
| | - Cong Tang
- College of Medical Informatics, Chongqing Medical University, Chongqing, 400016, China
| | - Li Yin
- Department of Orthopaedics, General Hospital of Western Theater Command, Chengdu, 610083, China
| | - Yunsheng Jiang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yang Huang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Yong Feng
- Department of Orthopedic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing, 400014, China
| | - Cheng Chen
- College of Medical Informatics, Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
3
|
Noddeland HK, Canbay V, Lind M, Savickas S, Jensen LB, Petersson K, Malmsten M, Koch J, Auf dem Keller U, Heinz A. Matrix metalloproteinase landscape in the imiquimod-induced skin inflammation mouse model. Biochimie 2024; 226:99-106. [PMID: 38513823 DOI: 10.1016/j.biochi.2024.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/09/2024] [Revised: 02/29/2024] [Accepted: 03/18/2024] [Indexed: 03/23/2024]
Abstract
Inflammation and autoimmunity are known as central processes in many skin diseases, including psoriasis. It is therefore important to develop pre-clinical models that describe disease-related aspects to enable testing of pharmaceutical drug candidates and formulations. A widely accepted pre-clinical model of psoriasis is the imiquimod (IMQ)-induced skin inflammation mouse model, where topically applied IMQ provokes local skin inflammation. In this study, we investigated the abundance of a subset of matrix metalloproteinases (MMPs) in skin from mice with IMQ-induced skin inflammation and skin from naïve mice using targeted proteomics. Our findings reveal a significant increase in the abundance of MMP-2, MMP-7, MMP-8, and MMP-13 after treatment with IMQ compared to the control skin, while MMP-3, MMP-9, and MMP-10 were exclusively detected in the IMQ-treated skin. The increased abundance and broader representation of MMPs in the IMQ-treated skin provide valuable insight into the pathophysiology of skin inflammation in the IMQ model, adding to previous studies on cytokine levels using conventional immunochemical methods. Specifically, the changes in the MMP profiles observed in the IMQ-treated skin resemble the MMP patterns found in skin lesions of individuals with psoriasis. Ultimately, the differences in MMP abundance under IMQ-induced inflammation as compared to non-inflamed control skin can be exploited as a model to investigate drug efficacy or performance of drug delivery systems.
Collapse
Affiliation(s)
- Heidi Kyung Noddeland
- LEO Foundation Center for Cutaneous Drug Delivery, Department of Pharmacy, University of Copenhagen, 2100, Copenhagen, Denmark; Explorative Formulation & Technologies, CMC Design and Development, LEO Pharma A/S, 2750, Ballerup, Denmark
| | - Vahap Canbay
- Technical University of Denmark, Department of Biotechnology and Biomedicine, 2800, Kongens Lyngby, Denmark
| | - Marianne Lind
- Explorative Formulation & Technologies, CMC Design and Development, LEO Pharma A/S, 2750, Ballerup, Denmark
| | - Simonas Savickas
- Technical University of Denmark, Department of Biotechnology and Biomedicine, 2800, Kongens Lyngby, Denmark
| | - Louise Bastholm Jensen
- Explorative Formulation & Technologies, CMC Design and Development, LEO Pharma A/S, 2750, Ballerup, Denmark
| | - Karsten Petersson
- Explorative Formulation & Technologies, CMC Design and Development, LEO Pharma A/S, 2750, Ballerup, Denmark
| | - Martin Malmsten
- LEO Foundation Center for Cutaneous Drug Delivery, Department of Pharmacy, University of Copenhagen, 2100, Copenhagen, Denmark; Department of Physical Chemistry 1, University of Lund, SE-22100, Lund, Sweden
| | - Janne Koch
- Translational Sciences, Research and Early Development, LEO Pharma A/S, 2750, Ballerup, Denmark
| | - Ulrich Auf dem Keller
- Technical University of Denmark, Department of Biotechnology and Biomedicine, 2800, Kongens Lyngby, Denmark; ETH Zürich, Department of Biology, Institute of Molecular Health Sciences, 8093, Zürich, Switzerland
| | - Andrea Heinz
- LEO Foundation Center for Cutaneous Drug Delivery, Department of Pharmacy, University of Copenhagen, 2100, Copenhagen, Denmark.
| |
Collapse
|
4
|
Hu HM, Lee HL, Liu CJ, Hsieh YH, Chen YS, Hsueh KC. Loss of MTA2-mediated downregulation of PTK7 inhibits hepatocellular carcinoma metastasis progression by modulating the FAK-MMP7 axis. ENVIRONMENTAL TOXICOLOGY 2024; 39:1897-1908. [PMID: 38050825 DOI: 10.1002/tox.24073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 09/17/2023] [Revised: 11/14/2023] [Accepted: 11/20/2023] [Indexed: 12/07/2023]
Abstract
The expression of metastasis tumor-associated protein 2 (MTA2) and protein tyrosine kinase 7 (PTK7) is associated with hepatocellular carcinoma (HCC) progression. However, the functional effect and mechanism through which MTA2 regulates PTK7-mediated HCC progression remains unclear. Here, we found that MTA2 knockdown significantly down-regulated PTK7 expression in HCC cells (SK-Hep-1 and PLC/PRF/5). Data from the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases show that the PTK7 expression level was higher in HCC tissues than in normal liver tissues. In HCC patients, the PTK7 expression level clearly correlated with tumor stage and grade, lower overall survival (OS) correlated positively with MTA2 level, and PTK7 expression acted as a downstream factor for MTA2 expression. In addition, matrix metalloproteinase 7 (MMP7) expression was closely regulated by PTK7, and the mRNA and protein expression levels of MTA2 and PTK7 correlated positively with lower OS. MMP7 downregulation by PTK7 knockdown clearly decreased the migration and invasion abilities of HCC cells. In HCC cells, recombinant human MMP7 reversed the PTK7 knockdown-induced suppression of migration and invasion. Furthermore, deactivation of FAK using siFAK or FAK inhibitor (PF-573228, PF) synergistically contributed to PTK7 knockdown-inhibited FAK activity, MMP7 expression, and the migration and invasion abilities of HCC cells. Collectively, our findings show that PTK7 mediates HCC progression by regulating the MTA2-FAK-MMP7 axis and may be a diagnostic value for HCC patients.
Collapse
Affiliation(s)
- Huang-Ming Hu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Medical University, Kaohsiung, Taiwan
- Department of Internal Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan
| | - Hsiang-Lin Lee
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Deptartment of Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chung-Jung Liu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Medical University, Kaohsiung, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Hsien Hsieh
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yong-Syuan Chen
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Kuan-Chun Hsueh
- Division of General Surgery, Department of Surgery, Tungs' Taichung Metroharbor Hospital, Taichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
5
|
Fan W, Cao D, Yang B, Wang J, Li X, Kitka D, Li TWH, You S, Shiao S, Gangi A, Posadas E, Di Vizio D, Tomasi ML, Seki E, Mato JM, Yang H, Lu SC. Hepatic prohibitin 1 and methionine adenosyltransferase α1 defend against primary and secondary liver cancer metastasis. J Hepatol 2024; 80:443-453. [PMID: 38086446 PMCID: PMC10922446 DOI: 10.1016/j.jhep.2023.11.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 03/30/2023] [Revised: 11/06/2023] [Accepted: 11/13/2023] [Indexed: 12/24/2023]
Abstract
BACKGROUND & AIMS The liver is a common site of cancer metastasis, most commonly from colorectal cancer, and primary liver cancers that have metastasized are associated with poor outcomes. The underlying mechanisms by which the liver defends against these processes are largely unknown. Prohibitin 1 (PHB1) and methionine adenosyltransferase 1A (MAT1A) are highly expressed in the liver. They positively regulate each other and their deletion results in primary liver cancer. Here we investigated their roles in primary and secondary liver cancer metastasis. METHODS We identified common target genes of PHB1 and MAT1A using a metastasis array, and measured promoter activity and transcription factor binding using luciferase reporter assays and chromatin immunoprecipitation, respectively. We examined how PHB1 or MAT1A loss promotes liver cancer metastasis and whether their loss sensitizes to colorectal liver metastasis (CRLM). RESULTS Matrix metalloproteinase-7 (MMP-7) is a common target of MAT1A and PHB1 and its induction is responsible for increased migration and invasion when MAT1A or PHB1 is silenced. Mechanistically, PHB1 and MAT1A negatively regulate MMP7 promoter activity via an AP-1 site by repressing the MAFG-FOSB complex. Loss of MAT1A or PHB1 also increased MMP-7 in extracellular vesicles, which were internalized by colon and pancreatic cancer cells to enhance their oncogenicity. Low hepatic MAT1A or PHB1 expression sensitized to CRLM, but not if endogenous hepatic MMP-7 was knocked down first, which lowered CD4+ T cells while increasing CD8+ T cells in the tumor microenvironment. Hepatocytes co-cultured with colorectal cancer cells express less MAT1A/PHB1 but more MMP-7. Consistently, CRLM raised distant hepatocytes' MMP-7 expression in mice and humans. CONCLUSION We have identified a PHB1/MAT1A-MAFG/FOSB-MMP-7 axis that controls primary liver cancer metastasis and sensitization to CRLM. IMPACT AND IMPLICATIONS Primary and secondary liver cancer metastasis is associated with poor outcomes but whether the liver has underlying defense mechanism(s) against metastasis is unknown. Here we examined the hypothesis that hepatic prohibitin 1 (PHB1) and methionine adenosyltransferase 1A (MAT1A) cooperate to defend the liver against metastasis. Our studies found PHB1 and MAT1A form a complex that suppresses matrix metalloproteinase-7 (MMP-7) at the transcriptional level and loss of either PHB1 or MAT1A sensitizes the liver to metastasis via MMP-7 induction. Strategies that target the PHB1/MAT1A-MMP-7 axis may be a promising approach for the treatment of primary and secondary liver cancer metastasis.
Collapse
Affiliation(s)
- Wei Fan
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center (CSMC), Los Angeles, CA 90048, USA
| | - DuoYao Cao
- Department of Biomedical Sciences, CSMC, Los Angeles, CA 90048, USA
| | - Bing Yang
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center (CSMC), Los Angeles, CA 90048, USA; Department of Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Jiaohong Wang
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center (CSMC), Los Angeles, CA 90048, USA
| | - Xiaomo Li
- Department of Pathology, CSMC, Los Angeles CA 90048, USA
| | - Diana Kitka
- Department of Biomedical Sciences, CSMC, Los Angeles, CA 90048, USA; Department of Surgery, Cedars-Sinai Cancer, CSMC, Los Angeles, CA, 90048, USA
| | - Tony W H Li
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center (CSMC), Los Angeles, CA 90048, USA
| | - Sungyong You
- Department of Biomedical Sciences, CSMC, Los Angeles, CA 90048, USA; Department of Surgery, Cedars-Sinai Cancer, CSMC, Los Angeles, CA, 90048, USA
| | - Stephen Shiao
- Department of Radiation Oncology, CSMC, LA, CA 90048, USA
| | | | | | - Dolores Di Vizio
- Department of Biomedical Sciences, CSMC, Los Angeles, CA 90048, USA; Department of Surgery, Cedars-Sinai Cancer, CSMC, Los Angeles, CA, 90048, USA
| | - Maria Lauda Tomasi
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center (CSMC), Los Angeles, CA 90048, USA
| | - Ekihiro Seki
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center (CSMC), Los Angeles, CA 90048, USA
| | - José M Mato
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticasy Digestivas (Ciberehd), Basque Research and Technology Alliance (BRTA), Technology, Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - Heping Yang
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center (CSMC), Los Angeles, CA 90048, USA
| | - Shelly C Lu
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center (CSMC), Los Angeles, CA 90048, USA.
| |
Collapse
|
6
|
Uddin MH, Al‐Hallak MN, Khan HY, Aboukameel A, Li Y, Bannoura SF, Dyson G, Kim S, Mzannar Y, Azar I, Odisho T, Mohamed A, Landesman Y, Kim S, Beydoun R, Mohammad RM, Philip PA, Shields AF, Azmi AS. Molecular analysis of XPO1 inhibitor and gemcitabine-nab-paclitaxel combination in KPC pancreatic cancer mouse model. Clin Transl Med 2023; 13:e1513. [PMID: 38131168 PMCID: PMC10739156 DOI: 10.1002/ctm2.1513] [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] [Academic Contribution Register] [Received: 07/27/2023] [Revised: 11/03/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND The majority of pancreatic ductal adenocarcinoma (PDAC) patients experience disease progression while on treatment with gemcitabine and nanoparticle albumin-bound (nab)-paclitaxel (GemPac) necessitating the need for a more effective treatment strategy for this refractory disease. Previously, we have demonstrated that nuclear exporter protein exportin 1 (XPO1) is a valid therapeutic target in PDAC, and the selective inhibitor of nuclear export selinexor (Sel) synergistically enhances the efficacy of GemPac in pancreatic cancer cells, spheroids and patient-derived tumours, and had promising activity in a phase I study. METHODS Here, we investigated the impact of selinexor-gemcitabine-nab-paclitaxel (Sel-GemPac) combination on LSL-KrasG12D/+ ; LSL-Trp53R172H/+ ; Pdx1-Cre (KPC) mouse model utilising digital spatial profiling (DSP) and single nuclear RNA sequencing (snRNAseq). RESULTS Sel-GemPac synergistically inhibited the growth of the KPC tumour-derived cell line. The Sel-GemPac combination reduced the 2D colony formation and 3D spheroid formation. In the KPC mouse model, at a sub-maximum tolerated dose (sub-MTD) , Sel-GemPac enhanced the survival of treated mice compared to controls (p < .05). Immunohistochemical analysis of residual KPC tumours showed re-organisation of tumour stromal architecture, suppression of proliferation and nuclear retention of tumour suppressors, such as Forkhead Box O3a (FOXO3a). DSP revealed the downregulation of tumour promoting genes such as chitinase-like protein 3 (CHIL3/CHI3L3/YM1) and multiple pathways including phosphatidylinositol 3'-kinase-Akt (PI3K-AKT) signalling. The snRNAseq demonstrated a significant loss of cellular clusters in the Sel-GemPac-treated mice tumours including the CD44+ stem cell population. CONCLUSION Taken together, these results demonstrate that the Sel-GemPac treatment caused broad perturbation of PDAC-supporting signalling networks in the KPC mouse model. HIGHLIGHTS The majority of pancreatic ductal adenocarcinoma (PDAC) patients experience disease progression while on treatment with gemcitabine and nanoparticle albumin-bound (nab)-paclitaxel (GemPac). Exporter protein exportin 1 (XPO1) inhibitor selinexor (Sel) with GemPac synergistically inhibited the growth of LSL-KrasG12D/+; LSL-Trp53R172H/+; Pdx1-Cre (KPC) mouse derived cell line and enhanced the survival of mice. Digital spatial profiling shows that Sel-GemPac causes broad perturbation of PDAC-supporting signalling in the KPC model.
