1
|
Neuman K, Zhang X, Schellberg B, Lewis LH, Koppes A, Koppes R. Electrical and magnetic stimulation separately modulates the extent and direction of neurite outgrowth in an ionically conductive hydrogel. J Neural Eng 2025; 22:026041. [PMID: 40014876 PMCID: PMC11962742 DOI: 10.1088/1741-2552/adbb1e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 02/05/2025] [Accepted: 02/27/2025] [Indexed: 03/01/2025]
Abstract
Objective. The use of conductive materials for aiding peripheral nerve regeneration is a promising method to recapitulate native conductance of nerve tissue and facilitate the delivery of exogeneous stimulation for enhanced recovery. This study systematically investigated the effects of applying electrical (ES) or magnetic stimulation (MS) to neurons within new ionically conductive hydrogels.Approach. The material properties of ionically conductive Gel-Amin hydrogels (Gelatin methacryloyl (GelMA) + Choline acrylate) were compared to those of GelMA hydrogels. Neonatal rat dorsal root ganglia (DRG) were encapsulated in both hydrogel formulations, subjected to ES or MS, and evaluated for differences in neuronal extension. Peripheral glia, Schwann cells (SCs), were subjected to the same stimuli and their secretion of various neurotrophic analytes were investigated.Main results. Gel-Amin hydrogels are 4x more ionically conductive than GelMA hydrogels. The application of electrical stimulation to the encapsulated cells led to a significant decrease (76%) in DRG outgrowth when encapsulated in GelMA versus the Gel-Amin hydrogel. In contrast, MS led to directional neurite extension in a direction perpendicular to the magnetic field gradient.Significance. We present here the first report of a controlled, direct comparison of ES and MS on whole DRG in synthetic materials. The combination of ES and MS decreased total neurite outgrowth but led to more directional growth. Aspects of the material and type of stimuli were noted to reduce several cytokine secretion levels from primary SC cultures. These results highlight the importance of understanding material and biophysical interactions to enhance peripheral nerve regeneration.
Collapse
Affiliation(s)
- Katelyn Neuman
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States of America
| | - Xiaoyu Zhang
- Department of Mechanical and Industrial Engineering, Northeastern University, Boston, MA, United States of America
| | - Bryan Schellberg
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States of America
| | - Laura H Lewis
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States of America
- Department of Mechanical and Industrial Engineering, Northeastern University, Boston, MA, United States of America
| | - Abigail Koppes
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States of America
- Department of Biology, Northeastern University, Boston, MA, United States of America
- Department of Bioengineering, Northeastern University, Boston, MA, United States of America
| | - Ryan Koppes
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States of America
| |
Collapse
|
2
|
Aswani SS, Aparna NS, Mohan MS, Boban PT, Saja K. Sesame oil downregulates the expression of ADAMTS-4 in high-fat diet-induced atherosclerosis. Prostaglandins Other Lipid Mediat 2024; 174:106862. [PMID: 38936541 DOI: 10.1016/j.prostaglandins.2024.106862] [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: 04/30/2024] [Revised: 06/06/2024] [Accepted: 06/18/2024] [Indexed: 06/29/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease forming plaques in medium and large-sized arteries. ADAMTS-4 (a disintegrin and metalloproteinase with thrombospondin motifs-4) is an extracellular-matrix remodelling enzyme involved in the degradation of versican in the arterial wall. Recent reports indicated that increased expression of ADAMTS-4 is associated with plaque progression and vulnerability. Bioactive components of dietary oil, like sesame oil, are reported to have anti-inflammatory and antioxidant properties. Here, we studied the effect of sesame oil on regulating ADAMTS-4 in high-fat diet-induced atherosclerosis rat model. Our results indicated that sesame oil supplementation improved the anti-inflammatory and anti-oxidative status of the body. It also reduced atherosclerotic plaque formation in high-fat diet-fed rats. Our results showed that the sesame oil supplementation significantly down-regulated the expression of ADAMTS-4 in serum and aortic samples. The versican, the large proteoglycan substrate of ADAMTS-4 in the aorta, was downregulated to normal control level on sesame oil supplementation. This study, for the first time, reveals that sesame oil could down-regulate the expression of ADAMTS-4 in high-fat diet-induced atherosclerosis, imparting a new therapeutic potential for sesame oil in the management of atherosclerosis.
Collapse
Affiliation(s)
- S S Aswani
- Department of Biochemistry, University of Kerala, Kariavattom, Thiruvananthapuram, Kerala 695581, India
| | - N S Aparna
- Department of Biochemistry, University of Kerala, Kariavattom, Thiruvananthapuram, Kerala 695581, India
| | - Mithra S Mohan
- Department of Biochemistry, University of Kerala, Kariavattom, Thiruvananthapuram, Kerala 695581, India
| | - P T Boban
- Department of Biochemistry, Government College Kariavattom, Thiruvananthapuram, Kerala 695581, India
| | - K Saja
- Department of Biochemistry, University of Kerala, Kariavattom, Thiruvananthapuram, Kerala 695581, India.
| |
Collapse
|
3
|
Ai Y, Ding Q, Wan Z, Tyagi S, Indeglia A, Murphy M, Tian B. Regulation of alternative polyadenylation isoforms of Timp2 is an effector event of RAS signaling in cell transformation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.26.613909. [PMID: 39386512 PMCID: PMC11463442 DOI: 10.1101/2024.09.26.613909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Alternative polyadenylation (APA) generates mRNA isoforms with different lengths of the 3' untranslated region (3' UTR). The tissue inhibitor of metalloproteinase 2 (TIMP2) plays a key role in extracellular matrix remodeling under various developmental and disease conditions. Both human and mouse genes encoding TIMP2 contain two highly conserved 3'UTR APA sites, leading to mRNA isoforms that differ substantially in 3'UTR size. APA of Timp2 is one of the most significantly regulated events in multiple cell differentiation lineages. Here we show that Timp2 APA is highly regulated in transformation of NIH3T3 cells by the oncogene HRAS G12V . Perturbations of isoform expression with long 3'UTR isoform-specific knockdown or genomic removal of the alternative UTR (aUTR) region indicate that the long 3'UTR isoform contributes to the secreted Timp2 protein much more than the short 3'UTR isoform. The short and long 3'UTR isoforms differ in subcellular localization to endoplasmic reticulum (ER). Strikingly, Timp2 aUTR enhances secreted protein expression but no effect on intracellular proteins in reporter assays. Furthermore, downregulation of Timp2 long isoform mitigates gene expression changes elicited by HRAS G12V . Together, our data indicate that regulation of Timp2 protein expression through APA isoform changes is an integral part of RAS-mediated cell transformation and 3'UTR isoforms of Timp2 can have distinct impacts on expression of secreted vs. intracellular proteins.
Collapse
|
4
|
Namazi M, Eftekhar SP, Mosaed R, Shiralizadeh Dini S, Hazrati E. Pulmonary Hypertension and Right Ventricle: A Pathophysiological Insight. CLINICAL MEDICINE INSIGHTS-CARDIOLOGY 2024; 18:11795468241274744. [PMID: 39257563 PMCID: PMC11384539 DOI: 10.1177/11795468241274744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 07/21/2024] [Indexed: 09/12/2024]
Abstract
Background Pulmonary hypertension (PH) is a pulmonary vascular disease characterized by elevated pulmonary vascular pressure. Long-term PH, irrespective of its etiology, leads to increased right ventricular (RV) pressure, RV hypertrophy, and ultimately, RV failure. Main body Research indicates that RV failure secondary to hypertrophy remains the primary cause of mortality in pulmonary arterial hypertension (PAH). However, the impact of PH on RV structure and function under increased overload remains incompletely understood. Several mechanisms have been proposed, including extracellular remodeling, RV hypertrophy, metabolic disturbances, inflammation, apoptosis, autophagy, endothelial-to-mesenchymal transition, neurohormonal dysregulation, capillary rarefaction, and ischemia. Conclusions Studies have demonstrated the significant role of oxidative stress in the development of RV failure. Understanding the interplay among these mechanisms is crucial for the prevention and management of RV failure in patients with PH.
Collapse
Affiliation(s)
- Mehrshad Namazi
- Trauma and Surgery Research Center, AJA University of Medical Sciences, Tehran, Iran
- Clinical Biomechanics and Ergonomics Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Seyed Parsa Eftekhar
- Trauma and Surgery Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Reza Mosaed
- Trauma and Surgery Research Center, AJA University of Medical Sciences, Tehran, Iran
| | | | - Ebrahim Hazrati
- Trauma and Surgery Research Center, AJA University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Hosseinzadeh A, Alinaghian N, Sheibani M, Seirafianpour F, Naeini AJ, Mehrzadi S. Melatonin: Current evidence on protective and therapeutic roles in gynecological diseases. Life Sci 2024; 344:122557. [PMID: 38479596 DOI: 10.1016/j.lfs.2024.122557] [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: 01/01/2024] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 03/17/2024]
Abstract
Melatonin, a potent antioxidant and free radical scavenger, has been demonstrated to be effective in gynecological conditions and female reproductive cancers. This review consolidates the accumulating evidence on melatonin's multifaceted protective effects in different pathological contexts. In gynecological conditions such as endometriosis, polycystic ovary syndrome (PCOS), and uterine leiomyoma, melatonin has shown promising effects in reducing oxidative stress, inflammation, and hormonal imbalances. It inhibits adhesion molecules' production, and potentially mitigates leukocyte adherence and inflammatory responses. Melatonin's regulatory effects on hormone production and insulin sensitivity in PCOS individuals make it a promising candidate for improving oocyte quality and menstrual irregularities. Moreover, melatonin exhibits significant antitumor effects by modulating various signaling pathways, promoting apoptosis, and suppressing metastasis in breast cancers and gynecological cancers, including ovarian, endometrial, and cervical cancers. Furthermore, melatonin's protective effects are suggested to be mediated by interactions with its receptors, estrogen receptors and other nuclear receptors. The regulation of clock-related genes and circadian clock systems may also contribute to its inhibitory effects on cancer cell growth. However, more comprehensive research is warranted to fully elucidate the underlying molecular mechanisms and establish melatonin as a potential therapeutic agent for these conditions.
Collapse
Affiliation(s)
- Azam Hosseinzadeh
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Nazila Alinaghian
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Sheibani
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Ali Jamshidi Naeini
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Saeed Mehrzadi
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Diwan R, Bhatt HN, Beaven E, Nurunnabi M. Emerging delivery approaches for targeted pulmonary fibrosis treatment. Adv Drug Deliv Rev 2024; 204:115147. [PMID: 38065244 PMCID: PMC10787600 DOI: 10.1016/j.addr.2023.115147] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 11/02/2023] [Accepted: 11/29/2023] [Indexed: 01/01/2024]
Abstract
Pulmonary fibrosis (PF) is a progressive, and life-threatening interstitial lung disease which causes scarring in the lung parenchyma and thereby affects architecture and functioning of lung. It is an irreversible damage to lung functioning which is related to epithelial cell injury, immense accumulation of immune cells and inflammatory cytokines, and irregular recruitment of extracellular matrix. The inflammatory cytokines trigger the differentiation of fibroblasts into activated fibroblasts, also known as myofibroblasts, which further increase the production and deposition of collagen at the injury sites in the lung. Despite the significant morbidity and mortality associated with PF, there is no available treatment that efficiently and effectively treats the disease by reversing their underlying pathologies. In recent years, many therapeutic regimens, for instance, rho kinase inhibitors, Smad signaling pathway inhibitors, p38, BCL-xL/ BCL-2 and JNK pathway inhibitors, have been found to be potent and effective in treating PF, in preclinical stages. However, due to non-selectivity and non-specificity, the therapeutic molecules also result in toxicity mediated severe side effects. Hence, this review demonstrates recent advances on PF pathology, mechanism and targets related to PF, development of various drug delivery systems based on small molecules, RNAs, oligonucleotides, peptides, antibodies, exosomes, and stem cells for the treatment of PF and the progress of various therapeutic treatments in clinical trials to advance PF treatment.
Collapse
Affiliation(s)
- Rimpy Diwan
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX 79902, United States; Department of Biomedical Engineering, College of Engineering, The University of Texas El Paso, El Paso, TX 79968, United States
| | - Himanshu N Bhatt
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX 79902, United States; Department of Biomedical Engineering, College of Engineering, The University of Texas El Paso, El Paso, TX 79968, United States
| | - Elfa Beaven
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX 79902, United States; Department of Biomedical Engineering, College of Engineering, The University of Texas El Paso, El Paso, TX 79968, United States
| | - Md Nurunnabi
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX 79902, United States; Department of Biomedical Engineering, College of Engineering, The University of Texas El Paso, El Paso, TX 79968, United States; The Border Biomedical Research Center, The University of Texas El Paso, El Paso, TX 79968, United States.
| |
Collapse
|
7
|
Zhou Y, Zhang T, Wang S, Yang R, Jiao Z, Lu K, Li H, Jiang W, Zhang X. Targeting of HBP1/TIMP3 axis as a novel strategy against breast cancer. Pharmacol Res 2023; 194:106846. [PMID: 37414199 DOI: 10.1016/j.phrs.2023.106846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/30/2023] [Accepted: 06/30/2023] [Indexed: 07/08/2023]
Abstract
Malignant proliferation and metastasis are the main causes of breast cancer death. The transcription factor high mobility group (HMG) box-containing protein 1 (HBP1) is an important tumor suppressor whose deletion or mutation is closely related to the appearance of tumors. Here, we investigated the role of HBP1 in breast cancer suppression. HBP1 enhances the activity of the tissue inhibitors of metalloproteinases 3 (TIMP3) promoter, thereby increasing protein and mRNA levels of TIMP3. TIMP3 increases the phosphatase and tensin homolog (PTEN) protein level by inhibiting its degradation and acts as a metalloproteinase inhibitor to inhibit the protein levels of MMP2/9. In this study, we demonstrated that the HBP1/TIMP3 axis plays a crucial role in inhibiting the tumorigenesis of breast cancer. HBP1 deletion interferes with the regulation of the axis and induces the occurrence and malignant progression of breast cancer. In addition, the HBP1/TIMP3 axis promotes the sensitivity of breast cancer to radiation therapy and hormone therapy. Our study opens new perspectives on the treatment and prognosis of breast cancer.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, PR China
| | - Tongjia Zhang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, PR China
| | - Shujie Wang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, PR China
| | - Ruixiang Yang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, PR China
| | - Zitao Jiao
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, PR China
| | - Kejia Lu
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, PR China
| | - Hui Li
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, PR China
| | - Wei Jiang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, PR China
| | - Xiaowei Zhang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, PR China.
| |
Collapse
|
8
|
Arepalli S, Wykoff CC, Abraham JR, Lunasco L, Yu H, Hu M, Srivastava SK, Reese JL, Brown D, Ehlers JP. Longitudinal analysis of aqueous humour cytokine expression and OCT-based imaging biomarkers in retinal vein occlusions treated with anti-vascular endothelial growth factor therapy in the IMAGINE study. Eye (Lond) 2023; 37:1928-1935. [PMID: 36220884 PMCID: PMC10275974 DOI: 10.1038/s41433-022-02265-2] [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: 11/08/2021] [Revised: 08/14/2022] [Accepted: 09/15/2022] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND/OBJECTIVES Retinal vein occlusion (RVO) is the second most common retinal vascular disorder. Despite promising advances with anti-VEGF therapy, select patients are unresponsive to therapy. A precision medicine-based approach for therapeutic decision-making based on underlying biomarkers may facilitate treatment based on the underlying pathway. This study aims to identify the baseline and longitudinal cytokine profiles of RVO-related macular oedema and correlating these expression profiles with higher order OCT features using a novel retinal segmentation and feature extraction platform. SUBJECTS/METHODS The IMAGINE study is a post-hoc assessment of aqueous humour cytokines with correlation to higher level analysis of imaging studies. OCT scans underwent machine learning enhanced segmentation of the internal limiting membrane (ILM), ellipsoid zone (EZ) and retinal pigment epithelium (RPE), as well as evaluating volumetric fluid metrics. Samples of aqueous humour were obtained at baseline, as well as months 4 and 9 prior to treatment. These samples were analysed for the expression of multiple cytokines. Patients were divided into Responders and Non-Responders based on OCT profiles. Additionally, patients were categorised as a Rebounder if their CST increased by 50% after initial improvement. RESULTS Twenty-six eyes were included. The OCT-based response schema identified 21 Responders (81%) and 5 Non-Responders (19%). VEGF levels directly correlated with intraretinal fluid volume and angiogenin was inversely correlated with fluid indices. Multiple cytokines, including ANGPTL4, were directly correlated with ellipsoid zone disruption. The baseline VEGF levels were significantly higher in all responders compared to Non-Responders (p = 0.02). Rebounders tended to have significantly decreased levels of angiogenin and TIMP-1 (p = 0.019, p = 0.015). CONCLUSIONS Cytokine expression was linked to specific OCT features and treatment response in RVO. Identification of an imaging phenotype that could serve as a surrogate for underlying active disease pathways could enhance treatment decision-making and precision medicine.
