1
|
Islam ME, Debnath KC, Moniruzzaman R, Okuyama K, Islam S, Dongre HN. Biological implications of decoding the extracellular matrix of vulva cancer. Oncol Rep 2025; 53:19. [PMID: 39670289 DOI: 10.3892/or.2024.8852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/15/2024] [Indexed: 12/14/2024] Open
Abstract
The present review aimed to elucidate the roles of extracellular matrix (ECM) components in the progression of vulvar squamous cell carcinoma (VSCC) and explore potential therapeutic avenues for this type of malignancy. This exploration holds promise for identifying precise molecular targets within the ECM milieu, thus facilitating the development of innovative therapeutic modalities tailored to disrupt these interactions and ultimately improve patient outcomes in VSCC. The dysregulated ECM serves as a potent driver of SCC tumor progression, orchestrating key processes such as angiogenesis, inflammation and stromal cell behavior. Yet, the exploration of ECM role in VSCC is still in its early stages. Recent research highlights the critical role of ECM organization and expression within the tumor microenvironment (TME) in influencing key aspects of VSCC, including tumor staging, grading, metastasis, invasion and patient survival. Cancer‑associated fibroblasts play a pivotal role in this dynamic by engaging in reciprocal interactions with VSCC cells, leading to significant ECM alterations and creating an immune‑suppressive TME. This hinders antitumor immunity and fosters therapeutic resistance in VSCC treatment. The dysregulated ECM in VSCC drives tumor progression, metastasis and affects patient survival. Targeting ECM, along with emerging therapies such as immune checkpoint blockade, offers promise for improved VSCC treatment outcomes.
Collapse
Affiliation(s)
- Mohammad Emranul Islam
- Department of Oral and Maxillofacial Surgery, City Dental College and Hospital, 1229 Dhaka, Bangladesh
| | - Kala Chand Debnath
- Department of Head and Neck Surgery, The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Rohan Moniruzzaman
- Department of Pathology, The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kohei Okuyama
- Department of Head and Neck Surgery, The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shajedul Islam
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Harsh Nitin Dongre
- Center for Cancer Biomarkers and Gade Laboratory for Pathology, Institute of Clinical Medicine, University of Bergen, 5021 Bergen, Norway
| |
Collapse
|
2
|
Morcos CA, Haiba NS, Bassily RW, Abu-Serie MM, El-Yazbi AF, Soliman OA, Khattab SN, Teleb M. Structure optimization and molecular dynamics studies of new tumor-selective s-triazines targeting DNA and MMP-10/13 for halting colorectal and secondary liver cancers. J Enzyme Inhib Med Chem 2024; 39:2423174. [PMID: 39513468 PMCID: PMC11552285 DOI: 10.1080/14756366.2024.2423174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 10/20/2024] [Accepted: 10/24/2024] [Indexed: 11/15/2024] Open
Abstract
A series of triazole-tethered triazines bearing pharmacophoric features of DNA-targeting agents and non-hydroxamate MMPs inhibitors were synthesized and screened against HCT-116, Caco-2 cells, and normal colonocytes by MTT assay. 7a and 7g surpassed doxorubicin against HCT-116 cells regarding potency (IC50 = 0.87 and 1.41 nM) and safety (SI = 181.93 and 54.41). 7g was potent against liver cancer (HepG-2; IC50 = 65.08 nM), the main metastatic site of CRC with correlation to MMP-13 expression. Both derivatives induced DNA damage at 2.67 and 1.87 nM, disrupted HCT-116 cell cycle and triggered apoptosis by 33.17% compared to doxorubicin (DNA damage at 0.76 nM and 40.21% apoptosis induction). 7g surpassed NNGH against MMP-10 (IC50 = 0.205 μM) and MMP-13 (IC50 = 0.275 μM) and downregulated HCT-116 VEGF related to CRC progression by 38%. Docking and MDs simulated ligands-receptors binding modes and highlighted SAR. Their ADMET profiles, drug-likeness and possible off-targets were computationally predicted.
Collapse
Affiliation(s)
- Christine A. Morcos
- Chemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Nesreen S. Haiba
- Department of Physics and Chemistry, Faculty of Education, Alexandria University, Alexandria, Egypt
| | - Rafik W. Bassily
- Chemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Marwa M. Abu-Serie
- Medical Biotechnology Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab, Egypt
| | - Amira F. El-Yazbi
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Omar A. Soliman
- Department of Human Genetics, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Sherine N. Khattab
- Chemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Mohamed Teleb
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Faculty of Pharmacy, Alamein International University (AIU), Alamein City, Egypt
| |
Collapse
|
3
|
Iannelli F, Lombardi R, Costantini S, Roca MS, Addi L, Bruzzese F, Di Gennaro E, Budillon A, Pucci B. Integrated proteomics and metabolomics analyses reveal new insights into the antitumor effects of valproic acid plus simvastatin combination in a prostate cancer xenograft model associated with downmodulation of YAP/TAZ signaling. Cancer Cell Int 2024; 24:381. [PMID: 39550583 PMCID: PMC11569608 DOI: 10.1186/s12935-024-03573-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 11/11/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND Despite advancements in therapeutic approaches, including taxane-based chemotherapy and androgen receptor-targeting agents, metastatic castration-resistant prostate cancer (mCRPC) remains an incurable tumor, highlighting the need for novel strategies that can target the complexities of this disease and bypass the development of drug resistance mechanisms. We previously demonstrated the synergistic antitumor interaction of valproic acid (VPA), an antiepileptic agent with histone deacetylase inhibitory activity, with the lipid-lowering drug simvastatin (SIM). This combination sensitizes mCRPC cells to docetaxel treatment both in vitro and in vivo by targeting the cancer stem cell compartment via mevalonate pathway/YAP axis modulation. METHODS Here, using a combined proteomic and metabolomic/lipidomic approach, we characterized tumor samples derived from 22Rv1 mCRPC cell-xenografted mice treated with or without VPA/SIM and performed an in-depth bioinformatics analysis. RESULTS We confirmed the specific impact of VPA/SIM on the Hippo-YAP signaling pathway, which is functionally related to the modulation of cancer-related extracellular matrix biology and metabolic reprogramming, providing further insights into the molecular mechanism of the antitumor effects of VPA/SIM. CONCLUSIONS In this study, we present an in-depth exploration of the potential to repurpose two generic, safe drugs for mCRPC treatment, valproic acid (VPA) and simvastatin (SIM), which already show antitumor efficacy in combination, primarily affecting the cancer stem cell compartment via MVP/YAP axis modulation. Bioinformatics analysis of the LC‒MS/MS and 1H‒NMR metabolomics/lipidomics results confirmed the specific impact of VPA/SIM on Hippo-YAP.
Collapse
Affiliation(s)
- Federica Iannelli
- Experimental Pharmacology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, 80131, Italy
| | - Rita Lombardi
- Experimental Animal Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, 80131, Italy
| | - Susan Costantini
- Experimental Pharmacology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, 80131, Italy
| | - Maria Serena Roca
- Experimental Pharmacology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, 80131, Italy
| | - Laura Addi
- Experimental Pharmacology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, 80131, Italy
| | - Francesca Bruzzese
- Experimental Animal Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, 80131, Italy
| | - Elena Di Gennaro
- Experimental Pharmacology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, 80131, Italy
| | - Alfredo Budillon
- Scientific Directorate, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Via M. Semmola, Napoli, 80131, Italy.
| | - Biagio Pucci
- Experimental Pharmacology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, 80131, Italy
| |
Collapse
|
4
|
Agraval H, Kandhari K, Yadav UCS. MMPs as potential molecular targets in epithelial-to-mesenchymal transition driven COPD progression. Life Sci 2024; 352:122874. [PMID: 38942362 DOI: 10.1016/j.lfs.2024.122874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/17/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is the third leading cause of mortality globally and the risk of developing lung cancer is six times greater in individuals with COPD who smoke compared to those who do not smoke. Matrix metalloproteinases (MMPs) play a crucial role in the pathophysiology of respiratory diseases by promoting inflammation and tissue degradation. Furthermore, MMPs are involved in key processes like epithelial-to-mesenchymal transition (EMT), metastasis, and invasion in lung cancer. While EMT has traditionally been associated with the progression of lung cancer, recent research highlights its active involvement in individuals with COPD. Current evidence underscores its role in orchestrating airway remodeling, fostering airway fibrosis, and contributing to the potential for malignant transformation in the complex pathophysiology of COPD. The precise regulatory roles of diverse MMPs in steering EMT during COPD progression needs to be elucidated. Additionally, the less-understood aspect involves how these MMPs bi-directionally activate or regulate various EMT-associated signaling cascades during COPD progression. This review article explores recent advancements in understanding MMPs' role in EMT during COPD progression and various pharmacological approaches to target MMPs. It also delves into the limitations of current MMP inhibitors and explores novel, advanced strategies for inhibiting MMPs, potentially offering new avenues for treating respiratory diseases.
Collapse
Affiliation(s)
- Hina Agraval
- Department of Medicine, National Jewish Health, Denver, CO 80206, USA
| | - Kushal Kandhari
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Umesh C S Yadav
- Special Center for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
5
|
Sastriques-Dunlop S, Elizondo-Benedetto S, Arif B, Meade R, Zaghloul MS, Luehmann H, Heo GS, English SJ, Liu Y, Zayed MA. Ketosis prevents abdominal aortic aneurysm rupture through C-C chemokine receptor type 2 downregulation and enhanced extracellular matrix balance. Sci Rep 2024; 14:1438. [PMID: 38228786 PMCID: PMC10791699 DOI: 10.1038/s41598-024-51996-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 01/12/2024] [Indexed: 01/18/2024] Open
Abstract
Abdominal aortic aneurysms (AAAs) are prevalent with aging, and AAA rupture is associated with increased mortality. There is currently no effective medical therapy to prevent AAA rupture. The monocyte chemoattractant protein (MCP-1)/C-C chemokine receptor type 2 (CCR2) axis critically regulates AAA inflammation, matrix-metalloproteinase (MMP) production, and extracellular matrix (ECM) stability. We therefore hypothesized that a diet intervention that can modulate CCR2 axis may therapeutically impact AAA risk of rupture. Since ketone bodies (KBs) can trigger repair mechanisms in response to inflammation, we evaluated whether systemic ketosis in vivo could reduce CCR2 and AAA progression. Male Sprague-Dawley rats underwent surgical AAA formation using porcine pancreatic elastase and received daily β-aminopropionitrile to promote AAA rupture. Rats with AAAs received either a standard diet, ketogenic diet (KD), or exogenous KBs (EKB). Rats receiving KD and EKB reached a state of ketosis and had significant reduction in AAA expansion and incidence of rupture. Ketosis also led to significantly reduced aortic CCR2 content, improved MMP balance, and reduced ECM degradation. Consistent with these findings, we also observed that Ccr2-/- mice have significantly reduced AAA expansion and rupture. In summary, this study demonstrates that CCR2 is essential for AAA expansion, and that its modulation with ketosis can reduce AAA pathology. This provides an impetus for future clinical studies that will evaluate the impact of ketosis on human AAA disease.
Collapse
Affiliation(s)
- Sergio Sastriques-Dunlop
- Section of Vascular Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Santiago Elizondo-Benedetto
- Section of Vascular Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Batool Arif
- Section of Vascular Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Rodrigo Meade
- Section of Vascular Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Mohamed S Zaghloul
- Section of Vascular Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Hannah Luehmann
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Gyu S Heo
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Sean J English
- Section of Vascular Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Yongjian Liu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Mohamed A Zayed
- Section of Vascular Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA.
- Division of Molecular Cell Biology, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University, St. Louis, MO, USA.
- Veterans Affairs St. Louis Health Care System, St. Louis, MO, USA.
| |
Collapse
|
6
|
Okamura K, Sato M, Suzuki T, Nohara K. Arsenite exposure induces premature senescence and senescence-associated secretory phenotype (SASP) in human hepatocyte-derived cell line Huh-7. Environ Health Prev Med 2024; 29:74. [PMID: 39756915 DOI: 10.1265/ehpm.24-00139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND Chronic arsenite exposure has been known to induce cancer in various organs; however, the underlying mechanisms remain elusive. The characteristic feature of carcinogenesis due to arsenic exposure is that the disease develops after a prolonged latent period, even after cessation of exposure. Our previous study revealed that arsenite exposure induces premature senescence in hepatic stellate cells and suggests that the senescence-associated secretory phenotype (SASP) factors from the senescent cells promote hepatic carcinogenesis. However, arsenite exposure in the liver occurs not only in hepatic stellate cells, but also in hepatocytes. Therefore, we examined whether arsenite exposure in hepatocytes also causes premature senescence and the enhancement of SASP factors. We also assessed whether those effects remained after cessation of arsenite exposure. METHODS Human hepatocyte-derived cell line Huh-7 was exposed to sodium arsenite for 72 hours to determine the concentration at which cell proliferation was inhibited. In the 5 µM of exposure, various cellular senescence markers and SASP factors were analyzed and compared with unexposed cells. We also examined whether those senescence markers and SASP factors were maintained after cessation of arsenite exposure. Finally, we explored whether the increased expression of SASP factor, which was upregulated in hepatocytes by arsenic exposure in this study, is related to the prognosis of human hepatocellular carcinoma. RESULTS After exposure to 5 µM of sodium arsenite for 72 hours, various senescent features, such as the induction of P21 mRNA, the reduction of LAMINB1 mRNA, morphological changes, phosphorylation of P53, and the presence of SA-β-gal positive cells were observed. Those changes were maintained after cessation of arsenite exposure. In addition, mRNA levels of SASP factors (MMP1, MMP3, MMP10, GDF15, PAI-1, and IL-6) were increased after arsenite exposure, and their high expression levels were maintained after cessation of arsenite exposure. Furthermore, by analyzing the TCGA database, we found that the increased expression levels of many SASP factors negatively correlated with prognosis. CONCLUSIONS Arsenite exposure induces premature senescence in hepatocyte-derived cells and increases SASP factors that are related to hepatic tumorigenesis. Once arsenite exposure induces premature senescence, the senescent cells remain even after cessation of exposure.