Collapse
Affiliation(s)
- Md. Hafiz Uddin
- Department of OncologyKarmanos Cancer InstituteWayne State University School of MedicineDetroitMichiganUSA
| | - Mohammad Najeeb Al‐Hallak
- Department of OncologyKarmanos Cancer InstituteWayne State University School of MedicineDetroitMichiganUSA
| | - Husain Yar Khan
- Department of OncologyKarmanos Cancer InstituteWayne State University School of MedicineDetroitMichiganUSA
| | - Amro Aboukameel
- Department of OncologyKarmanos Cancer InstituteWayne State University School of MedicineDetroitMichiganUSA
| | - Yiwei Li
- Department of OncologyKarmanos Cancer InstituteWayne State University School of MedicineDetroitMichiganUSA
| | - Sahar F. Bannoura
- Department of OncologyKarmanos Cancer InstituteWayne State University School of MedicineDetroitMichiganUSA
| | - Gregory Dyson
- Department of OncologyKarmanos Cancer InstituteWayne State University School of MedicineDetroitMichiganUSA
| | - Seongho Kim
- Department of OncologyKarmanos Cancer InstituteWayne State University School of MedicineDetroitMichiganUSA
| | - Yosef Mzannar
- Department of OncologyKarmanos Cancer InstituteWayne State University School of MedicineDetroitMichiganUSA
| | - Ibrahim Azar
- Department of OncologyKarmanos Cancer InstituteWayne State University School of MedicineDetroitMichiganUSA
| | | | - Amr Mohamed
- UH Seidman Cancer CenterUniversity Hospitals, Case Western Reserve UniversityClevelandOhioUSA
| | | | - Steve Kim
- Department of OncologyKarmanos Cancer InstituteWayne State University School of MedicineDetroitMichiganUSA
| | - Rafic Beydoun
- Department of OncologyKarmanos Cancer InstituteWayne State University School of MedicineDetroitMichiganUSA
- Department of PathologyWayne State University School of MedicineDetroitMichiganUSA
| | - Ramzi M. Mohammad
- Department of OncologyKarmanos Cancer InstituteWayne State University School of MedicineDetroitMichiganUSA
| | | | - Anthony F. Shields
- Department of OncologyKarmanos Cancer InstituteWayne State University School of MedicineDetroitMichiganUSA
| | - Asfar S. Azmi
- Department of OncologyKarmanos Cancer InstituteWayne State University School of MedicineDetroitMichiganUSA
| |
Collapse
|
7
|
Jones DC, Danaher P, Kim Y, Beechem JM, Gottardo R, Newell EW. An information theoretic approach to detecting spatially varying genes. CELL REPORTS METHODS 2023; 3:100507. [PMID: 37426750 PMCID: PMC10326450 DOI: 10.1016/j.crmeth.2023.100507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Academic Contribution Register] [Received: 02/06/2023] [Revised: 04/03/2023] [Accepted: 05/25/2023] [Indexed: 07/11/2023]
Abstract
A key step in spatial transcriptomics is identifying genes with spatially varying expression patterns. We adopt an information theoretic perspective to this problem by equating the degree of spatial coherence with the Jensen-Shannon divergence between pairs of nearby cells and pairs of distant cells. To avoid the notoriously difficult problem of estimating information theoretic divergences, we use modern approximation techniques to implement a computationally efficient algorithm designed to scale with in situ spatial transcriptomics technologies. In addition to being highly scalable, we show that our method, which we call maximization of spatial information (Maxspin), improves accuracy across several spatial transcriptomics platforms and a variety of simulations when compared with a variety of state-of-the-art methods. To further demonstrate the method, we generated in situ spatial transcriptomics data in a renal cell carcinoma sample using the CosMx Spatial Molecular Imager and used Maxspin to reveal novel spatial patterns of tumor cell gene expression.
Collapse
Affiliation(s)
| | | | - Youngmi Kim
- NanoString Technologies, Inc., Seattle, WA, USA
| | | | - Raphael Gottardo
- Fred Hutchinson Cancer Center, Seattle, WA, USA
- Biomedical Data Science Center, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
| | | |
Collapse
|
8
|
Liu H, Chen L, Wang C, Zhou H. Matrix metalloproteinase 7 is associated with clinical and pathological characteristics of salivary adenoid cystic carcinomas. Eur Arch Otorhinolaryngol 2023; 280:839-845. [PMID: 36066668 DOI: 10.1007/s00405-022-07630-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/25/2022] [Accepted: 08/24/2022] [Indexed: 01/25/2023]
Abstract
OBJECTIVES The aim of this study was to investigate the relationship between matrix metalloproteinase-7 (MMP-7) expression and the clinical and pathological characteristics of salivary adenoid cystic carcinomas (SACC) of the palatal minor salivary gland. METHODS In this study, 58 samples of SACC and 10 samples of normal salivary gland tissue were examined. Immunohistochemistry was used to detect MMP-7 and vascular endothelial growth factor A (VEGF-A) expression in SACC and normal tissues. The clinical and pathological characteristics of the patients with SACC were collected. RESULTS Of the 58 SACC samples, 44 were positive for MMP-7, and the expression rate was 75.9%. No expression was detected in the 10 normal salivary gland tissues. The level of MMP-7 expression in the SACC and normal samples was significantly different. The level of expression of MMP-7 in the SACC samples did not correlate with age, sex or pathological type but did correlate with pathological grade, nerve infiltration and clinical stage. There was a positive correlation between VEGF-A and MMP-7 expression. CONCLUSIONS The SACC samples showed high expression of MMP-7, which was associated with tumour differentiation, invasiveness and clinical stage. The detection of MMP-7 positively correlated with the detection of VEGF-A in SACC.
Collapse
Affiliation(s)
- Haitao Liu
- Department of Oral and Maxillofacial Surgery, First People's Hospital of Jiujiang City, Jiangxi Province, Jiujiang, 332000, China
| | - Linlin Chen
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Nanchang University, No.49 Fuzhou Road, Nanchang, 320049, Jiangxi, China.
| | - Chenliang Wang
- Department of Pathology, First People's Hospital of Jiujiang City, Jiangxi Province, Jiujiang, 332000, China
| | - Haibo Zhou
- Department of Oral and Maxillofacial Surgery, First People's Hospital of Jiujiang City, Jiangxi Province, Jiujiang, 332000, China
| |
Collapse
|
9
|
Schossig P, Coskun E, Arsenic R, Horst D, Sehouli J, Bergmann E, Andresen N, Sigler C, Busse A, Keller U, Ochsenreither S. Target Selection for T-Cell Therapy in Epithelial Ovarian Cancer: Systematic Prioritization of Self-Antigens. Int J Mol Sci 2023; 24:ijms24032292. [PMID: 36768616 PMCID: PMC9916968 DOI: 10.3390/ijms24032292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/23/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 01/26/2023] Open
Abstract
Adoptive T cell-receptor therapy (ACT) could represent a promising approach in the targeted treatment of epithelial ovarian cancer (EOC). However, the identification of suitable tumor-associated antigens (TAAs) as targets is challenging. We identified and prioritized TAAs for ACT and other immunotherapeutic interventions in EOC. A comprehensive list of pre-described TAAs was created and candidates were prioritized, using predefined weighted criteria. Highly ranked TAAs were immunohistochemically stained in a tissue microarray of 58 EOC samples to identify associations of TAA expression with grade, stage, response to platinum, and prognosis. Preselection based on expression data resulted in 38 TAAs, which were prioritized. Along with already published Cyclin A1, the TAAs KIF20A, CT45, and LY6K emerged as most promising targets, with high expression in EOC samples and several identified peptides in ligandome analysis. Expression of these TAAs showed prognostic relevance independent of molecular subtypes. By using a systematic vetting algorithm, we identified KIF20A, CT45, and LY6K to be promising candidates for immunotherapy in EOC. Results are supported by IHC and HLA-ligandome data. The described method might be helpful for the prioritization of TAAs in other tumor entities.
Collapse
Affiliation(s)
- Paul Schossig
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Ebru Coskun
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Ruza Arsenic
- Department of Pathology, Universitätsklinikum Heidelberg, Heidelberg University, 69120 Heidelberg, Germany
| | - David Horst
- Insitute of Pathology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Jalid Sehouli
- Department of Gynecology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
- Tumorbank Ovarian Cancer Network, 13353 Berlin, Germany
| | - Eva Bergmann
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Nadine Andresen
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Christian Sigler
- Charité Comprehensive Cancer Center, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Antonia Busse
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Ulrich Keller
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Sebastian Ochsenreither
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Charité Comprehensive Cancer Center, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
- Correspondence:
| |
Collapse
|
10
|
Alarcon NO, Jaramillo M, Mansour HM, Sun B. Therapeutic Cancer Vaccines—Antigen Discovery and Adjuvant Delivery Platforms. Pharmaceutics 2022; 14:pharmaceutics14071448. [PMID: 35890342 PMCID: PMC9325128 DOI: 10.3390/pharmaceutics14071448] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/20/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 12/15/2022] Open
Abstract
For decades, vaccines have played a significant role in protecting public and personal health against infectious diseases and proved their great potential in battling cancers as well. This review focused on the current progress of therapeutic subunit vaccines for cancer immunotherapy. Antigens and adjuvants are key components of vaccine formulations. We summarized several classes of tumor antigens and bioinformatic approaches of identification of tumor neoantigens. Pattern recognition receptor (PRR)-targeting adjuvants and their targeted delivery platforms have been extensively discussed. In addition, we emphasized the interplay between multiple adjuvants and their combined delivery for cancer immunotherapy.
Collapse
Affiliation(s)
- Neftali Ortega Alarcon
- Skaggs Pharmaceutical Sciences Center, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (N.O.A.); (M.J.); (H.M.M.)
| | - Maddy Jaramillo
- Skaggs Pharmaceutical Sciences Center, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (N.O.A.); (M.J.); (H.M.M.)
| | - Heidi M. Mansour
- Skaggs Pharmaceutical Sciences Center, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (N.O.A.); (M.J.); (H.M.M.)
- The University of Arizona Cancer Center, Tucson, AZ 85721, USA
- Department of Medicine, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
- BIO5 Institute, The University of Arizona, Tucson, AZ 85721, USA
| | - Bo Sun
- Skaggs Pharmaceutical Sciences Center, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (N.O.A.); (M.J.); (H.M.M.)
- The University of Arizona Cancer Center, Tucson, AZ 85721, USA
- BIO5 Institute, The University of Arizona, Tucson, AZ 85721, USA
- Correspondence: ; Tel.: +1-520-621-6420
| |
Collapse
|
11
|
Meng N, Li Y, Jiang P, Bu X, Ding J, Wang Y, Zhou X, Yu F, Zhang Y, Zhang J, Xia L. A Comprehensive Pan-Cancer Analysis of the Tumorigenic Role of Matrix Metallopeptidase 7 (MMP7) Across Human Cancers. Front Oncol 2022; 12:916907. [PMID: 35785154 PMCID: PMC9248742 DOI: 10.3389/fonc.2022.916907] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/10/2022] [Accepted: 05/05/2022] [Indexed: 12/12/2022] Open
Abstract
Growing evidence has shown the oncogenic function of matrix metallopeptidase 7 (MMP7) in various tumors. However, no systemic pan-cancer analysis on the association between MMP7 and different cancers based on big clinical data is available. TIMER2, GEPIA2, UALCAN, cBioPortal, String, Metascape, and other web databases were searched in the present study. Generally, MMP7 expression is significantly upregulated in most The Cancer Genome Atlas (TCGA) cancer types compared to the paired normal controls, yet is downregulated in tumor tissues of invasive breast carcinoma (BRCA), kidney chromophobe (KICH), kidney renal clear cell carcinoma (KIRC), liver hepatocellular carcinoma (LIHC), and skin cutaneous melanoma (SKCM). MMP7 protein expression is notably higher in the primary tumor tissues of colon cancer, lung adenocarcinoma (LUAD), and uterine corpus endometrial carcinoma (UCEC) than in normal tissues and is significantly lower in the primary tumor tissues of breast cancer, clear cell renal carcinoma, and ovarian cancer. Furthermore, MMP7 expression is strongly associated with pathological stages, clinical outcomes, tumor mutational burden (TMB), and microsatellite instability (TSI). Gene amplification was detected in most TCGA cancer types. In addition, the missense mutation is the primary type of MMP7 genetic alteration in tumors. Significant positive correlations between MMP7 expression and cancer-associated fibroblasts (CAFs) have been demonstrated in most TCGA cancers. MMP7 expression was also found to be positively correlated with infiltration of dendritic cells and macrophages in some specific tumor types. Functional enrichment analysis by the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways and gene ontology (GO) methods revealed that RNA processing and DNA damage checkpoints might reveal the pathogenetic mechanisms of MMP7. This pan-cancer analysis provides a clear panorama for the tumorigenic roles of MMP7 across different cancer types. Moreover, MMP7 could be a potential drug therapeutic target in such cancers.
Collapse
Affiliation(s)
- Nana Meng
- Department of Ophthalmology, Affiliated People’s Hospital, Jiangsu University, Zhenjiang, China
- Department of Ophthalmology, Zhenjiang Kangfu Eye Hospital, Zhenjiang, China
| | - Yaguang Li
- Department of Kidney Transplantation, Second Xiangya Hospital of Central South University, Changsha, China
| | - Pengcheng Jiang
- Department of General Surgery, Affiliated People’s Hospital, Jiangsu University, Zhenjiang, China
| | - Xuefeng Bu
- Department of General Surgery, Affiliated People’s Hospital, Jiangsu University, Zhenjiang, China
| | - Jifei Ding
- Department of Thoracic Surgery, Jiangsu Province Hospital on Integration of Chinese and Western Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Yan Wang
- Department of Orthopedic, Affiliated People’s Hospital, Jiangsu University, Zhenjiang, China
| | - Xiaodong Zhou
- Department of General Surgery, Affiliated People’s Hospital, Jiangsu University, Zhenjiang, China
| | - Feng Yu
- Department of General Surgery, Affiliated People’s Hospital, Jiangsu University, Zhenjiang, China
| | - Yongjun Zhang
- Department of General Surgery, Affiliated People’s Hospital, Jiangsu University, Zhenjiang, China
| | - Jie Zhang
- Department of General Surgery, Affiliated People’s Hospital, Jiangsu University, Zhenjiang, China
| | - Leizhou Xia
- Department of General Surgery, Affiliated People’s Hospital, Jiangsu University, Zhenjiang, China
- *Correspondence: Leizhou Xia,
| |
Collapse
|
12
|
Robust Validation and Comprehensive Analysis of a Novel Signature Derived from Crucial Metabolic Pathways of Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2022; 14:cancers14071825. [PMID: 35406597 PMCID: PMC8997486 DOI: 10.3390/cancers14071825] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/01/2022] [Revised: 03/25/2022] [Accepted: 03/29/2022] [Indexed: 02/01/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a malignant tumor with a dismal prognosis. PDAC have extensively reprogrammed metabolic characteristics influenced by interactions with normal cells, the effects of the tumor microenvironment and oncogene-mediated cell-autonomous pathways. In this study, we found that among all cancer hallmarks, metabolism played an important role in PDAC. Subsequently, a 16-gene prognostic signature was established with genes derived from crucial metabolic pathways, including glycolysis, bile acid metabolism, cholesterol homeostasis and xenobiotic metabolism (gbcx). The signature was used to distinguish overall survival in multiple cohorts from public datasets as well as a validation cohort followed up by us at Shanghai Cancer Center. Notably, the gbcx-related risk score (gbcxMRS) also accurately predicted poor PDAC subtypes, such as pure-basal-like and squamous types. At the same time, it also predicted PDAC recurrence. The gbcxMRS was also associated with immune cells, especially CD8 T cells, Treg cells. Furthermore, a high gbcxMRS may indicate high drug sensitivity to irinotecan and docetaxel and CTLA4 inhibitor immunotherapy. Taken together, these results indicate a robust and reproducible metabolic-related signature based on analysis of the overall pathogenesis of pancreatic cancer, which may have excellent prognostic and therapeutic implications for PDAC.
