1
|
Wu Y, Wang H, Xu H. Autophagy-lysosome pathway in insulin & glucagon homeostasis. Front Endocrinol (Lausanne) 2025; 16:1541794. [PMID: 39996055 PMCID: PMC11847700 DOI: 10.3389/fendo.2025.1541794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 01/22/2025] [Indexed: 02/26/2025] Open
Abstract
Lysosome, a highly dynamic organelle, is an important nutrient sensing center. They utilize different ion channels and transporters to complete the mission in degradation, trafficking, nutrient sensing and integration of various metabolic pathways to maintain cellular homeostasis. Glucose homeostasis relies on tightly regulated insulin secretion by pancreatic β cells, and their dysfunction is a hallmark of type 2 diabetes. Glucagon also plays an important role in hyperglycemia in diabetic patients. Currently, lysosome has been recognized as a nutrient hub to regulate the homeostasis of insulin and other hormones. In this review, we will discuss recent advances in understanding lysosome-mediated autophagy and lysosomal proteins involved in maintaining insulin and glucagon homeostasis, as well as their contributions to the etiology of diabetes.
Collapse
Affiliation(s)
- Yi Wu
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Shanghai Key Laboratory of Molecular Imaging, School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Hui Wang
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Huoyan Xu
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
| |
Collapse
|
2
|
Chiarelli R, Caradonna F, Naselli F. Autophagy and nutrigenomics: a winning team against chronic disease and tumors. Front Nutr 2024; 11:1409142. [PMID: 39703336 PMCID: PMC11655209 DOI: 10.3389/fnut.2024.1409142] [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: 03/29/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024] Open
Abstract
Autophagy, a vital cell process, has garnered attention for its role in various diseases and potential therapeutic interventions. Dysregulation of autophagy contributes to conditions such as metabolic diseases, neurodegenerative disorders, and cancer. In diseases such as diabetes, autophagy plays a crucial role in islet β-cell maintenance and glucose homeostasis, offering potential targets for therapeutic intervention. Nutrigenomics, which explores how dietary components interact with the genome, has emerged as a promising avenue for disease management. It sheds light on how diet influences gene expression and cellular processes, offering personalized approaches to disease prevention and management. Studies have showed the impact of specific dietary components, such as polyphenols and omega-3 fatty acids, on autophagy processes, suggesting their potential therapeutic benefits in neurodegenerative conditions and metabolic disorders. In cancer, autophagy's dual role in either suppressing tumorigenesis or promoting cancer cell survival underscores the importance of understanding its modulation through dietary interventions. Combined with conventional chemotherapy drugs, dietary compounds show synergistic effects in cancer treatment. Furthermore, phytochemicals such as indicaxanthin have been found to epigenetically regulate genes involved in autophagy, offering novel insights into personalized cancer therapies. This comprehensive review has the aim to study the autophagy in a combined view with nutrigenomics effects of some dietary molecules in maintaining cellular homeostasis and responding to pathological stimuli. Overall, the intersection of autophagy and nutrigenomics effect of bioactive compounds holds promise for developing targeted interventions for various diseases, emphasizing the significance of dietary interventions in disease prevention and management.
Collapse
Affiliation(s)
- Roberto Chiarelli
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy
| | - Fabio Caradonna
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy
- National Biodiversity Future Center (NBFC), Palermo, Italy
| | - Flores Naselli
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy
| |
Collapse
|
3
|
Rockhold JD, Marszalkowski H, Sannella M, Gibney K, Murphy L, Zukowski E, Kalantar GH, SantaCruz-Calvo S, Hart SN, Kuhn MK, Yu J, Stefanik O, Chase G, Proctor EA, Hasturk H, Nikolajczyk BS, Bharath LP. Everolimus alleviates CD4 + T cell inflammation by regulating autophagy and cellular redox homeostasis. GeroScience 2024; 46:5681-5699. [PMID: 38761287 PMCID: PMC11493941 DOI: 10.1007/s11357-024-01187-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 04/30/2024] [Indexed: 05/20/2024] Open
Abstract
Aging is associated with the onset and progression of multiple diseases, which limit health span. Chronic low-grade inflammation in the absence of overt infection is considered the simmering source that triggers age-associated diseases. Failure of many cellular processes during aging is mechanistically linked to inflammation; however, the overall decline in the cellular homeostasis mechanism of autophagy has emerged as one of the top and significant inducers of inflammation during aging, frequently known as inflammaging. Thus, physiological or pharmacological interventions aimed at improving autophagy are considered geroprotective. Rapamycin analogs (rapalogs) are known for their ability to inhibit mTOR and thus regulate autophagy. This study assessed the efficacy of everolimus, a rapalog, in regulating inflammatory cytokine production in T cells from older adults. CD4+ T cells from older adults were treated with a physiological dose of everolimus (0.01 µM), and indices of autophagy and inflammation were assessed to gain a mechanistic understanding of the effect of everolimus on inflammation. Everolimus (Ever) upregulated autophagy and broadly alleviated inflammatory cytokines produced by multiple T cell subsets. Everolimus's ability to alleviate the cytokines produced by Th17 subsets of T cells, such as IL-17A and IL-17F, was dependent on autophagy and antioxidant signaling pathways. Repurposing the antineoplastic drug everolimus for curbing inflammaging is promising, given the drug's ability to restore multiple cellular homeostasis mechanisms.
Collapse
Affiliation(s)
- Jack Donato Rockhold
- Department of Health Sciences and Nutrition, Merrimack College, North Andover, MA, USA
| | | | - Marco Sannella
- Department of Health Sciences and Nutrition, Merrimack College, North Andover, MA, USA
| | - Kaleigh Gibney
- Department of Health Sciences and Nutrition, Merrimack College, North Andover, MA, USA
| | - Lyanne Murphy
- Department of Biology, Merrimack College, North Andover, MA, USA
| | - Emelia Zukowski
- Department of Health Sciences and Nutrition, Merrimack College, North Andover, MA, USA
| | - Gabriella H Kalantar
- Dept of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY, USA
| | - Sara SantaCruz-Calvo
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
- Barnstable Brown Diabetes and Obesity Center, University of Kentucky, Lexington, KY, USA
| | - Samantha N Hart
- Barnstable Brown Diabetes and Obesity Center, University of Kentucky, Lexington, KY, USA
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA
| | - Madison K Kuhn
- Department of Neurosurgery, Pharmacology, and Biomedical Engineering and Center for Neural Engineering, Pennsylvania State University, Hershey, PA, USA
| | - Jingting Yu
- Razavi Newman Integrative Genomics and Bioinformatics Core, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Olivia Stefanik
- Department of Health Sciences and Nutrition, Merrimack College, North Andover, MA, USA
| | - Gabrielle Chase
- Department of Chemistry and Biochemistry, Merrimack College, North Andover, MA, USA
| | - Elizabeth A Proctor
- Department of Neurosurgery, Pharmacology, and Biomedical Engineering and Center for Neural Engineering, Pennsylvania State University, Hershey, PA, USA
- Department of Engineering Science & Mechanics, Pennsylvania State University, University Park, PA, USA
| | | | - Barbara S Nikolajczyk
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
- Barnstable Brown Diabetes and Obesity Center, University of Kentucky, Lexington, KY, USA
| | - Leena P Bharath
- Department of Health Sciences and Nutrition, Merrimack College, North Andover, MA, USA.
| |
Collapse
|
4
|
Zhang S, Gao Z, Feng L, Li M. Prevention and Treatment Strategies for Alzheimer's Disease: Focusing on Microglia and Astrocytes in Neuroinflammation. J Inflamm Res 2024; 17:7235-7259. [PMID: 39421566 PMCID: PMC11484773 DOI: 10.2147/jir.s483412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024] Open
Abstract
Alzheimer's disease (AD) is a fatal neurodegenerative disease characterized by its insidious onset and progressive development, making it the most common form of dementia. Despite its prevalence, the exact causes and mechanisms responsible for AD remain unclear. Recent studies have highlighted that inflammation in the central nervous system (CNS) plays a crucial role in both the initiation and progression of AD. Neuroinflammation, an immune response within the CNS triggered by glial cells in response to various stimuli, such as nerve injury, infection, toxins, or autoimmune reactions, has emerged as a significant factor alongside amyloid deposition and neurofibrillary tangles (NFTs) commonly associated with AD. This article aims to provide an overview of the most recent research regarding the involvement of neuroinflammation in AD, with a particular focus on elucidating the specific mechanisms involving microglia and astrocytes. By exploring these intricate processes, a new theoretical framework can be established to further probe the impact of neuroinflammation on the development and progression of AD. Through a deeper understanding of these underlying mechanisms, potential targets for therapeutic interventions and novel treatment strategies can be identified in the ongoing battle against AD.