Collapse
Affiliation(s)
- Kazuyuki Okamura
- Health and Environmental Risk Division, National Institute for Environmental Studies
| | - Miyuki Sato
- Health and Environmental Risk Division, National Institute for Environmental Studies
| | - Takehiro Suzuki
- Health and Environmental Risk Division, National Institute for Environmental Studies
| | - Keiko Nohara
- Health and Environmental Risk Division, National Institute for Environmental Studies
| |
Collapse
|
7
|
Qulu W, Mtshali A, Osman F, Ndlela N, Ntuli L, Mzobe G, Naicker N, Garrett N, Rompalo A, Mindel A, Ngcapu S, Liebenberg L. High-risk human papillomavirus prevalence among South African women diagnosed with other STIs and BV. PLoS One 2023; 18:e0294698. [PMID: 38032961 PMCID: PMC10688634 DOI: 10.1371/journal.pone.0294698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 11/07/2023] [Indexed: 12/02/2023] Open
Abstract
INTRODUCTION Human papillomavirus (HPV) infection is a leading cause of cervical cancer. Although this relies on infection and persistence of HPV in epithelial cells, often occurring in the context of other sexually transmitted infections (STIs) and bacterial vaginosis (BV), data on the relationships between these and their relative effects on epithelial barrier integrity in women remain sparse. This study describes the epidemiology of HPV combined with STI and/or BV prevalence and the relative impact on matrix metalloproteinases (MMPs) among South African women. METHODS Roche Linear Array was used for HPV genotyping in menstrual cup pellets of 243 HIV-negative women participating in the CAPRISA 083 cohort study. Vulvovaginal swabs were tested for Chlamydia trachomatis, Neisseria gonorrhoeae, and Trichomonas vaginalis using Xpert® CT/NG assay and lateral flow assay, and Gram staining was performed to diagnose BV using Nugent scoring criteria. Concentrations of 5 MMPs were measured in menstrual cup supernatants by multiplexed ELISA. Fisher's exact tests, Mann-Whitney U tests, and multivariable regression models determined associations between HPV infection, STI and/or BV, and MMP concentrations. RESULTS HPV was prevalent in 34% of women (83/243; median 23 years, interquartile range (IQR) 21-27 years). Low-risk (lr) (71%, 59/83) and high-risk (hr)-HPV infections (54.2%, 45/83) were common. Hr-HPV was frequently detected in STI and/or BV-positive women compared to women without STIs or BV (p = 0.029). In multivariable analysis, BV was associated with increased odds of hr-HPV detection (OR: 2.64, 95%CI: 1.02-6.87, p = 0.046). Furthermore, Gardasil®9 vaccine-type strains were more frequently detected in women diagnosed with STI and/or BV (55.2%, 32/58 vs 24%, 6/25; p = 0.009). Among STI and/or BV-positive women, HPV detection was significantly associated with increased MMP-10 concentrations (b = 0.55, 95% CI 0.79-1.01; p = 0.022). CONCLUSION Most women with hr-HPV had another STI and/or BV, emphasizing an urgent need for STI and BV screening and intensive scale-up of cervical cancer screening and HPV vaccination programmes. Furthermore, the study highlights the need for more extensive research to confirm and understand the relationship between HPV infection and barrier integrity.
Collapse
Affiliation(s)
- Wenkosi Qulu
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Andile Mtshali
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Farzana Osman
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| | - Nonsikelelo Ndlela
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Lungelo Ntuli
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Gugulethu Mzobe
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Nivashnee Naicker
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| | - Nigel Garrett
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
- School of Nursing and Public Health, Discipline of Public Health Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Anne Rompalo
- Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - Adrian Mindel
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| | - Sinaye Ngcapu
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Lenine Liebenberg
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
- Centre for Epidemic Response and Innovation, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
8
|
Sah DK, Arjunan A, Lee B, Jung YD. Reactive Oxygen Species and H. pylori Infection: A Comprehensive Review of Their Roles in Gastric Cancer Development. Antioxidants (Basel) 2023; 12:1712. [PMID: 37760015 PMCID: PMC10525271 DOI: 10.3390/antiox12091712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/14/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Gastric cancer (GC) is the fifth most common cancer worldwide and makes up a significant component of the global cancer burden. Helicobacter pylori (H. pylori) is the most influential risk factor for GC, with the International Agency for Research on Cancer classifying it as a Class I carcinogen for GC. H. pylori has been shown to persist in stomach acid for decades, causing damage to the stomach's mucosal lining, altering gastric hormone release patterns, and potentially altering gastric function. Epidemiological studies have shown that eliminating H. pylori reduces metachronous cancer. Evidence shows that various molecular alterations are present in gastric cancer and precancerous lesions associated with an H. pylori infection. However, although H. pylori can cause oxidative stress-induced gastric cancer, with antioxidants potentially being a treatment for GC, the exact mechanism underlying GC etiology is not fully understood. This review provides an overview of recent research exploring the pathophysiology of H. pylori-induced oxidative stress that can cause cancer and the antioxidant supplements that can reduce or even eliminate GC occurrence.
Collapse
Affiliation(s)
| | | | - Bora Lee
- Department of Biochemistry, Chonnam National University Medical School, Seoyang Ro 264, Jeonnam, Hwasun 58128, Republic of Korea; (D.K.S.); (A.A.)
| | - Young Do Jung
- Department of Biochemistry, Chonnam National University Medical School, Seoyang Ro 264, Jeonnam, Hwasun 58128, Republic of Korea; (D.K.S.); (A.A.)
| |
Collapse
|
9
|
Li Y, Wang C, Ma A, Rani AQ, Luo M, Li J, Liu X, Ma Q. Identification of HPV oncogene and host cell differentiation associated cellular heterogeneity in cervical cancer via single-cell transcriptomic analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.10.552878. [PMID: 37645794 PMCID: PMC10462038 DOI: 10.1101/2023.08.10.552878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Human Papillomaviruses (HPVs) are associated with around 5-10% of human cancer, notably nearly 99% of cervical cancer. The mechanisms HPV interacts with stratified epithelium (differentiated layers) during the viral life cycle, and oncogenesis remain unclear. In this study, we used single-cell transcriptome analysis to study viral gene and host cell differentiation-associated heterogeneity of HPV-positive cervical cancer tissue. We examined the HPV16 genes - E1, E6, and E7, and found they expressed differently across nine epithelial clusters. We found that three epithelial clusters had the highest proportion of HPV-positive cells (33.6%, 37.5%, and 32.4%, respectively), while two exhibited the lowest proportions (7.21% and 5.63%, respectively). Notably, the cluster with the most HPV-positive cells deviated significantly from normal epithelial layer markers, exhibiting functional heterogeneity and altered epithelial structuring, indicating that significant molecular heterogeneity existed in cancer tissues and that these cells exhibited unique/different gene signatures compared with normal epithelial cells. These HPV-positive cells, compared to HPV-negative, showed different gene expressions related to the extracellular matrix, cell adhesion, proliferation, and apoptosis. Further, the viral oncogenes E6 and E7 appeared to modify epithelial function via distinct pathways, thus contributing to cervical cancer progression. We investigated the HPV and host transcripts from a novel viewpoint focusing on layer heterogeneity. Our results indicated varied HPV expression across epithelial clusters and epithelial heterogeneity associated with viral oncogenes, contributing biological insights to this critical field of study.
Collapse
Affiliation(s)
- Yingjie Li
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Cankun Wang
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Anjun Ma
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Abdul Qawee Rani
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Mingjue Luo
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Jenny Li
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Xuefeng Liu
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
- The Departments of Pathology, Urology, and Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Qin Ma
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| |
Collapse
|
10
|
Li Y, Wang C, Ma A, Rani AQ, Luo M, Li J, Liu X, Ma Q. Identification of HPV oncogene and host cell differentiation associated cellular heterogeneity in cervical cancer via single-cell transcriptomic analysis. J Med Virol 2023; 95:e29060. [PMID: 37638381 DOI: 10.1002/jmv.29060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/11/2023] [Accepted: 08/16/2023] [Indexed: 08/29/2023]
Abstract
Human Papillomaviruses (HPVs) are associated with around 5%-10% of human cancer, notably nearly 99% of cervical cancer. The mechanisms HPV interacts with stratified epithelium (differentiated layers) during the viral life cycle, and oncogenesis remain unclear. In this study, we used single-cell transcriptome analysis to study viral gene and host cell differentiation-associated heterogeneity of HPV-positive cervical cancer tissue. We examined the HPV16 genes-E1, E6, and E7, and found they expressed differently across nine epithelial clusters. We found that three epithelial clusters had the highest proportion of HPV-positive cells (33.6%, 37.5%, and 32.4%, respectively), while two exhibited the lowest proportions (7.21% and 5.63%, respectively). Notably, the cluster with the most HPV-positive cells deviated significantly from normal epithelial layer markers, exhibiting functional heterogeneity and altered epithelial structuring, indicating that significant molecular heterogeneity existed in cancer tissues and that these cells exhibited unique/different gene signatures compared with normal epithelial cells. These HPV-positive cells, compared to HPV-negative, showed different gene expressions related to the extracellular matrix, cell adhesion, proliferation, and apoptosis. Further, the viral oncogenes E6 and E7 appeared to modify epithelial function via distinct pathways, thus contributing to cervical cancer progression. We investigated the HPV and host transcripts from a novel viewpoint focusing on layer heterogeneity. Our results indicated varied HPV expression across epithelial clusters and epithelial heterogeneity associated with viral oncogenes, contributing biological insights to this critical field of study.
Collapse
Affiliation(s)
- Yingjie Li
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Cankun Wang
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Anjun Ma
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, Ohio, USA
- The James Comprehensive Cancer Center, Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, Ohio, USA
| | - Abdul Qawee Rani
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Mingjue Luo
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Jenny Li
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Xuefeng Liu
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
- The Departments of Pathology, Urology, and Radiation Oncology, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Qin Ma
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, Ohio, USA
- The James Comprehensive Cancer Center, Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
11
|
Zayed M, Sastriques-Dunlop S, Elizondo-Benedetto S, Arif B, Meade R, Zaghloul M, Luehmann H, Heo G, English S, Liu Y. Ketosis Prevents Abdominal Aortic Aneurysm Rupture Through C-C Chemokine Receptor Type 2 Downregulation and Enhanced MMP Balance. RESEARCH SQUARE 2023:rs.3.rs-3054767. [PMID: 37461581 PMCID: PMC10350122 DOI: 10.21203/rs.3.rs-3054767/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
Abdominal aortic aneurysms (AAAs) are prevelant with aging, and AAA rupture is associated with high mortality. There is currently no effective medical therapy for AAA rupture. Previous work demonstrated that the monocyte chemoattractant protein (MCP-1) / C-C chemokine receptor type 2 (CCR2) axis critically regulates AAA inflammation, matrix-metalloproteinase (MMP) production, and extracellular matrix (ECM) stability. Here we similarly observed that Ccr2-/- mice have significantly reduced AAA expansion and rupture. We therefore hypothesized that a dietary modulation of the CCR2 axis may therapeutically impact AAA risk of rupture. Since ketone bodies (KBs) can trigger repair mechanisms in response to inflammation, we specifically evaluated whether systemic ketosis in vivo can reduce CCR2 and AAA progression. Male Sprague-Dawley rats underwent surgical AAA formation using porcine pancreatic elastase (PPE), and received daily β-aminopropionitrile (BAPN) to promote AAA rupture. Animals with AAAs received either a standard diet (SD), ketogenic diet (KD), or exogenous KBs (EKB). Animals recieving KD and EKB reached a state of ketosis, and had significant reduction in AAA expansion and incidence of rupture. Ketosis also led to significantly reduced aortic CCR2 content, improved MMP balance, and reduced ECM degradation. In summary, this study demonstrates that ketosis plays a crucial role in AAA pathobiology, and provides the impetus for future clinical studies investigating the potential benefit of ketosis for prevention of AAA expansion and rupture.
Collapse
Affiliation(s)
- Mohamed Zayed
- Washington University in St. Louis School of Medicine
| | | | | | - Batool Arif
- Washington University in St. Louis School of Medicine
| | - Rodrigo Meade
- Washington University in St. Louis School of Medicine
| | | | | | - Gyu Heo
- ashington University in St. Louis School of Medicine
| | - Sean English
- ashington University in St. Louis School of Medicine
| | | |
Collapse
|
12
|
Walsh C, Rajora MA, Ding L, Nakamura S, Endisha H, Rockel J, Chen J, Kapoor M, Zheng G. Protease-Activatable Porphyrin Molecular Beacon for Osteoarthritis Management. CHEMICAL & BIOMEDICAL IMAGING 2023; 1:66-80. [PMID: 37122828 PMCID: PMC10131263 DOI: 10.1021/cbmi.3c00005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 02/06/2023] [Accepted: 02/12/2023] [Indexed: 05/02/2023]
Abstract
Despite the substantial burden posed by osteoarthritis (OA) globally, difficult challenges remain in achieving early OA diagnosis and adopting effective disease-modifying treatments. In this study, we use a biomolecular approach to address these limitations by creating an inherently theranostic molecular beacon whose imaging and therapeutic capabilities are activated by early pathological changes in OA. This platform comprised (1) a peptide linker substrate for metalloproteinase-13 (MMP-13), a pathological protease upregulated in OA, which was conjugated to (2) a porphyrin moiety with inherent multimodal imaging, photodynamic therapy, and drug delivery capabilities, and (3) a quencher that silences the porphyrin's endogenous fluorescence and photoreactivity when the beacon is intact. In diseased OA tissue with upregulated MMP-13 expression, this porphyrin molecular beacon (PPMMP13B) was expected to undergo sequence-specific cleavage, yielding porphyrin fragments with restored fluorescence and photoreactivity that could, respectively, be used as a readout of MMP-13 activity within the joint for early OA imaging and disease-targeted photodynamic therapy. This study focused on the synthesis and characterization of PPMMP13B, followed by a proof-of-concept evaluation of its OA imaging and drug delivery potential. In solution, PPMMP13B demonstrated 90% photoactivity quenching in its intact form and robust MMP-13 activation, yielding a 13-fold increase in fluorescence post-cleavage. In vitro, PPMMP13B was readily uptaken and activated in an MMP-13 cell expression-dependent manner in primary OA synoviocytes without exuding significant cytotoxicity. This translated into effective intra-articular cartilage (to a 50 μm depth) and synovial uptake and activation of PPMMP13B in a destabilization of the medial meniscus OA mouse model, yielding strong fluorescence contrast (7-fold higher signal than background) at the diseased joint site. These results provide the foundation for further exploration of porphyrin molecular beacons for image-guided OA disease stratification, effective articular delivery of disease-modify agents, and OA photodynamic therapy.
Collapse
Affiliation(s)
- Connor Walsh
- Princess
Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
- Institute
of Biomedical Engineering, University of
Toronto, Toronto, ON M5S 3G9, Canada
| | - Maneesha A. Rajora
- Princess
Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
- Institute
of Biomedical Engineering, University of
Toronto, Toronto, ON M5S 3G9, Canada
| | - Lili Ding
- Princess
Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Sayaka Nakamura
- Schroeder
Arthritis Institute, University Health Network, Toronto, ON M5T 0S8, Canada
- Krembil
Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
| | - Helal Endisha
- Schroeder
Arthritis Institute, University Health Network, Toronto, ON M5T 0S8, Canada
- Krembil
Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
| | - Jason Rockel
- Schroeder
Arthritis Institute, University Health Network, Toronto, ON M5T 0S8, Canada
- Krembil
Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
| | - Juan Chen
- Princess
Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Mohit Kapoor
- Schroeder
Arthritis Institute, University Health Network, Toronto, ON M5T 0S8, Canada
- Krembil
Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
| | - Gang Zheng
- Princess
Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
- Institute
of Biomedical Engineering, University of
Toronto, Toronto, ON M5S 3G9, Canada
- Department
of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| |
Collapse
|
13
|
Hritcu OM, Bocaneti Daraban F, Bacusca FD, Pasca AS. Unusual Canine Cutaneous Melanoma Presenting Parietal Bone Metastasis: A Case Report. Vet Sci 2023; 10:vetsci10040282. [PMID: 37104437 PMCID: PMC10141863 DOI: 10.3390/vetsci10040282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 04/28/2023] Open
Abstract
Melanocytic tumour anatomic location is considered an important prognostic indicator. The cutaneous forms are generally considered benign and may show various biological behaviours. This work reports a rare case of canine cutaneous melanoma showing parietal bone metastasis. Bone invasion in melanocytic tumours is often described in oral or visceral melanomas, but not in cutaneous forms. The patient (dog, male, mixed breed, 12 years) was initially presented for the surgical removal of a cutaneous tumour located on the skin of the carpal region of the right forelimb. Four months after, the patient returned with enlarged lymph nodes and acute respiratory failure. The patient was euthanized due to a decline in physical condition. The necropsy showed metastases in the affected forelimb, regional lymph node, splanchnic organs, parietal bone and meninges. Histopathological examination of tumour tissue samples revealed a mixture of pigmented and non-pigmented spindle and epithelioid melanocytes, while according to immunohistochemistry, the tumours showed a strong immunopositivity for VEGF and MMP-10, and a moderate positivity for MMP-2 expression. This case shows that cutaneous melanocytic tumours may show an aggressive malignant form with positive immunohistochemical reactions for multiple invasiveness factors.
