1
|
Liu HJ, Gui LK, Wei H, Zhou XY, Liu ZL, Jin LJ. The role of NF-κB in diabetic cardiomyopathy. ALL LIFE 2024; 17. [DOI: 10.1080/26895293.2024.2397402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 08/20/2024] [Indexed: 01/03/2025] Open
Affiliation(s)
- Huang-Jun Liu
- Department of Cardiology, The First Affiliated Hospital of Yangtze University, Jingzhou, People’s Republic of China
| | - Le-Kun Gui
- Department of Cardiology, The First Affiliated Hospital of Yangtze University, Jingzhou, People’s Republic of China
- School of Medicine, Yangtze University, Jingzhou, People’s Republic of China
| | - Han Wei
- Department of Cardiology, The First Affiliated Hospital of Yangtze University, Jingzhou, People’s Republic of China
| | - Xing-Yu Zhou
- Department of Cardiology, The First Affiliated Hospital of Yangtze University, Jingzhou, People’s Republic of China
- School of Medicine, Yangtze University, Jingzhou, People’s Republic of China
| | - Zhen-Lan Liu
- Department of Anesthesiology, The First Affiliated Hospital of Yangtze University, Jingzhou, People’s Republic of China
| | - Li-Jun Jin
- Department of Cardiology, The First Affiliated Hospital of Yangtze University, Jingzhou, People’s Republic of China
| |
Collapse
|
2
|
Zamanian MY, Alsaab HO, Golmohammadi M, Yumashev A, Jabba AM, Abid MK, Joshi A, Alawadi AH, Jafer NS, Kianifar F, Obakiro SB. NF-κB pathway as a molecular target for curcumin in diabetes mellitus treatment: Focusing on oxidative stress and inflammation. Cell Biochem Funct 2024; 42:e4030. [PMID: 38720663 DOI: 10.1002/cbf.4030] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/05/2024] [Accepted: 04/25/2024] [Indexed: 08/03/2024]
Abstract
Diabetes mellitus (DM) is a collection of metabolic disorder that is characterized by chronic hyperglycemia. Recent studies have demonstrated the crucial involvement of oxidative stress (OS) and inflammatory reactions in the development of DM. Curcumin (CUR), a natural compound derived from turmeric, exerts beneficial effects on diabetes mellitus through its interaction with the nuclear factor kappa B (NF-κB) pathway. Research indicates that CUR targets inflammatory mediators in diabetes, including tumor necrosis factor α (TNF-α) and interleukin-6 (IL-6), by modulating the NF-κB signaling pathway. By reducing the expression of these inflammatory factors, CUR demonstrates protective effects in DM by improving pancreatic β-cells function, normalizing inflammatory cytokines, reducing OS and enhancing insulin sensitivity. The findings reveal that CUR administration effectively lowered blood glucose elevation, reinstated diminished serum insulin levels, and enhanced body weight in Streptozotocin -induced diabetic rats. CUR exerts its beneficial effects in management of diabetic complications through regulation of signaling pathways, such as calcium-calmodulin (CaM)-dependent protein kinase II (CaMKII), peroxisome proliferator-activated receptor gamma (PPAR-γ), NF-κB, and transforming growth factor β1 (TGFB1). Moreover, CUR reversed the heightened expression of inflammatory cytokines (TNF-α, Interleukin-1 beta (IL-1β), IL-6) and chemokines like MCP-1 in diabetic specimens, vindicating its anti-inflammatory potency in counteracting hyperglycemia-induced alterations. CUR diminishes OS, avert structural kidney damage linked to diabetic nephropathy, and suppress NF-κB activity. Furthermore, CUR exhibited a protective effect against diabetic cardiomyopathy, lung injury, and diabetic gastroparesis. Conclusively, the study posits that CUR could potentially offer therapeutic benefits in relieving diabetic complications through its influence on the NF-κB pathway.
Collapse
Affiliation(s)
- Mohammad Yasin Zamanian
- Department of Physiology, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hashem O Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, Taif, Saudi Arabia
| | - Maryam Golmohammadi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alexey Yumashev
- Department of Prosthetic Dentistry, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Abeer Mhussan Jabba
- Colleges of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Mohammed Kadhem Abid
- Department of Anesthesia, College of Health & Medical Technology, Al-Ayen University, Nasiriyah, Iraq
| | - Abhishek Joshi
- Department of Liberal Arts School of Liberal Arts, Uttaranchal University, Dehradun, India
| | - Ahmed Hussien Alawadi
- College of Technical Engineering, The Islamic University, Najaf, Iraq
- College of Technical Engineering, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| | - Noor S Jafer
- Department of Medical Laboratory Technologies, Al Rafidain University College, Bagdad, Iraq
| | - Farzaneh Kianifar
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Samuel Baker Obakiro
- Department of Pharmacology and Therapeutics, Faculty of Health Sciences, Busitema University, Mbale, Uganda
| |
Collapse
|
3
|
Rezaee A, Rahmanian P, Nemati A, Sohrabifard F, Karimi F, Elahinia A, Ranjbarpazuki A, Lashkarbolouki R, Dezfulian S, Zandieh MA, Salimimoghadam S, Nabavi N, Rashidi M, Taheriazam A, Hashemi M, Hushmandi K. NF-ĸB axis in diabetic neuropathy, cardiomyopathy and nephropathy: A roadmap from molecular intervention to therapeutic strategies. Heliyon 2024; 10:e29871. [PMID: 38707342 PMCID: PMC11066643 DOI: 10.1016/j.heliyon.2024.e29871] [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: 12/03/2023] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 05/07/2024] Open
Abstract
Diabetes mellitus (DM) is a metabolic illness defined by elevated blood glucose levels, mediating various tissue alterations, including the dysfunction of vital organs. Diabetes mellitus (DM) can lead to many consequences that specifically affect the brain, heart, and kidneys. These issues are known as neuropathy, cardiomyopathy, and nephropathy, respectively. Inflammation is acknowledged as a pivotal biological mechanism that contributes to the development of various diabetes consequences. NF-κB modulates inflammation and the immune system at the cellular level. Its abnormal regulation has been identified in several clinical situations, including cancer, inflammatory bowel illnesses, cardiovascular diseases, and Diabetes Mellitus (DM). The purpose of this review is to evaluate the potential impact of NF-κB on complications associated with DM. Enhanced NF-κB activity promotes inflammation, resulting in cellular harm and compromised organ performance. Phytochemicals, which are therapeutic molecules, can potentially decline the NF-κB level, therefore alleviating inflammation and the progression of problems correlated with DM. More importantly, the regulation of NF-κB can be influenced by various factors, such as TLR4 in DM. Highlighting these factors can facilitate the development of novel therapies in the future.