Collapse
Affiliation(s)
- Shenghao Zhang
- Department of Neurology, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin Province, 130021, People’s Republic of China
| | - Zhejianyi Gao
- Department of Orthopaedics, Fushun Hospital of Chinese Medicine, Fushun, Liaoning Province, 113008, People’s Republic of China
| | - Lina Feng
- Department of Neurology, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin Province, 130021, People’s Republic of China
- Shandong Key Laboratory of TCM Multi-Targets Intervention and Disease Control, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, 271000, People’s Republic of China
| | - Mingquan Li
- Department of Neurology, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin Province, 130021, People’s Republic of China
| |
Collapse
|
5
|
Hao M, Sebag SC, Qian Q, Yang L. Lysosomal physiology and pancreatic lysosomal stress in diabetes mellitus. EGASTROENTEROLOGY 2024; 2:e100096. [PMID: 39512752 PMCID: PMC11542681 DOI: 10.1136/egastro-2024-100096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Endocrine and exocrine functions of the pancreas control nutritional absorption, utilisation and systemic metabolic homeostasis. Under basal conditions, the lysosome is pivotal in regulating intracellular organelles and metabolite turnover. In response to acute or chronic stress, the lysosome senses metabolic flux and inflammatory challenges, thereby initiating the adaptive programme to re-establish cellular homeostasis. A growing body of evidence has demonstrated the pathophysiological relevance of the lysosomal stress response in metabolic diseases in diverse sets of tissues/organs, such as the liver and the heart. In this review, we discuss the pathological relevance of pancreatic lysosome stress in diabetes mellitus. We begin by summarising lysosomal biology, followed by exploring the immune and metabolic functions of lysosomes and finally discussing the interplay between lysosomal stress and the pathogenesis of pancreatic diseases. Ultimately, our review aims to enhance our understanding of lysosomal stress in disease pathogenesis, which could potentially lead to the discovery of innovative treatment methods for these conditions.
Collapse
Affiliation(s)
- Meihua Hao
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Sara C Sebag
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Qingwen Qian
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Ling Yang
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| |
Collapse
|
6
|
Zhao J, Duan L, Li J, Yao C, Wang G, Mi J, Yu Y, Ding L, Zhao Y, Yan G, Li J, Zhao Z, Wang X, Li M. New insights into the interplay between autophagy, gut microbiota and insulin resistance in metabolic syndrome. Biomed Pharmacother 2024; 176:116807. [PMID: 38795644 DOI: 10.1016/j.biopha.2024.116807] [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: 03/12/2024] [Revised: 05/20/2024] [Accepted: 05/20/2024] [Indexed: 05/28/2024] Open
Abstract
Metabolic syndrome (MetS) is a widespread and multifactorial disorder, and the study of its pathogenesis and treatment remains challenging. Autophagy, an intracellular degradation system that maintains cellular renewal and homeostasis, is essential for maintaining antimicrobial defense, preserving epithelial barrier integrity, promoting mucosal immune response, maintaining intestinal homeostasis, and regulating gut microbiota and microbial metabolites. Dysfunctional autophagy is implicated in the pathological mechanisms of MetS, involving insulin resistance (IR), chronic inflammation, oxidative stress, and endoplasmic reticulum (ER) stress, with IR being a predominant feature. The study of autophagy represents a valuable field of research with significant clinical implications for identifying autophagy-related signals, pathways, mechanisms, and treatment options for MetS. Given the multifactorial etiology and various potential risk factors, it is imperative to explore the interplay between autophagy and gut microbiota in MetS more thoroughly. This will facilitate the elucidation of new mechanisms underlying the crosstalk among autophagy, gut microbiota, and MetS, thereby providing new insights into the diagnosis and treatment of MetS.
Collapse
Affiliation(s)
- Jinyue Zhao
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Liyun Duan
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Jiarui Li
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Chensi Yao
- Molecular Biology Laboratory, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Guoqiang Wang
- The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Jia Mi
- The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Yongjiang Yu
- The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Lu Ding
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Yunyun Zhao
- The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Guanchi Yan
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Jing Li
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Zhixuan Zhao
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Xiuge Wang
- The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China.
| | - Min Li
- Molecular Biology Laboratory, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
7
|
Friuli M, Sepe C, Panza E, Travelli C, Paterniti I, Romano A. Autophagy and inflammation an intricate affair in the management of obesity and metabolic disorders: evidence for novel pharmacological strategies? Front Pharmacol 2024; 15:1407336. [PMID: 38895630 PMCID: PMC11184060 DOI: 10.3389/fphar.2024.1407336] [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: 03/26/2024] [Accepted: 05/06/2024] [Indexed: 06/21/2024] Open
Abstract
Unhealthy lifestyle habits including a sedentary life, the lack of physical activity, and wrong dietary habits are the major ones responsible for the constant increase of obesity and metabolic disorders prevalence worldwide; therefore, the scientific community pays significant attention to the pharmacotherapy of such diseases, beyond lifestyle interventions, the use of medical devices, and surgical approaches. The intricate interplay between autophagy and inflammation appears crucial to orchestrate fundamental aspects of cellular and organismal responses to challenging stimuli, including metabolic insults; hence, when these two processes are dysregulated (enhanced or suppressed) they produce pathologic effects. The present review summarizes the existing literature reporting the intricate affair between autophagy and inflammation in the context of metabolic disorders, including obesity, diabetes, and liver metabolic diseases (non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH)). The evidence collected so far suggests that an alteration of autophagy might lead to maladaptive metabolic and inflammatory responses thus exacerbating the severity of the disease, and the most prominent conclusion underlies that autophagy might exert a protective function by contributing to balance inflammation. However, the complex nature of obesity and metabolic disorders might represent a limit of the studies; indeed, although many pharmacological treatments, producing positive metabolic effects, are also able to modulate autophagic flux and inflammation, it is not clear if the final beneficial effect might occur only by their mechanism of action, rather than because of additionally involved pathways. Finally, although future studies are needed, the observation that anti-obesity and antidiabetic drugs already on the market, including incretin mimetic agents, facilitate autophagy by dampening inflammation, strongly contributes to the idea that autophagy might represent a druggable system for the development of novel pharmacological tools that might represent an attractive strategy for the treatment of obesity and metabolic disorders.
Collapse
Affiliation(s)
- Marzia Friuli
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, Rome, Italy
| | - Christian Sepe
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, Rome, Italy
| | - Elisabetta Panza
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Cristina Travelli
- Department of Pharmaceutical Sciences, University of Pavia, Pavia, Italy
| | - Irene Paterniti
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Adele Romano
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
8
|
Zhou J, Wang J, Wang J, Li D, Hou J, Li J, Bai Y, Gao J. An inulin-type fructan CP-A from Codonopsis pilosula attenuates experimental colitis in mice by promoting autophagy-mediated inactivation of NLRP3 inflammasome. Chin J Nat Med 2024; 22:249-264. [PMID: 38553192 DOI: 10.1016/s1875-5364(24)60556-x] [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: 10/11/2023] [Indexed: 04/02/2024]
Abstract
Inulin-type fructan CP-A, a predominant polysaccharide in Codonopsis pilosula, demonstrates regulatory effects on immune activity and anti-inflammation. The efficacy of CP-A in treating ulcerative colitis (UC) is, however, not well-established. This study employed an in vitro lipopolysaccharide (LPS)-induced colonic epithelial cell model (NCM460) and an in vivo dextran sulfate sodium (DSS)-induced colitis mouse model to explore CP-A's protective effects against experimental colitis and its underlying mechanisms. We monitored the clinical symptoms in mice using various parameters: body weight, disease activity index (DAI), colon length, spleen weight, and histopathological scores. Additionally, molecular markers were assessed through enzyme-linked immunosorbent assay (ELISA), quantitative real-time polymerase chain reaction (qRT-PCR), immunofluorescence (IF), immunohistochemistry (IHC), and Western blotting assays. Results showed that CP-A significantly reduced reactive oxygen species (ROS), tumor necrosis factor-alpha (TNF-α), and interleukins (IL-6, IL-1β, IL-18) in LPS-induced cells while increasing IL-4 and IL-10 levels and enhancing the expression of Claudin-1, ZO-1, and occludin proteins in NCM460 cells. Correspondingly, in vivo findings revealed that CP-A administration markedly improved DAI, reduced colon shortening, and decreased the production of myeloperoxidase (MPO), malondialdehyde (MDA), ROS, IL-1β, IL-18, and NOD-like receptor protein 3 (NLRP3) inflammasome-associated genes/proteins in UC mice. CP-A treatment also elevated glutathione (GSH) and superoxide dismutase (SOD) levels, stimulated autophagy (LC3B, P62, Beclin-1, and ATG5), and reinforced Claudin-1 and ZO-1 expression, thereby aiding in intestinal epithelial barrier repair in colitis mice. Notably, the inhibition of autophagy via chloroquine (CQ) diminished CP-A's protective impact against colitis in vivo. These findings elucidate that CP-A's therapeutic effect on experimental colitis possibly involves mitigating intestinal inflammation through autophagy-mediated NLRP3 inflammasome inactivation. Consequently, inulin-type fructan CP-A emerges as a promising drug candidate for UC treatment.