Collapse
Affiliation(s)
- Ozana-Maria Hritcu
- Faculty of Veterinary Medicine, Iasi University of Life Sciences Ion Ionescu de la Brad, Mihail Sadoveanu Alley, No.8, 700489 Iasi, Romania
| | - Florentina Bocaneti Daraban
- Faculty of Veterinary Medicine, Iasi University of Life Sciences Ion Ionescu de la Brad, Mihail Sadoveanu Alley, No.8, 700489 Iasi, Romania
| | - Fabian Dominic Bacusca
- Faculty of Veterinary Medicine, Iasi University of Life Sciences Ion Ionescu de la Brad, Mihail Sadoveanu Alley, No.8, 700489 Iasi, Romania
| | - Aurelian-Sorin Pasca
- Faculty of Veterinary Medicine, Iasi University of Life Sciences Ion Ionescu de la Brad, Mihail Sadoveanu Alley, No.8, 700489 Iasi, Romania
| |
Collapse
|
14
|
Transcriptional landscape of oncogene-induced senescence: a machine learning-based meta-analytic approach. Ageing Res Rev 2023; 85:101849. [PMID: 36621646 DOI: 10.1016/j.arr.2023.101849] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/28/2022] [Accepted: 01/04/2023] [Indexed: 01/07/2023]
Abstract
Oncogene-induced senescence (OIS) is highly heterogeneous, varying by oncogenic signals and cellular context. While its dual role, in the initial inhibition potentially later leading to promotion of tumors through the senescence-associated secretory phenotype, is still a matter of debate, it is undeniable that OIS is critical to understanding tumorigenesis. A major obstacle to OIS research is the absence of a universally accepted marker. Here, we present a robust OIS-specific transcriptomic secretory phenotype, termed oncogene-induced senescence secretory phenotype (OIS-SP), which can identify OIS across multiple biological contexts from in vitro datasets to in vivo human samples. We apply a meta-analytic machine learning pipeline to harmonize a deliberately varied selection of Ras-Raf-MEK-induced senescence datasets of differing origins, oncogenic signals and cell types. Finally we make use of bypass data to identify key genes and eliminate genes associated with quiescence, so identifying 40 OIS-SP genes. Within this set, we determined a robust core of five OIS-SP genes (FBLN1, CXCL12, EREG, CST1 and MMP10). Importantly, these 5 OIS-SP genes showed clear, consistent regulation patterns across various human Ras-Raf-MEK-mutated tumor tissues, which suggests that OIS-SP may be a novel cancer driver phenotype with an unexpectedly critical role in tumorigenesis.
Collapse
|
15
|
Dharavath B, Butle A, Pal A, Desai S, Upadhyay P, Rane A, Khandelwal R, Manavalan S, Thorat R, Sonawane K, Vaish R, Gera P, Bal M, D'Cruz AK, Nair S, Dutt A. Role of miR-944/MMP10/AXL- axis in lymph node metastasis in tongue cancer. Commun Biol 2023; 6:57. [PMID: 36650344 PMCID: PMC9845355 DOI: 10.1038/s42003-023-04437-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 01/06/2023] [Indexed: 01/18/2023] Open
Abstract
Occult lymph-node metastasis is a crucial predictor of tongue cancer mortality, with an unmet need to understand the underlying mechanism. Our immunohistochemical and real-time PCR analysis of 208 tongue tumors show overexpression of Matrix Metalloproteinase, MMP10, in 86% of node-positive tongue tumors (n = 79; p < 0.00001). Additionally, global profiling for non-coding RNAs associated with node-positive tumors reveals that of the 11 significantly de-regulated miRNAs, miR-944 negatively regulates MMP10 by targeting its 3'-UTR. We demonstrate that proliferation, migration, and invasion of tongue cancer cells are suppressed by MMP10 knockdown or miR-944 overexpression. Further, we show that depletion of MMP10 prevents nodal metastases using an orthotopic tongue cancer mice model. In contrast, overexpression of MMP10 leads to opposite effects upregulating epithelial-mesenchymal-transition, mediated by a tyrosine kinase gene, AXL, to promote nodal and distant metastasis in vivo. Strikingly, AXL expression is essential and sufficient to mediate the functional consequence of MMP10 overexpression. Consistent with our findings, TCGA-HNSC data suggests overexpression of MMP10 or AXL positively correlates with poor survival of the patients. In conclusion, our results establish that the miR-944/MMP10/AXL- axis underlies lymph node metastases with potential therapeutic intervention and prediction of nodal metastases in tongue cancer patients.
Collapse
Affiliation(s)
- Bhasker Dharavath
- Integrated Cancer Genomics Laboratory, Advanced Centre for Treatment, Research, and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra, 410210, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, 400094, India
| | - Ashwin Butle
- Integrated Cancer Genomics Laboratory, Advanced Centre for Treatment, Research, and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra, 410210, India
| | - Ankita Pal
- Integrated Cancer Genomics Laboratory, Advanced Centre for Treatment, Research, and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra, 410210, India
| | - Sanket Desai
- Integrated Cancer Genomics Laboratory, Advanced Centre for Treatment, Research, and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra, 410210, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, 400094, India
| | - Pawan Upadhyay
- Integrated Cancer Genomics Laboratory, Advanced Centre for Treatment, Research, and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra, 410210, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, 400094, India
| | - Aishwarya Rane
- Integrated Cancer Genomics Laboratory, Advanced Centre for Treatment, Research, and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra, 410210, India
| | - Risha Khandelwal
- Integrated Cancer Genomics Laboratory, Advanced Centre for Treatment, Research, and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra, 410210, India
| | - Sujith Manavalan
- Integrated Cancer Genomics Laboratory, Advanced Centre for Treatment, Research, and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra, 410210, India
| | - Rahul Thorat
- Laboratory Animal Facility, Advanced Centre for Treatment, Research and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra, 410210, India
| | - Kavita Sonawane
- Division of Head and Neck Oncology, Department of Surgical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Parel, Mumbai, 400012, India
| | - Richa Vaish
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, 400094, India
- Division of Head and Neck Oncology, Department of Surgical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Parel, Mumbai, 400012, India
| | - Poonam Gera
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, 400094, India
- Tissue Biorepository, Advanced Centre for Treatment Research and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra, 410210, India
| | - Munita Bal
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, 400094, India
- Department of Pathology, Tata Memorial Hospital, Tata Memorial Centre, Parel, Mumbai, 400012, India
| | - Anil K D'Cruz
- Division of Head and Neck Oncology, Department of Surgical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Parel, Mumbai, 400012, India
- Apollo Cancer Center, Apollo Hospitals, CBD Belapur, Navi Mumbai, 400614, India
| | - Sudhir Nair
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, 400094, India.
- Division of Head and Neck Oncology, Department of Surgical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Parel, Mumbai, 400012, India.
| | - Amit Dutt
- Integrated Cancer Genomics Laboratory, Advanced Centre for Treatment, Research, and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra, 410210, India.
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, 400094, India.
| |
Collapse
|
16
|
Monea M, Pop AM. The Use of Salivary Levels of Matrix Metalloproteinases as an Adjuvant Method in the Early Diagnosis of Oral Squamous Cell Carcinoma: A Narrative Literature Review. Curr Issues Mol Biol 2022; 44:6306-6322. [PMID: 36547091 PMCID: PMC9776994 DOI: 10.3390/cimb44120430] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/04/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is an aggressive malignancy with increased mortality, in which the early diagnosis is the most important step in increasing patients' survival rate. Extensive research has evaluated the role of saliva as a source of diagnostic biomarkers, among which matrix metalloproteinases (MMPs) have shown a valuable potential for detecting even early stages of OSCC. The aim of this review was to present recent clinical data regarding the significance of salivary MMPs in the detection of early malignant transformation of the oral mucosa. A narrative review was conducted on articles published in PubMed, Cochrane Library, Web of Science, EBSCO and SciELO databases, using specific terms. Our search revealed that MMP-1, MMP-2, MMP-3, MMP-8, MMP-9, MMP-10, MMP-12 and MMP-13 had significantly higher levels in saliva from patients with OSCC compared to controls. However, the strength of evidence is limited, as most information regarding their use as adjuvant diagnostic tools for OSCC comes from studies with a low number of participants, variable methodologies for saliva sampling and diagnostic assays, and insufficient adjustment for all covariates. MMP-1, MMP-3 and MMP-9 were considered the most promising candidates for salivary diagnosis of OSCC, but larger studies are needed in order to validate their clinical application.
Collapse
Affiliation(s)
- Monica Monea
- Department of Odontology and Oral Pathology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540139 Târgu Mureș, Romania
- Correspondence:
| | - Anca Maria Pop
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540139 Târgu Mureș, Romania
| |
Collapse
|
17
|
García-Vargas AM, Roque-Reyes YM, Arroyo-Villegas DM, Santiago-Negron D, Sánchez-Vázquez MM, Rivera-Torres A, Reyes-Meléndez AC, Cardona-Berdecía V, García-Maldonado M, Víquez OM, Martínez-Ferrer M. HLA-BAT1 alters migration, invasion and pro-inflammatory cytokines in prostate cancer. Front Oncol 2022; 12:969396. [PMID: 36505884 PMCID: PMC9727259 DOI: 10.3389/fonc.2022.969396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 10/27/2022] [Indexed: 11/24/2022] Open
Abstract
Prostate cancer (PCa) accounts for more than 1 in 5 diagnoses and is the second cause of cancer-related deaths in men. Although PCa may be successfully treated, patients may undergo cancer recurrence and there is a need for new biomarkers to improve the prediction of prostate cancer recurrence and improve treatment. Our laboratory demonstrated that HLA-B-associated transcript 1 (BAT1) was differentially expressed in patients with high Gleason scores when compared to low Gleason scores. BAT1 is an anti-inflammatory gene but its role in PCa has not been identified. The objective of this study is to understand the role of BAT1 in prostate cancer. In vitro studies showed that BAT1 down-regulation increased cell migration and invasion. In contrast, BAT1 overexpression decreased cell migration and invasion. RT-PCR analysis showed differential expression of pro-inflammatory cytokines (TNF-α and IL-6) and cell adhesion and migration genes (MMP10, MMP13, and TIMPs) in BAT1 overexpressed cells when compared to BAT1 siRNA cells. Our in vivo studies demonstrated up-regulation of TNF-α, IL-6, and MMP10 in tumors developed from transfected BAT1 shRNA cells when compared to tumors developed from BAT1 cDNA cells. These findings indicate that BAT1 down-regulation modulates TNF-α and IL-6 expression which may lead to the secretion of MMP-10 and inhibition of TIMP2.
Collapse
Affiliation(s)
- Aileen M. García-Vargas
- Department of Pharmacology and Toxicology, School of Medicine, University of Puerto Rico, Medical Sciences Campus, San Juan, PR, United States
- Division of Cancer Biology, University of Puerto Rico Comprehensive Cancer Center, San Juan, PR, United States
| | - Yarelis M. Roque-Reyes
- Division of Cancer Biology, University of Puerto Rico Comprehensive Cancer Center, San Juan, PR, United States
- Department of Biology, University of Puerto Rico, Rio Piedras Campus, San Juan, PR, United States
| | - Desiree M. Arroyo-Villegas
- Division of Cancer Biology, University of Puerto Rico Comprehensive Cancer Center, San Juan, PR, United States
- Department of Chemistry, University of Puerto Rico, Rio Piedras Campus, San Juan, PR, United States
| | - Daniel Santiago-Negron
- Division of Cancer Biology, University of Puerto Rico Comprehensive Cancer Center, San Juan, PR, United States
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University, Nashville, TN, United States
| | - María M. Sánchez-Vázquez
- Division of Cancer Biology, University of Puerto Rico Comprehensive Cancer Center, San Juan, PR, United States
| | - Alejandro Rivera-Torres
- Division of Cancer Biology, University of Puerto Rico Comprehensive Cancer Center, San Juan, PR, United States
- Department of Chemistry, University of Puerto Rico, Rio Piedras Campus, San Juan, PR, United States
| | - Andrea C. Reyes-Meléndez
- Division of Cancer Biology, University of Puerto Rico Comprehensive Cancer Center, San Juan, PR, United States
- Department of Biology, University of Puerto Rico, Rio Piedras Campus, San Juan, PR, United States
| | - Valerie Cardona-Berdecía
- Division of Cancer Biology, University of Puerto Rico Comprehensive Cancer Center, San Juan, PR, United States
- Department of Chemistry, University of Puerto Rico, Rio Piedras Campus, San Juan, PR, United States
| | - Miosotis García-Maldonado
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Puerto Rico, Medical Sciences Campus, San Juan, PR, United States
| | - Olga M. Víquez
- Research Biobank, University of Puerto Rico Comprehensive Cancer Center, San Juan, PR, United States
| | - Magaly Martínez-Ferrer
- Division of Cancer Biology, University of Puerto Rico Comprehensive Cancer Center, San Juan, PR, United States
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
18
|
Luo L, Li F, Gong B, Xi P, Xie W. A novel prognostic model based on cellular senescence-related gene signature for bladder cancer. Front Oncol 2022; 12:937951. [PMID: 36505846 PMCID: PMC9727082 DOI: 10.3389/fonc.2022.937951] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 10/31/2022] [Indexed: 11/24/2022] Open
Abstract
Background Cellular senescence plays crucial role in the progression of tumors. However, the expression patterns and clinical significance of cellular senescence-related genes in bladder cancer (BCa) are still not clearly clarified. This study aimed to establish a prognosis model based on senescence-related genes in BCa. Methods The transcriptional profile data and clinical information of BCa were downloaded from TCGA and GEO databases. The least absolute shrinkage and selection operator (LASSO), univariate and multivariate Cox regression analyses were performed to develop a prognostic model in the TCGA cohort. The GSE13507 cohort were used for validation. Gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and single-sample gene set enrichment analysis (ssGSEA) were performed to investigate underlying mechanisms. Results A six-gene signature (CBX7, EPHA3, STK40, TGFB1I1, SREBF1, MYC) was constructed in the TCGA databases. Patients were classified into high risk and low risk group in terms of the median risk score. Survival analysis revealed that patients in the higher risk group presented significantly worse prognosis. Receiver operating characteristic (ROC) curve analysis verified the moderate predictive power of the risk model based on the six senescence-related genes signature. Further analysis indicated that the clinicopathological features analysis were significantly different between the two risk groups. As expected, the signature presented prognostic significance in the GSE13507 cohort. Functional analysis indicated that immune-related pathways activity, immune cell infiltration and immune-related function were different between two risk groups. In addition, risk score were positively correlated with multiple immunotherapy biomarkers. Conclusion Our study revealed that a novel model based on senescence-related genes could serve as a reliable predictor of survival for patients with BCa.