Collapse
Affiliation(s)
- Sruthi Arepalli
- The Tony and Leona Campane Center for Excellence in Image-Guided Surgery and Advanced Imaging Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
- Vitreoretinal Service, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Charles C Wykoff
- Retinal Consultants of Texas, Retina Consultants of America, Houston, TX, USA
| | - Joseph R Abraham
- The Tony and Leona Campane Center for Excellence in Image-Guided Surgery and Advanced Imaging Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
- Vitreoretinal Service, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Leina Lunasco
- The Tony and Leona Campane Center for Excellence in Image-Guided Surgery and Advanced Imaging Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Hannah Yu
- Retinal Consultants of Texas, Retina Consultants of America, Houston, TX, USA
| | - Ming Hu
- The Tony and Leona Campane Center for Excellence in Image-Guided Surgery and Advanced Imaging Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
- Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Sunil K Srivastava
- The Tony and Leona Campane Center for Excellence in Image-Guided Surgery and Advanced Imaging Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
- Vitreoretinal Service, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jamie L Reese
- The Tony and Leona Campane Center for Excellence in Image-Guided Surgery and Advanced Imaging Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
| | - David Brown
- Retinal Consultants of Texas, Retina Consultants of America, Houston, TX, USA
| | - Justis P Ehlers
- The Tony and Leona Campane Center for Excellence in Image-Guided Surgery and Advanced Imaging Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA.
- Vitreoretinal Service, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
9
|
Zheng L, Yang X, Fan Q, Liu B, Hu W, Cui Y. Transcriptomic profiling identifies differentially expressed genes and related pathways associated with wound healing and cuproptosis-related genes in Ganxi goats. Front Vet Sci 2023; 10:1149333. [PMID: 37313229 PMCID: PMC10259478 DOI: 10.3389/fvets.2023.1149333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 04/10/2023] [Indexed: 06/15/2023] Open
Abstract
Introduction Wound healing is very important for the maintenance of immune barrier integrity, which has attracted wide attention in past 10 years. However, no studies on the regulation of cuproptosis in wound healing have been reported. Methods In this study, the skin injury model was constructed in Gnxi goats, and the function, regulatory network and hub genes of the skin before and after the injury were comprehensively analyzed by transcriptomics. Results The results showed that there were 1,438 differentially expressed genes (DEGs), genes up-regulated by 545 and genes down-regulated by 893, which were detected by comparing day 0 and day 5 posttraumatic skin. Based on GO-KEGG analysis, DEGs that were up-regulated tended to be enriched in lysosome, phagosome, and leukocyte transendothelial migration pathways, while down-regulated DEGs were significantly enriched in adrenergic signaling in cardiomyocytes and calcium signaling pathway. There were 166 overlapped genes (DE-CUGs) between DEGs and cuproptosis-related genes, with 72 up-regulated DE-CUGs and 94 down-regulated DE-CUGs. GOKEGG analysis showed that up-regulated DE-CUGs were significantly enriched in ferroptosis, leukocyte transendothelial migration and lysosome pathways, while down-regulated DE-CUGs were significantly enriched in Apelin signaling pathway and tyrosine metabolism pathways. By constructing and analyzing of protein-protein interaction (PPI) networks of DEGs and DE-CUGs, 10 hub DEGs (ENSCHIG00000020079, PLK1, AURKA, ASPM, CENPE, KIF20A, CCNB2, KIF2C, PRC1 and KIF4A) and 10 hub DE-CUGs (MMP2, TIMP1, MMP9, MMP14, TIMP3, MMP1, EDN1, GCAT, SARDH, and DCT) were obtained, respectively. Discussion This study revealed the hub genes and important wound healing pathways in Ganxi goats, and identified the correlation between wound healing and cuproptosis for the first time, and found that MMP2, TIMP1, MMP9, and EDN1 were the core genes associated. This study enriched the transcriptome data of wound healing in Ganxi goats and expanded the research direction of cuproptosis.
Collapse
Affiliation(s)
- Lucheng Zheng
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
- College of Life Science and Resources and Environment, Yichun University, Yichun, China
| | - Xue Yang
- College of Life Science and Resources and Environment, Yichun University, Yichun, China
| | - Qingcan Fan
- College of Life Science and Resources and Environment, Yichun University, Yichun, China
| | - Ben Liu
- College of Life Science and Resources and Environment, Yichun University, Yichun, China
| | - Wei Hu
- College of Life Science and Resources and Environment, Yichun University, Yichun, China
| | - Yan Cui
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| |
Collapse
|
10
|
Chen YF, Wu KJ, Siao LR, Tsai HY. Trilinolein, a Natural Triacylglycerol, Protects Cerebral Ischemia through Inhibition of Neuronal Apoptosis and Ameliorates Intimal Hyperplasia via Attenuation of Migration and Modulation of Matrix Metalloproteinase-2 and RAS/MEK/ERK Signaling Pathway in VSMCs. Int J Mol Sci 2022; 23:12820. [PMID: 36361610 PMCID: PMC9658252 DOI: 10.3390/ijms232112820] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/02/2022] [Accepted: 10/20/2022] [Indexed: 09/11/2023] Open
Abstract
Cerebrovascular disease is one of the leading causes of disability and death worldwide, and seeking a potential treatment is essential. Trilinolein (TriL) is a natural triacylglycerol presented in several plants. The effects of TriL on cerebrovascular diseases such as cerebral ischemia and carotid stenosis have never been studied. Accordingly, we investigated the protection of TriL on cerebral ischemia/reperfusion (I/R) and vascular smooth muscle cell (VSMC) migration in vivo and in vitro. The cerebral infarction area, the intima to media area (I/M ratio), and proliferating cell nuclear antigen (PCNA)-staining of the carotid artery were measured. Platelet-derived growth factor (PDGF)-BB-stimulated A7r5 cell migration and potential mechanisms of TriL were investigated by wound healing, transwell, and Western blotting. TriL (50, 100, and 200 mg/kg, p.o.) reduced: the cerebral infarction area; neurological deficit; TUNEL-positive apoptosis; intimal hyperplasia; and PCNA-positive cells in rodents. TriL (5, 10, and 20 µM) significantly inhibited PDGF-BB-stimulated A7r5 cell migration and reduced matrix metalloproteinase-2 (MMP-2), Ras, MEK, and p-ERK protein levels in PDGF-BB-stimulated A7r5 cells. TriL is protective in models of I/R-induced brain injury, carotid artery ligation-induced intimal hyperplasia, and VSMC migration both in vivo and in vitro. TriL could be potentially efficacious in preventing cerebral ischemia and cerebrovascular diseases.
Collapse
Affiliation(s)
- Yuh-Fung Chen
- Department of Pharmacology, China Medical University, Taichung 404333, Taiwan
- Department of Pharmacy, China Medical University Hospital, Taichung 404332, Taiwan
| | - Kuo-Jen Wu
- Department of Pharmacology, China Medical University, Taichung 404333, Taiwan
| | - Lian-Ru Siao
- Department of Pharmacology, China Medical University, Taichung 404333, Taiwan
| | - Huei-Yann Tsai
- Department of Pharmacy, China Medical University Hospital, Taichung 404332, Taiwan
| |
Collapse
|
11
|
Bunnell BA, Martin EC, Matossian MD, Brock CK, Nguyen K, Collins-Burow B, Burow ME. The effect of obesity on adipose-derived stromal cells and adipose tissue and their impact on cancer. Cancer Metastasis Rev 2022; 41:549-573. [PMID: 35999486 DOI: 10.1007/s10555-022-10063-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 08/16/2022] [Indexed: 11/24/2022]
Abstract
The significant increase in the incidence of obesity represents the next global health crisis. As a result, scientific research has focused on gaining deeper insights into obesity and adipose tissue biology. As a result of the excessive accumulation of adipose tissue, obesity results from hyperplasia and hypertrophy within the adipose tissue. The functional alterations in the adipose tissue are a confounding contributing factor to many diseases, including cancer. The increased incidence and aggressiveness of several cancers, including colorectal, postmenopausal breast, endometrial, prostate, esophageal, hematological, malignant melanoma, and renal carcinomas, result from obesity as a contributing factor. The increased morbidity and mortality of obesity-associated cancers are attributable to increased hormones, adipokines, and cytokines produced by the adipose tissue. The increased adipose tissue levels observed in obese patients result in more adipose stromal/stem cells (ASCs) distributed throughout the body. ASCs have been shown to impact cancer progression in vitro and in preclinical animal models. ASCs influence tumor biology via multiple mechanisms, including the increased recruitment of ASCs to the tumor site and increased production of cytokines and growth factors by ASCs and other cells within the tumor stroma. Emerging evidence indicates that obesity induces alterations in the biological properties of ASCs, subsequently leading to enhanced tumorigenesis and metastasis of cancer cells. As the focus of this review is the interaction and impact of ASCs on cancer, the presentation is limited to preclinical data generated on cancers in which there is a demonstrated role for ASCs, such as postmenopausal breast, colorectal, prostate, ovarian, multiple myeloma, osteosarcoma, cervical, bladder, and gastrointestinal cancers. Our group has investigated the interactions between obesity and breast cancer and the mechanisms that regulate ASCs and adipocytes in these different contexts through interactions between cancer cells, immune cells, and other cell types present in the tumor microenvironment (TME) are discussed. The reciprocal and circular feedback loop between obesity and ASCs and the mechanisms by which ASCs from obese patients alter the biology of cancer cells and enhance tumorigenesis will be discussed. At present, the evidence for ASCs directly influencing human tumor growth is somewhat limited, though recent clinical studies suggest there may be some link.
Collapse
Affiliation(s)
- Bruce A Bunnell
- Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX, 76107, USA.
| | - Elizabeth C Martin
- Department of Biological and Agricultural Engineering, Louisiana State University, Baton Rouge, LA, USA
| | - Margarite D Matossian
- Department of Microbiology, Immunology and Genetics, University of Chicago, IL, Chicago, USA
| | - Courtney K Brock
- Section of Hematology and Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Khoa Nguyen
- Section of Hematology and Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Bridgette Collins-Burow
- Section of Hematology and Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Matthew E Burow
- Section of Hematology and Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
12
|
Yamamoto K, Scavenius C, Meschis MM, Gremida AME, Mogensen EH, Thøgersen IB, Bonelli S, Scilabra SD, Jensen A, Santamaria S, Ahnström J, Bou-Gharios G, Enghild JJ, Nagase H. A top-down approach to uncover the hidden ligandome of low-density lipoprotein receptor-related protein 1 in cartilage. Matrix Biol 2022; 112:190-218. [PMID: 36028175 DOI: 10.1016/j.matbio.2022.08.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/26/2022] [Accepted: 08/17/2022] [Indexed: 11/29/2022]
Abstract
The low-density lipoprotein receptor-related protein 1 (LRP1) is a cell-surface receptor ubiquitously expressed in various tissues. It plays tissue-specific roles by mediating endocytosis of a diverse range of extracellular molecules. Dysregulation of LRP1 is involved in multiple conditions including osteoarthritis (OA) but little information is available about the specific profile of direct binding partners of LRP1 (ligandome) for each tissue, which would lead to a better understanding of its role in disease states. Here, we investigated adult articular cartilage where impaired LRP1-mediated endocytosis leads to tissue destruction. We used a top-down approach involving proteomic analysis of the LRP1 interactome in human chondrocytes, direct binding assays using purified LRP1 and ligand candidates, and validation in LRP1-deficient fibroblasts and human chondrocytes, as well as a novel Lrp1 conditional knockout (KO) mouse model. We found that inhibition of LRP1 and ligand interaction results in cell death, alteration of the entire secretome and transcriptional modulations in human chondrocytes. We identified a chondrocyte-specific LRP1 ligandome consisting of more than 50 novel ligand candidates. Surprisingly, 23 previously reported LRP1 ligands were not regulated by LRP1-mediated endocytosis in human chondrocytes. We confirmed direct LRP1 binding of HGFAC, HMGB1, HMGB2, CEMIP, SLIT2, ADAMTS1, TSG6, IGFBP7, SPARC and LIF, correlation between their affinity for LRP1 and the rate of endocytosis, and some of their intracellular localization. Moreover, a conditional LRP1 KO mouse model demonstrated a critical role of LRP1 in regulating the high-affinity ligands in cartilage in vivo. This systematic approach revealed the specificity and the extent of the chondrocyte LRP1 ligandome and identified potential novel therapeutic targets for OA.