Collapse
Affiliation(s)
- Aryan Rezaee
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Parham Rahmanian
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Amirreza Nemati
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farima Sohrabifard
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Fatemeh Karimi
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Ali Elahinia
- Department of Clinical Science, Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Ali Ranjbarpazuki
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Rozhin Lashkarbolouki
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Sadaf Dezfulian
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Mohammad Arad Zandieh
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6, Vancouver, BC, Canada
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Afshin Taheriazam
- Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| |
Collapse
|
4
|
Visanji M, Venegas-Pino DE, Werstuck GH. Understanding One Half of the Sex Difference Equation: The Modulatory Effects of Testosterone on Diabetic Cardiomyopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:551-561. [PMID: 38061627 DOI: 10.1016/j.ajpath.2023.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/31/2023] [Accepted: 11/20/2023] [Indexed: 12/20/2023]
Abstract
Diabetes is a prevalent disease, primarily characterized by high blood sugar (hyperglycemia). Significantly higher rates of myocardial dysfunction have been noted in individuals with diabetes, even in those without coronary artery disease or high blood pressure (hypertension). Numerous molecular mechanisms have been identified through which diabetes contributes to the pathology of diabetic cardiomyopathy, which presents as cardiac hypertrophy and fibrosis. At the cellular level, oxidative stress and inflammation in cardiomyocytes are triggered by hyperglycemia. Although males are generally more likely to develop cardiovascular disease than females, diabetic males are less likely to develop diabetic cardiomyopathy than are diabetic females. One reason for these differences may be the higher levels of serum testosterone in males compared with females. Although testosterone appears to protect against cardiomyocyte oxidative stress and exacerbate hypertrophy, its role in inflammation and fibrosis is much less clear. Additional preclinical and clinical studies will be required to delineate testosterone's effect on the diabetic heart.
Collapse
Affiliation(s)
- Mika'il Visanji
- Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | | | - Geoff H Werstuck
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
5
|
Hao J, Zhou J, Hu S, Zhang P, Wu H, Yang J, Zhao B, Liu H, Lin H, Chi J, Lou D. RTA 408 ameliorates diabetic cardiomyopathy by activating Nrf2 to regulate mitochondrial fission and fusion and inhibiting NF-κB-mediated inflammation. Am J Physiol Cell Physiol 2024; 326:C331-C347. [PMID: 38047307 DOI: 10.1152/ajpcell.00467.2023] [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: 09/19/2023] [Revised: 11/28/2023] [Accepted: 11/28/2023] [Indexed: 12/05/2023]
Abstract
Diabetic cardiomyopathy (dCM) is a major complication of diabetes; however, specific treatments for dCM are currently lacking. RTA 408, a semisynthetic triterpenoid, has shown therapeutic potential against various diseases by activating the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway. We established in vitro and in vivo models using high glucose toxicity and db/db mice, respectively, to simulate dCM. Our results demonstrated that RTA 408 activated Nrf2 and alleviated various dCM-related cardiac dysfunctions, both in vivo and in vitro. Additionally, it was found that silencing the Nrf2 gene eliminated the cardioprotective effect of RTA 408. RTA 408 ameliorated oxidative stress in dCM mice and high glucose-exposed H9C2 cells by activating Nrf2, inhibiting mitochondrial fission, exerting anti-inflammatory effects through the Nrf2/NF-κB axis, and ultimately suppressing apoptosis, thereby providing cardiac protection against dCM. These findings provide valuable insights for potential dCM treatments.NEW & NOTEWORTHY We demonstrated first that the nuclear factor erythroid 2-related factor 2 (Nrf2) activator RTA 408 has a protective effect against diabetic cardiomyopathy. We found that RTA 408 could stimulate the nuclear entry of Nrf2 protein, regulate the mitochondrial fission-fusion balance, and redistribute p65, which significantly alleviated the oxidative stress level in cardiomyocytes, thereby reducing apoptosis and inflammation, and protecting the systolic and diastolic functions of the heart.
Collapse
Affiliation(s)
- Jinjin Hao
- Department of Endocrinology, Shaoxing People's Hospital, Shaoxing, China
| | - Jiedong Zhou
- College of Medicine, Shaoxing University, Shaoxing, China
| | - Songqing Hu
- Zhejiang University School of Medicine, Hangzhou, China
| | - Peipei Zhang
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Haowei Wu
- Zhejiang University School of Medicine, Hangzhou, China
| | - Juntao Yang
- College of Medicine, Shaoxing University, Shaoxing, China
| | - Bingjie Zhao
- College of Medicine, Shaoxing University, Shaoxing, China
| | - Hanxuan Liu
- College of Medicine, Shaoxing University, Shaoxing, China
| | - Hui Lin
- The Affiliated Lihuili Hospital of Ningbo University, Ningbo, China
| | - Jufang Chi
- Department of Cardiology, Zhuji People's Hospital, Shaoxing, China
| | - Dajun Lou
- Department of Endocrinology, Shaoxing People's Hospital, Shaoxing, China
| |
Collapse
|
6
|
Leng J, Wang C, Liang Z, Qiu F, Zhang S, Yang Y. An updated review of YAP: A promising therapeutic target against cardiac aging? Int J Biol Macromol 2024; 254:127670. [PMID: 37913886 DOI: 10.1016/j.ijbiomac.2023.127670] [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: 07/11/2023] [Revised: 09/05/2023] [Accepted: 10/23/2023] [Indexed: 11/03/2023]
Abstract
The transcriptional co-activator Yes-associated protein (YAP) functions as a downstream effector of the Hippo signaling pathway and plays a crucial role in cardiomyocyte survival. In its non-phosphorylated activated state, YAP binds to transcription factors, activating the transcription of downstream target genes. It also regulates cell proliferation and survival by selectively binding to enhancers and activating target genes. However, the upregulation of the Hippo pathway in human heart failure inhibits cardiac regeneration and disrupts astrogenesis, thus preventing the nuclear translocation of YAP. Existing literature indicates that the Hippo/YAP axis contributes to inflammation and fibrosis, potentially playing a role in the development of cardiac, vascular and renal injuries. Moreover, it is a key mediator of myofibroblast differentiation and fibrosis in the infarcted heart. Given these insights, can we harness YAP's regenerative potential in a targeted manner? In this review, we provide a detailed discussion of the Hippo signaling pathway and consolidate concepts for the development and intervention of cardiac anti-aging drugs to leverage YAP signaling as a pivotal target.
