1
|
Zhang B, Enriquez-Sarano M, Schaff HV, Michelena HI, Roos CM, Hagler MA, Zhang H, Casaclang-Verzosa G, Huang R, Bartoo A, Ranadive S, Joyner MJ, Pislaru S, Nkomo VT, Kremers WK, Araoz PA, Singh G, Walters MA, Hawkinson J, Cunningham KY, Sung J, Dunagan B, Ye Z, Miller JD. Reactivation of Oxidized Soluble Guanylate Cyclase as a Novel Treatment Strategy to Slow Progression of Calcific Aortic Valve Stenosis: Preclinical and Randomized Clinical Trials to Assess Safety and Efficacy. Circulation 2025; 151:913-930. [PMID: 39989354 DOI: 10.1161/circulationaha.123.066523] [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: 10/03/2023] [Accepted: 01/27/2025] [Indexed: 02/25/2025]
Abstract
BACKGROUND Pharmacological treatments for fibrocalcific aortic valve stenosis (FCAVS) have been elusive for >50 years. Here, we tested the hypothesis that reactivation of oxidized sGC (soluble guanylate cyclase), the primary receptor for nitric oxide, with ataciguat is a safe and efficacious strategy to slow progression of FCAVS. METHODS We used quantitative real-time reverse transcription polymerase chain reaction, Western blotting, and immunohistochemistry to characterize sGC signaling and the biological effects of ataciguat on signaling cascades related to nitric oxide, calcification, and fibrosis in excised human aortic valve tissue, aortic valve interstitial cells, and mouse aortic valves. We then conducted randomized, placebo-controlled phase I (14-day safety/tolerance) and phase II (6-month efficacy) trials in patients with moderate aortic valve stenosis. RESULTS In excised human tissue, we found robust losses in sGC signaling despite upregulation of sGC subunits. In vitro, ataciguat increased sGC signaling and reduced BMP2 (bone morphogenetic protein 2) signaling in aortic valve interstitial cells. In mice with established FCAVS, treatment with ataciguat attenuated BMP signaling and slowed progression of valve calcification and dysfunction. In a phase I, randomized, placebo-controlled trial, treatment with ataciguat for 2 weeks was safe and well tolerated in patients with moderate FCAVS (https://www.clinicaltrials.gov; Unique identifier: NCT02049203). In a separate phase II, randomized, placebo-controlled trial, treatment with ataciguat for 6 months slowed the progression of aortic valve calcification and tended to slow the progression of valvular and ventricular dysfunction in patients with moderate FCAVS (https://www.clinicaltrials.gov; Unique identifier: NCT02481258). CONCLUSIONS Collectively, this study highlights the therapeutic potential of the targeted restoration of the diseased/inactive form of sGC for treatment of FCAVS. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT02049203. URL: https://www.clinicaltrials.gov; Unique identifier: NCT02481258.
Collapse
Affiliation(s)
- Bin Zhang
- Departments of Surgery (B.Z., C.M.R., M.A.H., H.Z., G.C.-V., J.S., B.D., J.D.M.), Mayo Clinic, Rochester, MN
| | - Maurice Enriquez-Sarano
- Cardiovascular Diseases (M.E.-S., H.I.M., R.H., S.P., V.T.N., Z.Y.), Mayo Clinic, Rochester, MN
| | - Hartzell V Schaff
- Cardiovascular Surgery (H.V.S., B.D., J.D.M.), Mayo Clinic, Rochester, MN
| | - Hector I Michelena
- Cardiovascular Diseases (M.E.-S., H.I.M., R.H., S.P., V.T.N., Z.Y.), Mayo Clinic, Rochester, MN
| | - Carolyn M Roos
- Departments of Surgery (B.Z., C.M.R., M.A.H., H.Z., G.C.-V., J.S., B.D., J.D.M.), Mayo Clinic, Rochester, MN
| | - Michael A Hagler
- Departments of Surgery (B.Z., C.M.R., M.A.H., H.Z., G.C.-V., J.S., B.D., J.D.M.), Mayo Clinic, Rochester, MN
| | - Heyu Zhang
- Departments of Surgery (B.Z., C.M.R., M.A.H., H.Z., G.C.-V., J.S., B.D., J.D.M.), Mayo Clinic, Rochester, MN
| | - Grace Casaclang-Verzosa
- Departments of Surgery (B.Z., C.M.R., M.A.H., H.Z., G.C.-V., J.S., B.D., J.D.M.), Mayo Clinic, Rochester, MN
| | - Runqing Huang
- Cardiovascular Diseases (M.E.-S., H.I.M., R.H., S.P., V.T.N., Z.Y.), Mayo Clinic, Rochester, MN
| | | | | | | | - Sorin Pislaru
- Cardiovascular Diseases (M.E.-S., H.I.M., R.H., S.P., V.T.N., Z.Y.), Mayo Clinic, Rochester, MN
| | - Vuyisile T Nkomo
- Cardiovascular Diseases (M.E.-S., H.I.M., R.H., S.P., V.T.N., Z.Y.), Mayo Clinic, Rochester, MN
| | - Walter K Kremers
- Quantitative Health Sciences (W.K.K.), Mayo Clinic, Rochester, MN
| | | | - Gurpreet Singh
- Department of Medicinal Chemistry (G.S., M.A.W., J.H.), University of Minnesota, Minneapolis, MN
| | - Michael A Walters
- Department of Medicinal Chemistry (G.S., M.A.W., J.H.), University of Minnesota, Minneapolis, MN
| | - Jon Hawkinson
- Department of Medicinal Chemistry (G.S., M.A.W., J.H.), University of Minnesota, Minneapolis, MN
| | - Kevin Y Cunningham
- Bioinformatics and Computational Biology Program (K.Y.C.), University of Minnesota, Minneapolis, MN
| | - Jaeyun Sung
- Departments of Surgery (B.Z., C.M.R., M.A.H., H.Z., G.C.-V., J.S., B.D., J.D.M.), Mayo Clinic, Rochester, MN
| | - Brandon Dunagan
- Departments of Surgery (B.Z., C.M.R., M.A.H., H.Z., G.C.-V., J.S., B.D., J.D.M.), Mayo Clinic, Rochester, MN
- Cardiovascular Surgery (H.V.S., B.D., J.D.M.), Mayo Clinic, Rochester, MN
| | - Zi Ye
- Cardiovascular Diseases (M.E.-S., H.I.M., R.H., S.P., V.T.N., Z.Y.), Mayo Clinic, Rochester, MN
| | - Jordan D Miller
- Departments of Surgery (B.Z., C.M.R., M.A.H., H.Z., G.C.-V., J.S., B.D., J.D.M.), Mayo Clinic, Rochester, MN
- Cardiovascular Surgery (H.V.S., B.D., J.D.M.), Mayo Clinic, Rochester, MN
- Physiology and Biomedical Engineering (J.D.M.), Mayo Clinic, Rochester, MN
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN (J.D.M.)
| |
Collapse
|
2
|
Al-Maskari R, Abdelrahman AM, Ali H, Manoj P, Al Suleimani Y. Nephroprotective effects of the soluble guanylyl cyclase stimulator, riociguat in doxorubicin-induced acute kidney injury in rats. Toxicol Rep 2024; 13:101800. [PMID: 39606778 PMCID: PMC11600010 DOI: 10.1016/j.toxrep.2024.101800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/21/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
This study aimed to investigate the potential protective effects of riociguat, a soluble guanylyl cyclase (sGC) stimulator, on kidney function and structure in rats with acute kidney injury (AKI) induced by the chemotherapeutic drug doxorubicin (DX). Rats were subjected to a single intraperitoneal injection of DX (13.5 mg/kg) on the 5th day, either alone or in combination with low-dose riociguat (3 mg/kg/day), or high-dose riociguat (10 mg/kg/day) for 8 consecutive days. Various markers related to kidney function, oxidative stress, and inflammation were measured in plasma and urine. Kidney tissues were examined histopathologically. DX-induced nephrotoxicity was characterized by increased plasma urea, creatinine, uric acid and neutrophil gelatinase-associated lipocalin (NGAL). DX also decreased creatinine clearance and albumin levels and increased urinary N-acetyl-β-D-glucosaminidase (NAG) activity. Furthermore, DX increased the inflammatory markers interleukin 1 beta (IL-1 β), interleukin 6 (IL-6) and tumor necrosis factor alpha (TNF-α). DX further induced oxidative stress injury evidenced by decreased glutathione reductase (GR) activity, total antioxidant capacity (TAC), superoxide dismutase (SOD) and catalase levels and increased malondialdehyde (MDA) levels. Concomitant treatment with riociguat ameliorated these DX-induced changes with parallel histopathological improvements but the effects were more favorable with high-dose riociguat. The observed renoprotective effects of riociguat can be partly attributed to the anti-inflammatory and anti-oxidant properties of this drug.
Collapse
Affiliation(s)
- Raya Al-Maskari
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, P.O. Box 35, Al Khod 123, Oman
| | - Aly M. Abdelrahman
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, P.O. Box 35, Al Khod 123, Oman
| | - Haytham Ali
- Department of Animal and Veterinary Sciences, College of Agricultural and Marine Sciences, Sultan Qaboos University, Muscat 123, Oman
| | - Priyadarsini Manoj
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, P.O. Box 35, Al Khod 123, Oman
| | - Yousuf Al Suleimani
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, P.O. Box 35, Al Khod 123, Oman
| |
Collapse
|
3
|
Khanna D, de Vries-Bouwstra J, Hoffmann-Vold AM, Kuwana M, Low AHL, Proudman S, Flack M, Kukreja A, Fagan N, Distler O. A Phase II study of avenciguat, a novel soluble guanylate cyclase activator, in patients with systemic sclerosis: Study design and rationale of the VITALISScE™ study. JOURNAL OF SCLERODERMA AND RELATED DISORDERS 2024:23971983241291923. [PMID: 39544899 PMCID: PMC11559521 DOI: 10.1177/23971983241291923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 11/17/2024]
Abstract
Introduction Systemic sclerosis is a rare autoimmune connective tissue disease characterised by (1) microvasculopathy; (2) immune dysregulation; and (3) progressive fibrosis of the skin and internal organs. Soluble guanylate cyclase plays an important role in maintaining vascular and immunological homeostasis and preventing organ fibrosis. Pharmacological modulation of soluble guanylate cyclase with soluble guanylate cyclase stimulators has shown anti-inflammatory and antifibrotic effects in animal models of systemic sclerosis, with a trend towards clinical efficacy in a Phase II study (RISE-SSc). However, the efficacy of soluble guanylate cyclase stimulators may be reduced under conditions of hypoxia and oxidative stress. Soluble guanylate cyclase activators have the potential to overcome this limitation. This paper describes the study design of VITALISScE™, a Phase II clinical trial assessing the efficacy, safety and tolerability of avenciguat, a novel soluble guanylate cyclase activator in patients with active systemic sclerosis at risk of progression. Methods The VITALISScE™ study (NCT05559580) is evaluating the action of avenciguat on all three aspects of systemic sclerosis pathophysiology. The primary endpoint is the rate of decline in forced vital capacity (mL) over 48 weeks. Secondary endpoints include absolute change from baseline at Week 48 in modified Rodnan skin score, Health Assessment Questionnaire Disability Index score and the proportion of responders based on the revised Composite Response Index in Systemic Sclerosis. Additional endpoints include a composite assessment of Raynaud's phenomenon, digital ulcer burden, functional outcomes and quality of life, safety, pharmacokinetics, and biomarkers associated with systemic sclerosis and the mechanism of action of avenciguat. Results VITALISScE™ is an ongoing, multicentre (180 sites; 38 countries), placebo-controlled, double-blind, parallel-group, Phase II clinical study. Recruitment is currently ongoing. Conclusions The VITALISScE™ study is assessing the efficacy, safety and tolerability of avenciguat in patients with active systemic sclerosis at risk of progression. Results will inform further development of avenciguat. Trial Registration VITALISScE™; EU CT No. 2022-500332-11-00; Clinicaltrials.gov: NCT05559580 (https://www.clinicaltrials.gov/study/NCT05559580).