Collapse
Affiliation(s)
- Kazuhiro Yamamoto
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, United Kingdom.
| | - Carsten Scavenius
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| | - Maria M Meschis
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, United Kingdom
| | - Abdulrahman M E Gremida
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, United Kingdom
| | - Emilie H Mogensen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| | - Ida B Thøgersen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| | - Simone Bonelli
- Fondazione RiMED - ISMETT via Ernesto Tricomi 5, 90127 Palermo, Italy
| | - Simone D Scilabra
- Fondazione RiMED - ISMETT via Ernesto Tricomi 5, 90127 Palermo, Italy
| | - Anders Jensen
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, United Kingdom
| | - Salvatore Santamaria
- Department of Immunology and Inflammation, Imperial College London, Du Cane Road, W12 0NN, London, United Kingdom
| | - Josefin Ahnström
- Department of Immunology and Inflammation, Imperial College London, Du Cane Road, W12 0NN, London, United Kingdom
| | - George Bou-Gharios
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, United Kingdom
| | - Jan J Enghild
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| | - Hideaki Nagase
- Kennedy Institute of Rheumatology, University of Oxford, Headington, Oxford OX3 7FY, United Kingdom
| |
Collapse
|
13
|
Jt S, M H, Wam B, Ac B, Sa N. Adenoviral vectors for cardiovascular gene therapy applications: a clinical and industry perspective. J Mol Med (Berl) 2022; 100:875-901. [PMID: 35606652 PMCID: PMC9126699 DOI: 10.1007/s00109-022-02208-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 04/29/2022] [Accepted: 05/10/2022] [Indexed: 11/29/2022]
Abstract
Abstract Despite the development of novel pharmacological treatments, cardiovascular disease morbidity and mortality remain high indicating an unmet clinical need. Viral gene therapy enables targeted delivery of therapeutic transgenes and represents an attractive platform for tackling acquired and inherited cardiovascular diseases in the future. Current cardiovascular gene therapy trials in humans mainly focus on improving cardiac angiogenesis and function. Encouragingly, local delivery of therapeutic transgenes utilising first-generation human adenovirus serotype (HAd)-5 is safe in the short term and has shown some efficacy in drug refractory angina pectoris and heart failure with reduced ejection fraction. Despite this success, systemic delivery of therapeutic HAd-5 vectors targeting cardiovascular tissues and internal organs is limited by negligible gene transfer to target cells, elimination by the immune system, liver sequestration, off-target effects, and episomal degradation. To circumvent these barriers, cardiovascular gene therapy research has focused on determining the safety and efficacy of rare alternative serotypes and/or genetically engineered adenoviral capsid protein-modified vectors following local or systemic delivery. Pre-clinical studies have identified several vectors including HAd-11, HAd-35, and HAd-20–42-42 as promising platforms for local and systemic targeting of vascular endothelial and smooth muscle cells. In the past, clinical gene therapy trials were often restricted by limited scale-up capabilities of gene therapy medicinal products (GTMPs) and lack of regulatory guidance. However, significant improvement of industrial GTMP scale-up and purification, development of novel producer cell lines, and issuing of GTMP regulatory guidance by national regulatory health agencies have addressed many of these challenges, creating a more robust framework for future adenoviral-based cardiovascular gene therapy. In addition, this has enabled the mass roll out of adenovirus vector-based COVID-19 vaccines. Key messages
First-generation HAd-5 vectors are widely used in cardiovascular gene therapy. HAd-5-based gene therapy was shown to lead to cardiac angiogenesis and improved function. Novel HAd vectors may represent promising transgene carriers for systemic delivery. Novel methods allow industrial scale-up of rare/genetically altered Ad serotypes. National regulatory health agencies have issued guidance on GMP for GTMPs.
Collapse
Affiliation(s)
- Schwartze Jt
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK.
| | - Havenga M
- Batavia Biosciences B.V., Bioscience Park Leiden, Zernikedreef 16, 2333, CL, Leiden, The Netherlands
| | - Bakker Wam
- Batavia Biosciences B.V., Bioscience Park Leiden, Zernikedreef 16, 2333, CL, Leiden, The Netherlands
| | - Bradshaw Ac
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Nicklin Sa
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
14
|
Choi D, Gonzalez Z, Ho SY, Bermudez A, Lin NY. Cell-cell adhesion impacts epithelia response to substrate stiffness: Morphology and gene expression. Biophys J 2022; 121:336-346. [PMID: 34864047 PMCID: PMC8790207 DOI: 10.1016/j.bpj.2021.11.2887] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 09/04/2021] [Accepted: 11/29/2021] [Indexed: 01/21/2023] Open
Abstract
Monolayer epithelial cells interact constantly with the substrate they reside on and their surrounding neighbors. As such, the properties of epithelial cells are profoundly governed by the mechanical and molecular cues that arise from both the substrate and contiguous cell neighbors. Although both cell-substrate and cell-cell interactions have been studied individually, these results are difficult to apply to native confluent epithelia, in which both jointly regulate the cell phenotype. Specifically, it remains poorly understood about the intertwined contributions from intercellular adhesion and substrate stiffness on cell morphology and gene expression, two essential microenvironment properties. Here, by adjusting the substrate modulus and altering the intercellular adhesion within confluent kidney epithelia, we found that cell-substrate and cell-cell interactions can mask each other's influence. For example, we found that epithelial cells exhibit an elongated morphological phenotype only when the substrate modulus and intercellular adhesions are both reduced, whereas their motility can be upregulated by either reduction. These results illustrate that combinatorial changes of the physical microenvironment are required to alter cell morphology and gene expression.
Collapse
Affiliation(s)
- David Choi
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, California,Corresponding author
| | - Zachary Gonzalez
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, California,Department of Physics and Astronomy, University of California, Los Angeles, California
| | - Sum Yat Ho
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, California,Department of Chemistry and Biochemistry, University of California, Los Angeles, California
| | - Alexandra Bermudez
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, California,Department of Bioengineering, University of California, Los Angeles, California
| | - Neil Y.C. Lin
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, California,Department of Bioengineering, University of California, Los Angeles, California,Institute for Quantitative and Computational Biosciences, University of California, Los Angeles
| |
Collapse
|
15
|
Russell JJ, Grisanti LA, Brown SM, Bailey CA, Bender SB, Chandrasekar B. Reversion inducing cysteine rich protein with Kazal motifs and cardiovascular diseases: The RECKlessness of adverse remodeling. Cell Signal 2021; 83:109993. [PMID: 33781845 DOI: 10.1016/j.cellsig.2021.109993] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/19/2022]
Abstract
The Reversion Inducing Cysteine Rich Protein With Kazal Motifs (RECK) is a glycosylphosphatidylinositol (GPI) anchored membrane-bound regulator of matrix metalloproteinases (MMPs). It is expressed throughout the body and plays a role in extracellular matrix (ECM) homeostasis and inflammation. In initial studies, RECK expression was found to be downregulated in various invasive cancers and associated with poor prognostic outcome. Restoring RECK, however, has been shown to reverse the metastatic phenotype. Downregulation of RECK expression is also reported in non-malignant diseases, such as periodontal disease, renal fibrosis, and myocardial fibrosis. As such, RECK induction has therapeutic potential in several chronic diseases. Mechanistically, RECK negatively regulates various matrixins involved in cell migration, proliferation, and adverse remodeling by targeting the expression and/or activation of multiple MMPs, A Disintegrin And Metalloproteinase Domain-Containing Proteins (ADAMs), and A Disintegrin And Metalloproteinase With Thrombospondin Motifs (ADAMTS). Outside of its role in remodeling, RECK has also been reported to exert anti-inflammatory effects. In cardiac diseases, for example, it has been shown to counteract several downstream effectors of Angiotensin II (Ang-II) that play a role in adverse cardiac and vascular remodeling, such as Interleukin-6 (IL-6)/IL-6 receptor (IL-6R)/glycoprotein 130 (IL-6 signal transducer) signaling and Epidermal Growth Factor Receptor (EGFR) transactivation. This review article focuses on the current understanding of the multifunctional effects of RECK and how its downregulation may contribute to adverse cardiovascular remodeling.
Collapse
Affiliation(s)
- Jacob J Russell
- Biomedical Sciences, University of Missouri, Columbia, MO, United States of America; Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States of America.
| | - Laurel A Grisanti
- Biomedical Sciences, University of Missouri, Columbia, MO, United States of America.
| | - Scott M Brown
- Biomedical Sciences, University of Missouri, Columbia, MO, United States of America; Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States of America.
| | - Chastidy A Bailey
- Biomedical Sciences, University of Missouri, Columbia, MO, United States of America; Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States of America.
| | - Shawn B Bender
- Biomedical Sciences, University of Missouri, Columbia, MO, United States of America; Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States of America; Dalton Cardiovascular Center, University of Missouri, Columbia, MO, United States of America.
| | - B Chandrasekar
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States of America; Medicine, University of Missouri School of Medicine, Columbia, MO, United States of America; Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, United States of America; Dalton Cardiovascular Center, University of Missouri, Columbia, MO, United States of America.
| |
Collapse
|
16
|
Fontes MMDS, Costa TC, Lopes MM, Souza RO, Carneiro LS, Paulino PVR, Chizzotti ML, Silva FF, Serão NVL, Duarte MDS. Intramuscular collagen characteristics and expression of related genes in skeletal muscle of cull cows receiving a high-energy diet. Meat Sci 2021; 177:108495. [PMID: 33756247 DOI: 10.1016/j.meatsci.2021.108495] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 03/01/2021] [Accepted: 03/08/2021] [Indexed: 11/28/2022]
Abstract
We aimed to investigate differences in the synthesis and metabolism of intramuscular collagen in the Longissimus thoracis (LT) muscle between heifers and cull-cows fed high-energy diet. Ten cull-cows, (74.9 ± 3.2 months age, weighing 536 ± 14.55 kg) and ten heifers (18.4 ± 3.2 months age, weighting 310.5 ± 14.5 kg) were fed with high-energy diets for 150 days. The total collagen content did not differ between treatments. Greater collagen solubility was observed in heifers than cull-cows, although no differences in lysyl oxidase activity were observed between treatments. No differences were observed for mRNA expression of CO1A1, MMP2, MMP9 and TIMP2 between treatments. However, cull-cows presented greater mRNA expression of COL3A1, TIMP1 and TIMP3 than heifers. Our data give no indication that feeding a high-energy diet to cull-cows decreases the concentration of intramuscular collagen in the LT muscle or increases its solubility in respect to the collagen solubility in LT muscles from heifers on the same diet.
Collapse
Affiliation(s)
- Marta Maria Dos Santos Fontes
- Department of Animal Science, Universidade Federal de Vicosa, 36570-900 Viçosa, MG, Brazil; Muscle Biology and Nutrigenomics Laboratory, Universidade Federal de Viçosa, 36570-900, Viçosa, MG, Brazil
| | - Thaís Correia Costa
- Department of Animal Science, Universidade Federal de Vicosa, 36570-900 Viçosa, MG, Brazil; Muscle Biology and Nutrigenomics Laboratory, Universidade Federal de Viçosa, 36570-900, Viçosa, MG, Brazil
| | - Mariana Mescouto Lopes
- Department of Animal Science, Universidade Federal de Vicosa, 36570-900 Viçosa, MG, Brazil; Muscle Biology and Nutrigenomics Laboratory, Universidade Federal de Viçosa, 36570-900, Viçosa, MG, Brazil
| | - Ranyeri Oliveira Souza
- Department of Animal Science, Universidade Federal de Vicosa, 36570-900 Viçosa, MG, Brazil; Muscle Biology and Nutrigenomics Laboratory, Universidade Federal de Viçosa, 36570-900, Viçosa, MG, Brazil
| | - Lorena Silva Carneiro
- Department of Animal Science, Universidade Federal de Vicosa, 36570-900 Viçosa, MG, Brazil; Muscle Biology and Nutrigenomics Laboratory, Universidade Federal de Viçosa, 36570-900, Viçosa, MG, Brazil
| | | | - Mário Luiz Chizzotti
- Department of Animal Science, Universidade Federal de Vicosa, 36570-900 Viçosa, MG, Brazil
| | - Fabyano Fonseca Silva
- Department of Animal Science, Universidade Federal de Vicosa, 36570-900 Viçosa, MG, Brazil
| | | | - Marcio de Souza Duarte
- Department of Animal Science, Universidade Federal de Vicosa, 36570-900 Viçosa, MG, Brazil; Muscle Biology and Nutrigenomics Laboratory, Universidade Federal de Viçosa, 36570-900, Viçosa, MG, Brazil.
| |
Collapse
|
17
|
Changes in Gene and Protein Expression of Metalloproteinase-2 and -9 and Their Inhibitors TIMP2 and TIMP3 in Different Parts of Fluoride-Exposed Rat Brain. Int J Mol Sci 2020; 22:ijms22010391. [PMID: 33396569 PMCID: PMC7796218 DOI: 10.3390/ijms22010391] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/22/2020] [Accepted: 12/29/2020] [Indexed: 02/06/2023] Open
Abstract
Fluoride (F) exposure decreases brain receptor activity and neurotransmitter production. A recent study has shown that chronic fluoride exposure during childhood can affect cognitive function and decrease intelligence quotient, but the mechanism of this phenomenon is still incomplete. Extracellular matrix (ECM) and its enzymes are one of the key players of neuroplasticity which is essential for cognitive function development. Changes in the structure and the functioning of synapses are caused, among others, by ECM enzymes. These enzymes, especially matrix metalloproteinases (MMPs) and their tissue inhibitors (TIMPs), are involved in both physiological processes, such as learning or memory, and pathological processes like glia scare formation, brain tissue regeneration, brain-blood barrier damage and inflammation. Therefore, in this study, we examined the changes in gene and protein expression of MMP2, MMP9, TIMP2 and TIMP3 in the prefrontal cortex, hippocampus, striatum and cerebellum of rats (Wistar) exposed to relatively low F doses (50 mg/L in drinking water) during the pre- and neonatal period. We found that exposure to F during pre- and postnatal period causes a change in the mRNA and protein level of MMP2, MMP9, TIMP2 and TIMP3 in the prefrontal cortex, striatum, hippocampus and cerebellum. These changes may be associated with many disorders that are observed during F intoxication. MMPs/TIMPs imbalance may contribute to cognitive impairments. Moreover, our results suggest that a chronic inflammatory process and blood-brain barrier (BBB) damage occur in rats’ brains exposed to F.
Collapse
|
18
|
Ding L, Hang C, Cheng S, Jia L, Mou L, Tang L, Zhang C, Xie Y, Zheng W, Zhang Y, Jiang X. A Soft, Conductive External Stent Inhibits Intimal Hyperplasia in Vein Grafts by Electroporation and Mechanical Restriction. ACS NANO 2020; 14:16770-16780. [PMID: 33030886 DOI: 10.1021/acsnano.0c04827] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Intimal hyperplasia (IH) in vein grafts (VGs) is a major issue in coronary artery bypass grafting (CABG) surgery. Although external stents can attenuate IH of VGs to some extent, none of the existing external stents have shown satisfactory clinical outcomes. Here we develop a flexible, biodegradable, and conductive external metal-polymer conductor stent (MPCS) that can electroporate the vessel wall and produce a protein that prevents IH. We designed the plasmid DNA encoding the tissue inhibitor of metalloproteinases-3 (TIMP-3) and lyophilized it on the inner surface of the MPCS to deliver into the adventitia and the middle layer of VGs for gene therapy. Coupled with its continuous mechanical support to prevent dilation after implanting, the MPCS can inhibit the IH of VGs significantly in the rabbit model. This proof-of-concept demonstration may aid the development of other implantable bioelectronics for electroporation gene therapy.
