1
|
Li W, Yang Y, Lin Y, Mu D. In Vitro Study of Thymosin Beta 4 Promoting Transplanted Fat Survival by Regulating Adipose-Derived Stem Cells. Aesthetic Plast Surg 2024; 48:2179-2189. [PMID: 38409346 DOI: 10.1007/s00266-024-03861-1] [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: 11/19/2023] [Accepted: 01/12/2024] [Indexed: 02/28/2024]
Abstract
BACKGROUND Autologous fat grafting (AFG) has emerged as a highly sought-after plastic surgery procedure, although its success has been hampered by the uncertain fat survival rate. Current evidence suggests that adipose-derived stem cells (ADSCs) may contribute to fat retention in AFG. In previous studies, it was confirmed that thymosin beta 4 (Tβ4) could enhance fat survival in vivo, although the precise mechanism remains unclear. METHODS ADSCs were isolated from patients undergoing liposuction and their proliferation, apoptosis, anti-apoptosis, and migration were analyzed under Tβ4 stimulation using cell counting kit-8, flow cytometry, wound healing assay, and real-time quantitative PCR. The mRNA levels of genes relating to angiogenesis and Hippo signaling were also determined. RESULTS Tβ4 at 100 ng/mL (p-value = 0.0171) and 1000 ng/mL (p-value = 0.0054) significantly increased ADSC proliferation from day 1 compared to the control group (0 ng/mL). In addition, the mRNA levels of proliferation-associated genes were elevated in the Tβ4 group. Furthermore, Tβ4 enhanced the anti-apoptotic ability of ADSCs when stimulated with Tβ4 and an apoptotic induction reagent (0 ng/mL vs. 1000 ng/mL, p-value = 0.011). Crucially, the mRNA expression levels of angiogenesis-related genes and critical genes in the Hippo pathway were affected by Tβ4 in ADSCs. CONCLUSIONS Tβ4 enhances adipose viability in AFG via facilitating ADSC proliferation and reducing apoptosis, and acts as a crucial positive regulator of ADSC-associated angiogenesis. Additionally, Tβ4 could be accountable for the phenotypic adjustment of ADSCs by regulating the Hippo pathway. NO LEVEL ASSIGNED This journal requires that authors assign a level of evidence to each submission to which Evidence-Based Medicine rankings are applicable. This excludes Review Articles, Book Reviews, and manuscripts that concern Basic Science, Animal Studies, Cadaver Studies, and Experimental Studies. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Collapse
Affiliation(s)
- Wandi Li
- Senior Department of Burns and Plastic Surgery, The Fourth Medical Center of PLA General Hospital, No. 51 Fucheng Road, Haidian District, Beijing, 100048, People's Republic of China
- Department of Aesthetic and Reconstructive Breast Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 33 Badachu Road, Shijingshan District, Beijing, 100144, People's Republic of China
| | - Yan Yang
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu District, Changsha, Hunan, 410008, People's Republic of China
| | - Yan Lin
- Department of Aesthetic and Reconstructive Breast Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 33 Badachu Road, Shijingshan District, Beijing, 100144, People's Republic of China
| | - Dali Mu
- Department of Aesthetic and Reconstructive Breast Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 33 Badachu Road, Shijingshan District, Beijing, 100144, People's Republic of China.
| |
Collapse
|
2
|
Lin C, Qiu L, Wang P, Zhang B, Yan L, Zhao C. Thymosin beta-4 participate in antibacterial immunity and wound healing in black tiger shrimp, Penaeus monodon. FISH & SHELLFISH IMMUNOLOGY 2023; 141:109065. [PMID: 37689229 DOI: 10.1016/j.fsi.2023.109065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/11/2023]
Abstract
Thymosin beta-4 (Tβ4) is a ubiquitous protein with multiple and diverse intracellular and extracellular functions in vertebrates, which play fundamental roles in innate immune against pathogens and wound healing. In this study, the full-length cDNA of Tβ4 was cloned from Penaeus monodon (designated as PmTβ4), using the technology of rapid amplification of cDNA ends (RACE). The cDNA of PmTβ4 was 1361 bp with an open reading frame (ORF) of 501 bp, which encoding a polypeptide of 166 amino acid. The Quantitative Real-time PCR (qRT-PCR) analysis results showed that PmTβ4 was ubiquitously expressed in all the tested shrimp tissues, with the highest expression level was detected in the hemolymph, while the lowest expression level in the muscle. The expression level of PmTβ4 was significantly up-regulated in hepatopancreas after challenged by Vibrio parahaemolyticus, Vibrio harveyi and Staphylococcus aureus. In vitro antimicrobial test showed that the recombinant protein of PmTβ4 (rPmTβ4) had broad-spectrum of antimicrobial activity, which could inhibit both the growth of gram-negative bacteria and gram-positive bacteria, including Vibrio vulnificus, V. parahaemolyticus, Streptococcus agalactiae, S. aureus and Aeromonas hydrophila. Moreover, rPmTβ4 had a certain binding ability to different bacteria, and this binding ability exhibits a strong dose-dependent effect. In vivo, PmTβ4 could facilitate external bacterial clearance in shrimp, and have beneficial to shrimp survival post V. parahaemolyticus infection. Furthermore, wound-healing assay was carried out to study the role of PmTβ4 in the process of wound healing. The results showed that the PmTβ4 expression was significantly up-regulated by injury treatment, and exerted positive effects to promote wound healing. In addition, PmTβ4 can significantly increase the expression level of superoxide dismutase (SOD) and Catalase (CAT) after injury treatment in shrimp, which would involve in scavenging reactive oxygen species (ROS) caused by the wound. In conclusion, these results indicated that PmTβ4 may play important roles in antibacterial immunity and wound healing in Penaeus monodon.
Collapse
Affiliation(s)
- Changhong Lin
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, PR China; College of Aqua-life Science and Technology, Shanghai Ocean University, Shanghai, PR China; Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture, Guangzhou, PR China
| | - Lihua Qiu
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, PR China; Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture, Guangzhou, PR China; Key Laboratory of Fishery Ecology and Environment, Guangdong Province, PR China
| | - Pengfei Wang
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, PR China; Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture, Guangzhou, PR China
| | - Bo Zhang
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, PR China; Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture, Guangzhou, PR China
| | - Lulu Yan
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, PR China; Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture, Guangzhou, PR China
| | - Chao Zhao
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, PR China; Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture, Guangzhou, PR China.
| |
Collapse
|
3
|
Stewart W, Hejl C, Guleria RS, Gupta S. Effect of thymosin β4 on lipopolysaccharide‑stimulated brain microvascular endothelial cell remodeling: A possible role in blood‑brain barrier injury. Exp Ther Med 2023; 26:468. [PMID: 37664684 PMCID: PMC10469577 DOI: 10.3892/etm.2023.12167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 07/07/2023] [Indexed: 09/05/2023] Open
Abstract
War veterans, in particular, are more prone to mental illness as they are more likely to have encountered multiple traumatic brain injuries (TBIs) whilst serving on active duty in war zone areas. A TBI is known to cause mortality or serious neurological disabilities among survivors and elicits a number of pathological processes, including neuroinflammation and blood brain barrier (BBB) disruption, leading to secondary brain damage and subsequent impairment of the neurovascular unit. Although several drugs exhibit promising effects for TBI, the repertoire of currently available therapeutic strategies remains limited. Thymosin 4 (Tβ4) is a 43-amino acid G-acting sequestering peptide that confers neuroprotective potential in TBI models. However, its role in BBB function remains unclear. Further research into the mechanism of BBB disruption induced by TBI and its specific role in neurovascular pathophysiology is necessary. In the present study, the protective effects of Tβ4 in lipopolysaccharide (LPS)-stimulated gene expression of several tight junction proteins, inflammatory genes, apoptotic genes, and adhesion genes in human brain microvascular endothelial cells (hBMVECs), one of the pivotal cell types in the BBB, were reported. The results suggested that pretreatment with Tβ4 reversed the LPS-induced damage of BBB components in hBMVECs. Furthermore, these results identified neuregulin 1 as a possible target for Tβ4. Therefore, it is proposed that Tβ4-mediated cellular signaling in hBMVEC may be vital for understanding the association between the BBB and TBI pathophysiology, which warrants further investigation.
Collapse
Affiliation(s)
- William Stewart
- Biomarkers & Genetics Core, VISN 17 Center of Excellence for Research on Returning War Veterans, Central Texas Veterans Health Care System, Waco, TX 76711, USA
| | - Christina Hejl
- Biomarkers & Genetics Core, VISN 17 Center of Excellence for Research on Returning War Veterans, Central Texas Veterans Health Care System, Waco, TX 76711, USA
| | - Rakeshwar S. Guleria
- Biomarkers & Genetics Core, VISN 17 Center of Excellence for Research on Returning War Veterans, Central Texas Veterans Health Care System, Waco, TX 76711, USA
| | - Sudhiranjan Gupta
- Biomarkers & Genetics Core, VISN 17 Center of Excellence for Research on Returning War Veterans, Central Texas Veterans Health Care System, Waco, TX 76711, USA
| |
Collapse
|
4
|
Tsai CY, Chen PH, Chen AL, Wang TSA. Spatiotemporal Investigation of Intercellular Heterogeneity via Multiple Photocaged Probes. Chemistry 2023; 29:e202301067. [PMID: 37382047 DOI: 10.1002/chem.202301067] [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: 04/03/2023] [Revised: 06/14/2023] [Accepted: 06/28/2023] [Indexed: 06/30/2023]
Abstract
Intercellular heterogeneity occurs widely under both normal physiological environments and abnormal disease-causing conditions. Several attempts to couple spatiotemporal information to cell states in a microenvironment were performed to decipher the cause and effect of heterogeneity. Furthermore, spatiotemporal manipulation can be achieved with the use of photocaged/photoactivatable molecules. Here, we provide a platform to spatiotemporally analyze differential protein expression in neighboring cells by multiple photocaged probes coupled with homemade photomasks. We successfully established intercellular heterogeneity (photoactivable ROS trigger) and mapped the targets (directly ROS-affected cells) and bystanders (surrounding cells), which were further characterized by total proteomic and cysteinomic analysis. Different protein profiles were shown between bystanders and target cells in both total proteome and cysteinome. Our strategy should expand the toolkit of spatiotemporal mapping for elucidating intercellular heterogeneity.
Collapse
Affiliation(s)
- Chun-Yi Tsai
- Department of Chemistry, National Taiwan University and Center for, Emerging Material and Advanced Devices, National Taiwan University, Taipei, 10617, Taiwan (R.O.C
| | - Po-Hsun Chen
- Department of Chemistry, National Taiwan University and Center for, Emerging Material and Advanced Devices, National Taiwan University, Taipei, 10617, Taiwan (R.O.C
| | - Ai-Lin Chen
- Department of Chemistry, National Taiwan University and Center for, Emerging Material and Advanced Devices, National Taiwan University, Taipei, 10617, Taiwan (R.O.C
| | - Tsung-Shing Andrew Wang
- Department of Chemistry, National Taiwan University and Center for, Emerging Material and Advanced Devices, National Taiwan University, Taipei, 10617, Taiwan (R.O.C
| |
Collapse
|
5
|
Zhang Y, Bailey JT, Xu E, Singh K, Lavaert M, Link VM, D'Souza S, Hafiz A, Cao J, Cao G, Sant'Angelo DB, Sun W, Belkaid Y, Bhandoola A, McGavern DB, Yang Q. Mucosal-associated invariant T cells restrict reactive oxidative damage and preserve meningeal barrier integrity and cognitive function. Nat Immunol 2022; 23:1714-1725. [PMID: 36411380 PMCID: PMC10202031 DOI: 10.1038/s41590-022-01349-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 10/03/2022] [Indexed: 11/22/2022]
Abstract
Increasing evidence indicates close interaction between immune cells and the brain, revising the traditional view of the immune privilege of the brain. However, the specific mechanisms by which immune cells promote normal neural function are not entirely understood. Mucosal-associated invariant T cells (MAIT cells) are a unique type of innate-like T cell with molecular and functional properties that remain to be better characterized. In the present study, we report that MAIT cells are present in the meninges and express high levels of antioxidant molecules. MAIT cell deficiency in mice results in the accumulation of reactive oxidative species in the meninges, leading to reduced expression of junctional protein and meningeal barrier leakage. The presence of MAIT cells restricts neuroinflammation in the brain and preserves learning and memory. Together, our work reveals a new functional role for MAIT cells in the meninges and suggests that meningeal immune cells can help maintain normal neural function by preserving meningeal barrier homeostasis and integrity.
Collapse
Affiliation(s)
- Yuanyue Zhang
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Jacob T Bailey
- Department of Immunology & Microbial Disease, Albany Medical College, Albany, NY, USA
| | - En Xu
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Kunal Singh
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Marieke Lavaert
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Verena M Link
- Metaorganism Immunity Section, Laboratory of Immune System Biology and Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shanti D'Souza
- Department of Immunology & Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Alex Hafiz
- Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Jian Cao
- Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Gaoyuan Cao
- Rutgers Institute for Translational Medicine and Science, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Derek B Sant'Angelo
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
- Department of Pediatrics, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Wei Sun
- Department of Immunology & Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Immune System Biology and Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Avinash Bhandoola
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Dorian B McGavern
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Qi Yang
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA.
- Rutgers Institute for Translational Medicine and Science, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA.