Collapse
|
13
|
Cao L, Zhang Y, Mi J, Shi Z, Fang Z, Jia D, Pan Z, Peng P. α-Hederin inhibits the platelet activating factor-induced metastasis of HCC cells through disruption of PAF/PTAFR axis cascaded STAT3/MMP-2 expression. Pharmacol Res 2022; 178:106180. [DOI: 10.1016/j.phrs.2022.106180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 01/18/2022] [Revised: 02/28/2022] [Accepted: 03/09/2022] [Indexed: 01/01/2023]
|
14
|
Kim HS, Kim MG, Min KW, Jung US, Kim DH. High MMP-11 expression associated with low CD8+ T cells decreases the survival rate in patients with breast cancer. PLoS One 2021; 16:e0252052. [PMID: 34038440 PMCID: PMC8153507 DOI: 10.1371/journal.pone.0252052] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/29/2021] [Accepted: 05/10/2021] [Indexed: 12/23/2022] Open
Abstract
Matrix metalloproteinase-11 (MMP-11) promote cancer invasion and metastasis through degrading the extracellular matrix. Protein degradation by MMP-11 in tumor cells may progressively suppress cancer surveillance activities with blocking immune response in breast cancer. The aim of study is to analyze clinicopathological parameters, molecular interactions and anticancer immune response in patients with MMP-11 expression and to provide candidate target drugs. We investigated the clinicopathologic parameters, specific gene sets, tumor antigenicity, and immunologic relevance according to MMP-11 expression in 226 and 776 breast cancer patients from the Hanyang University Guri Hospital (HUGH) cohort and The Cancer Genome Atlas (TCGA) data, respectively. We analyzed pathway networks and in vitro drug response. High MMP-11 expression was associated with worse survival rate in breast cancer from HUGH cohort and TCGA data (all p < 0.05). In analysis of immunologic gene sets, high MMP-11 expression was related to low immune response such as CD8+T cell, CD4+T cell and B cell. In silico cytometry, there was a decrease of cancer testis antigen and low tumor infiltrating lymphocyte in patient with high MMP-11 expression: activated dendritic cell, CD8+T cell, CD4+ memory T cell, and memory B cell. In pathway networks, MMP-11 was linked to the pathways including low immune response, response to growth hormone and catabolic process. We found that pictilisib and AZ960 effectively inhibited the breast cancer cell lines with high MMP-11 expression. Strategies making use of MMP-11-related hub genes could contribute to better clinical management/research for patients with breast cancer.
Collapse
Affiliation(s)
- Hyung Suk Kim
- Division of Breast Surgery, Department of Surgery, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Gyeonggi-do, Republic of Korea
| | - Min Gyu Kim
- Department of Surgery, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Gyeonggi-do, Republic of Korea
| | - Kyueng-Whan Min
- Department of Pathology, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Gyeonggi-do, Republic of Korea
| | - Un Suk Jung
- Department of Obstetrics and Gynecology, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Gyeonggi-do, Republic of Korea
- * E-mail: (USJ); (DHK)
| | - Dong-Hoon Kim
- Department of Pathology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- * E-mail: (USJ); (DHK)
| |
Collapse
|
15
|
Liao HY, Da CM, Liao B, Zhang HH. Roles of matrix metalloproteinase-7 (MMP-7) in cancer. Clin Biochem 2021; 92:9-18. [PMID: 33713636 DOI: 10.1016/j.clinbiochem.2021.03.003] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/11/2020] [Revised: 02/13/2021] [Accepted: 03/03/2021] [Indexed: 12/19/2022]
Abstract
Matrix metalloproteinase-7 (MMP-7) is a small proteolytic enzyme that secretes zinc and calcium endopeptidases. It can degrade a variety of extracellular matrix substrates and other substrates and plays important regulatory roles in many human pathophysiological processes. Since its discovery, MMP-7 has been recognized as a regulatory protein in wound healing, bone growth, and remodeling. Later, MMP-7 was reported to regulate the occurrence and development of cancers and mediate the proliferation, differentiation, metastasis, and invasion of several types of cancer cells via various mechanisms. Thus, matrix metalloproteinase-7 may be a promising tumor biomarker and therapeutic target. The expression of MMP-7 correlates with the clinical characteristics of cancer patients, and its expression profile is a new diagnostic and prognostic biomarker for a variety of human diseases. Hence, manipulating the expression or function of MMP-7 may be a potential treatment strategy for different diseases including cancers. This review summarizes the role played by MMP-7 in carcinogenesis of several human cancers, underlying mechanisms, and its clinical significance of the occurrence and development of cancers.
Collapse
Affiliation(s)
- Hai-Yang Liao
- The Second Clinical Medical College of Lanzhou University, 82 Cuiying Men, Lanzhou 730030, PR China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou 730000, PR China.
| | - Chao-Ming Da
- The Second Clinical Medical College of Lanzhou University, 82 Cuiying Men, Lanzhou 730030, PR China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou 730000, PR China.
| | - Bei Liao
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou 730000, PR China; The First Clinical Medical College of Lanzhou University, 1 Donggang Road, Lanzhou 730000, PR China
| | - Hai-Hong Zhang
- The Second Clinical Medical College of Lanzhou University, 82 Cuiying Men, Lanzhou 730030, PR China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou 730000, PR China.
| |
Collapse
|
16
|
Zhan Z, Jing Z, He B, Hosseini N, Westerhoff M, Choi EY, Garmire LX. Two-stage Cox-nnet: biologically interpretable neural-network model for prognosis prediction and its application in liver cancer survival using histopathology and transcriptomic data. NAR Genom Bioinform 2021; 3:lqab015. [PMID: 33778491 PMCID: PMC7985035 DOI: 10.1093/nargab/lqab015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/31/2020] [Revised: 02/01/2021] [Accepted: 02/24/2021] [Indexed: 12/11/2022] Open
Abstract
Pathological images are easily accessible data with the potential of prognostic biomarkers. Moreover, integration of heterogeneous data types from multi-modality, such as pathological image and gene expression data, is invaluable to help predicting cancer patient survival. However, the analytical challenges are significant. Here, we take the hepatocellular carcinoma (HCC) pathological image features extracted by CellProfiler, and apply them as the input for Cox-nnet, a neural network-based prognosis prediction model. We compare this model with the conventional Cox proportional hazards (Cox-PH) model, CoxBoost, Random Survival Forests and DeepSurv, using C-index and log-rank P-values. The results show that Cox-nnet is significantly more accurate than Cox-PH and Random Survival Forests models and comparable with CoxBoost and DeepSurv models, on pathological image features. Further, to integrate pathological image and gene expression data of the same patients, we innovatively construct a two-stage Cox-nnet model, and compare it with another complex neural-network model called PAGE-Net. The two-stage Cox-nnet complex model combining histopathology image and transcriptomic RNA-seq data achieves much better prognosis prediction, with a median C-index of 0.75 and log-rank P-value of 6e-7 in the testing datasets, compared to PAGE-Net (median C-index of 0.68 and log-rank P-value of 0.03). Imaging features present additional predictive information to gene expression features, as the combined model is more accurate than the model with gene expression alone (median C-index 0.70). Pathological image features are correlated with gene expression, as genes correlated to top imaging features present known associations with HCC patient survival and morphogenesis of liver tissue. This work proposes two-stage Cox-nnet, a new class of biologically relevant and interpretable models, to integrate multiple types of heterogenous data for survival prediction.
Collapse
Affiliation(s)
- Zhucheng Zhan
- School of Science and Engineering, Chinese University of Hong Kong, Shenzhen Campus, Shenzhen 518172, P.R. China
| | - Zheng Jing
- Department of Applied Statistics, University of Michigan, Ann Arbor, MI 48104, USA
| | - Bing He
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48104, USA
| | - Noshad Hosseini
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48104, USA
| | - Maria Westerhoff
- Department of Pathology, University of Michigan, Ann Arbor, MI 48104, USA
| | - Eun-Young Choi
- Department of Pathology, University of Michigan, Ann Arbor, MI 48104, USA
| | - Lana X Garmire
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48104, USA
| |
Collapse
|
17
|
Yadav PK, Gupta SK, Kumar S, Ghosh M, Yadav BS, Kumar D, Kumar A, Saini M, Kataria M. MMP-7 derived peptides with MHC class-I binding motifs from canine mammary tumor tissue elicit strong antigen-specific T-cell responses in BALB/c mice. Mol Cell Biochem 2020; 476:311-320. [PMID: 32970284 PMCID: PMC7511522 DOI: 10.1007/s11010-020-03908-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/01/2020] [Accepted: 09/07/2020] [Indexed: 11/27/2022]
Abstract
Matrix Metalloproteinases (MMPs)-induced altered proteolysis of extracellular matrix proteins and basement membrane holds the key for tumor progression and metastasis. Matrix metalloproteinases-7 (Matrilysin), the smallest member of the MMP family also performs quite alike; thus serves as a potential candidate for anti-tumor immunotherapy. Conversely, being an endogenous tumor-associated antigen (TAA), targeting MMP-7 for immunization is challenging. But MMP-7-based xenovaccine can surmount the obstacle of poor immunogenicity and immunological tolerance, often encountered in TAA-based conventional vaccine for anti-tumor immunotherapy. This paves the way for investigating the potential of MMP-7-derived major histocompatibility complex (MHC)-binding peptides to elicit precise epitope-specific T-cell responses towards their possible inclusion in anti-tumor vaccine formulations. Perhaps it also ushers the path of achieving multiple epitope-based broad and universal cellular immunity. In current experiment, an immunoinformatics approach has been employed to identify the putative canine matrix matelloproteinases-7 (cMMP-7)-derived peptides with MHC class-I-binding motifs which can elicit potent antigen-specific immune responses in BALB/c mice. Immunization with the cMMP-7 DNA vaccine induced a strong CD8+ cytotoxic T lymphocytes (CTLs) and Th1- type response, with high level of gamma interferon (IFN-γ) production in BALB/c mice. The two identified putative MHC-I-binding nonameric peptides (Peptide32-40 and Peptide175-183) from cMMP-7 induced significant lymphocyte proliferation along with the production of IFN-γ from CD8+ T-cells in mice immunized with cMMP-7 DNA vaccine. The current observation has depicted the immunogenic potential of the two cMMP-7-derived nonapeptides for their possible exploitation in xenovaccine-mediated anti-tumor immunotherapy in mouse model.
Collapse
Affiliation(s)
- Pavan Kumar Yadav
- ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, 243122, India.
- Faculty of Veterinary and Animal Sciences, Rajiv Gandhi South Campus, Banaras Hindu University, Mirzapur, Uttar Pradesh, 231001, India.
| | - Shishir Kumar Gupta
- ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, 243122, India
- Laboratory Animal Facility, CSIR-CDRI, Lucknow, Uttar Pradesh, 226031, India
| | - Saroj Kumar
- ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, 243122, India
- Faculty of Veterinary and Animal Sciences, Rajiv Gandhi South Campus, Banaras Hindu University, Mirzapur, Uttar Pradesh, 231001, India
| | - Mayukh Ghosh
- Faculty of Veterinary and Animal Sciences, Rajiv Gandhi South Campus, Banaras Hindu University, Mirzapur, Uttar Pradesh, 231001, India
| | - Brijesh Singh Yadav
- ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, 243122, India
- University of Information Science & Technology St. Paul the apostle Partizanska bb., 6000, Ohrid, Republic of Macedonia
| | - Dinesh Kumar
- ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, 243122, India
- College of Agriculture, Tikamgarh, Jawaharlal Nehru Krishi Vishwa Vidylaya, Jabalpur, Madhya Pradesh, 482004, India
| | - Ajay Kumar
- ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, 243122, India
| | - Mohini Saini
- ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, 243122, India
| | - Meena Kataria
- ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, 243122, India
| |
Collapse
|
18
|
Shao Y, Lu B. The crosstalk between circular RNAs and the tumor microenvironment in cancer metastasis. Cancer Cell Int 2020; 20:448. [PMID: 32943996 PMCID: PMC7488731 DOI: 10.1186/s12935-020-01532-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/06/2020] [Accepted: 09/01/2020] [Indexed: 02/06/2023] Open
Abstract
Background Carcinomas are highly heterogeneous with regard to various cancer cells within a tumor microenvironment (TME), which is composed of stromal cells, blood vessels, immunocytes, and modified extracellular matrix. Focus of the study Circular RNAs (circRNAs) are non-coding RNAs that are expressed in cancer and stromal cells. They are closely associated with cancer metastasis as their expression in tumor cells directs the latter to migrate to different organs. circRNAs packaged in exosomes might be involved in this process. This is particularly important as the TME acts in tandem with cancer cells to enhance their proliferation and metastatic capability. In this review, we focus on recent studies on the crosstalk between circRNAs and the TME during cancer metastasis. Conclusion We particularly emphasize the roles of the interaction between circRNAs and the TME in anoikis resistance, vessel co-option, and local circRNA expression in directing homing of exosome.
Collapse
Affiliation(s)
- Ying Shao
- Department of Surgical Pathology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang China
| | - Bingjian Lu
- Department of Surgical Pathology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang China
| |
Collapse
|
19
|
Wu Z, Wei ZH, Chen SH. LncUBE2R2-AS1 acts as a microRNA sponge of miR-302b to promote HCC progression via activation EGFR-PI3K-AKT signaling pathway. Cell Cycle 2020; 19:2426-2435. [PMID: 32835579 DOI: 10.1080/15384101.2020.1795991] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a main cause of cancer-related deaths globally. Long non-coding RNAs (lncRNAs) play important roles in diverse cancers. LncRNA-UBE2R2-AS1 has been reported to promote apoptosis in glioma cell. However, the expressions, functions, and mechanisms of action of UBE2R2-AS1 in HCC are still unclear. UBE2R2-AS1 is increased in HCC tissues and cell lines. Increased expression of UBE2R2-AS1 is associated with large tumor size, multiple tumor number, advanced TNM stage, and poor survival of HCC patients. Functional experiments showed that knockdown UBE2R2-AS1 inhibited HCC growth and metastasis through in vitro and in vivo experiments. Regarding the mechanism, UBE2R2-AS1/miR-302b/EGFR established the ceRNA network involved in the modulation of cell progression of HCC cells via activation of PI3K-AKT signaling pathway. Overall, UBE2R2-AS1 may exhibit an oncogenic function in HCC via acting as a sponge for miR-302b to up-regulate EGFR, and may serve as a potential therapeutic target and a prognostic biomarker for HCC patients.
