1
|
Novaes RD, Souza-E-Leite EG, Silva TD, Caetano-da-Silva JE, Caldas IS, Souza RLM, Marques MJ, Gonçalves RV. Pharmacological blockade of infection chronification modulates oxy-inflammation and prevents the activation of stress-induced premature senescence markers in schistosomiasis. Microb Pathog 2025; 199:107264. [PMID: 39732412 DOI: 10.1016/j.micpath.2024.107264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 12/17/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
Chronic inflammation, oxidative stress, and DNA damage are observed in schistosomiasis and premature aging. However, the potential of these events to trigger stress-induced premature senescence (SIPS) throughout schistosomiasis progression remains overlooked, especially in response to the first-line pharmacological treatment. Thus, we investigated the relationship between oxidative stress and SIPS sentinel markers in untreated Schistosoma mansoni-infected mice and those receiving praziquantel (Pz)-based reference treatment. Swiss mice were randomized into 5 groups: uninfected (followed by 60- and 180-days post-infection), acutely (60 days) and chronically (180 days) infected untreated, and infected treated with Pz followed until 180 days. Our results indicated that infection chronification was accompanied by the worsening of hepatic granulomatous inflammation, increased number of granulomas, IL-4, TGF-β, reactive oxygen species (ROS) levels, fibrosis, hepatocytes DNA damage, upregulation in SA-β-gal activity, p16 and p21 gene expression, and hepatocytes proliferation down-regulation in the absence of telomeric shortening. These abnormalities were blocked by Pz treatment, which prevented infection chronification and the decline in hepatocytes proliferative potential, stimulating granulomatous inflammation resolution. Taken together, our findings provide the evidence that progressive fibrosis, sustained production of high ROS levels, marked DNA damage and decline in p16 and p21 expression are associated with hepatocytes replication attenuation in the chronic phase of S. mansoni infection. Thus, pharmacological blockade of infection and granulomatous inflammation is essential to prevent these premature senescence markers associated with hepatocytes replicative disorders, stimulating liver regeneration in schistosomiasis mansoni.
Collapse
Affiliation(s)
- Rômulo D Novaes
- Instituto de Ciências Biomédicas (ICB), Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, 37130-001, Minas Gerais, Brazil; Departamento de Biologia Animal (DBA), Programa de Pós-Graduação em Biologia Animal (PPGBA), Universidade Federal de Viçosa (UFV), Viçosa, 36570-900, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas (PPGCB), Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, 37130-001, Minas Gerais, Brazil; Programa de Pós-Graduação em Biociências Aplicadas à Saúde (PPGB), Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, 37130-001, Minas Gerais, Brazil.
| | - Elda G Souza-E-Leite
- Instituto de Ciências Biomédicas (ICB), Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, 37130-001, Minas Gerais, Brazil; Programa de Pós-Graduação em Biociências Aplicadas à Saúde (PPGB), Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, 37130-001, Minas Gerais, Brazil
| | - Thiago D Silva
- Programa de Pós-Graduação em Ciências Biológicas (PPGCB), Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, 37130-001, Minas Gerais, Brazil; Programa de Pós-Graduação em Biociências Aplicadas à Saúde (PPGB), Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, 37130-001, Minas Gerais, Brazil
| | - José Edson Caetano-da-Silva
- Programa de Pós-Graduação em Ciências Biológicas (PPGCB), Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, 37130-001, Minas Gerais, Brazil
| | - Ivo S Caldas
- Instituto de Ciências Biomédicas (ICB), Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, 37130-001, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas (PPGCB), Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, 37130-001, Minas Gerais, Brazil
| | - Raquel L M Souza
- Instituto de Ciências Biomédicas (ICB), Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, 37130-001, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas (PPGCB), Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, 37130-001, Minas Gerais, Brazil
| | - Marcos J Marques
- Instituto de Ciências Biomédicas (ICB), Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, 37130-001, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas (PPGCB), Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, 37130-001, Minas Gerais, Brazil
| | - Reggiani V Gonçalves
- Departamento de Biologia Animal (DBA), Programa de Pós-Graduação em Biologia Animal (PPGBA), Universidade Federal de Viçosa (UFV), Viçosa, 36570-900, Minas Gerais, Brazil
| |
Collapse
|
2
|
Liu J, Ma X, Guo W, Lu B, Yue Y, Yang X, Wang R, Wu C, Zhang B, Li X, Luo X. Deacetylation of HnRNP U mediated by sirtuin1 ameliorates aged rat with liver fibrosis via inhibiting p53-related senescence and NLRP3-related inflammation. Int Immunopharmacol 2024; 141:113026. [PMID: 39216234 DOI: 10.1016/j.intimp.2024.113026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Senescence represents a major risk factor promoting liver fibrosis progression. Sirtuin 1 (SIRT1), an essential regulator of cellular senescence, may be involved in developing liver fibrosis. However, the role and mechanism of SIRT1 in liver fibrosis development were largely unknown. We constructed the liver fibrosis in aged rats induced by carbon tetrachloride (CCl4) and then transfected with GFP-SIRT1 adenoviral vectors. After that, we performed acetylomic analysis of liver tissue in aged rats to identify potential substrates of SIRT1. Furthermore, replicative senescent rat hepatocytes were pretreated with siRNA HnRNP U, SIRT1 adenoviral vectors, resveratrol, and siRNA SIRT1, following stimulation with H2O2. We found that the protein levels of SIRT1 and HnRNP U were down-regulation in aged rat liver fibrotic tissues, with an accumulation of NLRP3 inflammasome and activation of the p53/p21 pathway in liver tissue, as well as an increased level of plasma IL-1β secretion. In comparison, these effects were reversed by overexpressing SIRT1 with adenoviral vectors. Acetylation of HnRNP U and its sites at K28 and K787 might be potential targets for SIRT1-mediated liver fibrosis in aged rats. Silencing HnRNP U reduced H2O2-induced up-regulation expression of p53, p21, and NLRP3 inflammasome at protein levels. Additionally, H2O2 induced high acetylation of HnRNP U in senescent hepatocytes, whereas overexpressing SIRT1 with adenoviral vectors and resveratrol deacetylate HnRNP U to inhibit NLRP3 inflammasome and the p53/p21 pathway. Besides, the silence of SIRT1 aggravated H2O2-induced p53-related senescence and NLRP3-related inflammation in senescent hepatocytes. Our findings suggested that deacetylation of HnRNPU mediated by SIRT1 attenuated liver fibrosis in the elderly by inhibiting p53/p21 pathway and NLRP3-related inflammation.
Collapse
Affiliation(s)
- Jinying Liu
- Department of Gastroenterology, Henan Provincial People's Hospital, Henan University People's Hospital, People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Jinshui District, Zhengzhou 450003, China; Microbiome Laboratory, Henan Provincial People's Hospital, Henan University People's Hospital, People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Jinshui District, Zhengzhou 450003, China; Jiaozuo Coal Industry (Group) Co. Ltd. Central Hospital, Jiaozuo 410800, China.
| | - Xiaoli Ma
- Department of Gastroenterology, Henan Provincial People's Hospital, Henan University People's Hospital, People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Jinshui District, Zhengzhou 450003, China; Microbiome Laboratory, Henan Provincial People's Hospital, Henan University People's Hospital, People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Jinshui District, Zhengzhou 450003, China.
| | - Wang Guo
- Department of Gastroenterology, Henan Provincial People's Hospital, Henan University People's Hospital, People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Jinshui District, Zhengzhou 450003, China; Microbiome Laboratory, Henan Provincial People's Hospital, Henan University People's Hospital, People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Jinshui District, Zhengzhou 450003, China.
| | - Bingxin Lu
- Department of Gastroenterology, Henan Provincial People's Hospital, Henan University People's Hospital, People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Jinshui District, Zhengzhou 450003, China; Microbiome Laboratory, Henan Provincial People's Hospital, Henan University People's Hospital, People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Jinshui District, Zhengzhou 450003, China.
| | - Yanqin Yue
- Department of Gastroenterology, Henan Provincial People's Hospital, Henan University People's Hospital, People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Jinshui District, Zhengzhou 450003, China; Microbiome Laboratory, Henan Provincial People's Hospital, Henan University People's Hospital, People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Jinshui District, Zhengzhou 450003, China.
| | - Xingyuan Yang
- Department of Gastroenterology, Henan Provincial People's Hospital, Henan University People's Hospital, People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Jinshui District, Zhengzhou 450003, China; Microbiome Laboratory, Henan Provincial People's Hospital, Henan University People's Hospital, People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Jinshui District, Zhengzhou 450003, China.
| | - Rui Wang
- Department of Gastroenterology, Henan Provincial People's Hospital, Henan University People's Hospital, People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Jinshui District, Zhengzhou 450003, China; Microbiome Laboratory, Henan Provincial People's Hospital, Henan University People's Hospital, People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Jinshui District, Zhengzhou 450003, China.
| | - Caihong Wu
- Department of Clinical Laboratory, Hebei Petro China Central Hospital, Langfang 065000, China.
| | - Bingyong Zhang
- Department of Gastroenterology, Henan Provincial People's Hospital, Henan University People's Hospital, People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Jinshui District, Zhengzhou 450003, China.
| | - Xiuling Li
- Department of Gastroenterology, Henan Provincial People's Hospital, Henan University People's Hospital, People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Jinshui District, Zhengzhou 450003, China.
| | - Xiaoying Luo
- Department of Gastroenterology, Henan Provincial People's Hospital, Henan University People's Hospital, People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Jinshui District, Zhengzhou 450003, China; Microbiome Laboratory, Henan Provincial People's Hospital, Henan University People's Hospital, People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Jinshui District, Zhengzhou 450003, China.
| |
Collapse
|
3
|
Zhang Y, Chen P, Fang X. Proteomic and metabolomic analysis of GH deficiency-induced NAFLD in hypopituitarism: insights into oxidative stress. Front Endocrinol (Lausanne) 2024; 15:1371444. [PMID: 38836220 PMCID: PMC11148278 DOI: 10.3389/fendo.2024.1371444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/24/2024] [Indexed: 06/06/2024] Open
Abstract
Objective Individuals with hypopituitarism (HPs) have an increased risk of developing non-alcoholic fatty liver disease (NAFLD)/non-alcoholic steatohepatitis (NASH) due to growth hormone deficiency (GHD). We aimed to investigate the possible mechanisms underlying the relationship between GHD and NAFLD using proteomic and metabolomic insights. Methods Serum metabolic alternations were assessed in male HPs using untargeted metabolomics. A rat model of HP was established through hypophysectomy, followed by recombinant human growth hormone (rhGH) intervention. The mechanisms underlying GHD-mediated NAFLD were elucidated through the application of label-free proteomics and phosphorylation proteomics. Results Metabolomic analysis revealed that biomarkers of mitochondrial dysfunction and oxidative stress, such as alanine, lactate, and creatine, were significantly elevated in HPs compared to age-matched controls. In rats, hypophysectomy led to marked hepatic steatosis, lipid peroxidation, and reduced glutathione (GSH), which were subsequently modulated by rhGH replacement. Proteomic analysis identified cytochrome P450s, mitochondrial translation elongation, and PPARA activating genes as the major distinguishing pathways in hypophysectomized rats. The processes of fatty acid transport, synthesis, oxidation, and NADP metabolism were tightly described. An enhanced regulation of peroxisome β-oxidation and ω-oxidation, together with a decreased NADPH regeneration, may exacerbate oxidative stress. Phosphoproteome data showed downregulation of JAK2-STAT5B and upregulation of mTOR signaling pathway. Conclusions This study identified proteo-metabolomic signatures associated with the development of NAFLD in pituitary GHD. Evidence was found of oxidative stress imbalance resulting from abnormal fatty acid oxidation and NADPH regeneration, highlighting the role of GH deficiency in the development of NAFLD.
Collapse
Affiliation(s)
- Yuwen Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peizhan Chen
- Clinical Research Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuqian Fang
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Bao YN, Yang Q, Shen XL, Yu WK, Zhou L, Zhu QR, Shan QY, Wang ZC, Cao G. Targeting tumor suppressor p53 for organ fibrosis therapy. Cell Death Dis 2024; 15:336. [PMID: 38744865 PMCID: PMC11094089 DOI: 10.1038/s41419-024-06702-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 04/18/2024] [Accepted: 04/22/2024] [Indexed: 05/16/2024]
Abstract
Fibrosis is a reparative and progressive process characterized by abnormal extracellular matrix deposition, contributing to organ dysfunction in chronic diseases. The tumor suppressor p53 (p53), known for its regulatory roles in cell proliferation, apoptosis, aging, and metabolism across diverse tissues, appears to play a pivotal role in aggravating biological processes such as epithelial-mesenchymal transition (EMT), cell apoptosis, and cell senescence. These processes are closely intertwined with the pathogenesis of fibrotic disease. In this review, we briefly introduce the background and specific mechanism of p53, investigate the pathogenesis of fibrosis, and further discuss p53's relationship and role in fibrosis affecting the kidney, liver, lung, and heart. In summary, targeting p53 represents a promising and innovative therapeutic approach for the prevention and treatment of organ fibrosis.