Collapse
Affiliation(s)
- Jingzhi Leng
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China; School of Physical Education, Qingdao University, China
| | - Chuanzhi Wang
- College of Sports Science, South China Normal University, Guangzhou, China
| | - Zhide Liang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China; Qingdao Cancer Institute, Qingdao University, Qingdao, China
| | - Fanghui Qiu
- School of Physical Education, Qingdao University, China
| | - Shuangshuang Zhang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China; Qingdao Cancer Institute, Qingdao University, Qingdao, China; School of Physical Education, Qingdao University, China.
| | - Yuan Yang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China; Qingdao Cancer Institute, Qingdao University, Qingdao, China; School of Physical Education, Qingdao University, China.
| |
Collapse
|
7
|
Maiese K. Artificial Intelligence and Disease Signature Pathways: Driving Innovation to Elucidate Underlying Pathogenic Mechanisms. Curr Neurovasc Res 2024; 21:229-233. [PMID: 38910427 DOI: 10.2174/1567202621999240621122700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
|
8
|
Du H, Huangfu W, Liu Z, Jia G, Zhao F, Cheng W. 5-Demethylnobiletin Ameliorates Isoproterenol-Induced Cardiac Fibrosis and Apoptosis by Repressing the Sirt1/FOXO3a/NF-κB and Wnt/β-Catenin Pathways. Biol Pharm Bull 2024; 47:1774-1785. [PMID: 39477471 DOI: 10.1248/bpb.b24-00122] [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] [Indexed: 11/07/2024]
Abstract
Apoptosis and fibrosis are two main factors leading to heart failure. 5-Demethylnobiletin (5-OH-Nob) is a natural polymethoxyflavone derived from the peel of citrus fruits that has many biological effects, such as antioxidative stress and anti-inflammatory effects. Here, we aimed to probe the function and mechanism of 5-OH-Nob in myocardial damage. Primary rat cardiac fibroblasts were exposed to isoproterenol (ISO, 10 µM) to establish an in vitro model of cardiac damage, and ISO (30 mg/kg/d) was used to induce myocardial fibrosis in mice. 5-OH-Nob was used for treatment in vivo and ex vivo. Functional assays revealed that 5-OH-Nob alleviated the apoptosis and fibrosis of cardiac fibroblasts treated with ISO and increased cell viability (p < 0.05). In vivo, 5-OH-Nob treatment ameliorated cardiac injury in ISO-treated mice (p < 0.05). Mechanistically, 5-OH-Nob treatment enhanced Sirt1 expression and suppressed ISO-mediated activation of the FOXO3a/nuclear transcription factor-κB (NF-κB) and Wnt/β-catenin pathways. Furthermore, Sirt1 inhibition attenuated the protective effect of 5-OH-Nob on ISO-induced cardiac apoptosis and fibrosis. Overall, 5-demethylnobiletin mediates the Sirt1/FOXO3a/NF-κB and Wnt/β-catenin pathways to mitigate ISO-induced myocardial fibrosis and apoptosis.
Collapse
Affiliation(s)
- Haiyan Du
- Department of General Practice, Affiliated Hospital of Inner Mongolia Medical University
| | - Weizhong Huangfu
- Department of General Practice, Affiliated Hospital of Inner Mongolia Medical University
| | - Zhonghua Liu
- Department of General Practice, Affiliated Hospital of Inner Mongolia Medical University
| | - Gaopeng Jia
- Department of General Practice, Affiliated Hospital of Inner Mongolia Medical University
| | - Feng Zhao
- Department of General Practice, Affiliated Hospital of Inner Mongolia Medical University
| | - Wenjun Cheng
- Department of General Practice, Affiliated Hospital of Inner Mongolia Medical University
| |
Collapse
|
9
|
Maiese K. Cornerstone Cellular Pathways for Metabolic Disorders and Diabetes Mellitus: Non-Coding RNAs, Wnt Signaling, and AMPK. Cells 2023; 12:2595. [PMID: 37998330 PMCID: PMC10670256 DOI: 10.3390/cells12222595] [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: 09/21/2023] [Revised: 10/31/2023] [Accepted: 11/07/2023] [Indexed: 11/25/2023] Open
Abstract
Metabolic disorders and diabetes (DM) impact more than five hundred million individuals throughout the world and are insidious in onset, chronic in nature, and yield significant disability and death. Current therapies that address nutritional status, weight management, and pharmacological options may delay disability but cannot alter disease course or functional organ loss, such as dementia and degeneration of systemic bodily functions. Underlying these challenges are the onset of aging disorders associated with increased lifespan, telomere dysfunction, and oxidative stress generation that lead to multi-system dysfunction. These significant hurdles point to the urgent need to address underlying disease mechanisms with innovative applications. New treatment strategies involve non-coding RNA pathways with microRNAs (miRNAs) and circular ribonucleic acids (circRNAs), Wnt signaling, and Wnt1 inducible signaling pathway protein 1 (WISP1) that are dependent upon programmed cell death pathways, cellular metabolic pathways with AMP-activated protein kinase (AMPK) and nicotinamide, and growth factor applications. Non-coding RNAs, Wnt signaling, and AMPK are cornerstone mechanisms for overseeing complex metabolic pathways that offer innovative treatment avenues for metabolic disease and DM but will necessitate continued appreciation of the ability of each of these cellular mechanisms to independently and in unison influence clinical outcome.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, New York, NY 10022, USA
| |
Collapse
|
10
|
Maiese K. The impact of aging and oxidative stress in metabolic and nervous system disorders: programmed cell death and molecular signal transduction crosstalk. Front Immunol 2023; 14:1273570. [PMID: 38022638 PMCID: PMC10663950 DOI: 10.3389/fimmu.2023.1273570] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Life expectancy is increasing throughout the world and coincides with a rise in non-communicable diseases (NCDs), especially for metabolic disease that includes diabetes mellitus (DM) and neurodegenerative disorders. The debilitating effects of metabolic disorders influence the entire body and significantly affect the nervous system impacting greater than one billion people with disability in the peripheral nervous system as well as with cognitive loss, now the seventh leading cause of death worldwide. Metabolic disorders, such as DM, and neurologic disease remain a significant challenge for the treatment and care of individuals since present therapies may limit symptoms but do not halt overall disease progression. These clinical challenges to address the interplay between metabolic and neurodegenerative disorders warrant innovative strategies that can focus upon the underlying mechanisms of aging-related disorders, oxidative stress, cell senescence, and cell death. Programmed cell death pathways that involve autophagy, apoptosis, ferroptosis, and pyroptosis can play a critical role in metabolic and neurodegenerative disorders and oversee processes that include insulin resistance, β-cell function, mitochondrial integrity, reactive oxygen species release, and inflammatory cell activation. The silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), AMP activated protein kinase (AMPK), and Wnt1 inducible signaling pathway protein 1 (WISP1) are novel targets that can oversee programmed cell death pathways tied to β-nicotinamide adenine dinucleotide (NAD+), nicotinamide, apolipoprotein E (APOE), severe acute respiratory syndrome (SARS-CoV-2) exposure with coronavirus disease 2019 (COVID-19), and trophic factors, such as erythropoietin (EPO). The pathways of programmed cell death, SIRT1, AMPK, and WISP1 offer exciting prospects for maintaining metabolic homeostasis and nervous system function that can be compromised during aging-related disorders and lead to cognitive impairment, but these pathways have dual roles in determining the ultimate fate of cells and organ systems that warrant thoughtful insight into complex autofeedback mechanisms.