Collapse
Affiliation(s)
- Jiangtao Zhou
- School of Pharmaceutical Science, Shanxi Medical University, Jinzhong 030600, China
| | - Jun Wang
- School of Pharmaceutical Science, Shanxi Medical University, Jinzhong 030600, China
| | - Jiajing Wang
- School of Pharmaceutical Science, Shanxi Medical University, Jinzhong 030600, China
| | - Deyun Li
- School of Pharmaceutical Science, Shanxi Medical University, Jinzhong 030600, China
| | - Jing Hou
- School of Pharmaceutical Science, Shanxi Medical University, Jinzhong 030600, China
| | - Jiankuan Li
- School of Pharmaceutical Science, Shanxi Medical University, Jinzhong 030600, China
| | - Yun'e Bai
- School of Pharmaceutical Science, Shanxi Medical University, Jinzhong 030600, China
| | - Jianping Gao
- School of Pharmaceutical Science, Shanxi Medical University, Jinzhong 030600, China.
| |
Collapse
|
9
|
Bi J, Zhang C, Lu C, Mo C, Zeng J, Yao M, Jia B, Liu Z, Yuan P, Xu S. Age-related bone diseases: Role of inflammaging. J Autoimmun 2024; 143:103169. [PMID: 38340675 DOI: 10.1016/j.jaut.2024.103169] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/03/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024]
Abstract
Bone aging is characterized by an imbalance in the physiological and pathological processes of osteogenesis, osteoclastogenesis, adipogenesis, and chondrogenesis, resulting in exacerbated bone loss and the development of age-related bone diseases, including osteoporosis, osteoarthritis, rheumatoid arthritis, and periodontitis. Inflammaging, a novel concept in the field of aging research, pertains to the persistent and gradual escalation of pro-inflammatory reactions during the aging process. This phenomenon is distinguished by its low intensity, systemic nature, absence of symptoms, and potential for management. The mechanisms by which inflammaging contribute to age-related chronic diseases, particularly in the context of age-related bone diseases, remain unclear. The precise manner in which systemic inflammation induces bone aging and consequently contributes to the development of age-related bone diseases has yet to be fully elucidated. This article primarily examines the mechanisms underlying inflammaging and its association with age-related bone diseases, to elucidate the potential mechanisms of inflammaging in age-related bone diseases and offer insights for developing preventive and therapeutic strategies for such conditions.
Collapse
Affiliation(s)
- Jiaming Bi
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Caimei Zhang
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Caihong Lu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Chuzi Mo
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiawei Zeng
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Mingyan Yao
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, China; Department of Endocrinology, Baoding No.1 Central Hospital, Baoding, China
| | - Bo Jia
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhongjun Liu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Peiyan Yuan
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| | - Shuaimei Xu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
10
|
Guo L, Yao Q, Lv J, Li Z, Wang LA, Zhang J. Anti-Hyperglycemic Effect of the Brown Slime Cap Mushroom Chroogomphus rutilus (Agaricomycetes) Crude Polysaccharide In Vitro and In Vivo. Int J Med Mushrooms 2024; 26:1-12. [PMID: 38801084 DOI: 10.1615/intjmedmushrooms.2024053173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
The prevalence of diabetes is increasing worldwide, and it is very important to study new hypoglycemic active substances. In this study, we investigated the hypoglycemic effect of Chroogomphus rutilus crude polysaccharide (CRCP) in HepG2 cells and streptozotocin-induced diabetic mice. A glucose consumption experiment conducted in HepG2 cells demonstrated the in vitro hypoglycemic activity of CRCP. Furthermore, CRCP exhibited significant hypoglycemic effects and effectively ameliorated insulin resistance in insulin resistant HepG2 cells. In high-fat diet and streptozotocin-induced diabetic mice, after 4 weeks of CRCP administration, fasting blood glucose, fasting serum insulin, triglyceride, total cholesterol, low-density lipoprotein cholesterol, glutamate transaminase, alanine transaminase, and insulin resistance index significantly decreased, while high-density lipoprotein cholesterol and insulin sensitivity index (ISI) were markedly increased. Moreover, hematoxylin-eosin (HE) staining and immunofluorescence labeling of tissue sections indicated that CRCP attenuated the pathological damage of liver and pancreas in diabetic mice. These results indicate that CRCP is a potential hypoglycemic agent.
Collapse
Affiliation(s)
- Lichao Guo
- College of Life Science, Hebei Normal University, Shijiazhuang, People's Republic of China
| | - Qingguo Yao
- College of Chemical Engineering, Shijiazhuang University, Shijiazhuang, People's Republic of China
| | - Jianhua Lv
- College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, People's Republic of China
| | - Zhuang Li
- College of Life Science, Hebei Normal University, Shijiazhuang, People's Republic of China
| | - Li-An Wang
- College of Life Science, Hebei Normal University, Shijiazhuang, 050024, P.R. China
| | - Jinxiu Zhang
- College of Life Science, Hebei Normal University, Shijiazhuang, 050024, P.R. China
| |
Collapse
|
11
|
Liu T, Wang Q, Du Z, Yin L, Li J, Meng X, Xue D. The trigger for pancreatic disease: NLRP3 inflammasome. Cell Death Discov 2023; 9:246. [PMID: 37452057 PMCID: PMC10349060 DOI: 10.1038/s41420-023-01550-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 06/23/2023] [Accepted: 07/05/2023] [Indexed: 07/18/2023] Open
Abstract
NLRP3 inflammasome is a multiprotein complex expressed in a variety of cells to stimulate the production of inflammatory factors. Activation of NLRP3 inflammasome depends on a complex regulatory mechanism, and its pro-inflammatory function plays an important role in pancreatic diseases. In this literature review, we summarize the activation mechanism of NLRP3 and analyze its role in each of the four typical pancreatic diseases. Through this article, we provide a relatively comprehensive summary to the researchers in this field, and provide some targeted therapy routes.