Collapse
Affiliation(s)
- Dinesh Khanna
- Department of Internal Medicine, University of Michigan Scleroderma Clinic, Ann Arbor, MI, USA
| | | | - Anna-Maria Hoffmann-Vold
- Department of Rheumatology, Oslo University Hospital, Oslo, Norway
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Masataka Kuwana
- Department of Allergy and Rheumatology, Nippon Medical School Graduate School of Medicine, Tokyo, Japan
| | - Andrea Hsiu Ling Low
- Department of Rheumatology and Immunology, Singapore General Hospital, Singapore
- Duke-National University of Singapore Medical School, Singapore
| | - Susanna Proudman
- Discipline of Medicine, University of Adelaide and Rheumatology Unit, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Mary Flack
- TA Inflammation Medicine, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA
| | - Anjli Kukreja
- Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA
| | - Nora Fagan
- Global Biostatistics & Data Sciences, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA
| | - Oliver Distler
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
4
|
Zeng X, Zhang H, Xu T, Mei X, Wang X, Yang Q, Luo Z, Zeng Q, Xu D, Ren H. Vericiguat attenuates doxorubicin-induced cardiotoxicity through the PRKG1/PINK1/STING axis. Transl Res 2024; 273:90-103. [PMID: 39059761 DOI: 10.1016/j.trsl.2024.07.005] [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: 01/16/2024] [Revised: 05/18/2024] [Accepted: 07/14/2024] [Indexed: 07/28/2024]
Abstract
Doxorubicin (DOX) is restricted due to its severe cardiotoxicity. There is still a lack of viable and effective drugs to prevent or treat DOX-induced cardiotoxicity(DIC). Vericiguat is widely used to treat heart failure with reduced ejection fraction. However, it is not clear whether vericiguat can improve DIC. In the present study, we constructed a DIC model using mice and neonatal rat cardiomyocytes and found that vericiguat ameliorated DOX-induced cardiac insufficiency in mice, restored DOX-induced mitochondrial dysfunction in neonatal rat cardiomyocytes, and inhibited the expression of inflammatory factors. Further studies showed that vericiguat improved mitochondrial dysfunction and reduced mtDNA leakage into the cytoplasm by up-regulating PRKG1, which activated PINK1 and then inhibited the STING/IRF3 pathway to alleviate DIC. These findings demonstrate for the first time that vericiguat has therapeutic potential for the treatment of DIC.
Collapse
Affiliation(s)
- Xianghui Zeng
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Key Laboratory For Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China; Department of Cardiology, Ganzhou Hospital of Traditional Chinese Medicine, Ganzhou, Jiangxi, China
| | - Hao Zhang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Key Laboratory For Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China
| | - Tianyu Xu
- NHC Key Laboratory of Assisted Circulation, Department of Cardiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiyuan Mei
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Key Laboratory For Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China
| | - Xiao Wang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Key Laboratory For Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China
| | - Qiling Yang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Key Laboratory For Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China
| | - Zhen Luo
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Key Laboratory For Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China
| | - Qingchun Zeng
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Key Laboratory For Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China
| | - Dingli Xu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Key Laboratory For Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China.
| | - Hao Ren
- Key Laboratory For Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China; Department of Rheumatology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
5
|
Zhou Z, Mao X, Jiang C, Li W, Zhou T, Liu M, Sun S, Wang M, Dong N, Wu Q, Zhou H. Deficiencies in corin and atrial natriuretic peptide-mediated signaling impair endochondral ossification in bone development. Commun Biol 2024; 7:1380. [PMID: 39443661 PMCID: PMC11500007 DOI: 10.1038/s42003-024-07077-6] [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: 03/19/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024] Open
Abstract
Corin is a protease that activates atrial natriuretic peptide (ANP), a hormone in cardiovascular homeostasis. Structurally, ANP is similar to C-type natriuretic peptide (CNP) crucial in bone development. Here, we examine the role of corin and ANP in chondrocyte differentiation and bone formation. We show that in Corin and Nppa (encoding ANP) knockout (KO) mice, chondrocyte differentiation is impaired, resulting in shortened limb long bones. In adult mice, Corin and Nppa deficiency impairs bone density and microarchitecture. Molecular studies in cartilages from newborn Corin and Nppa KO mice and in cultured chondrocytes indicate that corin and ANP act in chondrocytes via cGMP-dependent protein kinase G signaling to inhibit mitogen-activated protein kinase phosphorylation and stimulate glycogen synthase kinase-3β phosphorylation and β-catenin upregulation. These results indicate that corin and ANP signaling regulates chondrocyte differentiation in bone development and homeostasis, suggesting that enhancing ANP signaling may improve bone quality in patients with osteoporosis.
Collapse
Affiliation(s)
- Zibin Zhou
- Department of Orthopedics, the Second Affiliated Hospital of Soochow University, Suzhou, China
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Xiaoyu Mao
- Department of Orthopedics, the Second Affiliated Hospital of Soochow University, Suzhou, China
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Chun Jiang
- Department of Orthopedics, the Second Affiliated Hospital of Soochow University, Suzhou, China
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Wenguo Li
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Tiantian Zhou
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Meng Liu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Shijin Sun
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Mengting Wang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
- NHC Key Laboratory of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ningzheng Dong
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China.
- NHC Key Laboratory of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Qingyu Wu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China.
| | - Haibin Zhou
- Department of Orthopedics, the Second Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
6
|
Li SJ, Liu AB, Yu YY, Ma JH. The role and mechanism of pyroptosis and potential therapeutic targets in non-alcoholic fatty liver disease (NAFLD). Front Cell Dev Biol 2024; 12:1407738. [PMID: 39022762 PMCID: PMC11251954 DOI: 10.3389/fcell.2024.1407738] [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: 03/27/2024] [Accepted: 06/10/2024] [Indexed: 07/20/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a clinical pathological syndrome characterized by the excessive accumulation of fat within liver cells, which can progress to end-stage liver disease in severe cases, posing a threat to life. Pyroptosis is a distinct, pro-inflammatory form of cell death, differing from traditional apoptosis. In recent years, there has been growing research interest in the association between pyroptosis and NAFLD, encompassing the mechanisms and functions of pyroptosis in the progression of NAFLD, as well as potential therapeutic targets. Controlled pyroptosis can activate immune cells, eliciting host immune responses to shield the body from harm. However, undue activation of pyroptosis may worsen inflammatory responses, induce cellular or tissue damage, disrupt immune responses, and potentially impact liver function. This review elucidates the involvement of pyroptosis and key molecular players, including NOD-like receptor thermal protein domain associated protein 3(NLRP3) inflammasome, gasdermin D (GSDMD), and the caspase family, in the pathogenesis and progression of NAFLD. It emphasizes the promising prospects of targeting pyroptosis as a therapeutic approach for NAFLD and offers valuable insights into future directions in the field of NAFLD treatment.
Collapse
Affiliation(s)
- Shu-Jing Li
- Department of Pediatrics Medical, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - An-Bu Liu
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yuan-Yuan Yu
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Jin-Hai Ma
- Department of Pediatrics Medical, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| |
Collapse
|
7
|
Zuo Y, Li B, Gao M, Xiong R, He R, Li N, Geng Q. Novel insights and new therapeutic potentials for macrophages in pulmonary hypertension. Respir Res 2024; 25:147. [PMID: 38555425 PMCID: PMC10981837 DOI: 10.1186/s12931-024-02772-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 03/13/2024] [Indexed: 04/02/2024] Open
Abstract
Inflammation and immune processes underlie pulmonary hypertension progression. Two main different activated phenotypes of macrophages, classically activated M1 macrophages and alternatively activated M2 macrophages, are both involved in inflammatory processes related to pulmonary hypertension. Recent advances suggest that macrophages coordinate interactions among different proinflammatory and anti-inflammatory mediators, and other cellular components such as smooth muscle cells and fibroblasts. In this review, we summarize the current literature on the role of macrophages in the pathogenesis of pulmonary hypertension, including the origin of pulmonary macrophages and their response to triggers of pulmonary hypertension. We then discuss the interactions among macrophages, cytokines, and vascular adventitial fibroblasts in pulmonary hypertension, as well as the potential therapeutic benefits of macrophages in this disease. Identifying the critical role of macrophages in pulmonary hypertension will contribute to a comprehensive understanding of this pathophysiological abnormality, and may provide new perspectives for pulmonary hypertension management.
Collapse
Affiliation(s)
- Yifan Zuo
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Boyang Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Minglang Gao
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Rui Xiong
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Ruyuan He
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| |
Collapse
|
8
|
Li S, Xiao H, Liu M, Wang Q, Sun C, Yao J, Cao N, Zhang H, Zhang G, Xiao X. Network pharmacology and experimental verification to explore the anti-superficial thrombophlebitis mechanism of Mailuo shutong pill. JOURNAL OF ETHNOPHARMACOLOGY 2024; 322:117668. [PMID: 38159829 DOI: 10.1016/j.jep.2023.117668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 12/13/2023] [Accepted: 12/25/2023] [Indexed: 01/03/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Mailuo shutong pill (MLST) has been widely used in clinical treatment of superficial thrombotic phlebitis (STP). Nevertheless, the major active components of MLST and the mechanism of synergistic action have not been reported. AIM OF THE STUDY The present study aimed to evaluate the improving effects and the underlying mechanism of MLST on mannitol-induced STP in rabbits. MATERIAL AND METHODS In this study, Ultrahigh-performance liquid chromatography electrospray ionization quadrupole-exactive orbitrap mass spectrometry (UHPLC-ESI-Q-Exactive-Orbitrap-MS) was used to analyze and identify the chemical composition of MLST and the prototype components absorbed into the blood. Then, according to the prototype components in serum, the targets and mechanisms of MLST were explored by applying network pharmacology. The rabbit model of STP was established by injecting 20% mannitol into bilateral auricular vein. The pathological changes of rabbit ear tissues, inflammatory factors, coagulation function and hemorheology were detected. In addition, molecular docking verified the interaction between the main active ingredient and the key target. Finally, the PI3K/AKT pathway and its regulated downstream pathways were verified by Western blot. RESULTS A total of 96 MLST components and 53 prototypical components absorbed into the blood were successfully identified. Based on network pharmacology, PI3K/AKT pathway and 10 chemical components closely related to this pathway were obtained. Hematoxylin-eosin (HE) staining results indicated that MLST effectively improved of the pathological damage of ear tissues. MLST decreased levels of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6 and C-reactive protein (CRP). The expression of platelets (PLT) and fibrinogen concentration (FIB) was decreased, while prothrombin time (PT) and activated partial thromboplastin time (APTT) were prolonged. In addition, the plasma viscosity and whole blood viscosity in the MLST groups were significantly decreased. The more important discovery was that the expressions of P-PI3K, VEGF, P-AKT, P-IκB-α, P-NF-κB, NLRP3, ASC, Cleaved IL-1β and Cleaved Caspase-1 were effectively reversed after treatment with MLST. CONCLUSIONS This study comprehensively analyzed and characterized the chemical composition of MLST and the prototypical components absorbed into the blood. This study strongly confirmed the pharmacodynamic effect of MLST on STP. More importantly, this pharmacodynamic effect was achieved through inhibition of the PI3K/AKT pathway and its regulated NF-κB and NLRP3 pathways.