Collapse
Affiliation(s)
- Yi-Ni Bao
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China
| | - Qiao Yang
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China
| | - Xin-Lei Shen
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China
| | - Wen-Kai Yu
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China
| | - Li Zhou
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China
| | - Qing-Ru Zhu
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China
| | - Qi-Yuan Shan
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China
| | - Zhi-Chao Wang
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China.
| |
Collapse
|
5
|
Hu Y, Yuan C, Abdulnaimu M, Memetmin J, Jie Z, Tuhuti A, Abudueini H, Guo Y. U-Shaped relationship of insulin-like growth factor I and incidence of nonalcoholic fatty liver in patients with pituitary neuroendocrine tumors: a cohort study. Front Endocrinol (Lausanne) 2024; 15:1290007. [PMID: 38370349 PMCID: PMC10869555 DOI: 10.3389/fendo.2024.1290007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 01/17/2024] [Indexed: 02/20/2024] Open
Abstract
Context Although the role of insulin-like growth factor I (IGF-1) in nonalcoholic fatty liver disease (NAFLD) has garnered attention in recent years, few studies have examined both reduced and elevated levels of IGF-1. Objective The aim of this study was to examine the potential relationship between IGF-1 levels and the risk of new-onset NAFLD in patients with pituitary neuroendocrine tumors (PitNET). Methods We employed multivariable Cox regression models and two-piecewise regression models to assess the association between IGF-1 and new-onset NAFLD. Hazard ratios (HRs) and their corresponding 95% confidence intervals (CIs) were calculated to quantify this association. Furthermore, a dose-response correlation between lgIGF-1 and the development of NAFLD was plotted. Additionally, we also performed subgroup analysis and a series sensitivity analysis. Results A total of 3,291 PitNET patients were enrolled in the present study, and the median duration of follow-up was 65 months. Patients with either reduced or elevated levels of IGF-1 at baseline were found to be at a higher risk of NAFLD compared to PitNET patients with normal IGF-1(log-rank test, P < 0.001). In the adjusted Cox regression analysis model (model IV), compared with participants with normal IGF-1, the HRs of those with elevated and reduced IGF-1 were 2.33 (95% CI 1.75, 3.11) and 2.2 (95% CI 1.78, 2.7). Furthermore, in non-adjusted or adjusted models, our study revealed a U-shaped relationship between lgIGF-1 and the risk of NAFLD. Moreover, the results from subgroup and sensitivity analyses were consistent with the main results. Conclusions There was a U-shaped trend between IGF-1 and new-onset NAFLD in patients with PitNET. Further evaluation of our discoveries is warranted.
Collapse
Affiliation(s)
- Yan Hu
- Graduate School, Xinjiang Medical University, Urumqi, China
| | - Chen Yuan
- Department of Endocrinology, People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Clinical Research Center for Diabetes, Urumqi, China
| | - Muila Abdulnaimu
- Department of Endocrinology, People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Clinical Research Center for Diabetes, Urumqi, China
| | - Jimilanmu Memetmin
- Department of Endocrinology, People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Clinical Research Center for Diabetes, Urumqi, China
| | - Zhang Jie
- Department of Endocrinology, People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Clinical Research Center for Diabetes, Urumqi, China
| | - Aihemaitijiang Tuhuti
- Department of Endocrinology, People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Clinical Research Center for Diabetes, Urumqi, China
| | - Hanikzi Abudueini
- Department of Endocrinology, People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Clinical Research Center for Diabetes, Urumqi, China
| | - Yanying Guo
- Department of Endocrinology, People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Clinical Research Center for Diabetes, Urumqi, China
| |
Collapse
|
6
|
Tanaka M, Kaji K, Nishimura N, Asada S, Koizumi A, Matsuda T, Yorioka N, Tsuji Y, Fujinaga Y, Sato S, Namisaki T, Akahane T, Yoshiji H. Blockade of angiotensin II modulates insulin-like growth factor 1-mediated skeletal muscle homeostasis in experimental steatohepatitis. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119649. [PMID: 38097064 DOI: 10.1016/j.bbamcr.2023.119649] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/29/2023] [Accepted: 12/01/2023] [Indexed: 12/18/2023]
Abstract
Sarcopenia is associated with mortality in patients with nonalcoholic steatohepatitis (NASH). Angiotensin II receptor blocker (ARB) has been suggested to prevent sarcopenia, but reports on its effect on NASH-derived skeletal muscle atrophy in conjunction with insulin-like growth factor 1 (IGF-1)-mediated muscle homeostasis are few. Our aim was to examine the combined effect of the ARB losartan and IGF-1 replacement on skeletal muscle atrophy in a methionine-choline deficient (MCD) diet-fed murine steatohepatitis model. The MCD-fed mice developed steatohepatitis and skeletal muscle atrophy, as indicated by the reduction of psoas muscle mass and attenuation of forelimb and hindlimb grip strength. Significantly suppressed steatohepatitis and skeletal muscle atrophy was observed after single treatment with ARB or IGF-1, and these effects were augmented after combination treatment. Treatment with ARB and IGF-1 effectively inhibited ubiquitin proteasome-mediated protein degradation by reducing forkhead box protein O1 (FOXO1) and FOXO3a transcriptional activity in the skeletal muscle. Combined ARB and IGF-1 decreased the intramuscular expression of proinflammatory cytokines (i.e., TNFα, IL6, and IL1β) and increased the Trolox equivalent antioxidant capacity and antioxidant enzymes (CAT, GPX1, SOD2, and CYTB). This antioxidant effect was based on downregulation of NADPH oxidase (NOX) 2, normalization of mitochondrial biogenesis and dynamics. Moreover, ARB increased the hepatic and plasma IGF-1 levels and improved steatohepatitis, leading to enhanced skeletal muscle protein synthesis mediated by IGF-1/ AKT/ mechanistic target of rapamycin signaling. Collectively, combined ARB and IGF-1 replacement could be a promising new therapeutic target for NASH-derived skeletal muscle wasting.
Collapse
Affiliation(s)
- Misako Tanaka
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Kosuke Kaji
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara 634-8521, Japan.
| | - Norihisa Nishimura
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Shohei Asada
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Aritoshi Koizumi
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Takuya Matsuda
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Nobuyuki Yorioka
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Yuki Tsuji
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Yukihisa Fujinaga
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Shinya Sato
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Tadashi Namisaki
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Takemi Akahane
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Hitoshi Yoshiji
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| |
Collapse
|
7
|
Chen Z, Li C, Huang H, Shi YL, Wang X. Research Progress of Aging-related MicroRNAs. Curr Stem Cell Res Ther 2024; 19:334-350. [PMID: 36892029 DOI: 10.2174/1574888x18666230308111043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 01/02/2023] [Accepted: 01/11/2023] [Indexed: 03/10/2023]
Abstract
Senescence refers to the irreversible state in which cells enter cell cycle arrest due to internal or external stimuli. The accumulation of senescent cells can lead to many age-related diseases, such as neurodegenerative diseases, cardiovascular diseases, and cancers. MicroRNAs are short non-coding RNAs that bind to target mRNA to regulate gene expression after transcription and play an important regulatory role in the aging process. From nematodes to humans, a variety of miRNAs have been confirmed to alter and affect the aging process. Studying the regulatory mechanisms of miRNAs in aging can further deepen our understanding of cell and body aging and provide a new perspective for the diagnosis and treatment of aging-related diseases. In this review, we illustrate the current research status of miRNAs in aging and discuss the possible prospects for clinical applications of targeting miRNAs in senile diseases.
Collapse
Affiliation(s)
- Zhongyu Chen
- School of Basic Medicine, Dali University, Dali, Yunnan, 671000, China
| | - Chenxu Li
- School of Basic Medicine, Dali University, Dali, Yunnan, 671000, China
| | - Haitao Huang
- School of Basic Medicine, Dali University, Dali, Yunnan, 671000, China
| | - Yi-Ling Shi
- School of Basic Medicine, Dali University, Dali, Yunnan, 671000, China
| | - Xiaobo Wang
- School of Basic Medicine, Dali University, Dali, Yunnan, 671000, China
- Key Laboratory of University Cell Biology, Dali, Yunnan, 671000, China
| |
Collapse
|
8
|
Gui R, Li W, Li Z, Wang H, Wu Y, Jiao W, Zhao G, Shen Y, Wang L, Zhang J, Chen S, Hao L, Cheng Y. Effects and potential mechanisms of IGF1/IGF1R in the liver fibrosis: A review. Int J Biol Macromol 2023; 251:126263. [PMID: 37567540 DOI: 10.1016/j.ijbiomac.2023.126263] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/13/2023]
Abstract
Liver fibrosis is a wound-healing response due to persistent liver damage and it may progress to cirrhosis and even liver cancer if no intervention is given. In the current cognition, liver fibrosis is reversible. So, it is of great significance to explore the related gene targets or biomarker for anti-fibrosis of liver. Insulin like growth factor 1 (IGF1) and IGF1 receptor (IGF1R) are mainly expressed in the liver tissues and play critical roles in the liver function. The present review summarized the role of IGF1/IGF1R and its signaling system in liver fibrosis and illustrated the potential mechanisms including DNA damage repair, cell senescence, lipid metabolism and oxidative stress that may be involved in this process according to the studies on the fibrosis of liver or other organs. In particular, the roles of IGF1 and IGF1R in DNA damage repair were elaborated, including membrane-localized and nucleus-localized IGF1R. In addition, for each of the potential mechanism in anti-fibrosis of liver, the signaling pathways of the IGF1/IGF1R mediated and the cell species in liver acted by IGF1 and IGF1R under different conditions were included. The data in this review will support for the study about the effect of IGF1/IGF1R on liver fibrosis induced by various factors, meanwhile, provide a basis for the study of liver fibrosis to focus on the communications between the different kinds of liver cells.
Collapse
Affiliation(s)
- Ruirui Gui
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Wanqiao Li
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Zhipeng Li
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Hongbin Wang
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Yuchen Wu
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Wenlin Jiao
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Gang Zhao
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Yannan Shen
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Luping Wang
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Jialu Zhang
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Sihan Chen
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Linlin Hao
- College of Animal Science, Jilin University, Changchun, Jilin 130062, China.
| | - Yunyun Cheng
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China.
| |
Collapse
|
9
|
Kim BH, Chung YH, Woo TG, Kang SM, Park S, Park BJ. Progerin, an Aberrant Spliced Form of Lamin A, Is a Potential Therapeutic Target for HGPS. Cells 2023; 12:2299. [PMID: 37759521 PMCID: PMC10527460 DOI: 10.3390/cells12182299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is an extremely rare genetic disorder caused by the mutant protein progerin, which is expressed by the abnormal splicing of the LMNA gene. HGPS affects systemic levels, with the exception of cognition or brain development, in children, showing that cellular aging can occur in the short term. Studying progeria could be useful in unraveling the causes of human aging (as well as fatal age-related disorders). Elucidating the clear cause of HGPS or the development of a therapeutic medicine could improve the quality of life and extend the survival of patients. This review aimed to (i) briefly describe how progerin was discovered as the causative agent of HGPS, (ii) elucidate the puzzling observation of the absence of primary neurological disease in HGPS, (iii) present several studies showing the deleterious effects of progerin and the beneficial effects of its inhibition, and (iv) summarize research to develop a therapy for HGPS and introduce clinical trials for its treatment.