- Department of Pediatrics, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA.
| |
Collapse
|
6
|
Sharma V, Manhas A, Gupta S, Dikshit M, Jagavelu K, Verma RS. Fabrication, characterization and in vivo assessment of cardiogel loaded chitosan patch for myocardial regeneration. Int J Biol Macromol 2022; 222:3045-3056. [DOI: 10.1016/j.ijbiomac.2022.10.079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 10/06/2022] [Accepted: 10/09/2022] [Indexed: 11/05/2022]
|
7
|
Thymosin β4 Protects against Cardiac Damage and Subsequent Cardiac Fibrosis in Mice with Myocardial Infarction. Cardiovasc Ther 2022; 2022:1308651. [PMID: 35712678 PMCID: PMC9187458 DOI: 10.1155/2022/1308651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 04/27/2022] [Accepted: 05/07/2022] [Indexed: 11/17/2022] Open
Abstract
Background Inflammation is a critical factor in the development and progression of myocardial infarction and cardiac fibrosis. Thymosin β4 (Tβ4) alleviates the disease process via protective antioxidant and anti-inflammatory mechanisms. Although Tβ4 has been shown to have a protective effect in myocardial infarction, its impact on cardiac fibrosis has not been well reported. In this study, we evaluated the influence of exogenous Tβ4 on myocardial infarction and cardiac fibrosis and explored the possible underlying mechanism. Methods Real-time quantitative reverse-transcription PCR (qRT-PCR), immunohistochemistry (IHC), and Western blot were used to analyze Tβ4 expression in acute myocardial infarction (AMI) cardiac tissues. The effects of intraperitoneal adeno-associated virus-Tβ4 (AAV-Tβ4) on ligation-induced AMI in mice were studied using cardiac function parameters, and RT-PCR, Western blot, HE staining, Masson staining, and IHC were used to assess the degree of myocardial fibrosis. The effects of Tβ4 were confirmed in vitro using mouse cardiac myocytes and myofibroblasts. Results Tβ4 was shown to be significantly elevated in mice AMI cardiac tissues. In mice, AAV-Tβ4 induced exogenous expression of Tβ4 significantly reduced oxidative damage, inflammation, cardiac dysfunction, and fibrosis. H2O2 inhibited mitophagy and increased inflammation in mouse cardiac myocytes via oxidative stress, and Tβ4 substantially reduced mitophagy inhibition and inflammasome activation in myocytes caused by H2O2. Furthermore, Tβ4 decreased cardiac myofibroblast growth and reduced TGF-β1-induced activation. Conclusions AAV-Tβ4 induced expression of Tβ4 reduced inflammation, heart damage, and eventual fibrosis in vivo. Tβ4 helped to reduce oxidative stress, promote mitophagy, and alleviate inflammation and fibrosis. Exogenous supplementation of Tβ4 might be a promising therapeutic agent for treating myocardial infarction as well as cardiac fibrosis.
Collapse
|
8
|
Maselli D, Matos RS, Johnson RD, Chiappini C, Camelliti P, Campagnolo P. Epicardial slices: an innovative 3D organotypic model to study epicardial cell physiology and activation. NPJ Regen Med 2022; 7:7. [PMID: 35039552 PMCID: PMC8764051 DOI: 10.1038/s41536-021-00202-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 11/30/2021] [Indexed: 11/26/2022] Open
Abstract
The epicardium constitutes an untapped reservoir for cardiac regeneration. Upon heart injury, the adult epicardium re-activates, leading to epithelial-to-mesenchymal transition (EMT), migration, and differentiation. While interesting mechanistic and therapeutic findings arose from lower vertebrates and rodent models, the introduction of an experimental system representative of large mammals would undoubtedly facilitate translational advancements. Here, we apply innovative protocols to obtain living 3D organotypic epicardial slices from porcine hearts, encompassing the epicardial/myocardial interface. In culture, our slices preserve the in vivo architecture and functionality, presenting a continuous epicardium overlaying a healthy and connected myocardium. Upon thymosin β4 treatment of the slices, the epicardial cells become activated, upregulating epicardial and EMT genes, resulting in epicardial cell mobilization and differentiation into epicardial-derived mesenchymal cells. Our 3D organotypic model enables to investigate the reparative potential of the adult epicardium, offering an advanced tool to explore ex vivo the complex 3D interactions occurring within the native heart environment.
Collapse
Affiliation(s)
- D Maselli
- Faculty of Health & Medical Sciences, School of Biosciences & Medicine, Section of Cardiovascular Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - R S Matos
- Faculty of Health & Medical Sciences, School of Biosciences & Medicine, Section of Cardiovascular Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - R D Johnson
- Faculty of Health & Medical Sciences, School of Biosciences & Medicine, Section of Cardiovascular Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - C Chiappini
- Centre for Craniofacial and Regenerative Biology, King's College London, SE1 9RT, London, United Kingdom
| | - P Camelliti
- Faculty of Health & Medical Sciences, School of Biosciences & Medicine, Section of Cardiovascular Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - P Campagnolo
- Faculty of Health & Medical Sciences, School of Biosciences & Medicine, Section of Cardiovascular Sciences, University of Surrey, Guildford, GU2 7XH, UK.
| |
Collapse
|
9
|
Lachowicz JI, Pichiri G, Piludu M, Fais S, Orrù G, Congiu T, Piras M, Faa G, Fanni D, Dalla Torre G, Lopez X, Chandra K, Szczepski K, Jaremko L, Ghosh M, Emwas AH, Castagnola M, Jaremko M, Hannappel E, Coni P. Thymosin β4 Is an Endogenous Iron Chelator and Molecular Switcher of Ferroptosis. Int J Mol Sci 2022; 23:551. [PMID: 35008976 PMCID: PMC8745404 DOI: 10.3390/ijms23010551] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/01/2022] [Accepted: 01/03/2022] [Indexed: 02/01/2023] Open
Abstract
Thymosin β4 (Tβ4) was extracted forty years agofrom calf thymus. Since then, it has been identified as a G-actin binding protein involved in blood clotting, tissue regeneration, angiogenesis, and anti-inflammatory processes. Tβ4 has also been implicated in tumor metastasis and neurodegeneration. However, the precise roles and mechanism(s) of action of Tβ4 in these processes remain largely unknown, with the binding of the G-actin protein being insufficient to explain these multi-actions. Here we identify for the first time the important role of Tβ4 mechanism in ferroptosis, an iron-dependent form of cell death, which leads to neurodegeneration and somehow protects cancer cells against cell death. Specifically, we demonstrate four iron2+ and iron3+ binding regions along the peptide and show that the presence of Tβ4 in cell growing medium inhibits erastin and glutamate-induced ferroptosis in the macrophage cell line. Moreover, Tβ4 increases the expression of oxidative stress-related genes, namely BAX, hem oxygenase-1, heat shock protein 70 and thioredoxin reductase 1, which are downregulated during ferroptosis. We state the hypothesis that Tβ4 is an endogenous iron chelator and take part in iron homeostasis in the ferroptosis process. We discuss the literature data of parallel involvement of Tβ4 and ferroptosis in different human pathologies, mainly cancer and neurodegeneration. Our findings confronted with literature data show that controlled Tβ4 release could command on/off switching of ferroptosis and may provide novel therapeutic opportunities in cancer and tissue degeneration pathologies.
Collapse
Affiliation(s)
- Joanna I. Lachowicz
- Department of Medical Sciences and Public Health, University of Cagliari, Cittadella Universitaria, 09042 Monserrato, Italy; (J.I.L.); (T.C.); (M.P.); (G.F.); (D.F.); (P.C.)
| | - Giusi Pichiri
- Department of Medical Sciences and Public Health, University of Cagliari, Cittadella Universitaria, 09042 Monserrato, Italy; (J.I.L.); (T.C.); (M.P.); (G.F.); (D.F.); (P.C.)
| | - Marco Piludu
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria, 09042 Monserrato, Italy
| | - Sara Fais
- Department of Surgical Science, OBL Oral Biotechnology Laboratory, University of Cagliari, 09124 Cagliari, Italy; (S.F.); (G.O.)
| | - Germano Orrù
- Department of Surgical Science, OBL Oral Biotechnology Laboratory, University of Cagliari, 09124 Cagliari, Italy; (S.F.); (G.O.)
| | - Terenzio Congiu
- Department of Medical Sciences and Public Health, University of Cagliari, Cittadella Universitaria, 09042 Monserrato, Italy; (J.I.L.); (T.C.); (M.P.); (G.F.); (D.F.); (P.C.)
| | - Monica Piras
- Department of Medical Sciences and Public Health, University of Cagliari, Cittadella Universitaria, 09042 Monserrato, Italy; (J.I.L.); (T.C.); (M.P.); (G.F.); (D.F.); (P.C.)
| | - Gavino Faa
- Department of Medical Sciences and Public Health, University of Cagliari, Cittadella Universitaria, 09042 Monserrato, Italy; (J.I.L.); (T.C.); (M.P.); (G.F.); (D.F.); (P.C.)
| | - Daniela Fanni
- Department of Medical Sciences and Public Health, University of Cagliari, Cittadella Universitaria, 09042 Monserrato, Italy; (J.I.L.); (T.C.); (M.P.); (G.F.); (D.F.); (P.C.)
| | - Gabriele Dalla Torre
- Kimika Fakultatea, Euskal Herriko Unibertsitatea UPV/EHU, Donostia International Physics Center (DIPC), P.K. 1072 Donostia Euskadi, 20080 San Sebastian, Spain; (G.D.T.); (X.L.)
| | - Xabier Lopez
- Kimika Fakultatea, Euskal Herriko Unibertsitatea UPV/EHU, Donostia International Physics Center (DIPC), P.K. 1072 Donostia Euskadi, 20080 San Sebastian, Spain; (G.D.T.); (X.L.)
| | - Kousik Chandra
- Smart-Health Initiative (SHI) and Red Sea Research Center (RSRC), Division of Biological and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia; (K.C.); (K.S.); (L.J.); (M.G.)
| | - Kacper Szczepski
- Smart-Health Initiative (SHI) and Red Sea Research Center (RSRC), Division of Biological and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia; (K.C.); (K.S.); (L.J.); (M.G.)
| | - Lukasz Jaremko
- Smart-Health Initiative (SHI) and Red Sea Research Center (RSRC), Division of Biological and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia; (K.C.); (K.S.); (L.J.); (M.G.)
| | - Mitra Ghosh
- Smart-Health Initiative (SHI) and Red Sea Research Center (RSRC), Division of Biological and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia; (K.C.); (K.S.); (L.J.); (M.G.)
| | - Abdul-Hamid Emwas
- Core Labs, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia;
| | - Massimo Castagnola
- Institute of Chemistry of Molecular Recognition, National Research Council (Consiglio Nazionale delle Ricerche), 00185 Rome, Italy;
- Laboratory of Proteomics and Metabolomics, IRCCS, Santa Lucia Foundation, 00143 Rome, Italy
| | - Mariusz Jaremko
- Smart-Health Initiative (SHI) and Red Sea Research Center (RSRC), Division of Biological and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia; (K.C.); (K.S.); (L.J.); (M.G.)
| | - Ewald Hannappel
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nuremberg, 91058 Erlangen, Germany;
| | - Pierpaolo Coni
- Department of Medical Sciences and Public Health, University of Cagliari, Cittadella Universitaria, 09042 Monserrato, Italy; (J.I.L.); (T.C.); (M.P.); (G.F.); (D.F.); (P.C.)
| |
Collapse
|
10
|
Tian Z, Yao N, Wang F, Ruan L. Thymosin β4 Suppresses LPS-Induced Murine Lung Fibrosis by Attenuating Oxidative Injury and Alleviating Inflammation. Inflammation 2021; 45:59-73. [PMID: 34414534 DOI: 10.1007/s10753-021-01528-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 07/27/2021] [Indexed: 11/29/2022]
Abstract
Inflammation plays a critical role in the progression of pulmonary fibrosis. Thymosin β4 (Tβ4) has antioxidant, anti-inflammatory, and antifibrotic effects. Although the potent protective role of Tβ4 in bleomycin-induced pulmonary fibrosis has been validated, the underlying mechanism is not clear; moreover, the influence of Tβ4 on lipopolysaccharide (LPS)-induced lung injury/fibrosis has not been reported. Expression of Tβ4 in fibrotic lung tissues was assessed by real-time quantitative reverse-transcription PCR (rt-PCR), immunohistochemistry (IHC), and western blotting. The effects of intraperitoneal adeno-associated virus-Tβ4 (AAV-Tβ4) on LPS-induced lung injury and fibrosis were observed through the evaluation of collagen deposition and α-smooth muscle actin (SMA) expression. In vitro tests with HPAEpiC and HLF-1 cells were performed to confirm the effects of Tβ4. In this study, we evaluated the role of Tβ4 in pulmonary fibrosis and explored the possible underlying mechanisms. Tβ4 was markedly upregulated in human or mouse fibrotic lung tissues. AAV-Tβ4 markedly alleviated LPS-induced oxidative damage, lung injury, inflammation, and fibrosis in mice. Our in vitro experiments also showed that LPS inhibited mitophagy and promoted inflammation via oxidative stress in HPAEpiC, and Tβ4 significantly attenuated LPS-induced mitophagy inhibition, inflammasome activation, and transforming growth factor-β (TGF)-β1-induced epithelial-mesenchymal transition (EMT) in HPAEpiC. Moreover, Tβ4 suppressed the proliferation and attenuated the TGF-β1-induced activation of HLF-1 cells. In conclusion, Tβ4 alleviates LPS-induced lung injury, inflammation, and subsequent fibrosis in mice, suggesting that Tβ4 has a protective role in the pathogenesis of pulmonary fibrosis. Tβ4 is involved in attenuating oxidative injury, promoting mitophagy, and alleviating inflammation and fibrosis. Modulation of Tβ4 might be a novel strategy for treating pulmonary fibrosis.