Collapse
Affiliation(s)
- Shirong Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - He Xiao
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. LTD., Linyi, 276005, China.
| | - Mingfei Liu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Qingguo Wang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Chenghong Sun
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. LTD., Linyi, 276005, China.
| | - Jingchun Yao
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. LTD., Linyi, 276005, China.
| | - Ningning Cao
- Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300250, China.
| | - Haifang Zhang
- Graduate School of Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Guimin Zhang
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. LTD., Linyi, 276005, China.
| | - Xuefeng Xiao
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
9
|
Fioretto BS, Rosa I, Andreucci E, Mencucci R, Marini M, Romano E, Manetti M. Pharmacological Stimulation of Soluble Guanylate Cyclase Counteracts the Profibrotic Activation of Human Conjunctival Fibroblasts. Cells 2024; 13:360. [PMID: 38391973 PMCID: PMC10887040 DOI: 10.3390/cells13040360] [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: 01/23/2024] [Revised: 02/09/2024] [Accepted: 02/17/2024] [Indexed: 02/24/2024] Open
Abstract
Conjunctival fibrosis is a serious clinical concern implicated in a wide spectrum of eye diseases, including outcomes of surgery for pterygium and glaucoma. It is mainly driven by chronic inflammation that stimulates conjunctival fibroblasts to differentiate into myofibroblasts over time, leading to abnormal wound healing and scar formation. Soluble guanylate cyclase (sGC) stimulation was found to suppress transforming growth factor β (TGFβ)-induced myofibroblastic differentiation in various stromal cells such as skin and pulmonary fibroblasts, as well as corneal keratocytes. Here, we evaluated the in vitro effects of stimulation of the sGC enzyme with the cell-permeable pyrazolopyridinylpyrimidine compound BAY 41-2272 in modulating the TGFβ1-mediated profibrotic activation of human conjunctival fibroblasts. Cells were pretreated with the sGC stimulator before challenging with recombinant human TGFβ1, and subsequently assayed for viability, proliferation, migration, invasiveness, myofibroblast marker expression, and contractile properties. Stimulation of sGC significantly counteracted TGFβ1-induced cell proliferation, migration, invasiveness, and acquisition of a myofibroblast-like phenotype, as shown by a significant downregulation of FAP, ACTA2, COL1A1, COL1A2, FN1, MMP2, TIMP1, and TIMP2 mRNA levels, as well as by a significant reduction in α-smooth muscle actin, N-cadherin, COL1A1, and FN-EDA protein expression. In addition, pretreatment with the sGC stimulator was capable of significantly dampening TGFβ1-induced acquisition of a contractile phenotype by conjunctival fibroblasts, as well as phosphorylation of Smad3 and release of the proinflammatory cytokines IL-1β and IL-6. Taken together, our findings are the first to demonstrate the effectiveness of pharmacological sGC stimulation in counteracting conjunctival fibroblast-to-myofibroblast transition, thus providing a promising scientific background to further explore the feasibility of sGC stimulators as potential new adjuvant therapeutic compounds to treat conjunctival fibrotic conditions.
Collapse
Affiliation(s)
- Bianca Saveria Fioretto
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (B.S.F.); (I.R.); (M.M.)
| | - Irene Rosa
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (B.S.F.); (I.R.); (M.M.)
| | - Elena Andreucci
- Section of Experimental Pathology and Oncology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale Morgagni 50, 50134 Florence, Italy;
| | - Rita Mencucci
- Eye Clinic, Careggi Hospital, Department of Neurosciences, Psychology, Pharmacology and Child Health (NEUROFARBA), University of Florence, Largo Brambilla 3, 50134 Florence, Italy;
| | - Mirca Marini
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (B.S.F.); (I.R.); (M.M.)
| | - Eloisa Romano
- Section of Internal Medicine, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy;
| | - Mirko Manetti
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (B.S.F.); (I.R.); (M.M.)
- Imaging Platform, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy
| |
Collapse
|
10
|
Wan Y, Zhou J, Zhang P, Lin X, Li H. Inhibition of spinal Rac1 attenuates chronic inflammatory pain by regulating the activation of astrocytes. Cell Signal 2024; 114:110972. [PMID: 37984604 DOI: 10.1016/j.cellsig.2023.110972] [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: 08/16/2023] [Revised: 10/20/2023] [Accepted: 11/13/2023] [Indexed: 11/22/2023]
Abstract
BACKGROUND Spinal astrocyte-mediated neuroinflammation is an important mechanism for the maintenance of chronic inflammatory pain. Previous studies have investigated that Ras-related C3 botulinum toxin substrate 1 (Rac1) is closely related to astrocyte activation after central nervous system injury. However, the role of Rac1 in astrocyte activation in chronic inflammatory pain has not been reported. METHODS Complete Freund's adjuvant (CFA)-induced chronic inflammatory pain model and LPS-stimulated astrocytes were used to investigate the role of Rac1 in astrocyte activation and the underlying mechanism. Rac1-interfering adeno-associated virus (AAV) targeting astrocytes was delivered to spinal astrocytes by intrathecal administration and a Rac1 specific inhibitor, NSC23766, was used to block cultured astrocytes. The glial fibrillary acidic protein (GFAP), proinflammatory cytokines, p-NF-κB, and nod-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome were detected by RT-qPCR, Western blotting, and immunofluorescence to investigate the activation of astrocytes. RESULTS CFA induced spinal astrocyte activation and increased the expression of active Rac1 in spinal astrocytes. Knockdown of astrocyte Rac1 alleviated chronic inflammatory pain and inhibited astrocyte activation. Inhibition of Rac1 activation in cultured astrocytes decreased the expression of GFAP and proinflammatory cytokines. Knockdown of Rac1 inhibited the increase of expression of NLRP3 inflammasome and phosphorylation of NF-κB in the spinal lumbar enlargement after CFA injection. Similarly, the inhibition of Rac1 suppressed the increase of NLRP3 inflammasome and p-NF-κB protein level after LPS stimulation. CONCLUSION Knockdown of astrocyte Rac1 attenuated CFA-induced hyperalgesia and astrocyte activation possibly by blocking the expression of NLRP3 inflammasome and phosphorylation of NF-κB.
Collapse
Affiliation(s)
- Yantong Wan
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Jieshu Zhou
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Panpan Zhang
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Xuemei Lin
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China.
| | - Hao Li
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China.
| |
Collapse
|
11
|
Zhang X, Yang Q, Zhang R, Zhang Y, Zeng W, Yu Q, Zeng M, Gan J, Li H, Yang L, Gao Q, Jiang X. Sodium Danshensu ameliorates cerebral ischemia/reperfusion injury by inhibiting CLIC4/NLRP3 inflammasome-mediated endothelial cell pyroptosis. Biofactors 2024; 50:74-88. [PMID: 37458329 DOI: 10.1002/biof.1991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 07/03/2023] [Indexed: 02/20/2024]
Abstract
Endothelial pyroptosis promotes cerebral ischemia/reperfusion injury (CIRI). Sodium Danshensu (SDSS) has been shown to attenuate CIRI and have anti-inflammatory properties in endothelial cells. However, the mechanism and effect of SDSS on alleviating endothelial pyroptosis after CIRI remains poorly understood. Thus, we aimed to investigate the efficacy and mechanism of SDSS in reducing endothelial pyroptosis. It has been shown that SDSS administration inhibited NLRP3 inflammasome-mediated pyroptosis. As demonstrated by protein microarrays, molecular docking, CETSA and ITDRFCETSA , SDSS bound strongly to CLIC4. Furthermore, SDSS can decrease its expression and inhibit its translocation. Its effectiveness was lowered by CLIC4 overexpression but not by knockdown. Overall The beneficial effect of SDSS against CIRI in this study can be ascribed to blocking endothelial pyroptosis by binding to CLIC4 and then inhibiting chloride efflux-dependent NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Xiaolu Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Qiuyue Yang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Ruifeng Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Yilin Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Wenyun Zeng
- Oncology Department, Ganzhou People's Hospital, Ganzhou, People's Republic of China
| | - Qun Yu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Miao Zeng
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Jiali Gan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Huhu Li
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Lin Yang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Qing Gao
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| |
Collapse
|
12
|
Kwanten W(WJ, Francque SM. The liver sinusoid in chronic liver disease: NAFLD and NASH. SINUSOIDAL CELLS IN LIVER DISEASES 2024:263-284. [DOI: 10.1016/b978-0-323-95262-0.00012-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
13
|
Zhu JY, Tang M, Li H, Shi YL, Li YM, Li YH, Ma XC, Duan QL, Mei YH, He HW, Zhang N, Peng ZG, Song DQ. Design, synthesis and triglyceride-lowering activity of tricyclic matrine derivatives for the intervention of non-alcoholic fatty liver disease. Bioorg Chem 2024; 142:106925. [PMID: 37890213 DOI: 10.1016/j.bioorg.2023.106925] [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: 07/24/2023] [Revised: 10/09/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023]
Abstract
Thirty new tricyclicmatrinic derivatives were successively synthesized and evaluated for their inhibitory activity on the accumulation of triglycerides (TG) in AML12 cells, using 12 N-m-trifluoromethylbenzenesulfonyl matrine (1) as the hit compound. Among the analogues, compound 7n possessing 11-trimethylbutylamine quaternary exerted the highest in vitro TG-lowering potency, as well as a good safety profile. 7n significantly attenuated the hepatic injury and steatosis, and ameliorated dyslipidemia and dysglycemia in the mice with non-alcoholic fatty liver disease (NAFLD) induced by a high-fat diet. Primary mechanism study revealed that upregulation of peroxisome proliferator-activated receptors α (PPARα)-carnitine palmitoyltransferase 1A (CPT1A) pathway mediated the efficacy of 7n. Our study provides powerful information for developing this kind of compound into a new class of anti-NAFLD candidates, and compound 7n is worthy of further investigation as an ideal lead compound.
Collapse
Affiliation(s)
- Jing-Yang Zhu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Mei Tang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Hu Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yu-Long Shi
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yi-Ming Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Ying-Hong Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xi-Can Ma
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Qiong-Lu Duan
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yu-Heng Mei
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Hong-Wei He
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Na Zhang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| | - Zong-Gen Peng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| | - Dan-Qing Song
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
14
|
Chen T, Kong B, Shuai W, Gong Y, Zhang J, Huang H. Vericiguat alleviates ventricular remodeling and arrhythmias in mouse models of myocardial infarction via CaMKII signaling. Life Sci 2023; 334:122184. [PMID: 37866806 DOI: 10.1016/j.lfs.2023.122184] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/04/2023] [Accepted: 10/15/2023] [Indexed: 10/24/2023]
Abstract
AIMS Maladaptive ventricular remodeling is a major cause of ventricular arrhythmias following myocardial infarction (MI) and adversely impacts the quality of life of affected patients. Vericiguat is a new soluble guanylate cyclase (sGC) activator with cardioprotective properties. However, its effects on MI-induced ventricular remodeling and arrhythmias are not fully comprehended; hence, our research evaluated the effect of vericiguat on mice post-MI. MATERIALS AND METHODS Mice were divided into four treatment groups: Sham, Sham+Veri, MI, and MI + Veri. For the MI groups and MI + Veri groups, the left anterior descending (LAD) coronary artery was occluded to induce MI. Conversely, the Sham group underwent mock surgery. Vericiguat was administered orally daily for 28 days to the Sham+Veri and MI + Veri groups. Additionally, H9c2 cells were cultured for further mechanistic studies. Assessment methods included echocardiography, pathological analysis, electrophysiological analysis, and Western blotting. KEY FINDINGS Vericiguat reduced cardiac dysfunction and infarct size after MI. It also mitigated MI-induced left ventricular fibrosis and cardiomyocyte apoptosis. Vericiguat normalized the expression of ion channel proteins (Kv4.3, Kv4.2, Kv2.1, Kv1.5, Kv7.1, KCNH2, Cav1.2) and the gap junction protein connexin 43, reducing the susceptibility to ventricular arrhythmia. Vericiguat significantly inhibited MI-induced calcium/calmodulin-dependent protein kinase II (CaMKII) pathway activation in mice. SIGNIFICANCE Vericiguat alleviated MI-induced left ventricular adverse remodeling and arrhythmias through modulation of the CamkII signaling pathway.