Collapse
Affiliation(s)
- Li Ding
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- Department of Biomedical Engineering and Shenzhen Bay Laboratory, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen, Guangdong 518055, China
- National Center for NanoScience and Technology and University of Chinese Academy of Sciences, Beijing 100190, China
| | - Chen Hang
- Department of Biomedical Engineering and Shenzhen Bay Laboratory, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen, Guangdong 518055, China
- National Center for NanoScience and Technology and University of Chinese Academy of Sciences, Beijing 100190, China
| | - Shiyu Cheng
- National Center for NanoScience and Technology and University of Chinese Academy of Sciences, Beijing 100190, China
| | - Liujun Jia
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Lei Mou
- Department of Biomedical Engineering and Shenzhen Bay Laboratory, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen, Guangdong 518055, China
- National Center for NanoScience and Technology and University of Chinese Academy of Sciences, Beijing 100190, China
| | - Lixue Tang
- Department of Biomedical Engineering and Shenzhen Bay Laboratory, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen, Guangdong 518055, China
- National Center for NanoScience and Technology and University of Chinese Academy of Sciences, Beijing 100190, China
| | - Chunliang Zhang
- National Center for NanoScience and Technology and University of Chinese Academy of Sciences, Beijing 100190, China
| | - Yangzhouyun Xie
- Department of Biomedical Engineering and Shenzhen Bay Laboratory, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen, Guangdong 518055, China
- National Center for NanoScience and Technology and University of Chinese Academy of Sciences, Beijing 100190, China
| | - Wenfu Zheng
- National Center for NanoScience and Technology and University of Chinese Academy of Sciences, Beijing 100190, China
| | - Yan Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Xingyu Jiang
- Department of Biomedical Engineering and Shenzhen Bay Laboratory, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen, Guangdong 518055, China
- National Center for NanoScience and Technology and University of Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
19
|
Escalona RM, Bilandzic M, Western P, Kadife E, Kannourakis G, Findlay JK, Ahmed N. TIMP-2 regulates proliferation, invasion and STAT3-mediated cancer stem cell-dependent chemoresistance in ovarian cancer cells. BMC Cancer 2020; 20:960. [PMID: 33023532 PMCID: PMC7542139 DOI: 10.1186/s12885-020-07274-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 08/09/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The metzincin family of metalloproteinases and the tissue inhibitors of metalloproteinases (TIMPs) are essential proteins required for biological processes during cancer progression. This study aimed to determine the role of TIMP-2 in ovarian cancer progression and chemoresistance by reducing TIMP-2 expression in vitro in Fallopian tube secretory epithelial (FT282) and ovarian cancer (JHOS2 and OVCAR4) cell lines. METHODS FT282, JHOS2 and OVCAR4 cells were transiently transfected with either single or pooled TIMP-2 siRNAs. The expression of different genes after TIMP-2 knock down (T2-KD) or in response to chemotherapy was determined at the mRNA level by quantitative real time PCR (qRT-PCR) and at the protein level by immunofluorescence. Sensitivity of the cell lines in response to chemotherapy after TIMP-2 knock down was investigated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and 5-Ethynyl-2'-deoxyuridine (EdU) assays. Cell invasion in response to TIMP-2 knockdown was determined by xCELLigence. RESULTS Sixty to 90 % knock down of TIMP-2 expression was confirmed in FT282, OVCAR4 and JHOS2 cell lines at the mRNA and protein levels. TIMP-2 knock down did not change the mRNA expression of TIMP-1 or TIMP-3. However, a significant downregulation of MMP-2 in T2-KD cells occurred at both the protein and activation levels, compared to Control (Cont; scrambled siRNA) and Parental cells (P, transfection reagent only). In contrast, membrane bound MT1-MMP protein levels were significantly upregulated in T2-KD compared to Cont and P cells. T2-KD cells exhibited enhanced proliferation and increased sensitivity to cisplatin and paclitaxel treatments. Enhanced invasion was observed in the T2-KD-JOSH2 and OVCAR4 cells but not in T2-KD-FT282 cells. Treatment with cisplatin or paclitaxel significantly elevated the expression of TIMP-2 in Cont cells but not in T2-KD cells, consistent with significantly elevated expression of chemoresistance and CSC markers and activation of STAT3. Furthermore, a potent inhibitor of STAT3 activation, Momelotinib, suppressed chemotherapy-induced activation of P-STAT3 in OVCAR4 cells with concomitant reductions in the expression of chemoresistance genes and CSC markers. CONCLUSIONS The above results suggest that TIMP-2 may have a novel role in ovarian cancer proliferation, invasion and chemoresistance.
Collapse
Affiliation(s)
- Ruth M Escalona
- Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, VIC, 3050, Australia.,Centre for Reproductive Health, Hudson Institute of Medical Research, and the Department of Molecular and Translational Science, Monash University, Melbourne, VIC, 3168, Australia.,Fiona Elsey Cancer Research Institute, Ballarat, 3353, Australia
| | - Maree Bilandzic
- Centre for Cancer Research, Hudson Institute of Medical Research, and the Department of Molecular and Translational Science, Monash University, Melbourne, VIC, 3168, Australia
| | - Patrick Western
- Centre for Reproductive Health, Hudson Institute of Medical Research, and the Department of Molecular and Translational Science, Monash University, Melbourne, VIC, 3168, Australia
| | - Elif Kadife
- Fiona Elsey Cancer Research Institute, Ballarat, 3353, Australia
| | - George Kannourakis
- Fiona Elsey Cancer Research Institute, Ballarat, 3353, Australia.,Federation University Australia, Vic, Ballarat, 3010, Australia
| | - Jock K Findlay
- Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, VIC, 3050, Australia.,Centre for Reproductive Health, Hudson Institute of Medical Research, and the Department of Molecular and Translational Science, Monash University, Melbourne, VIC, 3168, Australia
| | - Nuzhat Ahmed
- Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, VIC, 3050, Australia. .,Centre for Reproductive Health, Hudson Institute of Medical Research, and the Department of Molecular and Translational Science, Monash University, Melbourne, VIC, 3168, Australia. .,Fiona Elsey Cancer Research Institute, Ballarat, 3353, Australia. .,Federation University Australia, Vic, Ballarat, 3010, Australia.
| |
Collapse
|
20
|
Xi Y, Huang H, Zhao Z, Ma J, Chen Y. Tissue inhibitor of metalloproteinase 1 suppresses growth and differentiation of osteoblasts and differentiation of osteoclasts by targeting the AKT pathway. Exp Cell Res 2020; 389:111930. [PMID: 32113948 DOI: 10.1016/j.yexcr.2020.111930] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 02/17/2020] [Accepted: 02/26/2020] [Indexed: 01/01/2023]
Abstract
Tissue inhibitor of metalloproteinase 1 (TIMP1) has various biological activities including the regulation of cell growth and differentiation. However, its role in bone homeostasis and remodeling remains poorly understood. In this study, we investigate the effects of TIMP1 on osteoblast and osteoclast activity at both cellular and molecular level using siRNA-mediated knockdown technique. Our results show that knockdown of TIMP1 stimulates proliferation and survival, but decreases apoptosis in osteoblastic MC3T3-E1 cells, suggesting that TIMP1 inhibits cell growth. TIMP1 also dampens differentiation of committed osteoblasts, as well as osteoblastogenesis of bone marrow-derived mesenchymal stem cells (BMSCs). We further show that the modulation of TIMP1 on osteoblast activity is independent of its MMP inhibition. Importantly, we uncover that TIMP1 suppresses osteoblast growth and differentiation by targeting the AKT pathway, and this is associated with TIMP1-mediated induction of PTEN via its binding to the cell surface receptor CD44. Therefore, our results highlight a novel TIMP1/CD44/PTEN/AKT signaling nexus that functions as a suppressor of osteoblast activity. Moreover, we show that TIMP1 also inhibits osteoclast differentiation in osteoclast precursor RAW 264.7 cells by targeting the AKT. In conclusion, our results demonstrate that TIMP1 can act as a suppressor of growth and differentiation of osteoblasts and differentiation of osteoclasts through the negative regulation of the AKT pathway. We propose that TIMP1 may serve as a potential target for low bone mass-related skeletal diseases, such as osteoporosis.
Collapse
Affiliation(s)
- Yongming Xi
- Department of Orthopaedics, Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Hui Huang
- Department of Anesthesia, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zheng Zhao
- Department of Orthopaedics, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jinfeng Ma
- Department of Orthopaedics, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yan Chen
- Princess Margaret Cancer Center, University Health Network, Toronto, Canada.
| |
Collapse
|
21
|
Rai GP, Baird SK. Tissue inhibitor of matrix metalloproteinase-3 has both anti-metastatic and anti-tumourigenic properties. Clin Exp Metastasis 2020; 37:69-76. [PMID: 31894441 DOI: 10.1007/s10585-019-10017-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 12/23/2019] [Indexed: 02/06/2023]
Abstract
TIMP-3 is one of four tissue inhibitors of matrix metalloproteinases, the endogenous inhibitors of the matrix metalloproteinase enzymes. These enzymes have an important role in metastasis, in the invasion of cancer cells through the basement membrane and extracellular matrix. TIMP-1, -2 and -4 both promote and inhibit tumour development, in a context-dependent manner, however TIMP-3 is consistently anti-tumourigenic. TIMP-3 is also the only insoluble member of the family, being either bound to the extracellular matrix or the low density lipoprotein-related protein-1, through which it can be endocytosed. Levels of TIMP-3 have also been shown to be regulated by micro RNAs and promoter hypermethylation, resulting in frequent silencing in many tumour types, to the extent that its expression has been suggested as a prognostic marker in some tumours, being associated with lower levels of metastasis, or better response to treatment. TIMP-3 has been shown to have anti-metastatic effects, both through inhibition of matrix metalloproteinases and ADAM family members and downregulation of angiogenesis. This occurs via interactions with receptors including VEGF, via modulation of signaling pathways and due to protease inhibition. TIMP-3 has also been shown to reduce tumour growth rate, most often by inducing apoptosis by stabilisation of death receptors. A number of successful mechanisms of delivery of TIMP-3 to tumour or inflammatory sites have been investigated in vitro or in animal studies. It may therefore be worthwhile further exploring the use of TIMP-3 as a potential anti-metastatic or anti-tumorigenic therapy for many tumour types.
Collapse
Affiliation(s)
- Geetanjali P Rai
- Department of Pharmacology and Toxicology, School of Biomedical Sciences, University of Otago, PO Box 56, Dunedin, 9054, New Zealand
| | - Sarah K Baird
- Department of Pharmacology and Toxicology, School of Biomedical Sciences, University of Otago, PO Box 56, Dunedin, 9054, New Zealand.
| |
Collapse
|
22
|
Kostova E, Shubeska Stratrova S. Prognostic meaning of tissue inhibitors of matrix metalloproteinases TIMP-1 and TIMP-2 in patients with colorectal cancer. MAKEDONSKO FARMACEVTSKI BILTEN 2020. [DOI: 10.33320/maced.pharm.bull.2020.66.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The aim of this study was to analyze TIMP-1 and TIMP-2 serum levels in patients with colorectal cancer (CRC) and to correlate the results with the pathological stage of the disease and outcome in order to evaluate the role of TIMP-1 and TIMP-2 serum levels as prognostic markers.
The investigation has been made on 82 patients with operable CRC without distant metastases, who had undergone blood tests in order to determine the TIMP-1 and TIMP-2 serum levels in the following points of time: preoperatively, as well as 3, 6, 9 and 12 months postoperatively.
Significant differences were found between serum levels of TIMP-1 and TIMP-2 obtained preoperatively and postoperatively, as well as significant association of serum TIMP-1 levels obtained preoperatively in CRC patients in stage I and III, in the 3th and in the 6th month (p<0.001) postoperatively as defined points of time with the outcome of CRC patients. Serum TIMP-2 levels obtained preoperatively was significantly associated with the outcome of the CRC patients. Analysis of the obtained TIMP-1 and TIMP-2 serum levels in CRC patients showed statistically significant differences with: disease progression, occurrence of liver metastasis, prior to and post chemotherapy treatment.
The results derived a conclusion that the serum levels of TIMP-1 and TIMP-2 could be indicators for occurrence and progression of CRC, as well as valuable and useful markers for following the effects of chemotherapy treatment.
Keywords: colorectal cancer, TIMP-1, TIMP-2, prognosis
Collapse
Affiliation(s)
- Elena Kostova
- Department of Preclinical and Clinical Pharmacology and Toxicology, Medical Faculty, Ss. Cyril and Methodius University, 50 Divizija 6,1000 Skopje, Republic of North Macedonia
| | - Slavica Shubeska Stratrova
- University Clinic of Endocrinology, Diabetes and Metabolic disorders, Medical Faculty, Ss. Cyril and Methodius University, Mother Theresa 17, 1000 Skopje, Republic of North Macedonia
| |
Collapse
|
23
|
Qi JH, Bell B, Singh R, Batoki J, Wolk A, Cutler A, Prayson N, Ali M, Stoehr H, Anand-Apte B. Sorsby Fundus Dystrophy Mutation in Tissue Inhibitor of Metalloproteinase 3 (TIMP3) promotes Choroidal Neovascularization via a Fibroblast Growth Factor-dependent Mechanism. Sci Rep 2019; 9:17429. [PMID: 31757977 PMCID: PMC6874529 DOI: 10.1038/s41598-019-53433-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 11/01/2019] [Indexed: 12/11/2022] Open
Abstract
Choroidal neovascularization (CNV) leads to loss of vision in patients with Sorsby Fundus Dystrophy (SFD), an inherited, macular degenerative disorder, caused by mutations in the Tissue Inhibitor of Metalloproteinase-3 (TIMP3) gene. SFD closely resembles age-related macular degeneration (AMD), which is the leading cause of blindness in the elderly population of the Western hemisphere. Variants in TIMP3 gene have recently been identified in patients with AMD. A majority of patients with AMD also lose vision as a consequence of choroidal neovascularization (CNV). Thus, understanding the molecular mechanisms that contribute to CNV as a consequence of TIMP-3 mutations will provide insight into the pathophysiology in SFD and likely the neovascular component of the more commonly seen AMD. While the role of VEGF in CNV has been studied extensively, it is becoming increasingly clear that other factors likely play a significant role. The objective of this study was to test the hypothesis that basic Fibroblast Growth Factor (bFGF) regulates SFD-related CNV. In this study we demonstrate that mice expressing mutant TIMP3 (Timp3S179C/S179C) showed reduced MMP inhibitory activity with an increase in MMP2 activity and bFGF levels, as well as accentuated CNV leakage when subjected to laser injury. S179C mutant-TIMP3 in retinal pigment epithelial (RPE) cells showed increased secretion of bFGF and conditioned medium from these cells induced increased angiogenesis in endothelial cells. These studies suggest that S179C-TIMP3 may promote angiogenesis and CNV via a FGFR-1-dependent pathway by increasing bFGF release and activity.
Collapse
Affiliation(s)
- Jian Hua Qi
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Brent Bell
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Rupesh Singh
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Julia Batoki
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Alyson Wolk
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Alecia Cutler
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Nicholas Prayson
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Mariya Ali
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Heidi Stoehr
- Institute of Human Genetics, University of Regensburg, Regensburg, Germany
| | - Bela Anand-Apte
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, USA.
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Cleveland Clinic Foundation, Cleveland, OH, USA.
| |
Collapse
|
24
|
Sabol RA, Giacomelli P, Beighley A, Bunnell BA. Adipose Stem Cells and Cancer: Concise Review. Stem Cells 2019; 37:1261-1266. [DOI: 10.1002/stem.3050] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/11/2019] [Accepted: 06/04/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Rachel A. Sabol
- Center for Stem Cell Research; Tulane University School of Medicine; New Orleans Louisiana USA
| | - Paulina Giacomelli
- Center for Stem Cell Research; Tulane University School of Medicine; New Orleans Louisiana USA
| | - Adam Beighley
- Center for Stem Cell Research; Tulane University School of Medicine; New Orleans Louisiana USA
| | - Bruce A. Bunnell
- Center for Stem Cell Research; Tulane University School of Medicine; New Orleans Louisiana USA
- Department of Pharmacology; Tulane University; New Orleans Louisiana USA
- Division of Regenerative Medicine; Tulane National Primate Research Center; Covington Louisiana USA
| |
Collapse
|
25
|
Chen J, Hu Y, Chen L, Liu W, Mu Y, Liu P. The effect and mechanisms of Fuzheng Huayu formula against chronic liver diseases. Biomed Pharmacother 2019; 114:108846. [PMID: 30965233 DOI: 10.1016/j.biopha.2019.108846] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 03/18/2019] [Accepted: 04/01/2019] [Indexed: 12/12/2022] Open
Abstract
Fuzheng Huayu formula (FZHY) is a traditional Chinese medicine formula composed of Radix Salvia Miltiorrhizae, Cordyceps, Semen Persicae, Gynostemma Pentaphyllammak, Pollen Pini, Fructus Schisandrae Chinensis that is used to treat chronic liver diseases in China. We here describe the pharmacological actions of FZHY in this review. We also describe and provide updates on recent advances in the medical applications and the basic research of mechanisms of FZHY formula. FZHY has been shown to have no serious adverse reactions and exerts antifibrotic effects through targeting more than one molecule.