Collapse
Affiliation(s)
- Bae-Hoon Kim
- Rare Disease R&D Center, PRG S&T Co., Ltd., Busan 46274, Republic of Korea; (B.-H.K.); (Y.-H.C.); (T.-G.W.)
| | - Yeon-Ho Chung
- Rare Disease R&D Center, PRG S&T Co., Ltd., Busan 46274, Republic of Korea; (B.-H.K.); (Y.-H.C.); (T.-G.W.)
| | - Tae-Gyun Woo
- Rare Disease R&D Center, PRG S&T Co., Ltd., Busan 46274, Republic of Korea; (B.-H.K.); (Y.-H.C.); (T.-G.W.)
| | - So-Mi Kang
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan 46231, Republic of Korea; (S.-M.K.); (S.P.)
| | - Soyoung Park
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan 46231, Republic of Korea; (S.-M.K.); (S.P.)
| | - Bum-Joon Park
- Rare Disease R&D Center, PRG S&T Co., Ltd., Busan 46274, Republic of Korea; (B.-H.K.); (Y.-H.C.); (T.-G.W.)
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan 46231, Republic of Korea; (S.-M.K.); (S.P.)
| |
Collapse
|
10
|
Wang Y, Wang P, Yu Y, Huang E, Yao Y, Guo D, Peng H, Tian B, Zheng Q, Jia M, Wang J, Wu X, Cheng J, Liu H, Wang QK, Xu C. Hepatocyte Ninjurin2 promotes hepatic stellate cell activation and liver fibrosis through the IGF1R/EGR1/PDGF-BB signaling pathway. Metabolism 2023; 140:155380. [PMID: 36549436 DOI: 10.1016/j.metabol.2022.155380] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 11/25/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Liver fibrogenesis is orchestrated by the paracrine signaling interaction between several resident cell types regulating the activation of hepatic stellate cells (HSCs). However, the molecular mechanisms underlying paracrine regulation are largely unknown. The aim of this study is to elucidate the role of Ninjurin2 in the crosstalk between hepatocytes and HSCs and better understand the implications of Ninjurin2 in liver fibrosis. METHODS Ninj2 knockout mice (Ninj2-/-) and hepatocyte-specific Ninj2 overexpression mice (Ninj2Hep-tg) were constructed and followed by the induction of liver fibrosis using methionine- and choline-deficient (MCD) diet. The relationship between Ninjurin2 and liver fibrosis phenotype was evaluated in vivo by measurement of fibrotic markers and related genes. We used an in vitro transwell cell co-culture model to examine the impact of Ninjurin2 in hepatocytes on the crosstalk to HSCs. The interaction of Ninjurin2 and IGF1R and the regulation of PI3K-AKT-EGR1 were analyzed in vivo and in vitro. Finally, an inhibitory Ninjurin2 peptide was injected intravenously via the tail vein to investigate whether inhibiting of Ninjurin2 cascade can attenuate MCD diet-induced liver fibrosis in mice. RESULTS We found that hepatic Ninjurin2 expression was significantly increased in fibrotic human liver and MCD diet-induced liver injury mouse models. In the mouse model, hepatocyte-specific overexpression of Ninj2 exacerbates MCD-induced liver fibrosis, while global Ninj2 knockout reverses the phenotype. To mimic hepatocyte-HSC crosstalk during liver fibrosis, we used co-culture systems containing hepatocytes and HSCs and determined that Ninjurin2 overexpression in hepatocytes directly activates HSCs in vitro. Mechanistically, Ninjurin2 directly interacts with insulin-like growth factor 1 receptor (IGF1R) and increases the hepatocyte secretion of the fibrogenic cytokine, platelet-derived growth factor-BB (PDGF-BB) through IGF1R-PI3K-AKT-EGR1 cascade. Inhibition of PDGFRB signaling in HSCs can abolish the profibrogenic effect of Ninjurin2. In addition, we demonstrated that a specific inhibitory Ninjurin2 peptide containing an N-terminal adhesion motif mitigates liver fibrosis and improves hepatic function in the mouse models by negatively regulating the sensitivity of IGF1R to IGF1 in hepatocytes. CONCLUSION Hepatic Ninjurin2 plays a key role in liver fibrosis through paracrine regulation of PDGF-BB/PDGFRB signaling in HSCs, and the results suggesting Ninjurin2 may be a potential therapeutic target.
Collapse
Affiliation(s)
- Yifan Wang
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Pengyun Wang
- Liyuan Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China
| | - Yubing Yu
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Erwen Huang
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yufeng Yao
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Di Guo
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Huixin Peng
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Beijia Tian
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Qian Zheng
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Mengru Jia
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Jing Wang
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Xinna Wu
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Jianding Cheng
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Huiying Liu
- College of Pulmonary and Critical Medicine, Chinese PLA General Hospital, Beijing, China
| | - Qing K Wang
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China.
| | - Chengqi Xu
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China.
| |
Collapse
|
11
|
Li W, Yin X, Yan Y, Liu C, Li G. Kurarinone attenuates hydrogen peroxide-induced oxidative stress and apoptosis through activating the PI3K/Akt signaling by upregulating IGF1 expression in human ovarian granulosa cells. ENVIRONMENTAL TOXICOLOGY 2023; 38:28-38. [PMID: 36114797 DOI: 10.1002/tox.23659] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 08/28/2022] [Accepted: 08/29/2022] [Indexed: 06/15/2023]
Abstract
Dysregulated follicular development may lead to follicular atresia, and this is associated with oxidative stress in granulosa cells. Kurarinone is a natural compound possessing multiple activities, including antioxidative ability. However, the role of kurarinone in granulosa cell damage during follicular atresia remains unknown. Human ovarian granulosa KGN cells were treated with hydrogen peroxide (H2 O2 ) to induce cellular damage. Cytotoxicity was investigated by lactate dehydrogenase (LDH) release assay. Oxidative stress was evaluated by detection of reactive oxygen species (ROS) generation and oxidative biomarker levels. Cell apoptosis was evaluated by flow cytometry, a Cell Death Detection ELISA Kit, and a Caspase-3 Assay Kit. The downstream target and related signaling pathway were analyzed by western blotting. Kurarinone attenuated H2 O2 -induced LDH release in KGN cells. Kurarinone relieved H2 O2 -induced increase in ROS generation and malondialdehyde level as well as decrease in superoxide dismutase-1 activity and heme oxygenase 1 and NAD(P)H quinone dehydrogenase 1 mRNA levels. Kurarinone inhibited H2 O2 -induced apoptosis in KGN cells. Kurarinone targeted insulin-like growth factor 1 (IGF1) and upregulated IGF1 expression to activate the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) signaling. IGF1 silencing attenuated the suppressive effects of kurarinone on H2 O2 -induced oxidative stress and apoptosis in KGN cells. In conclusion, kurarinone attenuates H2 O2 -induced oxidative stress and apoptosis in KGN cells through activating the PI3K/Akt signaling by upregulating IGF1 expression, indicating the therapeutic potential of kurarinone in follicular atresia.
Collapse
Affiliation(s)
- Weiwei Li
- Department of Reproductive Medicine, Maternal and Child Care Center of Qinhuangdao, Qinhuangdao, China
| | - Xiurong Yin
- Department of Reproductive Medicine, Maternal and Child Care Center of Qinhuangdao, Qinhuangdao, China
| | - Yani Yan
- Department of Reproductive Medicine, Maternal and Child Care Center of Qinhuangdao, Qinhuangdao, China
| | - Cong Liu
- Department of Reproductive Medicine, Maternal and Child Care Center of Qinhuangdao, Qinhuangdao, China
| | - Gang Li
- Department of Surgical Anesthesiology, Maternal and Child Care Center of Qinhuangdao, Qinhuangdao, China
| |
Collapse
|
12
|
Nucleophagic Degradation of Progerin Ameliorates Defenestration in Liver Sinusoidal Endothelium Due to SIRT1-Mediated Deacetylation of Nuclear LC3. Cells 2022; 11:cells11233918. [PMID: 36497176 PMCID: PMC9738635 DOI: 10.3390/cells11233918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/24/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Progerin, a permanently farnesylated prelamin A protein in cell nuclei, is potentially implicated in the defenestration of liver sinusoidal endothelial cells (LSECs) and liver fibrogenesis. Autophagy regulates the degradation of nuclear components, called nucleophagy, in response to damage. However, little is known about the role of nucleophagy in LSEC defenestration. Herein, we aim to dissect the underlying mechanism of progerin and nucleophagy in LSEC phenotype. We found an abnormal accumulation of progerin and a loss of SIRT1 in the nucleus of intrahepatic cells in human fibrotic liver tissue. In vivo, nuclear progerin abnormally accumulated in defenestrated LSECs, along with a depletion of SIRT1 and Cav-1 during liver fibrogenesis, whereas these effects were reversed by the overexpression of SIRT1 with the adenovirus vector. In vitro, H2O2 induced the excessive accumulation of progeirn, with the depletion of Lamin B1 and Cav-1 to aggravate LSEC defenestration. NAC and mito-TEMPO, classical antioxidants, inhibited NOX2- and NOX4-dependent oxidative stress to improve the depletion of Lamin B1 and Cav-1 and promoted progerin-related nucleophagy, leading to a reverse in H2O2-induced LSEC defenestration. However, rapamycin aggravated the H2O2-induced depletion of Lamin B1 and Cav-1 due to excessive autophagy, despite promoting progerin nucleophagic degradation. In addition, overexpressing SIRT1 with the adenovirus vector inhibited oxidative stress to rescue the production of Lamin B1 and Cav-1. Moreover, the SIRT1-mediated deacetylation of nuclear LC3 promoted progerin nucleophagic degradation and subsequently inhibited the degradation of Lamin B1 and Cav-1, as well as improved F-actin remodeling, contributing to maintaining LSEC fenestrae. Hence, our findings indicate a new strategy for reversing LSEC defenestration by promoting progerin clearance via the SIRT1-mediated deacetylation of nuclear LC3.
Collapse
|
13
|
Kopchick JJ, Basu R, Berryman DE, Jorgensen JOL, Johannsson G, Puri V. Covert actions of growth hormone: fibrosis, cardiovascular diseases and cancer. Nat Rev Endocrinol 2022; 18:558-573. [PMID: 35750929 PMCID: PMC9703363 DOI: 10.1038/s41574-022-00702-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/19/2022] [Indexed: 12/20/2022]
Abstract
Since its discovery nearly a century ago, over 100,000 studies of growth hormone (GH) have investigated its structure, how it interacts with the GH receptor and its multiple actions. These include effects on growth, substrate metabolism, body composition, bone mineral density, the cardiovascular system and brain function, among many others. Recombinant human GH is approved for use to promote growth in children with GH deficiency (GHD), along with several additional clinical indications. Studies of humans and animals with altered levels of GH, from complete or partial GHD to GH excess, have revealed several covert or hidden actions of GH, such as effects on fibrosis, cardiovascular function and cancer. In this Review, we do not concentrate on the classic and controversial indications for GH therapy, nor do we cover all covert actions of GH. Instead, we stress the importance of the relationship between GH and fibrosis, and how fibrosis (or lack thereof) might be an emerging factor in both cardiovascular and cancer pathologies. We highlight clinical data from patients with acromegaly or GHD, alongside data from cellular and animal studies, to reveal novel phenotypes and molecular pathways responsible for these actions of GH in fibrosis, cardiovascular function and cancer.
Collapse
Affiliation(s)
- John J Kopchick
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA.
- The Diabetes Institute, Ohio University, Athens, OH, USA.
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA.
| | - Reetobrata Basu
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
- The Diabetes Institute, Ohio University, Athens, OH, USA
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
| | - Darlene E Berryman
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
- The Diabetes Institute, Ohio University, Athens, OH, USA
| | - Jens O L Jorgensen
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Gudmundur Johannsson
- Department of Endocrinology, Sahlgrenska University Hospital, Sahlgrenska Academy, University of Göteborg, Gothenburg, Sweden
| | - Vishwajeet Puri
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
- The Diabetes Institute, Ohio University, Athens, OH, USA
| |
Collapse
|
14
|
Xiang Y, You Z, Huang X, Dai J, Zhang J, Nie S, Xu L, Jiang J, Xu J. Oxidative stress-induced premature senescence and aggravated denervated skeletal muscular atrophy by regulating progerin-p53 interaction. Skelet Muscle 2022; 12:19. [PMID: 35906707 PMCID: PMC9335985 DOI: 10.1186/s13395-022-00302-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 07/21/2022] [Indexed: 11/17/2022] Open
Abstract
Background Progerin elevates atrophic gene expression and helps modify the nuclear membrane to cause severe muscle pathology, which is similar to muscle weakness in the elderly, to alter the development and function of the skeletal muscles. Stress-induced premature senescence (SIPS), a state of cell growth arrest owing to such stimuli as oxidation, can be caused by progerin. However, evidence for whether SIPS-induced progerin accumulation is connected to denervation-induced muscle atrophy is not sufficient. Methods Flow cytometry and a reactive oxygen species (ROS) as well as inducible nitric oxide synthase (iNOS) inhibitors were used to assess the effect of oxidation on protein (p53), progerin, and nuclear progerin–p53 interaction in the denervated muscles of models of mice suffering from sciatic injury. Loss-of-function approach with the targeted deletion of p53 was used to assess connection among SIPS, denervated muscle atrophy, and fibrogenesis. Results The augmentation of ROS and iNOS-derived NO in the denervated muscles of models of mice suffering from sciatic injury upregulates p53 and progerin. The abnormal accumulation of progerin in the nuclear membrane as well as the activation of nuclear progerin–p53 interaction triggered premature senescence in the denervated muscle cells of mice. The p53-dependent SIPS in denervated muscles contributes to their atrophy and fibrogenesis. Conclusion Oxidative stress-triggered premature senescence via nuclear progerin–p53 interaction that promotes denervated skeletal muscular atrophy and fibrogenesis. Supplementary Information The online version contains supplementary material available at 10.1186/s13395-022-00302-y.