Collapse
Affiliation(s)
- Tao Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, PR China; Cardiovascular Research Institute of Wuhan University, Wuhan 430060, Hubei, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei, PR China
| | - Bin Kong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, PR China; Cardiovascular Research Institute of Wuhan University, Wuhan 430060, Hubei, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei, PR China
| | - Wei Shuai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, PR China; Cardiovascular Research Institute of Wuhan University, Wuhan 430060, Hubei, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei, PR China
| | - Yang Gong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, PR China; Cardiovascular Research Institute of Wuhan University, Wuhan 430060, Hubei, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei, PR China
| | - Jingjing Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, PR China; Cardiovascular Research Institute of Wuhan University, Wuhan 430060, Hubei, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei, PR China
| | - He Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, PR China; Cardiovascular Research Institute of Wuhan University, Wuhan 430060, Hubei, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei, PR China.
| |
Collapse
|
15
|
Zamanian MY, Sadeghi Ivraghi M, Khachatryan LG, Vadiyan DE, Bali HY, Golmohammadi M. A review of experimental and clinical studies on the therapeutic effects of pomegranate ( Punica granatum) on non-alcoholic fatty liver disease: Focus on oxidative stress and inflammation. Food Sci Nutr 2023; 11:7485-7503. [PMID: 38107091 PMCID: PMC10724645 DOI: 10.1002/fsn3.3713] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/06/2023] [Accepted: 09/12/2023] [Indexed: 12/19/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is frequently linked to metabolic disorders and is prevalent in obese and diabetic patients. The pathophysiology of NAFLD involves multiple factors, including insulin resistance (IR), oxidative stress (OS), inflammation, and genetic predisposition. Recently, there has been an emphasis on the use of herbal remedies with many people around the world resorting to phytonutrients or nutraceuticals for treatment of numerous health challenges in various national healthcare settings. Pomegranate (Punica granatum) parts, such as juice, peel, seed and flower, have high polyphenol content and is well known for its antioxidant capabilities. Pomegranate polyphenols, such as hydrolyzable tannins, anthocyanins, and flavonoids, have high antioxidant capabilities that can help lower the OS and inflammation associated with NAFLD. The study aimed to investigate whether pomegranate parts could attenuate OS, inflammation, and other risk factors associated with NAFLD, and ultimately prevent the development of the disease. The findings of this study revealed that: 1. pomegranate juice contains hypoglycemic qualities that can assist manage blood sugar levels, which is vital for avoiding and treating NAFLD. 2. Polyphenols from pomegranate flowers increase paraoxonase 1 (PON1) mRNA and protein levels in the liver, which can help protect liver enzymes and prevent NAFLD. 3. Punicalagin (PU) is one of the major ellagitannins found in pomegranate, and PU-enriched pomegranate extract (PE) has been shown to inhibit HFD-induced hyperlipidemia and hepatic lipid deposition in rats. 4. Pomegranate fruit consumption, which is high in antioxidants, can decrease the activity of AST and ALT (markers of liver damage), lower TNF-α (a marker of inflammation), and improve overall antioxidant capacity in NAFLD patients. Overall, the polyphenols in pomegranate extracts have antioxidant, anti-inflammatory, hypoglycemic, and protective effects on liver enzymes, which can help prevent and manage NAFLD effects on liver enzymes, which can help prevent and manage NAFLD.
Collapse
Affiliation(s)
- Mohammad Yassin Zamanian
- Department of Physiology, School of MedicineHamadan University of Medical SciencesHamadanIran
- Department of Pharmacology and Toxicology, School of PharmacyHamadan University of Medical SciencesHamadanIran
| | | | - Lusine G. Khachatryan
- Department of Pediatric Diseases, N.F. Filatov Clinical Institute of Children's HealthI.M. Sechenov First Moscow State Medical University (Sechenov University)MoscowRussia
| | - Diana E. Vadiyan
- Institute of Dentistry, Department of Pediatric, Preventive Dentistry and OrthodonticsI.M. Sechenov First Moscow State Medical University (Sechenov University)MoscowRussia
| | | | | |
Collapse
|
16
|
Cui D, Wang Z, Dang Q, Wang J, Qin J, Song J, Zhai X, Zhou Y, Zhao L, Lu G, Liu H, Liu G, Liu R, Shao C, Zhang X, Liu Z. Spliceosome component Usp39 contributes to hepatic lipid homeostasis through the regulation of autophagy. Nat Commun 2023; 14:7032. [PMID: 37923718 PMCID: PMC10624899 DOI: 10.1038/s41467-023-42461-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 10/11/2023] [Indexed: 11/06/2023] Open
Abstract
Regulation of alternative splicing (AS) enables a single transcript to yield multiple isoforms that increase transcriptome and proteome diversity. Here, we report that spliceosome component Usp39 plays a role in the regulation of hepatocyte lipid homeostasis. We demonstrate that Usp39 expression is downregulated in hepatic tissues of non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) subjects. Hepatocyte-specific Usp39 deletion in mice leads to increased lipid accumulation, spontaneous steatosis and impaired autophagy. Combined analysis of RNA immunoprecipitation (RIP-seq) and bulk RNA sequencing (RNA-seq) data reveals that Usp39 regulates AS of several autophagy-related genes. In particular, deletion of Usp39 results in alternative 5' splice site selection of exon 6 in Heat shock transcription factor 1 (Hsf1) and consequently its reduced expression. Importantly, overexpression of Hsf1 could attenuate lipid accumulation caused by Usp39 deficiency. Taken together, our findings indicate that Usp39-mediated AS is required for sustaining autophagy and lipid homeostasis in the liver.
Collapse
Affiliation(s)
- Donghai Cui
- Key Laboratory of Experimental Teratology, Ministry of Education, School of Basic Medical Science, Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, China
- Advanced Medical Research Institute, Shandong University, Jinan, China
| | - Zixiang Wang
- Key Laboratory of Experimental Teratology, Ministry of Education, School of Basic Medical Science, Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, China
- Advanced Medical Research Institute, Shandong University, Jinan, China
| | - Qianli Dang
- Key Laboratory of Experimental Teratology, Ministry of Education, School of Basic Medical Science, Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, China
- Advanced Medical Research Institute, Shandong University, Jinan, China
| | - Jing Wang
- Key Laboratory of Experimental Teratology, Ministry of Education, School of Basic Medical Science, Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, China
- Advanced Medical Research Institute, Shandong University, Jinan, China
| | - Junchao Qin
- Key Laboratory of Experimental Teratology, Ministry of Education, School of Basic Medical Science, Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, China
- Advanced Medical Research Institute, Shandong University, Jinan, China
| | - Jianping Song
- Department of General Surgery, The Second Hospital, Shandong University, Jinan, China
| | - Xiangyu Zhai
- Department of General Surgery, The Second Hospital, Shandong University, Jinan, China
| | - Yachao Zhou
- Key Laboratory of Experimental Teratology, Ministry of Education, School of Basic Medical Science, Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, China
- Advanced Medical Research Institute, Shandong University, Jinan, China
| | - Ling Zhao
- Key Laboratory of Experimental Teratology, Ministry of Education, School of Basic Medical Science, Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, China
| | - Gang Lu
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Hongbin Liu
- Center for Reproductive Medicine, Shandong University, Jinan, China
| | - Gang Liu
- Nephrology Research Institute of Shandong University, The Second Hospital of Shandong University, Jinan, China
| | - Runping Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Changshun Shao
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China.
| | - Xiyu Zhang
- Key Laboratory of Experimental Teratology, Ministry of Education, School of Basic Medical Science, Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, China.
| | - Zhaojian Liu
- Key Laboratory of Experimental Teratology, Ministry of Education, School of Basic Medical Science, Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, China.
- Advanced Medical Research Institute, Shandong University, Jinan, China.
- Nephrology Research Institute of Shandong University, The Second Hospital of Shandong University, Jinan, China.
| |
Collapse
|
17
|
Fernández-Sáez EM, Losarcos M, Becerril S, Valentí V, Moncada R, Martín M, Burrell MA, Catalán V, Gómez-Ambrosi J, Mugueta C, Colina I, Silva C, Escalada J, Frühbeck G, Rodríguez A. Uroguanylin prevents hepatic steatosis, mitochondrial dysfunction and fibrosis in obesity-associated NAFLD. Metabolism 2023; 147:155663. [PMID: 37517791 DOI: 10.1016/j.metabol.2023.155663] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/19/2023] [Accepted: 07/24/2023] [Indexed: 08/01/2023]
Abstract
BACKGROUND The biological mediators supporting the resolution of liver steatosis, inflammation and fibrosis after bariatric surgery in patients with obesity and NAFLD remain unclear. We sought to analyze whether uroguanylin and guanylin, two gut hormones involved in the regulation of satiety, food preference and adiposity, are involved in the amelioration of obesity-associated NAFLD after bariatric surgery. METHODS Proguanylin (GUCA2A) and prouroguanylin (GUCA2B) were measured in 214 participants undergoing bariatric surgery with biopsy-proven NAFLD diagnosis. Pathways involved in lipid metabolism, mitochondrial network and fibrogenesis were evaluated in liver biopsies (n = 137). The effect of guanylin and uroguanylin on these metabolic functions was assessed in HepG2 hepatocytes and LX-2 hepatic stellate cells (HSC) under lipotoxic and profibrogenic conditions. RESULTS Plasma and hepatic expression of GUCA2B were decreased in obesity-associated NAFLD. Both GUCA2A and GUCA2B levels were increased after sleeve gastrectomy and Roux-en-Y gastric bypass in parallel to the improved liver function. The liver of patients with type 2 diabetes showed impaired mitochondrial β-oxidation, biogenesis, dynamics as well as increased fibrosis. Uroguanylin diminished the lipotoxicity in palmitate-treated HepG2 hepatocytes, evidenced by decresased steatosis and lipogenic factors, as well as increased mitochondrial network expression, AMPK-induced β-oxidation and oxygen consumption rate. Additionally, uroguanylin, but not guanylin, reversed HSC myofibroblast transdifferentiation as well as fibrogenesis after TGF-β1 stimulation. CONCLUSIONS Uroguanylin constitutes a protective factor against lipotoxicity, mitochondrial dysfunction and fibrosis. Increased GUCA2B levels might contribute to improve liver injury in patients with obesity-associated NAFLD after bariatric surgery.