Collapse
Affiliation(s)
- Zhe Wu
- Department of Hepatology Surgery, 900 Hospital of the Joint Logistics Team , Fuzhou, Fujian, China
| | - Zhi-Hong Wei
- Department of Hepatology Surgery, 900 Hospital of the Joint Logistics Team , Fuzhou, Fujian, China
| | - Shao-Hua Chen
- Department of Hepatology Surgery, 900 Hospital of the Joint Logistics Team , Fuzhou, Fujian, China
| |
Collapse
|
20
|
Kumar P, Siripini S, Sreedhar AS. The matrix metalloproteinase 7 (MMP7) links Hsp90 chaperone with acquired drug resistance and tumor metastasis. Cancer Rep (Hoboken) 2020; 5:e1261. [PMID: 32761892 PMCID: PMC9780424 DOI: 10.1002/cnr2.1261] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/16/2020] [Revised: 05/12/2020] [Accepted: 06/01/2020] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Cancer emergence is associated with a series of cellular transformations that include acquired drug resistance followed by tumor metastasis. Matrix metalloproteinases (MMPs) and Hsp90 chaperone are implicated in tumor progression, however, they are not studied in the context of drug resistance. AIMS In the present study, we aimed at understanding the cross-talk between acquired drug resistance and tumor progression, linking MMP7 and Hsp90. METHODS AND RESULTS We have developed an in vitro model system for acquired drug resistance and studied the correlation between MMP7 and Hsp90. We demonstrate that enhanced drug efflux activity correlates with the induced expression and activity of MMP7, and enhanced metastatic potential of cells, however, in Hsp90-dependent manner. The MMP7 overexpression alone could enhance the drug efflux activity marginally, and metastasis significantly. However, challenging these cells with 17AAG has significantly increased the drug efflux activity and, in contrast, decreased the metastatic potential. Evaluating our in vitro findings in mice xenografts revealed that MMP7 overexpression facilitates altered homing properties. However, these cells, in response to 17AAG treatment, exhibited increased localized tumor growth but decreased tumor metastasis. CONCLUSION We demonstrated a cross-talk between Hsp90 and MMP7 in regulating the acquired drug resistance and tumor progression. Our findings provide novel insights on targeting drug resistant-tumors.
Collapse
Affiliation(s)
- Pankaj Kumar
- CSIR‐Centre for Cellular and Molecular BiologyHyderabadIndia
| | | | | |
Collapse
|
21
|
Abbes A, Zayani Y, Zidi W, Hammami MB, Mebazaa A, El Euch D, Ben Ammar A, Sanhaji H, El May MV, Mokni M, Feki M, Allal-Elasmi M. Matrix metalloproteinase-7 could be a predictor for acute inflammation in psoriatic patients. Cytokine 2020; 134:155195. [PMID: 32663776 DOI: 10.1016/j.cyto.2020.155195] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/15/2020] [Revised: 06/03/2020] [Accepted: 07/04/2020] [Indexed: 11/15/2022]
Abstract
PURPOSE The pathogenesis of psoriasis is characterized by a disruption of extracellular matrix (ECM) in which matrix metalloproteinases (MMPs) participate actively. We aimed to determine MMP-7 level and its association with the inflammatory response in order to determine its usefulness as a biomarker for psoriasis prediction. We also aimed to determine its distribution in uninvolved and involved psoriatic skin to evaluate the probable role of MMP-7 in psoriasis pathogenesis. MATERIALS AND METHODS We recruited 108 psoriatic patients and 133 healthy controls. MMP-7, tissue inhibitors of metalloproteinases (TIMPs) and interleukin-6 (IL-6) levels were measured by Enzyme-Linked Immunosorbent Assay (ELISA) assay. MMP-7 expression was detected by Immunohistochemistry (IHC) study. RESULTS ECM turnover and inflammatory biomarker levels were significantly higher in psoriatic patients. MMP-7 revealed to be independently associated to psoriasis even after adjustment for different models. The area under the curve (AUC) of MMP-7 and inflammation Z-score were similar. MMP-7 was positively correlated with IL-6 and inflammation Z-score. Psoriasis severity (PASI) was correlated significantly with IL-6 (p = 0.007). The MMP-7 expression was detected in the epidermis of involved and uninvolved psoriatic skin. In involved skin, MMP-7 was expressed by basal and mostly suprabasal keratinocytes. In uninvolved skin, expression of MMP-7 was restricted to basal keratinocytes. CONCLUSION MMP-7 is independently associated to psoriasis disease and to inflammatory response which make it a potential biomarker for this dermatosis.
Collapse
Affiliation(s)
- Arbia Abbes
- University of Tunis El Manar, Faculty of Medicine of Tunis, LR99ES11, Laboratory of Biochemistry, Rabta Hospital, Tunis, Tunisia; University of Tunis El Manar, Faculty of Sciences of Tunis, Tunisia
| | - Yosra Zayani
- University of Tunis El Manar, Faculty of Medicine of Tunis, LR99ES11, Laboratory of Biochemistry, Rabta Hospital, Tunis, Tunisia
| | - Wiem Zidi
- University of Tunis El Manar, Faculty of Medicine of Tunis, LR99ES11, Laboratory of Biochemistry, Rabta Hospital, Tunis, Tunisia
| | - Mohamed Bassem Hammami
- University of Tunis El Manar, Faculty of Medicine of Tunis, LR99ES11, Laboratory of Biochemistry, Rabta Hospital, Tunis, Tunisia
| | - Amel Mebazaa
- Department of Dermatology, Rabta Hospital, Tunis, Tunisia
| | | | - Awatef Ben Ammar
- Research Unit 17ES/13 Laboratory of Histology and Embryology, Faculty of Medicine, University of Tunis El Manar, Tunis, Tunisia
| | - Haifa Sanhaji
- University of Tunis El Manar, Faculty of Medicine of Tunis, LR99ES11, Laboratory of Biochemistry, Rabta Hospital, Tunis, Tunisia
| | - Michele Veronique El May
- Research Unit 17ES/13 Laboratory of Histology and Embryology, Faculty of Medicine, University of Tunis El Manar, Tunis, Tunisia
| | - Mourad Mokni
- Department of Dermatology, Rabta Hospital, Tunis, Tunisia
| | - Moncef Feki
- University of Tunis El Manar, Faculty of Medicine of Tunis, LR99ES11, Laboratory of Biochemistry, Rabta Hospital, Tunis, Tunisia
| | - Monia Allal-Elasmi
- University of Tunis El Manar, Faculty of Medicine of Tunis, LR99ES11, Laboratory of Biochemistry, Rabta Hospital, Tunis, Tunisia.
| |
Collapse
|
22
|
Liu Z, Tan RJ, Liu Y. The Many Faces of Matrix Metalloproteinase-7 in Kidney Diseases. Biomolecules 2020; 10:biom10060960. [PMID: 32630493 PMCID: PMC7356035 DOI: 10.3390/biom10060960] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/01/2020] [Revised: 06/19/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022] Open
Abstract
Matrix metalloproteinase-7 (MMP-7) is a secreted zinc-dependent endopeptidase that is implicated in regulating kidney homeostasis and diseases. MMP-7 is produced as an inactive zymogen, and proteolytic cleavage is required for its activation. MMP-7 is barely expressed in normal adult kidney but upregulated in acute kidney injury (AKI) and chronic kidney disease (CKD). The expression of MMP-7 is transcriptionally regulated by Wnt/β-catenin and other cues. As a secreted protein, MMP-7 is present and increased in the urine of patients, and its levels serve as a noninvasive biomarker for predicting AKI prognosis and monitoring CKD progression. Apart from degrading components of the extracellular matrix, MMP-7 also cleaves a wide range of substrates, such as E-cadherin, Fas ligand, and nephrin. As such, it plays an essential role in regulating many cellular processes, such as cell proliferation, apoptosis, epithelial-mesenchymal transition, and podocyte injury. The function of MMP-7 in kidney diseases is complex and context-dependent. It protects against AKI by priming tubular cells for survival and regeneration but promotes kidney fibrosis and CKD progression. MMP-7 also impairs podocyte integrity and induces proteinuria. In this review, we summarized recent advances in our understanding of the regulation, role, and mechanisms of MMP-7 in the pathogenesis of kidney diseases. We also discussed the potential of MMP-7 as a biomarker and therapeutic target in a clinical setting.
Collapse
Affiliation(s)
- Zhao Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China;
| | - Roderick J. Tan
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA;
| | - Youhua Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China;
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Correspondence:
| |
Collapse
|
23
|
Hu PS, Wang YC, Liao CH, Hsia NY, Wu MF, Yang JS, Yu CC, Chang WS, Bau DAT, Tsai CW. The Association of MMP7 Genotype With Pterygium. In Vivo 2020; 34:51-56. [PMID: 31882462 DOI: 10.21873/invivo.11744] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/15/2019] [Revised: 10/05/2019] [Accepted: 10/10/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND/AIM In literature, few studies have examined the diagnostic or prognostic potential of matrix metalloproteinases (MMP) in pterygium, whose formation and progression are closely related to imbalance in the extracellular microenvironment. In this study, we investigated the contribution of MMP7 promoter (A-181G and C-153T) polymorphic genotypes to pterygium risk. MATERIALS AND METHODS A total of 134 cases and 268 controls were collected and their MMP7 genotypes at A-181G and C-153T were examined by polymerase chain reaction-restriction fragment length polymorphism methodology. RESULTS The AA, AG and GG genotypes at MMP7 promoter A-181G were non-significantly differentially distributed between the two groups at 85.8, 11.2 and 3.0%, respectively, in pterygium cases and 88.4, 9.7 and 1.9% in controls, respectively (p for trend=0.6822). There was no polymorphic genotype for MMP7 C-153T among our Taiwanese cohort. CONCLUSION A-181G and C-153T genotypes at MMP7 do not have a direct role in determining Taiwanese susceptibility to pterygium.
Collapse
Affiliation(s)
- Pei-Shin Hu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C.,Department of Ophthalmology, Changhua Christian Hospital, Changhua, Taiwan, R.O.C.,Terry Fox Cancer Research Laboratory, Translational Medicine Center, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Yun-Chi Wang
- Terry Fox Cancer Research Laboratory, Translational Medicine Center, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Cheng-Hsi Liao
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C.,Terry Fox Cancer Research Laboratory, Translational Medicine Center, China Medical University Hospital, Taichung, Taiwan, R.O.C.,National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Ning-Yi Hsia
- Terry Fox Cancer Research Laboratory, Translational Medicine Center, China Medical University Hospital, Taichung, Taiwan, R.O.C.,Department of Ophthalmology, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Meng-Feng Wu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C.,Terry Fox Cancer Research Laboratory, Translational Medicine Center, China Medical University Hospital, Taichung, Taiwan, R.O.C.,National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Jai-Sing Yang
- Terry Fox Cancer Research Laboratory, Translational Medicine Center, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Chien-Chih Yu
- Terry Fox Cancer Research Laboratory, Translational Medicine Center, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Wen-Shin Chang
- Terry Fox Cancer Research Laboratory, Translational Medicine Center, China Medical University Hospital, Taichung, Taiwan, R.O.C.
| | - DA-Tian Bau
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C. .,Terry Fox Cancer Research Laboratory, Translational Medicine Center, China Medical University Hospital, Taichung, Taiwan, R.O.C.,Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan, R.O.C
| | - Chia-Wen Tsai
- Terry Fox Cancer Research Laboratory, Translational Medicine Center, China Medical University Hospital, Taichung, Taiwan, R.O.C.
| |
Collapse
|
24
|
Yadav PK, Gupta SK, Kumar S, Ghosh M, Yadav BS, Kumar D, Kumar A, Saini M, Kataria M. IL-18 immunoadjuvanted xenogeneic canine MMP-7 DNA vaccine overcomes immune tolerance and supresses the growth of murine mammary tumor. Int Immunopharmacol 2020; 82:106370. [PMID: 32155464 DOI: 10.1016/j.intimp.2020.106370] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/13/2020] [Revised: 02/27/2020] [Accepted: 03/02/2020] [Indexed: 12/22/2022]
Abstract
The development of the tumorigenesis and angiogenesis through proteolytic cleavage of extracellular matrix protein and basement membranes is promoted by Matrix metelloproteinases-7 (MMP-7). Consequently, MMP-7 is presumed as potential target for mammary cancer immunotherapy. However, MMP-7 is an endogenous tumor associated antigen (TAA); therefore, immunization is challenging. In current study, a potent anti-tumor immune response has been elicited through recombinant bivalent plasmid pVIVO2.IL18.cMMP7 which subside the highly metastatic 4 T1 cell line induced mammary tumors and efficiently negate the existing challenge of using MMP-7 as immunotherapeutic target. Balb/c mice were immunized with canine MMP-7 (cMMP-7) using interleukine-18 (IL-18), as an immunoadjuvant, to explore the potential of the combination regarding elicitation of a potent anti-tumor immune response. Mice vaccinated with pVIVO2.IL18.cMMP7 DNA plasmid reduced the tumor growth significantly along with augmentation of the immune response to fight against tumor antigen as depicted by substantial enrichment of CD4+ and CD8+ population in splenocytes, infiltration of immune system cells in tumor tissue and enhanced survival time of mice. Further, splenocyte supernatant examination of the cytokines revealed that Th1 cytokines (IFN-γ and IL-2) were remarkably up-regulated demonstrating the stimulation of cell-mediated immune response. Thus the current observations vividly portray that administration of xenogeneic MMP-7 DNA vaccine bypasses the tolerance barrier.
Collapse
Affiliation(s)
- Pavan Kumar Yadav
- ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, Uttar Pradesh, India; Faculty of Veterinary and Animal Sciences, Rajiv Gandhi South Campus, Banaras Hindu University, Mirzapur 231001, Uttar Pradesh, India.
| | - Shishir Kumar Gupta
- ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, Uttar Pradesh, India; Laboratory Animal Facility, CSIR-CDRI, Lucknow 226031, Uttar Pradesh, India
| | - Saroj Kumar
- ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, Uttar Pradesh, India; Faculty of Veterinary and Animal Sciences, Rajiv Gandhi South Campus, Banaras Hindu University, Mirzapur 231001, Uttar Pradesh, India
| | - Mayukh Ghosh
- Faculty of Veterinary and Animal Sciences, Rajiv Gandhi South Campus, Banaras Hindu University, Mirzapur 231001, Uttar Pradesh, India
| | - Brijesh Singh Yadav
- ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, Uttar Pradesh, India; University of Information Science & Technology St. Paul the apostle Partizanska bb., 6000 Ohrid, The Former Yugolav Republic of Macedonia
| | - Dinesh Kumar
- ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, Uttar Pradesh, India; College of Agriculture, Tikamgarh, Jawaharlal Nehru Krishi Vishwa Vidylaya, Jabalpur 482004, Madhya Pradesh, India
| | - Ajay Kumar
- ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, Uttar Pradesh, India
| | - Mohini Saini
- ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, Uttar Pradesh, India
| | - Meena Kataria
- ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, Uttar Pradesh, India
| |
Collapse
|
25
|
The Role of Matrix Metalloproteinases in the Epithelial-Mesenchymal Transition of Hepatocellular Carcinoma. Anal Cell Pathol (Amst) 2019; 2019:9423907. [PMID: 31886121 PMCID: PMC6899323 DOI: 10.1155/2019/9423907] [Citation(s) in RCA: 211] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/03/2019] [Accepted: 10/08/2019] [Indexed: 02/07/2023] Open
Abstract
The epithelial-mesenchymal transition (EMT) is a transformation process mandatory for the local and distant progression of many malignant tumors, including hepatocellular carcinoma (HCC). Matrix metalloproteinases (MMPs) play significant roles in cellular regeneration, programmed death, angiogenesis, and many other essential tissular functions, involved in the normal development and also in pathological processes, such as the EMT. This paper reviews the roles of MMPs in the EMT involved in HCC invasion, as well as the ancillary roles that MMP cross-activation and tissue inhibitors play in modulating this process. While gelatinases MMP-2 and MMP-9 are the MMPs commonly cited in the EMT of HCC, MMPs belonging to other classes have been proven to be involved in this process, favoring not only invasion and metastasis (MMP-1, MMP-3, MMP-7, MMP-10, MMP-11, MMP-13, MMP-14, MMP-16, MMP-26, and MMP-28) but also angiogenesis (MMP-8 and MMP-10). There is also data suggesting that other MMPs with a suspected or demonstrated role in the EMT of other cancers may also have some degree of involvement in HCC. The auto- and cross-activation of MMPs may complicate this issue, as pinpointing the extent of implication of each MMP may be extremely difficult. The homeostasis between MMPs and their tissue inhibitors is essential in preventing tumor progression, and the disturbance of this stability is another entailed factor in the EMT of HCC, which is addressed herein.