Collapse
Affiliation(s)
- Yaoxian Xiang
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China.,NHC Key Laboratory of Hand Reconstruction, (Fudan University), Shanghai, People's Republic of China.,Shanghai Key Laboratory of Peripheral Nerve and Microsurgery, Shanghai, People's Republic of China
| | - Zongqi You
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China.,NHC Key Laboratory of Hand Reconstruction, (Fudan University), Shanghai, People's Republic of China.,Shanghai Key Laboratory of Peripheral Nerve and Microsurgery, Shanghai, People's Republic of China
| | - Xinying Huang
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China.,NHC Key Laboratory of Hand Reconstruction, (Fudan University), Shanghai, People's Republic of China.,Shanghai Key Laboratory of Peripheral Nerve and Microsurgery, Shanghai, People's Republic of China.,Shanghai Medical College of Fudan University, Shanghai, People's Republic of China
| | - Junxi Dai
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China.,NHC Key Laboratory of Hand Reconstruction, (Fudan University), Shanghai, People's Republic of China.,Shanghai Key Laboratory of Peripheral Nerve and Microsurgery, Shanghai, People's Republic of China
| | - Junpeng Zhang
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Shuqi Nie
- Shanghai Medical College of Fudan University, Shanghai, People's Republic of China
| | - Lei Xu
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China.,NHC Key Laboratory of Hand Reconstruction, (Fudan University), Shanghai, People's Republic of China.,Shanghai Key Laboratory of Peripheral Nerve and Microsurgery, Shanghai, People's Republic of China
| | - Junjian Jiang
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China. .,NHC Key Laboratory of Hand Reconstruction, (Fudan University), Shanghai, People's Republic of China. .,Shanghai Key Laboratory of Peripheral Nerve and Microsurgery, Shanghai, People's Republic of China.
| | - Jianguang Xu
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China. .,NHC Key Laboratory of Hand Reconstruction, (Fudan University), Shanghai, People's Republic of China. .,Shanghai Key Laboratory of Peripheral Nerve and Microsurgery, Shanghai, People's Republic of China. .,School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China.
| |
Collapse
|
15
|
Hu WS, Liao WY, Chang CH, Chen TS. Paracrine IGF-1 Activates SOD2 Expression and Regulates ROS/p53 Axis in the Treatment of Cardiac Damage in D-Galactose-Induced Aging Rats after Receiving Mesenchymal Stem Cells. J Clin Med 2022; 11:4419. [PMID: 35956039 PMCID: PMC9369306 DOI: 10.3390/jcm11154419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/23/2022] [Accepted: 07/28/2022] [Indexed: 12/27/2022] Open
Abstract
Aging is one of the causative agents associated with heart failure. Cell-based therapies show potential in the treatment of cardiac aging due to the characteristics of stem cells, including differentiation and the paracrine effect. This study aimed to investigate in detail the mechanism related to biomolecules released from mesenchymal stem cells in the treatment of cardiac aging. In vitro and in vivo models were designed to explore the above hypothesis. Experimental results from the in vitro model indicated that the elevation of oxidative stress, the expression of aging marker p53, and the suppression of antioxidant marker SOD2 could be found in D-galactose-stressed H9c2 cardiomyoblasts. The co-culture of D-galactose-stressed H9c2 with mesenchymal stem cells significantly improved the above pathological signaling. An animal model revealed that the change in cardiac structure, the accumulation of fibrotic collagen, and the activation of the above pathological signaling could be observed in heart tissues of D-galactose-stressed rats. After the rats had received mesenchymal stem cells, all the pathological conditions were significantly improved in D-galactose-stressed hearts. Further evidence indicated that the release of the survival marker IGF-1 was detected in a stem-cell-conditioned medium. Significant increases in cell viability and the expression of SOD2, as well as a reduction in oxidative stress and the suppression of p53, were found in D-galactose-stressed H9c2 cells cultured with a stem-cell-conditioned medium, whereas the depletion of IGF-1 in stem-cell-conditioned medium diminished the antiaging effect on H9c2 cells. In conclusion, the paracrine release of IGF-1 from mesenchymal stem cells increases the expression of antioxidant marker SOD2, and the expression of SOD2 reduces oxidative stress as well as suppresses p53, leading to a reduction in cardiac senescence in D-galactose-stressed rats.
Collapse
Affiliation(s)
- Wei-Syun Hu
- School of Medicine, College of Medicine, China Medical University, Taichung City 40042, Taiwan;
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University Hospital, Taichung City 40447, Taiwan
| | - Wei-Yu Liao
- Traditional Chinese Medicine Department, En Chu Kong Hospital, New Taipei City 40237, Taiwan;
- Department of Cosmetic Science, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan
| | - Chin-Hsien Chang
- Traditional Chinese Medicine Department, En Chu Kong Hospital, New Taipei City 40237, Taiwan;
- Department of Cosmetic Science, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan
- College of Chinese Medicine, China Medical University, Taichung City 40402, Taiwan
| | - Tung-Sheng Chen
- Graduate Program of Biotechnology and Pharmaceutical Industries, National Taiwan Normal University, Taipei 11677, Taiwan
| |
Collapse
|
16
|
Ebert T, Tran N, Schurgers L, Stenvinkel P, Shiels PG. Ageing - Oxidative stress, PTMs and disease. Mol Aspects Med 2022; 86:101099. [PMID: 35689974 DOI: 10.1016/j.mam.2022.101099] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 05/10/2022] [Indexed: 12/15/2022]
Abstract
Post-translational modifications (PTMs) have been proposed as a link between the oxidative stress-inflammation-ageing trinity, thereby affecting several hallmarks of ageing. Phosphorylation, acetylation, and ubiquitination cover >90% of all the reported PTMs. Several of the main PTMs are involved in normal "healthy" ageing and in different age-related diseases, for instance neurodegenerative, metabolic, cardiovascular, and bone diseases, as well as cancer and chronic kidney disease. Ultimately, data from human rare progeroid syndromes, but also from long-living animal species, imply that PTMs are critical regulators of the ageing process. Mechanistically, PTMs target epigenetic and non-epigenetic pathways during ageing. In particular, epigenetic histone modification has critical implications for the ageing process and can modulate lifespan. Therefore, PTM-based therapeutics appear to be attractive pharmaceutical candidates to reduce the burden of ageing-related diseases. Several phosphorylation and acetylation inhibitors have already been FDA-approved for the treatment of other diseases and offer a unique potential to investigate both beneficial effects and possible side-effects. As an example, the most well-studied senolytic compounds dasatinib and quercetin, which have already been tested in Phase 1 pilot studies, also act as kinase inhibitors, targeting cellular senescence and increasing lifespan. Future studies need to carefully determine the best PTM-based candidates for the treatment of the "diseasome of ageing".
Collapse
Affiliation(s)
- Thomas Ebert
- Karolinska Institute, Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Stockholm, Sweden; University of Leipzig Medical Center, Medical Department III - Endocrinology, Nephrology, Rheumatology, Leipzig, Germany.
| | - Ngoc Tran
- University of Glasgow, Wolfson Wohl Cancer Research Centre, College of Medical, Veterinary & Life Sciences, Institute of Cancer Sciences, Glasgow, UK
| | - Leon Schurgers
- Department of Biochemistry, Cardiovascular Research School Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Peter Stenvinkel
- Karolinska Institute, Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Stockholm, Sweden
| | - Paul G Shiels
- University of Glasgow, Wolfson Wohl Cancer Research Centre, College of Medical, Veterinary & Life Sciences, Institute of Cancer Sciences, Glasgow, UK
| |
Collapse
|
17
|
Luo J, Li L, Chang B, Zhu Z, Deng F, Hu M, Yu Y, Lu X, Chen Z, Zuo D, Zhou J. Mannan-Binding Lectin via Interaction With Cell Surface Calreticulin Promotes Senescence of Activated Hepatic Stellate Cells to Limit Liver Fibrosis Progression. Cell Mol Gastroenterol Hepatol 2022; 14:75-99. [PMID: 35381393 PMCID: PMC9117817 DOI: 10.1016/j.jcmgh.2022.03.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 03/28/2022] [Accepted: 03/28/2022] [Indexed: 12/18/2022]
Abstract
BACKGROUND & AIMS Liver fibrosis represents a hallmark of most chronic liver diseases (CLD) triggered by recurrent liver injury and subsequent myofibroblast transdifferentiations of resident hepatic stellate cells (HSCs). Mannan-binding lectin (MBL) is potentially involved in hepatic fibrosis in CLD through unclear mechanisms. Therefore, we investigated the crosstalk between MBL and HSCs, and the consequent effects on fibrosis progression. METHODS Samples from patients with liver cirrhosis were collected. MBL deficiency (MBL-/-) and wild-type (WT) C57BL/6J mice were used to construct a CCl4-induced liver fibrosis model. Administration of MBL-expressing, liver-specific, adeno-associated virus was performed to restore hepatic MBL expression in MBL-/- mice. The human HSC line LX-2 was used for in vitro experiments. RESULTS MBL levels in patients with liver cirrhosis were correlated with disease severity. In the CCl4-induced liver fibrosis model, MBL-/- mice showed severer liver fibrosis accompanied by reduced senescent activated HSCs in liver tissue compared with WT mice, which could be inhibited by administering MBL-expressing, liver-specific, adeno-associated virus. Moreover, depleting senescent cells with senolytic treatment could abrogate these differences owing to MBL absence. Furthermore, MBL could interact directly with calreticulin associated with low-density lipoprotein receptor-related protein 1 on the cell surface of HSCs, which further promotes senescence in HSCs by up-regulating the mammalian target of rapamycin/p53/p21 signaling pathway. CONCLUSIONS MBL as a newfound senescence-promoting modulator and its crosstalk with HSCs in the liver microenvironment is essential for the control of hepatic fibrosis progression, suggesting its potential therapeutic use in treating CLD associated with liver fibrosis.
Collapse
Affiliation(s)
- Jialiang Luo
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Lei Li
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Bo Chang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhengyumeng Zhu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Fan Deng
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Mengyao Hu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yu Yu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiao Lu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhengliang Chen
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Daming Zuo
- Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China.
| | - Jia Zhou
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
18
|
Regulation of Cr(VI)-Induced Premature Senescence in L02 Hepatocytes by ROS-Ca2+-NF-κB Signaling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7295224. [PMID: 35222804 PMCID: PMC8881123 DOI: 10.1155/2022/7295224] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/18/2022] [Accepted: 01/29/2022] [Indexed: 02/07/2023]
Abstract
Stress-induced premature senescence may be involved in the pathogeneses of acute liver injury. Hexavalent chromium [Cr(VI)], a common environmental pollutant related to liver injury, likely leads to premature senescence in L02 hepatocytes. However, the underlying mechanisms regarding hepatocyte premature senility in Cr(VI) exposure remain poorly understood. In this study, we found that chronic exposure of L02 hepatocytes to Cr(VI) led to premature senescence characterized by increased β-galactosidase activity, senescence-associated heterochromatin foci, G1 phase arrest, and decreased cell proliferation. Additionally, Cr(VI)-induced senescent L02 hepatocytes showed upregulated inflammation-related factors, such as IL-6 and fibroblast growth factor 23 (FGF23), which also exhibited reactive oxygen species (ROS) accumulation derived from mitochondria accompanied with increased concentration of intracellular calcium ions (Ca2+) and activity of nuclear factor kappa B (NF-κB). Of note is that ROS inhibition by N-acetyl-Lcysteine pretreatment not only alleviated Cr(VI)-induced premature senescence but also reduced the elevated intracellular Ca2+, activated NF-κB, and secretion of IL-6/FGF23. Intriguingly, the toxic effect of Cr(VI) upon premature senescence of L02 hepatocytes and increased levels of IL-6/FGF23 could be partially reversed by the intracellular Ca2+ chelator BAPTA-AM pretreatment. Furthermore, by utilizing the NF-κB inhibitor pyrrolidine dithiocarbamate (PDTC), we confirmed that NF-κB mediated IL-6/FGF23 to regulate the Cr(VI)-induced L02 hepatocyte premature senescence, whilst the concentration of intracellular Ca2+ was not influenced by PDTC. To the best of our knowledge, our data reports for the first time the role of ROS-Ca2+-NF-κB signaling pathway in Cr(VI)-induced premature senescence. Our results collectively shed light on further exploration of innovative intervention strategies and treatment targeting Cr(VI)-induced chronic liver damage related to premature senescence.