Collapse
Affiliation(s)
- Kenneth Maiese
- Innovation and Commercialization, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
11
|
Ishioka M, Nihashi Y, Sunagawa Y, Umezawa K, Shimosato T, Kagami H, Morimoto T, Takaya T. Myogenetic Oligodeoxynucleotide Induces Myocardial Differentiation of Murine Pluripotent Stem Cells. Int J Mol Sci 2023; 24:14380. [PMID: 37762684 PMCID: PMC10532123 DOI: 10.3390/ijms241814380] [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: 08/05/2023] [Revised: 09/16/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
An 18-base myogenetic oligodeoxynucleotide (myoDN), iSN04, acts as an anti-nucleolin aptamer and induces myogenic differentiation of skeletal muscle myoblasts. This study investigated the effect of iSN04 on murine embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). In the undifferentiated state, iSN04 inhibited the proliferation of ESCs and iPSCs but did not affect the expression of pluripotent markers. In the differentiating condition, iSN04 treatment of ESCs/iPSCs from day 5 onward dramatically induced differentiation into Nkx2-5+ beating cardiomyocytes with upregulation of Gata4, Isl1, and Nkx2-5, whereas iSN04 treatment from earlier stages completely inhibited cardiomyogenesis. RNA sequencing revealed that iSN04 treatment from day 5 onward contributes to the generation of cardiac progenitors by modulating the Wnt signaling pathway. Immunostaining showed that iSN04 suppressed the cytoplasmic translocation of nucleolin and restricted it to the nucleoli. These results demonstrate that nucleolin inhibition by iSN04 facilitates the terminal differentiation of cardiac mesoderm into cardiomyocytes but interferes with the differentiation of early mesoderm into the cardiac lineage. This is the first report on the generation of cardiomyocytes from pluripotent stem cells using a DNA aptamer. Since iSN04 did not induce hypertrophic responses in primary-cultured cardiomyocytes, iSN04 would be useful and safe for the regenerative therapy of heart failure using stem cell-derived cardiomyocytes.
Collapse
Affiliation(s)
- Mina Ishioka
- Department of Agriculture, Graduate School of Science and Technology, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan; (M.I.); (T.S.)
| | - Yuma Nihashi
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology, Central 5-41, 1-1-1 Higashi, Tsukuba 305-8565, Ibaraki, Japan;
| | - Yoichi Sunagawa
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (Y.S.); (T.M.)
| | - Koji Umezawa
- Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan; (K.U.); (H.K.)
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan
| | - Takeshi Shimosato
- Department of Agriculture, Graduate School of Science and Technology, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan; (M.I.); (T.S.)
- Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan; (K.U.); (H.K.)
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan
| | - Hiroshi Kagami
- Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan; (K.U.); (H.K.)
| | - Tatsuya Morimoto
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (Y.S.); (T.M.)
| | - Tomohide Takaya
- Department of Agriculture, Graduate School of Science and Technology, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan; (M.I.); (T.S.)
- Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan; (K.U.); (H.K.)
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan
| |
Collapse
|
12
|
Yu YN, Ren YY, Shao ZL, Chen BL, Cui BY, Chao CY, Guo LJ, Guo S, Zhang MX, Wang SX, Zhu ML, Yin YL, Li P. Perillaldehyde improves diabetic cardiomyopathy by upregulating miR-133a-3p to regulate GSK-3β. Eur J Pharmacol 2023; 953:175836. [PMID: 37329971 DOI: 10.1016/j.ejphar.2023.175836] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/04/2023] [Accepted: 06/06/2023] [Indexed: 06/19/2023]
Abstract
Diabetic cardiomyopathy (DCM) is part of the most important causes of death from cardiovascular disease. Perillaldehyde (PAE), a major component of the herb perilla, has been shown to ameliorate doxorubicin-induced cardiotoxicity, but it is unclear whether PAE exerts beneficial effects on DCM. Exploring the potential molecular mechanisms of PAE for the treatment of DCM through network pharmacology and molecular docking. The SD rat type 1 diabetes model was established by a single intraperitoneal injection of streptozotocin (60 mg/kg), the cardiac function indexes of each group were detected by echocardiography; the morphological changes, apoptosis, protein expression of P-GSK-3β (S9), collagen I (Col-Ⅰ), collagen III (Col-Ⅲ) and alpha-smooth muscle actin (α-SMA), and miR-133a-3p expression levels were detected. An DCM model of H9c2 cells was established in vitro and transfected with Mimic and Inhibitor of miR-133a-3p. The results showed that PAE ameliorated cardiac dysfunction, reduced fasting glucose and cardiac weight index, and improved myocardial injury and apoptosis in DCM rats. It reduced high glucose-induced apoptosis, promoted migration and improved mitochondrial division injury in H9c2 cells. PAE decreased P-GSK-3β (S9), Col-Ⅰ, Col-Ⅲ and α-SMA protein expression and upregulated miR-133a-3p expression levels. After miR-133a-3p Inhibitor treatment, the expression of P-GSK-3β (S9) and α-SMA expression were significantly increased; after miR-133a-3p Mimic treatment, the expression of P-GSK-3β (S9) and α-SMA decreased significantly in H9c2 cells. It suggests that the mechanism of action of PAE to improve DCM may be related to the upregulation of miR-133a-3p and inhibition of P-GSK-3β expression.
Collapse
Affiliation(s)
- Ya-Nan Yu
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, Xinxiang, 453003, China
| | - Yuan-Yuan Ren
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, Xinxiang, 453003, China
| | - Zhen-Lei Shao
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, Xinxiang, 453003, China
| | - Bu-Lei Chen
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, Xinxiang, 453003, China
| | - Bao-Yue Cui
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, Xinxiang, 453003, China
| | | | - Li-Juan Guo
- Department of Oncology, Xinxiang Medical University First Affiliated Hospital, Xinxiang, 453119, China
| | - Shuang Guo
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, 437100, China
| | - Ming-Xiang Zhang
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, Xinxiang, 453003, China
| | - Shuang-Xi Wang
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, Xinxiang, 453003, China.
| | - Mo-Li Zhu
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, Xinxiang, 453003, China.
| | - Ya-Ling Yin
- School of Basic Medical Sciences, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and Pathophysiology, Xinxiang Medical University, Xinxiang, 453003, China.
| | - Peng Li
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
13
|
Jin B, Wang J, Chen Y, Zuo W, Hong B, Li J, Huang F, Zhang M, Wang Y. Focal adhesion kinase induces cardiac remodeling through NF-κB-mediated inflammatory responses in diabetic cardiomyopathy. Int Immunopharmacol 2023; 120:110280. [PMID: 37216798 DOI: 10.1016/j.intimp.2023.110280] [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: 09/06/2022] [Revised: 12/11/2022] [Accepted: 05/01/2023] [Indexed: 05/24/2023]
Abstract
BACKGROUND Hyperglycemia-induced chronic inflammation is a crucial risk factor that causes undesirable cardiac alternations in diabetic cardiomyopathy (DCM). Focal adhesion kinase (FAK) is a non-receptor protein tyrosine kinase that primarily regulates cell adhesion and migration. Based on recent studies, FAK is involved in inflammatory signaling pathway activation in cardiovascular diseases. Here, we evaluated the possibility of FAK as a therapeutic target for DCM. METHODS A small molecular selective FAKinhibitor, PND-1186 (PND), was used to evaluate the effect of FAK on DCM in both high glucose-stimulated cardiomyocytes and streptozotocin (STZ)-induced type 1 diabetes mellitus (T1DM) mice. RESULTS Increased FAK phosphorylation was found in the hearts of STZ-induced T1DM mice. PND treatment significantly decreased the expression of inflammatory cytokines and fibrogenic markers in cardiac specimens of diabetic mice. Notably, these reductions were correlated with improved cardiac systolic function. Furthermore, PND suppressed transforming growth factor-β-activated kinase 1 (TAK1) phosphorylation and NF-κB activation in the hearts of diabetic mice. Cardiomyocytes were identified as the main contributor to FAK-mediated cardiac inflammation and the involvement of FAK in cultured primary mouse cardiomyocytes and H9c2 cells was identified. Both FAK inhibition or FAK deficiency prevented hyperglycemia-induced inflammatory and fibrotic responses in cardiomyocytes owing to the inhibition of NF-κB. Herein, FAK activation was revealed to FAK directly binding to TAK1, leading to activation of TAK1 and downstream NF-κB signaling pathway. CONCLUSIONS FAK is a key regulator of diabetes-associated myocardial inflammatory injury by directly targeting to TAK1.