Collapse
Affiliation(s)
| | - Maite Losarcos
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
| | - Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Víctor Valentí
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain; Department of Surgery, Clínica Universidad de Navarra, Pamplona, Spain
| | - Rafael Moncada
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain; Department of Anesthesia, Clínica Universidad de Navarra, Pamplona, Spain
| | - Marina Martín
- Department of Pathology, Anatomy and Physiology, University of Navarra, Pamplona, Spain
| | - María A Burrell
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain; Department of Pathology, Anatomy and Physiology, University of Navarra, Pamplona, Spain
| | - Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Carmen Mugueta
- Department of Biochemistry, Clínica Universidad de Navarra, Pamplona, Spain
| | - Inmaculada Colina
- Department of Internal Medicine, Clínica Universidad de Navarra, Pamplona, Spain
| | - Camilo Silva
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain; Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Javier Escalada
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain; Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain; Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.
| |
Collapse
|
18
|
Hu XH, Chen L, Wu H, Tang YB, Zheng QM, Wei XY, Wei Q, Huang Q, Chen J, Xu X. Cell therapy in end-stage liver disease: replace and remodel. Stem Cell Res Ther 2023; 14:141. [PMID: 37231461 DOI: 10.1186/s13287-023-03370-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 04/26/2023] [Indexed: 05/27/2023] Open
Abstract
Liver disease is prevalent worldwide. When it reaches the end stage, mortality rises to 50% or more. Although liver transplantation has emerged as the most efficient treatment for end-stage liver disease, its application has been limited by the scarcity of donor livers. The lack of acceptable donor organs implies that patients are at high risk while waiting for suitable livers. In this scenario, cell therapy has emerged as a promising treatment approach. Most of the time, transplanted cells can replace host hepatocytes and remodel the hepatic microenvironment. For instance, hepatocytes derived from donor livers or stem cells colonize and proliferate in the liver, can replace host hepatocytes, and restore liver function. Other cellular therapy candidates, such as macrophages and mesenchymal stem cells, can remodel the hepatic microenvironment, thereby repairing the damaged liver. In recent years, cell therapy has transitioned from animal research to early human studies. In this review, we will discuss cell therapy in end-stage liver disease treatment, especially focusing on various cell types utilized for cell transplantation, and elucidate the processes involved. Furthermore, we will also summarize the practical obstacles of cell therapy and offer potential solutions.
Collapse
Affiliation(s)
- Xin-Hao Hu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Lan Chen
- Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Hao Wu
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Yang-Bo Tang
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Qiu-Min Zheng
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China
| | - Xu-Yong Wei
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Qiang Wei
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Qi Huang
- Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jian Chen
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| | - Xiao Xu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| |
Collapse
|
19
|
Recombinant Mouse Prolactin Confers Partial Protection Against Toxoplasma gondii Infection in a Pre-treated Experimental Murine Model. Acta Parasitol 2023; 68:182-193. [PMID: 36542295 DOI: 10.1007/s11686-022-00651-0] [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: 04/09/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Toxoplasmosis is one of the most important health-threatening diseases with worldwide distribution and global impact. It is caused by Toxoplasma gondii (T. gondii), an intracellular apicomplexan parasite that can evade the host immune responses and establish a chronic infection. The available treatments are not efficient against this stage and have many adverse effects. There are no available effective vaccines, apart from Toxovax®, which is used in sheep to prevent abortion. Studies documented that prolactin (PRL) had in vivo and in vitro anti-Toxoplasma effects. Detailed research was recommended about the mechanisms of such inhibitory effects. AIM This study was designed to assess the possible protective role of the recombinant prolactin (rPRL) against T. gondii. MATERIALS AND METHODS Sixty experimentally infected mice with T. gondii were used. The treated mice received rPRL for five days before infection. Serum prolactin levels were measured; survival rate was monitored; number, size, and DNA of T. gondii cysts in the brain were measured; and histopathological and immunological studies were done. RESULTS There was a significant increase in the survival rate of the rPRL-treated mice, a significant decrease in the number, size, and DNA amount of T. gondii cysts in the brain with a noticeable improvement of histopathological lesions in the brain and liver tissues when compared to the infected untreated group. These effects seem to be achieved through stimulating humoral and cell-mediated immune responses that were evident by the significant rise in serum levels of anti-Toxoplasma IgM, IFN-γ, and TNF-α. CONCLUSION The rPRL elicited robust immune responses, which provided efficient protection against murine T. gondii infection.
Collapse
|
20
|
Qin H, Wang C, He Y, Lu A, Li T, Zhang B, Shen J. Silencing miR-146a-5p Protects against Injury-Induced Osteoarthritis in Mice. Biomolecules 2023; 13:123. [PMID: 36671508 PMCID: PMC9856058 DOI: 10.3390/biom13010123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 01/10/2023] Open
Abstract
Osteoarthritis (OA), the most prevalent joint disease and the leading cause of disability, remains an incurable disease largely because the etiology and pathogenesis underlying this degenerative process are poorly understood. Low-grade inflammation within joints is a well-established factor that disturbs joint homeostasis and leads to an imbalance between anabolic and catabolic processes in articular cartilage; however, the complexity of the network between inflammatory factors that often involves positive and negative feedback loops makes current anti-cytokine therapy ineffective. MicroRNAs (miRNAs) have emerged as key regulators to control inflammation, and aberrant miRNAs expression has recently been linked to OA pathophysiology. In the present study, we characterized transcriptomic profiles of miRNAs in primary murine articular chondrocytes in response to a proinflammatory cytokine, IL-1β, and identified miR-146a-5p as the most responsive miRNA to IL-1β. miR-146a-5p was also found to be upregulated in human OA cartilage. We further demonstrated that knockdown of miR-146a-5p antagonized IL-1β-mediated inflammatory responses and IL-1β-induced catabolism in vitro, and silencing of miR-146a in chondrocytes ameliorated articular cartilage destruction and reduced OA-evoked pain in an injury-induced murine OA model. Moreover, parallel RNA sequencing revealed that differentially expressed genes in response to IL-1β were enriched in pathways related to inflammatory processes, cartilage matrix homeostasis, and cell metabolism. Bioinformatic analyses of putative miR-146a-5p gene targets and following prediction of protein-protein interactions suggest a functional role of miR-146a-5p in mediating inflammatory processes and regulation of cartilage homeostasis. Our genetic and transcriptomic data define a crucial role of miR-146a-5p in OA pathogenesis and implicate modulation of miR-146a-5p in articular chondrocytes as a potential therapeutic strategy to alleviate OA.
Collapse
Affiliation(s)
- Haocheng Qin
- Department of Orthopaedic Surgery, School of Medicine, Washington University, St. Louis, MO 63110, USA
- The 2nd Xiangya Hospital, Central South University, Changsha 410021, China
| | - Cuicui Wang
- Department of Orthopaedic Surgery, School of Medicine, Washington University, St. Louis, MO 63110, USA
| | - Yonghua He
- Department of Orthopaedic Surgery, School of Medicine, Washington University, St. Louis, MO 63110, USA
| | - Aiwu Lu
- Department of Orthopaedic Surgery, School of Medicine, Washington University, St. Louis, MO 63110, USA
| | - Tiandao Li
- Department of Developmental Biology, Center of Regenerative Medicine, Washington University, St. Louis, MO 63110, USA
| | - Bo Zhang
- Department of Developmental Biology, Center of Regenerative Medicine, Washington University, St. Louis, MO 63110, USA
| | - Jie Shen
- Department of Orthopaedic Surgery, School of Medicine, Washington University, St. Louis, MO 63110, USA
| |
Collapse
|
21
|
Felli E, Nulan Y, Selicean S, Wang C, Gracia-Sancho J, Bosch J. Emerging Therapeutic Targets for Portal Hypertension. CURRENT HEPATOLOGY REPORTS 2023; 22:51-66. [PMID: 36908849 PMCID: PMC9988810 DOI: 10.1007/s11901-023-00598-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 01/18/2023] [Indexed: 02/13/2023]
Abstract
Purpose of Review Portal hypertension is responsible of the main complications of cirrhosis, which carries a high mortality. Recent treatments have improved prognosis, but this is still far from ideal. This paper reviews new potential therapeutic targets unveiled by advances of key pathophysiologic processes. Recent Findings Recent research highlighted the importance of suppressing etiologic factors and a safe lifestyle and outlined new mechanisms modulating portal pressure. These include intrahepatic abnormalities linked to inflammation, fibrogenesis, vascular occlusion, parenchymal extinction, and angiogenesis; impaired regeneration; increased hepatic vascular tone due to sinusoidal endothelial dysfunction with insufficient NO availability; and paracrine liver cell crosstalk. Moreover, pathways such as the gut-liver axis modulate splanchnic vasodilatation and systemic inflammation, exacerbate liver fibrosis, and are being targeted by therapy. We have summarized studies of new agents addressing these targets. Summary New agents, alone or in combination, allow acting in complementary mechanisms offering a more profound effect on portal hypertension while simultaneously limiting disease progression and favoring regression of fibrosis and of cirrhosis. Major changes in treatment paradigms are anticipated.
Collapse
Affiliation(s)
- Eric Felli
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, 3012 Bern, Switzerland
- Department for BioMedical Research, Hepatology, University of Bern, 3012 Bern, Switzerland
| | - Yelidousi Nulan
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, 3012 Bern, Switzerland
- Department for BioMedical Research, Hepatology, University of Bern, 3012 Bern, Switzerland
| | - Sonia Selicean
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, 3012 Bern, Switzerland
- Department for BioMedical Research, Hepatology, University of Bern, 3012 Bern, Switzerland
| | - Cong Wang
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, 3012 Bern, Switzerland
- Department for BioMedical Research, Hepatology, University of Bern, 3012 Bern, Switzerland
| | - Jordi Gracia-Sancho
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, 3012 Bern, Switzerland
- Department for BioMedical Research, Hepatology, University of Bern, 3012 Bern, Switzerland
- Liver Vascular Biology Research Group, CIBEREHD, IDIBAPS Research Institute, 08036 Barcelona, Spain
| | - Jaume Bosch
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, 3012 Bern, Switzerland
| |
Collapse
|
22
|
The Soluble Guanylate Cyclase Stimulator BAY 41-2272 Attenuates Transforming Growth Factor β1-Induced Myofibroblast Differentiation of Human Corneal Keratocytes. Int J Mol Sci 2022; 23:ijms232315325. [PMID: 36499651 PMCID: PMC9737374 DOI: 10.3390/ijms232315325] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/01/2022] [Accepted: 12/03/2022] [Indexed: 12/07/2022] Open
Abstract
Corneal transparency, necessary for vision and depending on the high organization of stromal extracellular matrix, is maintained by keratocytes. Severe or continuous corneal injuries determine exaggerated healing responses resulting in the formation of irreversible fibrotic scars and vision impairment. Soluble guanylate cyclase (sGC) stimulation demonstrated antifibrotic effects in both experimental fibrosis and human lung and skin fibroblasts. Here, we assessed whether sGC stimulation with BAY 41-2272 could attenuate transforming growth factor β1 (TGFβ1)-induced myofibroblast differentiation of human corneal keratocytes. Cells were challenged with TGFβ1, with/without BAY 41-2272 preincubation, and subsequently assessed for viability, proliferation, migration, chemoinvasion, as well for the expression of myofibroblast/fibroblast activation markers and contractile abilities. Treatment with BAY 41-2272 did not affect keratocyte viability, while preincubation of cells with the sGC stimulator was able to inhibit TGFβ1-induced proliferation, wound healing capacity, and invasiveness. BAY 41-2272 was also able to attenuate TGFβ1-induced myofibroblast-like profibrotic phenotype of keratocytes, as demonstrated by the significant decrease in ACTA2, COL1A1, COL1A2, FN1 and PDPN gene expression, as well as in α-smooth muscle actin, α-1 chain of type I collagen, podoplanin, vimentin and N-cadherin protein expression. Finally, BAY 41-2272 significantly counteracted the TGFβ1-induced myofibroblast-like ability of keratocytes to contract collagen gels, reduced phosphorylated Smad3 protein levels, and attenuated gene expression of proinflammatory cytokines. Collectively, our data show for the first time that BAY 41-2272 is effective in counteracting keratocyte-to-myofibroblast transition, thus providing the rationale for the development of sGC stimulators as novel promising modulators of corneal scarring and fibrosis.