Collapse
Affiliation(s)
- Zhen Tian
- Department of Ultrasound, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China. .,Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China. .,Department of Ultrasound, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China.
| | - Naijuan Yao
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Fei Wang
- Department of Ultrasound, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Litao Ruan
- Department of Ultrasound, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| |
Collapse
|
11
|
Poniedziałek-Kempny K, Gajda B, Rajska I, Gajda L, Smorąg Z. Effect of thymosin β on maturation of pig oocytes and quality of in vitro produced embryos. JOURNAL OF ANIMAL AND FEED SCIENCES 2020. [DOI: 10.22358/jafs/128208/2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
12
|
Ramamoorthy H, Abraham P, Isaac B. Melatonin protects against tenofovir-induced nephrotoxicity in rats by targeting multiple cellular pathways. Hum Exp Toxicol 2020; 40:826-850. [PMID: 33146023 DOI: 10.1177/0960327120968860] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Nephrotoxicity is a dose-limiting side effect of long-term use of tenofovir, a reverse transcriptase inhibitor that is used for the treatment of HIV infection and chronic hepatitis B infection. Identifying an agent that prevents tenofovir disoproxil fumarate (TDF)-induced renal injury can lead to its better tolerance, and a more effective treatment can be achieved. The present study is aimed at investigating whether melatonin, a potent antioxidant and anti-inflammatory agent, protects against TDF nephrotoxicity in rats and to determine its cellular targets. Rats were divided into groups and treated as follows. Group I (control): Rats in this group (n = 6) received sterile water only by gavage for 35 days. Group II: Rats (n = 6) in this group received 600 mg/kg body weight TDF in sterile water by gavage for 35 days. Group III: Rats (n = 6) in this group received once daily 20 mg/kg bodyweight melatonin i.p. 2 h before the administration of 600 mg/kg body weight TDF in sterile water by gavage for 35 days. Group IV: Rats were pretreated daily with 20 mg/kg body weight melatonin i.p. 2 h before the administration of sterile water by gavage. All the rats were sacrificed on the 36th day, after overnight fast. Melatonin pretreatment protected the rats against TDF nephrotoxicity both histologically and biochemically. Biochemically, melatonin pretreatment attenuated TDF-induced, oxidative stress, nitrosative stress, mitochondrial pathway of apoptosis, PARP overactivation and preserved proximal tubular function (p < 0.01). This suggests that melatonin may be useful in ameliorating TDF nephrotoxicity.
Collapse
Affiliation(s)
| | - Premila Abraham
- Department of Biochemistry, Christian Medical College, Vellore, Tamil Nadu, India
| | - Bina Isaac
- Department of Anatomy, Christian Medical College, Vellore, Tamil Nadu, India
| |
Collapse
|
13
|
Qu Y, Wang Q, Fu S, Guo X, Luan J, Mu D. The Effect of Thymosin beta4 on the Survival of Autologous Fat Grafting: A Preliminary Study. Aesthet Surg J 2020; 40:NP519-NP529. [PMID: 32144415 DOI: 10.1093/asj/sjaa062] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Autologous fat grafting is a common procedure to improve tissue deficiencies. However, the survival rate of fat grafting is unpredictable. Thymosin beta 4 (Tß4), a multifunctional peptide containing 43 amino acids, is effective in angiogenesis, inhibiting apoptosis and inflammation. OBJECTIVES The authors initially investigated the potential effect of Tß4 in fat grafting. METHODS Adipose tissue premixed exogenous Tß4 were transplanted into rabbit ears. Rabbits were randomly assigned to 3 groups: group A, 5 μg/mL Tß4; group B, 10 μg/mL Tß4; and group C, phosphate-buffered saline buffer as a blank control. The fat grafts were subjected to magnetic resonance imaging at 2, 4, and 12 weeks in vivo. Each harvested graft was analyzed at 3 time points after transplantation. RESULTS The fat grafts in the Tß4-treated groups showed better volume and weight retention, greater adipose tissue integrity, adipocyte viability, and angiogenesis. The results of dynamic contrast-enhanced magnetic resonance imaging also showed that the experimental groups increased microcirculation perfusion of the grafts. CONCLUSIONS The study proved that Tß4 could improve adipose tissue survival and neovascularization. It may be useful for fat grafting as a potential protective reagent.
Collapse
Affiliation(s)
- Yaping Qu
- Plastic Surgery Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Qian Wang
- Plastic Surgery Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Su Fu
- Plastic Surgery Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Xiaoshuang Guo
- Plastic Surgery Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Jie Luan
- Plastic Surgery Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Dali Mu
- Plastic Surgery Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| |
Collapse
|
14
|
Coni P, Piras M, Mateddu A, Piludu M, Orru G, Scano A, Cabras T, Piras V, Lachowicz JI, Jaremko M, Faa G, Castagnola M, Pichiri G. Thymosin β4 cytoplasmic/nuclear translocation as a new marker of cellular stress. A Caco2 case study. RSC Adv 2020; 10:12680-12688. [PMID: 35497634 PMCID: PMC9051466 DOI: 10.1039/c9ra10365a] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/20/2020] [Indexed: 01/22/2023] Open
Abstract
Biomarkers of cell stress are important for proper diagnosis, and in studies of how cells respond to drug treatment. Biomarkers that respond early to pharmacological treatment could improve therapy by tailoring the treatment to the needs of the patient. Thymosin beta-4 (Tβ4) plays a significant role in many aspects of cellular metabolism because of its actin-sequestering properties. Other physiological functions of Tβ4 have been also reported. Among these, Tβ4 may play a crucial role during cellular stress. We addressed the relevance of Tβ4 in cellular stress conditions by using different treatments (serum starvation, DMSO, and butyrate administration) in a colon adenocarcinoma cell line (CaCo2), a cell line frequently used for in vitro experimental studies of Tβ4. In this study, different stress stimuli were analyzed and the obtained results were compared using immunocytochemistry, and molecular and biochemical methods. Taken together, the data clearly indicate that the Tβ4 peptide is involved in adaptive and defensive cellular mechanisms, and that different stress inducers lead to a similar Tβ4 cytoplasmic/nuclear translocation. The translocation of Tβ4 between the cytoplasm and the nucleus of the cell seems characteristic of a possible molecular response to cellular stress exerted by this peptide. Biomarkers of cell stress are important for proper diagnosis, and in studies of how cells respond to drug treatment.![]()
Collapse
|
15
|
Renga G, Oikonomou V, Moretti S, Stincardini C, Bellet MM, Pariano M, Bartoli A, Brancorsini S, Mosci P, Finocchi A, Rossi P, Costantini C, Garaci E, Goldstein AL, Romani L. Thymosin β4 promotes autophagy and repair via HIF-1α stabilization in chronic granulomatous disease. Life Sci Alliance 2019; 2:2/6/e201900432. [PMID: 31719116 PMCID: PMC6851533 DOI: 10.26508/lsa.201900432] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 10/31/2019] [Accepted: 11/04/2019] [Indexed: 02/07/2023] Open
Abstract
This study demonstrates that thymosin β4 stabilizes HIF-1a to promote autophagy and up-regulate genes involved in tissue and mucosal barrier protection in chronic granulomatous disease. Chronic granulomatous disease (CGD) is a genetic disorder of the NADPH oxidase characterized by increased susceptibility to infections and hyperinflammation associated with defective autophagy and increased inflammasome activation. Herein, we demonstrate that thymosin β4 (Tβ4), a g-actin sequestering peptide with multiple and diverse intracellular and extracellular activities affecting inflammation, wound healing, fibrosis, and tissue regeneration, promoted in human and murine cells noncanonical autophagy, a form of autophagy associated with phagocytosis and limited inflammation via the death-associated protein kinase 1. We further show that the hypoxia inducible factor-1 (HIF-1)α was underexpressed in CGD but normalized by Tβ4 to promote autophagy and up-regulate genes involved in mucosal barrier protection. Accordingly, inflammation and granuloma formation were impaired and survival increased in CGD mice with colitis or aspergillosis upon Tβ4 treatment or HIF-1α stabilization. Thus, the promotion of endogenous pathways of inflammation resolution through HIF-1α stabilization is druggable in CGD by Tβ4.
Collapse
Affiliation(s)
- Giorgia Renga
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Vasilis Oikonomou
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Silvia Moretti
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | - Marina M Bellet
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Marilena Pariano
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Andrea Bartoli
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | - Paolo Mosci
- Internal Medicine, Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | - Andrea Finocchi
- Department of Pediatrics, Unit of Immune and Infectious Diseases, Children's Hospital Bambino Gesù, Rome, Italy
| | - Paolo Rossi
- Department of Pediatrics, Unit of Immune and Infectious Diseases, Children's Hospital Bambino Gesù, Rome, Italy
| | - Claudio Costantini
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Enrico Garaci
- University San Raffaele and Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele, Rome, Italy
| | - Allan L Goldstein
- Department of Biochemistry and Molecular Medicine, the George Washington University, School of Medicine and Health Sciences, Washington, DC, USA
| | - Luigina Romani
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
16
|
Aksu U, Yaman OM, Guner I, Guntas G, Sonmez F, Tanriverdi G, Eser M, Cakiris A, Akyol S, Seçkin İ, Uzun H, Yelmen N, Sahin G. The Protective Effects of Thymosin-β-4 in a Rat Model of Ischemic Acute Kidney Injury. J INVEST SURG 2019; 34:601-609. [PMID: 31702404 DOI: 10.1080/08941939.2019.1672841] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Despite the progress in the treatment of acute kidney injury (AKI), current curative approaches fail to provide adequate treatment. In this study, we aimed to investigate the possible protective effects of thymosin-β-4(Tβ4) on an ischemic AKI model in rats. METHODS Rats were randomly assigned into four groups (n = 8/group): The control group (sham-operated), the ischemia-reperfusion (I/R) group; renal ischemia (90 min) by infrarenal abdominal aortic occlusion followed by reperfusion (3 h), the Tβ4 + I/R group; treated with Tβ4 before I/R, and the I/Tβ4/R group; treated with Tβ4 just before reperfusion. Besides renal function determination (creatinine (Cr) and blood urea nitrogen (BUN)); histological evaluation was also conducted. Renal tissue caspase-9, matrix metalloproteinase (MMP-9) activities, and hyaluronan levels were measured. Additionally, renal tissue oxidative stress (lipid hydroperoxide, malondialdehyde, superoxide dismutase, glutathione, pro-oxidant-antioxidant balance, ferric reducing antioxidant power, nitric oxide), inflammation (tumor necrosis factor-α, interleukin-1β, interleukin-6, nuclear factor-κβ) were evaluated. RESULTS I/R increased the level of caspase-9, MMP-9 activity, and hyaluronan (p < 0.001) and these were significantly decreased in both Tβ4 groups. Moreover, I/R led to increases in oxidative stress and inflammation parameters (p < 0.001) while the levels of antioxidants were decreased. Nevertheless, Tβ4 in both groups were able to restore oxidative stress and inflammation parameters. Furthermore, Tβ4 attenuated histologic injury caused by I/R (p < 0.01) and diminished serum urea-creatinine levels (p < 0.001). CONCLUSION These results suggest that Tβ4 has significant improving effects in ischemic acute kidney injury. This beneficial effect might be a result of the inhibition of extracellular matrix remodeling and apoptosis cascade via modulation in renal redox status and inflammation.
Collapse
Affiliation(s)
- Ugur Aksu
- Department of Biology, Science Faculty, Istanbul University, Istanbul, Turkey
| | - Onur M Yaman
- Department of Physiology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Ibrahim Guner
- Department of Physiology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Gulcan Guntas
- Department of Nursery, School of Health, University of Kırklareli, Kırklareli, Turkey
| | - Fuat Sonmez
- Department of Physiology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Gamze Tanriverdi
- Department of Histology and Embriology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Mediha Eser
- Department of Histology and Embriology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Aris Cakiris
- Institute of Health Sciences, Genetics, Istanbul University, Istanbul, Turkey
| | - Sibel Akyol
- Department of Physiology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - İsmail Seçkin
- Department of Histology and Embriology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Hafize Uzun
- Department of Biochemistry, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Nermin Yelmen
- Department of Physiology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Gulderen Sahin
- Department of Physiology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| |
Collapse
|
17
|
Chiasson V, Takano APC, Guleria RS, Gupta S. Deficiency of MicroRNA miR-1954 Promotes Cardiac Remodeling and Fibrosis. J Am Heart Assoc 2019; 8:e012880. [PMID: 31640463 PMCID: PMC6898847 DOI: 10.1161/jaha.119.012880] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 09/18/2019] [Indexed: 12/17/2022]
Abstract
Background Cardiac fibrosis occurs because of disruption of the extracellular matrix network leading to myocardial dysfunction. Angiotensin II (AngII) has been implicated in the development of cardiac fibrosis. Recently, microRNAs have been identified as an attractive target for therapeutic intervention in cardiac pathologies; however, the underlying mechanism of microRNAs in cardiac fibrosis remains unclear. Next-generation sequencing analysis identified a novel characterized microRNA, miR-1954, that was significantly reduced in AngII-infused mice. The finding led us to hypothesize that deficiency of miR-1954 triggers cardiac fibrosis. Methods and Results A transgenic mouse was created using α-MHC (α-myosin heavy chain) promoter and was challenged with AngII infusion. AngII induced cardiac hypertrophy and remodeling. The in vivo overexpression of miR-1954 showed significant reduction in cardiac mass and blood pressure in AngII-infused mice. Further analysis showed significant reduction in cardiac fibrotic genes, hypertrophy marker genes, and an inflammatory gene and restoration of a calcium-regulated gene (Atp2a2 [ATPase sarcoplasmic/endoplasmic reticulum Ca2+ transporting 2]; also known as SERCA2), but no changes were observed in apoptotic genes. THBS1 (thrombospondin 1) is indicated as a target gene for miR-1954. Conclusions Our findings provide evidence, for the first time, that miR-1954 plays a critical role in cardiac fibrosis by targeting THBS1. We conclude that promoting the level of miR-1954 would be a promising strategy for the treatment of cardiac fibrosis.