Collapse
Affiliation(s)
- Jiamei Chen
- Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai Key Laboratory of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yonghong Hu
- Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai Key Laboratory of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Long Chen
- Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai Key Laboratory of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Wei Liu
- Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai Key Laboratory of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yongping Mu
- Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai Key Laboratory of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ping Liu
- Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai Key Laboratory of Traditional Chinese Medicine, Shanghai, 201203, China; E-Institute of Shanghai Municipal Education Commission Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
26
|
Vanichkitrungruang S, Chuang CY, Hawkins CL, Hammer A, Hoefler G, Malle E, Davies MJ. Oxidation of human plasma fibronectin by inflammatory oxidants perturbs endothelial cell function. Free Radic Biol Med 2019; 136:118-134. [PMID: 30959171 DOI: 10.1016/j.freeradbiomed.2019.04.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 02/18/2019] [Accepted: 04/01/2019] [Indexed: 01/08/2023]
Abstract
Dysfunction of endothelial cells of the artery wall is an early event in cardiovascular disease and atherosclerosis. The cause(s) of this dysfunction are unresolved, but accumulating evidence suggests that oxidants arising from chronic low-grade inflammation are contributory agents, with increasing data implicating myeloperoxidase (MPO, released by activated leukocytes), and the oxidants it generates (e.g. HOCl and HOSCN). As these are formed extracellularly and react rapidly with proteins, we hypothesized that MPO-mediated damage to the matrix glycoprotein fibronectin (FN) would modulate FN structure and function, and its interactions with human coronary artery endothelial cells (HCAEC). Exposure of human plasma FN to HOCl resulted in modifications to FN and its functional epitopes. A dose-dependent loss of methionine and tryptophan residues, together with increasing concentrations of methionine sulfoxide, and modification of the cell-binding fragment (CBF) and heparin-binding fragment (HBF) domains was detected with HOCl, but not HOSCN. FN modification resulted in a loss of HCAEC adhesion, impaired cell spreading and reduced cell proliferation. Exposure to HCAEC to HOCl-treated FN altered the expression of HCAEC genes associated with extracellular matrix (ECM) synthesis and adhesion. Modifications were detected on HCAEC-derived ECM pre-treated with HOCl, but not HOSCN, with a loss of antibody recognition of the CBF, HBF and extra-domain A. Co-localization of epitopes arising from MPO-generated HOCl and cell-derived FN was detected in human atherosclerotic lesions. Damage was also detected on FN extracted from lesions. These data support the hypothesis that HOCl, but not HOSCN, targets and modifies FN resulting in arterial wall endothelial cell dysfunction.
Collapse
Affiliation(s)
- Siriluck Vanichkitrungruang
- The Heart Research Institute, Newtown, NSW, Australia; Faculty of Medicine, The University of Sydney, NSW, Australia; Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Denmark
| | - Christine Y Chuang
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Denmark
| | - Clare L Hawkins
- The Heart Research Institute, Newtown, NSW, Australia; Faculty of Medicine, The University of Sydney, NSW, Australia; Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Denmark
| | - Astrid Hammer
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Gerald Hoefler
- Institute of Pathology, Diagnostic & Research Center for Molecular BioMedicine, Medical University of Graz, Graz, Austria
| | - Ernst Malle
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Michael J Davies
- The Heart Research Institute, Newtown, NSW, Australia; Faculty of Medicine, The University of Sydney, NSW, Australia; Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Denmark.
| |
Collapse
|
27
|
Zajkowska M, Gacuta E, Kozłowska S, Lubowicka E, Głażewska EK, Chrostek L, Szmitkowski M, Pawłowski P, Zbucka-Krętowska M, Ławicki S. Diagnostic power of VEGF, MMP-9 and TIMP-1 in patients with breast cancer. A multivariate statistical analysis with ROC curve. Adv Med Sci 2019; 64:1-8. [PMID: 30227310 DOI: 10.1016/j.advms.2018.07.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 04/13/2018] [Accepted: 07/31/2018] [Indexed: 02/07/2023]
Abstract
PURPOSE Vascular endothelial growth factor is an important factor in promoting angiogenesis in malignant processes, matrix metalloproteinase-9 in the degradation of extracellular matrix, which enhances metastasis, and tissue inhibitor of metalloproteinase-1 is its inhibitor. The aim of this study was to investigate the diagnostic power of these parameters in comparison to CA15-3 in breast cancer patients and in relation to the control group. MATERIALS/METHODS The study included 120 breast cancer patients, 60 patients with benign breast tumors and 60 healthy women. Plasma levels of tested parameters were determined by enzyme-linked immunosorbent assay, CA15-3 by chemiluminescent microparticle immuno assay. RESULTS Tissue inhibitor of metalloproteinase-1 showed the highest value of sensitivity in breast cancer group (86.25%) and, more importantly, highest value in breast cancer stage I (85%). Vascular endothelial growth factor also showed high sensitivity (stage I and II-75%, III-85%, IV-70% and 76.25% in total breast cancer group) and the highest specificity (85%) from all tested parameters. It was also the only parameter which had statistically significant area under curve in all stages. In the total breast cancer group all tested parameters showed statistically significant area under curve, but the maximum range was obtained for combination: 'vascular endothelial growth factor + CA15-3'. Vascular endothelial growth factor seems to be the best candidate for diagnosing breast cancer stage I and for differentiating between breast cancer and non-carcinoma cases. CONCLUSIONS The combined analysis of tested parameters and CA15-3 resulted in an increase in sensitivity and area under curve values, which provides hope for developing new panel of biomarkers that may be used in diagnosing breast cancer in the future.
Collapse
Affiliation(s)
- Monika Zajkowska
- Department of Biochemical Diagnostics, Medical University of Bialystok, Bialystok, Poland.
| | - Ewa Gacuta
- Department of Perinatology, Medical University of Bialystok, Bialystok, Poland
| | - Sylwia Kozłowska
- Department of Laboratory Diagnostics, Public Health Care Hospital, Bialystok, Poland
| | - Emilia Lubowicka
- Department of Esthetic Medicine, Medical University of Bialystok, Bialystok, Poland
| | - Edyta K Głażewska
- Department of Esthetic Medicine, Medical University of Bialystok, Bialystok, Poland
| | - Lech Chrostek
- Department of Biochemical Diagnostics, Medical University of Bialystok, Bialystok, Poland
| | - Maciej Szmitkowski
- Department of Biochemical Diagnostics, Medical University of Bialystok, Bialystok, Poland
| | - Przemysław Pawłowski
- Department of Medical Pathomorphology, Medical University of Bialystok, Bialystok, Poland
| | - Monika Zbucka-Krętowska
- Department of Department of Re-productivness and Gynecological Endocrinology, Medical University of Bialystok, Bialystok, Poland
| | - Sławomir Ławicki
- Department of Biochemical Diagnostics, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
28
|
Anand-Apte B, Chao JR, Singh R, Stöhr H. Sorsby fundus dystrophy: Insights from the past and looking to the future. J Neurosci Res 2019; 97:88-97. [PMID: 30129971 PMCID: PMC6241301 DOI: 10.1002/jnr.24317] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 07/13/2018] [Accepted: 07/26/2018] [Indexed: 12/17/2022]
Abstract
Sorsby fundus dystrophy (SFD), an autosomal dominant, fully penetrant, degenerative disease of the macula, is manifested by symptoms of night blindness or sudden loss of visual acuity, usually in the third to fourth decades of life due to choroidal neovascularization (CNV). SFD is caused by specific mutations in the Tissue Inhibitor of Metalloproteinase-3, (TIMP3) gene. The predominant histo-pathological feature in the eyes of patients with SFD are confluent 20-30 m thick, amorphous deposits found between the basement membrane of the retinal pigment epithelium (RPE) and the inner collagenous layer of Bruch's membrane. SFD is a rare disease but it has generated significant interest because it closely resembles the exudative or "wet" form of the more common age-related macular degeneration (AMD). In addition, in both SFD and AMD donor eyes, sub-retinal deposits have been shown to accumulate TIMP3 protein. Understanding the molecular functions of wild-type and mutant TIMP3 will provide significant insights into the patho-physiology of SFD and perhaps AMD. This review summarizes the current knowledge on TIMP3 and how mutations in TIMP3 cause SFD to provide insights into how we can study this disease going forward. Findings from these studies could have potential therapeutic implications for both SFD and AMD.
Collapse
Affiliation(s)
- Bela Anand-Apte
- Department of Ophthalmic Research, Cole Eye Institute,
Cleveland Clinic Foundation, Cleveland Ohio; Department of Ophthalmology and
Department of Molecular Medicine, Lerner Research Institute, Cleveland Clinic Lerner
College of Medicine, Cleveland, OH,
| | - Jennifer R. Chao
- Department of Ophthalmology, University of Washington,
Seattle, WA 98109,
| | - Ruchira Singh
- Department of Ophthalmology (Flaum Eye Institute) and
Biomedical Genetics, 3Center for Visual Science, UR Stem Cell and Regenerative
Medicine Institute University of Rochester, Rochester, NY, USA, ruchira
| | - Heidi Stöhr
- Institute of Human Genetics, Universität
Regensburg, Regensburg, Germany,
| |
Collapse
|
29
|
Kim HS, Vargas A, Eom YS, Li J, Yamamoto KL, Craft CM, Lee EJ. Tissue inhibitor of metalloproteinases 1 enhances rod survival in the rd1 mouse retina. PLoS One 2018; 13:e0197322. [PMID: 29742163 PMCID: PMC5942829 DOI: 10.1371/journal.pone.0197322] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 04/29/2018] [Indexed: 01/06/2023] Open
Abstract
Retinitis pigmentosa (RP), an inherited retinal degenerative disease, is characterized by a progressive loss of rod photoreceptors followed by loss of cone photoreceptors. Previously, when tissue inhibitor of metalloproteinase 1 (TIMP1), a key extracellular matrix (ECM) regulator that binds to and inhibits activation of Matrix metallopeptidase 9 (MMP9) was intravitreal injected into eyes of a transgenic rhodopsin rat model of RP, S334ter-line3, we discovered cone outer segments are partially protected. In parallel, we reported that a specific MMP9 and MMP2 inhibitor, SB-3CT, interferes with mechanisms leading to rod photoreceptor cell death in an MMP9 dependent manner. Here, we extend our initial rat studies to examine the potential of TIMP1 as a treatment in retinal degeneration by investigating neuroprotective effects in a classic mouse retinal degeneration model, rdPde6b-/- (rd1). The results clearly demonstrate that intravitreal injections of TIMP1 produce extended protection to delay rod photoreceptor cell death. The mean total number of rods in whole-mount retinas was significantly greater in TIMP-treated rd1 retinas (postnatal (P) 30, P35 (P<0.0001) and P45 (P<0.05) than in saline-treated rd1 retinas. In contrast, SB-3CT did not delay rod cell death, leading us to further investigate alternative pathways that do not involve MMPs. In addition to inducing phosphorylated ERK1/2, TIMP1 significantly reduces BAX activity and delays attenuation of the outer nuclear layer (ONL). Physiological responses using scotopic electroretinograms (ERG) reveal b-wave amplitudes from TIMP1-treated retinas are significantly greater than from saline-treated rd1 retinas (P<0.05). In later degenerative stages of rd1 retinas, photopic b-wave amplitudes from TIMP1-treated rd1 retinas are significantly larger than from saline-treated rd1 retinas (P<0.05). Our findings demonstrate that TIMP1 delays photoreceptor cell death. Furthermore, this study provides new insights into how TIMP1 works in the mouse animal model of RP.
Collapse
Affiliation(s)
- Hwa Sun Kim
- MDA Vision Research, USC Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America
| | - Andrew Vargas
- MDA Vision Research, USC Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America
| | - Yun Sung Eom
- MDA Vision Research, USC Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America
| | - Justin Li
- MDA Vision Research, USC Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America
| | - Kyra L. Yamamoto
- MDA Vision Research, USC Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America
| | - Cheryl Mae Craft
- MDA Vision Research, USC Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America
- Department of Integrative Anatomical Sciences, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America
| | - Eun-Jin Lee
- MDA Vision Research, USC Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
30
|
English WR, Ireland-Zecchini H, Baker AH, Littlewood TD, Bennett MR, Murphy G. Tissue Inhibitor of Metalloproteinase-3 (TIMP-3) induces FAS dependent apoptosis in human vascular smooth muscle cells. PLoS One 2018; 13:e0195116. [PMID: 29617412 PMCID: PMC5884528 DOI: 10.1371/journal.pone.0195116] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 03/17/2018] [Indexed: 02/08/2023] Open
Abstract
Over expression of Tissue Inhibitor of Metalloproteinases-3 (TIMP-3) in vascular smooth muscle cells (VSMCs) induces apoptosis and reduces neointima formation occurring after saphenous vein interposition grafting or coronary stenting. In studies to address the mechanism of TIMP-3-driven apoptosis in human VSMCs we find that TIMP-3 increased activation of caspase-8 and apoptosis was inhibited by expression of Cytokine response modifier A (CrmA) and dominant negative FAS-Associated protein with Death Domain (FADD). TIMP-3 induced apoptosis did not cause mitochondrial depolarisation, increase activation of caspase-9 and was not inhibited by over-expression of B-cell Lymphoma 2 (Bcl2), indicating a mitochondrial independent/type-I death receptor pathway. TIMP-3 increased levels of the First Apoptosis Signal receptor (FAS) and depletion of FAS with shRNA showed TIMP-3-induced apoptosis was FAS dependent. TIMP-3 induced formation of the Death-Inducing Signalling Complex (DISC), as detected by immunoprecipitation and by immunofluorescence. Cellular-FADD-like IL-1 converting enzyme-Like Inhibitory Protein (c-FLIP) localised with FAS at the cell periphery in the absence of TIMP-3 and this localisation was lost on TIMP-3 expression with c-FLIP adopting a perinuclear localisation. Although TIMP-3 inhibited FAS shedding, this did not increase total surface levels of FAS but instead increased FAS levels within localised regions at the cell surface. A Disintegrin And Metalloproteinase 17 (ADAM17) is inhibited by TIMP-3 and depletion of ADAM17 with shRNA significantly decreased FAS shedding. However ADAM17 depletion did not induce apoptosis or replicate the effects of TIMP-3 by increasing localised clustering of cell surface FAS. ADAM17-depleted cells could activate caspase-3 when expressing levels of TIMP-3 that were otherwise sub-apoptotic, suggesting a partial role for ADAM17 mediated ectodomain shedding in TIMP-3 mediated apoptosis. We conclude that TIMP-3 induced apoptosis in VSMCs is highly dependent on FAS and is associated with changes in FAS and c-FLIP localisation, but is not solely dependent on shedding of the FAS ectodomain.