Collapse
|
26
|
The role of matrix metalloproteinases in osteoarthritis pathogenesis: An updated review. Life Sci 2019; 234:116786. [DOI: 10.1016/j.lfs.2019.116786] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/20/2019] [Revised: 08/19/2019] [Accepted: 08/20/2019] [Indexed: 12/22/2022]
|
27
|
Dimitrova I, Tacheva T, Mindov I, Petrov B, Aleksandrova E, Valkanov S, Gulubova M, Vlaykova T. Serum levels of MMP-7 in primary brain cancers and brain metastases. BIOTECHNOL BIOTEC EQ 2019. [DOI: 10.1080/13102818.2019.1626282] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Ivelina Dimitrova
- Department of Surgery and Neurosurgery, Medical Faculty, Trakia University, Stara Zagora, Bulgaria
| | - Tanya Tacheva
- Department of Chemistry and Biochemistry, Medical Faculty, Trakia University, Stara Zagora, Bulgaria
| | - Ivan Mindov
- Department of Surgery and Neurosurgery, Medical Faculty, Trakia University, Stara Zagora, Bulgaria
| | - Bozhidar Petrov
- Department of Surgery and Neurosurgery, Medical Faculty, Trakia University, Stara Zagora, Bulgaria
| | - Elina Aleksandrova
- Department of General and Clinical Pathology, Medical Faculty, Trakia University, Stara Zagora, Bulgaria
| | - Stefan Valkanov
- Department of Surgery and Neurosurgery, Medical Faculty, Trakia University, Stara Zagora, Bulgaria
| | - Maya Gulubova
- Department of General and Clinical Pathology, Medical Faculty, Trakia University, Stara Zagora, Bulgaria
| | - Tatyana Vlaykova
- Department of Chemistry and Biochemistry, Medical Faculty, Trakia University, Stara Zagora, Bulgaria
| |
Collapse
|
28
|
Liao CH, Chang WS, Tsai CW, Hu PS, Wu HC, Hsu SW, Chen GL, Yueh TC, Shen TC, Hsia TC, Bau DAT. Association of Matrix Metalloproteinase-7 Genotypes with the Risk of Bladder Cancer. In Vivo 2019; 32:1045-1050. [PMID: 30388078 DOI: 10.21873/invivo.11345] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/20/2018] [Revised: 05/29/2018] [Accepted: 05/01/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND/AIM The breakage of matrix metalloproteinases (MMPs) has been reported to be one of the mechanisms required for tumor invasion, and the expression of MMP-7 in serum is correlated with poor prognosis of urinary bladder cancer patients. However, the role of the MMP-7 genotypes has been seldom examined among bladder cancer patients. Therefore, this study aimed at examining the promoter polymorphic MMP-7 genotypes A-181G and C-153T among Taiwanese bladder cancer patients and evaluate the contribution of the genotypic variants of MMP-7 to bladder cancer risk in Taiwan. MATERIALS AND METHODS Three hundred and seventy-five bladder cancer patients and the same number of gender- and age-matched healthy controls were genotyped for A-181G and C-153T in the promoter of MMP-7 via polymerase chain reaction-restriction fragment length polymorphism methodology. RESULTS The frequencies of AA, AG and GG at A-181G of the promoter of MMP-7 were 89.1, 8.8 and 2.1% in the bladder cancer patient group and 87.5, 10.9 and 1.6% in the matched healthy control group, respectively (p for trend=0.5475). There was no polymorphic genotype for MMP-7 C-153T among the Taiwanese population. The comparisons in allelic frequency distribution also support the findings that the G allele may not be the determinant allele for bladder cancer in Taiwan. In addition, the results showed that there is no significant association of the bladder risk with the MMP-7 A-181G genotype, even after adjustment for the possible confounding factors. Furthermore, there is no interaction of the genotypes of MMP-7 with age, gender, smoking and alcohol consumption on bladder cancer risk. CONCLUSION The results of this study suggest that the two MMP-7 polymorphisms, - A-181G and C-153T, do not play a major role in determining personal susceptibility to bladder cancer in Taiwan.
Collapse
Affiliation(s)
- Cheng-Hsi Liao
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C.,Department of Surgery, Taichung Armed Forces General Hospital, Taichung, Taiwan, R.O.C.,National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Wen-Shin Chang
- Terry Fox Cancer Research Laboratory, Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Chia-Wen Tsai
- Terry Fox Cancer Research Laboratory, Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Pei-Shin Hu
- Department of Ophthalmology, Changhua Christian Hospital, Changhua, Taiwan, R.O.C
| | - Hsi-Chin Wu
- Terry Fox Cancer Research Laboratory, Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Shih-Wei Hsu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C.,Department of Surgery, Taichung Armed Forces General Hospital, Taichung, Taiwan, R.O.C.,National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Guan-Liang Chen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C.,National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Te-Cheng Yueh
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C.,Department of Surgery, Taichung Armed Forces General Hospital, Taichung, Taiwan, R.O.C.,National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Te-Chun Shen
- Terry Fox Cancer Research Laboratory, Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Te-Chun Hsia
- Terry Fox Cancer Research Laboratory, Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - DA-Tian Bau
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C. .,Terry Fox Cancer Research Laboratory, Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan, R.O.C.,Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan, R.O.C
| |
Collapse
|
29
|
Zhang H, Wang Y, Chen T, Zhang Y, Xu R, Wang W, Cheng M, Chen Q. Aberrant Activation Of Hedgehog Signalling Promotes Cell Migration And Invasion Via Matrix Metalloproteinase-7 In Ovarian Cancer Cells. J Cancer 2019; 10:990-1003. [PMID: 30854105 PMCID: PMC6400802 DOI: 10.7150/jca.26478] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/04/2018] [Accepted: 12/14/2018] [Indexed: 12/31/2022] Open
Abstract
Background: Accumulating evidence indicates that aberrant activation of the Hedgehog (Hh) signalling pathway by Glioma-associated oncogene (Gli) transcription factors is involved in the aggressive progression of cancers, including ovarian cancer. Whereas the molecular mechanism underlying this phenomenon remains unelucidated. Matrix metalloproteinase-7 (MMP-7) facilitates degradation of the extracellular matrix, promoting the invasion of tumour cells, and is associated with cancer progression and metastasis. In previous reports, we identified a set of genes regulated by Hh signalling in ovarian tumour cells among which MMP-7 was identified as a potential Hh target gene candidate. However, establishing an association between Hh signalling activation and MMP-7 expression requires further validation, and the function of this regulation remains unknown. Methods: A cDNA microarray was utilized to identify potential downstream targets of Hh signalling. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) was used to evaluate mRNA expression, and immunoblotting (IB) was conducted to evaluate protein expression. The invasive and migratory capabilities of tumour cells were tested with the transwell and wound healing assays, respectively. The mRNA levels of Gli2 and MMP-7 in normal ovarian tissues and cancerous tissues in various stages together with the corresponding clinical information were acquired from the indicated GEO datasets to elucidate associations between MMP-7 expression and cancer progression and prognosis. Additionally, immunohistochemistry (IHC) was performed in multiple ovarian cancers, benign tumours and normal tissues to evaluate Gli2 and MMP-7 protein expression. Results: MMP-7 expression was regulated by the Hh ligand, antagonist and downstream transcript factor Gli2, demonstrating this gene as an Hh target. MMP-7 facilitated the invasion and migration of ovarian tumour cells, indicating its key function in ovarian cancer progression. IHC analysis demonstrated abnormally increased Gli2 and MMP-7 expression levels in benign tumours and ovarian cancer tissues. Moreover, high MMP-7 levels were significantly associated with poor overall survival (OS) and poor progression-free survival (PFS) in ovarian cancer patients. Conclusion: Aberrant activation of the Hh-Gli-MMP-7 signalling axis is essential for acceleration of the progression and metastasis of human ovarian cancer, implicating its use as a novel therapeutic target of ovarian cancer. In addition, MMP-7 can potentially serve as a prognostic marker of ovarian cancer.
Collapse
Affiliation(s)
- Hong Zhang
- Department of Obstetrics & Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Yiting Wang
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Tingtao Chen
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Yan Zhang
- Department of Obstetrics & Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Rong Xu
- Department of Obstetrics & Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Wanwan Wang
- Department of Obstetrics & Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Minzhang Cheng
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Qi Chen
- Department of Obstetrics & Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| |
Collapse
|
30
|
Yueh TC, Wu CN, Hung YW, Chang WS, Fu CK, Pei JS, Wu MH, Lai YL, Lee YM, Yen ST, Li HT, Tsai CW, Bau DAT. The Contribution of MMP-7 Genotypes to Colorectal Cancer Susceptibility in Taiwan. Cancer Genomics Proteomics 2018; 15:207-212. [PMID: 29695403 DOI: 10.21873/cgp.20079] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/14/2018] [Revised: 03/22/2018] [Accepted: 03/23/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND/AIM Matrix metalloproteinases (MMPs) play important roles in inflammation and carcinogenesis, but the genotypic role of MMP-7 has never been investigated in colorectal cancer (CRC) among the Taiwanese. Therefore, in this study we aimed to evaluate the contribution of MMP-7 genotypes to the risk of CRC in Taiwan. MATERIALS AND METHODS In this case-control study, MMP-7 A-181G and C-153T promoter genotypes were determined and their association with CRC risk were investigated among 362 CRC patients and 362 age- and gender-matched healthy controls. In addition, the interaction of MMP-7 genotypes and personal behaviors were also examined. RESULTS The percentages of variant AG and GG for MMP-7 A-181G genotypes were 10.5% and 1.7% in the CRC group and 11.9% and 2.2% in the control group, respectively (p for trend=0.7145). The allelic frequency distribution analysis showed that the variant G allele of MMP-7 A-181G conferred a slight but non-significant decreased CRC susceptibility to the wild-type C allele (odds ratio (OR)=0.86, 95% confidence interval (CI)=0.64-1.31, p=0.37). Taiwanese all harbour the CC genotype at MMP-7 C-153T. As for the gene-lifestyle interaction, there were no obvious joint effects of MMP-7 A-181G genotype on the risk of CRC among ever smoker, alcohol drinker, non-smoker or non-drinker subgroups. No statistically significant correlation was observed between MMP-7 A-181G genotypic distributions and age, gender, tumor size, location or metastasis status. CONCLUSION The genotypes of MMP-7 A-181G may play an indirect role in determining personal susceptibility to CRC and prognosis. The further genotyping work on MMP-7 and other genes (such as other MMPs, oncogenes and tumor suppression genes) on CRC susceptibility and prognosis, should be taken into consideration spontaneously in the precision medicine era.
Collapse
Affiliation(s)
- Te-Cheng Yueh
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C.,Taichung Armed Forces General Hospital, Taichung, Taiwan, R.O.C.,Terry Fox Cancer Research Laboratory, Translational Medical Research Center, China Medical University Hospital, Taichung, Taiwan, R.O.C.,National Defence Medical Center, Taipei, Taiwan, R.O.C
| | - Cheng-Nan Wu
- Department of Medical Laboratory Science and Biotechnology, Central Taiwan University of Science and Technology, Taichung, Taiwan, R.O.C
| | - Yi-Wen Hung
- Department of Medicine Research, Taichung Veterans General Hospital, Taichung, Taiwan, R.O.C
| | - Wen-Shin Chang
- Terry Fox Cancer Research Laboratory, Translational Medical Research Center, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Chun-Kai Fu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C.,Taichung Armed Forces General Hospital, Taichung, Taiwan, R.O.C
| | - Jen-Sheng Pei
- Department of Pediatrics, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan, R.O.C
| | - Ming-Hsien Wu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C.,Taichung Armed Forces General Hospital, Taichung, Taiwan, R.O.C
| | - Yi-Liang Lai
- Taichung Armed Forces General Hospital, Taichung, Taiwan, R.O.C
| | - Yi-Min Lee
- Department of Medical Laboratory Science and Biotechnology, Central Taiwan University of Science and Technology, Taichung, Taiwan, R.O.C
| | - Shiou-Ting Yen
- Terry Fox Cancer Research Laboratory, Translational Medical Research Center, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Hsin-Ting Li
- Terry Fox Cancer Research Laboratory, Translational Medical Research Center, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Chia-Wen Tsai
- Terry Fox Cancer Research Laboratory, Translational Medical Research Center, China Medical University Hospital, Taichung, Taiwan, R.O.C. .,Department of Medical Laboratory Science and Biotechnology, Central Taiwan University of Science and Technology, Taichung, Taiwan, R.O.C
| | - DA-Tian Bau
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C. .,Terry Fox Cancer Research Laboratory, Translational Medical Research Center, China Medical University Hospital, Taichung, Taiwan, R.O.C.,Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan, R.O.C
| |
Collapse
|
31
|
Gao X, Liu J, Liu X, Li L, Zheng J. Cleavage and phosphorylation: important post-translational modifications of galectin-3. Cancer Metastasis Rev 2018; 36:367-374. [PMID: 28378189 DOI: 10.1007/s10555-017-9666-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 12/29/2022]
Abstract
As the unique chimeric member of the β-galactoside-binding protein family, galectin-3 is a multivalent and multifunctional oncogenic protein involved in multiple physiological and pathological processes, including cell growth, cell differentiation, cell adhesion, RNA splicing, cell apoptosis, and malignant transformation. Post-translational modifications can effectively increase a protein's functional diversity, either by degradation or adding chemical modifications, thus regulating activity, localization, and ligand interaction. In order to clearly understand the functional mechanisms of galectin-3 involved in normal cell biology and pathogenesis, here, we have summarized the previously reported post-translational modifications of galectin-3, including cleavage and phosphorylation. Cleavage of galectin-3 by MMPs, PSA, and proteases from parasites generated intact carbohydrate-recognition domain and N-terminal peptides of varying lengths that retained lectin binding activity but lost multivalence. Serine and tyrosine phosphorylation of galectin-3 by c-Abl, CKI, and GSK-3β could regulate its localization and associated signal transduction. Accordingly, cleavage and phosphorylation play an important role in regulating galectin-3 function via altering its multivalence, localization, and ligand interaction.