Collapse
|
23
|
He H, Yang W, Su N, Zhang C, Dai J, Han F, Singhal M, Bai W, Zhu X, Zhu J, Liu Z, Xia W, Liu X, Zhang C, Jiang K, Huang W, Chen D, Wang Z, He X, Kirchhoff F, Li Z, Liu C, Huan J, Wang X, Wei W, Wang J, Augustin HG, Hu J. Activating NO-sGC crosstalk in the mouse vascular niche promotes vascular integrity and mitigates acute lung injury. J Exp Med 2022; 220:213673. [PMID: 36350314 PMCID: PMC9984546 DOI: 10.1084/jem.20211422] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/20/2022] [Accepted: 10/27/2022] [Indexed: 11/11/2022] Open
Abstract
Disruption of endothelial cell (ECs) and pericytes interactions results in vascular leakage in acute lung injury (ALI). However, molecular signals mediating EC-pericyte crosstalk have not been systemically investigated, and whether targeting such crosstalk could be adopted to combat ALI remains elusive. Using comparative genome-wide EC-pericyte crosstalk analysis of healthy and LPS-challenged lungs, we discovered that crosstalk between endothelial nitric oxide and pericyte soluble guanylate cyclase (NO-sGC) is impaired in ALI. Indeed, stimulating the NO-sGC pathway promotes vascular integrity and reduces lung edema and inflammation-induced lung injury, while pericyte-specific sGC knockout abolishes this protective effect. Mechanistically, sGC activation suppresses cytoskeleton rearrangement in pericytes through inhibiting VASP-dependent F-actin formation and MRTFA/SRF-dependent de novo synthesis of genes associated with cytoskeleton rearrangement, thereby leading to the stabilization of EC-pericyte interactions. Collectively, our data demonstrate that impaired NO-sGC crosstalk in the vascular niche results in elevated vascular permeability, and pharmacological activation of this crosstalk represents a promising translational therapy for ALI.
Collapse
Affiliation(s)
- Hao He
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Wu Yang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Nan Su
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Chuankai Zhang
- Department of Burn and Plastic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianing Dai
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Feng Han
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Mahak Singhal
- Laboratory of AngioRhythms, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Wenjuan Bai
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaolan Zhu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Jing Zhu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Zhen Liu
- University of Chinese Academy of Sciences, Beijing, China,Chinese Academy of Sciences Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Wencheng Xia
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Xiaoting Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Chonghe Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Kai Jiang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Wenhui Huang
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg, Germany
| | - Dan Chen
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Zhaoyin Wang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Xueyang He
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Frank Kirchhoff
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg, Germany
| | - Zhenyu Li
- Texas A&M Health Science Center, Bryan, TX
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Jingning Huan
- Department of Burn and Plastic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaohong Wang
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Wu Wei
- University of Chinese Academy of Sciences, Beijing, China,Chinese Academy of Sciences Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Jing Wang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hellmut G. Augustin
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany,Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Junhao Hu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China,Correspondence to Junhao Hu:
| |
Collapse
|
24
|
Microglial NLRP3 inflammasome activates neurotoxic astrocytes in depression-like mice. Cell Rep 2022; 41:111532. [DOI: 10.1016/j.celrep.2022.111532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 07/22/2022] [Accepted: 09/29/2022] [Indexed: 11/17/2022] Open
|
25
|
Ataei Ataabadi E, Golshiri K, Jüttner AA, de Vries R, Van den Berg‐Garrelds I, Nagtzaam NMA, Khan HN, Leijten FPJ, Brandt RMC, Dik WA, van der Pluijm I, Danser AHJ, Sandner P, Roks AJM. Soluble guanylate cyclase activator BAY 54-6544 improves vasomotor function and survival in an accelerated ageing mouse model. Aging Cell 2022; 21:e13683. [PMID: 36029161 PMCID: PMC9470884 DOI: 10.1111/acel.13683] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/08/2022] [Accepted: 07/17/2022] [Indexed: 01/24/2023] Open
Abstract
DNA damage is a causative factor in ageing of the vasculature and other organs. One of the most important vascular ageing features is reduced nitric oxide (NO)soluble guanylate cyclase (sGC)-cyclic guanosine monophosphate (cGMP) signaling. We hypothesized that the restoration of NO-sGC-cGMP signaling with an sGC activator (BAY 54-6544) may have beneficial effects on vascular ageing and premature death in DNA repair-defective mice undergoing accelerated ageing. Eight weeks of treatment with a non-pressor dosage of BAY 54-6544 restored the decreased in vivo microvascular cutaneous perfusion in progeroid Ercc1∆/- mice to the level of wild-type mice. In addition, BAY 54-6544 increased survival of Ercc1∆/- mice. In isolated Ercc1∆/- aorta, the decreased endothelium-independent vasodilation was restored after chronic BAY 54-6544 treatment. Senescence markers p16 and p21, and markers of inflammation, including Ccl2, Il6 in aorta and liver, and circulating IL-6 and TNF-α were increased in Ercc1∆/- , which was lowered by the treatment. Expression of antioxidant genes, including Cyb5r3 and Nqo1, was favorably changed by chronic BAY 54-6544 treatment. In summary, BAY 54-6544 treatment improved the vascular function and survival rates in mice with accelerated ageing, which may have implication in prolonging health span in progeria and normal ageing.
Collapse
Affiliation(s)
- Ehsan Ataei Ataabadi
- Division of Pharmacology and Vascular Medicine, Department of Internal MedicineErasmus MCRotterdamthe Netherlands
| | - Keivan Golshiri
- Division of Pharmacology and Vascular Medicine, Department of Internal MedicineErasmus MCRotterdamthe Netherlands
| | - Annika A. Jüttner
- Division of Pharmacology and Vascular Medicine, Department of Internal MedicineErasmus MCRotterdamthe Netherlands
| | - René de Vries
- Division of Pharmacology and Vascular Medicine, Department of Internal MedicineErasmus MCRotterdamthe Netherlands
| | - Ingrid Van den Berg‐Garrelds
- Division of Pharmacology and Vascular Medicine, Department of Internal MedicineErasmus MCRotterdamthe Netherlands
| | - Nicole M. A. Nagtzaam
- Laboratory Medical Immunology, Department of ImmunologyErasmus MCRotterdamthe Netherlands
| | - Hina N. Khan
- Department of Molecular GeneticsErasmus MC Rotterdamthe Netherlands
| | - Frank P. J. Leijten
- Division of Pharmacology and Vascular Medicine, Department of Internal MedicineErasmus MCRotterdamthe Netherlands
| | | | - Willem A. Dik
- Laboratory Medical Immunology, Department of ImmunologyErasmus MCRotterdamthe Netherlands
| | - Ingrid van der Pluijm
- Department of Molecular GeneticsErasmus MC Rotterdamthe Netherlands
- Department of Vascular SurgeryErasmus MC Rotterdamthe Netherlands
| | - A. H. Jan Danser
- Division of Pharmacology and Vascular Medicine, Department of Internal MedicineErasmus MCRotterdamthe Netherlands
| | - Peter Sandner
- Bayer AG, Pharmaceuticals R&D, Pharma Research Center Wuppertal, Germany & Hannover Medical SchoolInstitute of PharmacologyHannoverGermany
| | - Anton J. M. Roks
- Division of Pharmacology and Vascular Medicine, Department of Internal MedicineErasmus MCRotterdamthe Netherlands
| |
Collapse
|
26
|
Aydin P, Magden ZBA, Uzuncakmak SK, Halici H, Akgun N, Mendil AS, Mokhtare B, Cadirci E. Avanafil as a Novel Therapeutic Agent Against LPS-Induced Acute Lung Injury via Increasing CGMP to Downregulate the TLR4-NF-κB-NLRP3 Inflammasome Signaling Pathway. Lung 2022; 200:561-572. [PMID: 36040529 DOI: 10.1007/s00408-022-00564-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/14/2022] [Indexed: 01/16/2023]
Abstract
AIM We demonstrate the effect of PDE5 inhibitors in cases of acute lung injury via the relationship between cGMP/NO and the TLR4-NF-κB-NLRP3 pathway. MATERIALS AND METHODS This study was performed with 30 male Wistar albino rats. Lipopolysaccharide (LPS) was administered intratracheally to the rats and acute lung injury (ALI) was induced. Twelve hours after LPS administration, avanafil, prepared at suitable doses according to the body weights of the animals, was administered by oral gavage. Lung tissue samples of all groups were examined histopathologically and by immunochemical staining (IL-1β, iNOS, TLR4, and NF-κB). The iNOS, NLRP3, and IL-1B mRNA expression levels in the lung tissues were measured by RT-PCR. The left upper lobes of the rat lungs were dried at 70 °C for 48 h and lung water content was calculated. RESULT Statistically significant increases in iNOS, NLRP3, and IL-1β mRNA expressions were observed in the rats with ALI compared to the healthy controls (p < 0.0001). Those increased expressions were reduced at both doses of avanafil (p < 0.0001). This reduction was found to be greater at 20 mg/kg (p < 0.0001). IL-1β, iNOS, TLR4, and NF-κB immunopositivity was moderate/severe in the ALI group and mild in the group with ALI + avanafil at 20 mg/kg (p < 0.05). When the wet/dry lung ratios were calculated, a statistically significant increase was seen in the ALI group compared to the healthy rats (p < 0.05). That increase was decreased with both avanafil doses (p < 0.05). CONCLUSION We suggest that avanafil may prevent the progression of ALI and be effective in its treatment. We hope that this study will be supported by future clinical studies to yield a new indication for avanafil.
Collapse
Affiliation(s)
- Pelin Aydin
- Department of Anesthesiology and Reanimation, Educational and Research Hospital, Erzurum, Turkey. .,Department of Pharmacology, Faculty of Medicine, Ataturk University, Ataturk University Campus, Ataturk District, Erzurum, 25240, Yakutiye, Turkey.
| | - Zeynep Berna Aksakalli Magden
- Department of Pharmacology, Faculty of Medicine, Ataturk University, Ataturk University Campus, Ataturk District, Erzurum, 25240, Yakutiye, Turkey
| | | | - Hamza Halici
- Department of Pharmacology, Faculty of Medicine, Ataturk University, Ataturk University Campus, Ataturk District, Erzurum, 25240, Yakutiye, Turkey.,Department of Hınıs Vocational Training School, Ataturk University, Erzurum, Turkey
| | - Nurullah Akgun
- Department of Pharmacology, Faculty of Medicine, Ataturk University, Ataturk University Campus, Ataturk District, Erzurum, 25240, Yakutiye, Turkey
| | - Ali Sefa Mendil
- Department of Pathology, Faculty of Veterinary Medicine, Erciyes University, Kayseri, Turkey
| | - Behzad Mokhtare
- Department of Pathology, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
| | - Elif Cadirci
- Department of Pharmacology, Faculty of Medicine, Ataturk University, Ataturk University Campus, Ataturk District, Erzurum, 25240, Yakutiye, Turkey
| |
Collapse
|
27
|
Lee HJ. Therapeutic Potential of the Combination of Pentoxifylline and Vitamin-E in Inflammatory Bowel Disease: Inhibition of Intestinal Fibrosis. J Clin Med 2022; 11:jcm11164713. [PMID: 36012952 PMCID: PMC9410449 DOI: 10.3390/jcm11164713] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 08/07/2022] [Accepted: 08/10/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Although intestinal fibrosis is a consequence of recurrent inflammation in Inflammatory bowel disease (IBD), alleviating inflammation alone does not prevent the progression of fibrosis, suggesting that the development of direct anti-fibrotic agents is necessary. This study aimed to evaluate the anti-fibrotic properties of combination treatment with pentoxifylline (PTX) and vitamin E (Vit-E) on human primary intestinal myofibroblasts (HIMFs) and the therapeutic potential of the combination therapy in murine models of IBD. Methods: HIMFs were pretreated with PTX, Vit-E, or both, and incubated with TGF-β1. We performed Western blot, qPCR, collagen staining, and immunofluorescence to estimate the anti-fibrotic effects of PTX and Vit-E. The cytotoxicity of these was investigated through MTT assay. To induce murine models of IBD for in vivo study, C57BL/6 mice were treated with repeated cycles of dextran sulfate sodium (DSS), developing chronic colitis. We examined whether the combined PTX and Vit-E treatment would effectively ameliorate colonic fibrosis in vivo. Results: We found that the co-treatment with PTX and Vit-E suppressed TGF-β1-induced expression of fibrogenic markers, with decreased expression of pERK, pSmad2, and pJNK, more than either treatment alone in HIMFs. Neither PTX nor Vit-E showed any significant cytotoxicity in given concentrations. Consistently with the in vitro results, the co-administration with PTX and Vit-E effectively attenuated colonic fibrosis with recovery from thickening and shortening of colon in murine models of IBD. Conclusions: These findings demonstrated that the combination of PTX and Vit-E exhibits significant anti-fibrotic effects in both HIMFs and in vivo IBD models, providing a promising therapy for IBD.