Collapse
Affiliation(s)
- William R. English
- Cancer Research UK Cambridge Research Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, United Kingdom
- Department of Oncology and Metabolism, University of Sheffield School of Medicine, Sheffield, United Kingdom
- * E-mail:
| | - Heather Ireland-Zecchini
- Microscopy Core Facility, Cancer Research UK Cambridge Research Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, United Kingdom
| | - Andrew H. Baker
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | | | - Martin R. Bennett
- Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Gillian Murphy
- Cancer Research UK Cambridge Research Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, United Kingdom
| |
Collapse
|
31
|
Han Q, Zhang W, Meng J, Ma L, Li A. LncRNA-LET inhibits cell viability, migration and EMT while induces apoptosis by up-regulation of TIMP2 in human granulosa-like tumor cell line KGN. Biomed Pharmacother 2018; 100:250-256. [PMID: 29432996 DOI: 10.1016/j.biopha.2018.01.162] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 01/04/2018] [Accepted: 01/29/2018] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a common endocrine disease characterized by hyperandrogenism, irregular menses, and polycystic ovaries. Several long non-coding RNAs (lncRNAs) are aberrantly expressed in PCOS patients; however, little is known about the effects of the lncRNA-low expression in tumor (lncRNA-LET) on PCOS. We aimed to explore the effects of lncRNA-LET on human granulosa-like tumor cell line, KGN. METHODS Expression of lncRNA-LET in normal IOSE80 cells and granulosa cells was determined by qRT-PCR. KGN cell viability, apoptosis and migration were measured by trypan blue exclusion method, flow cytometry assay and wound healing assay, respectively. TGF-β1 was used to induce epithelial-mesenchymal transition (EMT) process. LncRNA-LET expression and mRNA expressions of TIMP2 and EMT-related proteins were measured by qRT-PCR. Western blot analysis was used to measure the protein expression of apoptosis-related proteins, EMT-related proteins, TIMP2, and the proteins in the Wnt/β-catenin and Notch signaling pathways. RESULTS lncRNA-LET was down-regulated in KGN cells, and its overexpression inhibited cell viability and migration, and promoted apoptosis in KGN cells. Overexpression of lncRNA-LET increased the expression of E-cadherin and decreased the expressions of N-cadherin and vimentin in KGN cells. These effects of lncRNA-LET on KGN cells were reversed by TIMP2 suppression. Overexpression of TIMP2 inhibited cell viability, migration and EMT process, and increased apoptosis by activating the Wnt/β-catenin and Notch pathways. CONCLUSION Overexpression of lncRNA-LET inhibits cell viability, migration and EMT process, and increases apoptosis in KGN cells by up-regulating the expression of TIMP2 and activating the Wnt/β-catenin and notch signaling pathways.
Collapse
Affiliation(s)
- Qingfang Han
- Department of Obstetrics, The Affiliated Hospital of Qingdao University, Qingdao 266555, China.
| | - Wenke Zhang
- Department of Obstetrics, The Affiliated Hospital of Qingdao University, Qingdao 266555, China
| | - Jinlai Meng
- Department of Obstetrics, Shandong Provincial Hospital, Jinan 250021, China
| | - Li Ma
- Department of Nutrition, The Affiliated Hospital of Qingdao University, Qingdao 266555, China
| | - Aihua Li
- Department of Obstetrics and Gynecology, Liaocheng People's Hospital, Liaocheng 252000, China
| |
Collapse
|
32
|
Wang X, Khalil RA. Matrix Metalloproteinases, Vascular Remodeling, and Vascular Disease. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 81:241-330. [PMID: 29310800 DOI: 10.1016/bs.apha.2017.08.002] [Citation(s) in RCA: 421] [Impact Index Per Article: 52.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc-dependent endopeptidases that degrade various proteins in the extracellular matrix (ECM). Typically, MMPs have a propeptide sequence, a catalytic metalloproteinase domain with catalytic zinc, a hinge region or linker peptide, and a hemopexin domain. MMPs are commonly classified on the basis of their substrates and the organization of their structural domains into collagenases, gelatinases, stromelysins, matrilysins, membrane-type (MT)-MMPs, and other MMPs. MMPs are secreted by many cells including fibroblasts, vascular smooth muscle (VSM), and leukocytes. MMPs are regulated at the level of mRNA expression and by activation through removal of the propeptide domain from their latent zymogen form. MMPs are often secreted in an inactive proMMP form, which is cleaved to the active form by various proteinases including other MMPs. MMPs degrade various protein substrates in ECM including collagen and elastin. MMPs could also influence endothelial cell function as well as VSM cell migration, proliferation, Ca2+ signaling, and contraction. MMPs play a role in vascular tissue remodeling during various biological processes such as angiogenesis, embryogenesis, morphogenesis, and wound repair. Alterations in specific MMPs could influence arterial remodeling and lead to various pathological disorders such as hypertension, preeclampsia, atherosclerosis, aneurysm formation, as well as excessive venous dilation and lower extremity venous disease. MMPs are often regulated by endogenous tissue inhibitors of metalloproteinases (TIMPs), and the MMP/TIMP ratio often determines the extent of ECM protein degradation and tissue remodeling. MMPs may serve as biomarkers and potential therapeutic targets for certain vascular disorders.
Collapse
Affiliation(s)
- Xi Wang
- Vascular Surgery Research Laboratories, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
33
|
Sorsby fundus dystrophy - A review of pathology and disease mechanisms. Exp Eye Res 2017; 165:35-46. [PMID: 28847738 DOI: 10.1016/j.exer.2017.08.014] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 08/23/2017] [Accepted: 08/23/2017] [Indexed: 01/29/2023]
Abstract
Sorsby fundus dystrophy (SFD) is an autosomal dominant macular dystrophy with an estimated prevalence of 1 in 220,000 and an onset of disease around the 4th to 6th decade of life. Similar to age-related macular degeneration (AMD), ophthalmoscopy reveals accumulation of protein/lipid deposits under the retinal pigment epithelium (RPE), referred to as drusen, in the eyes of patients with SFD. SFD is caused by variants in the gene for tissue inhibitor of metalloproteinases-3 (TIMP3), which has been found in drusen-like deposits of SFD patients. TIMP3 is constitutively expressed by RPE cells and, in healthy eyes, resides in Bruch's membrane. Most SFD-associated TIMP3 variants involve the gain or loss of a cysteine residue. This suggests the protein aberrantly forms intermolecular disulphide bonds, resulting in the formation of TIMP3 dimers. It has been demonstrated that SFD-associated TIMP3 variants are more resistant to turnover, which is thought to be a result of dimerisation and thought to explain the accumulation of TIMP3 in drusen-like deposits at the level of Bruch's membrane. An important function of TIMP3 within the outer retina is to regulate the thickness of Bruch's membrane. TIMP3 performs this function by inhibiting the activity of matrix metalloproteinases (MMPs), which have the function of catalysing breakdown of the extracellular matrix. TIMP3 has an additional function to inhibit vascular endothelial growth factor (VEGF) signalling and thereby to inhibit angiogenesis. However, it is unclear whether SFD-associated TIMP3 variant proteins retain these functions. In this review, we discuss the current understanding of the potential mechanisms underlying development of SFD and summarise all known SFD-associated TIMP3 variants. Cell culture models provide an invaluable way to study disease and identify potential treatments. These allow a greater understanding of RPE physiology and pathophysiology, including the ability to study the blood-retinal barrier as well as other RPE functions such as phagocytosis of photoreceptor outer segments. This review describes some examples of such recent in vitro studies and how they might provide new insights into degenerative diseases like SFD. Thus far, most studies on SFD have been performed using ARPE-19 cells or other, less suitable, cell-types. Now, induced pluripotent stem cell (iPSC) technologies allow the possibility to non-invasively collect somatic cells, such as dermal fibroblast cells and reprogram those to produce iPSCs. Subsequent differentiation of iPSCs can generate patient-derived RPE cells that carry the same disease-associated variant as RPE cells in the eyes of the patient. Use of these patient-derived RPE cells in novel cell culture systems should increase our understanding of how SFD and similar macular dystrophies develop.
Collapse
|
34
|
Matrix Metalloproteinase Inhibitors as Investigational and Therapeutic Tools in Unrestrained Tissue Remodeling and Pathological Disorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 148:355-420. [PMID: 28662828 DOI: 10.1016/bs.pmbts.2017.04.003] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Matrix metalloproteinases (MMPs) are zinc-dependent proteolytic enzymes that degrade various proteins in the extracellular matrix (ECM). MMPs may also regulate the activity of membrane receptors and postreceptor signaling mechanisms and thereby affect cell function. The MMP family includes collagenases, gelatinases, stromelysins, matrilysins, membrane-type MMPs, and other MMPs. Inactive proMMPs are cleaved by other MMPs or proteases into active MMPs, which interact with various protein substrates in ECM and cell surface. MMPs regulate important biological processes such as vascular remodeling and angiogenesis and may be involved in the pathogenesis of cardiovascular disorders such as hypertension, atherosclerosis, and aneurysm. The role of MMPs is often assessed by measuring their mRNA expression, protein levels, and proteolytic activity using gel zymography. MMP inhibitors are also used to assess the role of MMPs in different biological processes and pathological conditions. MMP activity is regulated by endogenous tissue inhibitors of metalloproteinases (TIMPs), and the MMP/TIMP balance could determine the net MMP activity, ECM turnover, and tissue remodeling. Also, several synthetic MMP inhibitors have been developed. Synthetic MMP inhibitors include a large number of zinc-binding globulins (ZBGs), in addition to non-ZBGs and mechanism-based inhibitors. MMP inhibitors have been proposed as potential tools in the management of osteoarthritis, cancer, and cardiovascular disorders. However, most MMP inhibitors have broad-spectrum actions on multiple MMPs and could cause undesirable musculoskeletal side effects. Currently, doxycycline is the only MMP inhibitor approved by the Food and Drug Administration. New generation biological and synthetic MMP inhibitors may show greater MMP specificity and fewer side effects and could be useful in targeting specific MMPs, reducing unrestrained tissue remodeling, and the management of MMP-related pathological disorders.
Collapse
|
35
|
Valencia AM, Cai CL, Tan J, Duggan TJ, Valencia GB, Aranda JV, Beharry KD. Intravitreal bevacizumab alters type IV collagenases and exacerbates arrested alveologenesis in the neonatal rat lungs. Exp Lung Res 2017; 43:120-133. [PMID: 28409646 DOI: 10.1080/01902148.2017.1306897] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Purpose/Aim: Intravitreal bevacizumab (Avastin) is an irreversible vascular endothelial growth factor (VEGF) inhibitor used off-label to treat severe retinopathy of prematurity in extremely low gestational age neonates. VEGF and matrix metalloproteinases (MMPs) and the tissue inhibitors of metalloproteinases (TIMPs) participate in lung maturation. We tested the hypothesis that intravitreal bevacizumab enters the systemic circulation and has long-lasting effects on lung MMPs. MATERIALS AND METHODS Neonatal rats were exposed to: (1) hyperoxia (50% O2); (2) intermittent hypoxia (IH) (50% O2 with brief episodes of 12% O2); or (3) room air (RA) from birth (P0) to P14. At P14, the time of eye opening in rats, a single dose of Avastin (0.125 mg) was injected into the vitreous cavity of the left eye. A control group received equivalent volume saline. At P23 and P45, lung MMP-2 and MMP-9, and TIMP-1, and TIMP-2 were assessed in the lungs. RESULTS At P23, Avastin increased MMP-2, MMP-9, and TIMP-1 levels in the hyperoxia group but decreased TIMP-1 levels in the IH group. The ratios of MMP-2/TIMP-1 and MMP-9/TIMP-1 were significantly elevated at P23 in the IH group treated with Avastin. At P45, the levels of MMP-2 and MMP-9 remained elevated in the hyperoxia and IH groups treated with Avastin, while a rebound increase in TIMP-1 levels was noted in the IH group. CONCLUSIONS Avastin treatment in IH has lasting alterations in the balance between MMPs and their tissue inhibitors. These changes may lead to impaired alveologenesis and tissue damage consistent with bronchopulmonary dysplasia/chronic lung disease.
Collapse
Affiliation(s)
- Arwin M Valencia
- a Department of Pediatrics, Division of Neonatal-Perinatal Medicine , State University of New York, Downstate Medical Center , Brooklyn , New York , USA.,b Department of Pediatrics, Division of Neonatal-Perinatal Medicine , Sumerlin Hospital Medical Center, Valley Healthcare System , Las Vegas , Nevada , USA
| | - Charles L Cai
- a Department of Pediatrics, Division of Neonatal-Perinatal Medicine , State University of New York, Downstate Medical Center , Brooklyn , New York , USA
| | - Jeffrey Tan
- c Department of Ophthalmology , State University of New York, Downstate Medical Center , Brooklyn, New York , USA.,d SUNY Eye Institute , New York , New York , USA
| | - Thomas J Duggan
- a Department of Pediatrics, Division of Neonatal-Perinatal Medicine , State University of New York, Downstate Medical Center , Brooklyn , New York , USA
| | - Gloria B Valencia
- a Department of Pediatrics, Division of Neonatal-Perinatal Medicine , State University of New York, Downstate Medical Center , Brooklyn , New York , USA
| | - Jacob V Aranda
- a Department of Pediatrics, Division of Neonatal-Perinatal Medicine , State University of New York, Downstate Medical Center , Brooklyn , New York , USA.,c Department of Ophthalmology , State University of New York, Downstate Medical Center , Brooklyn, New York , USA.,d SUNY Eye Institute , New York , New York , USA
| | - Kay D Beharry
- a Department of Pediatrics, Division of Neonatal-Perinatal Medicine , State University of New York, Downstate Medical Center , Brooklyn , New York , USA.,c Department of Ophthalmology , State University of New York, Downstate Medical Center , Brooklyn, New York , USA.,d SUNY Eye Institute , New York , New York , USA
| |
Collapse
|
36
|
Shi WZ, Ju JY, Xiao HJ, Xue F, Wu J, Pan MM, Ni WF. Dynamics of MMP‑9, MMP‑2 and TIMP‑1 in a rat model of brain injury combined with traumatic heterotopic ossification. Mol Med Rep 2017; 15:2129-2135. [PMID: 28259914 PMCID: PMC5365022 DOI: 10.3892/mmr.2017.6275] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 12/12/2016] [Indexed: 01/12/2023] Open
Abstract
The present study aimed to detect early changes in the concentration of matrix metalloproteinase-9 (MMP-9), matrix metalloproteinase-2 (MMP-2) and tissue inhibitor of metalloproteinase-1 (TIMP-1) in a rat model of brain injury combined with traumatic heterotopic ossification (HO). A total of 132 male Sprague-Dawley rats were used to establish the experimental and control groups. Anatomy and sample collection were conducted on postoperative days 1, 2, 3, 4, 5, 6 and 7. Hematoxylin and eosin and immunohistochemical staining were performed for local tissues. MMP-9, MMP-2 and TIMP-1 levels and gene expression level were measured by ELISA and reverse transcription-quantitative polymerase chain reaction. Radiological investigation of the rat lower limbs was conducted at weeks 5 and 10 following modeling to observe the occurrence of HO. The incidence of HO for rats in the experimental group was higher compared with the control group. The serum MMP-9 levels of the experimental group were notably higher on postoperative days 5–7 compared with the control group. The MMP-9 gene expression of the experimental group was higher on postoperative days 3–7 compared with the control group. The TIMP-1 gene expression levels were markedly higher compared with the control group at each time point. Thus, an increase in inflammatory response is closely associated with brain injury, in addition to an increase in the number of inflammatory cells with the incidence of HO. The pathological elevation of MMP-9 and the altered dynamic equilibrium between MMP-9 and TIMP-1 contributed to the degradation, remodeling and calcification of the extracellular matrix, resulting in the induction of osteoblast precursor cells in HO. MMP-9 is a predictive marker of HO.