Collapse
Affiliation(s)
| | | | - Rakeshwar S. Guleria
- Department of Medical PhysiologyTexas A&M UniversityTempleTX
- VISN 17 Center of Excellence on Returning War VeteransWacoTX
| | - Sudhiranjan Gupta
- Department of BiologyBaylor UniversityWacoTX
- Department of Medical PhysiologyTexas A&M UniversityTempleTX
- VISN 17 Center of Excellence on Returning War VeteransWacoTX
| |
Collapse
|
18
|
Huang W, Huang C, Ding H, Luo J, Liu Y, Fan R, Xiao F, Fan X, Jiang Z. Involvement of miR-145 in the development of aortic dissection via inducing proliferation, migration, and apoptosis of vascular smooth muscle cells. J Clin Lab Anal 2019; 34:e23028. [PMID: 31489719 PMCID: PMC6977357 DOI: 10.1002/jcla.23028] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 08/02/2019] [Accepted: 08/05/2019] [Indexed: 01/22/2023] Open
Abstract
Aim The current study aimed to examine miR‐145's contribution to thoracic aortic dissection (AD) development by modulating the biological functions of vascular smooth muscle cells (VSMCs). Methods The concentration of circulating miR‐145 was determined in patients with AD and healthy controls using quantitative polymerase chain reaction (qPCR). Aortic specimens were obtained from both individuals with Stanford type A AD undergoing surgical treatment and deceased organ donors (serving as controls) whose causes of death were nonvascular diseases. Then, qPCR and fluorescence in situ hybridization were applied to assess miR‐145 amounts and location, respectively. Furthermore, qPCR and immunoblot were employed to determine SMAD3 (the target gene of miR‐145, involved in the TGF‐β pathway) amounts at the gene and protein levels, respectively. Moreover, in vitro transfection of VSMCs with miR‐145 mimics or inhibitors was conducted. Finally, the 3‐(4,5‐Dimethylthiazol‐2‐yl)‐2,5‐diphenyltetrazolium bromide (MTT) assay, Transwell assay and flow cytometry were employed for detecting VSMC proliferation, migration, and apoptosis, respectively. Results The amounts of miR‐145 in plasma and aortic specimens were markedly reduced in the AD group in comparison with control values (P < .05). miR‐145 was mostly located in VSMCs. Proliferation and apoptosis of VSMCs were significantly induced in vitro by the downregulation of miR‐145. Also, miR‐145 modulated SMAD3 expression. Conclusions miR‐145 was found to be downregulated in patients with AD, which induced the proliferation, migration, and apoptosis of VSMCs by targeting SMAD3. This suggested the involvement of miR‐145 in the pathogenesis of AD.
Collapse
Affiliation(s)
- Wenhui Huang
- Institute of Cardiovascular Disease and Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical School, University of South China, Hengyang, China.,Department of Cardiology, Vascular Center, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Cheng Huang
- Department of Cardiology, Vascular Center, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Huanyu Ding
- Department of Cardiology, Vascular Center, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jianfang Luo
- Department of Cardiology, Vascular Center, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yuan Liu
- Department of Cardiology, Vascular Center, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ruixin Fan
- Department of Cardiovascular Surgery, Vascular Center, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Fei Xiao
- Department of Cardiovascular Surgery, Vascular Center, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xiaoping Fan
- Department of Cardiovascular Surgery, Vascular Center, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhisheng Jiang
- Institute of Cardiovascular Disease and Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
19
|
Chen Q, Shen Z, Mao Y, Li Q, Liu Y, Mei M, Qiu F, Wang M. Inhibition of microRNA-34a mediates protection of thymosin beta 4 in endothelial progenitor cells against advanced glycation endproducts by targeting B-cell lymphoma 2. Can J Physiol Pharmacol 2019; 97:945-951. [PMID: 31397599 DOI: 10.1139/cjpp-2018-0743] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The aim of our work was to test whether thymosin beta 4 protected endothelial progenitor cells against apoptosis induced by advanced glycation endproducts and investigate the underlying mechanism. Treatment with thymosin beta 4 or transfection with microRNA-34a inhibitor enhanced cell viability, reduced apoptosis, abated oxidative stress, and attenuated mitochondrial dysfunction in endothelial progenitor cells exposed to advanced glycation endproducts. Incubation with advanced glycation endproducts led to increased levels of microRNA-34a, which was attenuated by treatment with thymosin beta 4. Transfection with microRNA-34a reversed the beneficial effect of thymosin beta 4 against injuries induced by advanced glycation endproducts. The microRNA-34a could directly bind to the 3'UTRs of the mRNA of B-cell lymphoma 2, and thymosin beta 4 treatment upregulated B-cell lymphoma 2 expression in endothelial progenitor cells exposed to advanced glycation endproducts. More importantly, knockdown of B-cell lymphoma 2 abolished the protection of thymosin beta 4 and microRNA-34a inhibitor against advanced glycation endproducts. In conclusion, inhibition of microRNA-34a mediated protection of thymosin beta 4 in endothelial progenitor cells against advanced glycation endproducts by targeting B-cell lymphoma 2, which was helpful for understanding the therapeutic potential of thymosin beta 4 for diabetic patients.
Collapse
Affiliation(s)
- Qi Chen
- Department of Cardiology, Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhida Shen
- Department of Cardiology, Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yanjun Mao
- Shanghai Pulmonary Hospital, Shanghai, China
| | - Qinfeng Li
- Department of Cardiology, Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yu Liu
- Department of Cardiology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, China
| | - Menghan Mei
- Department of Blood Transfusion, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Fuyu Qiu
- Department of Cardiology, Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Meihui Wang
- Department of Cardiology, Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
20
|
Suliman I, Abdelgelil N, Kassamali F, Hassanein TI. The Effects of Hepatic Steatosis on the Natural History of HBV Infection. Clin Liver Dis 2019; 23:433-450. [PMID: 31266618 DOI: 10.1016/j.cld.2019.05.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Fatty liver prevalence is increasing and becoming a global health burden. Chronic hepatitis B infection (CHB) is one of the most common chronic viral infections. Steatosis in CHB patients increases risk of cirrhosis and hepatocellular carcinoma. Data from studies on the interaction between CHB and nonalcoholic fatty liver disease are not conclusive. Liver biopsy is the gold standard for diagnosis of fatty liver; however, noninvasive diagnostic tests have been developed to diagnose and predict fibrosis in CHB/NAFLD. Treatment guidelines are not clear.
Collapse
Affiliation(s)
- Idrees Suliman
- Blake Medical Center Internal Medicine, 2020 59th St W, Bradenton, FL 34209, USA
| | - Noha Abdelgelil
- Southern California Research Center, 131 Orange Avenue, Suite 101, Coronado, CA 92118, USA
| | - Farah Kassamali
- St. Mary's Medical Center, 450 Stanyan St, San Francisco, CA 94117, USA
| | - Tarek I Hassanein
- Southern California Liver Centers, 131 Orange Avenue, Suite 101, Coronado, CA 92118, USA.
| |
Collapse
|
21
|
Kassem KM, Vaid S, Peng H, Sarkar S, Rhaleb NE. Tβ4-Ac-SDKP pathway: Any relevance for the cardiovascular system? Can J Physiol Pharmacol 2019; 97:589-599. [PMID: 30854877 PMCID: PMC6824425 DOI: 10.1139/cjpp-2018-0570] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The last 20 years witnessed the emergence of the thymosin β4 (Tβ4)-N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP) pathway as a new source of future therapeutic tools to treat cardiovascular and renal diseases. In this review article, we attempted to shed light on the numerous experimental findings pertaining to the many promising cardiovascular therapeutic avenues for Tβ4 and (or) its N-terminal derivative, Ac-SDKP. Specifically, Ac-SDKP is endogenously produced from the 43-amino acid Tβ4 by 2 successive enzymes, meprin α and prolyl oligopeptidase. We also discussed the possible mechanisms involved in the Tβ4-Ac-SDKP-associated cardiovascular biological effects. In infarcted myocardium, Tβ4 and Ac-SDKP facilitate cardiac repair after infarction by promoting endothelial cell migration and myocyte survival. Additionally, Tβ4 and Ac-SDKP have antifibrotic and anti-inflammatory properties in the arteries, heart, lungs, and kidneys, and stimulate both in vitro and in vivo angiogenesis. The effects of Tβ4 can be mediated directly through a putative receptor (Ku80) or via its enzymatically released N-terminal derivative Ac-SDKP. Despite the localization and characterization of Ac-SDKP binding sites in myocardium, more studies are needed to fully identify and clone Ac-SDKP receptors. It remains promising that Ac-SDKP or its degradation-resistant analogs could serve as new therapeutic tools to treat cardiac, vascular, and renal injury and dysfunction to be used alone or in combination with the already established pharmacotherapy for cardiovascular diseases.
Collapse
Affiliation(s)
- Kamal M Kassem
- a Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA
- b Internal Medicine Department, University of Cincinnati Medical Center, Cincinnati, OH 45219, USA
| | - Sonal Vaid
- a Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA
- c Internal Medicine Department, St. Vincent Indianapolis Hospital, Indianapolis, IN 46260, USA
| | - Hongmei Peng
- a Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA
| | - Sarah Sarkar
- a Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA
| | - Nour-Eddine Rhaleb
- a Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA
- d Department of Physiology, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
22
|
Chen W, Yan F, Qin S, Dong H. Molecular cloning, expression analysis, and the immune-related role of a thymosin β in the goldfish, Carassius auratus. FISH PHYSIOLOGY AND BIOCHEMISTRY 2019; 45:427-437. [PMID: 30361821 DOI: 10.1007/s10695-018-0574-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 09/28/2018] [Indexed: 06/08/2023]
Abstract
β-Thymosins play critical roles in the regulation of many important physiological processes, but their function in teleost fishes remains poorly understood. In this study, the full-length cDNA coding for a thymosin β (Tβ) was cloned and identified in goldfish, Carassius auratus (gfTβ). The gfTβ cDNA consisted of 653 bp with an open reading frame of 135 bp that encodes a 44 amino acid polypeptide. Sequence analysis revealed one thymosin domain and a highly conserved actin-binding motif (18LKKTET23). Expression of gfTβ transcript was detected ubiquitously in all tissues examined, with relatively higher levels in the brain, intestine, spleen, gill, skin, kidney, and testis. Cadmium and H2O2 exposure induced increases in gfTβ transcript levels in the liver and spleen. Moreover, gfTβ transcription was upregulated in response to LPS challenge in the spleen while Poly I:C treatment did not affect gfTβ expression. In vivo injection of recombinant gfTβ generated from an Escherichia coli system induced expression of T lymphocyte-related genes (RAG1 and CD8α). These results suggest that gfTβ may be involved in the immune response of teleost fishes via modulation of T lymphocyte development.
Collapse
Affiliation(s)
- Wenbo Chen
- Department of Biology, Institute of Resources and Environment, Henan Polytechnic University, Jiaozuo, 454000, Henan, China.
| | - Fangfang Yan
- Department of Biology, Institute of Resources and Environment, Henan Polytechnic University, Jiaozuo, 454000, Henan, China
| | - Shaozong Qin
- Department of Biology, Institute of Resources and Environment, Henan Polytechnic University, Jiaozuo, 454000, Henan, China
| | - Haiyan Dong
- Department of Medicine, Huzhou University, Huzhou, 313000, Zhejiang, China.
| |
Collapse
|
23
|
Renga G, Oikonomou V, Stincardini C, Pariano M, Borghi M, Costantini C, Bartoli A, Garaci E, Goldstein AL, Romani L. Thymosin β4 limits inflammation through autophagy. Expert Opin Biol Ther 2019; 18:171-175. [PMID: 30063848 DOI: 10.1080/14712598.2018.1473854] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Thymosin β4 (Tβ4) is a thymic hormone with multiple and different intracellular and extracellular activities affecting wound healing, inflammation, fibrosis and tissue regeneration. As the failure to resolve inflammation leads to uncontrolled inflammatory pathology which underlies many chronic diseases, the endogenous pathway through which Tβ4 may promote inflammation resolution becomes of great interest. In this review, we discuss data highlighting the efficacy of Tβ4 in resolving inflammation by restoring autophagy. AREAS COVERED The authors provide an overview of the Tβ4's anti-inflammatory properties in several pathologies and provide preliminary evidence on the ability of Tβ4 to resolve inflammation via the promotion of non-canonical autophagy associated with the activation of the DAP kinase anti-inflammatory function. EXPERT OPINION Based on its multitasking activity in various animal studies, including tissue repair and prevention of chronic inflammation, Tβ4 may represent a potential, novel treatment for inflammatory diseases associated with defective autophagy.
Collapse
Affiliation(s)
- Giorgia Renga
- a Department of Experimental Medicine , University of Perugia , Perugia , Italy
| | - Vasilis Oikonomou
- a Department of Experimental Medicine , University of Perugia , Perugia , Italy
| | - Claudia Stincardini
- a Department of Experimental Medicine , University of Perugia , Perugia , Italy
| | - Marilena Pariano
- a Department of Experimental Medicine , University of Perugia , Perugia , Italy
| | - Monica Borghi
- a Department of Experimental Medicine , University of Perugia , Perugia , Italy
| | - Claudio Costantini
- a Department of Experimental Medicine , University of Perugia , Perugia , Italy
| | - Andrea Bartoli
- a Department of Experimental Medicine , University of Perugia , Perugia , Italy
| | - Enrico Garaci
- b University San Raffaele and IRCCS San Raffaele , Rome , Italy
| | - Allan L Goldstein
- c Department of Biochemistry and Molecular Medicine , the George Washington University, School of Medicine and Health Sciences , Washington , DC , USA
| | - Luigina Romani
- a Department of Experimental Medicine , University of Perugia , Perugia , Italy
| |
Collapse
|
24
|
Gupta S, Li L. The role of Thymosin β4 in angiotensin II-induced cardiomyocytes growth. Expert Opin Biol Ther 2019; 18:105-110. [PMID: 30063846 DOI: 10.1080/14712598.2018.1494718] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
INTRODUCTION Thymosin beta-4 (Tβ4) is an actin sequestering protein and is furthermore involved in diverse biological processes including cell proliferation, differentiation, wound healing, stem- or progenitor cell differentiation, and modulates inflammatory mediators. Tβ4 also attenuates fibrosis. However, the role of Tβ4 in cardiomyocytes hypertrophy is unknown. AREAS COVERED In this review, we will discuss the role of Tβ4 in cardiac remodeling that specifically includes cardiac hypertrophy and fibrosis only. Our review will further cover a new signaling pathway, the wingless and integrated-1 (Wnt) pathway in cardiac remodeling. In rat neonatal and adult cardiomyocytes stimulated with angiotensin II (Ang II), we showed that Tβ4 has the ability to reduce cell sizes, attenuate hypertrophy marker genes expression, along with a panel of WNT-associated gene expressions induced by Ang II. Selected target gene WNT1-inducible-signaling pathway protein 1 (WISP-1) was identified by Tβ4. Data further confirmed that WISP-1 overexpression promoted cardiomyocytes growth and was reversed by Tβ4 pretreatment. EXPERT OPINION Our data suggested that Tβ4 protects cardiomyocytes from hypertrophic response by targeting WISP-1. The new role of Tβ4 in cardiac hypertrophy advances our understanding, and the mechanism of action of Tβ4 may provide a solid foundation for the treatment of cardiac disease.