Collapse
|
19
|
Zhao T, Zhu Y, Yao L, Liu L, Li N. IGF-1 alleviates CCL4-induced hepatic cirrhosis and dysfunction of intestinal barrier through inhibition TLR4/NF-κB signaling mediated by down-regulation HMGB1. Ann Hepatol 2021; 26:100560. [PMID: 34653689 DOI: 10.1016/j.aohep.2021.100560] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/23/2021] [Accepted: 04/24/2021] [Indexed: 02/04/2023]
Abstract
INTRODUCTION AND OBJECTIVES Cirrhosis has gradually become a serious public health issue, especially the national prevalence of cirrhosis was 29.2% in northwest China. Recent evidence has revealed that intestinal barrier (IB) dysfunction results from and contributes to cirrhosis. Our previous results have indicated that insulin-like growth factors (IGF-1) improved the impaired IB function and downregulated high mobility group protein box-1 (HMGB-1). Nevertheless, the role of the IGF-1/HMGB1 axis in cirrhosis remains largely unknown. MATERIALS AND METHODS Western blotting and qRT-PCR were used to detect protein and mRNA levels of related genes. The levels of AST, ALT, IL-1β, and TNF-α were examined using commercial kits. Immunofluorescence was used to evaluate the expression of HMGB1 in tissues. RESULTS In carbon tetrachloride (CCl4)-treated rat, the levels of AST (380.12 vs. 183.97), ALT (148.12 vs. 53.56), IL-1β (155.94 vs. 55.60), and TNF-α (155.00 vs. 48.90) were significantly increased compared with the control group, while IGF-1 treatment significantly alleviated CCL4-induced inflammatory response and IB dysfunction by downregulating HMGB1-mediated the TLR4/MyD88/NF-κB signaling pathway. In vitro experiments, HMGB1 treatment promoted inflammatory cytokines secretion and reduced cell viability and tight junctions by activating the TLR4/MyD88/NF-κB signaling pathway in Caco-2 cells, but IGF-1 alleviated these effects. CONCLUSION Our findings suggest that IGF-1 might serve as a potential therapeutic target for cirrhosis and IB dysfunction via inactivation of the TLR4/MyD88/NF-κB pathway through down-regulation HMGB1.
Collapse
Affiliation(s)
- Tianyu Zhao
- Department of Infectious Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China
| | - Ying Zhu
- Integrated Traditional Chinese and Western Medicine Liver Center, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China.
| | - Liying Yao
- Department of Infectious Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China
| | - Liu Liu
- Integrated Traditional Chinese and Western Medicine Liver Center, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China
| | - Na Li
- Department of Infectious Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China
| |
Collapse
|
20
|
Gao H, Liu X, Tian K, Meng Y, Yu C, Peng Y. Insight into the Protective Effect of Salidroside against H 2O 2-Induced Injury in H9C2 Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:1060271. [PMID: 34887995 PMCID: PMC8651377 DOI: 10.1155/2021/1060271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/17/2021] [Accepted: 10/31/2021] [Indexed: 11/18/2022]
Abstract
Salidroside is the important active ingredient of Rhodiola species, which shows a wide range of pharmacological activities such as antioxidative stress, anti-inflammation, and antiliver fibrosis. In this paper, we aimed to study the protective effect and mechanism of salidroside against H2O2-induced oxidative damage in H9C2 cells by determining cell proliferation rate, intracellular reactive oxygen species (ROS) level, antioxidant enzyme activities, and the expression of apoptosis-related proteins. The results showed that salidroside significantly alleviated cell growth inhibition induced by H2O2 treatment in H9C2 cells, decreased the levels of intracellular ROS and malondialdehyde (MDA), and increased the activity of superoxide dismutase (SOD) and catalase (CAT); meanwhile, salidroside upregulated the expression of Bcl-2 while downregulated the expression of Bax, p53, and caspase-3 in H2O2-treated H9C2 cells. Furthermore, the antiapoptotic effect of salidroside was almost eliminated by the knockdown of Bcl-2. In the further exploration, the Bcl-2 expression was decreased by the p53 overexpression and increased by p53 knockdown in H2O2-treated H9C2 cells. Consequently, salidroside could protect H9C2 cells against H2O2-induced oxidative damage, and the underlying mechanism may be related to scavenging intracellular ROS, increasing the activities of intracellular antioxidant enzymes and inhibiting the expression of apoptosis-related proteins.
Collapse
Affiliation(s)
- Hui Gao
- Department of Pharmacology, School of Medicine, Shaoxing University, Shaoxing 312000, China
- Department of Pharmacology, School of Medicine, Jishou University, Jishou 416000, China
| | - Xueping Liu
- Department of Pharmacology, School of Medicine, Guangxi University of Science and Technology, Liuzhou 545006, China
| | - Kunming Tian
- Department of Environmental Toxicity, Zunyi Medical University, Zunyi 563006, China
| | - Yichong Meng
- Department of Pharmacology, School of Medicine, Shaoxing University, Shaoxing 312000, China
| | - Cuicui Yu
- Tibet Agricultural Science and Technology Innovation Park, Lhasa, 850000 Tibet, China
| | - Yingfu Peng
- Department of Pharmacology, School of Medicine, Jishou University, Jishou 416000, China
| |
Collapse
|
21
|
Thai SF, Jones CP, Robinette BL, Ren H, Vallanat B, Fisher AA, Kitchin KT. Effects of Silver Nanoparticles and Silver Nitrate on mRNA and microRNA Expression in Human Hepatocellular Carcinoma Cells (HepG2). JOURNAL OF NANOSCIENCE AND NANOTECHNOLOGY 2021; 21:5414-5428. [PMID: 33980351 PMCID: PMC10563035 DOI: 10.1166/jnn.2021.19481] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
In order to understand toxicity of nano silver, human hepatocellular carcinoma (HepG2) cells were treated either with silver nitrate (AgNO₃) or with nano silver capped with glutathione (Ag-S) at various concentration. Differentially expressed genelists for mRNA and microRNA were obtained through Illumina RNA sequencing and DEseq data analyses. Both treatments showed non-linear dose response relationships for mRNA and microRNA. Gene expression analysis showed signaling pathways common to both nano Ag-S and AgNO₃, such as cell cycle regulation, DNA damage response and cancer related pathways. But, nano Ag-S caused signaling pathway changes that were not altered by AgNO₃ such as NRF2-mediated oxidative stress response inflammation, cell membrane signaling, and cell proliferation. Nano Ag-S also affected p53 signaling, survival, apoptosis, tissue repair, lipid synthesis, angiogenesis, liver fibrosis and tumor development. Several of the pathways affected by nano Ag-S are hypothesized as major contributors to nanotoxicity. MicroRNA target filter analysis revealed additional affected pathways that were not reflected in the mRNA expression response alone, including DNA damage signaling, genomic stability, ROS, cell cycle, ubiquitination, DNA methylation, cell proliferation and fibrosis for AgNO₃; and cell cycle regulation, P53 signaling, cell proliferation, survival, apoptosis, tissue repair and so on for nano Ag-S. These pathways may be mediated by microRNA repression of protein translation.Our study clearly showed that the addition of microRNA profiling increased the numbers of signaling pathways discovered that affected by the treatments on HepG2 cells and gave US a better picture of the effects of these reagents in the cells.
Collapse
Affiliation(s)
- Sheau-Fung Thai
- Center for Computational Toxicology and Exposure, US Environmental Protection Agency, 109 TWAlexander Dr, Durham NC 27709, USA
| | - Carlton P Jones
- Center for Computational Toxicology and Exposure, US Environmental Protection Agency, 109 TWAlexander Dr, Durham NC 27709, USA
| | - Brian L Robinette
- Center for Computational Toxicology and Exposure, US Environmental Protection Agency, 109 TWAlexander Dr, Durham NC 27709, USA
| | - Hongzu Ren
- Center for Public Health and Environmental Assessment, US Environmental Production Agency, 109 TW Alexander Dr., Durham NC 27709, USA
| | | | - Anna A Fisher
- Center for Public Health and Environmental Assessment, US Environmental Production Agency, 109 TW Alexander Dr., Durham NC 27709, USA
| | - Kirk T Kitchin
- US Environmental Protection Agency, Retired from EPA, Durham NC 27709, USA
| |
Collapse
|
22
|
Wang J, Jia R, Gong H, Celi P, Zhuo Y, Ding X, Bai S, Zeng Q, Yin H, Xu S, Liu J, Mao X, Zhang K. The Effect of Oxidative Stress on the Chicken Ovary: Involvement of Microbiota and Melatonin Interventions. Antioxidants (Basel) 2021; 10:1422. [PMID: 34573054 PMCID: PMC8472688 DOI: 10.3390/antiox10091422] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/30/2021] [Accepted: 09/03/2021] [Indexed: 12/12/2022] Open
Abstract
The poultry ovary is used as a classic model to study ovarian biology and ovarian cancer. Stress factors induced oxidative stress to cause follicle atresia, which may be a fundamental reason for the reduction in fertility in older laying hens or in aging women. In the present study, we set out to characterize the relationships between oxidative stress and ovarian function. Layers (62 weeks of age; BW = 1.42 ± 0.12 kg) were injected with tert-butyl hydroperoxide (tBHP) at 0 (CON) and 800 μmol/kg BW (oxidative stress group, OS) for 24 days and the role of melatonin (Mel) on tBHP-induced ovary oxidative stress was assessed through ovary culture in vitro. The OS (800 μmol/kg BW tert-butyl hydroperoxide) treatment decreased the reproduction performance and ovarian follicle numbers. OS decreased the expression of SIRT1 and increased the P53 and FoxO1 expression of the ovary. A decreased Firmicutes to Bacteroidetes ratio, enriched Marinifilaceae (family), Odoribacter (genus) and Bacteroides_plebeius (species) were observed in the cecum of the OS group. Using Mel in vitro enhanced the follicle numbers and decreased the ovary cell apoptosis induced by tBHP. In addition, it increased the expression of SIRT1 and decreased the P53 and FoxO1 expression. These findings indicated that oxidative stress could decrease the laying performance, ovarian function and influence gut microbiota and body metabolites in the layer model, while the melatonin exerts an amelioration the ovary oxidative stress through SIRT1-P53/FoxO1 pathway.