Collapse
Affiliation(s)
- Bo Jin
- Department of Gastroenterology, The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jiong Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yi Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wei Zuo
- Department of Gastroenterology, The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, China
| | - Bo Hong
- Department of Gastroenterology, The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, China
| | - Jie Li
- Department of Gastroenterology, The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, China
| | - Fang Huang
- Department of Gastroenterology, The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, China
| | - Mengpei Zhang
- Department of Gastroenterology, The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, China.
| | - Yi Wang
- Department of Gastroenterology, The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
14
|
Rami M, Rahdar S, Ahmadi Hekmatikar A, Awang Daud DM. Highlighting the novel effects of high-intensity interval training on some histopathological and molecular indices in the heart of type 2 diabetic rats. Front Endocrinol (Lausanne) 2023; 14:1175585. [PMID: 37274326 PMCID: PMC10235768 DOI: 10.3389/fendo.2023.1175585] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/04/2023] [Indexed: 06/06/2023] Open
Abstract
Background Type 2 diabetes is one of the most common metabolic diseases in recent years and has become an important risk factor for cardiovascular disorders. The first goal is to reduce type 2 diabetes, and in the case of cardiovascular disease, the second goal is to reduce and manage that disorder. Materials and methods The rats were divided into 4 groups: Healthy Control (n=8), Diabetes Control (n=8), Diabetes Training (n=8), and Healthy Training (n=8). The protocol consisted of 8 weeks of High-intensity interval (5 sessions per week), where the training started with 80% of the peak speed in the first week, and 10% was added to this speed every week. To measure the level of B-catenin, c-MYC, GSK3B, and Bcl-2 proteins using the western blot method, cardiac pathological changes were measured using hematoxylin and eosin staining, Masson's trichrome and PAS staining and apoptosis using the TUNEL method. Findings Histological results showed that diabetes causes significant pathological hypertrophy, fibrosis, and severe apoptosis in heart tissue. HIIT training significantly reduced pathological hypertrophy and fibrosis in heart tissue, and the rate of cardiomyocyte apoptosis was greatly reduced. This research showed that diabetes disorder increases the levels of B-catenin and c-Myc proteins and causes a decrease in the expression of GSK3B and Bcl-2 proteins. After eight weeks of HIIT training, the levels of B-catenin and c-Myc proteins decreased significantly, and the levels of GSK3B and Bcl-2 proteins increased. Conclusion This study showed that HIIT could be a suitable strategy to reduce cardiomyopathy in type 2 diabetic rats. However, it is suggested that in future studies, researchers should perform different intensities and exercises to promote exercise goals in type 2 diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Mohammad Rami
- Department of Sport Physiology, Faculty of Sport Sciences, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Samane Rahdar
- Department of Basic Sciences, Histology section, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Amirhoseein Ahmadi Hekmatikar
- Department of Physical Education and Sport Sciences, Faculty of Humanities, Tarbiat Modares University, Tehran, Iran
| | - D. Maryama Awang Daud
- Health Through Exercise and Active Living (HEAL) Research Unit, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University Malaysia Sabah, Kota Kinabalu, Sabah, Malaysia
| |
Collapse
|
15
|
Balatskyi VV, Sowka A, Dobrzyn P, Piven OO. WNT/β-catenin pathway is a key regulator of cardiac function and energetic metabolism. Acta Physiol (Oxf) 2023; 237:e13912. [PMID: 36599355 DOI: 10.1111/apha.13912] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 10/24/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023]
Abstract
The WNT/β-catenin pathway is a master regulator of cardiac development and growth, and its activity is low in healthy adult hearts. However, even this low activity is essential for maintaining normal heart function. Acute activation of the WNT/β-catenin signaling cascade is considered to be cardioprotective after infarction through the upregulation of prosurvival genes and reprogramming of metabolism. Chronically high WNT/β-catenin pathway activity causes profibrotic and hypertrophic effects in the adult heart. New data suggest more complex functions of β-catenin in metabolic maturation of the perinatal heart, establishing an adult pattern of glucose and fatty acid utilization. Additionally, low basal activity of the WNT/β-catenin cascade maintains oxidative metabolism in the adult heart, and this pathway is reactivated by physiological or pathological stimuli to meet the higher energy needs of the heart. This review summarizes the current state of knowledge of the organization of canonical WNT signaling and its function in cardiogenesis, heart maturation, adult heart function, and remodeling. We also discuss the role of the WNT/β-catenin pathway in cardiac glucose, lipid metabolism, and mitochondrial physiology.
Collapse
Affiliation(s)
- Volodymyr V Balatskyi
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Adrian Sowka
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Pawel Dobrzyn
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Oksana O Piven
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
- Department of Human Genetics, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| |
Collapse
|
16
|
Ni B, Sun M, Zhao J, Wang J, Cao Z. The role of β-catenin in cardiac diseases. Front Pharmacol 2023; 14:1157043. [PMID: 37033656 PMCID: PMC10073558 DOI: 10.3389/fphar.2023.1157043] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/13/2023] [Indexed: 04/11/2023] Open
Abstract
The Wnt/β-catenin signaling pathway is a classical Wnt pathway that regulates the stability and nuclear localization of β-catenin and plays an important role in adult heart development and cardiac tissue homeostasis. In recent years, an increasing number of researchers have implicated the dysregulation of this signaling pathway in a variety of cardiac diseases, such as myocardial infarction, arrhythmias, arrhythmogenic cardiomyopathy, diabetic cardiomyopathies, and myocardial hypertrophy. The morbidity and mortality of cardiac diseases are increasing, which brings great challenges to clinical treatment and seriously affects patient health. Thus, understanding the biological roles of the Wnt/β-catenin pathway in these diseases may be essential for cardiac disease treatment and diagnosis to improve patient quality of life. In this review, we summarize current research on the roles of β-catenin in human cardiac diseases and potential inhibitors of Wnt/β-catenin, which may provide new strategies for cardiac disease therapies.