Collapse
Affiliation(s)
- Wei-Zhe Shi
- Department of Orthopedics, Southern Medical University Affiliated Fengxian Hospital, Shanghai 200000, P.R. China
| | - Jin-Yong Ju
- Department of Orthopedics, Fengxian Central Hospital Graduate Training Base, Liaoning Medical University, Shanghai 310000, P.R. China
| | - Hai-Jun Xiao
- Department of Orthopedics, Southern Medical University Affiliated Fengxian Hospital, Shanghai 200000, P.R. China
| | - Feng Xue
- Department of Orthopedics, Shanghai Fengxian District Central Hospital, Shanghai 310000, P.R. China
| | - Jiang Wu
- Department of Orthopedics, Changsha Eighth Hospital, Changsha, Hunan 430000, P.R. China
| | - Ming-Mang Pan
- Department of Orthopedics, Southern Medical University Affiliated Fengxian Hospital, Shanghai 200000, P.R. China
| | - Wei-Feng Ni
- Department of Orthopedics, Shanghai Fengxian District Central Hospital, Shanghai 310000, P.R. China
| |
Collapse
|
37
|
Yıldırım-Buharalıoğlu G, Bond M, Sala-Newby GB, Hindmarch CCT, Newby AC. Regulation of Epigenetic Modifiers, Including KDM6B, by Interferon-γ and Interleukin-4 in Human Macrophages. Front Immunol 2017; 8:92. [PMID: 28228757 PMCID: PMC5296311 DOI: 10.3389/fimmu.2017.00092] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 01/19/2017] [Indexed: 11/25/2022] Open
Abstract
Background Interferon-γ (IFN-γ) or interleukin-4 (IL-4) drives widely different transcriptional programs in macrophages. However, how IFN-γ and IL-4 alter expression of histone-modifying enzymes involved in epigenetic regulation and how this affects the resulting phenotypic polarization is incompletely understood. Methods and results We investigated steady-state messenger RNA levels of 84 histone-modifying enzymes and related regulators in colony-stimulating factor-1 differentiated primary human macrophages using quantitative polymerase chain reaction. IFN-γ or IL-4 treatment for 6–48 h changed 11 mRNAs significantly. IFN-γ increased CIITA, KDM6B, and NCOA1, and IL-4 also increased KDM6B by 6 h. However, either cytokine decreased AURKB, ESCO2, SETD6, SUV39H1, and WHSC1, whereas IFN-γ alone decreased KAT2A, PRMT7, and SMYD3 mRNAs only after 18 h, which coincided with decreased cell proliferation. Rendering macrophages quiescent by growth factor starvation or adenovirus-mediated overexpression of p27kip1 inhibited expression of AURKB, ESCO2, SUV39H1, and WHSC1, and mRNA levels were restored by overexpressing the S-phase transcription factor E2F1, implying their expression, at least partly, depended on proliferation. However, CIITA, KDM6B, NCOA1, KAT2A, PRMT7, SETD6, and SMYD3 were regulated independently of effects on proliferation. Silencing KDM6B, the only transcriptional activator upregulated by both IFN-γ and IL-4, pharmacologically or with short hairpin RNA, blunted a subset of responses to each cytokine. Conclusion These findings demonstrate that IFN-γ or IL-4 can regulate the expression of histone acetyl transferases and histone methyl transferases independently of effects on proliferation and that upregulation of the histone demethylase, KDM6B, assists phenotypic polarization by both cytokines.
Collapse
Affiliation(s)
| | - Mark Bond
- Chair of Vascular Cell Biology, School of Clinical Sciences, University of Bristol , Bristol , UK
| | - Graciela B Sala-Newby
- Chair of Vascular Cell Biology, School of Clinical Sciences, University of Bristol , Bristol , UK
| | - Charles C T Hindmarch
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada; Department of Physiology, Faculty of Medicine, University of Malaya, Lembah Pantai, Kuala Lumpur, Malaysia
| | - Andrew C Newby
- Chair of Vascular Cell Biology, School of Clinical Sciences, University of Bristol , Bristol , UK
| |
Collapse
|
38
|
Ławicki S, Głażewska EK, Sobolewska M, Będkowska GE, Szmitkowski M. Plasma Levels and Diagnostic Utility of Macrophage Colony-Stimulating Factor, Matrix Metalloproteinase-9, and Tissue Inhibitor of Metalloproteinases-1 as New Biomarkers of Breast Cancer. Ann Lab Med 2017; 36:223-9. [PMID: 26915610 PMCID: PMC4773262 DOI: 10.3343/alm.2016.36.3.223] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 01/26/2016] [Accepted: 01/29/2016] [Indexed: 12/21/2022] Open
Abstract
Background Macrophage colony-stimulating factor (M-CSF), matrix metalloproteinase-9 (MMP-9), and its specific tissue inhibitor - tissue inhibitor of metalloproteinases-1 (TIMP-1) may play an important role in the pathogenesis and spread of cancer. We investigated the plasma levels of M-CSF, MMP-9, and TIMP-1 in comparison with a commonly accepted tumor marker CA 15-3 in breast cancer patients and in control groups. Methods The cohort included 110 breast cancer patients in groups at stages I-IV. The control group consisted of 50 healthy volunteers and 50 benign tumor patients. Plasma levels of M-CSF, MMP-9, and TIMP-1 were determined by using ELISA, while CA 15-3 concentrations were determined by using chemiluminescent microparticle immunoassay (CMIA). Results The results showed significant differences in concentrations of the analyzed parameters and in levels of CA 15-3 between the groups of breast cancer patients and the two control groups. Diagnosis using these markers was equal to that using CA 15-3 in terms of sensitivity, predictive values of positive and negativetest results (PPV, NPV) and area under the ROC curve (AUC) in the studied groups. The diagnostic specificities of MMP-9, TIMP-1, M-CSF, and CA 15-3 showed equally high values (95%). The combined use of all tested parameters with CA 15-3 resulted in increased sensitivity, NPV, and AUC, especially in the combination of M-CSF with tumor markers (76%, 64%, and 0.8653). Conclusions These findings suggest the tested parameters are useful in the diagnosis of breast cancer patients (except stage I), when combined with CA 15-3.
Collapse
Affiliation(s)
- Sławomir Ławicki
- Department of Biochemical Diagnostics, Medical University, Białystok, Poland.
| | | | - Monika Sobolewska
- Department of Biochemical Diagnostics, Medical University, Białystok, Poland
| | | | - Maciej Szmitkowski
- Department of Biochemical Diagnostics, Medical University, Białystok, Poland
| |
Collapse
|
39
|
Ghorai A, Sarma A, Chowdhury P, Ghosh U. PARP-1 depletion in combination with carbon ion exposure significantly reduces MMPs activity and overall increases TIMPs expression in cultured HeLa cells. Radiat Oncol 2016; 11:126. [PMID: 27659937 PMCID: PMC5034624 DOI: 10.1186/s13014-016-0703-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 09/13/2016] [Indexed: 12/11/2022] Open
Abstract
Background Hadron therapy is an innovative technique where cancer cells are precisely killed leaving surrounding healthy cells least affected by high linear energy transfer (LET) radiation like carbon ion beam. Anti-metastatic effect of carbon ion exposure attracts investigators into the field of hadron biology, although details remain poor. Poly(ADP-ribose) polymerase-1 (PARP-1) inhibitors are well-known radiosensitizer and several PARP-1 inhibitors are in clinical trial. Our previous studies showed that PARP-1 depletion makes the cells more radiosensitive towards carbon ion than gamma. The purpose of the present study was to investigate combining effects of PARP-1 inhibition with carbon ion exposure to control metastatic properties in HeLa cells. Methods Activities of matrix metalloproteinases-2, 9 (MMP-2, MMP-9) were measured using the gelatin zymography after 85 MeV carbon ion exposure or gamma irradiation (0- 4 Gy) to compare metastatic potential between PARP-1 knock down (HsiI) and control cells (H-vector - HeLa transfected with vector without shRNA construct). Expression of MMP-2, MMP-9, tissue inhibitor of MMPs such as TIMP-1, TIMP-2 and TIMP-3 were checked by immunofluorescence and western blot. Cell death by trypan blue, apoptosis and autophagy induction were studied after carbon ion exposure in each cell-type. The data was analyzed using one way ANOVA and 2-tailed paired-samples T-test. Results PARP-1 silencing significantly reduced MMP-2 and MMP-9 activities and carbon ion exposure further diminished their activities to less than 3 % of control H-vector. On the contrary, gamma radiation enhanced both MMP-2 and MMP-9 activities in H-vector but not in HsiI cells. The expression of MMP-2 and MMP-9 in H-vector and HsiI showed different pattern after carbon ion exposure. All three TIMPs were increased in HsiI, whereas only TIMP-1 was up-regulated in H-vector after irradiation. Notably, the expressions of all TIMPs were significantly higher in HsiI than H-vector at 4 Gy. Apoptosis was the predominant mode of cell death and no autophagic death was observed. Conclusions Our study demonstrates for the first time that PARP-1 inhibition in combination with carbon ion synergistically decreases MMPs activity along with overall increase of TIMPs. These data open up the possibilities of improvement of carbon ion therapy with PARP-1 inhibition to control highly metastatic cancers. Electronic supplementary material The online version of this article (doi:10.1186/s13014-016-0703-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Atanu Ghorai
- Department of Biochemistry & Biophysics, University of Kalyani, Kalyani, 741235, India.,Present address: Department of Biological Sciences, Tata Institute of Fundamental Research (TIFR), Homi Bhabha Road, Colaba, Mumbai, 400005, India
| | - Asitikantha Sarma
- Inter-University Accelerator Center (IUAC), Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Priyanka Chowdhury
- Department of Biochemistry & Biophysics, University of Kalyani, Kalyani, 741235, India
| | - Utpal Ghosh
- Department of Biochemistry & Biophysics, University of Kalyani, Kalyani, 741235, India.
| |
Collapse
|
40
|
Nguyen TTTN, Shynlova O, Lye SJ. Matrix Metalloproteinase Expression in the Rat Myometrium During Pregnancy, Term Labor, and Postpartum. Biol Reprod 2016; 95:24. [PMID: 27251092 PMCID: PMC5029434 DOI: 10.1095/biolreprod.115.138248] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 05/16/2016] [Indexed: 01/08/2023] Open
Abstract
Pregnancy, spontaneous term labor (TL), and postpartum (PP) involution are associated with changes in the cellular and extracellular matrix composition of the uterus. Both the uterine smooth muscle (myometrium) and the infiltrating peripheral blood leukocytes involved in the activation of labor secrete extracellular matrix-degrading enzymes (matrix metalloproteinases, MMPs) that can modulate cellular behavior and barrier function. MMP expression is induced by mechanical stretch in several tissues. We hypothesized that the expression and activity of myometrial MMPs and their tissue inhibitors (TIMPs) are modulated in preparation for TL and PP involution and are regulated by mechanical stretch of uterine walls imposed by the growing fetus. Myometrial tissues were collected from bilaterally and unilaterally pregnant rats across gestation, TL, and PP. Total RNA and proteins were subjected to real-time PCR and immunoblotting, respectively, and tissue localization and activity was examined by immunohistochemistry and in situ zymography. We found that Mmp7, Mmp11, and Mmp12 mRNA levels were upregulated during TL and PP, while Mmp2, Mmp3, Mmp8, Mmp9, Mmp10, and Mmp13 mRNAs were only upregulated during PP. Timp1–Timp4 were stably expressed throughout gestation with some fluctuations PP. Active MMP2 was induced in the empty uterine horn during gestation and in the gravid PP uterus, suggesting negative regulation by biological mechanical stretch. We conclude that specific subsets of uterine MMPs are differentially regulated in the rat myometrium in preparation for two major events: TL and PP uterine involution.
Collapse
Affiliation(s)
- Tina Tu-Thu Ngoc Nguyen
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada Department of Physiology, University of Toronto, Canada
| | - Oksana Shynlova
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada Department of Obstetrics and Gynecology, University of Toronto, Canada
| | - Stephen J Lye
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada Department of Physiology, University of Toronto, Canada Department of Obstetrics and Gynecology, University of Toronto, Canada
| |
Collapse
|
41
|
Garziano A, Urciuolo F, Imparato G, Martorina F, Corrado B, Netti P. A micro-perfusion bioreactor for on line investigation of ECM remodeling under hydrodynamic and biochemical stimulation. LAB ON A CHIP 2016; 16:855-867. [PMID: 26860053 DOI: 10.1039/c5lc01481f] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Tissue-on-chip (TOC) systems aim at replicating complex biological dynamics in vitro with the potential either to improve the understanding of human biology or to develop more accurate therapeutic strategies. To replicate faithfully the intricate interrelationships between cells and their surrounding microenvironment, the three-dimensional (3D) tissue model must possess a responsive extracellular matrix (ECM). ECM remodeling plays a pivotal role in guiding cells and tissues functions and such aspect is somewhat denied during in vitro studies. For this purpose, we fabricated a micro-perfusion bioreactor capable to sustain the viability of 3D engineered tissue models recapitulating the process of the native ECM deposition and assembly. Engineered human dermis micro-tissue precursors (HD-μTP) were used as building blocks to generate a final tissue. HD-μTP were loaded in the perfusion space of the micro-perfusion bioreactor and, under the superimposition of different fluid dynamic regimes and biochemical stimulation, they synthesized new collagen proteins that were, then, assembled in the perfusion space forming a continuum of cells embedded in their own ECM. The micro-perfusion bioreactor was fabricated to allow the on-line monitoring of the oxygen consumption and the assembly of the newly formed collagen network via real time acquisition of the second harmonic generation (SHG) signal. The possibility to detect the collagen reorganization due to both fluid dynamic and biochemical stimulation, let us to define the optimal perfusion configuration in order to obtain a TOC system based on an endogenous and responsive ECM.