Collapse
Affiliation(s)
- Xiaoge Gao
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, 221002, People's Republic of China
| | - Jingjie Liu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, 221002, People's Republic of China
| | - Xiangye Liu
- Jiangsu Province Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu Province, 221004, People's Republic of China
| | - Liantao Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, 221002, People's Republic of China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221002, People's Republic of China
| | - Junnian Zheng
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, 221002, People's Republic of China. .,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221002, People's Republic of China. .,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, 221002, People's Republic of China.
| |
Collapse
|
32
|
Detection of Matrix Metalloproteinase Activity by Bioluminescence via Intein-Mediated Biotinylation of Luciferase. SENSORS 2018; 18:s18030875. [PMID: 29543764 PMCID: PMC5877304 DOI: 10.3390/s18030875] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 02/12/2018] [Revised: 03/13/2018] [Accepted: 03/14/2018] [Indexed: 11/16/2022]
Abstract
We report bioluminescence analysis of matrix metalloproteinase (MMP) activity in biological substances using a surface-bound luciferase probe. Intein-fused luciferase protein enables site-specific biotinylation of luciferase in the presence of N-terminus cysteine-biotin via intein-mediated splicing process, resulting in a strong association with high bioluminescence signal onto a NeutrAvidin-coated surface. When the peptide substrate for MMP-7 was inserted into a region between luciferase and intein, the biotinylated probe detected MMP-7 activity by cleaving the peptide, and surface-induced bioluminescence signal was strongly reduced in the MMP-secreted media or mouse tissue extracts, compared with that in MMP-deficient control set. Our approach is anticipated to be useful for generating biotinylated proteins and for their applications in diagnosing MMP activity in human diseases.
Collapse
|
33
|
Bolenz C, Knauf D, John A, Erben P, Steidler A, Schneider SW, Günes C, Gorzelanny C. Decreased Invasion of Urothelial Carcinoma of the Bladder by Inhibition of Matrix-Metalloproteinase 7. Bladder Cancer 2018; 4:67-75. [PMID: 29430508 PMCID: PMC5798526 DOI: 10.3233/blc-170124] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/14/2022]
Abstract
Objectives: To measure and to modulate the invasive potential of urothelial carcinoma of the bladder (UCB) cells in a standardized preclinical setting using broad-spectrum matrix-metalloproteinase (MMPs) inhibitors and specific targeting of MMP7. Materials and Methods: MMP expression levels in UCB cells were determined by quantitative real-time PCR (qRT-PCR) and gel zymographies of cell supernatants (MMP9, MMP2 and MMP1) and cell lysates (MMP7). The invasiveness of human UCB cells (HT1197 and T24/83) and human benign urothelial cells (UROtsa) was modulated by a broad-spectrum MMP inhibitor (4-Aminobenzoyl-Gly-Pro-D-Leu-D-Ala hydroxamic acid; AHA) and by MMP7 specific siRNAs. MMP7 knockdown efficiency was assessed by qRT-PCR and western blot. Invasive potential of UCB cells was measured by a standardized trans-epithelial electrical resistance (TEER) assay. Results: Different MMP secretion profiles were measured in UCB cells. The active form of MMP7 was exclusively detected in HT1197 cells. Characteristic TEER breakdown patterns were observed in UCB cells when compared to benign cells. Invasive potentials were significantly higher in HT1197 cells than in T24/83 and in UROtsa cells [14.8±5.75 vs. 1.5±0.56 and 1.2±0.15, respectively; p < 0.01]. AHA treatment reduced the invasive potential of HT1197 cells. Also the specific downregulation of MMP7 by siRNA lowered the HT1197 cell invasiveness [20±1.0 vs. 16±2.8; p < 0.05]. Neither AHA nor MMP-7 siRNA transfection altered the invasive potential of T24/83 cells. Conclusions: Invasion of UCB is partially dependent on MMPs. Specific targeting of MMP7 by siRNA reduces the invasive potential in a subgroup of UCB cells. Therefore, MMP7 represents a potential therapeutic target which warrants further investigation.
Collapse
Affiliation(s)
| | - Daniel Knauf
- Department of Urology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany
| | - Axel John
- Department of Urology, University of Ulm, Ulm, Germany
| | - Philipp Erben
- Department of Urology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany
| | - Annette Steidler
- Department of Urology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany
| | - Stefan W Schneider
- Department of Dermatology and Venerology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Cagatay Günes
- Department of Urology, University of Ulm, Ulm, Germany
| | - Christian Gorzelanny
- Department of Experimental Dermatology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany.,Department of Dermatology and Venerology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
34
|
Delker DA, Wood AC, Snow AK, Samadder NJ, Samowitz WS, Affolter KE, Boucher KM, Pappas LM, Stijleman IJ, Kanth P, Byrne KR, Burt RW, Bernard PS, Neklason DW. Chemoprevention with Cyclooxygenase and Epidermal Growth Factor Receptor Inhibitors in Familial Adenomatous Polyposis Patients: mRNA Signatures of Duodenal Neoplasia. Cancer Prev Res (Phila) 2018; 11:4-15. [PMID: 29109117 PMCID: PMC5754246 DOI: 10.1158/1940-6207.capr-17-0130] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/01/2017] [Revised: 08/31/2017] [Accepted: 10/02/2017] [Indexed: 12/20/2022]
Abstract
To identify gene expression biomarkers and pathways targeted by sulindac and erlotinib given in a chemoprevention trial with a significant decrease in duodenal polyp burden at 6 months (P < 0.001) in familial adenomatous polyposis (FAP) patients, we biopsied normal and polyp duodenal tissues from patients on drug versus placebo and analyzed the RNA expression. RNA sequencing was performed on biopsies from the duodenum of FAP patients obtained at baseline and 6-month endpoint endoscopy. Ten FAP patients on placebo and 10 on sulindac and erlotinib were selected for analysis. Purity of biopsied polyp tissue was calculated from RNA expression data. RNAs differentially expressed between endpoint polyp and paired baseline normal were determined for each group and mapped to biological pathways. Key genes in candidate pathways were further validated by quantitative RT-PCR. RNA expression analyses of endpoint polyp compared with paired baseline normal for patients on placebo and drug show that pathways activated in polyp growth and proliferation are blocked by this drug combination. Directly comparing polyp gene expression between patients on drug and placebo also identified innate immune response genes (IL12 and IFNγ) preferentially expressed in patients on drug. Gene expression analyses from tissue obtained at endpoint of the trial demonstrated inhibition of the cancer pathways COX2/PGE2, EGFR, and WNT. These findings provide molecular evidence that the drug combination of sulindac and erlotinib reached the intended tissue and was on target for the predicted pathways. Furthermore, activation of innate immune pathways from patients on drug may have contributed to polyp regression. Cancer Prev Res; 11(1); 4-15. ©2017 AACRSee related editorial by Shureiqi, p. 1.
Collapse
Affiliation(s)
- Don A Delker
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah
| | - Austin C Wood
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Angela K Snow
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - N Jewel Samadder
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Wade S Samowitz
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
- Department of Pathology, University of Utah, Salt Lake City, Utah
| | - Kajsa E Affolter
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
- Department of Pathology, University of Utah, Salt Lake City, Utah
| | - Kenneth M Boucher
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Lisa M Pappas
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Inge J Stijleman
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Priyanka Kanth
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah
| | - Kathryn R Byrne
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah
| | - Randall W Burt
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Philip S Bernard
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
- Department of Pathology, University of Utah, Salt Lake City, Utah
| | - Deborah W Neklason
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah.
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| |
Collapse
|
35
|
Agrawal L, Engel KB, Greytak SR, Moore HM. Understanding preanalytical variables and their effects on clinical biomarkers of oncology and immunotherapy. Semin Cancer Biol 2017; 52:26-38. [PMID: 29258857 DOI: 10.1016/j.semcancer.2017.12.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/26/2017] [Revised: 12/07/2017] [Accepted: 12/13/2017] [Indexed: 12/20/2022]
Abstract
Identifying a suitable course of immunotherapy treatment for a given patient as well as monitoring treatment response is heavily reliant on biomarkers detected and quantified in blood and tissue biospecimens. Suboptimal or variable biospecimen collection, processing, and storage practices have the potential to alter clinically relevant biomarkers, including those used in cancer immunotherapy. In the present review, we summarize effects reported for immunologically relevant biomarkers and highlight preanalytical factors associated with specific analytical platforms and assays used to predict and gauge immunotherapy response. Given that many of the effects introduced by preanalytical variability are gene-, transcript-, and protein-specific, biospecimen practices should be standardized and validated for each biomarker and assay to ensure accurate results and facilitate clinical implementation of newly identified immunotherapy approaches.
Collapse
Affiliation(s)
- Lokesh Agrawal
- Biorepositories and Biospecimen Research Branch (BBRB), Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, 9609 Medical Center Drive, Bethesda, Maryland, USA
| | | | | | - Helen M Moore
- Biorepositories and Biospecimen Research Branch (BBRB), Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, 9609 Medical Center Drive, Bethesda, Maryland, USA.
| |
Collapse
|
36
|
The long noncoding RNA lnc-EGFR stimulates T-regulatory cells differentiation thus promoting hepatocellular carcinoma immune evasion. Nat Commun 2017; 8:15129. [PMID: 28541302 PMCID: PMC5529670 DOI: 10.1038/ncomms15129] [Citation(s) in RCA: 262] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/16/2015] [Accepted: 03/01/2017] [Indexed: 12/12/2022] Open
Abstract
Long noncoding RNAs play a pivotal role in T-helper cell development but little is known about their roles in Treg differentiation and functions during the progression of hepatocellular carcinoma (HCC). Here, we show that lnc-epidermal growth factor receptor (EGFR) upregulation in Tregs correlates positively with the tumour size and expression of EGFR/Foxp3, but negatively with IFN-γ expression in patients and xenografted mouse models. Lnc-EGFR stimulates Treg differentiation, suppresses CTL activity and promotes HCC growth in an EGFR-dependent manner. Mechanistically, lnc-EGFR specifically binds to EGFR and blocks its interaction with and ubiquitination by c-CBL, stabilizing it and augmenting activation of itself and its downstream AP-1/NF-AT1 axis, which in turn elicits EGFR expression. Lnc-EGFR links an immunosuppressive state to cancer by promoting Treg cell differentiation, thus offering a potential therapeutic target for HCC. The role of long noncoding RNAs in regulating T-cell differentiation within the tumour microenvironment is unclear. Here the authors identify a lncRNA that, through direct interactions with EGFR, promotes T-regulatory cell differentiation within the microenvironment of hepatocellular carcinoma, thus promoting tumour growth via immune suppression.
Collapse
|
37
|
Chen S, Hu X, Zhu W, Jia C, Han X, Yuan J, Sun Z, Yang Z, Geng T, Cui H. Reactivation of development-related genes by the DNA methylation inhibitor 5-Aza-2΄-deoxycytidine in chicken embryo fibroblasts. Poult Sci 2017; 96:1007-1014. [PMID: 28158798 DOI: 10.3382/ps/pew378] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/19/2016] [Accepted: 09/14/2016] [Indexed: 11/20/2022] Open
|
38
|
Meng F, Yang H, Jack C, Zhang H, Moller A, Spivey D, Page RC, Tierney DL, Crowder MW. Biochemical characterization and zinc binding group (ZBGs) inhibition studies on the catalytic domain of MMP7 (cdMMP7). J Inorg Biochem 2016; 165:7-17. [DOI: 10.1016/j.jinorgbio.2016.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/14/2016] [Revised: 10/05/2016] [Accepted: 10/13/2016] [Indexed: 02/08/2023]
|
39
|
Yuan S, Wang L, Chen X, Fan B, Yuan Q, Zhang H, Yang D, Wang S. Triptolide inhibits the migration and invasion of human prostate cancer cells via Caveolin-1/CD147/MMPs pathway. Biomed Pharmacother 2016; 84:1776-1782. [DOI: 10.1016/j.biopha.2016.10.104] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/03/2016] [Revised: 10/25/2016] [Accepted: 10/30/2016] [Indexed: 12/15/2022] Open
|
40
|
Löffler MW, Chandran PA, Laske K, Schroeder C, Bonzheim I, Walzer M, Hilke FJ, Trautwein N, Kowalewski DJ, Schuster H, Günder M, Carcamo Yañez VA, Mohr C, Sturm M, Nguyen HP, Riess O, Bauer P, Nahnsen S, Nadalin S, Zieker D, Glatzle J, Thiel K, Schneiderhan-Marra N, Clasen S, Bösmüller H, Fend F, Kohlbacher O, Gouttefangeas C, Stevanović S, Königsrainer A, Rammensee HG. Personalized peptide vaccine-induced immune response associated with long-term survival of a metastatic cholangiocarcinoma patient. J Hepatol 2016; 65:849-855. [PMID: 27397612 PMCID: PMC5756536 DOI: 10.1016/j.jhep.2016.06.027] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 02/19/2016] [Revised: 06/16/2016] [Accepted: 06/29/2016] [Indexed: 01/06/2023]
Abstract
BACKGROUND & AIMS We report a novel experimental immunotherapeutic approach in a patient with metastatic intrahepatic cholangiocarcinoma. In the 5year course of the disease, the initial tumor mass, two local recurrences and a lung metastasis were surgically removed. Lacking alternative treatment options, aiming at the induction of anti-tumor T cells responses, we initiated a personalized multi-peptide vaccination, based on in-depth analysis of tumor antigens (immunopeptidome) and sequencing. METHODS Tumors were characterized by immunohistochemistry, next-generation sequencing and mass spectrometry of HLA ligands. RESULTS Although several tumor-specific neo-epitopes were predicted in silico, none could be validated by mass spectrometry. Instead, a personalized multi-peptide vaccine containing non-mutated tumor-associated epitopes was designed and applied. Immunomonitoring showed vaccine-induced T cell responses to three out of seven peptides administered. The pulmonary metastasis resected after start of vaccination showed strong immune cell infiltration and perforin positivity, in contrast to the previous lesions. The patient remains clinically healthy, without any radiologically detectable tumors since March 2013 and the vaccination is continued. CONCLUSIONS This remarkable clinical course encourages formal clinical studies on adjuvant personalized peptide vaccination in cholangiocarcinoma. LAY SUMMARY Metastatic cholangiocarcinomas, cancers that originate from the liver bile ducts, have very limited treatment options and a fatal prognosis. We describe a novel therapeutic approach in such a patient using a personalized multi-peptide vaccine. This vaccine, developed based on the characterization of the patient's tumor, evoked detectable anti-tumor immune responses, associating with long-term tumor-free survival.