Collapse
|
17
|
Wang X, He K, Ma L, Wu L, Yang Y, Li Y. Puerarin attenuates isoproterenol‑induced myocardial hypertrophy via inhibition of the Wnt/β‑catenin signaling pathway. Mol Med Rep 2022; 26:306. [PMID: 35946454 PMCID: PMC9437969 DOI: 10.3892/mmr.2022.12822] [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: 03/23/2022] [Accepted: 07/20/2022] [Indexed: 11/06/2022] Open
Abstract
Myocardial hypertrophy (MH) is an independent risk factor for cardiovascular disease, which in turn lead to arrhythmia or heart failure. Therefore, attention must be paid to formulation of therapeutic strategies for MH. Puerarin is a key bioactive ingredient isolated from Pueraria genera of plants that is beneficial for the treatment of MH. However, its molecular mechanism of action has not been fully determined. In the present study, 40 µM puerarin was demonstrated to be a safe dose for human AC16 cells using Cell Counting Kit‑8 assay. The protective effects of puerarin against MH were demonstrated in AC16 cells stimulated with isoproterenol (ISO). These effects were characterized by a significant decrease in surface area of cells (assessed using fluorescence staining) and mRNA and protein expression levels of MH‑associated biomarkers, including atrial and brain natriuretic peptide, assessed using reverse transcription‑quantitative PCR and western blotting, as well as β‑myosin heavy chain mRNA expression levels. Mechanistically, western blotting demonstrated that puerarin inhibited activation of the Wnt signaling pathway. Puerarin also significantly decreased phosphorylation of p65; this was mediated via crosstalk between the Wnt and NF‑κB signaling pathways. An inhibitor (Dickkopf‑1) and activator (IM‑12) of the Wnt signaling pathway were used to demonstrate that puerarin‑mediated effects alleviated ISO‑induced MH via the Wnt signaling pathway. The results of the present study demonstrated that puerarin pre‑treatment may be a potential therapeutic strategy for preventing ISO‑induced MH and managing MH in the future.
Collapse
Affiliation(s)
- Xiaoying Wang
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Kai He
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Linlin Ma
- College of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai 201318, P.R. China
| | - Lan Wu
- College of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai 201318, P.R. China
| | - Yan Yang
- Institute of Edible Fungi, Shanghai Academy of Agricultural Sciences, Shanghai 201106, P.R. China
| | - Yanfei Li
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| |
Collapse
|
18
|
Vallee A, Lecarpentier Y, Vallée JN. WNT/β-catenin pathway and circadian rhythms in obsessive-compulsive disorder. Neural Regen Res 2022; 17:2126-2130. [PMID: 35259818 PMCID: PMC9083179 DOI: 10.4103/1673-5374.332133] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The neuropsychiatric disease named obsessive-compulsive disorder is composed by obsessions and/or compulsions. Obsessive-compulsive disorder etiologies are undefined. However, numerous mechanisms in several localizations are implicated. Some studies showed that both glutamate, inflammatory factors and oxidative stress could have main functions in obsessive-compulsive disorder. Glycogen synthase kinase-3β, the major negative controller of the WNT/β-catenin pathway is upregulated in obsessive-compulsive disorder. In obsessive-compulsive disorder, some studies presented the actions of the different circadian clock genes. WNT/β-catenin pathway and circadian clock genes appear to be intricate. Thus, this review focuses on the interaction between circadian clock genes and the WNT/β-catenin pathway in obsessive-compulsive disorder.
Collapse
Affiliation(s)
- Alexandre Vallee
- Department of Clinical Research and Innovation (DRCI), Foch Hospital, Suresnes, France
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien (GHEF), Meaux, France
| | - Jean-Noël Vallée
- Laboratoire de Mathématiques et Applications (LMA), Université de Poitiers, Poitiers; Centre Hospitalier Universitaire (CHU) Amiens Picardie, Université Picardie Jules Verne (UPJV), Amiens, France
| |
Collapse
|
19
|
Zhang D, Li Y, Wang W, Lang X, Zhang Y, Zhao Q, Yan J, Zhang Y. NOX1 promotes myocardial fibrosis and cardiac dysfunction via activating the TLR2/NF-κB pathway in diabetic cardiomyopathy. Front Pharmacol 2022; 13:928762. [PMID: 36225554 PMCID: PMC9549956 DOI: 10.3389/fphar.2022.928762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is a prevalent complication in patients with diabetes, resulting in high morbidity and mortality. However, the molecular mechanisms of diabetic cardiomyopathy have yet to be fully elucidated. In this study, we investigated a novel target, NOX1, an isoform of superoxide-producing NADPH oxidase with key functional involvement in the pathophysiology of DCM. The DCM rat model was established by a high-fat diet combined with streptozotocin injections. DCM rats elicited myocardial fibrosis exacerbation, which was accompanied by a marked elevation of NOX1 expression in cardiac tissue. In particular, a specific NOX1 inhibitor, ML171, effectively decreased myocardial fibrosis and protected against cardiac dysfunction in DCM rats. Rat neonatal cardiac fibroblasts were incubated with high glucose (HG, 33 mM) as an in vitro model of DCM. We also observed that the expression of NOX1 was upregulated in HG-cultured cardiac fibroblasts. Silencing of NOX1 was found to attenuate myocardial fibrosis and oxidative stress in HG-induced cardiac fibroblasts. Furthermore, the upregulation of NOX1 by hyperglycemia induced activation of the TLR2/NF-κB pathway both in vitro and in vivo, whereas these effects were significantly attenuated with NOX1 gene silencing and further enhanced with NOX1 gene overexpression. In summary, we demonstrated that NOX1 induced activation of the TLR2/NF-κB pathway and increased reactive oxygen species production accumulation, which ultimately increased myocardial fibrosis and deteriorated cardiac function in diabetic cardiomyopathy. Our study revealed that NOX1 was a potential therapeutic target for DCM.