Collapse
Affiliation(s)
- Lianmin Luo
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Fenghua Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Binbin Gong
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Ping Xi
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Wenjie Xie
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China,*Correspondence: Wenjie Xie,
| |
Collapse
|
19
|
PAI-1 is a potential transcriptional silencer that supports bladder cancer cell activity. Sci Rep 2022; 12:12186. [PMID: 35842542 PMCID: PMC9288475 DOI: 10.1038/s41598-022-16518-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 07/11/2022] [Indexed: 12/12/2022] Open
Abstract
The extracellular activity of Plasminogen activator inhibitor-1 (PAI-1) is well described, acting as an inhibitor of tissue plasminogen activator and urokinase-type plasminogen activator, impacting fibrinolysis. Recent studies have revealed a pro-tumorigenic role of PAI-1 in human cancers, via the regulation of angiogenesis and tumor cell survival. In this study, immunohistochemical staining of 939 human bladder cancer specimens showed that PAI-1 expression levels correlated with tumor grade, tumor stage and overall survival. The typical subcellular localization of PAI-1 is cytoplasmic, but in approximately a quarter of the cases, PAI-1 was observed to be localized to both the tumor cell cytoplasm and the nucleus. To investigate the potential function of nuclear PAI-1 in tumor biology we applied chromatin immunoprecipitation (ChIP)-sequencing, gene expression profiling, and rapid immunoprecipitation mass spectrometry to a pair of bladder cancer cell lines. ChIP-sequencing revealed that PAI-1 can bind DNA at distal intergenic regions, suggesting a role as a transcriptional coregulator. The downregulation of PAI-1 in bladder cancer cell lines caused the upregulation of numerous genes, and the integration of ChIP-sequence and RNA-sequence data identified 57 candidate genes subject to PAI-1 regulation. Taken together, the data suggest that nuclear PAI-1 can influence gene expression programs and support malignancy.
Collapse
|
20
|
González-Zamora J, Hernandez M, Recalde S, Bezunartea J, Montoliu A, Bilbao-Malavé V, Orbe J, Rodríguez JA, Llorente-González S, Fernández-Robredo P, García-Layana A. Matrix Metalloproteinase 10 Contributes to Choroidal Neovascularisation. Biomedicines 2022; 10:biomedicines10071557. [PMID: 35884862 PMCID: PMC9313238 DOI: 10.3390/biomedicines10071557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022] Open
Abstract
Age-related macular degeneration (AMD) is currently the main cause of severe visual loss among older adults in developed countries. The pathophysiology has not been clarified, but oxidative stress is believed to play a major role. Matrix metalloproteinases (MMP) may play a prominent role in several steps of the pathophysiology of AMD, especially in its neovascular form; therefore, there is of great interest in understanding their role in choroidal neovascularisation. This study aimed to elucidate the role of MMP10 in the development of choroidal neovascularisation (CNV). We have demonstrated that MMP10 was expressed by retinal pigment epithelium cells and endothelial cells of the neovascular membrane, in cell culture, mouse and human retina. MMP10 expression and activity increased under oxidative stress conditions in ARPE-19 cells. MMP10-/- mice developed smaller laser-induced areas of CNV. Furthermore, to exclude a systemic MMP10 imbalance in these patients, plasma MMP10 concentrations were assessed in an age- and sex-matched sample of 52 control patients and 52 patients with neovascular AMD and no significant differences were found between the groups, demonstrating that MMP10 induction is a local phenomenon. Our findings suggest that MMP10 participates in the development of choroidal neovascularisation and promotes MMP10 as a possible new therapeutic target.
Collapse
Affiliation(s)
- Jorge González-Zamora
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Clinica Universidad de Navarra, 31008 Pamplona, Spain; (J.G.-Z.); (S.R.); (J.B.); (A.M.); (V.B.-M.); (S.L.-G.); (A.G.-L.)
| | - María Hernandez
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Clinica Universidad de Navarra, 31008 Pamplona, Spain; (J.G.-Z.); (S.R.); (J.B.); (A.M.); (V.B.-M.); (S.L.-G.); (A.G.-L.)
- Navarra Institute for Health Research, IdiSNA, 31008 Pamplona, Spain; (J.O.); (J.A.R.)
- Correspondence: (M.H.); (P.F.-R.)
| | - Sergio Recalde
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Clinica Universidad de Navarra, 31008 Pamplona, Spain; (J.G.-Z.); (S.R.); (J.B.); (A.M.); (V.B.-M.); (S.L.-G.); (A.G.-L.)
- Navarra Institute for Health Research, IdiSNA, 31008 Pamplona, Spain; (J.O.); (J.A.R.)
| | - Jaione Bezunartea
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Clinica Universidad de Navarra, 31008 Pamplona, Spain; (J.G.-Z.); (S.R.); (J.B.); (A.M.); (V.B.-M.); (S.L.-G.); (A.G.-L.)
- Navarra Institute for Health Research, IdiSNA, 31008 Pamplona, Spain; (J.O.); (J.A.R.)
| | - Ana Montoliu
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Clinica Universidad de Navarra, 31008 Pamplona, Spain; (J.G.-Z.); (S.R.); (J.B.); (A.M.); (V.B.-M.); (S.L.-G.); (A.G.-L.)
| | - Valentina Bilbao-Malavé
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Clinica Universidad de Navarra, 31008 Pamplona, Spain; (J.G.-Z.); (S.R.); (J.B.); (A.M.); (V.B.-M.); (S.L.-G.); (A.G.-L.)
| | - Josune Orbe
- Navarra Institute for Health Research, IdiSNA, 31008 Pamplona, Spain; (J.O.); (J.A.R.)
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, CIMA-Universidad de Navarra, CIBERCV, 31008 Pamplona, Spain
| | - José A. Rodríguez
- Navarra Institute for Health Research, IdiSNA, 31008 Pamplona, Spain; (J.O.); (J.A.R.)
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, CIMA-Universidad de Navarra, CIBERCV, 31008 Pamplona, Spain
| | - Sara Llorente-González
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Clinica Universidad de Navarra, 31008 Pamplona, Spain; (J.G.-Z.); (S.R.); (J.B.); (A.M.); (V.B.-M.); (S.L.-G.); (A.G.-L.)
- Navarra Institute for Health Research, IdiSNA, 31008 Pamplona, Spain; (J.O.); (J.A.R.)
| | - Patricia Fernández-Robredo
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Clinica Universidad de Navarra, 31008 Pamplona, Spain; (J.G.-Z.); (S.R.); (J.B.); (A.M.); (V.B.-M.); (S.L.-G.); (A.G.-L.)
- Navarra Institute for Health Research, IdiSNA, 31008 Pamplona, Spain; (J.O.); (J.A.R.)
- Correspondence: (M.H.); (P.F.-R.)
| | - Alfredo García-Layana
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Clinica Universidad de Navarra, 31008 Pamplona, Spain; (J.G.-Z.); (S.R.); (J.B.); (A.M.); (V.B.-M.); (S.L.-G.); (A.G.-L.)
- Navarra Institute for Health Research, IdiSNA, 31008 Pamplona, Spain; (J.O.); (J.A.R.)
| |
Collapse
|
21
|
Altered differentiation of endometrial mesenchymal stromal fibroblasts is associated with endometriosis susceptibility. Commun Biol 2022; 5:600. [PMID: 35725766 PMCID: PMC9209414 DOI: 10.1038/s42003-022-03541-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 05/31/2022] [Indexed: 01/10/2023] Open
Abstract
Cellular development is tightly regulated as mature cells with aberrant functions may initiate pathogenic processes. The endometrium is a highly regenerative tissue, shedding and regenerating each month. Endometrial stromal fibroblasts are regenerated each cycle from mesenchymal stem cells and play a pivotal role in endometriosis, a disease characterised by endometrial cells that grow outside the uterus. Why the cells of some women are more capable of developing into endometriosis lesions is not clear. Using isolated, purified and cultured endometrial cells of mesenchymal origin from 19 women with (n = 10) and without (n = 9) endometriosis we analysed the transcriptome of 33,758 individual cells and compared these to clinical characteristics and in vitro growth profiles. We show purified mesenchymal cell cultures include a mix of mesenchymal stem cells and two endometrial stromal fibroblast subtypes with distinct transcriptomic signatures indicative of varied progression through the differentiation processes. The fibroblast subgroup characterised by incomplete differentiation was predominantly (81%) derived from women with endometriosis and exhibited an altered in vitro growth profile. These results uncover an inherent difference in endometrial cells of women with endometriosis and highlight the relevance of cellular differentiation and its potential to contribute to disease susceptibility. Comparing single cell transcriptome data to clinical characteristics and in vitro growth profiles uncovers a potential role for divergent mesenchymal-derived stromal fibroblast maturation in endometriosis susceptibility.
Collapse
|
22
|
Xu Y, Xiong Q, Yang Y, Weng N, Li J, Liu J, Yang X, Zeng Z, Zhang Z, Zhu Q. Serum Nardilysin as a Prognostic Biomarker in Pancreatic Ductal Adenocarcinoma. J Clin Med 2022; 11:jcm11113101. [PMID: 35683488 PMCID: PMC9181681 DOI: 10.3390/jcm11113101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/20/2022] [Accepted: 05/25/2022] [Indexed: 11/29/2022] Open
Abstract
Background: Nardilysin, (N-arginine dibasic convertase, NRDC) has been reported to play an important role in cancer progression, and is associated with tumor proliferation signals and inflammatory signals, such as tumor necrosis factor-a (TNF-a) and heparin-binding epidermal growth factor-like growth factor (HB-EGF), through the activation of disintegrin and metalloproteinase (ADAM) proteases. NRDC has recently been revealed to be involved in the tumorigenesis of various types of cancer, including intrahepatic cholangiocarcinoma, malignant cerebral infarction, esophageal squamous cell carcinoma, and gastric cancer. However, the expression profiles and biological relevance of NRDC in pancreatic ductal adenocarcinoma have rarely been reported. Methods: We analyzed the NRDC expression profile in pancreatic ductal adenocarcinoma by enzyme-linked immunosorbent assay (ELISA) and identified NRDC as a circulating biomarker in the serum of 112 pancreatic ductal adenocarcinoma patients. The diagnostic value of NRDC was analyzed by the area under the curve (AUC) and the receiver operating characteristic (ROC) test. Results: Our results demonstrated that the clinical prognosis significance of NRDC with the clinical characteristics in pancreatic ductal adenocarcinoma (PDAC). NRDC was notably decreased in PDAC patient serum compared with the control group (p < 0.001). Furthermore, the present study found that the NRDC expression level was correlated with T grade (p < 0.001), metastasis(p < 0.001), differentiation(p < 0.001), and TNM stage (p = 0.011). Further bioinformatics analysis revealed that NRDC correlated with proliferation and migration pathways; in particular, it mediated cell-matrix adhesion-dependent activation in pancreatic ductal adenocarcinoma. Conclusions: Serum NRDC may play a useful diagnostic biomarker to evaluate the aggressive clinical features in PAAD patients.
Collapse
|
23
|
Filipiak-Duliban A, Brodaczewska K, Majewska A, Kieda C. Spheroid culture models adequately imitate distinctive features of the renal cancer or melanoma microenvironment. In Vitro Cell Dev Biol Anim 2022; 58:349-364. [PMID: 35536385 DOI: 10.1007/s11626-022-00685-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/24/2022] [Indexed: 11/05/2022]
Abstract
Tumor development studies should adapt to cancer cells' specific mechanisms in connection with their microenvironment. Standard two-dimensional cultures and gas composition are not relevant to the real cancer environment. Existing three-dimensional models are often requiring sophisticated conditions. Here, we propose and characterize, in two cancer models, melanoma (B16F10) and kidney cancer (RenCa), a three-dimensional culture method, reporting the presence of hypoxia-related genes/proteins and aggressiveness mechanisms (epithelial mesenchymal transition and cancer stem cells). We validate the designed three-dimensional method by comparing it with in vivo growing tumors. The developed method brings simplicity and data reproducibility. Melanoma spheroid-growing cells reached a cell cycle arrest at the G0/G1 phase and showed induction of hypoxia. Spheroid-recovered RenCa cells were enriched in proliferating cells and displayed delayed hypoxia. Moreover, the responses to hypoxia observed in spheroids were validated by in vivo tumor studies for both lines. Three-dimensional shapes induced cancer stem cells in renal cancer, whereas epithelial to mesenchymal transition occurred in the melanoma model. Such distinction in the use of different aggressiveness-leading pathways was observed in in vivo melanoma vs kidney tumors. Thus, this 3D culture model approach is adequate to uncover crucial molecular pathways using distinct mechanisms to reach aggressiveness; i.e., B16F10 cells perform epithelial to mesenchymal transition while RenCa cells dedifferentiate into cancer stem cells. Such three-dimensional models help mimic the in vivo tumor features, i.e., hypoxia and aggressiveness mechanisms as validated here by next-generation sequencing analysis, and are proposed for further alternative methods to in vivo studies.
Collapse
Affiliation(s)
- Aleksandra Filipiak-Duliban
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland. .,Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland.
| | - Klaudia Brodaczewska
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland
| | - Aleksandra Majewska
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland.,Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Claudine Kieda
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland.,Center for Molecular Biophysics UPR 4301 CNRS, 45071, Orleans, France
| |
Collapse
|
24
|
Oncogenic Mutation BRAF V600E Changes Phenotypic Behavior of THLE-2 Liver Cells through Alteration of Gene Expression. Int J Mol Sci 2022; 23:ijms23031548. [PMID: 35163468 PMCID: PMC8836259 DOI: 10.3390/ijms23031548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/12/2022] [Accepted: 01/17/2022] [Indexed: 12/10/2022] Open
Abstract
The accumulation of mutations in cancer driver genes, such as tumor suppressors or proto-oncogenes, affects cellular homeostasis. Disturbances in the mechanism controlling proliferation cause significant augmentation of cell growth and division due to the loss of sensitivity to the regulatory signals. Nowadays, an increasing number of cases of liver cancer are observed worldwide. Data provided by the International Cancer Genome Consortium (ICGC) have indicated many alterations within gene sequences, whose roles in tumor development are not well understood. A comprehensive analysis of liver cancer (virus-associated hepatocellular carcinoma) samples has identified new and rare mutations in B-Raf proto-oncogene (BRAF) in Japanese HCC patients, as well as BRAF V600E mutations in French HCC patients. However, their function in liver cancer has never been investigated. Here, using functional analysis and next generation sequencing, we demonstrate the tumorigenic effect of BRAF V600E on hepatocytes (THLE-2 cell line). Moreover, we identified genes such as BMP6, CXCL11, IL1B, TBX21, RSAD2, MMP10, and SERPIND1, which are possibly regulated by the BRAF V600E-mediated, mitogen-activated protein kinases/extracellular signal-regulated kinases (MAPK/ERK) signaling pathway. Through several functional assays, we demonstrate that BRAF L537M, D594A, and E648G mutations alone are not pathogenic in liver cancer. The investigation of genome mutations and the determination of their impact on cellular processes and functions is crucial to unraveling the molecular mechanisms of liver cancer development.