Collapse
Affiliation(s)
- Sudhiranjan Gupta
- a Department of Medical Physiology , Texas A&M University; Central Texas Veterans Health Care System , Temple , TX , USA
| | - Li Li
- a Department of Medical Physiology , Texas A&M University; Central Texas Veterans Health Care System , Temple , TX , USA
| |
Collapse
|
25
|
Hao Q, He L, Zhou J, Yuan Y, Ma X, Pang Z, Li W, Zhang Y, Zhang W, Zhang C, Li M. A dimeric thymosin beta 4 with novel bio-activity protects post-ischemic cardiac function by accelerating vascular endothelial cell proliferation. Int J Cardiol 2018; 261:146-154. [PMID: 29550018 DOI: 10.1016/j.ijcard.2018.03.052] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 02/24/2018] [Accepted: 03/12/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Thymosin beta 4 (Tβ4) is a 43-amino-acid peptide with protective properties in myocardium injury. Previously, we produced a recombinant human dimeric Tβ4 (DTβ4). Here, the cardioprotective effects of DTβ4 and the molecular mechanisms underlying its enhanced activity were investigated. METHODS AND RESULTS Echocardiography measurements showed that the cardioprotective effect of DTβ4 in myocardial infarction mice was significantly higher than that of wild-type Tβ4. Corresponding in vitro analyses demonstrated that the enhanced cardioprotection provided by DTβ4 was largely due to increased stimulation of angiogenesis. HPLC analysis, western blotting and qRT-PCR indicated that the enhanced pro-angiogenesis activity of DTβ4 was independent of the protein half-life and the known downstream pathways of wild-type Tβ4. Transcriptome deep sequencing (RNA-seq), BrdU incorporation assays, flow cytometry analysis and RNA interference demonstrated that the enhanced angiogenic activity of DTβ4 depended on MALAT1 (metastasis-associated lung adenocarcinoma transcript 1)-induced proliferation of vascular endothelial cells, which has not been reported for wild-type Tβ4. Moreover, transcription factor activation screening, luciferase promoter reporter assay and immunoprecipitation assay demonstrated that DTβ4 enhanced MALAT1 transcription by inhibiting the degradation of prospero-related homeobox 1 (PROX1). CONCLUSION This study demonstrates the potential applications and the novel bioactivity of the Tβ4 dimer. Moreover, to construct the dimer represents a new method for production of bioactive peptides that may have novel activities.
Collapse
Affiliation(s)
- Qiang Hao
- Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xian, China
| | - Lei He
- Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xian, China
| | - Jiming Zhou
- Department of Cardiology, 153 Central Hospital of People's Liberation Army, Zhengzhou, China; Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xian, China
| | - Yuan Yuan
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xian, China
| | - Xiaowen Ma
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xian, China
| | - Zhijun Pang
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xian, China
| | - Weina Li
- Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xian, China
| | - Yingqi Zhang
- Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xian, China
| | - Wei Zhang
- Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xian, China.
| | - Cun Zhang
- Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xian, China.
| | - Meng Li
- Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xian, China.
| |
Collapse
|
26
|
Abstract
Background Cardiac fibrosis occurs because of disruption of the extracellular matrix network leading to myocardial dysfunction. Angiotensin II has been implicated in the development of cardiac fibrosis. Recently, microRNAs have been identified as an attractive target for therapeutic intervention in cardiac pathologies; however, the underlying mechanism of microRNAs in cardiac fibrosis remains unclear. MicroRNA‐130a (miR‐130a) has been shown to participate in angiogenesis and cardiac arrhythmia; however, its role in cardiac fibrosis is unknown. Methods and Results In this study, we found that miR‐130a was significantly upregulated in angiotensin II‐infused mice. The in vivo inhibition of miR‐130a by locked nucleic acid– based anti‐miR‐130a in mice significantly reduced angiotensin II‐induced cardiac fibrosis. Upregulation of miR‐130a was confirmed in failing human hearts. Overexpressing miR‐130a in cardiac fibroblasts promoted profibrotic gene expression and myofibroblasts differentiation, and the inhibition of miR‐130a reversed the processes. Using the constitutive and dominant negative constructs of peroxisome proliferator‐activated receptor γ 3‐′untranslated region (UTR), data revealed that the protective mechanism was associated with restoration of peroxisome proliferator‐activated receptor γ level leading to the inhibition of angiotensin II‐induced cardiac fibrosis. Conclusions Our findings provide evidence that miR‐130a plays a critical role in cardiac fibrosis by directly targeting peroxisome proliferator‐activated receptor γ. We conclude that inhibition of miR‐130a would be a promising strategy for the treatment of cardiac fibrosis.
Collapse
Affiliation(s)
- Li Li
- Department of Medical Physiology, Texas A & M Health Science Center, Central Texas Veterans Health Care System, Temple, TX.,Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | - Kelsey R Bounds
- Division of Nephrology and Hypertension, Department of Internal Medicine, Baylor Scott White Health, Temple, TX
| | - Piyali Chatterjee
- Division of Nephrology and Hypertension, Department of Internal Medicine, Baylor Scott White Health, Temple, TX
| | - Sudhiranjan Gupta
- Department of Medical Physiology, Texas A & M Health Science Center, Central Texas Veterans Health Care System, Temple, TX
| |
Collapse
|
27
|
Yuan J, Shen Y, Yang X, Xie Y, Lin X, Zeng W, Zhao Y, Tian M, Zha Y. Thymosin β4 alleviates renal fibrosis and tubular cell apoptosis through TGF-β pathway inhibition in UUO rat models. BMC Nephrol 2017; 18:314. [PMID: 29047363 PMCID: PMC5648500 DOI: 10.1186/s12882-017-0708-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 08/24/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Thymosin β4 (Tβ4) is closely associated with the cytoskeleton, inflammation, wound healing, angiogenesis, apoptosis, and myocardial regeneration, but the effects of Tβ4 treatment on chronic renal tubular interstitial fibrosis (CRTIF) are poorly known. This study aimed to examine the effects of Tβ4 on the renal apoptosis and the expression of transforming growth factor (TGF-β), E-cadherin, and α-smooth muscle actin (α-SMA) in CRTIF rat models. METHODS Male SD rats were randomized into four groups (sham group, unilateral ureteral obstruction (UUO) group, UUO + low-dose Tβ4 group, and UUO + high-dose Tβ4 group). The pathological changes of kidney tissue and its function were assessed two weeks after UUO. In renal interstitial tissue,TGF-β, E-cadherin and α-SMA expression was detected by western blot. In tubular epithelial cells, E-cadherin and α-SMA expression was detected using Real-time qPCR and western blot. Cell apoptosis of rat renal interstitial tissue and tubular epithelial cells was evaluated by immunofluorescence and western blot. RESULTS Two weeks after UUO, no differences in blood urea nitrogen and creatinine were observed between the four groups (P > 0.05). Compared to the UUO group, Tβ4 treatment decreased the 24-h proteinuria (P < 0.001) and reduced the area of pathological change (P < 0.01); this effect was more apparent in the UUO + high-dose Tβ4 group. Compared to the UUO group, a significant decrease in TGF-β and α-SMA protein expression was observed in the high-dose Tβ4 group. The level of E-cadherin protein was lower in the UUO group than the Tβ4 groups, and high-dose Tβ4 treatment further increased E-cadherin expression and improved cell apoptosis in the renal interstitial tissue. Analysis of in vitro tubular epithelial cells showed that α-SMA mRNA and protein expression decreased, while E-cadherin mRNA and protein expression increased by Tβ4 treatment. Similarly, these changes were more significant in the UUO + high-dose Tβ4 group. Tβ4 treatment improved the apoptosis of In vitro tubular epithelial cells compared with pure TGF-β stimulation, and equally, the decrease of apoptosis was more apparent in the TGF-β + high-dose Tβ4 group. CONCLUSIONS Tβ4 treatment might alleviate the renal fibrosis and apoptosis of tubular epithelial cells through TGF-β pathway inhibition in UUO rats with CRTIF.
Collapse
Affiliation(s)
- Jing Yuan
- Department of Nephrology Guizhou Provincial People’s Hospital, Guiyang, 550002 China
| | - Yan Shen
- Department of Nephrology Guizhou Provincial People’s Hospital, Guiyang, 550002 China
| | - Xia Yang
- Department of Nephrology Guizhou Provincial People’s Hospital, Guiyang, 550002 China
| | - Ying Xie
- Department of Nephrology Guizhou Provincial People’s Hospital, Guiyang, 550002 China
| | - Xin Lin
- Department of Nephrology Guizhou Provincial People’s Hospital, Guiyang, 550002 China
| | - Wen Zeng
- Department of Nephrology Guizhou Provincial People’s Hospital, Guiyang, 550002 China
| | - Yingting Zhao
- Department of Nephrology Guizhou Provincial People’s Hospital, Guiyang, 550002 China
| | - Maolu Tian
- Department of Nephrology Guizhou Provincial People’s Hospital, Guiyang, 550002 China
| | - Yan Zha
- Department of Nephrology Guizhou Provincial People’s Hospital, Guiyang, 550002 China
| |
Collapse
|
28
|
Gorabi AM, Tafti SHA, Soleimani M, Panahi Y, Sahebkar A. Cells, Scaffolds and Their Interactions in Myocardial Tissue Regeneration. J Cell Biochem 2017; 118:2454-2462. [DOI: 10.1002/jcb.25912] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 01/25/2017] [Indexed: 01/08/2023]
Affiliation(s)
| | | | - Masoud Soleimani
- Faculty of Medical Sciences; Hematology Department; Tarbiat Modarres University; Tehran Iran
| | - Yunes Panahi
- Chemical Injuries Research Center; Baqiyatallah University of Medical Sciences; Tehran Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center; Mashhad University of Medical Sciences; Mashhad Iran
| |
Collapse
|
29
|
Varasteh-kojourian M, Abrishamchi P, Matin MM, Asili J, Ejtehadi H, Khosravitabar F. Antioxidant, cytotoxic and DNA protective properties of Achillea eriophora DC. and Achillea biebersteinii Afan. extracts: A comparative study. AVICENNA JOURNAL OF PHYTOMEDICINE 2017; 7:157-168. [PMID: 28348971 PMCID: PMC5355821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 02/15/2016] [Accepted: 05/08/2016] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Achillea is a traditional medicinal herb which contains different phenol and flavonoid compounds that are responsible for Achillea pharmacological effects. We aimed to determine phenol and flavonoid contents, besides antioxidant activities of different extracts from Achillea eriophoraa (A. eriophora) DC. and Achillea biebersteinii (A. biebersteinii) Afan. (endemic species in Iran) and to investigate their effects on human cells. MATERIALS AND METHODS Achillea extracts, were prepared by maceration and shaking methods, from different parts (aerial parts, stem, leaves and inflorescence) of two species using methanol and ethanol as solvents. Total phenol and flavonoid contents were measured by spectrophotometry, and antioxidant activities of the extracts were determined by DPPH radical scavenging, BCB and TBARS assays. Cytotoxicity and antioxidant activities of the extracts were investigated in Human Foreskin Fibroblast (HFF3) cells using MTT, comet and H2O2 assays. RESULTS Methanol extracts of A. biebersteinii prepared from leaves and inflorescence by maceration method exhibited maximum phenol (1657.58 ± 36.45 mg GAE/100 g DW) and flavonoid (264.00 ± 62.16 mg QUE/100 g DW) contents. Leaf methanol extract showed significantly higher antioxidant activity (0.0276 ± 0.003, 0.16 ± 0.016 and 13.96 ± 0.26 mg/ml for DPPH, BCB and TBARS IC50s, respectively) than those of the other extracts. Leaf extract of A. biebersteinii was not cytotoxic even at the highest examined dose (512 µg/ml) and inhibited cell toxicity induced by H2O2 (98% viability for the cells pretreated with plant extract in the presence of H2O2). Comet assay also confirmed high DNA protective activity of leaf extracts. CONCLUSION Achillea extracts possess remarkable antioxidant activity, and could be good natural alternatives to synthetic antioxidants in pharmaceutical and food industries.