Collapse
Affiliation(s)
- Jianping Wang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (R.J.); (H.G.); (Y.Z.); (X.D.); (S.B.); (Q.Z.); (H.Y.); (S.X.); (X.M.); (K.Z.)
| | - Ru Jia
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (R.J.); (H.G.); (Y.Z.); (X.D.); (S.B.); (Q.Z.); (H.Y.); (S.X.); (X.M.); (K.Z.)
| | - Haojie Gong
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (R.J.); (H.G.); (Y.Z.); (X.D.); (S.B.); (Q.Z.); (H.Y.); (S.X.); (X.M.); (K.Z.)
| | - Pietro Celi
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville 3010, Australia;
| | - Yong Zhuo
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (R.J.); (H.G.); (Y.Z.); (X.D.); (S.B.); (Q.Z.); (H.Y.); (S.X.); (X.M.); (K.Z.)
| | - Xuemei Ding
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (R.J.); (H.G.); (Y.Z.); (X.D.); (S.B.); (Q.Z.); (H.Y.); (S.X.); (X.M.); (K.Z.)
| | - Shiping Bai
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (R.J.); (H.G.); (Y.Z.); (X.D.); (S.B.); (Q.Z.); (H.Y.); (S.X.); (X.M.); (K.Z.)
| | - Qiufeng Zeng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (R.J.); (H.G.); (Y.Z.); (X.D.); (S.B.); (Q.Z.); (H.Y.); (S.X.); (X.M.); (K.Z.)
| | - Huadong Yin
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (R.J.); (H.G.); (Y.Z.); (X.D.); (S.B.); (Q.Z.); (H.Y.); (S.X.); (X.M.); (K.Z.)
| | - Shengyu Xu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (R.J.); (H.G.); (Y.Z.); (X.D.); (S.B.); (Q.Z.); (H.Y.); (S.X.); (X.M.); (K.Z.)
| | - Jingbo Liu
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, China;
| | - Xiangbing Mao
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (R.J.); (H.G.); (Y.Z.); (X.D.); (S.B.); (Q.Z.); (H.Y.); (S.X.); (X.M.); (K.Z.)
| | - Keying Zhang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (R.J.); (H.G.); (Y.Z.); (X.D.); (S.B.); (Q.Z.); (H.Y.); (S.X.); (X.M.); (K.Z.)
| |
Collapse
|
23
|
Wang M, Liu H, Zhang X, Zhao W, Li D, Xu C, Wu Z, Xie F, Li X. Lack of Mof reduces acute liver injury by enhancing transcriptional activation of Igf1. J Cell Physiol 2021; 236:6559-6570. [PMID: 33634483 DOI: 10.1002/jcp.30332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/25/2021] [Accepted: 02/08/2021] [Indexed: 12/30/2022]
Abstract
Acute liver injury (ALI) is a rapid pathological process that may cause severe liver disease and may even be life-threatening. During ALI, the function of males absent on the first (MOF) has not yet been elucidated. In this study, we unveiled the expression pattern of MOF during carbon tetrachloride (CCl4 )-induced ALI and role of MOF in the regulation of liver regeneration. In the process of ALI, MOF is significantly overexpressed in the liver injury area. Knockdown of Mof attenuated CCl4 -induced ALI, and promoted liver cell proliferation, hepatic stellate cell activation and aggregation to the injured area, and liver fibrosis. Simultaneously, overexpression of Mof aggravated liver dysfunction caused by ALI. By directly binding to the promoter, MOF suppressed the transcriptional activation of Igf1. Knockdown of Mof promotes the expression of Igf1 and activates the Insulin-like growth factor 1 signaling pathway in the liver. Through this pathway, Knockdown of Mof reduces CCl4 -induced ALI and promotes liver regeneration. Our results provide the first demonstration for MOF contributing to ALI. Further understanding of the role of MOF in ALI may lead to new therapeutic strategies for ALI.
Collapse
Affiliation(s)
- Meng Wang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Advanced Medical Research Institute, Shandong University, Qingdao, Shandong, China
- Department of Cell and Neurobiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Haoyu Liu
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Advanced Medical Research Institute, Shandong University, Qingdao, Shandong, China
| | - Xu Zhang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Advanced Medical Research Institute, Shandong University, Qingdao, Shandong, China
| | - Wenbo Zhao
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Danyang Li
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Advanced Medical Research Institute, Shandong University, Qingdao, Shandong, China
- Department of Rehabilitation, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Chengpeng Xu
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Advanced Medical Research Institute, Shandong University, Qingdao, Shandong, China
| | - Zhen Wu
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Advanced Medical Research Institute, Shandong University, Qingdao, Shandong, China
| | - Fei Xie
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Advanced Medical Research Institute, Shandong University, Qingdao, Shandong, China
| | - Xiangzhi Li
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Advanced Medical Research Institute, Shandong University, Qingdao, Shandong, China
| |
Collapse
|
24
|
Fang J, Liang W. ASCs -derived exosomes loaded with vitamin A and quercetin inhibit rapid senescence-like response after acute liver injury. Biochem Biophys Res Commun 2021; 572:125-130. [PMID: 34364291 DOI: 10.1016/j.bbrc.2021.07.059] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/10/2021] [Accepted: 07/16/2021] [Indexed: 12/29/2022]
Abstract
Acute liver injury is a short-term burst of liver cell damage, which has many causes and complex mechanisms. Despite the unique ability of the liver to heal itself, there is still no effective treatment except liver transplantation for chronic liver injury or even liver failure caused by acute liver injury. Stem cell-derived exosomes are ideal drug carriers due to their unique immunomodulatory effects and structural characteristics. In this study, quercetin and vitamin A loaded adipose mesenchymal stem cells (ASCs)-derived exosomes were constructed and used to treat acute liver injury induced by CCl4 in mice. Quercetin enhances the therapeutic efficacy of exosomes, while vitamin A enhances the liver targeting of exosomes, and it was found that quercetin and vitamin A loaded mesenchymal stem cell exosomes reduce rapid senescence-like response induced by acute liver injury.
Collapse
Affiliation(s)
- Jia Fang
- Clinical Research Service Center, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, 450000, China.
| | - Wulong Liang
- The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, China.
| |
Collapse
|
25
|
Alpha lipoic acid priming enhances the hepatoprotective effect of adipose derived stem cells in CCl4 induced hepatic injury in-vitro. Eur J Pharmacol 2021; 906:174201. [PMID: 34118221 DOI: 10.1016/j.ejphar.2021.174201] [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: 10/26/2020] [Revised: 05/17/2021] [Accepted: 05/21/2021] [Indexed: 11/20/2022]
Abstract
Mesenchymal stem cells are known to support hepatic defense against liver fibrosis. However, the fibrosis induced oxidative microenvironment affects the proliferative, regenerative, and angiogenic properties of mesenchymal stem cells. Alpha lipoic acid (ALA) is a strong anti-oxidant which has been shown to ameliorate the adverse effects of fibrosis that otherwise can lead to severe liver problems like cirrhosis and liver failure. Here, we studied the protective role of ALA primed adipose derived stem cells (ADSCs) against carbon tetrachloride (CCl4) induced hepatotoxicity in primary hepatocytes in-vitro. Priming of ADSCs helped to abrogate the damaging effects of fibrosis induced oxidative stress as evidenced by significantly reduced levels of alkaline phosphatase (ALP), Alanine Aminotransferase (ALAT) along with decreased lactate dehydrogenase (LDH) release and improved superoxide dismutase (SOD) activity. ALA and ADSCs synergistically down-regulated the expression of Bax gene, an apoptosis regulator while enhancing cell proliferation by up-regulating the expression of Bcl2l1 gene. This treatment improved the expression of albumin (Alb), cytokeratin-8 (Ck8), and hepatic nuclear factor alpha (Hnf4α). Cytochrome P450 2E1 (Cyp2e1) and Alpha fetoprotein (Afp) were down-regulated to lessen the damage caused by CCl4 treatment. Furthermore, paracrine release of several growth factors like hepatocyte growth factor (HGF), vascular endothelial growth factor (VEGF), interleukin-6 (IL-6), tumor necrosis factor alpha (TNFα), and insulin growth factor (IGF) reinforced the improved response of primary hepatocytes against CCl4 induced hepatotoxicity in the presence of ALA primed ADSCs. This study suggests that ALA priming may improve the therapeutic potential of ADSCs against chronic liver problems by activating the nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream antioxidant factors heme oxygenase 1 (HO-1) and quinone acceptor oxidoreductase-1 (NQO1).
Collapse
|
26
|
Armandi A, Rosso C, Caviglia GP, Bugianesi E. Insulin Resistance across the Spectrum of Nonalcoholic Fatty Liver Disease. Metabolites 2021; 11:155. [PMID: 33800465 PMCID: PMC8000048 DOI: 10.3390/metabo11030155] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/01/2021] [Accepted: 03/01/2021] [Indexed: 02/07/2023] Open
Abstract
Insulin resistance (IR) is defined as a lower-than-expected response to insulin action from target tissues, leading to the development of type 2 diabetes through the impairment of both glucose and lipid metabolism. IR is a common condition in subjects with nonalcoholic fatty liver disease (NAFLD) and is considered one of the main factors involved in the pathogenesis of nonalcoholic steatohepatitis (NASH) and in the progression of liver disease. The liver, the adipose tissue and the skeletal muscle are major contributors for the development and worsening of IR. In this review, we discuss the sites and mechanisms of insulin action and the IR-related impairment along the spectrum of NAFLD, from simple steatosis to progressive NASH and cirrhosis.
Collapse
Affiliation(s)
| | | | | | - Elisabetta Bugianesi
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (A.A.); (C.R.); (G.P.C.)
| |
Collapse
|
27
|
Luo X, Bai Y, He S, Sun S, Jiang X, Yang Z, Lu D, Wei P, Liang Y, Peng C, Wang Y, Sheng R, Han S, Li X, Zhang B. Sirtuin 1 ameliorates defenestration in hepatic sinusoidal endothelial cells during liver fibrosis via inhibiting stress-induced premature senescence. Cell Prolif 2021; 54:e12991. [PMID: 33522656 PMCID: PMC7941223 DOI: 10.1111/cpr.12991] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 12/02/2020] [Accepted: 12/26/2020] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE Premature senescence is related to progerin and involves in endothelial dysfunction and liver diseases. Activating sirtuin 1 (SIRT1) ameliorates liver fibrosis. However, the mechanisms of premature senescence in defenestration of hepatic sinusoidal endothelial cells (HSECs) and how SIRT1 affects HSECs fenestrae remain elusive. METHODS We employed the CCl4 -induced liver fibrogenesis rat models and cultured primary HSECs in vitro, administered with the SIRT1-adenovirus vector, the activator of SIRT1 and knockdown NOX2. We measured the activity of senescence-associated β-galactosidase (SA-β-gal) in HSECs. Meanwhile, the protein expression of SIRT1, NOX2, progerin, Lamin A/C, Ac p53 K381 and total p53 was detected by Western blot, co-immunoprecipitation and immunofluorescence. RESULTS In vivo, premature senescence was triggered by oxidative stress during CCl4 -induced HSECs defenestration and liver fibrogenesis, whereas overexpressing SIRT1 with adenovirus vector lessened premature senescence to relieve CCl4 -induced HSECs defenestration and liver fibrosis. In vitro, HSECs fenestrae disappeared, with emerging progerin-associated premature senescence; these effects were aggravated by H2 O2 . Nevertheless, knockdown of NOX2, activation of SIRT1 with resveratrol and SIRT1-adenovirus vector inhibited progerin-associated premature senescence to maintain fenestrae through deacetylating p53. Furthermore, more Ac p53 K381 and progerin co-localized with the abnormal accumulation of actin filament (F-actin) in the nuclear envelope of H2 O2 -treated HSECs; in contrast, these effects were rescued by overexpressing SIRT1. CONCLUSION SIRT1-mediated deacetylation maintains HSECs fenestrae and attenuates liver fibrogenesis through inhibiting oxidative stress-induced premature senescence.