Collapse
Affiliation(s)
- Dandan Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
| | - Yilan Li
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
| | - Weijie Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
| | - Xueyan Lang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
| | - Yanxiu Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
| | - Qianqian Zhao
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
| | - Jingru Yan
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
| | - Yao Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
- *Correspondence: Yao Zhang,
| |
Collapse
|
20
|
Chen X, Li Y, Yuan X, Yuan W, Li C, Zeng Y, Lian Y, Qiu X, Qin Y, Zhang G, Liu X, Luo C, Luo JD, Hou N. Methazolamide Attenuates the Development of Diabetic Cardiomyopathy by Promoting β-Catenin Degradation in Type 1 Diabetic Mice. Diabetes 2022; 71:795-811. [PMID: 35043173 DOI: 10.2337/db21-0506] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 12/21/2021] [Indexed: 11/13/2022]
Abstract
Methazolamide (MTZ), a carbonic anhydrase inhibitor, has been shown to inhibit cardiomyocyte hypertrophy and exert a hypoglycemic effect in patients with type 2 diabetes and diabetic db/db mice. However, whether MTZ has a cardioprotective effect in the setting of diabetic cardiomyopathy is not clear. We investigated the effects of MTZ in a mouse model of streptozotocin-induced type 1 diabetes mellitus (T1DM). Diabetic mice received MTZ by intragastric gavage (10, 25, or 50 mg/kg, daily for 16 weeks). In the diabetic group, MTZ significantly reduced both random and fasting blood glucose levels and improved glucose tolerance in a dose-dependent manner. MTZ ameliorated T1DM-induced changes in cardiac morphology and dysfunction. Mechanistic analysis revealed that MTZ blunted T1DM-induced enhanced expression of β-catenin. Similar results were observed in neonatal rat cardiomyocytes (NRCMs) and adult mouse cardiomyocytes treated with high glucose or Wnt3a (a β-catenin activator). There was no significant change in β-catenin mRNA levels in cardiac tissues or NRCMs. MTZ-mediated β-catenin downregulation was recovered by MG132, a proteasome inhibitor. Immunoprecipitation and immunofluorescence analyses showed augmentation of AXIN1-β-catenin interaction by MTZ in T1DM hearts and in NRCMs treated with Wnt3a; thus, MTZ may potentiate AXIN1-β-catenin linkage to increase β-catenin degradation. Overall, MTZ may alleviate cardiac hypertrophy by mediating AXIN1-β-catenin interaction to promote degradation and inhibition of β-catenin activity. These findings may help inform novel therapeutic strategy to prevent heart failure in patients with diabetes.
Collapse
Affiliation(s)
- Xiaoqing Chen
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Yilang Li
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Xun Yuan
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Wenchang Yuan
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Conglin Li
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Yue Zeng
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Yuling Lian
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Xiaoxia Qiu
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
- Zhujiang Hospital of Southern Medical University, Guangzhou, People's Republic of China
| | - Yuan Qin
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Guiping Zhang
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Xiawen Liu
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Chengfeng Luo
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Jian-Dong Luo
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Ning Hou
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
21
|
Qin R, Zhao Q, Han B, Zhu HP, Peng C, Zhan G, Huang W. Indole-Based Small Molecules as Potential Therapeutic Agents for the Treatment of Fibrosis. Front Pharmacol 2022; 13:845892. [PMID: 35250597 PMCID: PMC8888875 DOI: 10.3389/fphar.2022.845892] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 01/19/2022] [Indexed: 12/17/2022] Open
Abstract
Indole alkaloids are widely distributed in nature and have been particularly studied because of their diverse biological activities, such as anti-inflammatory, anti-tumor, anti-bacterial, and anti-oxidant activities. Many kinds of indole alkaloids have been applied to clinical practice, proving that indole alkaloids are beneficial scaffolds and occupy a crucial position in the development of novel agents. Fibrosis is an end-stage pathological condition of most chronic inflammatory diseases and is characterized by excessive deposition of fibrous connective tissue components, ultimately resulting in organ dysfunction and even failure with significant morbidity and mortality. Indole alkaloids and indole derivatives can alleviate pulmonary, myocardial, renal, liver, and islet fibrosis through the suppression of inflammatory response, oxidative stress, TGF-β/Smad pathway, and other signaling pathways. Natural indole alkaloids, such as isorhynchophylline, evodiamine, conophylline, indirubin, rutaecarpine, yohimbine, and vincristine, are reportedly effective in organ fibrosis treatment. In brief, indole alkaloids with a wide range of pharmacological bioactivities are important candidate drugs for organ fibrosis treatment. The present review discusses the potential of natural indole alkaloids, semi-synthetic indole alkaloids, synthetic indole derivatives, and indole-contained metabolites in organ fibrosis treatment.
Collapse
Affiliation(s)
- Rui Qin
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qian Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hong-Ping Zhu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Antibiotics Research and Re-Evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Gu Zhan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Wei Huang, ; Gu Zhan,
| | - Wei Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Wei Huang, ; Gu Zhan,
| |
Collapse
|
22
|
Tang Z, Wang P, Dong C, Zhang J, Wang X, Pei H. Oxidative Stress Signaling Mediated Pathogenesis of Diabetic Cardiomyopathy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5913374. [PMID: 35103095 PMCID: PMC8800599 DOI: 10.1155/2022/5913374] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 01/03/2022] [Indexed: 02/07/2023]
Abstract
As a serious cardiovascular complication, diabetic cardiomyopathy (DCM) refers to diabetes-related changes in myocardial structure and function, which is obviously different from those cardiomyopathy secondary to hypertension, coronary heart disease, and valvular disease. The clinical features of DCM are left ventricular hypertrophy, myocardial fibrosis, and impaired diastolic function. DCM will lead to cardiac dysfunction, eventually progress to cardiac arrhythmia, heart failure, and sudden cardiac death. At present, the pathogenesis of DCM is complex and not fully elucidated, and oxidative stress (OS), inflammatory response, glucolipid metabolism disorder, etc., are considered as the potential pathophysiological mechanisms. As a consequence, there is no specific and effective treatment for DCM. OS refers to the imbalance between reactive oxygen species (ROS) accumulation and scavenging, oxidation, and antioxidants in vivo, which is widely studied in DCM. Numerous studies have pointed out that regulating the OS signaling pathways and reducing the generation and accumulation of ROS are potential directions for the treatment of DCM. This review summarizes the major OS signaling pathways that are related to the pathogenesis of DCM, providing ideas about further research and therapy.