Collapse
Affiliation(s)
| | - Parvaneh Abrishamchi
- Department of Biology, Faculty of Sciences, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Maryam M. Matin
- Department of Biology, Faculty of Sciences, Ferdowsi University of Mashhad, Mashhad, Iran
- Cell and Molecular Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Javad Asili
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Ejtehadi
- Department of Biology, Faculty of Sciences, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Fatemeh Khosravitabar
- Department of Biology, Faculty of Sciences, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
30
|
Karbiener M, Darnhofer B, Frisch MT, Rinner B, Birner-Gruenberger R, Gugatschka M. Comparative proteomics of paired vocal fold and oral mucosa fibroblasts. J Proteomics 2017; 155:11-21. [PMID: 28099887 PMCID: PMC5389448 DOI: 10.1016/j.jprot.2017.01.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/22/2016] [Accepted: 01/12/2017] [Indexed: 12/12/2022]
Abstract
Injuries of the vocal folds frequently heal with scar formation, which can have lifelong detrimental impact on voice quality. Current treatments to prevent or resolve scars of the vocal fold mucosa are highly unsatisfactory. In contrast, the adjacent oral mucosa is mostly resistant to scarring. These differences in healing tendency might relate to distinct properties of the fibroblasts populating oral and vocal fold mucosae. We thus established the in vitro cultivation of paired, near-primary vocal fold fibroblasts (VFF) and oral mucosa fibroblasts (OMF) to perform a basic cellular characterization and comparative cellular proteomics. VFF were significantly larger than OMF, proliferated more slowly, and exhibited a sustained TGF-β1-induced elevation of pro-fibrotic interleukin 6. Cluster analysis of the proteomic data revealed distinct protein repertoires specific for VFF and OMF. Further, VFF displayed a broader protein spectrum, particularly a more sophisticated array of factors constituting and modifying the extracellular matrix. Conversely, subsets of OMF-enriched proteins were linked to cellular proliferation, nuclear events, and protection against oxidative stress. Altogether, this study supports the notion that fibroblasts sensitively adapt to the functional peculiarities of their respective anatomical location and presents several molecular targets for further investigation in the context of vocal fold wound healing. BIOLOGICAL SIGNIFICANCE Mammalian vocal folds are a unique but delicate tissue. A considerable fraction of people is affected by voice problems, yet many of the underlying vocal fold pathologies are sparsely understood at the molecular level. One such pathology is vocal fold scarring - the tendency of vocal fold injuries to heal with scar formation -, which represents a clinical problem with highly suboptimal treatment modalities. This study employed proteomics to obtain comprehensive insight into the protein repertoire of vocal fold fibroblasts, which are the cells that predominantly synthesize the extracellular matrix in both physiological and pathophysiological conditions. Protein profiles were compared to paired fibroblasts from the oral mucosa, a neighboring tissue that is remarkably resistant to scarring. Bioinformatic analyses of the data revealed a number of pathways as well as single proteins (e.g. ECM-remodeling factors, transcription factors, enzymes) that were significantly different between the two fibroblast types. Thereby, this study has revealed novel interesting molecular targets which can be analyzed in the future for their impact on vocal fold wound healing.
Collapse
Affiliation(s)
- Michael Karbiener
- Department of Phoniatrics, ENT University Hospital, Medical University of Graz, Austria.
| | - Barbara Darnhofer
- Research Unit, Functional Proteomics and Metabolic Pathways, Institute of Pathology, Medical University of Graz, Austria; Omics Center Graz, BioTechMed-Graz, Austria; Austrian Centre of Industrial Biotechnology (ACIB), Austria
| | - Marie-Therese Frisch
- Core Facility Alternative Biomodels und Preclinical Imaging, Division of Biomedical Research, Medical University of Graz, Austria
| | - Beate Rinner
- Core Facility Alternative Biomodels und Preclinical Imaging, Division of Biomedical Research, Medical University of Graz, Austria
| | - Ruth Birner-Gruenberger
- Research Unit, Functional Proteomics and Metabolic Pathways, Institute of Pathology, Medical University of Graz, Austria; Omics Center Graz, BioTechMed-Graz, Austria; Austrian Centre of Industrial Biotechnology (ACIB), Austria
| | - Markus Gugatschka
- Department of Phoniatrics, ENT University Hospital, Medical University of Graz, Austria
| |
Collapse
|
31
|
Zheng XY, Lv YF, Li S, Li Q, Zhang QN, Zhang XT, Hao ZM. Recombinant adeno-associated virus carrying thymosin β 4 suppresses experimental colitis in mice. World J Gastroenterol 2017; 23:242-255. [PMID: 28127198 PMCID: PMC5236504 DOI: 10.3748/wjg.v23.i2.242] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 10/04/2016] [Accepted: 11/13/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the protective effect of a recombinant adeno-associated virus carrying thymosin β4 (AAV-Tβ4) on murine colitis via intracolonic administration.
METHODS AAV-Tβ4 was prepared and intracolonically used to mediate the secretory expression of Tβ4 in mouse colons. Dextran sulfate sodium (DSS) was applied to induce the murine ulcerative colitis, and 2,4,6-trinitrobenzene sulfonic acid (TNBS) was used to establish a mouse colitis model resembling Crohn’s disease. The disease severity and colon injuries were observed and graded to reveal the effects of AAV-Tβ4 on colitis. The activities of myeloperoxidase (MPO) and superoxide dismutase (SOD) and the content of malondialdehyde (MDA) were determined using biochemical assays. Colonic levels of tumor necrosis factor-α (TNF-α), interleukin (IL)-1β and IL-10 were measured using ELISA, and mucosal epithelial cell apoptosis and proliferation were detected by TUNEL assay and immunochemistry, respectively.
RESULTS Recombinant AAVs efficiently delivered LacZ and Tβ4 into the colonic tissues of the mice, and AAV-Tβ4 led to a strong expression of Tβ4 in mouse colons. In both the DSS and TNBS colitis models, AAV-Tβ4-treated mice displayed distinctly attenuated colon injuries and reduced apoptosis rate of colonic mucosal epithelia. AAV-Tβ4 significantly reduced inflammatory cell infiltrations and relieved oxidative stress in the inflamed colons of the mice, as evidenced by decreases in MPO activity and MDA content and increases in SOD activity. AAV-Tβ4 also modulated colonic TNF-α, IL-1β and IL-10 levels and suppressed the compensatory proliferation of colonic epithelial cells in DSS- and TNBS-treated mice.
CONCLUSION Tβ4 exerts a protective effect on murine colitis, indicating that AAV-Tβ4 could potentially be developed into a promising agent for the therapy of inflammatory bowel diseases.
Collapse
|
32
|
Liang J, Cai W, Han T, Jing L, Ma Z, Gao Y. The expression of thymosin β4 in chronic hepatitis B combined nonalcoholic fatty liver disease. Medicine (Baltimore) 2016; 95:e5763. [PMID: 28033294 PMCID: PMC5207590 DOI: 10.1097/md.0000000000005763] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 11/12/2016] [Accepted: 11/30/2016] [Indexed: 12/16/2022] Open
Abstract
The aim of the study was to detect the expression level of thymosin β4 (Tβ4) in serum and tissues of patients with chronic hepatitis B (CHB) combined nonalcoholic fatty liver disease (NAFLD). The effects of Tβ4 in hepatic steatosis, chronic inflammation, and fibrosis development in CHB combined NAFLD patients were also discussed. The study included 46 patients in the case group with CHB and NAFLD and 42 patients in the control group with CHB. ELISA was applied to detect serum Tβ4 and TNF-α level. Furthermore, the correlation analysis of Tβ4 levels with biochemical index, pathological index, and TNF-α level was performed. The Tβ4 immunohistochemical levels of different inflammation fibrosis levels were compared, and the correlation analysis with TNF expression was performed. The Tβ4 levels in patients with CHB combined NAFLD showed no statistical difference when compared to the control group. In patients with CHB combined NAFLD group, the Tβ4 level had no correlation with ALT, AST, TG, FGP, hepatitis B virus (HBV)-DNA levels, and fat grading, but had negative correlation with inflammation score and fibrosis score (P <0.01). The immunohistochemical results of hepatic tissues showed that the expression intensity of severe inflammation fibrosis group had statistical significance compared with that of slight group, and the Tβ4 expression both in serum and in liver tissue negatively correlated with TNF-α expression. Tβ4 could be involved in the regulation of chronic inflammation and fibrosis and plays a defense role in the disease progression of CHB combined NAFLD patients.
Collapse
Affiliation(s)
- Jing Liang
- Department of Gastroenterology and Hepatology, Tianjin Third Central Hospital
- Tianjin Key Laboratory of Artificial Cell
- Artificial Cell Engineering Technology Research Center of Public Health Ministry, Tianjin, China
| | - Wenjuan Cai
- Department of Pathology, Tianjin First Central Hospital
| | - Tao Han
- Department of Gastroenterology and Hepatology, Tianjin Third Central Hospital
| | - Li Jing
- Molecular Biology Laboratory, Tianjin Third Central Hospital
| | - Zhe Ma
- Department of Pathology, Tianjin Third Central Hospital
| | - Yingtang Gao
- Molecular Biology Laboratory, Tianjin Third Central Hospital
| |
Collapse
|
33
|
Xiang MSW, Kikuchi K. Endogenous Mechanisms of Cardiac Regeneration. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 326:67-131. [PMID: 27572127 DOI: 10.1016/bs.ircmb.2016.04.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Zebrafish possess a remarkable capacity for cardiac regeneration throughout their lifetime, providing a model for investigating endogenous cellular and molecular mechanisms regulating myocardial regeneration. By contrast, adult mammals have an extremely limited capacity for cardiac regeneration, contributing to mortality and morbidity from cardiac diseases such as myocardial infarction and heart failure. However, the viewpoint of the mammalian heart as a postmitotic organ was recently revised based on findings that the mammalian heart contains multiple undifferentiated cell types with cardiogenic potential as well as a robust regenerative capacity during a short period early in life. Although it occurs at an extremely low level, continuous cardiomyocyte turnover has been detected in adult mouse and human hearts, which could potentially be enhanced to restore lost myocardium in damaged human hearts. This review summarizes and discusses recent advances in the understanding of endogenous mechanisms of cardiac regeneration.
Collapse
Affiliation(s)
- M S W Xiang
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst NSW, Australia
| | - K Kikuchi
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst NSW, Australia; St. Vincent's Clinical School, University of New South Wales, Kensington NSW, Australia.
| |
Collapse
|
34
|
Abstract
Treatment with thymosin beta 4 (Tβ4) reduces infarct volume and preserves cardiac function in preclinical models of cardiac ischemic injury. These effects stem in part from decreased infarct size, but additional benefits are likely due to specific antifibrotic and proangiogenic activities. Injected or transgenic Tβ4 increase blood vessel growth in large and small animal models, consistent with Tβ4 converting hibernating myocardium to an actively contractile state following ischemia. Tβ4 and its degradation products have antifibrotic effects in in vitro assays and in animal models of fibrosis not related to cardiac injury. This large number of pleiotropic effects results from Tβ4's many interactions with cellular signaling pathways, particularly indirect regulation of cellular motility and movement via the SRF-MRTF-G-actin transcriptional pathway. Variation in effects and effect sizes in animal models may potentially be due to variable distribution of Tβ4. Preclinical studies of PK/PD relationships and a reliable pharmacodynamic biomarker would facilitate clinical development of Tβ4.
Collapse
Affiliation(s)
- G T Pipes
- Cardiovascular Drug Discovery, Discovery Biology Research & Development, Bristol-Myers Squibb, Pennington, NJ, United States.
| | - J Yang
- Cardiovascular Drug Discovery, Discovery Biology Research & Development, Bristol-Myers Squibb, Pennington, NJ, United States
| |
Collapse
|
35
|
Kim J, Jung Y. Thymosin Beta 4 Is a Potential Regulator of Hepatic Stellate Cells. VITAMINS AND HORMONES 2016; 102:121-149. [PMID: 27450733 DOI: 10.1016/bs.vh.2016.04.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Liver fibrosis, a major characteristic of chronic liver disease, is inappropriate tissue remodeling caused by prolonged parenchymal cell injury and inflammation. During liver injury, hepatic stellate cells (HSCs) undergo transdifferentiation from quiescent HSCs into activated HSCs, which promote the deposition of extracellular matrix proteins, leading to liver fibrosis. Thymosin beta 4 (Tβ4), a major actin-sequestering protein, is the most abundant member of the highly conserved β-thymosin family and controls cell morphogenesis and motility by regulating the dynamics of the actin cytoskeleton. Tβ4 is known to be involved in various cellular responses, including antiinflammation, wound healing, angiogenesis, and cancer progression. Emerging evidence suggests that Tβ4 is expressed in the liver; however, its biological roles are poorly understood. Herein, we introduce liver fibrogenesis and recent findings regarding the function of Tβ4 in various tissues and discuss the potential role of Tβ4 in liver fibrosis with a special focus on the effects of exogenous and endogenous Tβ4. Recent studies have revealed that activated HSCs express Tβ4 in vivo and in vitro. Treatment with the exogenous Tβ4 peptide inhibits the proliferation and migration of activated HSCs and reduces liver fibrosis, indicating it has an antifibrotic action. Meanwhile, the endogenously expressed Tβ4 in activated HSCs is shown to promote HSCs activation. Although the role of Tβ4 has not been elucidated, it is apparent that Tβ4 is associated with HSC activation. Therefore, understanding the potential roles and regulatory mechanisms of Tβ4 in liver fibrosis may provide a novel treatment for patients.
Collapse
Affiliation(s)
- J Kim
- Pusan National University, Pusan, Republic of Korea
| | - Y Jung
- Pusan National University, Pusan, Republic of Korea.
| |
Collapse
|
36
|
Abstract
No agent has been identified that significantly accelerates the repair of chronic dermal wounds in humans. Thymosin beta 4 (Tβ4) is a small, abundant, naturally occurring regenerative protein that is found in body fluids and inside cells. It was found to have angiogenic and antiinflammatory activity and to be high in platelets that aggregate at the wound site. Thus we used Tβ4 initially in dermal healing. It has since been shown to have many activities important in tissue protection, repair, and regeneration. Tβ4 increases the rate of dermal healing in various preclinical animal models, including diabetic and aged animals, and is active for burns as well. Tβ4 also accelerated the rate of repair in phase 2 trials with patients having pressure ulcers, stasis ulcers, and epidermolysis bullosa wounds. It is safe and well tolerated and will likely have additional uses in the skin and in injured organs for tissue repair and regeneration.