Collapse
Affiliation(s)
- Markus W Löffler
- University Hospital Tübingen, Department of General, Visceral and Transplant Surgery, Hoppe-Seyler-Str. 3, 72076 Tübingen, Germany; University of Tübingen, Interfaculty Institute for Cell Biology, Department of Immunology, Auf der Morgenstelle 15, 72076 Tübingen, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) partner site Tübingen, Germany.
| | - P Anoop Chandran
- University of Tübingen, Interfaculty Institute for Cell Biology, Department of Immunology, Auf der Morgenstelle 15, 72076 Tübingen, Germany
| | - Karoline Laske
- University of Tübingen, Interfaculty Institute for Cell Biology, Department of Immunology, Auf der Morgenstelle 15, 72076 Tübingen, Germany; Current address: Immatics Biotechnologies GmbH, Paul Ehrlich Str. 15, 72076 Tübingen, Germany
| | - Christopher Schroeder
- University Hospital Tübingen, Institute of Medical Genetics and Applied Genomics, Calwerstr. 7, 72076 Tübingen, Germany
| | - Irina Bonzheim
- University Hospital Tübingen, Institute of Pathology, Liebermeisterstr. 8, 72076 Tübingen, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) partner site Tübingen, Germany
| | - Mathias Walzer
- University of Tübingen, Interfaculty Institute for Cell Biology, Department of Immunology, Auf der Morgenstelle 15, 72076 Tübingen, Germany; University of Tübingen, Center for Bioinformatics, Sand 14, 72076 Tübingen, Germany; University of Tübingen, Dept. of Computer Science, Sand 14, 72076 Tübingen, Germany
| | - Franz J Hilke
- University Hospital Tübingen, Institute of Medical Genetics and Applied Genomics, Calwerstr. 7, 72076 Tübingen, Germany
| | - Nico Trautwein
- University of Tübingen, Interfaculty Institute for Cell Biology, Department of Immunology, Auf der Morgenstelle 15, 72076 Tübingen, Germany
| | - Daniel J Kowalewski
- University of Tübingen, Interfaculty Institute for Cell Biology, Department of Immunology, Auf der Morgenstelle 15, 72076 Tübingen, Germany; Current address: Immatics Biotechnologies GmbH, Paul Ehrlich Str. 15, 72076 Tübingen, Germany
| | - Heiko Schuster
- University of Tübingen, Interfaculty Institute for Cell Biology, Department of Immunology, Auf der Morgenstelle 15, 72076 Tübingen, Germany
| | - Marc Günder
- University of Tübingen, Interfaculty Institute for Cell Biology, Department of Immunology, Auf der Morgenstelle 15, 72076 Tübingen, Germany
| | - Viviana A Carcamo Yañez
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany
| | - Christopher Mohr
- University of Tübingen, Center for Bioinformatics, Sand 14, 72076 Tübingen, Germany; University of Tübingen, Dept. of Computer Science, Sand 14, 72076 Tübingen, Germany
| | - Marc Sturm
- University Hospital Tübingen, Institute of Medical Genetics and Applied Genomics, Calwerstr. 7, 72076 Tübingen, Germany
| | - Huu-Phuc Nguyen
- University Hospital Tübingen, Institute of Medical Genetics and Applied Genomics, Calwerstr. 7, 72076 Tübingen, Germany
| | - Olaf Riess
- University Hospital Tübingen, Institute of Medical Genetics and Applied Genomics, Calwerstr. 7, 72076 Tübingen, Germany
| | - Peter Bauer
- University Hospital Tübingen, Institute of Medical Genetics and Applied Genomics, Calwerstr. 7, 72076 Tübingen, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) partner site Tübingen, Germany
| | - Sven Nahnsen
- University of Tübingen, Center for Bioinformatics, Sand 14, 72076 Tübingen, Germany; University of Tübingen, Quantitative Biology Center (QBiC), Auf der Morgenstelle 10, 72076 Tübingen, Germany
| | - Silvio Nadalin
- University Hospital Tübingen, Department of General, Visceral and Transplant Surgery, Hoppe-Seyler-Str. 3, 72076 Tübingen, Germany
| | - Derek Zieker
- University Hospital Tübingen, Department of General, Visceral and Transplant Surgery, Hoppe-Seyler-Str. 3, 72076 Tübingen, Germany
| | - Jörg Glatzle
- University Hospital Tübingen, Department of General, Visceral and Transplant Surgery, Hoppe-Seyler-Str. 3, 72076 Tübingen, Germany; Current address: Klinikum Konstanz, Luisenstr. 7, 78464 Konstanz, Germany
| | - Karolin Thiel
- University Hospital Tübingen, Department of General, Visceral and Transplant Surgery, Hoppe-Seyler-Str. 3, 72076 Tübingen, Germany
| | - Nicole Schneiderhan-Marra
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany
| | - Stephan Clasen
- University Hospital Tübingen, Department of Diagnostic and Interventional Radiology, Hoppe-Seyler-Str. 3, 72076 Tübingen, Germany
| | - Hans Bösmüller
- University Hospital Tübingen, Institute of Pathology, Liebermeisterstr. 8, 72076 Tübingen, Germany
| | - Falko Fend
- University Hospital Tübingen, Institute of Pathology, Liebermeisterstr. 8, 72076 Tübingen, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) partner site Tübingen, Germany
| | - Oliver Kohlbacher
- University of Tübingen, Center for Bioinformatics, Sand 14, 72076 Tübingen, Germany; University of Tübingen, Dept. of Computer Science, Sand 14, 72076 Tübingen, Germany; University of Tübingen, Quantitative Biology Center (QBiC), Auf der Morgenstelle 10, 72076 Tübingen, Germany; Max Planck Institute for Developmental Biology, Spemannstr. 35, 72076 Tübingen, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) partner site Tübingen, Germany
| | - Cécile Gouttefangeas
- University of Tübingen, Interfaculty Institute for Cell Biology, Department of Immunology, Auf der Morgenstelle 15, 72076 Tübingen, Germany
| | - Stefan Stevanović
- University of Tübingen, Interfaculty Institute for Cell Biology, Department of Immunology, Auf der Morgenstelle 15, 72076 Tübingen, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) partner site Tübingen, Germany
| | - Alfred Königsrainer
- University Hospital Tübingen, Department of General, Visceral and Transplant Surgery, Hoppe-Seyler-Str. 3, 72076 Tübingen, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) partner site Tübingen, Germany
| | - Hans-Georg Rammensee
- University of Tübingen, Interfaculty Institute for Cell Biology, Department of Immunology, Auf der Morgenstelle 15, 72076 Tübingen, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) partner site Tübingen, Germany
| |
Collapse
|
41
|
Nallar SC, Kalvakolanu DV. GRIM-19: A master regulator of cytokine induced tumor suppression, metastasis and energy metabolism. Cytokine Growth Factor Rev 2016; 33:1-18. [PMID: 27659873 DOI: 10.1016/j.cytogfr.2016.09.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/25/2016] [Accepted: 09/14/2016] [Indexed: 12/31/2022]
Abstract
Cytokines induce cell proliferation or growth suppression depending on the context. It is increasingly becoming clear that success of standard radiotherapy and/or chemotherapeutics to eradicate solid tumors is dependent on IFN signaling. In this review we discuss the molecular mechanisms of tumor growth suppression by a gene product isolated in our laboratory using a genome-wide expression knock-down strategy. Gene associated with retinoid-IFN-induced mortality -19 (GRIM-19) functions as non-canonical tumor suppressor by antagonizing oncoproteins. As a component of mitochondrial respiratory chain, GRIM-19 influences the degree of "Warburg effect" in cancer cells as many advanced and/or aggressive tumors show severely down-regulated GRIM-19 levels. In addition, GRIM-19 appears to regulate innate and acquired immune responses in mouse models. Thus, GRIM-19 is positioned at nodes that favor cell protection and/or prevent aberrant cell growth.
Collapse
Affiliation(s)
- Shreeram C Nallar
- Department of Microbiology and Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Dhan V Kalvakolanu
- Department of Microbiology and Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
42
|
Feng HY, Chen YC. Role of bile acids in carcinogenesis of pancreatic cancer: An old topic with new perspective. World J Gastroenterol 2016; 22:7463-77. [PMID: 27672269 PMCID: PMC5011662 DOI: 10.3748/wjg.v22.i33.7463] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 04/09/2016] [Revised: 06/30/2016] [Accepted: 07/20/2016] [Indexed: 02/06/2023] Open
Abstract
The role of bile acids in colorectal cancer has been well documented, but their role in pancreatic cancer remains unclear. In this review, we examined the risk factors of pancreatic cancer. We found that bile acids are associated with most of these factors. Alcohol intake, smoking, and a high-fat diet all lead to high secretion of bile acids, and bile acid metabolic dysfunction is a causal factor of gallstones. An increase in secretion of bile acids, in addition to a long common channel, may result in bile acid reflux into the pancreatic duct and to the epithelial cells or acinar cells, from which pancreatic adenocarcinoma is derived. The final pathophysiological process is pancreatitis, which promotes dedifferentiation of acinar cells into progenitor duct-like cells. Interestingly, bile acids act as regulatory molecules in metabolism, affecting adipose tissue distribution, insulin sensitivity and triglyceride metabolism. As a result, bile acids are associated with three risk factors of pancreatic cancer: obesity, diabetes and hypertriglyceridemia. In the second part of this review, we summarize several studies showing that bile acids act as cancer promoters in gastrointestinal cancer. However, more question are raised than have been solved, and further oncological and physiological experiments are needed to confirm the role of bile acids in pancreatic cancer carcinogenesis.
Collapse
|
43
|
Whitington T, Gao P, Song W, Ross-Adams H, Lamb AD, Yang Y, Svezia I, Klevebring D, Mills IG, Karlsson R, Halim S, Dunning MJ, Egevad L, Warren AY, Neal DE, Grönberg H, Lindberg J, Wei GH, Wiklund F. Gene regulatory mechanisms underpinning prostate cancer susceptibility. Nat Genet 2016; 48:387-97. [PMID: 26950096 DOI: 10.1038/ng.3523] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/22/2015] [Accepted: 02/08/2016] [Indexed: 12/29/2022]
Abstract
Molecular characterization of genome-wide association study (GWAS) loci can uncover key genes and biological mechanisms underpinning complex traits and diseases. Here we present deep, high-throughput characterization of gene regulatory mechanisms underlying prostate cancer risk loci. Our methodology integrates data from 295 prostate cancer chromatin immunoprecipitation and sequencing experiments with genotype and gene expression data from 602 prostate tumor samples. The analysis identifies new gene regulatory mechanisms affected by risk locus SNPs, including widespread disruption of ternary androgen receptor (AR)-FOXA1 and AR-HOXB13 complexes and competitive binding mechanisms. We identify 57 expression quantitative trait loci at 35 risk loci, which we validate through analysis of allele-specific expression. We further validate predicted regulatory SNPs and target genes in prostate cancer cell line models. Finally, our integrated analysis can be accessed through an interactive visualization tool. This analysis elucidates how genome sequence variation affects disease predisposition via gene regulatory mechanisms and identifies relevant genes for downstream biomarker and drug development.
Collapse
Affiliation(s)
- Thomas Whitington
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Ping Gao
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Wei Song
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Helen Ross-Adams
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Alastair D Lamb
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.,Department of Urology, Addenbrooke's Hospital, Cambridge, UK
| | - Yuehong Yang
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Ilaria Svezia
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Daniel Klevebring
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Ian G Mills
- Prostate Cancer Research Group, Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo and Oslo University Hospital, Oslo, Norway.,Department of Molecular Oncology, Institute of Cancer Research, Oslo University Hospital, Oslo, Norway.,Prostate Cancer UK/Movember Centre of Excellence for Prostate Cancer Research, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Robert Karlsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Silvia Halim
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.,Cancer Research UK Beatson Institute, Glasgow, UK
| | - Mark J Dunning
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Lars Egevad
- Department of Pathology and Cytology, Karolinska University Hospital, Stockholm, Sweden.,Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Anne Y Warren
- Department of Pathology, Addenbrooke's Hospital, Cambridge, UK
| | - David E Neal
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Henrik Grönberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Johan Lindberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Gong-Hong Wei
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Fredrik Wiklund
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
44
|
Su G, Letcher RJ, Crump D, Farmahin R, Giesy JP, Kennedy SW. Sunlight Irradiation of Highly Brominated Polyphenyl Ethers Generates Polybenzofuran Products That Alter Dioxin-responsive mRNA Expression in Chicken Hepatocytes. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2016; 50:2318-2327. [PMID: 26854739 DOI: 10.1021/acs.est.5b04939] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 06/05/2023]
Abstract
We report on two highly brominated polyphenyl ether flame retardants, tetradecabromo-1,4- diphenoxybenzene (TeDB-DiPhOBz) and 2,2',3,3',4,4',5,5',6,6'-decabromodiphenyl ether (BDE-209), that formed photolytic degradation products in tetrahydrofuran (THF)/hexane solvent after 21 days of natural sunlight irradiation (SI). These degradation products of SI-TeDB-DiPhOBz and SI-BDE-209 included the numerous polybrominated homologue groups of polybenzofurans and dibenzofurans, respectively. Formation of similar polybenzofuran and dibenzofuran products was also observed following a 3 month exposure of the solid powder forms of TeDB-DiPhOBz and BDE-209 to natural SI. These resulting degradation product mixtures were administered to chicken embryonic hepatocytes (CEH) to determine effects on mRNA expression levels of 27 dioxin-responsive genes. For the solvent-based SI study, equivalent concentrations of 1 or 25 μM of SI-TeDB-DiPhOBz or 1 or 10 μM of SI-BDE-209 resulted in gene expression profiles that were similar to those of the most potent dioxin-like compound, 2,3,7,8-tetrachlorodibenzo-p-dioxin. In addition, a concentration-dependent induction of CYP1A4 and CYP1A5 mRNA was observed following exposure to SI-TeDB-DiPhOBz and SI-BDE-209. Based on ECthreshold values for CYP1A4/5 mRNA expression, relative potency (ReP) values were 1 × 10(-6) and 1 × 10(-5) for SI-TeDB-DiPhOBz and SI-BDE-209, respectively. The SI TeDB-DiPhOBz and BDE-209 powder degradation product mixture also significantly induced CYP1A4 mRNA levels in CEH. Our findings clearly show that the environmental stability of TeDB-DiPhOBz and BDE-209, and possibly other highly brominated polyphenyl ethers, is of great concern from a dioxin-like degradation products and toxicity perspective.