Collapse
Affiliation(s)
- Zhaobing Tang
- Department of Rehabilitation Medicine, The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Peng Wang
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Chao Dong
- Department of Rehabilitation Medicine, The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Juan Zhang
- Emei Rehabilitation and Sanatorium Center of PLA, Leshan 614201, China
| | - Xiong Wang
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Haifeng Pei
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu 610083, China
| |
Collapse
|
23
|
Peng M, Liu H, Ji Q, Ma P, Niu Y, Ning S, Sun H, Pang X, Yang Y, Zhang Y, Han J, Hao G. Fufang Xueshuantong Improves Diabetic Cardiomyopathy by Regulating the Wnt/ β-Catenin Pathway. Int J Endocrinol 2022; 2022:3919161. [PMID: 36237833 PMCID: PMC9553353 DOI: 10.1155/2022/3919161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 07/02/2022] [Accepted: 08/12/2022] [Indexed: 11/30/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is one of the main complications of diabetic patients and the major reason for the high prevalence of heart failure in diabetic patients. Fufang Xueshuantong (FXST) is a traditional Chinese medicine formula commonly used in the treatment of diabetic retinopathy and stable angina pectoris. However, the role of FXST in DCM has not yet been clarified. This study was conducted to investigate the effects of FXST on diabetic myocardial lesions and reveal its molecular mechanism. The rats were intraperitoneally injected with 65 mg/kg streptozotocin (STZ) to induce diabetes mellitus (DM). DM rats were given saline or FXST. The rats in the control group were intraperitoneally injected with an equal amount of sodium citrate buffer and gavaged with saline. After 12 weeks, echocardiography, heart weight index (HWI), and myocardial pathological changes were determined. The expression of transforming growth factor-beta1 (TGF-β1), collagen I, and collagen III was examined using immunofluorescence staining and western blot. The expressions of Wnt/β-catenin signaling pathway-related proteins and mRNA were detected by western blot and real-time PCR. The results showed that FXST significantly improved cardiac function, ameliorated histopathological changes, and decreased HWI in the DM rats. FXST significantly inhibited the expression of myocardial TGF-β1, collagen I, and collagen III in DM rats. Furthermore, FXST significantly inhibited the Wnt/β-catenin pathway. Taken together, FXST has a protective effect on DCM, which might be mediated by suppressing the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Meizhong Peng
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Hanying Liu
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qingxuan Ji
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Pan Ma
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yiting Niu
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Shangqiu Ning
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Huihui Sun
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xinxin Pang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yuqian Yang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yuting Zhang
- Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jing Han
- Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Gaimei Hao
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- Gansu Provincial Hospital of Traditional Chinese Medicine, Gansu, China
| |
Collapse
|
24
|
Xu S, Xiaojing L, Xinyue S, Wei C, Honggui L, Shiwen X. Pig lung fibrosis is active in the subacute CdCl 2 exposure model and exerts cumulative toxicity through the M1/M2 imbalance. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 225:112757. [PMID: 34509164 DOI: 10.1016/j.ecoenv.2021.112757] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 08/30/2021] [Accepted: 09/03/2021] [Indexed: 06/13/2023]
Abstract
Environmental pollutant cadmium (Cd) can cause macrophage dysfunction, and the imbalance of M1/M2 is involved in the process of tissue fibrosis. In order to explore the effect of subacute CdCl2 exposure on pig lung tissue fibers and its mechanism, based on the establishment of this model, ICP-MS, H&E staining, Masson staining, Immunofluorescence, RT-PCR, and Western Blot methods were used to detect related indicators. The results found that lung tissue fibrosis, Cd content significantly increased, lung tissue ion disturbance, miR-20a-3p down-regulation, M1/M2 imbalance, LXA4/FPR2 content decreased, MDA content increased, NF-κB/NLRP3, TGFβ pathway, PPARγ/Wnt pathway activated, and the expression of fibrosis-related factors increased. The above results indicate that subacute CdCl2 exposure increase Cd content in the pig lungs, which leads to M1/M2 imbalance and down-regulates the content of LXA4/FPR2, further activates the oxidative stress/NF-κB/NLRP3 pathway, thereby activating the TGFβ and PPARγ/Wnt pathways to induce fibrosis. This study aims to reveal the toxic effects of CdCl2 and will provide new insights into the toxicology of Cd.
Collapse
Affiliation(s)
- Shi Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Li Xiaojing
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Sun Xinyue
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Cui Wei
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Liu Honggui
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China.
| | - Xu Shiwen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
25
|
Tuleta I, Frangogiannis NG. Fibrosis of the diabetic heart: Clinical significance, molecular mechanisms, and therapeutic opportunities. Adv Drug Deliv Rev 2021; 176:113904. [PMID: 34331987 PMCID: PMC8444077 DOI: 10.1016/j.addr.2021.113904] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/19/2021] [Accepted: 07/24/2021] [Indexed: 01/02/2023]
Abstract
In patients with diabetes, myocardial fibrosis may contribute to the pathogenesis of heart failure and arrhythmogenesis, increasing ventricular stiffness and delaying conduction. Diabetic myocardial fibrosis involves effects of hyperglycemia, lipotoxicity and insulin resistance on cardiac fibroblasts, directly resulting in increased matrix secretion, and activation of paracrine signaling in cardiomyocytes, immune and vascular cells, that release fibroblast-activating mediators. Neurohumoral pathways, cytokines, growth factors, oxidative stress, advanced glycation end-products (AGEs), and matricellular proteins have been implicated in diabetic fibrosis; however, the molecular links between the metabolic perturbations and activation of a fibrogenic program remain poorly understood. Although existing therapies using glucose- and lipid-lowering agents and neurohumoral inhibition may act in part by attenuating myocardial collagen deposition, specific therapies targeting the fibrotic response are lacking. This review manuscript discusses the clinical significance, molecular mechanisms and cell biology of diabetic cardiac fibrosis and proposes therapeutic targets that may attenuate the fibrotic response, preventing heart failure progression.
Collapse
Affiliation(s)
- Izabela Tuleta
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx NY, USA
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx NY, USA.
| |
Collapse
|
26
|
Hu J, Lu X, Zhang X, Shao X, Wang Y, Chen J, Zhao B, Li S, Xu C, Wei C. Exogenous spermine attenuates myocardial fibrosis in diabetic cardiomyopathy by inhibiting endoplasmic reticulum stress and the canonical Wnt signaling pathway. Cell Biol Int 2020; 44:1660-1670. [PMID: 32304136 DOI: 10.1002/cbin.11360] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 03/30/2020] [Accepted: 04/11/2020] [Indexed: 12/13/2022]
Abstract
Myocardial fibrosis is one of the main pathological manifestations of diabetic cardiomyopathy (DCM). Spermine (SPM), a product of polyamine metabolism, plays an important role in many cardiac diseases including hypertrophy, ischemia, and infarction, but its role in diabetic myocardial fibrosis has not been clarified. This study aimed to investigate the role of polyamine metabolism, specifically SPM, in diabetic myocardial fibrosis and to explore the related mechanisms. We used intraperitoneal injection of streptozotocin (STZ, 60 mg/kg) in Wistar rats and high glucose (HG, 40 mM) stimulated cardiac fibroblasts (CFs) to established a type 1 diabetes (T1D) model in vivo and in vitro, which were pretreated with exogenous SPM (5 mg/kg per day and 5 μM). The results showed that hyperglycemia induced the expression of the key polyamine synthesis enzyme ornithine decarboxylase (ODC) decreased and the key catabolic enzyme spermidine/spermine N1 -acetyltransferase (SSAT) increased compared with those in the control group. The body weight, blood insulin level, and cardiac ejection function were decreased, while blood glucose, heart weight, the ratio of heart weight to body weight, myocardial interstitial collagen deposition, and endoplasmic reticulum stress (ERS)-related protein (glucose-regulated protein-78, glucose-regulated protein-94, activating transcription factor-4, and C/EBP homology protein) expression in the T1D group were all significantly increased. HG also caused an increased expression of Wnt3, β-catenin (in cytoplasm and nucleus), while Axin2 and phosphorylated β-catenin decreased. Exogenous SPM improved the above changes caused by polyamine metabolic disorders. In conclusion, polyamine metabolism disorder occurs in the myocardial tissue of diabetic rats, causing myocardial fibrosis and ERS. Exogenous SPM plays a myocardial protective role via inhibiting of ERS and the canonical Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Jing Hu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Xiaoxiao Lu
- Department of Physical Diagnostics, First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Xinying Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Xiaoting Shao
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Yuehong Wang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Junting Chen
- Department of Anesthesiology, Harbin Medical University Fourth Hospital, Harbin, China
| | - Bingbing Zhao
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Siwei Li
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Changqing Xu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Can Wei
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| |
Collapse
|