Collapse
Affiliation(s)
- Tianming Liu
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Qiang Wang
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Zhiwei Du
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Lu Yin
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Jiachen Li
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Xianzhi Meng
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
| | - Dongbo Xue
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
12
|
Wu SY, Wu HT, Wang YC, Chang CJ, Shan YS, Wu SR, Chiu YC, Hsu CL, Juan HF, Lan KY, Chu CW, Lee YR, Lan SH, Liu HS. Secretory autophagy promotes RAB37-mediated insulin secretion under glucose stimulation both in vitro and in vivo. Autophagy 2023; 19:1239-1257. [PMID: 36109708 PMCID: PMC10012902 DOI: 10.1080/15548627.2022.2123098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
High blood glucose is one of the risk factors for metabolic disease and INS (insulin) is the key regulatory hormone for glucose homeostasis. Hypoinsulinemia accompanied with hyperglycemia was diagnosed in mice with pancreatic β-cells exhibiting autophagy deficiency; however, the underlying mechanism remains elusive. The role of secretory autophagy in the regulation of metabolic syndrome is gaining more attention. Our data demonstrated that increased macroautophagic/autophagic activity leads to induction of insulin secretion in β-cells both in vivo and in vitro under high-glucose conditions. Moreover, proteomic analysis of purified autophagosomes from β-cells identified a group of vesicular transport proteins participating in insulin secretion, implying that secretory autophagy regulates insulin exocytosis. RAB37, a small GTPase, regulates vesicle biogenesis, trafficking, and cargo release. We demonstrated that the active form of RAB37 increased MAP1LC3/LC3 lipidation (LC3-II) and is essential for the promotion of insulin secretion by autophagy, but these phenomena were not observed in rab37 knockout (rab37-/-) cells and mice. Unbalanced insulin and glucose concentration in the blood was improved by manipulating autophagic activity using a novel autophagy inducer niclosamide (an antihelminthic drug) in a high-fat diet (HFD)-obesity mouse model. In summary, we reveal that secretory autophagy promotes RAB37-mediated insulin secretion to maintain the homeostasis of insulin and glucose both in vitro and in vivo.
Collapse
Affiliation(s)
- Shan-Ying Wu
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hung-Tsung Wu
- Department of Internal Medicine, School of Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Ching Wang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chih-Jen Chang
- Department of Family Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yan-Shen Shan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shang-Rung Wu
- Institute of Oral Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yen-Chi Chiu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chia-Lang Hsu
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei, Taiwan
| | - Hsueh-Fen Juan
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei, Taiwan
| | - Kai-Ying Lan
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chi-Wen Chu
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University
| | - Ying-Ray Lee
- Department of Microbiology and Immunology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Sheng-Hui Lan
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University.,Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsiao-Sheng Liu
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Center for Cancer Research, Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,M.Sc. Program in Tropical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
13
|
Mesbah Mohamed M, Ahmed Rashed L, Ahmed El-Boghdady N, Mohamed Said M. Bone Marrow-Derived Mesenchymal Stem Cells and Pioglitazone or Exendin-4 Synergistically Improve Insulin Resistance via Multiple Modulatory Mechanisms in High-Fat Diet/Streptozotocin-Induced Diabetes in Rats. Rep Biochem Mol Biol 2023; 12:42-58. [PMID: 37724145 PMCID: PMC10505456 DOI: 10.52547/rbmb.12.1.42] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/18/2023] [Indexed: 09/20/2023]
Abstract
Background Diabetes mellitus (DM) is a metabolic disease, characterized by hyperglycemia resulting from defects in insulin secretion and/or insulin action. The current study was designed to assess the therapeutic potential of bone marrow-derived mesenchymal stem cells (BM-MSCs) alone and in combination with pioglitazone (Pz) or exendin-4 (Ex) in high-fat diet/streptozotocin (HFD/STZ)-induced diabetes in rats. Methods The rats were subjected to the HFD for three weeks before being injected with a single low dosage of STZ (35 mg/kg bw). The animals were assigned to different treatment groups after type II diabetes mellitus (T2DM) induction was confirmed. Results Severe insulin resistance was verified in untreated HFD/STZ T2DM rats, along with the exaggeration of oxidative stress, inflammation, apoptosis, and autophagy suppression in the adipose tissues. Monotherapy of HFD/T2DM rats with BM-MSCs and Pz or Ex alleviated diabetic complications by increasing insulin sensitivity, decreasing apoptosis and inflammation as evidenced by a decrease in serum tumor necrosis factor-alpha, caspase-3, and nuclear factor-kappa B (NF-κB) genes expression and Janus kinase (JNK) protein expression, and enhancing autophagy as revealed by upregulation in beclin and LC3, as well as peroxisome proliferator-activated receptor-γ coactivator-1 alpha (PGC-1α) genes expression in the adipose tissues. An augmented ameliorative efficacy was recorded in combined treatments. The biochemical and molecular results were confirmed by histological investigation of pancreatic tissues. Conclusions Combining Pz or Ex with BM-MSCs is a synergistic therapeutic option that reduces insulin resistance and subsequent complications in T2DM via multiple molecular mechanisms.
Collapse
Affiliation(s)
| | - Laila Ahmed Rashed
- Department of Medical Biochemistry, Faculty of Medicine, Cairo University, Cairo, Egypt.
| | | | - Mahmoud Mohamed Said
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
14
|
Wang H, Banerjee N, Wang G, Firoze Khan M. Autophagy dysregulation in trichloroethene-mediated inflammation and autoimmune response. Toxicology 2023; 487:153468. [PMID: 36849104 PMCID: PMC9998359 DOI: 10.1016/j.tox.2023.153468] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 02/23/2023] [Accepted: 02/25/2023] [Indexed: 02/27/2023]
Abstract
Trichloroethene (TCE), an organic solvent extensively used for degreasing metals, can cause inflammatory autoimmune disorders [i.e., systemic lupus erythematosus (SLE) and autoimmune hepatitis] from both environmental and occupational exposure. Autophagy has emerged as a pivotal pathogenic factor in various autoimmune diseases. However, role of autophagy dysregulation in TCE-mediated autoimmunity is largely unknown. Here, we investigate whether autophagy dysregulation contributes to pathogenesis of TCE-mediated autoimmune responses. Using our established mouse model, we observed TCE-treated mice had elevated MDA-protein adducts, microtubule-associated protein light chain 3 conversion (LC3-II/LC3-I), beclin-1, phosphorylation of AMP-activated protein kinase (AMPK) and inhibition of mammalian target of rapamycin (mTOR) phosphorylation in the livers of MRL+ /+ mice. Suppression of oxidative stress with antioxidant N-acetylcysteine (NAC) effectively blocked TCE-mediated induction of autophagy markers. On the other hand, pharmacological autophagy induction with rapamycin significantly reduced TCE-mediated hepatic inflammation (NLRP3, ASC, Caspase1 and IL1-β mRNA levels), systemic cytokines (IL-12 and IL-17) and autoimmune responses (ANA and anti-dsDNA levels). Taken together, these results suggest that autophagy plays a protective role against TCE-mediated hepatic inflammation and autoimmunity in MRL+ /+ mice. These novel findings on the regulation of autophagy could help in designing therapeutic strategies for chemical exposure-mediated autoimmune responses.
Collapse
Affiliation(s)
- Hui Wang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Nivedita Banerjee
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Gangduo Wang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - M Firoze Khan
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States.
| |
Collapse
|
15
|
Hegdekar N, Sarkar C, Bustos S, Ritzel RM, Hanscom M, Ravishankar P, Philkana D, Wu J, Loane DJ, Lipinski MM. Inhibition of autophagy in microglia and macrophages exacerbates innate immune responses and worsens brain injury outcomes. Autophagy 2023:1-19. [PMID: 36652438 DOI: 10.1080/15548627.2023.2167689] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Excessive and prolonged neuroinflammation following traumatic brain injury (TBI) contributes to long-term tissue damage and poor functional outcomes. However, the mechanisms contributing to exacerbated inflammatory responses after brain injury remain poorly understood. Our previous work showed that macroautophagy/autophagy flux is inhibited in neurons following TBI in mice and contributes to neuronal cell death. In the present study, we demonstrate that autophagy is also inhibited in activated microglia and infiltrating macrophages, and that this potentiates injury-induced neuroinflammatory responses. Macrophage/microglia-specific knockout of the essential autophagy gene Becn1 led to overall increase in neuroinflammation after TBI. In particular, we observed excessive activation of the innate immune responses, including both the type-I interferon and inflammasome pathways. Defects in microglial and macrophage autophagy following injury were associated with decreased phagocytic clearance of danger/damage-associated molecular patterns (DAMP) responsible for activation of the cellular innate immune responses. Our data also demonstrated a role for precision autophagy in targeting and degradation of innate immune pathways components, such as the NLRP3 inflammasome. Finally, inhibition of microglial/macrophage autophagy led to increased neurodegeneration and worse long-term cognitive outcomes after TBI. Conversely, increasing autophagy by treatment with rapamycin decreased inflammation and improved outcomes in wild-type mice after TBI. Overall, our work demonstrates that inhibition of autophagy in microglia and infiltrating macrophages contributes to excessive neuroinflammation following brain injury and in the long term may prevent resolution of inflammation and tissue regeneration.Abbreviations: Becn1/BECN1, beclin 1, autophagy related; CCI, controlled cortical impact; Cybb/CYBB/NOX2: cytochrome b-245, beta polypeptide; DAMP, danger/damage-associated molecular patterns; Il1b/IL1B/Il-1β, interleukin 1 beta; LAP, LC3-associated phagocytosis; Map1lc3b/MAP1LC3/LC3, microtubule-associated protein 1 light chain 3 beta; Mefv/MEFV/TRIM20: Mediterranean fever; Nos2/NOS2/iNOS: nitric oxide synthase 2, inducible; Nlrp3/NLRP3, NLR family, pyrin domain containing 3; Sqstm1/SQSTM1/p62, sequestosome 1; TBI, traumatic brain injury; Tnf/TNF/TNF-α, tumor necrosis factor; Ulk1/ULK1, unc-51 like kinase 1.