Collapse
Affiliation(s)
- Guanyong Su
- Ecotoxicology and Wildlife Health Division, Environment and Climate Change Canada, National Wildlife Research Centre, Carleton University , Ottawa, Ontario K1A 0H3, Canada
- Department of Chemistry, Carleton University , Ottawa, Ontario K1S 5B6, Canada
| | - Robert J Letcher
- Ecotoxicology and Wildlife Health Division, Environment and Climate Change Canada, National Wildlife Research Centre, Carleton University , Ottawa, Ontario K1A 0H3, Canada
- Department of Chemistry, Carleton University , Ottawa, Ontario K1S 5B6, Canada
| | - Doug Crump
- Ecotoxicology and Wildlife Health Division, Environment and Climate Change Canada, National Wildlife Research Centre, Carleton University , Ottawa, Ontario K1A 0H3, Canada
| | - Reza Farmahin
- Ecotoxicology and Wildlife Health Division, Environment and Climate Change Canada, National Wildlife Research Centre, Carleton University , Ottawa, Ontario K1A 0H3, Canada
- Centre for Advanced Research in Environmental Genomics, Department of Biology, University of Ottawa , Ottawa, Ontario K1N 6N5, Canada
| | - John P Giesy
- Department of Veterinary Biomedical Sciences and Toxicology Centre, University of Saskatchewan , Saskatoon, Saskatchewan S7N 5B3, Canada
- Department of Zoology and Center for Integrative Toxicology, Michigan State University , East Lansing, Michigan 48824, United States
| | - Sean W Kennedy
- Ecotoxicology and Wildlife Health Division, Environment and Climate Change Canada, National Wildlife Research Centre, Carleton University , Ottawa, Ontario K1A 0H3, Canada
- Centre for Advanced Research in Environmental Genomics, Department of Biology, University of Ottawa , Ottawa, Ontario K1N 6N5, Canada
| |
Collapse
|
45
|
Zhang X, Huang S, Guo J, Zhou L, You L, Zhang T, Zhao Y. Insights into the distinct roles of MMP-11 in tumor biology and future therapeutics (Review). Int J Oncol 2016; 48:1783-93. [PMID: 26892540 DOI: 10.3892/ijo.2016.3400] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/19/2015] [Accepted: 01/21/2016] [Indexed: 11/06/2022] Open
Abstract
The biological processes of cancer cells such as tumorigenesis, proliferation, angiogenesis, apoptosis and invasion are greatly influenced by the surrounding microenvironment. The ability of solid malignant tumors to alter the microenvironment represents an important characteristic through which tumor cells are able to acquire specific functions necessary for their malignant biological behaviors. Matrix metalloproteinases (MMPs) are a family of zinc-dependent endopeptidases with the capacity of remodeling extracellular matrix (ECM) by degrading almost all ECM proteins, which plays essential roles during the invasion and metastasis process of solid malignant tumors, including allowing tumor cells to modify the ECM components and release cytokines, ultimately facilitating protease-dependent tumor progression. MMP-11, also named stromelysin-3, is a member of the stromelysin subgroup belonging to MMPs superfamily, which has been detected in cancer cells, stromal cells and adjacent microenvironment. Differently, MMP-11 exerts a dual effect on tumors. On the one hand MMP-11 promotes cancer development by inhibiting apoptosis as well as enhancing migration and invasion of cancer cells, on the other hand MMP-11 plays a negative role against cancer development via suppressing metastasis in animal models. Overexpression of MMP-11 was discovered in sera of cancer patients compared with normal control group as well as in multiple tumor tissue specimens, such as gastric cancer, breast cancer, and pancreatic cancer. At present, some evidence supports that MMP-11 may work as a significant tumor biomarker for early detection of cancer, tumor staging, prognostic analysis, monitoring recurrence during follow-up and also a potential target for immunotherapy against cancer. In view of the importance of MMP-11 in modifying tumor microenvironment and potent antitumoral effects on solid tumors, there is an urgent need for a deeper understanding of how MMP-11 modulates tumor progression, and exploring its potential clinical application.
Collapse
Affiliation(s)
- Xu Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, P.R. China
| | - Shuai Huang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, P.R. China
| | - Junchao Guo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, P.R. China
| | - Li Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, P.R. China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, P.R. China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, P.R. China
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, P.R. China
| |
Collapse
|
46
|
Roscilli G, Cappelletti M, De Vitis C, Ciliberto G, Di Napoli A, Ruco L, Mancini R, Aurisicchio L. Circulating MMP11 and specific antibody immune response in breast and prostate cancer patients. J Transl Med 2014; 12:54. [PMID: 24564996 PMCID: PMC3936832 DOI: 10.1186/1479-5876-12-54] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/30/2013] [Accepted: 02/04/2014] [Indexed: 01/04/2023] Open
Abstract
Background Tumor Associated Antigens are characterized by spontaneous immune response in cancer patients as a consequence of overexpression and epitope-presentation on MHC class I/II machinery. Matrix Metalloprotease 11 (MMP11) expression has been associated with poor prognosis for several cancer types, including breast and prostate cancer. Methods MMP11 expression was determined by immunoistochemistry in breast and prostate cancer samples. Circulating MMP11 protein as well as the spontaneous immune responses against MMP11 were analyzed in a set of breast and prostate cancer patients. Results In plasma samples MMP11 protein was present in 5/13 breast cancer patients and in 1/12 prostate cancer patients. An antibody response was observed in 7/13 breast cancer patients and in 3/12 prostate cancer patients. Conclusions These findings further suggest MMP11 as a promising biomarker for these tumor types and a suitable target for cancer immunotherapy strategies.
Collapse
|
47
|
Huang Y, Yu H, Lei H, Xie C, Zhong Y. Matrix metalloproteinase 7 is a useful marker for 5-fluorouracil-based adjuvant chemotherapy in stage II and stage III colorectal cancer patients. Med Oncol 2014; 31:824. [PMID: 24469951 DOI: 10.1007/s12032-013-0824-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/23/2013] [Accepted: 12/18/2013] [Indexed: 12/23/2022]
Abstract
Matrix metalloproteinase 7 (MMP7) was reported to be a negative regulator in Fas-mediated apoptosis. The mechanism of cell killing associated with 5-FU treatment in colon cancer was also closely related to Fas-induced apoptosis, which implied that the expression level of MMP7 in colorectal cancer may be associated with the sensitivity of 5-FU treatment. To prove the hypothesis, first we verified the negative relevance between the colorectal cancer cells apoptosis in response to 5-FU treatment and MMP7 level by MTT and flow cytometry assay in vitro. Further, we found the apoptosis was in a positive relation with the Fas ligand level collected from the medium, suggesting a Fas-induced apoptosis. We found that increased level of MMP7 resulted in the enhanced drug resistance in SW620 colon cancer cells treated with 5-FU in vitro. Besides, we analyzed the influence of MMP7 on prognosis of 76 patients with TNM stage II-III colorectal cancers who had undergone curative resections and received 5-FU-based adjuvant chemotherapy. The expression of MMP7 was detected by IHC, and the relationship between the expression of MMP7 and disease-free survival was analyzed by univariate analysis and multivariate analysis. Patients with higher expression of MMP7 showed inferior disease-free survival (p=0.007), and high expression of MMP7 was a significant independent unfavorable prognostic factor (p=0.012). These data suggested that MMP7 is a useful marker for 5-FU chemotherapy sensitivity in patients with stage II-III colorectal cancer.
Collapse
Affiliation(s)
- Yong Huang
- Department of Radio-Chemo Therapy, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | | | | | | | | |
Collapse
|
48
|
Li HD, Huang C, Huang KJ, Wu WD, Jiang T, Cao J, Feng ZZ, Qiu ZJ. STAT3 knockdown reduces pancreatic cancer cell invasiveness and matrix metalloproteinase-7 expression in nude mice. PLoS One 2011; 6:e25941. [PMID: 21991388 PMCID: PMC3185063 DOI: 10.1371/journal.pone.0025941] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/20/2011] [Accepted: 09/13/2011] [Indexed: 01/15/2023] Open
Abstract
Aims Transducer and activator of transcription-3 (STAT3) plays an important role in tumor cell invasion and metastasis. The aim of the present study was to investigate the effects of STAT3 knockdown in nude mouse xenografts of pancreatic cancer cells and underlying gene expression. Methods A STAT3 shRNA lentiviral vector was constructed and infected into SW1990 cells. qRT-PCR and western immunoblot were performed to detect gene expression. Nude mouse xenograft assays were used to assess changes in phenotypes of these stable cells in vivo. HE staining was utilized to evaluate tumor cell invasion and immunohistochemistry was performed to analyze gene expression. Results STAT3 shRNA successfully silenced expression of STAT3 mRNA and protein in SW1990 cells compared to control cells. Growth rate of the STAT3-silenced tumor cells in nude mice was significantly reduced compared to in the control vector tumors and parental cells-generated tumors. Tumor invasion into the vessel and muscle were also suppressed in the STAT3-silenced tumors compared to controls. Collagen IV expression was complete and continuous surrounding the tumors of STAT3-silenced SW1990 cells, whereas collagen IV expression was incomplete and discontinuous surrounding the control tumors. Moreover, microvessel density was significantly lower in STAT3-silenced tumors than parental or control tumors of SW1990 cells. In addition, MMP-7 expression was reduced in STAT3-silenced tumors compared to parental SW1990 xenografts and controls. In contrast, expression of IL-1β and IgT7α was not altered. Conclusion These data clearly demonstrate that STAT3 plays an important role in regulation of tumor growth, invasion, and angiogenesis, which could be act by reducing MMP-7 expression in pancreatic cancer cells.
Collapse
Affiliation(s)
- Hai dong Li
- Department of General Surgery, Affiliated First People's Hospital, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Pancreas Disease, Shanghai, China
- Pancreatic Cancer Center of Shanghai Jiao Tong University, Shanghai, China
| | - Chen Huang
- Department of General Surgery, Affiliated First People's Hospital, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Pancreas Disease, Shanghai, China
- Pancreatic Cancer Center of Shanghai Jiao Tong University, Shanghai, China
| | - Ke jian Huang
- Department of General Surgery, Affiliated First People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wei dong Wu
- Department of General Surgery, Affiliated First People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Tao Jiang
- Department of General Surgery, Affiliated First People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Cao
- Department of General Surgery, Affiliated First People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zhen zhong Feng
- Department of Pathology, Shanghai Jiao Tong University-Affiliated First People's Hospital, Shanghai, China
| | - Zheng jun Qiu
- Department of General Surgery, Affiliated First People's Hospital, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Pancreas Disease, Shanghai, China
- Pancreatic Cancer Center of Shanghai Jiao Tong University, Shanghai, China
- * E-mail:
| |
Collapse
|
49
|
Nolting A, Dugast AS, Rihn S, Luteijn R, Carrington MF, Kane K, Jost S, Toth I, Nagami E, Faetkenheuer G, Hartmann P, Altfeld M, Alter G. MHC class I chain-related protein A shedding in chronic HIV-1 infection is associated with profound NK cell dysfunction. Virology 2010; 406:12-20. [PMID: 20667578 PMCID: PMC2932841 DOI: 10.1016/j.virol.2010.05.014] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/12/2010] [Revised: 03/01/2010] [Accepted: 05/12/2010] [Indexed: 11/24/2022]
Abstract
Natural killer (NK) cells play a critical role in host defense against viral infections. However chronic HIV-1 infection is associated with an accumulation of dysfunctional NK cells, that poorly control viral replication. The underlying mechanisms for this NK cell mediated dysfunction are not understood. Certain tumors evade NK cell mediated detection by dampening NK cell activity through the downregulation of NKG2D, via the release of soluble NKG2D-ligands, resulting in a potent suppression of NK cell function. Here we show that chronic HIV-1 infection is associated with a specific defect in NKG2D-mediated NK cell activation, due to reduced expression and transcription of NKG2D. Reduced NKG2D expression was associated with elevated levels of the soluble form of the NKG2D-ligand, MICA, in patient sera, likely released by HIV+CD4+ T cells. Thus, like tumors, HIV-1 may indirectly suppress NK cell recognition of HIV-1-infected CD4+ T cells by enhancing NKG2D-ligand secretion into the serum resulting in a profound impairment of NK cell function.
Collapse
Affiliation(s)
- Anne Nolting
- Ragon Institute of Massachusetts General Hospital, Harvard University and Massachusetts Institute of Technology (former known as Partners AIDS Research Center of Massachusetts General Hospital) Boston, MA
- Infectious Diseases Department of the University Hospital of Cologne, Germany
| | - Anne-Sophie Dugast
- Ragon Institute of Massachusetts General Hospital, Harvard University and Massachusetts Institute of Technology (former known as Partners AIDS Research Center of Massachusetts General Hospital) Boston, MA
| | - Suzannah Rihn
- Ragon Institute of Massachusetts General Hospital, Harvard University and Massachusetts Institute of Technology (former known as Partners AIDS Research Center of Massachusetts General Hospital) Boston, MA
| | - Rutger Luteijn
- Ragon Institute of Massachusetts General Hospital, Harvard University and Massachusetts Institute of Technology (former known as Partners AIDS Research Center of Massachusetts General Hospital) Boston, MA
| | - Mary F. Carrington
- Ragon Institute of Massachusetts General Hospital, Harvard University and Massachusetts Institute of Technology (former known as Partners AIDS Research Center of Massachusetts General Hospital) Boston, MA
| | - Katherine Kane
- Ragon Institute of Massachusetts General Hospital, Harvard University and Massachusetts Institute of Technology (former known as Partners AIDS Research Center of Massachusetts General Hospital) Boston, MA
| | - Stephanie Jost
- Ragon Institute of Massachusetts General Hospital, Harvard University and Massachusetts Institute of Technology (former known as Partners AIDS Research Center of Massachusetts General Hospital) Boston, MA
| | - Ildiko Toth
- Ragon Institute of Massachusetts General Hospital, Harvard University and Massachusetts Institute of Technology (former known as Partners AIDS Research Center of Massachusetts General Hospital) Boston, MA
| | - Ellen Nagami
- Ragon Institute of Massachusetts General Hospital, Harvard University and Massachusetts Institute of Technology (former known as Partners AIDS Research Center of Massachusetts General Hospital) Boston, MA
| | - Gerd Faetkenheuer
- Infectious Diseases Department of the University Hospital of Cologne, Germany
| | - Pia Hartmann
- Infectious Diseases Department of the University Hospital of Cologne, Germany
| | - Marcus Altfeld
- Ragon Institute of Massachusetts General Hospital, Harvard University and Massachusetts Institute of Technology (former known as Partners AIDS Research Center of Massachusetts General Hospital) Boston, MA
| | - Galit Alter
- Ragon Institute of Massachusetts General Hospital, Harvard University and Massachusetts Institute of Technology (former known as Partners AIDS Research Center of Massachusetts General Hospital) Boston, MA
| |
Collapse
|
50
|
Abstract
Renal cell carcinoma (RCC) remains a significant health concern that frequently presents as metastatic disease at the time of initial diagnosis. Current first-line therapeutics for the advanced-stage RCC include antiangiogenic drugs that have yielded high rates of objective clinical response; however, these tend to be transient in nature, with many patients becoming refractory to chronic treatment with these agents. Adjuvant immunotherapies remain viable candidates to sustain disease-free and overall patient survival. In particular, vaccines designed to optimize the activation, maintenance, and recruitment of specific immunity within or into the tumor site continue to evolve. Based on the integration of increasingly refined immunomonitoring systems in both translational models and clinical trials, allowing for the improved understanding of treatment mechanism(s) of action, further refined (combinational) vaccine protocols are currently being developed and evaluated. This review provides a brief history of RCC vaccine development, discusses the successes and limitations in such approaches, and provides a rationale for developing combinational vaccine approaches that may provide improved clinical benefits to patients with RCC.
Collapse
Affiliation(s)
- Nina Chi
- Department of immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | | | | | | |
Collapse
|