Collapse
Affiliation(s)
- A Garziano
- Center for Advanced Biomaterials for Health Care@CRIB Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci n. 53, 80125 Napoli, Italy. and Department of Chemical, Materials and Industrial Production Engineering (DICMAPI), University of Napoli Federico II, P.le Tecchio 80, 80125 Napoli, Italy and Interdisciplinary Research Centre on Biomaterials (CRIB), University of Napoli Federico II, P.le Tecchio 80, 80125 Napoli, Italy
| | - F Urciuolo
- Center for Advanced Biomaterials for Health Care@CRIB Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci n. 53, 80125 Napoli, Italy.
| | - G Imparato
- Center for Advanced Biomaterials for Health Care@CRIB Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci n. 53, 80125 Napoli, Italy.
| | - F Martorina
- Center for Advanced Biomaterials for Health Care@CRIB Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci n. 53, 80125 Napoli, Italy.
| | - B Corrado
- Center for Advanced Biomaterials for Health Care@CRIB Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci n. 53, 80125 Napoli, Italy. and Department of Chemical, Materials and Industrial Production Engineering (DICMAPI), University of Napoli Federico II, P.le Tecchio 80, 80125 Napoli, Italy and Interdisciplinary Research Centre on Biomaterials (CRIB), University of Napoli Federico II, P.le Tecchio 80, 80125 Napoli, Italy
| | - P Netti
- Center for Advanced Biomaterials for Health Care@CRIB Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci n. 53, 80125 Napoli, Italy. and Department of Chemical, Materials and Industrial Production Engineering (DICMAPI), University of Napoli Federico II, P.le Tecchio 80, 80125 Napoli, Italy and Interdisciplinary Research Centre on Biomaterials (CRIB), University of Napoli Federico II, P.le Tecchio 80, 80125 Napoli, Italy
| |
Collapse
|
42
|
Tissue inhibitor of metalloproteinase-3 (TIMP3) promotes endothelial apoptosis via a caspase-independent mechanism. Apoptosis 2016; 20:523-34. [PMID: 25558000 DOI: 10.1007/s10495-014-1076-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Tissue inhibitor of metalloproteinases-3 (TIMP3) is a tumor suppressor and a potent inhibitor of angiogenesis. TIMP3 exerts its anti-angiogenic effect via a direct interaction with vascular endothelial growth factor (VEGF) receptor-2 (KDR) and inhibition of proliferation, migration and tube formation of endothelial cells (ECs). TIMP3 has also been shown to induce apoptosis in some cancer cells and vascular smooth muscle cells via MMP inhibition and caspase-dependent mechanisms. In this study, we examined the molecular mechanisms of TIMP3-mediated apoptosis in endothelial cells. We have previously demonstrated that mice developed smaller tumors with decreased vascularity when injected with breast carcinoma cells overexpressing TIMP3, than with control breast carcinoma cells. TIMP3 overexpression resulted in increased apoptosis in human breast carcinoma (MDA-MB435) in vivo but not in vitro. However, TIMP3 could induce apoptosis in ECs in vitro. The apoptotic activity of TIMP3 in ECs appears to be independent of MMP inhibitory activity. Furthermore, the equivalent expression of functional TIMP3 promoted apoptosis and caspase activation in ECs expressing KDR (PAE/KDR), but not in ECs expressing PDGF beta-receptor (PAE/β-R). Surprisingly, the apoptotic activity of TIMP3 appears to be independent of caspases. TIMP3 inhibited matrix-induced focal adhesion kinase (FAK) tyrosine phosphorylation and association with paxillin and disrupted the incorporation of β3 integrin, FAK and paxillin into focal adhesion contacts on the matrix, which were not affected by caspase inhibitors. Thus, TIMP3 may induce apoptosis in ECs by triggering a caspase-independent cell death pathway and targeting a FAK-dependent survival pathway.
Collapse
|
43
|
Perturbations of the cerebrovascular matrisome: A convergent mechanism in small vessel disease of the brain? J Cereb Blood Flow Metab 2016; 36:143-57. [PMID: 25853907 PMCID: PMC4758555 DOI: 10.1038/jcbfm.2015.62] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 03/17/2015] [Indexed: 02/08/2023]
Abstract
The term matrisome refers to the ensemble of proteins constituting the extracellular matrix (ECM) (core matrisome) as well as the proteins associated with the ECM. Every organ has an ECM with a unique composition that not only provides the support and anchorage for cells, but also controls fundamental cellular processes as diverse as differentiation, survival, proliferation, and polarity. The current knowledge of the matrisome of small brain vessels is reviewed with a focus on the basement membrane (BM), a specialized form of ECM located at the interface between endothelial cells, contractile cells (smooth muscle cells and pericytes), and astrocyte endfeet—a very strategic location in the communication pathway between the cerebral microcirculation and astrocytes. We discuss some of the most recent genetic data and relevant findings from experimental models of nonamyloid cerebral small vessel disease (SVD). We propose the concept that perturbations of the cerebrovascular matrisome is a convergent pathologic pathway in monogenic forms of SVD, and is likely relevant to the sporadic disease.
Collapse
|
44
|
Li J, Shen T, Wu G, Wei Q, Mao D, Shi F. Potential roles of matrix metalloproteinases and characteristics of ovarian development in neonatal guinea pigs. Tissue Cell 2015; 47:478-88. [DOI: 10.1016/j.tice.2015.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 07/01/2015] [Accepted: 07/20/2015] [Indexed: 12/31/2022]
|
45
|
Abdelnaseer M, Elfayomi N, Esmail EH, Kamal MM, Hamdy A, Samie RMA, Elsawy E. Relationship between matrix metalloproteinase-9 and common carotid artery intima media thickness. Neurol Sci 2015; 37:117-122. [PMID: 26319043 DOI: 10.1007/s10072-015-2358-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 08/03/2015] [Indexed: 11/26/2022]
Abstract
Atherosclerosis causes significant morbidity and mortality. Carotid intima media thickness (IMT) predicts future ischaemic strok e incidence. Matrix metalloproteinases (MMPs) play a considerable role in atherosclerosis and hold therapeutic promise as well. To investigate the relationship between serum level of matrix metalloproteinase-9 (MMP-9) and common carotid artery intima media thickness (CCA-IMT) in patients with ischaemic stroke and asymptomatic subjects. Thirty patients with a previous ischaemic stroke and 30 asymptomatic volunteers were recruited. Assessment of vascular risk factors, serum level of MMP-9 and CCA-IMT on both sides was performed. The IMT of both CCAs correlated positively with the serum MMP-9 level in asymptomatic subjects (p = 0.000), even after adjustment for other risk factors. In the patients group, this positive correlation was significant for the right but not for the left CCA (right CCA: p = 0.023, left CCA: p = 0.0284). Fasting blood sugar correlated positively with serum levels of MMP-9 in asymptomatic subjects (p = 0.005) but did not correlate positively in patients. There was no significant correlation between MMP-9 and age or other investigated laboratory risk factors in either the patient or asymptomatic groups. MMP-9 is positively correlated with CCA-IMT both in stroke patients and asymptomatic subjects. This may indicate that MMP-9 is a possible therapeutic target for stroke prevention.
Collapse
Affiliation(s)
- Maged Abdelnaseer
- Neurology Department, Cairo University, 49 Misr Hilwan Elzraai Street, Maadi, Cairo, 2218, Egypt
| | - Nervana Elfayomi
- Neurology Department, Cairo University, 49 Misr Hilwan Elzraai Street, Maadi, Cairo, 2218, Egypt
| | - Eman H Esmail
- Neurology Department, Cairo University, 49 Misr Hilwan Elzraai Street, Maadi, Cairo, 2218, Egypt.
| | - Manal M Kamal
- Chemical and Clinical Pathology Department, Cairo University, Cairo, Egypt
| | - Ahmed Hamdy
- Internal Medicine Department, Cairo University, Cairo, Egypt
| | | | - Enji Elsawy
- Neurology Department, Cairo University, 49 Misr Hilwan Elzraai Street, Maadi, Cairo, 2218, Egypt
| |
Collapse
|
46
|
Yamagami K, Oka T, Wang Q, Ishizu T, Lee JK, Miwa K, Akazawa H, Naito AT, Sakata Y, Komuro I. Pirfenidone exhibits cardioprotective effects by regulating myocardial fibrosis and vascular permeability in pressure-overloaded hearts. Am J Physiol Heart Circ Physiol 2015; 309:H512-22. [DOI: 10.1152/ajpheart.00137.2015] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 05/28/2015] [Indexed: 12/12/2022]
Abstract
Although cardiac fibrosis causes heart failure, its molecular mechanisms remain elusive. In this study, we investigated the mechanisms of cardiac fibrosis and examined the effects of the antifibrotic drug pirfenidone (PFD) on chronic heart failure. To understand the responsible mechanisms, we generated an in vivo pressure-overloaded heart failure model via transverse aortic constriction (TAC) and examined the effects of PFD on chronic-phase cardiac fibrosis and function. In the vehicle group, contractile dysfunction and left ventricle fibrosis progressed further from 4 to 8 wk after TAC but were prevented by PFD treatment beginning 4 wk after TAC. We isolated cardiac fibroblasts and vascular endothelial cells from the left ventricles of adult male mice and investigated the cell-type-specific effects of PFD. Transforming growth factor-β induced upregulated collagen 1 expression via p38 phosphorylation and downregulated claudin 5 (Cldn5) expression in cardiac fibroblasts and endothelial cells, respectively; both processes were inhibited by PFD. Moreover, PFD inhibited changes in the collagen 1 and Cldn5 expression levels, resulting in reduced fibrosis and serum albumin leakage into the interstitial space during the chronic phase in TAC hearts. In conclusion, PFD inhibited cardiac fibrosis by suppressing both collagen expression and the increased vascular permeability induced by pressure overload.
Collapse
Affiliation(s)
- Kiyoshi Yamagami
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Toru Oka
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
- CREST, Japan Science and Technology Agency, Tokyo, Japan
| | - Qi Wang
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Takamaru Ishizu
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Jong-Kook Lee
- Department of Cardiovascular Regenerative Medicine, Osaka University Graduate School of Medicine, Suita, Japan
- CREST, Japan Science and Technology Agency, Tokyo, Japan
| | - Keiko Miwa
- Department of Cardiovascular Regenerative Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hiroshi Akazawa
- Department of Cardiovascular Medicine, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
- CREST, Japan Science and Technology Agency, Tokyo, Japan
| | - Atsuhiko T. Naito
- Department of Cardiovascular Medicine, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
- CREST, Japan Science and Technology Agency, Tokyo, Japan
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
- CREST, Japan Science and Technology Agency, Tokyo, Japan
| |
Collapse
|
47
|
Brighenti M. MicroRNA and MET in lung cancer. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:68. [PMID: 25992367 DOI: 10.3978/j.issn.2305-5839.2015.01.26] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 01/16/2015] [Indexed: 12/28/2022]
Abstract
MicroRNAs (miRNAs) are a class of small non-protein coding RNAs that modulate important cellular functions via their post-transcriptional regulation of messenger RNAs (mRNAs). Recent evidences from multiple tumor types and model systems implicate miRNA dysregulation as a common mechanism of tumorigenesis, cancer progression and resistance to therapy. Several miRNAs are dysregulated in cancers and a single miRNA can have multiple targets involved in different oncogenic pathways. MET, the tyrosine kinase receptor for hepatocyte growth factor (HGF), has a central role in lung cancer development and in acquired resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors; it has been predicted and shown to be the target gene of multiple miRNAs, which play a crucial role in controlling its activity in a stimulatory or inhibitory sense. In this review we will focus on the most important and recent studies about the role of miRNAs in the control of MET expression, reporting also the progress made using miRNAs for therapy of lung cancer.
Collapse
|
48
|
Tyagi SC, Joshua IG. Exercise and nutrition in myocardial matrix metabolism, remodeling, regeneration, epigenetics, microcirculation, and muscle. Can J Physiol Pharmacol 2015; 92:521-3. [PMID: 24959992 DOI: 10.1139/cjpp-2014-0197] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Remodeling and myocardial matrix metabolism contributes to cardiac endothelium-myocyte (perivascular fibrosis), myocyte-myocyte (interstitial fibrosis), and mitochondrion-myocyte (fusion and fission) coupling. Matrix metalloproteinases (MMPs), and tissue inhibitor of metalloproteinases (TIMPs) play differential roles in different tissues and diseases. For example, although present in the heart, MMP-3 is known as stromelysin (i.e., stromal tissue enzyme). Interestingly, TIMP-3 causes apoptosis. Exercise and nutrition are synergistic in the mitigation of diseases: exercise releases exosomes containing miRNAs. Nutrition/vitamins B6 and B12 regulate the metabolism of homocysteine (an epigenetic byproduct of DNA/RNA/protein methylation). Thus, epigenetic silencing is an important therapeutic target. The statistical analysis of cohorts may be less indicative for the treatment of a disease, particularly if the 2 twins are different in terms of responding to the medicine for the same disease, therefore, personalized medicine is the future of therapy.
Collapse
Affiliation(s)
- Suresh C Tyagi
- Department of Physiology and Biophysics, University of Louisville School of Medicine, 323 East Chestnut Street, Louisville, KY 40202, USA
| | | |
Collapse
|
49
|
Chaturvedi P, Kalani A, Familtseva A, Kamat PK, Metreveli N, Tyagi SC. Cardiac tissue inhibitor of matrix metalloprotease 4 dictates cardiomyocyte contractility and differentiation of embryonic stem cells into cardiomyocytes: Road to therapy. Int J Cardiol 2015; 184:350-363. [PMID: 25745981 PMCID: PMC4417452 DOI: 10.1016/j.ijcard.2015.01.091] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 01/08/2015] [Accepted: 01/24/2015] [Indexed: 02/08/2023]
Abstract
BACKGROUND TIMP4 (Tissue Inhibitors of Matrix Metalloprotease 4), goes down in failing hearts and mice lacking TIMP4 show poor regeneration capacity after myocardial infarction (MI). This study is based on our previous observation that administration of cardiac inhibitor of metalloproteinase (~TIMP4) attenuates oxidative stress and remodeling in failing hearts. Therefore, we hypothesize that TIMP4 helps in cardiac regeneration by augmenting contractility and inducing the differentiation of cardiac progenitor cells into cardiomyocytes. METHODS To validate this hypothesis, we transfected mouse cardiomyocytes with TIMP4 and TIMP4-siRNA and performed contractility studies in the TIMP4 transfected cardiomyocytes as compared to siRNA-TIMP4 transfected cardiomyocytes. We evaluated the calcium channel gene serca2a (sarcoplasmic reticulum calcium ATPase2a) and mir122a which tightly regulates serca2a to explain the changes in contractility. We treated mouse embryonic stem cells with cardiac extract and cardiac extract minus TIMP4 (using TIMP4 monoclonal antibody) to examine the effect of TIMP4 on differentiation of cardiac progenitor cells. RESULTS Contractility was augmented in the TIMP4 transfected cardiomyocytes as compared to siRNA-TIMP4 transfected cardiomyocytes. There was elevated expression of serca2a in the TIMP4 transformed myocytes and down regulation of mir122a. The cells treated with cardiac extract containing TIMP4 showed cardiac phenotype in terms of Ckit+, GATA4+ and Nkx2.5 expression. CONCLUSION This is a novel report suggesting that TIMP4 augments contractility and induces differentiation of progenitor cells into cardiac phenotype. In view of the failure of MMP9 inhibitors for cardiac therapy, TIMP4 provides an alternative approach, being an indigenous molecule and a natural inhibitor of MMP9.
Collapse
Affiliation(s)
- Pankaj Chaturvedi
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, KY, USA.
| | - Anuradha Kalani
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, KY, USA
| | - Anastasia Familtseva
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, KY, USA
| | - Pradip Kumar Kamat
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, KY, USA
| | - Naira Metreveli
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, KY, USA
| | - Suresh C Tyagi
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, KY, USA
| |
Collapse
|
50
|
Wang M, Kim SH, Monticone RE, Lakatta EG. Matrix metalloproteinases promote arterial remodeling in aging, hypertension, and atherosclerosis. Hypertension 2015; 65:698-703. [PMID: 25667214 DOI: 10.1161/hypertensionaha.114.03618] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Mingyi Wang
- From the Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Biomedical Research Center (BRC), Baltimore, MD.
| | - Soo Hyuk Kim
- From the Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Biomedical Research Center (BRC), Baltimore, MD
| | - Robert E Monticone
- From the Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Biomedical Research Center (BRC), Baltimore, MD
| | - Edward G Lakatta
- From the Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Biomedical Research Center (BRC), Baltimore, MD.
| |
Collapse
|