Collapse
Affiliation(s)
- Nivedita Hegdekar
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Chinmoy Sarkar
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sabrina Bustos
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rodney M Ritzel
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Neurology, McGovern Medical School, University of Texas, Houston, Tx, USA
| | - Marie Hanscom
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Prarthana Ravishankar
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Deepika Philkana
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Junfang Wu
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - David J Loane
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, USA.,School of Biochemistry and Immunology, Trinity College, Dublin, Ireland
| | - Marta M Lipinski
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
16
|
Sousa ESA, Queiroz LAD, Guimarães JPT, Pantoja KC, Barros RS, Epiphanio S, Martins JO. The influence of high glucose conditions on macrophages and its effect on the autophagy pathway. Front Immunol 2023; 14:1130662. [PMID: 37122742 PMCID: PMC10130370 DOI: 10.3389/fimmu.2023.1130662] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 03/13/2023] [Indexed: 05/02/2023] Open
Abstract
Introduction Macrophages are central cells in mediating the inflammatory response. Objective and Methods We evaluated the effect of high glucose conditions on the inflammatory profile and the autophagy pathway in Bone-Marrow Derived Macrophages (BMDM) from diabetic (D-BMDM) (alloxan: 60mg/kg, i.v.) and non-diabetic (ND-BMDM) C57BL/6 mice. BMDM were cultured in medium with normal glucose (5.5 mM), or high glucose (25 mM) concentration and were primed with Nigericin (20µM) stimulated with LPS (100 ng/mL) at times of 30 minutes; 2; 4; 6 and 24 hours, with the measurement of IL-6, IL-1β and TNF-α cytokines. Results We have further identified changes in the secretion of pro-inflammatory cytokines IL-6, IL-1β and TNF-α, where BMDM showed increased secretion of these cytokines after LPS + Nigericin stimulation. In addition, changes were observed in the autophagy pathway, where the increase in the autophagic protein LC3b and Beclin-1 occurred by macrophages of non-diabetic animals in hyperglycemic medium, without LPS stimulation. D-BMDM showed a reduction on the expression of LC3b and Beclin-1, suggesting an impaired autophagic process in these cells. Conclusion The results suggest that hyperglycemia alters the inflammatory pathways in macrophages stimulated by LPS, playing an important role in the inflammatory response of diabetic individuals.
Collapse
Affiliation(s)
- Emanuella S. A. Sousa
- Laboratory of Immunoendocrinology, School of Pharmaceutical Sciences, Department of Clinical and Toxicological Analyses, University of São Paulo, São Paulo, Brazil
| | - Luiz A. D. Queiroz
- Laboratory of Immunoendocrinology, School of Pharmaceutical Sciences, Department of Clinical and Toxicological Analyses, University of São Paulo, São Paulo, Brazil
| | - João P. T. Guimarães
- Laboratory of Immunoendocrinology, School of Pharmaceutical Sciences, Department of Clinical and Toxicological Analyses, University of São Paulo, São Paulo, Brazil
| | - Kamilla C. Pantoja
- Laboratory of Immunoendocrinology, School of Pharmaceutical Sciences, Department of Clinical and Toxicological Analyses, University of São Paulo, São Paulo, Brazil
| | - Rafael S. Barros
- Laboratory of Immunoendocrinology, School of Pharmaceutical Sciences, Department of Clinical and Toxicological Analyses, University of São Paulo, São Paulo, Brazil
| | - Sabrina Epiphanio
- Laboratory of Malaria Cellular and Molecular Immunopathology, School of Pharmaceutical Sciences, Department of Clinical and Toxicological Analyses, University of São Paulo, São Paulo, Brazil
| | - Joilson O. Martins
- Laboratory of Immunoendocrinology, School of Pharmaceutical Sciences, Department of Clinical and Toxicological Analyses, University of São Paulo, São Paulo, Brazil
- *Correspondence: Joilson O. Martins,
| |
Collapse
|
17
|
Orphan GPR26 Counteracts Early Phases of Hyperglycemia-Mediated Monocyte Activation and Is Suppressed in Diabetic Patients. Biomedicines 2022; 10:biomedicines10071736. [PMID: 35885041 PMCID: PMC9312814 DOI: 10.3390/biomedicines10071736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/07/2022] [Accepted: 07/13/2022] [Indexed: 11/17/2022] Open
Abstract
Diabetes is the ninth leading cause of death, with an estimated 1.5 million deaths worldwide. Type 2 diabetes (T2D) results from the body’s ineffective use of insulin and is largely the result of excess body weight and physical inactivity. T2D increases the risk of cardiovascular diseases, retinopathy, and kidney failure by two-to three-fold. Hyperglycemia, as a hallmark of diabetes, acts as a potent stimulator of inflammatory condition by activating endothelial cells and by dysregulating monocyte activation. G-protein couple receptors (GPCRs) can both exacerbate and promote inflammatory resolution. Genome-wide association studies (GWAS) indicate that GPCRs are differentially regulated in inflammatory and vessel cells from diabetic patients. However, most of these GPCRs are orphan receptors, for which the mechanism of action in diabetes is unknown. Our data indicated that orphan GPCR26 is downregulated in the PBMC isolated from T2D patients. In contrast, GPR26 was initially upregulated in human monocytes and PBMC treated with high glucose (HG) levels and then decreased upon chronic and prolonged HG exposure. GPR26 levels were decreased in T2D patients treated with insulin compared to non-insulin treated patients. Moreover, GPR26 inversely correlated with the BMI and the HbA1c of diabetic compared to non-diabetic patients. Knockdown of GPR26 enhanced monocyte ROS production, MAPK signaling, pro-inflammatory activation, monocyte adhesion to ECs, and enhanced the activity of Caspase 3, a pro-apoptotic molecule. The same mechanisms were activated by HG and exacerbated when GPR26 was knocked down. Hence, our data indicated that GPR26 is initially activated to protect monocytes from HG and is inhibited under chronic hyperglycemic conditions.
Collapse
|
18
|
Zhou J, Wang F, Jia L, Chai R, Wang H, Wang X, Li J, Wang K, Zhang P, Yang H. 2,4-dichlorophenoxyacetic acid induces ROS activation in NLRP3 inflammatory body-induced autophagy disorder in microglia and the protective effect of Lycium barbarum polysaccharide. ENVIRONMENTAL TOXICOLOGY 2022; 37:1136-1151. [PMID: 35099110 DOI: 10.1002/tox.23471] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 12/26/2021] [Accepted: 01/16/2022] [Indexed: 06/14/2023]
Abstract
The pesticide 2,4-dichlorophenoxyacetic acid (2,4-D) exerts neurotoxic effects; however, its action mechanism remains unclear. Here, we used BV2 cells as a model and divided them into six groups: control group (serum-free medium), lipopolysaccharide (LPS) (1 μg/mL), 2,4-D (1.2 μmol/mL), Lycium barbarum polysaccharide (LBP; 300 μg/mL LBP), LPS (1 μg/mL) + LBP (300 μg/mL), and 2,4-D (1.2 μmol/mL) + LBP (300 μg/mL) with dimethyl sulfoxide as the solvent. Our results showed that 2,4-D treatment decreased superoxide dismutase and glutathione peroxidase activities and increased malondialdehyde content. The percentage of microglial activation (co-expression of ionized calcium-binding adaptor protein-1 + CD68) in the LPS and 2,4-D groups and the levels of tumor necrosis factor alpha, interleukin (IL) 1 beta, IL-6, and IL-18 in the cell supernatant were increased. The protein and mRNA levels of Nod-like receptor protein 3 (NLRP3), apoptosis-associated speck-like protein, caspase-1, IL-1β, IL-18, and p62 increased, whereas those of LC3II/I and Beclin-1 decreased in the 2,4-D group. The protein expression and mRNA levels of NLRP3, cleaved caspase-1, IL-1β, IL-18, and p62 decreased significantly, whereas the protein expression and mRNA levels of LC3II/I and Beclin-1 increased in small interfering RNA of NLRP3-treated BV2 cells stimulated with 2,4-D and LPS. In conclusion, 2,4-D enhanced cell migration, promoted oxidative stress, induced excessive release of mitochondrial reactive oxygen species, promoted microglial cell activation, released inflammatory factors, activated NLRP3 inflammasomes, and inhibited autophagy. Meanwhile, LBP reduced inflammation and the release of mitochondrial reactive oxygen species, inhibited NLRP3 inflammasome activation, and regulated autophagy, thereby playing a neuroprotective role.