Collapse
Affiliation(s)
- Xiaoying Luo
- Department of GastroenterologyHenan Provincial People's HospitalPeople's Hospital of Zhengzhou UniversitySchool of Clinical MedicineHenan UniversityZhengzhouChina
- Microbiome LaboratoryHenan Provincial People's HospitalPeople's Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yangqiu Bai
- Department of GastroenterologyHenan Provincial People's HospitalPeople's Hospital of Zhengzhou UniversitySchool of Clinical MedicineHenan UniversityZhengzhouChina
| | - Shuli He
- Department of GastroenterologyHenan Provincial People's HospitalPeople's Hospital of Zhengzhou UniversitySchool of Clinical MedicineHenan UniversityZhengzhouChina
| | - Suofeng Sun
- Department of GastroenterologyHenan Provincial People's HospitalPeople's Hospital of Zhengzhou UniversitySchool of Clinical MedicineHenan UniversityZhengzhouChina
- Microbiome LaboratoryHenan Provincial People's HospitalPeople's Hospital of Zhengzhou UniversityZhengzhouChina
| | - Xiaoke Jiang
- Department of GastroenterologyHenan Provincial People's HospitalPeople's Hospital of Zhengzhou UniversitySchool of Clinical MedicineHenan UniversityZhengzhouChina
| | - Zhiyu Yang
- Department of GastroenterologyHenan Provincial People's HospitalPeople's Hospital of Zhengzhou UniversitySchool of Clinical MedicineHenan UniversityZhengzhouChina
- Microbiome LaboratoryHenan Provincial People's HospitalPeople's Hospital of Zhengzhou UniversityZhengzhouChina
| | - Di Lu
- Department of GastroenterologyHenan Provincial People's HospitalPeople's Hospital of Zhengzhou UniversitySchool of Clinical MedicineHenan UniversityZhengzhouChina
- Microbiome LaboratoryHenan Provincial People's HospitalPeople's Hospital of Zhengzhou UniversityZhengzhouChina
| | - Peiru Wei
- Department of GastroenterologyHenan Provincial People's HospitalPeople's Hospital of Zhengzhou UniversitySchool of Clinical MedicineHenan UniversityZhengzhouChina
| | - Yuan Liang
- Department of GastroenterologyHenan Provincial People's HospitalPeople's Hospital of Zhengzhou UniversitySchool of Clinical MedicineHenan UniversityZhengzhouChina
| | - Cong Peng
- Department of GastroenterologyHenan Provincial People's HospitalPeople's Hospital of Zhengzhou UniversitySchool of Clinical MedicineHenan UniversityZhengzhouChina
| | - Yaru Wang
- Department of GastroenterologyHenan Provincial People's HospitalPeople's Hospital of Zhengzhou UniversitySchool of Clinical MedicineHenan UniversityZhengzhouChina
| | - Ruli Sheng
- Department of GastroenterologyHenan Provincial People's HospitalPeople's Hospital of Zhengzhou UniversitySchool of Clinical MedicineHenan UniversityZhengzhouChina
| | - Shuangyin Han
- Department of GastroenterologyHenan Provincial People's HospitalPeople's Hospital of Zhengzhou UniversitySchool of Clinical MedicineHenan UniversityZhengzhouChina
| | - Xiuling Li
- Department of GastroenterologyHenan Provincial People's HospitalPeople's Hospital of Zhengzhou UniversitySchool of Clinical MedicineHenan UniversityZhengzhouChina
| | - Bingyong Zhang
- Department of GastroenterologyHenan Provincial People's HospitalPeople's Hospital of Zhengzhou UniversitySchool of Clinical MedicineHenan UniversityZhengzhouChina
| |
Collapse
|
28
|
Mata R, Yao Y, Cao W, Ding J, Zhou T, Zhai Z, Gao C. The Dynamic Inflammatory Tissue Microenvironment: Signality and Disease Therapy by Biomaterials. RESEARCH 2021; 2021:4189516. [PMID: 33623917 PMCID: PMC7879376 DOI: 10.34133/2021/4189516] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 12/22/2020] [Indexed: 12/14/2022]
Abstract
Tissue regeneration is an active multiplex process involving the dynamic inflammatory microenvironment. Under a normal physiological framework, inflammation is necessary for the systematic immunity including tissue repair and regeneration as well as returning to homeostasis. Inflammatory cellular response and metabolic mechanisms play key roles in the well-orchestrated tissue regeneration. If this response is dysregulated, it becomes chronic, which in turn causes progressive fibrosis, improper repair, and autoimmune disorders, ultimately leading to organ failure and death. Therefore, understanding of the complex inflammatory multiple player responses and their cellular metabolisms facilitates the latest insights and brings novel therapeutic methods for early diseases and modern health challenges. This review discusses the recent advances in molecular interactions of immune cells, controlled shift of pro- to anti-inflammation, reparative inflammatory metabolisms in tissue regeneration, controlling of an unfavorable microenvironment, dysregulated inflammatory diseases, and emerging therapeutic strategies including the use of biomaterials, which expand therapeutic views and briefly denote important gaps that are still prevailing.
Collapse
Affiliation(s)
- Rani Mata
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.,Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yuejun Yao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Wangbei Cao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Jie Ding
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Tong Zhou
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Zihe Zhai
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.,Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
29
|
Baig MT, Ghufran H, Mehmood A, Azam M, Humayun S, Riazuddin S. Vitamin E pretreated Wharton's jelly-derived mesenchymal stem cells attenuate CCl 4-induced hepatocyte injury in vitro and liver fibrosis in vivo. Biochem Pharmacol 2021; 186:114480. [PMID: 33617844 DOI: 10.1016/j.bcp.2021.114480] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 02/02/2021] [Accepted: 02/16/2021] [Indexed: 12/22/2022]
Abstract
Oxidative microenvironment in fibrotic liver alleviates the efficacious outcome of mesenchymal stem cells (MSCs)-based cell therapy. Recent evidence suggests that pharmacological pretreatment is a rational approach to harness the MSCs with higher therapeutic potential. Here, we investigated whether Vitamin E pretreatment can boost the antifibrotic effects of Wharton's jelly-derived MSCs (WJMSCs). We used rat liver-derived hepatocytes injured by CCl4 treatment in co-culture system with Vitamin E pretreated-WJMSCs (Vit E-WJMSCs) to evaluate the hepatoprotective effect of Vit E-WJMSCs. After 24 h of co-culturing, we found that Vit E-WJMSCs rescued injured hepatocytes as hepatocyte injury-associated medium (AST, ALT, and ALP) and mRNA (Cyp2e1, Hif1-α, and Il-1β) markers reduced to normal levels. Subsequently, CCl4-induced liver fibrosis rat models were employed to examine the antifibrotic potential of Vit E-WJMSCs. After 1 month of cell transplantation, it was revealed that Vit E-WJMSCs transplantation ceased fibrotic progression, as evident by improved hepatic architecture and functions, more significantly in comparison to naïve WJMSCs. In addition, Vit E-WJMSCs transplantation decreased the expressions of fibrosis-associated gene (Tgf-β1, α-Sma, and Col1α1) markers in the liver parenchyma. Intriguingly, the results of tracing experiments discovered that more WJMSCs engrafted in the Vit E-WJMSCs treated rat livers compared to naïve WJMSCs treated livers. These findings implicate that pretreatment of WJMSCs with Vitamin E improves their tolerance to hostile niche of fibrotic liver; thereby further enhancing their efficacy for hepatic fibrosis.
Collapse
Affiliation(s)
- Maria Tayyab Baig
- National Centre of Excellence in Molecular Biology, University of Punjab, 87-West Canal Bank Road, Lahore, Pakistan
| | - Hafiz Ghufran
- National Centre of Excellence in Molecular Biology, University of Punjab, 87-West Canal Bank Road, Lahore, Pakistan
| | - Azra Mehmood
- National Centre of Excellence in Molecular Biology, University of Punjab, 87-West Canal Bank Road, Lahore, Pakistan
| | - Maryam Azam
- National Centre of Excellence in Molecular Biology, University of Punjab, 87-West Canal Bank Road, Lahore, Pakistan
| | - Shamsa Humayun
- Fatima Jinnah Medical University, Sir Ganga Ram Hospital, Lahore, Pakistan
| | - Sheikh Riazuddin
- National Centre of Excellence in Molecular Biology, University of Punjab, 87-West Canal Bank Road, Lahore, Pakistan; Allama Iqbal Medical Research Centre, Jinnah Burn and Reconstructive Surgery Centre, Lahore, Pakistan.
| |
Collapse
|
30
|
Stanley TL, Fourman LT, Zheng I, McClure CM, Feldpausch MN, Torriani M, Corey KE, Chung RT, Lee H, Kleiner DE, Hadigan CM, Grinspoon SK. Relationship of IGF-1 and IGF-Binding Proteins to Disease Severity and Glycemia in Nonalcoholic Fatty Liver Disease. J Clin Endocrinol Metab 2021; 106:e520-e533. [PMID: 33125080 PMCID: PMC7823253 DOI: 10.1210/clinem/dgaa792] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Indexed: 12/11/2022]
Abstract
CONTEXT Growth hormone (GH) and IGF-1 help regulate hepatic glucose and lipid metabolism, and reductions in these hormones may contribute to development of nonalcoholic fatty liver disease (NAFLD). OBJECTIVE To assess relationships between hepatic expression of IGF1 and IGF-binding proteins (IGFBPs) and measures of glycemia and liver disease in adults with NAFLD. Secondarily to assess effects of GH-releasing hormone (GHRH) on circulating IGFBPs. DESIGN Analysis of data from a randomized clinical trial of GHRH. SETTING Two US academic medical centers. PARTICIPANTS Participants were 61 men and women 18 to 70 years of age with HIV-infection, ≥5% hepatic fat fraction, including 39 with RNA-Seq data from liver biopsy. MAIN OUTCOME MEASURES Hepatic steatosis, inflammation, and fibrosis by histopathology and measures of glucose homeostasis. RESULTS Hepatic IGF1 mRNA was significantly lower in individuals with higher steatosis and NAFLD Activity Score (NAS) and was inversely related to glucose parameters, independent of circulating IGF-1. Among the IGFBPs, IGFBP2 and IGFBP4 were lower and IGFBP6 and IGFBP7 (also known as IGFBP-related protein 1) were higher with increasing steatosis. Hepatic IGFBP6 and IGFBP7 mRNA levels were positively associated with NAS. IGFBP7 mRNA increased with increasing fibrosis. Hepatic IGFBP1 mRNA was inversely associated with glycemia and insulin resistance, with opposite relationships present for IGFBP3 and IGFBP7. GHRH increased circulating IGFBP-1 and IGFBP-3, but decreased IGFBP-2 and IGFBP-6. CONCLUSIONS These data demonstrate novel relationships of IGF-1 and IGFBPs with NAFLD severity and glucose control, with divergent roles seen for different IGFBPs. Moreover, the data provide new information on the complex effects of GHRH on IGFBPs.
Collapse
Affiliation(s)
- Takara L Stanley
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Lindsay T Fourman
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Isabel Zheng
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Colin M McClure
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Meghan N Feldpausch
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Martin Torriani
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Kathleen E Corey
- Liver Center, Gastroenterology Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Raymond T Chung
- Liver Center, Gastroenterology Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Hang Lee
- Biostatistics Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - David E Kleiner
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Colleen M Hadigan
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Steven K Grinspoon
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Correspondence and Reprint Requests: Steven K. Grinspoon, MD, Professor of Medicine, Harvard Medical School, MGH Endowed Chair in Neuroendocrinology and Metabolism, Chief, Metabolism Unit, Massachusetts General Hospital, 55 Fruit Street 5LON207, Boston, MA 02114, United States. E-mail:
| |
Collapse
|
31
|
Stavropoulos A, Varras M, Philippou A, Vasilakaki T, Varra VK, Varra FN, Tsavari A, Lazaris AC, Koutsilieris M. Immunohistochemical expression of insulin-like growth factor-1Ec in primary endometrial carcinoma: Association with PTEN, p53 and survivin expression. Oncol Lett 2020; 20:395. [PMID: 33193855 PMCID: PMC7656117 DOI: 10.3892/ol.2020.12258] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 09/16/2020] [Indexed: 12/21/2022] Open
Abstract
Chronic hyperinsulinemia due to insulin resistance and elevated levels of insulin-like growth factor (IGF)-1 and IGF-2 are suggestive of a significantly higher risk of endometrial carcinoma. There is a wealth of evidence showing differential expression of IGF-1 isoforms in various types of cancer. In the present study, 99 archived endometrial carcinoma tissue sections were retrospectively assessed by immunohistochemistry for IGF-1Ec isoform expression. Expression of IGF-1Ec was also assessed in nine cases of non-neoplastic endometrial tissue adjacent to the tumor, in 30 cases with normal endometrium and in 30 cases with endometrial hyperplasia. Furthermore, the association between IGF-1Ec and the concurrent expression of phosphatase and tensin homologue deleted on chromosome 10 (PTEN), p53 or survivin was assessed, as well as their combined expression in association with clinicopathological variables. In endometrial carcinoma, IGF-1Ec expression was high in non-endometrioid carcinoma (serous papillary or clear cell carcinoma) compared with that in endometrioid adenocarcinoma. IGF-1Ec expression was also high in the presence of tumoral necrosis. Furthermore, there was a significant correlation between the histological differentiation and the sum of staining intensity and the number of IGF-1Ec immunopositive cells in endometrial carcinoma. There was a moderate negative correlation between co-expression of IGF-1Ec and PTEN, for both the number of immunopositive cells (P=0.006, ρ=−0.343) and the sum of staining (scores and intensity; P=0.006, ρ=−0.343). Furthermore, there was a positive correlation between the sum of staining (scores and intensity) and co-expression of IGF-1Ec and survivin (P=0.043, ρ=0.225). However, there was no association between concomitant expression of IGF-1Ec and p53. These results emphasized the importance of IGF-1Ec expression during development of non-estrogen dependent endometrial adenocarcinoma. IGF-1Ec and PTEN may function opposingly during endometrial carcinogenesis. By contrast, IGF-1Ec and survivin may share common molecular pathways and may promote, in parallel, tumoral development.