Collapse
Affiliation(s)
- Hyun Joo Lee
- Division of Gastroenterology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, 59 Yatap-ro, Bundang-gu, Seongnam 13496, Korea; ; Tel.: +82-31-881-7075
- Division of Gastroenterology, Department of Internal Medicine, Graduate School, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam 13488, Korea
| |
Collapse
|
28
|
Pérez S, Rius-Pérez S. Macrophage Polarization and Reprogramming in Acute Inflammation: A Redox Perspective. Antioxidants (Basel) 2022; 11:antiox11071394. [PMID: 35883885 PMCID: PMC9311967 DOI: 10.3390/antiox11071394] [Citation(s) in RCA: 119] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/07/2022] [Accepted: 07/15/2022] [Indexed: 12/12/2022] Open
Abstract
Macrophage polarization refers to the process by which macrophages can produce two distinct functional phenotypes: M1 or M2. The balance between both strongly affects the progression of inflammatory disorders. Here, we review how redox signals regulate macrophage polarization and reprogramming during acute inflammation. In M1, macrophages augment NADPH oxidase isoform 2 (NOX2), inducible nitric oxide synthase (iNOS), synaptotagmin-binding cytoplasmic RNA interacting protein (SYNCRIP), and tumor necrosis factor receptor-associated factor 6 increase oxygen and nitrogen reactive species, which triggers inflammatory response, phagocytosis, and cytotoxicity. In M2, macrophages down-regulate NOX2, iNOS, SYNCRIP, and/or up-regulate arginase and superoxide dismutase type 1, counteract oxidative and nitrosative stress, and favor anti-inflammatory and tissue repair responses. M1 and M2 macrophages exhibit different metabolic profiles, which are tightly regulated by redox mechanisms. Oxidative and nitrosative stress sustain the M1 phenotype by activating glycolysis and lipid biosynthesis, but by inhibiting tricarboxylic acid cycle and oxidative phosphorylation. This metabolic profile is reversed in M2 macrophages because of changes in the redox state. Therefore, new therapies based on redox mechanisms have emerged to treat acute inflammation with positive results, which highlights the relevance of redox signaling as a master regulator of macrophage reprogramming.
Collapse
|
29
|
Lactobacillus paracasei CCFM1223 Protects against Lipopolysaccharide-Induced Acute Liver Injury in Mice by Regulating the “Gut–Liver” Axis. Microorganisms 2022; 10:microorganisms10071321. [PMID: 35889040 PMCID: PMC9319883 DOI: 10.3390/microorganisms10071321] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/26/2022] [Accepted: 06/28/2022] [Indexed: 12/12/2022] Open
Abstract
Background: Lactobacillus paracasei CCFM1223, a probiotic previously isolated from the healthy people’s intestine, exerts the beneficial influence of preventing the development of inflammation. Methods: The aim of this research was to explore the beneficial effects of L. paracasei CCFM1223 to prevent lipopolysaccharide (LPS)-induced acute liver injury (ALI) and elaborate on its hepatoprotective mechanisms. Results: L. paracasei CCFM1223 pretreatment remarkably decreased the activities of serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) in mice with LPS treatment and remarkably recovered LPS-induced the changes in inflammatory cytokines (tumor necrosis factor-α (TNF-α), transforming growth factor-β (TGF-β), interleukin (IL)-1β, IL-6, IL-17, IL-10, and LPS) and antioxidative enzymes activities (total antioxidant capacity (T-AOC), superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), and catalase (CAT)). Metagenomic analysis showed that L. paracasei CCFM1223 pretreatment remarkably increased the relative abundance of Catabacter compared with the LPS group but remarkably reduced the relative abundance of [Eubacterium] xylanophilumgroup, ASF356, LachnospiraceaeNK4A136group, and Lachnoclostridium, which is closely associated with the inflammation cytokines and antioxidative enzymes. Furthermore, L. paracasei CCFM1223 pretreatment remarkably increased the colonic, serum, and hepatic IL-22 levels in ALI mice. In addition, L. paracasei CCFM1223 pretreatment remarkably down-regulated the hepatic Tlr4 and Nf-kβ transcriptions and significantly up-regulated the hepatic Tlr9, Tak1, Iκ-Bα, and Nrf2 transcriptions in ALI mice. Conclusions: L. paracasei CCFM1223 has a hepatoprotective function in ameliorating LPS-induced ALI by regulating the “gut–liver” axis.
Collapse
|
30
|
Sun K, Kong F, Lin F, Li F, Sun J, Ren C, Zheng B, Shi J. Vericiguat Modulates Osteoclast Differentiation and Bone Resorption via a Balance between VASP and NF- κB Pathways. Mediators Inflamm 2022; 2022:1625290. [PMID: 35757109 PMCID: PMC9225892 DOI: 10.1155/2022/1625290] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/27/2022] [Accepted: 06/06/2022] [Indexed: 11/18/2022] Open
Abstract
Bone homeostasis has been a dynamic equilibrium between osteoclasts (OCs) and osteoblasts (OBs). However, excessive activation of OCs could disturb the bone homeostasis. As a result, effective medical interventions for patients are greatly demanding. NO/guanylate cyclase (GC)/cGMP signaling cascade has been previously reported to regulate bone metabolism, and GC plays a significantly critical role. Vericiguat, as a novel oral soluble guanylate cyclase (sGC) stimulator, has been firstly reported in 2020 to treat patients with heart failure. Nevertheless, the biological effects of Vericiguat on the function of OCs have not yet been explored. In this present study, we found that Vericiguat with the concentration between 0 and 8 μM was noncytotoxic to bone marrow-derived monocyte-macrophage lineage (BMMs). Vericiguat could enhance the differentiation of OCs at concentration of 500 nM, whereas it inhibited OC differentiation at 8 μM. In addition, Vericiguat also showed dual effects on OC fusion and bone resorption in a dose-dependent manner. Furthermore, a molecular assay suggested that the dual regulatory effects of Vericiguat on OCs were mediated by the bidirectional activation of the IκB-α/NF-κB signaling pathway. Taken together, our present study demonstrated the dual effects of Vericiguat on the formation of functional OCs. The regulatory effects of Vericiguat on OCs were achieved by the bidirectional modulation of the IκB-α/NF-κB signaling pathway, and a potential balance between the IκB-α/NF-κB signaling pathway and sGC/cGMP/VASP may exist.
Collapse
Affiliation(s)
- Kaiqiang Sun
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Fanqi Kong
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Feng Lin
- Department of Orthopedic Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Shandong, China
| | - Fudong Li
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Jingchuan Sun
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Changzhen Ren
- Department of General Practice, The 960th Hospital of PLA, Jinan, China
| | - Bing Zheng
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Jiangang Shi
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| |
Collapse
|
31
|
Yu L, Hong W, Lu S, Li Y, Guan Y, Weng X, Feng Z. The NLRP3 Inflammasome in Non-Alcoholic Fatty Liver Disease and Steatohepatitis: Therapeutic Targets and Treatment. Front Pharmacol 2022; 13:780496. [PMID: 35350750 PMCID: PMC8957978 DOI: 10.3389/fphar.2022.780496] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 02/17/2022] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is among the most prevalent primary liver diseases worldwide and can develop into various conditions, ranging from simple steatosis, through non-alcoholic steatohepatitis (NASH), to fibrosis, and eventually cirrhosis and hepatocellular carcinoma. Nevertheless, there is no effective treatment for NAFLD due to the complicated etiology. Recently, activation of the NLPR3 inflammasome has been demonstrated to be a contributing factor in the development of NAFLD, particularly as a modulator of progression from initial hepatic steatosis to NASH. NLRP3 inflammasome, as a caspase-1 activation platform, is critical for processing key pro-inflammatory cytokines and pyroptosis. Various stimuli involved in NAFLD can activate the NLRP3 inflammasome, depending on the diverse cellular stresses that they cause. NLRP3 inflammasome-related inhibitors and agents for NAFLD treatment have been tested and demonstrated positive effects in experimental models. Meanwhile, some drugs have been applied in clinical studies, supporting this therapeutic approach. In this review, we discuss the activation, biological functions, and treatment targeting the NLRP3 inflammasome in the context of NAFLD progression. Specifically, we focus on the different types of therapeutic agents that can inhibit the NLRP3 inflammasome and summarize their pharmacological effectiveness for NAFLD treatment.
Collapse
Affiliation(s)
- Lili Yu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China.,Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, China.,The Third Clinical College of Xinxiang Medical University, Xinxiang, China.,Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, China
| | - Wei Hong
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China.,Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, China
| | - Shen Lu
- The Third Clinical College of Xinxiang Medical University, Xinxiang, China
| | - Yanrong Li
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China.,Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, China
| | - Yaya Guan
- The Third Clinical College of Xinxiang Medical University, Xinxiang, China
| | - Xiaogang Weng
- The Third Clinical College of Xinxiang Medical University, Xinxiang, China
| | - Zhiwei Feng
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China.,Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, China.,Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
32
|
The Role and Mechanism of Oxidative Stress and Nuclear Receptors in the Development of NAFLD. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6889533. [PMID: 34745420 PMCID: PMC8566046 DOI: 10.1155/2021/6889533] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 10/11/2021] [Indexed: 12/12/2022]
Abstract
The overproduction of reactive oxygen species (ROS) and consequent oxidative stress contribute to the pathogenesis of acute and chronic liver diseases. It is now acknowledged that nonalcoholic fatty liver disease (NAFLD) is characterized as a redox-centered disease due to the role of ROS in hepatic metabolism. However, the underlying mechanisms accounting for these alternations are not completely understood. Several nuclear receptors (NRs) are dysregulated in NAFLD, and have a direct influence on the expression of a set of genes relating to the progress of hepatic lipid homeostasis and ROS generation. Meanwhile, the NRs act as redox sensors in response to metabolic stress. Therefore, targeting NRs may represent a promising strategy for improving oxidation damage and treating NAFLD. This review summarizes the link between impaired lipid metabolism and oxidative stress and highlights some NRs involved in regulating oxidant/antioxidant turnover in the context of NAFLD, shedding light on potential therapies based on NR-mediated modulation of ROS generation and lipid accumulation.