Collapse
Affiliation(s)
- Jian Zhou
- Department of Occupational and Environmental Health, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
- Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Faxuan Wang
- Department of Occupational and Environmental Health, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
- Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Leina Jia
- Department of Occupational and Environmental Health, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
- Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Ru Chai
- Department of Occupational and Environmental Health, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
- Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Hengquan Wang
- Department of Occupational and Environmental Health, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
- Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Xiaolan Wang
- Department of Occupational and Environmental Health, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
- Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Jiangping Li
- Department of Occupational and Environmental Health, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
- Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Kai Wang
- Department of Occupational and Environmental Health, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
- Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Pengju Zhang
- Department of Occupational and Environmental Health, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
- Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Huifang Yang
- Department of Occupational and Environmental Health, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
- Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| |
Collapse
|
19
|
Zhang Y, Aisker G, Dong H, Halemahebai G, Zhang Y, Tian L. Urolithin A suppresses glucolipotoxicity-induced ER stress and TXNIP/NLRP3/IL-1β inflammation signal in pancreatic β cells by regulating AMPK and autophagy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 93:153741. [PMID: 34656886 DOI: 10.1016/j.phymed.2021.153741] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 08/28/2021] [Accepted: 09/05/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Pancreatic inflammation plays a key role in diabetes pathogenesis and progression. Urolithin A (UA), an intestinal flora metabolite of pomegranate, has anti-diabetic, anti-inflammatory and kidney protection effects among others. However, its effects on pancreatic inflammation and the potential mechanisms have not been clearly established. PURPOSE This study aimed at investigating the molecular mechanisms of UA anti-pancreatic inflammation under a diabetic environment. METHODS Diabetes induction in male C57BL/6 mice was achieved by a high fat diet and intraperitoneal streptozotocin injections. Then, diabetic mice were orally administered with UA for 8 weeks. In vitro, endoplasmic reticulum stress and MIN6 pancreatic β cell inflammation were induced using 25 mM glucose and 0.5 mM palmitic acid. The effects of UA were evaluated by immunohistochemistry, Western blot, and enzyme linked immunosorbent assays. Finally, the underlying mechanisms were elucidated using an autophagy inhibitor (chloroquine, CQ) and an AMPK inhibitor (dorsomorphin dihydrochloride). RESULTS UA significantly inhibited IL-1β secretion and TXNIP/NLRP3 expression in the pancreas of diabetic mice and in MIN6 pancreatic cells. UA downregulated the ER stress protein, p-PERK, and promoted AMPK phosphorylation. UA activated autophagy to inhibit TXNIP/NLRP3 IL-1β inflammatory signal, an effect that was reversed by CQ. Dorsomorphin 2HCL, reversed the autophagy-activation and anti-inflammatory effects of UA. Verapamil, clinically applied as an antiarrhythmic drug, is a TXNIP inhibitor for prevention of beta cell loss and diabetes development, but limited by its cardiac toxicity. In this study, verapamil (as positive control) inhibited NLRP3 /IL-1β signaling in MIN6 cells. Inhibitory effects of UA on TXNIP and IL-1β were weaker than those of verapamil (both at 50 μM, p < 0.05, p < 0.01). Conversely, inhibitory effects of UA on p62 were stronger, relative to those of verapamil (p < 0.05), and there were no differences in AMPK activation and LC3 enhancement effects between UA and verapamil. CONCLUSION UA is a potential anti-pancreatic inflammation agent that activates AMPK and autophagy to inhibit endoplasmic reticulum stress associated TXNIP/NLRP3/IL-1β signal pathway.
Collapse
Affiliation(s)
- YanZhi Zhang
- Department of Pharmacology, College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China.
| | - Gulimila Aisker
- Department of Pharmacology, College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | - Huaiyang Dong
- Department of Pharmacology, College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | - Gulihaixia Halemahebai
- Department of Pharmacology, College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | - Yan Zhang
- Department of Pediatrics, Xinjiang Military General Hospital, Urumqi, China
| | - Linai Tian
- Third Clinical College, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
20
|
Wu LX, Xu YC, Hogstrand C, Zhao T, Wu K, Xu YH, Liu W, Luo Z. Lipophagy mediated glucose-induced changes of lipid deposition and metabolism via ROS dependent AKT-Beclin1 activation. J Nutr Biochem 2021; 100:108882. [PMID: 34655756 DOI: 10.1016/j.jnutbio.2021.108882] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 07/29/2021] [Accepted: 09/20/2021] [Indexed: 12/12/2022]
Abstract
High dietary carbohydrate intake leads to lipid accumulation in the intestinal tract, but the molecular mechanism remains unknown. In the present study, using yellow catfish (Pelteobagrus fulvidraco) as a model, we found that (1) high carbohydrate diets (HCD) and high glucose (HG) increased lipid deposition, up-regulated lipogenesis and fatty acid β-oxidation, activated autophagy and induced oxidative stress in the intestinal tissues and intestinal epithelial cells (IECs); (2) lipophagy alleviated HG-induced lipid accumulation via the up-regulation of fatty acid β-oxidation; (3) Akt interacted directly with Beclin1; (4) HG suppressed Akt1 phosphorylation, downregulated Akt1-mediated phosphorylation of Beclin1, activated lipophagy and alleviated the increment of TG deposition induced by HG with S87 and S292 being the key phosphorylation residues of Beclin1 in response to HG; (5) ROS generation mediated HG-induced activation of lipophagy and HG-induced suppression of AKT phosphorylation, activated AMPK and alleviated HG-induced increase of TG deposition. Our study provides mechanistic evidence that high carbohydrate- and glucose-induced lipophagy in intestine and IECs is associated with ROS-AKT-Beclin1-dependent activation of autophagy, which alleviates glucose-induced lipid accumulation. Our findings are important since the regulation of autophagy can be used as potential molecular targets for the prevention and treatment of lipotoxicity in the intestine of vertebrates, including humans.
Collapse
Affiliation(s)
- Li-Xiang Wu
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan, China
| | - Yi-Chuang Xu
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan, China
| | - Christer Hogstrand
- Diabetes and Nutritional Sciences Division, School of Medicine, King's College London, London, United Kingdom
| | - Tao Zhao
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan, China
| | - Kun Wu
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan, China
| | - Yi-Huan Xu
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan, China
| | - Wei Liu
- Laboratory of Fish Nutrition, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, China
| | - Zhi Luo
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan, China.
| |
Collapse
|
21
|
Yao D, GangYi Y, QiNan W. Autophagic dysfunction of β cell dysfunction in type 2 diabetes, a double-edged sword. Genes Dis 2021; 8:438-447. [PMID: 34179308 PMCID: PMC8209341 DOI: 10.1016/j.gendis.2020.03.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 03/07/2020] [Accepted: 03/12/2020] [Indexed: 12/19/2022] Open
Abstract
Diabetes is an age-related disease, most of which is type 2 diabetes, and islet β cell dysfunction and insulin resistance are the main mechanisms of type 2 diabetes. Recent studies have revealed that autophagy plays an important role in maintaining the structure and function of islet beta cells and inhibiting insulin resistance and apoptosis induced by oxidative stress. In this review, we discussed the positive and negative effects of autophagy and its dysfunction on type 2 diabetes mellitus, which is the so-called double-edged sword, analysed its possible mechanism, and identified possible research hot spots.