Collapse
|
25
|
Kumar AA, Buckley BJ, Ranson M. The Urokinase Plasminogen Activation System in Pancreatic Cancer: Prospective Diagnostic and Therapeutic Targets. Biomolecules 2022; 12:152. [PMID: 35204653 PMCID: PMC8961517 DOI: 10.3390/biom12020152] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/13/2022] [Accepted: 01/16/2022] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer is a highly aggressive malignancy that features high recurrence rates and the poorest prognosis of all solid cancers. The urokinase plasminogen activation system (uPAS) is strongly implicated in the pathophysiology and clinical outcomes of patients with pancreatic ductal adenocarcinoma (PDAC), which accounts for more than 90% of all pancreatic cancers. Overexpression of the urokinase-type plasminogen activator (uPA) or its cell surface receptor uPAR is a key step in the acquisition of a metastatic phenotype via multiple mechanisms, including the increased activation of cell surface localised plasminogen which generates the serine protease plasmin. This triggers multiple downstream processes that promote tumour cell migration and invasion. Increasing clinical evidence shows that the overexpression of uPA, uPAR, or of both is strongly associated with worse clinicopathological features and poor prognosis in PDAC patients. This review provides an overview of the current understanding of the uPAS in the pathogenesis and progression of pancreatic cancer, with a focus on PDAC, and summarises the substantial body of evidence that supports the role of uPAS components, including plasminogen receptors, in this disease. The review further outlines the clinical utility of uPAS components as prospective diagnostic and prognostic biomarkers for PDAC, as well as a rationale for the development of novel uPAS-targeted therapeutics.
Collapse
Affiliation(s)
- Ashna A. Kumar
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia; (A.A.K.); (B.J.B.)
- School of Chemistry and Molecular Biosciences, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Benjamin J. Buckley
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia; (A.A.K.); (B.J.B.)
- School of Chemistry and Molecular Biosciences, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Marie Ranson
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia; (A.A.K.); (B.J.B.)
- School of Chemistry and Molecular Biosciences, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| |
Collapse
|
26
|
Shigeta S, Watanabe Y, Suzuki F, Nagase S, Shibuya Y, Ishibashi M, Nagai T, Shiga N, Toyoshima M, Tokunaga H, Shimada M, Yaegashi N. MicroRNA-152 Regulates Endometrial Serous Carcinoma Cell Motility by Suppressing Matrix Metalloproteinase 10 Expression. TOHOKU J EXP MED 2022; 256:249-258. [DOI: 10.1620/tjem.256.249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Shogo Shigeta
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine
| | - Yoh Watanabe
- Division of Obstetrics and Gynecology, Tohoku Medical and Pharmaceutical University
| | - Fumihiko Suzuki
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine
| | - Satoru Nagase
- Department of Obstetrics and Gynecology, Yamagata University Faculty of Medicine
| | - Yusuke Shibuya
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine
| | - Masumi Ishibashi
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine
| | - Tomoyuki Nagai
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine
| | - Naomi Shiga
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine
| | | | - Hideki Tokunaga
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine
| | - Muneaki Shimada
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine
| | - Nobuo Yaegashi
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine
| |
Collapse
|
27
|
Matrix Metalloproteinases Shape the Tumor Microenvironment in Cancer Progression. Int J Mol Sci 2021; 23:ijms23010146. [PMID: 35008569 PMCID: PMC8745566 DOI: 10.3390/ijms23010146] [Citation(s) in RCA: 197] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer progression with uncontrolled tumor growth, local invasion, and metastasis depends largely on the proteolytic activity of numerous matrix metalloproteinases (MMPs), which affect tissue integrity, immune cell recruitment, and tissue turnover by degrading extracellular matrix (ECM) components and by releasing matrikines, cell surface-bound cytokines, growth factors, or their receptors. Among the MMPs, MMP-14 is the driving force behind extracellular matrix and tissue destruction during cancer invasion and metastasis. MMP-14 also influences both intercellular as well as cell-matrix communication by regulating the activity of many plasma membrane-anchored and extracellular proteins. Cancer cells and other cells of the tumor stroma, embedded in a common extracellular matrix, interact with their matrix by means of various adhesive structures, of which particularly invadopodia are capable to remodel the matrix through spatially and temporally finely tuned proteolysis. As a deeper understanding of the underlying functional mechanisms is beneficial for the development of new prognostic and predictive markers and for targeted therapies, this review examined the current knowledge of the interplay of the various MMPs in the cancer context on the protein, subcellular, and cellular level with a focus on MMP14.
Collapse
|
28
|
Łaniewski P, Herbst-Kralovetz MM. Bacterial vaginosis and health-associated bacteria modulate the immunometabolic landscape in 3D model of human cervix. NPJ Biofilms Microbiomes 2021; 7:88. [PMID: 34903740 PMCID: PMC8669023 DOI: 10.1038/s41522-021-00259-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/11/2021] [Indexed: 12/20/2022] Open
Abstract
Bacterial vaginosis (BV) is an enigmatic polymicrobial condition characterized by a depletion of health-associated Lactobacillus and an overgrowth of anaerobes. Importantly, BV is linked to adverse gynecologic and obstetric outcomes: an increased risk of sexually transmitted infections, preterm birth, and cancer. We hypothesized that members of the cervicovaginal microbiota distinctly contribute to immunometabolic changes in the human cervix, leading to these sequelae. Our 3D epithelial cell model that recapitulates the human cervical epithelium was infected with clinical isolates of cervicovaginal bacteria, alone or as a polymicrobial community. We used Lactobacillus crispatus as a representative health-associated commensal and four common BV-associated species: Gardnerella vaginalis, Prevotella bivia, Atopobium vaginae, and Sneathia amnii. The immunometabolic profiles of these microenvironments were analyzed using multiplex immunoassays and untargeted global metabolomics. A. vaginae and S. amnii exhibited the highest proinflammatory potential through induction of cytokines, iNOS, and oxidative stress-associated compounds. G. vaginalis, P. bivia, and S. amnii distinctly altered physicochemical barrier-related proteins and metabolites (mucins, sialic acid, polyamines), whereas L. crispatus produced an antimicrobial compound, phenyllactic acid. Alterations to the immunometabolic landscape correlate with symptoms and hallmarks of BV and connected BV with adverse women’s health outcomes. Overall, this study demonstrated that 3D cervical epithelial cell colonized with cervicovaginal microbiota faithfully reproduce the immunometabolic microenvironment previously observed in clinical studies and can successfully be used as a robust tool to evaluate host responses to commensal and pathogenic bacteria in the female reproductive tract.
Collapse
Affiliation(s)
- Paweł Łaniewski
- Department of Basic Medical Sciences, College of Medicine - Phoenix, University of Arizona, Phoenix, AZ, 85004, USA
| | - Melissa M Herbst-Kralovetz
- Department of Basic Medical Sciences, College of Medicine - Phoenix, University of Arizona, Phoenix, AZ, 85004, USA. .,Department of Obstetrics and Gynecology, College of Medicine - Phoenix, University of Arizona, Phoenix, AZ, 85004, USA.
| |
Collapse
|
29
|
Di Martino JS, Akhter T, Bravo-Cordero JJ. Remodeling the ECM: Implications for Metastasis and Tumor Dormancy. Cancers (Basel) 2021; 13:4916. [PMID: 34638400 PMCID: PMC8507703 DOI: 10.3390/cancers13194916] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/17/2021] [Accepted: 09/19/2021] [Indexed: 12/24/2022] Open
Abstract
While most primary tumors can be effectively treated, therapeutics fail to efficiently eliminate metastases. Metastases arise from cancer cells that leave the primary tumor and seed distant sites. Recent studies have shown that cancer cells disseminate early during tumor progression and can remain dormant for years before they resume growth. In these metastatic organs, cancer cells reside in microenvironments where they interact with other cells, but also with the extracellular matrix (ECM). The ECM was long considered to be an inert, non-cellular component of tissues, providing their architecture. However, in recent years, a growing body of evidence has shown that the ECM is a key driver of cancer progression, and it can exert effects on tumor cells, regulating their metastatic fate. ECM remodeling and degradation is required for the early steps of the metastatic cascade: invasion, tumor intravasation, and extravasation. Similarly, ECM molecules have been shown to be important for metastatic outgrowth. However, the role of ECM molecules on tumor dormancy and their contribution to the dormancy-supportive niches is not well understood. In this perspective article, we will summarize the current knowledge of ECM and its role in tumor metastasis and dormancy. We will discuss how a better understanding of the individual components of the ECM niche and their roles mediating the dormant state of disseminated tumor cells (DTCs) will advance the development of new therapies to target dormant cells and prevent metastasis outgrowth.
Collapse
Affiliation(s)
| | | | - Jose Javier Bravo-Cordero
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (J.S.D.M.); (T.A.)
| |
Collapse
|
30
|
Kratochvilova A, Ramesova A, Vesela B, Svandova E, Lesot H, Gruber R, Matalova E. Impact of FasL Stimulation on Sclerostin Expression and Osteogenic Profile in IDG-SW3 Osteocytes. BIOLOGY 2021; 10:biology10080757. [PMID: 34439989 PMCID: PMC8389703 DOI: 10.3390/biology10080757] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/27/2021] [Accepted: 08/05/2021] [Indexed: 12/26/2022]
Abstract
Simple Summary FasL used to be considered as a classical ligand triggering cell death (apoptosis) via its receptor, Fas and thefollowing caspase cascade. As such, it is known to be involved in regulation within the bone. Recently, however, the knowledge has expanded about the non-apoptotic and caspase-independent engagement of the Fas/FasL pathway. The present investigation identified that stimulation of osteocytic IDG-SW3 cells by FasL leads to a dramatic decrease in expression of the major osteocytic marker, sclerostin. Additionally, other key components of the osteogenic pathways were impacted, notably in a caspase-independent manner. Such findings are of importance for basic biology as well as biomedical applications since osteocytes are the major population within adult bones and Fas signalling is one of therapeutical targets, e.g., in the anti-osteoporotic treatment. Abstract The Fas ligand (FasL) is known from programmed cell death, the immune system, and recently also from bone homeostasis. As such, Fas signalling is a potential target of anti-osteoporotic treatment based on the induction of osteoclastic cell death. Less attention has been paid to osteocytes, although they represent the majority of cells within the mature bone and are the key regulators. To determine the impact of FasL stimulation on osteocytes, differentiated IDG-SW3 cells were challenged by FasL, and their osteogenic expression profiles were evaluated by a pre-designed PCR array. Notably, the most downregulated gene was the one for sclerostin, which is the major marker of osteocytes and a negative regulator of bone formation. FasL stimulation also led to significant changes (over 10-fold) in the expression of other osteogenic markers: Gdf10, Gli1, Ihh, Mmp10, and Phex. To determine whether these alterations involved caspase-dependent or caspase-independent mechanisms, the IDG-SW3 cells were stimulated by FasL with and without a caspase inhibitor: Q-VD-OPh. The alterations were also detected in the samples treated by FasL along with Q-VD-OPh, pointing to the caspase-independent impact of FasL stimulation. These results contribute to an understanding of the recently emerging pleiotropic effects of Fas/FasL signalling and specify its functions in bone cells.
Collapse
Affiliation(s)
- Adela Kratochvilova
- Laboratory of Odontogenesis and Osteogenesis, Institute of Animal Physiology and Genetics, Academy of Sciences, 60200 Brno, Czech Republic; (A.K.); (A.R.); (B.V.); (E.S.); (H.L.)
| | - Alice Ramesova
- Laboratory of Odontogenesis and Osteogenesis, Institute of Animal Physiology and Genetics, Academy of Sciences, 60200 Brno, Czech Republic; (A.K.); (A.R.); (B.V.); (E.S.); (H.L.)
| | - Barbora Vesela
- Laboratory of Odontogenesis and Osteogenesis, Institute of Animal Physiology and Genetics, Academy of Sciences, 60200 Brno, Czech Republic; (A.K.); (A.R.); (B.V.); (E.S.); (H.L.)
| | - Eva Svandova
- Laboratory of Odontogenesis and Osteogenesis, Institute of Animal Physiology and Genetics, Academy of Sciences, 60200 Brno, Czech Republic; (A.K.); (A.R.); (B.V.); (E.S.); (H.L.)
| | - Herve Lesot
- Laboratory of Odontogenesis and Osteogenesis, Institute of Animal Physiology and Genetics, Academy of Sciences, 60200 Brno, Czech Republic; (A.K.); (A.R.); (B.V.); (E.S.); (H.L.)
| | - Reinhard Gruber
- Department of Oral Biology, University Clinic of Dentistry, Medical University Vienna, Sensengasse 2a, 1090 Vienna, Austria;
| | - Eva Matalova
- Laboratory of Odontogenesis and Osteogenesis, Institute of Animal Physiology and Genetics, Academy of Sciences, 60200 Brno, Czech Republic; (A.K.); (A.R.); (B.V.); (E.S.); (H.L.)
- Institute of Physiology, Faculty of Veterinary Medicine, Veterinary University Brno, 61200 Brno, Czech Republic
- Correspondence:
| |
Collapse
|
31
|
Nichols P, Urriola J, Miller S, Bjorkman T, Mahady K, Vegh V, Nasrallah F, Winter C. Blood-brain barrier dysfunction significantly correlates with serum matrix metalloproteinase-7 (MMP-7) following traumatic brain injury. NEUROIMAGE-CLINICAL 2021; 31:102741. [PMID: 34225019 PMCID: PMC8264212 DOI: 10.1016/j.nicl.2021.102741] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/20/2021] [Accepted: 06/21/2021] [Indexed: 01/02/2023]
Abstract
Matrix metalloproteinase (MMP) 7 is elevated in traumatic brain injury. Blood brain barrier dysfunction as measured by DCE MRI can be expressed as KTrans. MMP-7 shows a strong correlation with BBB dysfunction shown on MRI. MMP-7 shows potential to function as a serum biomarker.
Objectives To determine if radiological evidence of blood brain barrier (BBB) dysfunction, measured using Dynamic Contrast Enhanced MRI (DCE-MRI), correlates with serum matrix metalloproteinase (MMP) levels in traumatic brain injury (TBI) patients, and thereby, identify a potential biomarker for BBB dysfunction. Patients and Methods 20 patients with a mild, moderate, or severe TBI underwent a DCE-MRI scan and BBB dysfunction was interpreted from KTrans. KTrans is a measure of capillary permeability that reflects the efflux of gadolinium contrast into the extra-cellar space. The serum samples were concurrently collected and later analysed for MMP-1, −2, −7, −9, and −10 levels using an ELISA assay. Statistical correlations between MMP levels and the KTrans value were calculated. Multiple testing was corrected using the Benjamin–Hochberg method to control the false‐discovery rate (FDR). Results Serum MMP-1 values ranged from 1.5 to 49.6 ng/ml (12 ± 12.7), MMP-2 values from 58.3 to 174.1 ng/ml (109.5 ± 26.7), MMP-7 from 1.5 to 31.5 ng/mL (10 ± 7.4), MMP-9 from 128.6 to 1917.5 ng/ml (647.7 ± 749.6) and MMP-10 from 0.1 to 0.6 ng/mL (0.3 ± 0.2). Non-parametric Spearman correlation analysis on the data showed significant positive relationship between KTrans and MMP-7 (r = 0.55, p < 0.01). Correlations were also found between KTrans and MMP-1 (r = 0.74, p < 0.0002) and MMP-2 (r = 0.5, p < 0.025) but the actual MMP values were not above reference ranges, limiting the interpretation of results. Statistically significant correlations between KTrans and either MMP-9 or −10 were not found. Conclusion This is the first study to show a correlation between DCE measures and MMP values in patients with a TBI. Our results support the suggestion that serum MMP-7 may be considered as a peripheral biomarker quantifying BBB dysfunction in TBI patients.