Collapse
Affiliation(s)
- Aggelis Stavropoulos
- Fourth Obstetrics and Gynecology Department, 'Elena Venizelou' General Hospital, Athens 11521, Greece
| | - Michail Varras
- Fifth Obstetrics and Gynecology Department, 'Elena Venizelou' General Hospital, Athens 11521, Greece
| | - Anastassios Philippou
- Department of Physiology, Medical School, National Kapodistrian University, Athens 11527, Greece
| | - Thivi Vasilakaki
- Pathology Department, 'Tzaneio' General Hospital, Piraeus 18536, Greece
| | | | - Fani-Niki Varra
- Pharmacy Department, Frederick University, Nicosia 1036, Cyprus
| | | | - Andreas C Lazaris
- First Pathology Department, Medical School, National Kapodistrian University, Athens 11527, Greece
| | - Michael Koutsilieris
- Department of Physiology, Medical School, National Kapodistrian University, Athens 11527, Greece
| |
Collapse
|
32
|
Zhang J, Ye J, Yuan C, Fu Q, Zhang F, Zhu X, Wang L, Gao P, Shu G, Wang S, Liu Q, Jiang Q. Hydrogen sulfide is a regulator of mammary gland development in prepubescent female mice. Mol Med Rep 2020; 22:4061-4069. [PMID: 33000185 DOI: 10.3892/mmr.2020.11462] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 07/09/2020] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to investigate the effects of exogenous H2S on mammary gland development in pubescent mice and to explore the underlying mechanism. The mouse mammary epithelial cell line HC11, along with C57BL/6J mice, were treated with different concentrations of sodium hydrosulfide (NaHS), which is a donor of H2S. The HC11 cell viability, pubescent mammary gland development, and the involvement of proliferative proteins and pathways were assessed by CCK‑8 assay, EdU assay, whole mount staining, H&E staining, western blotting and reverse transcription‑quantitative PCR. Both in vitro and in vivo, a low concentration of NaHS (100 µM in vitro; 9 mg/kg in vivo) significantly promoted the viability of HC11 cells and the development of mammary glands by increasing the expression of the proliferative markers cyclin D1/3 and proliferating cell nuclear antigen. However, a high concentration of NaHS (1,000 µM in vitro; 18 mg/kg in vivo) inhibited HC11 cell viability, mammary gland development and the expression levels of proteins involved in proliferation. Subsequent experiments revealed that NaHS regulated the phosphatidylinositol 3‑kinase (PI3K)/protein kinase B (Akt)‑mammalian target of rapamycin (mTOR) signaling pathway during this process. In vivo, intraperitoneal injection of low concentration NaHS (9 mg/kg) activated the PI3K/Akt‑mTOR pathway in mammary glands of pubescent mice, increased the secretion of insulin‑like growth factor 1 (IGF‑1) and estradiol (E2), and then stimulated mammary gland ductal development. Whereas a high concentration of NaHS (18 mg/kg) elicited the opposite effects to those of low‑dose NaHS. In conclusion, the present study demonstrated that exogenous H2S supplied by NaHS may exert bidirectional effects on mammary gland ductal development; promoting ductal development at a low concentration and inhibiting it at a high concentration. The effects of H2S may occur via the intracellular PI3K/Akt‑mTOR signaling pathway, or by regulation of the secretion of IGF‑1 and E2.
Collapse
Affiliation(s)
- Jing Zhang
- College of Animal Science, Shanxi Agricultural University, Jinzhong, Shanxi 030801, P.R. China
| | - Jiayi Ye
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
| | - Cong Yuan
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
| | - Qin Fu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
| | - Fenglin Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
| | - Xiaotong Zhu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
| | - Lina Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
| | - Ping Gao
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
| | - Gang Shu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
| | - Songbo Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
| | - Qiang Liu
- College of Animal Science, Shanxi Agricultural University, Jinzhong, Shanxi 030801, P.R. China
| | - Qingyan Jiang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
| |
Collapse
|
33
|
Shen J, Zhu X, Liu H. MiR-483 induces senescence of human adipose-derived mesenchymal stem cells through IGF1 inhibition. Aging (Albany NY) 2020; 12:15756-15770. [PMID: 32805717 PMCID: PMC7467354 DOI: 10.18632/aging.103818] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 07/06/2020] [Indexed: 12/19/2022]
Abstract
Human adipose-derived mesenchymal stem cells (hADSCs) are an ideal source of seed cells for regenerative applications and tissue engineering. However, long-term in vitro culture of hADSCs reduces their quantity and quality, which lessens their value in research and clinical applications. The molecular mechanisms underlying this biological process are poorly defined. Recently identified microRNAs (miRNAs) have emerged as critical modulators of cellular senescence. In this study, we examined the changes in hADSCs undergoing senescence. Significant miR-483-3p upregulation was noted during in vitro passaging of hADSCs, which correlated with the adipogenic differentiation and cellular senescence. Knockdown of miR-483-3p retarded the adipogenic differentiation potential of hADSCs and reduced cellular senescence. Dual-luciferase reporter assays identified insulin-like growth factor-1 (IGF1) as the target gene of miR-483-3p. IGF1 inhibition confirmed its inhibitory effects on replicative senescence in hADSCs. In conclusion, our study revealed essential regulatory roles of miR-483-3p in the adipogenesis and aging of hADSCs mediated by targeting IGF1.
Collapse
Affiliation(s)
- Junyan Shen
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, China
| | - Xiaoqi Zhu
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, China
| | - Hailiang Liu
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, China
| |
Collapse
|
34
|
Poudel SB, Dixit M, Neginskaya M, Nagaraj K, Pavlov E, Werner H, Yakar S. Effects of GH/IGF on the Aging Mitochondria. Cells 2020; 9:cells9061384. [PMID: 32498386 PMCID: PMC7349719 DOI: 10.3390/cells9061384] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 12/11/2022] Open
Abstract
The mitochondria are key organelles regulating vital processes in the eukaryote cell. A decline in mitochondrial function is one of the hallmarks of aging. Growth hormone (GH) and the insulin-like growth factor-1 (IGF-1) are somatotropic hormones that regulate cellular homeostasis and play significant roles in cell differentiation, function, and survival. In mammals, these hormones peak during puberty and decline gradually during adulthood and aging. Here, we review the evidence that GH and IGF-1 regulate mitochondrial mass and function and contribute to specific processes of cellular aging. Specifically, we discuss the contribution of GH and IGF-1 to mitochondrial biogenesis, respiration and ATP production, oxidative stress, senescence, and apoptosis. Particular emphasis was placed on how these pathways intersect during aging.
Collapse
Affiliation(s)
- Sher Bahadur Poudel
- David B. Kriser Dental Center, Department of Molecular Pathobiology, New York University College of Dentistry New York, NY 10010–4086, USA; (S.B.P.); (M.D.); (M.N.); (E.P.)
| | - Manisha Dixit
- David B. Kriser Dental Center, Department of Molecular Pathobiology, New York University College of Dentistry New York, NY 10010–4086, USA; (S.B.P.); (M.D.); (M.N.); (E.P.)
| | - Maria Neginskaya
- David B. Kriser Dental Center, Department of Molecular Pathobiology, New York University College of Dentistry New York, NY 10010–4086, USA; (S.B.P.); (M.D.); (M.N.); (E.P.)
| | - Karthik Nagaraj
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; (K.N.); (H.W.)
| | - Evgeny Pavlov
- David B. Kriser Dental Center, Department of Molecular Pathobiology, New York University College of Dentistry New York, NY 10010–4086, USA; (S.B.P.); (M.D.); (M.N.); (E.P.)
| | - Haim Werner
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; (K.N.); (H.W.)
| | - Shoshana Yakar
- David B. Kriser Dental Center, Department of Molecular Pathobiology, New York University College of Dentistry New York, NY 10010–4086, USA; (S.B.P.); (M.D.); (M.N.); (E.P.)
- Correspondence: ; Tel.: +212-998-9721
| |
Collapse
|
35
|
Banerjee J, Roy S, Dhas Y, Mishra N. Senescence-associated miR-34a and miR-126 in middle-aged Indians with type 2 diabetes. Clin Exp Med 2020; 20:149-158. [PMID: 31732824 DOI: 10.1007/s10238-019-00593-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 11/01/2019] [Indexed: 12/16/2022]
Abstract
Rapid urbanization and unhealthy dietary patterns critically increase the risk of type 2 diabetes (T2D) in middle-aged Indians. However, despite recent evidence of senescence-associated microRNAs (SA-miRNAs) in regulating complex pathways of ageing, their expressions in middle-aged Indians with T2D remain unexplored. Hence we aimed to investigate the changes in expressions of SA-miRNAs miR-34a and miR-126 in middle-aged T2D patients. A total of 30 T2D patients and 30 controls were recruited of age 31-50 years. The expressions of plasma miR-34a and miR-126 were determined by quantitative PCR. Oxidized LDL (OxLDL) and malondialdehyde (MDA) levels were quantified using enzyme-linked immunosorbent assay (ELISA). The effect of different glucose concentrations on miR-34a, miR-126, senescence-associated, and oxidative stress-responsive genes were also studied in an in vitro model of mice pancreatic β-cells. MiR-34a was significantly upregulated, whereas miR-126 was nonsignificantly reduced in T2D patients as compared to controls. T2D patients showed elevated levels of oxidative stress markers than controls. Analysis of cultured mice pancreatic β-cells exposed to high glucose showed significant upregulation of miR-34a, miR-126, p53, and superoxide dismutase 2 (SOD2). We found that circulating miR-34a levels and oxidative stress markers levels were elevated in the middle-aged Indians with T2D as compared to controls. The presence of diabetes may aggravate the normal ageing process in the middle-aged Indians. These SA-miRNAs can also be used to check the cellular dysfunctions and ageing of pancreatic β-cells.
Collapse
Affiliation(s)
- Joyita Banerjee
- Symbiosis School of Biological Sciences (Formerly Symbiosis School of Biomedical Sciences), Symbiosis International (Deemed University), Lavale, Pune, 412115, India
| | - Swagata Roy
- Symbiosis School of Biological Sciences (Formerly Symbiosis School of Biomedical Sciences), Symbiosis International (Deemed University), Lavale, Pune, 412115, India
| | - Yogita Dhas
- Symbiosis School of Biological Sciences (Formerly Symbiosis School of Biomedical Sciences), Symbiosis International (Deemed University), Lavale, Pune, 412115, India
| | - Neetu Mishra
- Symbiosis School of Biological Sciences (Formerly Symbiosis School of Biomedical Sciences), Symbiosis International (Deemed University), Lavale, Pune, 412115, India.
| |
Collapse
|
36
|
PAS kinase deficiency reduces aging effects in mice. Aging (Albany NY) 2020; 12:2275-2301. [PMID: 31974316 PMCID: PMC7041766 DOI: 10.18632/aging.102745] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 01/07/2020] [Indexed: 12/23/2022]
Abstract
Several signaling pathways may be affected during aging. All are regulated by nutrient levels leading to a decline in mitochondrial function and autophagy and to an increase in oxidative stress. PAS Domain Kinase (PASK) is a nutrient and bioenergetic sensor. We have previously found that PASK plays a role in the control of hepatic metabolic balance and mitochondrial homeostasis. To investigate PASK’s role in hepatic oxidative stress during aging, we analyzed the mitochondrial function, glucose tolerance, insulin resistance, and lipid-related parameters in aged PASK-deficient mice. Hepatic Pask mRNA decreased in step with aging, being undetectable in aged wild-type (WT) mice. Aged PASK-deficient mice recorded lower levels of ROS/RNS compared to aged WT. The regulators of mitochondrial biogenesis, PGC1a, SIRT1 and NRF2, decreased in aged WT, while aged PASK-deficient mice recorded a higher expression of NRF2, GCLm and HO1 proteins and CS activity under fasted conditions. Additionally, aged PASK-deficient mice recorded an overexpression of the longevity gene FoxO3a, and maintained elevated PCNA protein, suggesting that hepatic cell repair mechanisms might be functional. PASK-deficient mice have better insulin sensitivity and no glucose intolerance, as confirmed by a normal HOMA-IR index. PASK may be a good target for reducing damage during aging.
Collapse
|