Collapse
Affiliation(s)
- H K Kleinman
- George Washington University, Washington, DC, United States.
| | - G Sosne
- Kresge Eye Institute, Wayne State University School of Medicine, Detroit, MI, United States
| |
Collapse
|
37
|
Li J, Zhang Y, Liu Y, Zhang Y, Xiang Z, Qu F, Yu Z. A thymosin beta-4 is involved in production of hemocytes and immune defense of Hong Kong oyster, Crassostrea hongkongensis. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 57:1-9. [PMID: 26695126 DOI: 10.1016/j.dci.2015.12.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 12/10/2015] [Accepted: 12/11/2015] [Indexed: 06/05/2023]
Abstract
Thymosin beta-4 (Tβ4) is a ubiquitous protein with multiple and diverse intracellular and extracellular functions in vertebrates. In this study, the full-length cDNA of Tβ4 was cloned and identified in Crassostrea hongkongensis, designated as ChTβ4. The full-length cDNA of ChTβ4 consists of 530 bp with an open reading frame of 126 bp encoding a 41 amino acid polypeptide. SMART analysis indicated that there is one thymosin domain and a highly conserved actin-binding motif (18LKKTET23) in ChTβ4. In vivo injection of recombinant ChTβ4 protein could significantly increase total hemocytes count in oysters, and knockdown of the expression of ChTβ4 resulted in a significant decrease in the circulating hemocytes. Tissue distribution analysis revealed a ubiquitous presence of ChTβ4, with the highest expression in hemocytes. The upregulated transcripts of ChTβ4 in response to bacterial challenge and tissue injury suggest that ChTβ4 is involved in both innate immunity against pathogen infection and wound healing. Moreover, bacteria-clearance experiment showed ChTβ4 could facilitate the clearance of injected bacteria in oysters. In vivo injection with ChTβ4 resulted in reduction of the intracellular ROS in hemocytes, which was associated with increased expression of antioxidant enzymes Cu/Zn superoxide dismutase (SOD), Catalase, and Glutathione Peroxidase (GPX) by pre-treatment with ChTβ4. These results suggest that ChTβ4 is a thymosin beta-4 homolog and plays a vital role in the immune defense of C. hongkongensis.
Collapse
Affiliation(s)
- Jun Li
- Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, China
| | - Yuehuan Zhang
- Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, China
| | - Ying Liu
- Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, China; University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
| | - Yang Zhang
- Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, China.
| | - Zhiming Xiang
- Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, China
| | - Fufa Qu
- Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, China
| | - Ziniu Yu
- Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, China.
| |
Collapse
|
38
|
Li L, Guleria RS, Thakur S, Zhang CL, Pan J, Baker KM, Gupta S. Thymosin β4 Prevents Angiotensin II-Induced Cardiomyocyte Growth by Regulating Wnt/WISP Signaling. J Cell Physiol 2016; 231:1737-44. [PMID: 26627308 DOI: 10.1002/jcp.25275] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 12/01/2015] [Indexed: 11/08/2022]
Abstract
Thymosin beta-4 (Tβ4) is a ubiquitous protein with many properties relating to cell proliferation and differentiation that promotes wound healing and modulates inflammatory mediators. However, the role of Tβ4 in cardiomyocyte hypertrophy is currently unknown. The purpose of this study was to determine the cardio-protective effect of Tβ4 in angiotensin II (Ang II)-induced cardiomyocyte growth. Neonatal rat ventricular cardiomyocytes (NRVM) were pretreated with Tβ4 followed by Ang II stimulation. Cell size, hypertrophy marker gene expression and Wnt signaling components, β-catenin, and Wnt-induced secreted protein-1 (WISP-1) were evaluated by quantitative real-time PCR, Western blotting and fluorescent microscopy. Pre-treatment of Tβ4 resulted in reduction of cell size, hypertrophy marker genes and Wnt-associated gene expression, and protein levels; induced by Ang II in cardiomyocyte. WISP-1 was overexpressed in NRVM and, the effect of Tβ4 in Ang II-induced cardiomyocyte growth was evaluated. WISP-1 overexpression promoted cardiomyocytes growth and was reversed by pretreatment with Tβ4. This is the first report which demonstrates that Tβ4 targets Wnt/WISP-1 to protect Ang II-induced cardiomyocyte growth. J. Cell. Physiol. 231: 1737-1744, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Li Li
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M Health Science Center, Temple, Texas.,Baylor Scott and White Health, Temple, Texas.,Central Texas Veterans Health Care System, Temple, Texas.,Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | - Rakeshwar S Guleria
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M Health Science Center, Temple, Texas.,Baylor Scott and White Health, Temple, Texas.,Central Texas Veterans Health Care System, Temple, Texas
| | - Suresh Thakur
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M Health Science Center, Temple, Texas.,Baylor Scott and White Health, Temple, Texas.,Central Texas Veterans Health Care System, Temple, Texas
| | - Cheng-Lin Zhang
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | - Jing Pan
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M Health Science Center, Temple, Texas.,Baylor Scott and White Health, Temple, Texas.,Central Texas Veterans Health Care System, Temple, Texas
| | - Kenneth M Baker
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M Health Science Center, Temple, Texas.,Baylor Scott and White Health, Temple, Texas.,Central Texas Veterans Health Care System, Temple, Texas
| | - Sudhiranjan Gupta
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M Health Science Center, Temple, Texas.,Baylor Scott and White Health, Temple, Texas.,Central Texas Veterans Health Care System, Temple, Texas
| |
Collapse
|
39
|
Lee SI, Yi JK, Bae WJ, Lee S, Cha HJ, Kim EC. Thymosin Beta-4 Suppresses Osteoclastic Differentiation and Inflammatory Responses in Human Periodontal Ligament Cells. PLoS One 2016; 11:e0146708. [PMID: 26789270 PMCID: PMC4720371 DOI: 10.1371/journal.pone.0146708] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 12/21/2015] [Indexed: 12/19/2022] Open
Abstract
Background Recent reports suggest that thymosin beta-4 (Tβ4) is a key regulator for wound healing and anti-inflammation. However, the role of Tβ4 in osteoclast differentiation remains unclear. Purpose The purpose of this study was to evaluate Tβ4 expression in H2O2-stimulated human periodontal ligament cells (PDLCs), the effects of Tβ4 activation on inflammatory response in PDLCs and osteoclastic differentiation in mouse bone marrow-derived macrophages (BMMs), and identify the underlying mechanism. Methods Reverse transcription-polymerase chain reactions and Western blot analyses were used to measure mRNA and protein levels, respectively. Osteoclastic differentiation was assessed in mouse bone marrow-derived macrophages (BMMs) using conditioned medium (CM) from H2O2-treated PDLCs. Results Tβ4 was down-regulated in H2O2-exposed PDLCs in dose- and time-dependent manners. Tβ4 activation with a Tβ4 peptide attenuated the H2O2-induced production of NO and PGE2 and up-regulated iNOS, COX-2, and osteoclastogenic cytokines (TNF-α, IL-1β, IL-6, IL-8, and IL-17) as well as reversed the effect on RANKL and OPG in PDLCs. Tβ4 peptide inhibited the effects of H2O2 on the activation of ERK and JNK MAPK, and NF-κB in PDLCs. Furthermore, Tβ4 peptide inhibited osteoclast differentiation, osteoclast-specific gene expression, and p38, ERK, and JNK phosphorylation and NF-κB activation in RANKL-stimulated BMMs. In addition, H2O2 up-regulated Wnt5a and its cell surface receptors, Frizzled and Ror2 in PDLCs. Wnt5a inhibition by Wnt5a siRNA enhanced the effects of Tβ4 on H2O2-mediated induction of pro-inflammatory cytokines and osteoclastogenic cytokines as well as helping osteoclastic differentiation whereas Wnt5a activation by Wnt5a peptide reversed it. Conclusion In conclusion, this study demonstrated, for the first time, that Tβ4 was down-regulated in ROS-stimulated PDLCs as well as Tβ4 activation exhibited anti-inflammatory effects and anti-osteoclastogenesis in vitro. Thus, Tβ4 activation might be a therapeutic target for inflammatory osteolytic disease, such as periodontitis.
Collapse
Affiliation(s)
- Sang-Im Lee
- Department of Dental Hygiene, School of Health Sciences, Dankook University, Cheonan, Republic of Korea
| | - Jin-Kyu Yi
- Department of Conservative Dentistry, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Won-Jung Bae
- Department of Oral and Maxillofacial Pathology and Research Center for Tooth and Periodontal Regeneration (MRC), School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Soojung Lee
- Department of Oral Physiology, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Hee-Jae Cha
- Department of Parasitology and Genetics, College of Medicine, Kosin University Busan, Republic of Korea
| | - Eun-Cheol Kim
- Department of Oral and Maxillofacial Pathology and Research Center for Tooth and Periodontal Regeneration (MRC), School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
40
|
Epelboin Y, Quéré C, Pernet F, Pichereau V, Corporeau C. Energy and Antioxidant Responses of Pacific Oyster Exposed to Trace Levels of Pesticides. Chem Res Toxicol 2015; 28:1831-41. [DOI: 10.1021/acs.chemrestox.5b00269] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Yanouk Epelboin
- Ifremer, UMR 6539
(CNRS, UBO, IRD, Ifremer), Laboratoire des sciences de l’Environnement
Marin (LEMAR), 29280 Plouzané, France
| | - Claudie Quéré
- Ifremer, UMR 6539
(CNRS, UBO, IRD, Ifremer), Laboratoire des sciences de l’Environnement
Marin (LEMAR), 29280 Plouzané, France
| | - Fabrice Pernet
- Ifremer, UMR 6539
(CNRS, UBO, IRD, Ifremer), Laboratoire des sciences de l’Environnement
Marin (LEMAR), 29280 Plouzané, France
| | - Vianney Pichereau
- UBO,
UMR 6539 (CNRS, UBO, IRD, Ifremer), Laboratoire des sciences de l’Environnement
Marin (LEMAR), Institut Universitaire Européen de la Mer (IUEM), Université de Brest, 29280 Plouzané, France
| | - Charlotte Corporeau
- Ifremer, UMR 6539
(CNRS, UBO, IRD, Ifremer), Laboratoire des sciences de l’Environnement
Marin (LEMAR), 29280 Plouzané, France
| |
Collapse
|
41
|
Conte E, Iemmolo M, Fruciano M, Fagone E, Gili E, Genovese T, Esposito E, Cuzzocrea S, Vancheri C. Effects of thymosin β4 and its N-terminal fragment Ac-SDKP on TGF-β-treated human lung fibroblasts and in the mouse model of bleomycin-induced lung fibrosis. Expert Opin Biol Ther 2015; 15 Suppl 1:S211-21. [PMID: 26098610 DOI: 10.1517/14712598.2015.1026804] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
UNLABELLED Thymosin β4 (Tβ4) and its amino-terminal fragment comprising N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP) have been reported to act as anti-inflammatory and anti-fibrotic agents in vitro and in vivo. In recent papers, we have shown that Tβ4 exerts a widely protective role in mice treated with bleomycin, and in particular, we have demonstrated its inhibitory effects on both inflammation and early fibrosis. OBJECTIVES In this study, the putative anti-proliferative and anti-fibrogenic effects of Tβ4 and Ac-SDKP were evaluated in vitro. In addition, the effects of Tβ4 up to 21 days were evaluated in the bleomycin mouse model of lung fibrosis. METHODS We utilized both control and TGF-β-stimulated primary human lung fibroblasts isolated from both idiopathic pulmonary fibrosis (IPF) and control tissues. The in vivo effects of Tβ4 were assessed in CD1 mice treated with bleomycin. RESULTS In the in vitro experiments, we observed significant anti-proliferative effects of Ac-SDKP in IPF fibroblasts. In those cells, Ac-SDKP significantly inhibited TGF-β-induced α-SMA and collagen expression, hallmarks of fibroblast differentiation into myofibroblasts triggered by TGF-β. In vivo, despite its previously described protective role in mice treated with bleomycin at 7 days, Tβ4 failed to prevent fibrosis induced by the drug at 14 and 21 days. CONCLUSION We conclude that, compared to Tβ4, Ac-SDKP may have greater potential as an anti-fibrotic agent in the lung. Further in vivo experiments are warranted.
Collapse
Affiliation(s)
- Enrico Conte
- University of Catania, Department of Clinical and Experimental Medicine , Via Santa Sofia 78, 95123 Catania , Italy +39 095 378 1254 ; +39 095 378 1427 ;
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Taverna D, Boraldi F, De Santis G, Caprioli RM, Quaglino D. Histology-directed and imaging mass spectrometry: An emerging technology in ectopic calcification. Bone 2015; 74:83-94. [PMID: 25595835 PMCID: PMC4355241 DOI: 10.1016/j.bone.2015.01.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Revised: 12/24/2014] [Accepted: 01/07/2015] [Indexed: 01/18/2023]
Abstract
The present study was designed to demonstrate the potential of an optimized histology directed protein identification combined with imaging mass spectrometry technology to reveal and identify molecules associated to ectopic calcification in human tissue. As a proof of concept, mineralized and non-mineralized areas were compared within the same dermal tissue obtained from a patient affected by Pseudoxanthoma elasticum, a genetic disorder characterized by calcification only at specific sites of soft connective tissues. Data have been technically validated on a contralateral dermal tissue from the same subject and compared with those from control healthy skin. Results demonstrate that this approach 1) significantly reduces the effects generated by techniques that, disrupting tissue organization, blend data from affected and unaffected areas; 2) demonstrates that, abolishing differences due to inter-individual variability, mineralized and non-mineralized areas within the same sample have a specific protein profile and have a different distribution of molecules; and 3) avoiding the bias of focusing on already known molecules, reveals a number of proteins that have been never related to the disease nor to the calcification process, thus paving the way for the selection of new molecules to be validated as pathogenic or as potential pharmacological targets.