Collapse
Affiliation(s)
- Paul Nichols
- Department of Neurosurgery, Royal Brisbane and Women's Hospital, Australia.
| | - Javier Urriola
- Queensland Brain Institute, The University of Queensland, Australia; Australian e-Health Research Centre, CSIRO, Brisbane, Australia
| | - Stephanie Miller
- University of Queensland Centre for Clinical Research, The University of Queensland, Royal Brisbane and Women's Hospital, Australia
| | - Tracey Bjorkman
- University of Queensland Centre for Clinical Research, The University of Queensland, Royal Brisbane and Women's Hospital, Australia
| | - Kate Mahady
- Department of Radiology, Royal Brisbane and Women's Hospital, Australia
| | - Viktor Vegh
- The Centre for Advanced Imaging, The University of Queensland, Australia; The ARC Centre for Innovation in Biomedical Imaging Technology, Brisbane, Australia
| | - Fatima Nasrallah
- Queensland Brain Institute, The University of Queensland, Australia
| | - Craig Winter
- Department of Neurosurgery, Royal Brisbane and Women's Hospital, Australia; Faculty of Medicine, The University of Queensland, Australia; School of Clinical Sciences, Queensland University of Technology, Australia
| |
Collapse
|
32
|
Sah N, Kuehu DL, Khadka VS, Deng Y, Jha R, Wasti S, Mishra B. RNA sequencing-based analysis of the magnum tissues revealed the novel genes and biological pathways involved in the egg-white formation in the laying hen. BMC Genomics 2021; 22:318. [PMID: 33932994 PMCID: PMC8088581 DOI: 10.1186/s12864-021-07634-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 04/21/2021] [Indexed: 02/06/2023] Open
Abstract
Background The mechanism of egg formation in the oviduct of laying hens is tightly controlled; each segment of the oviduct contributes a unique component of the egg. Several genes/proteins are involved in the synthesis of a completely healthy egg. This implies a time- and tissue-specific expression of genes and proteins in the different oviductal segments. We used hens at different physiological stages and time points to understand the transcriptional regulation of egg-white (albumen) synthesis and secretion onto the eggs in the magnum of laying hens. This study used Next-Generation Sequencing and quantitative real-time PCR (qPCR) to detect the novel genes and the cognate biological pathways that regulate the major events during the albumen formation. Results Magnum tissues collected from laying (n = 5 each at 3 h post-ovulation, p.o. and 15–20 h p.o.), non-laying (n = 4), and molting (n = 5) hens were used for differential gene expression analyses. A total of 540 genes (152 upregulated and 388 down-regulated) were differentially expressed at 3 h p.o. in the magnum of laying hens. Kyoto Encyclopedia of Genes and Genomes pathways analysis of the 152 upregulated genes revealed that glycine, serine, and threonine metabolism was the most-enriched biological pathway. Furthermore, the top two most enriched keywords for the upregulated genes were amino-acid biosynthesis and proteases. Nine candidate genes associated with albumen formation were validated with qPCR to have differential expression in laying, non-laying, and molting hens. Proteases such as TMPRSS9, CAPN2, MMP1, and MMP9 (protein maturation, ECM degradation, and angiogenesis); enzymes such as PSPH, PHGDH, and PSAT1 (amino-acid biosynthesis); RLN3, ACE, and REN (albumen synthesis, secretion and egg transport); and AVD, AvBD11, and GPX3 (antimicrobial and antioxidants) were recognized as essential molecules linked to albumen deposition in the magnum. Conclusions This study revealed some novel genes that participate in the signaling pathways for egg-white synthesis and secretion along with some well-known functional genes. These findings help to understand the mechanisms involved in albumen biosynthesis. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-07634-x.
Collapse
Affiliation(s)
- Nirvay Sah
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, HI, 96822, Honolulu, USA
| | - Donna Lee Kuehu
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Vedbar Singh Khadka
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, 96813, USA
| | - Youping Deng
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, 96813, USA
| | - Rajesh Jha
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, HI, 96822, Honolulu, USA
| | - Sanjeev Wasti
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, HI, 96822, Honolulu, USA
| | - Birendra Mishra
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, HI, 96822, Honolulu, USA.
| |
Collapse
|
33
|
Piperigkou Z, Kyriakopoulou K, Koutsakis C, Mastronikolis S, Karamanos NK. Key Matrix Remodeling Enzymes: Functions and Targeting in Cancer. Cancers (Basel) 2021; 13:1441. [PMID: 33809973 PMCID: PMC8005147 DOI: 10.3390/cancers13061441] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/05/2021] [Accepted: 03/17/2021] [Indexed: 12/13/2022] Open
Abstract
Tissue functionality and integrity demand continuous changes in distribution of major components in the extracellular matrices (ECMs) under normal conditions aiming tissue homeostasis. Major matrix degrading proteolytic enzymes are matrix metalloproteinases (MMPs), plasminogen activators, atypical proteases such as intracellular cathepsins and glycolytic enzymes including heparanase and hyaluronidases. Matrix proteases evoke epithelial-to-mesenchymal transition (EMT) and regulate ECM turnover under normal procedures as well as cancer cell phenotype, motility, invasion, autophagy, angiogenesis and exosome formation through vital signaling cascades. ECM remodeling is also achieved by glycolytic enzymes that are essential for cancer cell survival, proliferation and tumor progression. In this article, the types of major matrix remodeling enzymes, their effects in cancer initiation, propagation and progression as well as their pharmacological targeting and ongoing clinical trials are presented and critically discussed.
Collapse
Affiliation(s)
- Zoi Piperigkou
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 265 04 Patras, Greece; (K.K.); (C.K.)
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), 265 04 Patras, Greece
| | - Konstantina Kyriakopoulou
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 265 04 Patras, Greece; (K.K.); (C.K.)
| | - Christos Koutsakis
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 265 04 Patras, Greece; (K.K.); (C.K.)
| | | | - Nikos K. Karamanos
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 265 04 Patras, Greece; (K.K.); (C.K.)
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), 265 04 Patras, Greece
| |
Collapse
|
34
|
Jiang S, Fu R, Shi J, Wu H, Mai J, Hua X, Chen H, Liu J, Lu M, Li N. CircRNA-Mediated Regulation of Angiogenesis: A New Chapter in Cancer Biology. Front Oncol 2021; 11:553706. [PMID: 33777729 PMCID: PMC7988083 DOI: 10.3389/fonc.2021.553706] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 01/07/2021] [Indexed: 12/15/2022] Open
Abstract
Angiogenesis is necessary for carcinoma progression and is regulated by a variety of pro- and anti-angiogenesis factors. CircRNAs are RNA molecules that do not have a 5'-cap or a 3'-polyA tail and are involved in a variety of biological functions. While circRNA-mediated regulation of tumor angiogenesis has received much attention, the detailed biological regulatory mechanism remains unclear. In this review, we investigated circRNAs in tumor angiogenesis from multiple perspectives, including its upstream and downstream factors. We believe that circRNAs have natural advantages and great potential for the diagnosis and treatment of tumors, which deserves further exploration.
Collapse
Affiliation(s)
- Shaotao Jiang
- Department of HBP SURGERY II, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Rongdang Fu
- Department of Hepatic Surgery, The First People's Hospital of Foshan, Affiliated Foshan Hospital of Sun Yat-sen University, Foshan, China
| | - Jiewei Shi
- Department of General Surgery, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Huijie Wu
- Department of Obstetrics, The First People's Hospital of Foshan, Affiliated Foshan Hospital of Sun Yat-sen University, Foshan, China
| | - Jialuo Mai
- Department of HBP SURGERY II, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xuefeng Hua
- Department of HBP SURGERY II, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Huan Chen
- Department of HBP SURGERY II, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jie Liu
- Department of HBP SURGERY II, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Minqiang Lu
- Department of HBP SURGERY II, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Ning Li
- Department of HBP SURGERY II, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
35
|
Jiang AN, Liu JT, Zhao K, Wu H, Wang S, Yan K, Yang W. Specific Inhibitor of Matrix Metalloproteinase Decreases Tumor Invasiveness After Radiofrequency Ablation in Liver Tumor Animal Model. Front Oncol 2020; 10:561805. [PMID: 33330030 PMCID: PMC7709861 DOI: 10.3389/fonc.2020.561805] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 10/19/2020] [Indexed: 12/26/2022] Open
Abstract
Objective To determine whether the specific inhibitor of matrix metalloproteinase (MMP)-batimastat (BB-94)-could decrease the progression of liver tumor after radiofrequency ablation (RFA) and achieve better therapeutic efficacy in an animal model. Methods In vitro experiments, the proliferation of H22 liver tumor cells was detected by CCK 8 assay and cell migration was detected by Transwell method. In vivo experiments, H22 murine liver tumors were used. First, 32 mice with one tumor were randomized into four groups (n = 8 each group): control (PBS only), RFA alone (65°C, 5 min), BB-94 (30 mg/kg), RFA+BB-94. The growth rate of the residual tumor and the end point survival were calculated and the pathologic changes were evaluated. Secondly, a total of 48 tumors in 24 animals (paired tumors) were randomized into three groups (n = 8 each group): control, RFA alone, RFA+BB-94. Each mouse was implanted with two tumors subcutaneously, one tumor was treated by RFA and the other was evaluated for distant metastasis after applying BB-94. Results In vitro, the proliferation assay demonstrated higher proliferation ability after heat treatment (0.82 ± 0.07 vs 1.27 ± 0.08, P = 0.008), and it could be inhibited by BB-94 (1.27 ± 0.08 vs 0.67 ± 0.06, P = 0.001). In the cell migration assay, the H22 cells demonstrated enhanced tumor invasiveness in the heat group than the control group (33.7 ± 2.1 vs 19.7 ± 4.9, P = 0.011). And it could be significantly suppressed after BB-94 incubation (33.7 ± 2.1 vs 23.0 ± 4.6, P = 0.009). With one tumor animal, the growth rate of the residual tumor in the BB-94+RFA group was slower than that in the RFA alone group (P = 0.003). And combination of BB-94 could significantly prolong the survival of the mice (40.3 ± 1.4d vs 47.1 ± 1.3d, P = 0.002). The expression of CD31 and VEGF at the coagulation margin were decreased after combined with BB-94. With two tumors animal, the growth of metastasis tumor in the BB-94+RFA group was slower than that in the RFA group (P < 0.001). Conclusion BB-94 combined with RFA reduced the invasiveness of the liver tumor and improved the end-point survival. Our data suggested that targeting the MMP process with the specific inhibition could help to increase overall ablation efficacy.
Collapse
Affiliation(s)
- An-Na Jiang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Ultrasound, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jing-Tao Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pharmacy, Peking University Cancer Hospital & Institute, Beijing, China
| | - Kun Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Ultrasound, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hao Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Ultrasound, Peking University Cancer Hospital & Institute, Beijing, China
| | - Song Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Ultrasound, Peking University Cancer Hospital & Institute, Beijing, China
| | - Kun Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Ultrasound, Peking University Cancer Hospital & Institute, Beijing, China
| | - Wei Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Ultrasound, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
36
|
Sabino F, Madzharova E, Auf dem Keller U. Cell density-dependent proteolysis by HtrA1 induces translocation of zyxin to the nucleus and increased cell survival. Cell Death Dis 2020; 11:674. [PMID: 32826880 PMCID: PMC7442833 DOI: 10.1038/s41419-020-02883-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 08/04/2020] [Accepted: 08/04/2020] [Indexed: 01/01/2023]
Abstract
Proteases modulate critical processes in cutaneous tissue repair to orchestrate inflammation, cell proliferation and tissue remodeling. However, the functional consequences and implications in healing impairments of most cleavage events are not understood. Using iTRAQ-based Terminal Amine Isotopic Labeling of Substrates (TAILS) we had characterized proteolytic signatures in a porcine wound healing model and identified two neo-N termini derived from proteolytic cleavage of the focal adhesion protein and mechanotransducer zyxin. Here, we assign these proteolytic events to the activity of either caspase-1 or serine protease HtrA1 and analyze the biological relevance of the resultant zyxin truncations. By cellular expression of full-length and truncated zyxin proteins, we demonstrate nuclear translocation of a C-terminal zyxin fragment that could also be generated in vitro by HtrA1 cleavage and provide evidence for its anti-apoptotic activities, potentially by regulating the expression of modulators of cell proliferation, protein synthesis and genome stability. Targeted degradomics correlated endogenous generation of the same zyxin fragment with increased cell density in human primary dermal fibroblasts. Hence, this newly identified HtrA1-zyxin protease signaling axis might present a novel mechanism to transiently enhance cell survival in environments of increased cell density like in wound granulation tissue.
Collapse
Affiliation(s)
- Fabio Sabino
- Technical University of Denmark, Department of Biotechnology and Biomedicine, Søltofts Plads, 2800, Kongens Lyngby, Denmark
- ETH Zurich, Department of Biology, Institute of Molecular Health Sciences, Otto-Stern-Weg 7, 8093, Zurich, Switzerland
| | - Elizabeta Madzharova
- ETH Zurich, Department of Biology, Institute of Molecular Health Sciences, Otto-Stern-Weg 7, 8093, Zurich, Switzerland
| | - Ulrich Auf dem Keller
- Technical University of Denmark, Department of Biotechnology and Biomedicine, Søltofts Plads, 2800, Kongens Lyngby, Denmark.
- ETH Zurich, Department of Biology, Institute of Molecular Health Sciences, Otto-Stern-Weg 7, 8093, Zurich, Switzerland.
| |
Collapse
|
37
|
Zhu X, Wang F, Wu X, Li Z, Wang Z, Ren X, Zhou Y, Song F, Liang Y, Zeng Z, Liao W, Ding Y, Liao W, Liang L. FBX8 promotes metastatic dormancy of colorectal cancer in liver. Cell Death Dis 2020; 11:622. [PMID: 32796813 PMCID: PMC7427987 DOI: 10.1038/s41419-020-02870-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 07/28/2020] [Accepted: 07/28/2020] [Indexed: 12/17/2022]
Abstract
Patients with colorectal cancer (CRC) often develop malignant regrowth of metastatic dormant tumor cells in liver years after primary treatment. FBX8 is involved in suppressing tumor metastasis. Short-term chemotherapy experiments and liver metastasis mice model of orthotopic injection into the cecum were performed to construct the dormant models. GST-pull-down assay, Co-IP and immunofluorescence were used to confirm the bindings among FBX8 and its substrates. FBX8 upregulated the expression of epithelial and stemness markers, while downregulated the expression of mesenchymal and proliferative markers associated with tumor cell dormancy. FBX8 promoted the maintenance of metastatic dormancy of CRC cells. Mechanistically, FBX8 directly bound to HIF-1α, CDK4 and C-myc through its Sec7 domain and led to the ubiquitin degradation of these proteins, thereby inhibiting cell cycle progression, proliferation, angiogenesis, and metastasis. Clinically, FBX8 expression was negatively correlated with the HIF-1α, CDK4, and c-Myc in CRC tissues. Our study reveals a novel mechanism of FBX8 in regulating tumor metastatic dormancy in liver and provides new strategies for the treatment of CRC metastasis.