Collapse
Affiliation(s)
- Ding Yao
- Endocrinology and Nephrology Department, Chongqing University Cancer Hospital and Chongqing Cancer Institute and Chongqing Cancer Hospital, Chongqing, 400030, PR China
| | - Yang GangYi
- Endocrinology Department, The Second Affiliated Hospital of the Chongqing Medical University, Chongqing, 400010, PR China
| | - Wu QiNan
- Endocrinology and Nephrology Department, Chongqing University Cancer Hospital and Chongqing Cancer Institute and Chongqing Cancer Hospital, Chongqing, 400030, PR China
| |
Collapse
|
22
|
Li X, Wan T, Li Y. Role of FoxO1 in regulating autophagy in type 2 diabetes mellitus (Review). Exp Ther Med 2021; 22:707. [PMID: 34007316 PMCID: PMC8120662 DOI: 10.3892/etm.2021.10139] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 02/09/2021] [Indexed: 12/11/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a major chronic disease that is characterized by pancreatic β-cell dysfunction and insulin resistance. Autophagy is a highly conserved intracellular recycling pathway and is involved in regulating intracellular homeostasis. Transcription factor Forkhead box O1 (FoxO1) also regulates fundamental cellular processes, including cell differentiation, metabolism and apoptosis, and proliferation to cellular stress. Increasing evidence suggest that autophagy and FoxO1 are involved in the pathogenesis of T2DM, including β-cell viability, apoptosis, insulin secretion and peripheral insulin resistance. Recent studies have demonstrated that FoxO1 improves insulin resistance by regulating target tissue autophagy. The present review summarizes current literature on the role of autophagy and FoxO1 in T2DM. The participation of FoxO1 in the development and occurrence of T2DM via autophagy is also discussed.
Collapse
Affiliation(s)
- Xiudan Li
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Tingting Wan
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yanbo Li
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
23
|
Geng K, Ma X, Jiang Z, Huang W, Gao C, Pu Y, Luo L, Xu Y, Xu Y. Innate Immunity in Diabetic Wound Healing: Focus on the Mastermind Hidden in Chronic Inflammatory. Front Pharmacol 2021; 12:653940. [PMID: 33967796 PMCID: PMC8097165 DOI: 10.3389/fphar.2021.653940] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 02/24/2021] [Indexed: 02/06/2023] Open
Abstract
A growing body of evidence suggests that the interaction between immune and metabolic responses is essential for maintaining tissue and organ homeostasis. These interacting disorders contribute to the development of chronic diseases associated with immune-aging such as diabetes, obesity, atherosclerosis, and nonalcoholic fatty liver disease. In Diabetic wound (DW), innate immune cells respond to the Pathogen-associated molecular patterns (PAMAs) and/or Damage-associated molecular patterns (DAMPs), changes from resting to an active phenotype, and play an important role in the triggering and maintenance of inflammation. Furthermore, the abnormal activation of innate immune pathways secondary to immune-aging also plays a key role in DW healing. Here, we review studies of innate immune cellular molecular events that identify metabolic disorders in the local microenvironment of DW and provide a historical perspective. At the same time, we describe some of the recent progress, such as TLR receptor-mediated intracellular signaling pathways that lead to the activation of NF-κB and the production of various pro-inflammatory mediators, NLRP3 inflammatory via pyroptosis, induction of IL-1β and IL-18, cGAS-STING responds to mitochondrial injury and endoplasmic reticulum stress, links sensing of metabolic stress to activation of pro-inflammatory cascades. Besides, JAK-STAT is also involved in DW healing by mediating the action of various innate immune effectors. Finally, we discuss the great potential of targeting these innate immune pathways and reprogramming innate immune cell phenotypes in DW therapy.
Collapse
Affiliation(s)
- Kang Geng
- Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, China.,State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Avenida Wai Long, Taipa, China.,Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, China.,Department of Plastic and Burn Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,National Key Clinical Construction Specialty, Luzhou, China
| | - Xiumei Ma
- Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, China.,State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Avenida Wai Long, Taipa, China.,Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, China
| | - Zongzhe Jiang
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, China
| | - Wei Huang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, China
| | - Chenlin Gao
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, China
| | - Yueli Pu
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, China
| | - Lifang Luo
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, China
| | - Youhua Xu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, China.,State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Avenida Wai Long, Taipa, China
| | - Yong Xu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, China.,State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Avenida Wai Long, Taipa, China.,Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, China.,Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
24
|
Arruri VK, Gundu C, Kalvala AK, Sherkhane B, Khatri DK, Singh SB. Carvacrol abates NLRP3 inflammasome activation by augmenting Keap1/Nrf-2/p62 directed autophagy and mitochondrial quality control in neuropathic pain. Nutr Neurosci 2021; 25:1731-1746. [PMID: 33641628 DOI: 10.1080/1028415x.2021.1892985] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Objectives: We aimed to evaluate the effect of carvacrol (CRC), a phenolic monoterpene with high nutritional value on NLRP3 activation against chronic constriction injury (CCI) of sciatic nerve induced neuropathic pain (NP) in rats and in lipopolysacharide (LPS) induced neuroinflammation in neuro2a (N2A) cells. Methods: NP was induced in male SD rats by performing CCI and CRC (30 and 60 mg/kg, p.o) was administered for 14 days. Behavioural and functional parameters were evaluated using standard procedures. Various molecular experimentations were conducted to evaluate the efficacy of CRC against CCI induced neuropathy and in LPS (1 μg/ml) primed and ATP (5 μM) treated N2A cells.Results: CCI resulted in marked development of hyperalgesia and allodynia. Further, CCI rats, LPS and ATP treated N2A cells showed enhanced expression of NLRP3, ASC, Caspase-1 and IL-1β. In addition, CCI rats exhibited diminished levels of Nrf-2 with an increase in Keap1 expression. Also, CCI animals manifested with compromised mitochondrial function along with decreased autophagy markers and enhanced p62 levels when compared to sham rats. However, CRC administration significantly ameliorated these changes suggesting NLRP3 inhibition by CRC may be attributed to activation of autophagy via Keap1/Nrf-2/p62 forward feedback loop and augmentation of mitochondrial quality control. Intriguingly, pretreatment of CRC (50 and 100 μM) to LPS and ATP treated N2A cells resulted in decreased colocalization of NLRP3 and ASC.Discussion: These findings revealed the neuroprotective potential of CRC against CCI induced NP and delineate the critical role of autophagy and mitochondrial quality control in NLRP3 regulation.
Collapse
Affiliation(s)
- Vijay Kumar Arruri
- Neuropharmacology Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Balanagar, India
| | - Chayanika Gundu
- Neuropharmacology Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Balanagar, India
| | - Anil Kumar Kalvala
- Neuropharmacology Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Balanagar, India
| | - Bhoomika Sherkhane
- Neuropharmacology Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Balanagar, India
| | - Dharmendra Kumar Khatri
- Neuropharmacology Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Balanagar, India
| | - Shashi Bala Singh
- Neuropharmacology Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Balanagar, India
| |
Collapse
|
25
|
Gonnella R, Gilardini Montani MS, Guttieri L, Romeo MA, Santarelli R, Cirone M. IRE1 Alpha/XBP1 Axis Sustains Primary Effusion Lymphoma Cell Survival by Promoting Cytokine Release and STAT3 Activation. Biomedicines 2021; 9:biomedicines9020118. [PMID: 33513694 PMCID: PMC7912693 DOI: 10.3390/biomedicines9020118] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/21/2021] [Accepted: 01/25/2021] [Indexed: 12/11/2022] Open
Abstract
Primary Effusion Lymphoma (PEL) is a highly aggressive B cell lymphoma associated with Kaposi's Sarcoma-associated Herpesvirus (KSHV). It is characterized by a high level of basal Endoplasmic Reticulum (ER) stress, Unfolded Protein Response (UPR) activation and constitutive phosphorylation of oncogenic pathways such as the Signal Transducer and activator of Transcription (STAT3). In this study, we found that the inositol requiring kinase (IRE) 1alpha/X-box binding protein (XBP1) axis of UPR plays a key role in the survival of PEL cells, while double stranded RNA-activated protein kinase-like ER kinase (PERK) and activating transcription factor (ATF) 6 slightly influence it, in correlation with the capacity of the IRE1alpha/XBP1 axis to induce the release of interleukin (IL)-6, IL-10 and Vascular-Endothelial Growth Factor (VEGF). Moreover, we found that IRE1alpha/XBP1 inhibition reduced STAT3 Tyr705 phosphorylation and induced a pro-survival autophagy in PEL cells. In conclusion, this study suggests that targeting the IRE1alpha/XBP1 axis represents a promising strategy against PEL cells and that the cytotoxic effect of this treatment may be potentiated by autophagy inhibition.