Collapse
Affiliation(s)
- Domenico Taverna
- Department of Chemistry and Chemical Technologies, University of Calabria, Arcavacata di Rende, Italy
| | - Federica Boraldi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Giorgio De Santis
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Richard M Caprioli
- Departments of Biochemistry, Medicine, Pharmacology and Chemistry and the Mass Spectrometry Research Center, Vanderbilt University Medical Center, Nashville, USA
| | - Daniela Quaglino
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
43
|
Santhakumar R, Vidyasekar P, Verma RS. Cardiogel: a nano-matrix scaffold with potential application in cardiac regeneration using mesenchymal stem cells. PLoS One 2014; 9:e114697. [PMID: 25521816 PMCID: PMC4270637 DOI: 10.1371/journal.pone.0114697] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 11/13/2014] [Indexed: 01/05/2023] Open
Abstract
3-Dimensional conditions for the culture of Bone Marrow-derived Stromal/Stem Cells (BMSCs) can be generated with scaffolds of biological origin. Cardiogel, a cardiac fibroblast-derived Extracellular Matrix (ECM) has been previously shown to promote cardiomyogenic differentiation of BMSCs and provide protection against oxidative stress. To determine the matrix composition and identify significant proteins in cardiogel, we investigated the differences in the composition of this nanomatrix and a BMSC-derived ECM scaffold, termed as ‘mesogel’. An optimized protocol was developed that resulted in efficient decellularization while providing the maximum yield of ECM. The proteins were sequentially solubilized using acetic acid, Sodium Dodecyl Sulfate (SDS) and Dithiothreitol (DTT). These proteins were then analyzed using surfactant-assisted in-solution digestion followed by nano-liquid chromatography and tandem mass spectrometry (nLC-MS/MS). The results of these analyses revealed significant differences in their respective compositions and 17 significant ECM/matricellular proteins were differentially identified between cardiogel and mesogel. We observed that cardiogel also promoted cell proliferation, adhesion and migration while enhancing cardiomyogenic differentiation and angiogenesis. In conclusion, we developed a reproducible method for efficient extraction and solubilization of in vitro cultured cell-derived extracellular matrix. We report several important proteins differentially identified between cardiogel and mesogel, which can explain the biological properties of cardiogel. We also demonstrated the cardiomyogenic differentiation and angiogenic potential of cardiogel even in the absence of any external growth factors. The transplantation of Bone Marrow derived Stromal/Stem Cells (BMSCs) cultured on such a nanomatrix has potential applications in regenerative therapy for Myocardial Infarction (MI).
Collapse
Affiliation(s)
- Rajalakshmi Santhakumar
- Stem cell and Molecular Biology Lab, Department of Biotechnology, Indian Institute of Technology Madras (IITM), Chennai, Tamil Nadu, India
| | - Prasanna Vidyasekar
- Stem cell and Molecular Biology Lab, Department of Biotechnology, Indian Institute of Technology Madras (IITM), Chennai, Tamil Nadu, India
| | - Rama Shanker Verma
- Stem cell and Molecular Biology Lab, Department of Biotechnology, Indian Institute of Technology Madras (IITM), Chennai, Tamil Nadu, India
- * E-mail:
| |
Collapse
|
44
|
Wei C, Kim IK, Li L, Wu L, Gupta S. Thymosin Beta 4 protects mice from monocrotaline-induced pulmonary hypertension and right ventricular hypertrophy. PLoS One 2014; 9:e110598. [PMID: 25412097 PMCID: PMC4239012 DOI: 10.1371/journal.pone.0110598] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 09/19/2014] [Indexed: 01/25/2023] Open
Abstract
Pulmonary hypertension (PH) is a progressive vascular disease of pulmonary arteries that impedes ejection of blood by the right ventricle. As a result there is an increase in pulmonary vascular resistance and pulmonary arterial pressure causing right ventricular hypertrophy (RVH) and RV failure. The pathology of PAH involves vascular cell remodeling including pulmonary arterial endothelial cell (PAEC) dysfunction and pulmonary arterial smooth muscle cell (PASMC) proliferation. Current therapies are limited to reverse the vascular remodeling. Investigating a key molecule is required for development of new therapeutic intervention. Thymosin beta-4 (Tβ4) is a ubiquitous G-actin sequestering protein with diverse biological function and promotes wound healing and modulates inflammatory responses. However, it remains unknown whether Tβ4 has any protective role in PH. The purpose of this study is to evaluate the whether Tβ4 can be used as a vascular-protective agent. In monocrotaline (MCT)-induced PH mouse model, we showed that mice treated with Tβ4 significantly attenuated the systolic pressure and RVH, compared to the MCT treated mice. Our data revealed for the first time that Tβ4 selectively targets Notch3-Col 3A-CTGF gene axis in preventing MCT-induced PH and RVH. Our study may provide pre-clinical evidence for Tβ4 and may consider as vasculo-protective agent for the treatment of PH induced RVH.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Collagen Type III/genetics
- Collagen Type III/metabolism
- Connective Tissue Growth Factor/genetics
- Connective Tissue Growth Factor/metabolism
- Disease Models, Animal
- Endothelial Cells/drug effects
- Hypertension, Pulmonary/chemically induced
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/prevention & control
- Hypertrophy, Right Ventricular/chemically induced
- Hypertrophy, Right Ventricular/pathology
- Hypertrophy, Right Ventricular/prevention & control
- Injections, Intraperitoneal
- Lung/drug effects
- Lung/metabolism
- Lung/pathology
- Male
- Mice
- Monocrotaline/toxicity
- Receptor, Notch3
- Receptors, Notch/genetics
- Receptors, Notch/metabolism
- Signal Transduction/drug effects
- Thymosin/administration & dosage
- Thymosin/pharmacology
Collapse
Affiliation(s)
- Chuanyu Wei
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A & M Health Science Center and Scott & White, Central Texas Veterans Health Care System, Temple, Texas, United States of America
| | - Il-Kwon Kim
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A & M Health Science Center and Scott & White, Central Texas Veterans Health Care System, Temple, Texas, United States of America
| | - Li Li
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A & M Health Science Center and Scott & White, Central Texas Veterans Health Care System, Temple, Texas, United States of America
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | - Liling Wu
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | - Sudhiranjan Gupta
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A & M Health Science Center and Scott & White, Central Texas Veterans Health Care System, Temple, Texas, United States of America
- * E-mail:
| |
Collapse
|
45
|
Wong SW, Sun S, Cho M, Lee KKH, Mak AFT. H2O2 Exposure Affects Myotube Stiffness and Actin Filament Polymerization. Ann Biomed Eng 2014; 43:1178-88. [PMID: 25371376 DOI: 10.1007/s10439-014-1178-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 10/29/2014] [Indexed: 01/24/2023]
Abstract
Skeletal muscles often experience oxidative stress in anaerobic metabolism and ischemia-reperfusion. This paper reports how oxidative stress affects the stiffness of cultured murine myotubes and their actin filaments polymerization dynamics. H2O2 was applied as an extrinsic oxidant to C2C12 myotubes. Atomic force microscopy results showed that short exposures to H2O2 apparently increased the stiffness of myotubes, but that long exposures made the cells softer. The turning point seemed to take place somewhere between 1 and 2 h of H2O2 exposure. We found that the stiffness change was probably due to actin filaments being favored for depolymerization after prolong H2O2 treatments, especially when the exposure duration exceeded 1 h and the exposure concentration reached 1.0 mM. Such depolymerization effect was associated with the down-regulation of thymosin beta 4, as well as the up-regulation of both cofilin2 and profilin1 after prolong H2O2 treatments.
Collapse
Affiliation(s)
- Sing Wan Wong
- Division of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong
| | | | | | | | | |
Collapse
|
46
|
Zamperone A, Pietronave S, Colangelo D, Antonini S, Locatelli M, Travaglia F, Coïsson JD, Arlorio M, Prat M. Protective effects of clovamide against H2O2-induced stress in rat cardiomyoblasts H9c2 cell line. Food Funct 2014; 5:2542-51. [PMID: 25133994 DOI: 10.1039/c4fo00195h] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Cocoa contains phenolic compounds with known antioxidant and antiradical properties beneficial in different pathologies, including cardiovascular diseases. Herein, we have evaluated the protective effects of clovamide, a minor cocoa component, against oxidative stress induced in the rat cardiomyoblast cell line, also comparing it to its bio-isosteric form, rosmarinic acid, and to the main monomeric flavan-3-ol from low-molecular-weight polyphenol in cocoa, i.e. epicatechin. At nano-micro-molar concentrations, the three compounds inhibited the production of reactive oxygen species and apoptosis, evaluated under different aspects, namely, annexin V positivity, DNA fragmentation, caspase release and activation. These molecules can, thus, be considered for their bioactive beneficial activity in the context of cardiovascular pathologies and, particularly, in the protection towards oxidative stress that follows ischemic injury. Clovamide may, thus, be the primary compound for the development of innovative nutraceutical strategies towards cardiovascular diseases.
Collapse
Affiliation(s)
- Andrea Zamperone
- Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
ZHU TINGTING, PARK HEECHUL, SON KYUNGMI, KWON JIHYUN, PARK JONGCHUL, YANG HYEONGCHEOL. Effects of thymosin β4 on wound healing of rat palatal mucosa. Int J Mol Med 2014; 34:816-21. [DOI: 10.3892/ijmm.2014.1832] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 06/25/2014] [Indexed: 11/06/2022] Open
|
48
|
Yi X, Li X, Zhou Y, Ren S, Wan W, Feng G, Jiang X. Hepatocyte growth factor regulates the TGF-β1-induced proliferation, differentiation and secretory function of cardiac fibroblasts. Int J Mol Med 2014; 34:381-90. [PMID: 24840640 PMCID: PMC4094591 DOI: 10.3892/ijmm.2014.1782] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Accepted: 05/09/2014] [Indexed: 01/10/2023] Open
Abstract
Cardiac fibroblast (CF) proliferation and transformation into myofibroblasts play important roles in cardiac fibrosis during pathological myocardial remodeling. In this study, we demonstrate that hepatocyte growth factor (HGF), an antifibrotic factor in the process of pulmonary, renal and liver fibrosis, is a negative regulator of cardiac fibroblast transformation in response to transforming growth factor-β1 (TGF-β1). HGF expression levels were significantly reduced in the CFs following treatment with 5 ng/ml TGF-β1 for 48 h. The overexpression of HGF suppressed the proliferation, transformation and the secretory function of the CFs following treatment with TGF-β1, as indicated by the attenuated expression levels of α-smooth muscle actin (α-SMA) and collagen I and III, whereas the knockdown of HGF had the opposite effect. Mechanistically, we identified that the phosphorylation of c-Met, Akt and total protein of TGIF was significantly inhibited by the knockdown of HGF, but was significantly enhanced by HGF overexpression. Collectively, these results indicate that HGF activates the c-Met-Akt-TGIF signaling pathway, inhibiting CF proliferation and transformation in response to TGF-β1 stimulation.
Collapse
Affiliation(s)
- Xin Yi
- Department of Cardiology, Renmin Hospital of Wuhan University and Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xiaoyan Li
- Department of Cardiology, Renmin Hospital of Wuhan University and Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yanli Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University and Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Shan Ren
- Department of Cardiology, Renmin Hospital of Wuhan University and Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Weiguo Wan
- Department of Cardiology, Renmin Hospital of Wuhan University and Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Gaoke Feng
- Department of Cardiology, Renmin Hospital of Wuhan University and Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xuejun Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University and Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
49
|
Thakur S, Li L, Gupta S. NF-κB-mediated integrin-linked kinase regulation in angiotensin II-induced pro-fibrotic process in cardiac fibroblasts. Life Sci 2014; 107:68-75. [PMID: 24802124 DOI: 10.1016/j.lfs.2014.04.030] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 04/24/2014] [Accepted: 04/25/2014] [Indexed: 12/22/2022]
Abstract
AIMS Cardiac fibrosis is a final outcome of many clinical conditions that lead to cardiac failure and is characterized by a progressive substitution of cellular elements by extracellular-matrix proteins, such as collagen type I, collagen type II, connective tissue growth factor (CTGF), etc. The aim of this study was to identify the mechanisms responsible for angiotensin II (Ang II)-stimulated cardiac fibrosis using rat neonatal cardiac fibroblasts. MAIN METHODS Neonatal fibroblasts were transfected with IκBα mutant, constitutively active (ca) integrin-linked kinase (ILK), dominant negative of ILK and small interfering RNA (siRNA) of ILK in the presence and absence of Ang-II stimulation. The pro-fibrotic gene expression and protein levels were determined by quantitative real time PCR and western blotting using their specific probes and antibodies. NF-κB translocation was determined by immunocytochemistry and confocal microscopy images were analyzed. KEY FINDINGS Our results indicate that overexpression of ILK promotes a pro-fibrotic process by upregulating collagen type I and CTGF genes via activation of nuclear factor-κB (NF-κB) in cardiac fibroblasts. Inactivation of either NF-κB by the super-repressor IκBα or ILK by siRNA significantly attenuates the pro-fibrotic process. Moreover, ILK overexpression triggers NF-κB-p65 translocation to the nucleus, and ILK inhibition prevents the translocation in cardiac fibroblasts stimulated with Ang II. SIGNIFICANCE Our data suggest that the Ang II-stimulated pro-fibrotic process is regulated by a complex mechanism involving crosstalk between ILK and NF-κB activation. This dual mechanism may play a critical role in the progression of cardiac fibrosis.
Collapse
Affiliation(s)
- Suresh Thakur
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A & M Health Science Center; Scott & White; Central Texas Veterans Health Care System, Temple, TX, USA
| | - Li Li
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A & M Health Science Center; Scott & White; Central Texas Veterans Health Care System, Temple, TX, USA
| | - Sudhiranjan Gupta
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A & M Health Science Center; Scott & White; Central Texas Veterans Health Care System, Temple, TX, USA.
| |
Collapse
|
50
|
Thymosin β4-sulfoxide attenuates inflammatory cell infiltration and promotes cardiac wound healing. Nat Commun 2013; 4:2081. [PMID: 23820300 PMCID: PMC3797509 DOI: 10.1038/ncomms3081] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 05/30/2013] [Indexed: 12/19/2022] Open
Abstract
The downstream consequences of inflammation in the adult mammalian heart are formation of a non-functional scar, pathological remodelling and heart failure. In zebrafish, hydrogen peroxide (H2O2) released from a wound is the initial instructive chemotactic cue for the infiltration of inflammatory cells, however, the identity of a subsequent resolution signal(s), to attenuate chronic inflammation, remains unknown. Here we reveal that Thymosin β4-Sulfoxide inhibits interferon-γ, and increases monocyte dispersal and cell death, lies downstream of H2O2 in the wounded fish and triggers depletion of inflammatory macrophages at the injury site. This function is conserved in the mouse and observed after cardiac injury, where it promotes wound healing and reduced scarring. In human T cell/CD14+ monocyte co-cultures, Tβ4-SO inhibits IFN-γ and increases monocyte dispersal and cell death, likely by stimulating superoxide production. Thus, Tβ4-SO is a putative target for therapeutic modulation of the immune response, resolution of fibrosis and cardiac repair.
Collapse
|