Collapse
|
33
|
Liang W, Xie BK, Ding PW, Wang M, Yuan J, Cheng X, Liao YH, Yu M. Sacubitril/Valsartan Alleviates Experimental Autoimmune Myocarditis by Inhibiting Th17 Cell Differentiation Independently of the NLRP3 Inflammasome Pathway. Front Pharmacol 2021; 12:727838. [PMID: 34603042 PMCID: PMC8479108 DOI: 10.3389/fphar.2021.727838] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 09/03/2021] [Indexed: 12/11/2022] Open
Abstract
Sacubitril/valsartan (Sac/Val) is a recently approved drug that is commonly used for treatment of heart failure. Several studies indicated that Sac/Val also regulated the secretion of inflammatory factors. However, the effect and mechanism of this drug modulation of inflammatory immune responses are uncertain. In this study, an experimental autoimmune myocarditis (EAM) mouse model was established by injection of α-myosin-heavy chain peptides. The effect of oral Sac/Val on EAM was evaluated by histological staining of heart tissues, measurements of cardiac troponin T and inflammatory markers (IL-6 and hsCRP). The effects of Sac/Val on NLRP3 inflammasome activation and Th1/Th17 cell differentiation were also determined. To further explore the signaling pathways, the expressions of cardiac soluble guanylyl cyclase (sGC) and NF-κB p65 were investigated. The results showed that Sac/Val downregulated the inflammatory response and attenuated the severity of EAM, but did not influence NLRP3 inflammasomes activation. Moreover, Sac/Val treatment inhibited cardiac Th17 cell differentiation, and this might be associated with sGC/NF-κB p65 signaling pathway. These findings indicate the potential use of Sac/Val for treatment of myocarditis.
Collapse
Affiliation(s)
- Wei Liang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bai-Kang Xie
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pei-Wu Ding
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Wang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Yuan
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Hua Liao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Miao Yu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
34
|
Mi J, Yang Y, Yao H, Huan Z, Xu C, Ren Z, Li W, Tang Y, Fu R, Ge X. Inhibition of heat shock protein family A member 8 attenuates spinal cord ischemia-reperfusion injury via astrocyte NF-κB/NLRP3 inflammasome pathway : HSPA8 inhibition protects spinal ischemia-reperfusion injury. J Neuroinflammation 2021; 18:170. [PMID: 34362408 PMCID: PMC8349068 DOI: 10.1186/s12974-021-02220-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 07/15/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Astrocyte over-activation and extensive neuron loss are the main characteristic pathological features of spinal cord ischemia-reperfusion injury (SCII). Prior studies have placed substantial emphasis on the role of heat shock protein family A member 8 (HSPA8) on postischemic myocardial inflammation and cardiac dysfunction. However, it has never been determined whether HSPA8 participates in astrocyte activation and thus mediated neuroinflammation associated with SCII. METHODS The left renal artery ligation-induced SCII rat models and oxygen-glucose deprivation and reoxygenation (OGD/R)-induced rat primary cultured astrocytes were established. The lentiviral vector encoding short hairpin RNA targeting HSPA8 was delivered to the spinal cord by intrathecal administration or to culture astrocytes. Then, the spinal neuron survival, gliosis, and nod-like receptor pyrin domain-containing 3 (NLRP3) inflammasome and its related pro-inflammatory cytokines were analyzed. RESULTS SCII significantly enhanced the GFAP and HSPA8 expression in the spinal cord, resulting in blood-brain barrier breakdown and the dramatical loss of spinal neuron and motor function. Moreover, injury also increased spinal nuclear factor-kappa B (NF-κB) p65 phosphorylation, NLRP3 inflammasome-mediated caspase-1 activation, and subsequent interleukin (IL)-1β as well as IL-18 secretion. Silencing the HSPA8 expression efficiently ameliorated the spinal cord tissue damage and promoted motor function recovery after SCII, through blockade of the astrocyte activation and levels of phosphorylated NF-κB, NLRP3, caspase-1, IL-1β, and IL-18. Further in vitro studies confirmed that HSPA8 knockdown protected astrocytes from OGD/R-induced injury via the blockade of NF-κB and NLRP3 inflammasome activation. CONCLUSION Our findings indicate that knockdown of HSPA8 inhibits spinal astrocytic damage after SCII, which may provide a promising therapeutic strategy for SCII treatment.
Collapse
Affiliation(s)
- Jingyi Mi
- Department of Sports Medicine, Wuxi 9Th Affiliated Hospital of Soochow University, Wuxi, 214000, Jiangsu, China
| | - Yang Yang
- Department of Neurosurgery, Central Hospital of Jinzhou, Jinzhou, 121001, Liaoning, China
| | - Hao Yao
- Department of ICU, Wuxi 9Th Affiliated Hospital of Soochow University, Wuxi, 214000, Jiangsu, China
| | - Zhirong Huan
- Department of ICU, Wuxi 9Th Affiliated Hospital of Soochow University, Wuxi, 214000, Jiangsu, China
| | - Ce Xu
- Department of ICU, Wuxi 9Th Affiliated Hospital of Soochow University, Wuxi, 214000, Jiangsu, China
| | - Zhiheng Ren
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Wenfu Li
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Ying Tang
- Department of Microbiology, Biochemistry, & Molecular Genetics, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, 07103, NJ, USA
- Brain Research Centre and Department of Biology, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
| | - Rao Fu
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China.
| | - Xin Ge
- Department of ICU, Wuxi 9Th Affiliated Hospital of Soochow University, Wuxi, 214000, Jiangsu, China.
- Orthopedic Institution of Wuxi City, Wuxi, 214000, Jiangsu, China.
| |
Collapse
|
35
|
Zhang Y, Wen J, Liu D, Qiu Z, Zhu Q, Li R, Zhang Y. Demethylenetetrahydroberberine alleviates nonalcoholic fatty liver disease by inhibiting the NLRP3 inflammasome and oxidative stress in mice. Life Sci 2021; 281:119778. [PMID: 34192596 DOI: 10.1016/j.lfs.2021.119778] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 05/08/2021] [Accepted: 06/19/2021] [Indexed: 12/20/2022]
Abstract
AIMS Demethylenetetrahydroberberine (DMTHB) is a novel derivative of berberine and demethyleneberberine. This research explored the pharmacological effects and molecular mechanisms of DMTHB on nonalcoholic fatty liver disease (NAFLD). MATERIALS AND METHODS C57BL/6 mice were induced by a methionine- and choline- deficient (MCD) diet and L02 cells were induced by palmitic acid to establish NAFLD animal and cell models. qPCR and western blotting were used to detect the expression of genes and proteins associated with pharmacological mechanism. A biotin-labeled DMTHB pulldown assay was used to further clarify the pharmacological targets. KEY FINDINGS Our results indicated that DMTHB significantly alleviates NAFLD in mice. Biochemical assays showed that serum alanine aminotransferase, aspartate aminotransferase and hepatic lipids were significantly decreased in MCD-induced NAFLD mice orally administered of DMTHB (50 mg/kg or 150 mg/kg body weight daily) for 30 d. qPCR and ELISA analysis demonstrated that DMTHB reduced the expression of serum proinflammatory cytokines, such as TNF-α, IL-1β and IL-6. Moreover, pull-down assays and compound-centric chemical proteomics illustrated that DMTHB inhibited NOD-like receptor protein 3 (NLRP3) inflammasome signaling. In addition, DMTHB also attenuated oxidative stress and endoplasmic reticulum stress by downregulation CYP2E-1 and ATF-4 expression. Moreover, DMTHB treatment ameliorated the liver fibrosis in MCD-induced NAFLD mice by suppressing the expression of TGF-β1, α-SMA and collagen 1A1. SIGNIFICANCE DMTHB targeted the NLRP3 inflammasome to suppress inflammation and inhibited CYP2E1 to reduce oxidative stress and ER stress. Consequently, DMTHB may have therapeutic benefits in the treatment of NAFLD in the clinic.
Collapse
Affiliation(s)
- Yuanqiang Zhang
- State Key Laboratory of Natural Medicines, Department of Biochemistry, China Pharmaceutical University, Nanjing, China
| | - Jing Wen
- State Key Laboratory of Natural Medicines, Department of Biochemistry, China Pharmaceutical University, Nanjing, China
| | - Dongqing Liu
- State Key Laboratory of Natural Medicines, Department of Biochemistry, China Pharmaceutical University, Nanjing, China
| | - Zhen Qiu
- State Key Laboratory of Natural Medicines, Department of Biochemistry, China Pharmaceutical University, Nanjing, China
| | - Qianqian Zhu
- State Key Laboratory of Natural Medicines, Department of Biochemistry, China Pharmaceutical University, Nanjing, China
| | - Ruiyan Li
- State Key Laboratory of Natural Medicines, Department of Biochemistry, China Pharmaceutical University, Nanjing, China
| | - Yubin Zhang
- State Key Laboratory of Natural Medicines, Department of Biochemistry, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
36
|
Wu R, Wang X, Shao Y, Jiang Y, Zhou Y, Lu C. NFATc4 mediates ethanol-triggered hepatocyte senescence. Toxicol Lett 2021; 350:10-21. [PMID: 34192554 DOI: 10.1016/j.toxlet.2021.06.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/18/2021] [Accepted: 06/23/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Hepatocyte senescence is a core event that mediates the occurrence and development of alcoholic liver disease. Nuclear factor of activated T-cells 4 (NFATc4) is a key driver of nonalcoholic steatohepatitis. However, little was known about the implication of NFATc4 for alcoholic liver disease. This study was aimed to investigate the role of NFATc4 in hepatocyte senescence and further elucidate the underlying mechanism. METHODS Real-time PCR, Western blot, immunofluorescence staining, and enzyme-linked immunosorbent assay were performed to explore the role of NFATc4 in hepatocyte senescence. RESULTS NFATc4 was induced in ethanol-incubated hepatocytes. NFATc4 knockdown recovered cell viability and reduced the release of aspartate transaminase, alanine transaminase, and lactic dehydrogenase from ethanol-incubated hepatocytes. NFATc4 knockdown protected mice from alcoholic liver injury and inflammation. NFATc4 knockdown counteracted ethanol-induced hepatocyte senescence, evidenced by decreased senescence-associated β-galactosidase positivity and reduced p16, p21, HMGA1, and γH2AX, which was validated in in vivo studies. Peroxisome proliferator-activated receptor (PPAR)γ was inhibited by NFATc4 in ethanol-treated hepatocytes. PPARγ deficiency abrogated the inhibitory effects of NFATc4 knockdown on hepatocyte senescence, oxidative stress, and hepatic steatosis in mice with alcoholic liver disease. CONCLUSIONS This work discovered that ethanol enhanced NFATc4 expression, which further triggered hepatocyte senescence via repression of PPARγ.
Collapse
Affiliation(s)
- Ruoman Wu
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Xinqi Wang
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Yunyun Shao
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Yiming Jiang
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Ying Zhou
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Chunfeng Lu
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
37
|
Mitochondrial Mutations and Genetic Factors Determining NAFLD Risk. Int J Mol Sci 2021; 22:ijms22094459. [PMID: 33923295 PMCID: PMC8123173 DOI: 10.3390/ijms22094459] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/21/2021] [Accepted: 04/22/2021] [Indexed: 02/07/2023] Open
Abstract
NAFLD (non-alcoholic fatty liver disease) is a widespread liver disease that is often linked with other life-threatening ailments (metabolic syndrome, insulin resistance, diabetes, cardiovascular disease, atherosclerosis, obesity, and others) and canprogress to more severe forms, such as NASH (non-alcoholic steatohepatitis), cirrhosis, and HCC (hepatocellular carcinoma). In this review, we summarized and analyzed data about single nucleotide polymorphism sites, identified in genes related to NAFLD development and progression. Additionally, the causative role of mitochondrial mutations and mitophagy malfunctions in NAFLD is discussed. The role of mitochondria-related metabolites of the urea cycle as a new non-invasive NAFLD biomarker is discussed. While mitochondria DNA mutations and SNPs (single nucleotide polymorphisms) canbe used as effective diagnostic markers and target for treatments, age and ethnic specificity should be taken into account.
Collapse
|