Collapse
Affiliation(s)
- Xiaohui Zhu
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangzhou, 510515, Guangdong Province, People's Republic of China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, 510515, Guangzhou, Guangdong Province, People's Republic of China
| | - Feifei Wang
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangzhou, 510515, Guangdong Province, People's Republic of China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, 510515, Guangzhou, Guangdong Province, People's Republic of China
| | - Xuehui Wu
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangzhou, 510515, Guangdong Province, People's Republic of China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, 510515, Guangzhou, Guangdong Province, People's Republic of China
| | - Zhou Li
- The First Clinical Medical Department, Southern Medical University, 510515, Guangzhou, Guangdong Province, People's Republic of China
| | - Zhizhi Wang
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, Guangdong Province, People's Republic of China
| | - Xiaoli Ren
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangzhou, 510515, Guangdong Province, People's Republic of China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, 510515, Guangzhou, Guangdong Province, People's Republic of China
| | - Yangshu Zhou
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangzhou, 510515, Guangdong Province, People's Republic of China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, 510515, Guangzhou, Guangdong Province, People's Republic of China
| | - Fuyao Song
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangzhou, 510515, Guangdong Province, People's Republic of China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, 510515, Guangzhou, Guangdong Province, People's Republic of China
| | - Yunshi Liang
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangzhou, 510515, Guangdong Province, People's Republic of China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, 510515, Guangzhou, Guangdong Province, People's Republic of China
| | - Zhicheng Zeng
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangzhou, 510515, Guangdong Province, People's Republic of China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, 510515, Guangzhou, Guangdong Province, People's Republic of China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, Guangdong Province, People's Republic of China
| | - Yanqing Ding
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangzhou, 510515, Guangdong Province, People's Republic of China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, 510515, Guangzhou, Guangdong Province, People's Republic of China
| | - Wenting Liao
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangzhou, 510515, Guangdong Province, People's Republic of China. .,Guangdong Province Key Laboratory of Molecular Tumor Pathology, 510515, Guangzhou, Guangdong Province, People's Republic of China.
| | - Li Liang
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangzhou, 510515, Guangdong Province, People's Republic of China. .,Guangdong Province Key Laboratory of Molecular Tumor Pathology, 510515, Guangzhou, Guangdong Province, People's Republic of China.
| |
Collapse
|
38
|
TNM staging for GIT cancers is correlated with the level of MMPs and TGF-β1. Clin Exp Med 2020; 20:545-555. [PMID: 32772210 DOI: 10.1007/s10238-020-00651-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 07/31/2020] [Indexed: 02/08/2023]
Abstract
Gastrointestinal (GIT) cancers represent the third common cancers worldwide, characterized by rapid progression and higher mortality rate. Matrix metalloproteinases (MMPs) play an important role in cancer metastases. The present study was conducted to estimate and evaluate the role of MMP-7, -9, -10 and -12 and TGF β1 along with conventional biomarkers (CEA and CA19-9) in gastric (GC), pancreatic (PC) and colorectal cancer (CRC) staging system according to tumor size (T), included lymph node (N) and metastasis (M). Seventy-five patients were divided into GC group (n = 25), PC group (n = 25), CRC group (n = 25) and twenty-five healthy subjects (control group). Serum levels of MMP-7, -10 and -12 were assayed simultaneously using luminex multiplex technique. Also, MMP-9, TGF-β1, CA19-9 and CEA were determined by ELISA. MMP-7,-9,-10, -12, TGF-β1 and CEA levels were significantly (p < 0.001) higher in GIT cancer groups compared with control. CA19-9 was significantly (p < 0.001) higher in PC and CRC groups compared with control. MMP-9 was positively correlated with TNM staging in PC patients. MMP-12 was negatively correlated with T in PC and positively correlated with M in CRC group. CA 19-9 was positively correlated with M grade in CRC. Depending on the estimated cutoff values of area under receiver curve; CA19-9 and MMP-7 were excellent diagnostic markers in PC, CEA and MMP-7 were excellent in CRC, and MMP-7 and MMP-9 were excellent in GC. Our findings indicated the clinical utility of MMPs in diagnosis and TNM staging of GIT cancers along with CEA and CA19-9.
Collapse
|
39
|
Aguirre JE, Beswick EJ, Grim C, Uribe G, Tafoya M, Chacon Palma G, Samedi V, McKee R, Villeger R, Fofanov Y, Cong Y, Yochum G, Koltun W, Powell D, Pinchuk IV. Matrix metalloproteinases cleave membrane-bound PD-L1 on CD90+ (myo-)fibroblasts in Crohn's disease and regulate Th1/Th17 cell responses. Int Immunol 2020; 32:57-68. [PMID: 31633754 DOI: 10.1093/intimm/dxz060] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 09/30/2019] [Indexed: 01/01/2023] Open
Abstract
Increased T helper (Th)1/Th17 immune responses are a hallmark of Crohn's disease (CD) immunopathogenesis. CD90+ (myo-)fibroblasts (MFs) are abundant cells in the normal (N) intestinal mucosa contributing to mucosal tolerance via suppression of Th1 cell activity through cell surface membrane-bound PD-L1 (mPD-L1). CD-MFs have a decreased level of mPD-L1. Consequently, mPD-L1-mediated suppression of Th1 cells by CD-MFs is decreased, yet the mechanism responsible for the reduction in mPDL-1 is unknown. Increased expression of matrix metalloproteinases (MMPs) has been reported in CD. Herein we observed that when compared to N- and ulcerative colitis (UC)-MFs, CD-MFs increase in LPS-inducible levels of MMP-7 and -9 with a significant increase in both basal and inducible MMP-10. A similar pattern of MMP expression was observed in the CD-inflamed mucosa. Treatment of N-MFs with a combination of recombinant human MMP-7, -9 and -10 significantly decreased mPD-L1. In contrast, inhibition of MMP activity with MMP inhibitors or anti-MMP-10 neutralizing antibodies restores mPD-L1 on CD-MFs. CD-MFs demonstrated reduced capacity to suppress Th1 and Th17 responses from activated CD4+ T cells. By contrast, supplementation of the CD-MF:T-cell co-cultures with MMP inhibitors or anti-MMP neutralizing antibodies restored the CD-MF-mediated suppression. Our data suggest that (i) increased MMP-10 expression by CD-MFs and concomitant cleavage of PD-L1 from the surface of CD-MFs are likely to be one of the factors contributing to the decrease of mPD-L1-mediated suppression of Th1/Th17 cells in CD; and (ii) MMPs are likely to have a significant role in the intestinal mucosal immune responses.
Collapse
Affiliation(s)
- Jose E Aguirre
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA.,Institute of Translational Science, University of Texas Medical Branch, Galveston, TX, USA
| | - Ellen J Beswick
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Carl Grim
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Gabriela Uribe
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA.,Department of Medicine at PennState Health Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Marissa Tafoya
- Department of Pathology, University of New Mexico, Albuquerque, NM, USA
| | | | - Von Samedi
- School of Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Rohini McKee
- Department of Surgery at the University of New Mexico, Albuquerque, NM, USA
| | - Romain Villeger
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Yuriy Fofanov
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Yingzi Cong
- Microbiology and Immunology at the University of Texas Medical Branch, Galveston, TX, USA
| | - Gregory Yochum
- Department of Biochemistry and Molecular Biology, PennState Health Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Walter Koltun
- Department of Colorectal Surgery at PennState Health Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Don Powell
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA.,Institute of Translational Science, University of Texas Medical Branch, Galveston, TX, USA
| | - Irina V Pinchuk
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA.,Institute of Translational Science, University of Texas Medical Branch, Galveston, TX, USA.,Department of Medicine at PennState Health Milton S. Hershey Medical Center, Hershey, PA, USA.,Microbiology and Immunology at the University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
40
|
Liu J, Chen Z, Huang M, Tang S, Wang Q, Hu P, Gupta P, Ashby CR, Chen ZS, Zhang L. Plasminogen activator inhibitor (PAI) trap3, an exocellular peptide inhibitor of PAI-1, attenuates the rearrangement of F-actin and migration of cancer cells. Exp Cell Res 2020; 391:111987. [DOI: 10.1016/j.yexcr.2020.111987] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 03/27/2020] [Accepted: 03/29/2020] [Indexed: 12/25/2022]
|
41
|
Wu Y, Meng D, You Y, Sun R, Yan Q, Bao J, Sun Y, Yun D, Li Y, Sun D. Increased expression of RBMS3 predicts a favorable prognosis in human gallbladder carcinoma. Oncol Rep 2020; 44:55-68. [PMID: 32627033 PMCID: PMC7251710 DOI: 10.3892/or.2020.7594] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 03/22/2020] [Indexed: 01/18/2023] Open
Abstract
Multiple regions in the short arm of chromosome 3 are frequently deleted in a variety of solid tumors including gallbladder carcinoma (GBC). RNA binding motif, single‑stranded interacting protein 3 (RBMS3), a tumor suppressor gene (TSG), is located in this region. However, the role of RBMS3 in GBC remains unclear. Reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) and western blotting were performed to evaluate the mRNA and protein expression levels of RBMS3 in 41 fresh frozen GBC tissues and paired normal tissues. An immunohistochemical assay was performed on a tissue microarray (TMA, consisting of 125 cases GBC and 47 normal controls). Microvessel density (MVD) counts were determined using CD34 immunohistochemical staining. Moreover, univariate and multivariate analyses were performed to determine the correlations between RBMS3 expression, MVD and patient prognosis. Cellular functions including proliferation, clonogenicity and apoptosis, were assessed to further identify in vitro roles of RBMS3. It was revealed that both mRNA and protein expression levels of RBMS3 were significantly lower in GBC tissues than in normal controls. Multivariate Cox regression analyses demonstrated cytoplasmic RBMS3 expression as an independent prognostic factor correlated with GBC angiogenesis, histopathological differentiation and TNM stage. Kaplan‑Meier curves revealed that patients with lower cytoplasmic RBMS3 levels had a significantly worse OS than patients with higher cytoplasmic RBMS3 expression. Additionally, ectopic expression of RBMS3 markedly suppressed GBC cell proliferation and clonogenicity and promoted apoptosis in vitro. These findings indicated the potential of cytoplasmic RBMS3 as a tumor prognostic biomarker and a promising therapeutic target for GBC.
Collapse
Affiliation(s)
- Youliang Wu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Delong Meng
- Department of Molecular Biology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yexiang You
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Ruochuan Sun
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Qiang Yan
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Junjun Bao
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Yanjun Sun
- Department of General Surgery, The Armed Police Corps Hospital of Anhui, Hefei, Anhui 230041, P.R. China
| | - Dapeng Yun
- Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yongxiang Li
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Dengqun Sun
- Department of General Surgery, The Armed Police Corps Hospital of Anhui, Hefei, Anhui 230041, P.R. China
| |
Collapse
|
42
|
Li Y, Wang W, Li L, Khalil RA. MMPs and ADAMs/ADAMTS inhibition therapy of abdominal aortic aneurysm. Life Sci 2020; 253:117659. [PMID: 32283055 DOI: 10.1016/j.lfs.2020.117659] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 04/05/2020] [Accepted: 04/07/2020] [Indexed: 12/12/2022]
Abstract
Abdominal aortic aneurysm (AAA) is a chronic vascular degenerative disease featured by progressive dilation and remodeling of the vascular wall, which may lead to aortic rupture and high mortality. The occurrence and development of AAA involve multiple mechanisms, including extracellular matrix degradation, chronic inflammation, oxidative stress, apoptosis of vascular smooth muscle cells and innate immunity. Extracellular matrix degradation is considered as the most important mechanism causing AAA. Matrix metalloproteinases (MMPs) are key factors in this process, contributing greatly to the occurrence and development of AAA. But whether the zinc-dependent endopeptidases (ADAM/ADAMTS) are involved in this process is very little known. This study is a review about the role of MMPs and ADAM/ADAMT as well as the existing MMP inhibitors in abdominal aortic aneurysm, with the purpose of providing reference for the clinical treatment of abdominal aortic aneurysm.
Collapse
Affiliation(s)
- Yongqi Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China; Graduate School of Comprehensive Human Sciences, University of Tsukuba, Japan
| | - Weicheng Wang
- Emergency Center, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Lei Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China; Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
43
|
Furuya H, Chan OT, Hokutan K, Tsukikawa Y, Chee K, Kozai L, Chan KS, Dai Y, Wong RS, Rosser CJ. Prognostic Significance of Lymphocyte Infiltration and a Stromal Immunostaining of a Bladder Cancer Associated Diagnostic Panel in Urothelial Carcinoma. Diagnostics (Basel) 2019; 10:diagnostics10010014. [PMID: 31905599 PMCID: PMC7168167 DOI: 10.3390/diagnostics10010014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 12/20/2019] [Accepted: 12/22/2019] [Indexed: 01/08/2023] Open
Abstract
We set out to expand on our previous work in which we reported the epithelial expression pattern of a urine-based bladder cancer-associated diagnostic panel (A1AT, ANG, APOE, CA9, IL8, MMP9, MMP10, PAI1, SDC1, and VEGFA). Since many of the analytes in the bladder cancer-associated diagnostic signature were chemokines, cytokines, or secreted proteins, we set out to report the stromal staining pattern of the diagnostic signature as well as CD3+ (T-cell) cell and CD68+ (macrophage) cell staining in human bladder tumors as a snapshot of the tumor immune landscape. Immunohistochemical staining was performed on 213 tumor specimens and 74 benign controls. Images were digitally captured and quantitated using Aperio (Vista, CA). The expression patterns were correlated with tumor grade, tumor stage, and outcome measures. We noted a positive correlation of seven of the 10 proteins (excluding A1AT and IL8 which had a negative association and VEGFA had no association) in bladder cancer. The overexpression of MMP10 was associated with higher grade disease, while overexpression of MMP10, PAI1, SDC1 and ANG were associated with high stage bladder cancer and CA9 was associated with low stage bladder cancer. Increased tumor infiltration of CD68+ cells were associated with higher stage disease. Overall survival was significantly reduced in bladder cancer patients' whose tumors expressed eight or more of the 10 proteins that comprise the bladder cancer diagnostic panel. These findings confirm that the chemokines, cytokines, and secreted proteins in a urine-based diagnostic panel are atypically expressed, not only in the epithelial component of bladder tumors, but also in the stromal component of bladder tumors and portends a worse overall survival. Thus, when assessing immunohistochemical staining, it is important to report staining patterns within the stroma as well as the entire stroma itself.
Collapse
Affiliation(s)
- Hideki Furuya
- Clinical and Translational Research Program, University of Hawaii Cancer Center, Honolulu, HI 96813, USA; (H.F.); (K.H.); (Y.T.); (K.C.)
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, HI 96822, USA
- Department of Surgery, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA;
| | - Owen T.M. Chan
- Clinical and Translational Research Program, University of Hawaii Cancer Center, Honolulu, HI 96813, USA; (H.F.); (K.H.); (Y.T.); (K.C.)
| | - Kanani Hokutan
- Clinical and Translational Research Program, University of Hawaii Cancer Center, Honolulu, HI 96813, USA; (H.F.); (K.H.); (Y.T.); (K.C.)
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, HI 96822, USA
| | - Yutaro Tsukikawa
- Clinical and Translational Research Program, University of Hawaii Cancer Center, Honolulu, HI 96813, USA; (H.F.); (K.H.); (Y.T.); (K.C.)
| | - Keanu Chee
- Clinical and Translational Research Program, University of Hawaii Cancer Center, Honolulu, HI 96813, USA; (H.F.); (K.H.); (Y.T.); (K.C.)
| | - Landon Kozai
- John A. Burn School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA;
| | - Keith S. Chan
- Department of Pathology, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA;
| | - Yunfeng Dai
- Department of Biostatistics, University of Florida, Gainesville, FL 32611, USA;
| | - Regan S. Wong
- Department of Surgery, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA;
| | - Charles J. Rosser
- Clinical and Translational Research Program, University of Hawaii Cancer Center, Honolulu, HI 96813, USA; (H.F.); (K.H.); (Y.T.); (K.C.)
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, HI 96822, USA
- Department of Surgery, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA;
- Correspondence:
| |
Collapse
|
44
|
Oktay K, Santaliz-Casiano A, Patel M, Marino N, Storniolo AMV, Torun H, Acar B, Madak Erdogan Z. A Computational Statistics Approach to Evaluate Blood Biomarkers for Breast Cancer Risk Stratification. Discov Oncol 2019; 11:17-33. [PMID: 31858384 DOI: 10.1007/s12672-019-00372-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 11/25/2019] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is the second leading cause of cancer mortality among women. Mammography and tumor biopsy followed by histopathological analysis are the current methods to diagnose breast cancer. Mammography does not detect all breast tumor subtypes, especially those that arise in younger women or women with dense breast tissue, and are more aggressive. There is an urgent need to find circulating prognostic molecules and liquid biopsy methods for breast cancer diagnosis and reducing the mortality rate. In this study, we systematically evaluated metabolites and proteins in blood to develop a pipeline to identify potential circulating biomarkers for breast cancer risk. Our aim is to identify a group of molecules to be used in the design of portable and low-cost biomarker detection devices. We obtained plasma samples from women who are cancer free (healthy) and women who were cancer free at the time of blood collection but developed breast cancer later (susceptible). We extracted potential prognostic biomarkers for breast cancer risk from plasma metabolomics and proteomics data using statistical and discriminative power analyses. We pre-processed the data to ensure the quality of subsequent analyses, and used two main feature selection methods to determine the importance of each molecule. After further feature elimination based on pairwise dependencies, we measured the performance of logistic regression classifier on the remaining molecules and compared their biological relevance. We identified six signatures that predicted breast cancer risk with different specificity and selectivity. The best performing signature had 13 factors. We validated the difference in level of one of the biomarkers, SCF/KITLG, in plasma from healthy and susceptible individuals. These biomarkers will be used to develop low-cost liquid biopsy methods toward early identification of breast cancer risk and hence decreased mortality. Our findings provide the knowledge basis needed to proceed in this direction.
Collapse
Affiliation(s)
- Kaan Oktay
- VAVlab, Electrical & Electronics Engineering Department, Bogazici University, Istanbul, Turkey
| | | | - Meera Patel
- Susan G. Komen Tissue Bank at the IU Simon Cancer Center, Indianapolis, IN, USA
| | - Natascia Marino
- Susan G. Komen Tissue Bank at the IU Simon Cancer Center, Indianapolis, IN, USA.,Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Anna Maria V Storniolo
- Susan G. Komen Tissue Bank at the IU Simon Cancer Center, Indianapolis, IN, USA.,Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Hamdi Torun
- Faculty of Engineering and Environment, University of Northumbria, Newcastle upon Tyne, UK
| | - Burak Acar
- VAVlab, Electrical & Electronics Engineering Department, Bogazici University, Istanbul, Turkey
| | - Zeynep Madak Erdogan
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA. .,Department of Food Sciences and Human Nutrition, University of Illinois Urbana-Champaign, Urbana, IL, USA. .,National Center for Supercomputing Applications, University of Illinois Urbana-Champaign, Urbana, IL, USA. .,Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL, USA. .,Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA. .,Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
45
|
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] [Scholar 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
|
46
|
Chuang HM, Chen YS, Harn HJ. The Versatile Role of Matrix Metalloproteinase for the Diverse Results of Fibrosis Treatment. Molecules 2019; 24:molecules24224188. [PMID: 31752262 PMCID: PMC6891433 DOI: 10.3390/molecules24224188] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 11/14/2019] [Accepted: 11/15/2019] [Indexed: 12/11/2022] Open
Abstract
Fibrosis is a type of chronic organ failure, resulting in the excessive secretion of extracellular matrix (ECM). ECM protects wound tissue from infection and additional injury, and is gradually degraded during wound healing. For some unknown reasons, myofibroblasts (the cells that secrete ECM) do not undergo apoptosis; this is associated with the continuous secretion of ECM and reduced ECM degradation even during de novo tissue formation. Thus, matrix metalloproteinases (MMPs) are considered to be a potential target of fibrosis treatment because they are the main groups of ECM-degrading enzymes. However, MMPs participate not only in ECM degradation but also in the development of various biological processes that show the potential to treat diseases such as stroke, cardiovascular diseases, and arthritis. Therefore, treatment involving the targeting of MMPs might impede typical functions. Here, we evaluated the links between these MMP functions and possible detrimental effects of fibrosis treatment, and also considered possible approaches for further applications.
Collapse
Affiliation(s)
- Hong-Meng Chuang
- Buddhist Tzu Chi Bioinnovation Center, Tzu Chi Foundation, Hualien 970, Taiwan; (H.-M.C.); (Y.-S.C.)
- Department of Medical Research, Hualien Tzu Chi Hospital, Hualien 970, Taiwan
| | - Yu-Shuan Chen
- Buddhist Tzu Chi Bioinnovation Center, Tzu Chi Foundation, Hualien 970, Taiwan; (H.-M.C.); (Y.-S.C.)
- Department of Medical Research, Hualien Tzu Chi Hospital, Hualien 970, Taiwan
| | - Horng-Jyh Harn
- Buddhist Tzu Chi Bioinnovation Center, Tzu Chi Foundation, Hualien 970, Taiwan; (H.-M.C.); (Y.-S.C.)
- Department of Pathology, Hualien Tzu Chi Hospital & Tzu Chi University, Hualien 970, Taiwan
- Correspondence: ; Tel.: +03-8561825 (ext. 15615)
| |
Collapse
|
47
|
Gao Z, Luo X, Liu L, Lin L, Tong M, Yang T. Recombinant
Treponema pallidum
protein Tp47 induces angiogenesis by modulating the matrix metalloproteinase/tissue inhibitor of metalloproteinase balance in endothelial cells. J Eur Acad Dermatol Venereol 2019; 33:1958-1970. [PMID: 31166625 DOI: 10.1111/jdv.15725] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 05/08/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Z.‐X. Gao
- Center of Clinical Laboratory Zhongshan Hospital School of Medicine Xiamen University Xiamen China
- Institute of Infectious Disease School of Medicine Xiamen University Xiamen China
| | - X. Luo
- Center of Clinical Laboratory Zhongshan Hospital School of Medicine Xiamen University Xiamen China
| | - L.‐L. Liu
- Center of Clinical Laboratory Zhongshan Hospital School of Medicine Xiamen University Xiamen China
- Institute of Infectious Disease School of Medicine Xiamen University Xiamen China
| | - L.‐R. Lin
- Center of Clinical Laboratory Zhongshan Hospital School of Medicine Xiamen University Xiamen China
- Institute of Infectious Disease School of Medicine Xiamen University Xiamen China
| | - M.‐L. Tong
- Center of Clinical Laboratory Zhongshan Hospital School of Medicine Xiamen University Xiamen China
| | - T.‐C. Yang
- Center of Clinical Laboratory Zhongshan Hospital School of Medicine Xiamen University Xiamen China
- Institute of Infectious Disease School of Medicine Xiamen University Xiamen China
| |
Collapse
|
48
|
Zhu L, Zheng X, Du Y, Xing Y, Xu K, Cui L. Matrix metalloproteinase-7 may serve as a novel biomarker for cervical cancer. Onco Targets Ther 2018; 11:4207-4220. [PMID: 30050312 PMCID: PMC6055895 DOI: 10.2147/ott.s160998] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background The biological and clinical significance of matrix metalloproteinase-7 (MMP-7) in cervical cancer remains unknown. Here, we investigated the function of MMP-7 in cervical cancer cells and evaluated its clinical significance in both tissues and serum from cervical cancer patients. Methods First, we analyzed the expression of MMP-7 in cervical cancer using Oncomine microarray data and examined its expression in cervical tissues by quantitative real-time polymerase chain reaction and Western blotting. Second, we utilized gene silencing to explore the role of MMP-7 in cells. Finally, we examined the MMP-7 levels in patients with cervical cancer and normal serum by enzyme-linked immunosorbent assay. Moreover, we further investigated the relationship between MMP-7 expression and pathological features. Results The mRNA and protein MMP-7 levels were higher in cervical cancer tissues than in healthy controls. Silencing of MMP-7 significantly decreased cervical cancer cell proliferation, migration, and invasion. The serum MMP-7 levels were significantly higher in cervical cancer patients than in healthy subjects (P<0.01). Further, higher MMP-7 expression was associated with increased lymph metastasis (P=0.021), pathological grade (P=0.039, P=0.047), and clinical stage (P=0.049, P=0.046). Conclusion MMP-7 appears to act as an oncogene in cervical cancer cells and is involved in cell proliferation, migration, and invasion. MMP-7 expression was significantly higher in the tissue and serum of cervical cancer patients compared to healthy individuals and was correlated with increased pathalogical grade, clinical stage, and lymph metastasis. Therefore, our data provide novel evidence that MMP-7 may be a clinically relevant biomarker for cervical cancer.
Collapse
Affiliation(s)
- Linyan Zhu
- Department of Obstetrics and Gynaecology, Ningbo First Hospital, Ningbo, Zhejiang, People's Republic of China,
| | - Xiaojiao Zheng
- Department of Obstetrics and Gynaecology, Ningbo First Hospital, Ningbo, Zhejiang, People's Republic of China,
| | - Yongming Du
- Department of Obstetrics and Gynaecology, Ningbo First Hospital, Ningbo, Zhejiang, People's Republic of China,
| | - Yan Xing
- Department of Obstetrics and Gynaecology, Ningbo First Hospital, Ningbo, Zhejiang, People's Republic of China,
| | - Kejun Xu
- Department of Obstetrics and Gynaecology, Ningbo First Hospital, Ningbo, Zhejiang, People's Republic of China,
| | - Lining Cui
- Department of Obstetrics and Gynaecology, Ningbo First Hospital, Ningbo, Zhejiang, People's Republic of China,
| |
Collapse
|
49
|
Increased FSHD region gene1 expression reduces in vitro cell migration, invasion, and angiogenesis, ex vivo supported by reduced expression in tumors. Biosci Rep 2017; 37:BSR20171062. [PMID: 28947680 PMCID: PMC5665614 DOI: 10.1042/bsr20171062] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 09/19/2017] [Accepted: 09/20/2017] [Indexed: 11/17/2022] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) region gene 1 (FRG1) is a candidate gene for FSHD. FRG1 regulates various muscle-related functions, but studies have proposed its role in development and angiogenesis also, where it is involved with tumor-associated molecules. Therefore, we decided to look into its role in tumor progression, tumor angiogenesis, and its impact on cellular properties. Cell proliferation, migration, invasion and in vitro angiogenesis assays were performed to decipher the effect of FRG1 on endothelial and epithelial cell functions. Q-RT PCR was done for human embyonic kidney (HEK293T) cells with altered FRG1 levels to identify associated molecules. Further, immunohistochemistry was done to identify FRG1 expression levels in various cancers and its association with tumor angiogenesis. Subsequently, inference was drawn from Oncomine and Kaplan-Meier plotter analysis, for FRG1 expression in different cancers. Ectopic expression of FRG1 affected cell migration and invasion in both HEK293T and human umbilical vein endothelial cells (HUVECs). In HUVECs, FRG1 overexpression led to reduced angiogenesis in vitro No effect was observed in cell proliferation in both the cell types. Q-RT PCR data revealed reduction in granulocyte-colony stimulating factor (G-CSF) expression with FRG1 overexpression and increased expression of matrix metalloproteinase 10 (MMP10) with FRG1 knockdown. Immunohistochemistry analysis showed reduced FRG1 levels in tumors which were supported by in silico analysis data. These findings suggest that reduction in FRG1 expression in gastric, colon and oral cavity tumor might have a role in tumor progression, by regulating cell migration and invasiveness. To elucidate a better understanding of molecular signaling involving FRG1 in angiogenesis regulation, further study is required.
Collapse
|
50
|
Upadhyay P, Gardi N, Desai S, Chandrani P, Joshi A, Dharavath B, Arora P, Bal M, Nair S, Dutt A. Genomic characterization of tobacco/nut chewing HPV-negative early stage tongue tumors identify MMP10 asa candidate to predict metastases. Oral Oncol 2017; 73:56-64. [PMID: 28939077 PMCID: PMC5628952 DOI: 10.1016/j.oraloncology.2017.08.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 07/27/2017] [Accepted: 08/06/2017] [Indexed: 02/08/2023]
Abstract
OBJECTIVES Nodal metastases status among early stage tongue squamous cell cancer patients plays a decisive role in the choice of treatment, wherein about 70% patients can be spared from surgery with an accurate prediction of negative pathological lymph node status. This underscores an unmet need for prognostic biomarkers to stratify the patients who are likely to develop metastases. MATERIALS AND METHODS We performed high throughput sequencing of fifty four samples derived from HPV negative early stage tongue cancer patients habitual of chewing betel nuts, areca nuts, lime or tobacco using whole exome (n=47) and transcriptome (n=17) sequencing that were analyzed using in-house computational tools. Additionally, gene expression meta-analyses were carried out for 253 tongue cancer samples. The candidate genes were validated using qPCR and immuno-histochemical analysis in an extended set of 50 early primary tongue cancer samples. RESULTS AND CONCLUSION Somatic analysis revealed a classical tobacco mutational signature C:G>A:T transversion in 53% patients that were mutated in TP53, NOTCH1, CDKN2A, HRAS, USP6, PIK3CA, CASP8, FAT1, APC, and JAK1. Similarly, significant gains at genomic locus 11q13.3 (CCND1, FGF19, ORAOV1, FADD), 5p15.33 (SHANK2, MMP16, TERT), and 8q24.3 (BOP1); and, losses at 5q22.2 (APC), 6q25.3 (GTF2H2) and 5q13.2 (SMN1) were observed in these samples. Furthermore, an integrated gene-expression analysis of 253 tongue tumors suggested an upregulation of metastases-related pathways and over-expression of MMP10 in 48% tumors that may be crucial to predict nodal metastases in early tongue cancer patients. In overall, we present the first descriptive portrait of somatic alterations underlying the genome of tobacco/nut chewing HPV-negative early tongue cancer, and identify MMP10 asa potential prognostic biomarker to stratify those likely to develop metastases.
Collapse
Affiliation(s)
- Pawan Upadhyay
- Integrated Genomics Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai 410210, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Nilesh Gardi
- Integrated Genomics Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai 410210, India
| | - Sanket Desai
- Integrated Genomics Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai 410210, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Pratik Chandrani
- Integrated Genomics Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai 410210, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Asim Joshi
- Integrated Genomics Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai 410210, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Bhaskar Dharavath
- Integrated Genomics Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai 410210, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Priyanca Arora
- Division of Head and Neck Oncology, Department of Surgical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai 400012, India
| | - Munita Bal
- Department of Pathology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai 400012, India
| | - Sudhir Nair
- Division of Head and Neck Oncology, Department of Surgical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai 400012, India
| | - Amit Dutt
- Integrated Genomics Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai 410210, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India.
| |
Collapse
|