Collapse
Affiliation(s)
- Roberta Gonnella
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (R.G.); (M.S.G.M.); (M.A.R.); (R.S.)
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00161 Rome, Italy;
| | - Maria Saveria Gilardini Montani
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (R.G.); (M.S.G.M.); (M.A.R.); (R.S.)
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00161 Rome, Italy;
| | - Luisa Guttieri
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00161 Rome, Italy;
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Maria Anele Romeo
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (R.G.); (M.S.G.M.); (M.A.R.); (R.S.)
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00161 Rome, Italy;
| | - Roberta Santarelli
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (R.G.); (M.S.G.M.); (M.A.R.); (R.S.)
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00161 Rome, Italy;
| | - Mara Cirone
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (R.G.); (M.S.G.M.); (M.A.R.); (R.S.)
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00161 Rome, Italy;
- Correspondence:
| |
Collapse
|
26
|
DE Nunzio C, Giglio S, Baldassarri V, Cirombella R, Mallel G, Nacchia A, Tubaro A, Vecchione A. Impairment of autophagy may represent the molecular mechanism behind the relationship between obesity and inflammation in patients with BPH and LUTS. Minerva Urol Nephrol 2020; 73:631-637. [PMID: 33200897 DOI: 10.23736/s2724-6051.20.03992-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Aim of this study was to evaluate the roles of inflammation and autophagy in obese patients with benign prostatic hyperplasia (BPH) and lower urinary tract symptoms (LUTS). METHODS We analyzed 150 surgical specimens from patients underwent transurethral resection of the prostate (TURP) for LUTS/BPH (Median age 70.3±8.1 years, median BMI 25.7±4.0 kg/m2 and median PSA 6.0±5.4 ng/mL). All surgical specimens were investigated for the presence inflammatory infiltrates, according to the standardized classification of chronic prostatitis of the National Institute of Health. The inflammatory score (IS Score) was calculated. High IS score was defined as ≥7. Each sample was stained for anti-LC3B (cell signaling) and for anti-P62/SQSTM1 (MBL) according to manufacturer's suggestions and scored as follow: 0 (no dots); 1 (detectable dots in 5-25% of cells); 2 (readily detectable dots in 25-75% of cells); 3 (dots in >75% of cells). High percentage of p62 or LC3B was defined as >25%, whereas low percentage of p62 or LC3B was defined as <25% of cells with dots. RESULTS Overall 74/150 (49.3%) patients were overweight or obese (BMI >25 kg/m2). Obese patients presented a higher inflammatory score. Obese/overweight patients presented a lower percentage of LC3B (58/74; 78.4%) and higher of p62 (49/74; 66.2%) compared to those of normal weight, which it means a deactivated autophagy (P<0.05). At multivariate analysis LC3B (OR=0.22; CI: 0.069-0.70; P=0.01) percentage and BMI (OR=1.118; CI: 1.001-1.250; P=0.04) were independent risk factors of prostatic inflammation (IS≥7). CONCLUSIONS Here we confirm the association between obesity and prostatic inflammatory infiltrates and present the first evidence of autophagy deregulation in obese patients with LUTS/BPH. Further studies should better investigate this relationship and provide new possible therapeutic targets.
Collapse
Affiliation(s)
| | - Simona Giglio
- Unit of Urology and Surgical Pathology, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Sapienza University, Rome, Italy
| | | | - Roberto Cirombella
- Unit of Urology and Surgical Pathology, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Sapienza University, Rome, Italy
| | - Giuseppe Mallel
- Unit of Urology and Surgical Pathology, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Sapienza University, Rome, Italy
| | | | - Andrea Tubaro
- Sant'Andrea Hospital, Sapienza University, Rome, Italy
| | - Andrea Vecchione
- Sant'Andrea Hospital, Sapienza University, Rome, Italy.,Unit of Urology and Surgical Pathology, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Sapienza University, Rome, Italy
| |
Collapse
|
27
|
Yang SJ, Han AR, Choi HR, Hwang K, Kim EA, Choi SY, Cho SW. N-Adamantyl-4-methylthiazol-2-amine suppresses glutamate-induced autophagic cell death via PI3K/Akt/mTOR signaling pathways in cortical neurons. BMB Rep 2020. [PMID: 32635984 PMCID: PMC7607153 DOI: 10.5483/bmbrep.2020.53.10.059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We recently reported that N-adamantyl-4-methylthiazol-2-amine (KHG26693) attenuates glutamate-induced oxidative stress and inflammation in the brain. In this study, we investigated KHG 26693 as a therapeutic agent against glutamate-induced autophagic death of cortical neurons. Treatment with KHG26693 alone did not affect the viability of cultured cortical neurons but was protective against glutamate-induced cytotoxicity in a concentration-dependent manner. KHG26693 attenuated the glutamate-induced increase in protein levels of LC3, beclin-1, and p62. Whereas glutamate decreased the phosphorylation of PI3K, Akt, and mTOR, these levels were restored by treatment with KHG26693. These results suggest that KHG26693 inhibits glutamate-induced autophagy by regulating PI3K/Akt/mTOR signaling. Finally, KHG26693 treatment also attenuated glutamate-induced increases in reactive oxygen species, glutathione, glutathione peroxidase, and superoxide dismutase levels in cortical neurons, indicating that KHG26693 also protects cortical neurons against glutamate-induced autophagy by regulating the reactive oxygen species scavenging system.
Collapse
Affiliation(s)
- Seung-Ju Yang
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365, Korea
| | - A Reum Han
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Hye-Rim Choi
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365, Korea
| | - Kyouk Hwang
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Eun-A Kim
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chunchon 24252, Korea
| | - Sung-Woo Cho
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| |
Collapse
|
28
|
Zhao T, Ma J, Li L, Teng W, Tian Y, Ma Y, Wang W, Yan W, Jiao P. MKP-5 Relieves Lipotoxicity-Induced Islet β-Cell Dysfunction and Apoptosis via Regulation of Autophagy. Int J Mol Sci 2020; 21:ijms21197161. [PMID: 32998359 PMCID: PMC7582937 DOI: 10.3390/ijms21197161] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 09/16/2020] [Accepted: 09/25/2020] [Indexed: 01/28/2023] Open
Abstract
Mitogen-activated protein kinase phosphatase-5 (MKP-5) is a regulator of extracellular signaling that is known to regulate lipid metabolism. In this study, we found that obesity caused by a high-fat diet (HFD) decreased the expression of MKP-5 in the pancreas and primary islet cells derived from mice. Then, we further investigated the role of MKP-5 in the protection of islet cells from lipotoxicity by modulating MKP-5 expression. As a critical inducer of lipotoxicity, palmitic acid (PA) was used to treat islet β-cells. We found that MKP-5 overexpression restored PA-mediated autophagy inhibition in Rin-m5f cells and protected these cells from PA-induced apoptosis and dysfunction. Consistently, a lack of MKP-5 aggravated the adverse effects of lipotoxicity. Islet cells from HFD-fed mice were infected using recombinant adenovirus expressing MKP-5 (Ad-MKP-5), and we found that Ad-MKP-5 was able to alleviate HFD-induced apoptotic protein activation and relieve the HFD-mediated inhibition of functional proteins. Notably, HFD-mediated impairments in autophagic flux were restored by Ad-MKP-5 transduction. Furthermore, the autophagy inhibitor 3-methyladenine (3-MA) was used to treat Rin-m5f cells, confirming that the MKP-5 overexpression suppressed apoptosis, dysfunction, inflammatory response, and oxidative stress induced by PA via improving autophagic signaling. Lastly, employing c-Jun amino-terminal kinas (JNK), P38, or extracellular-regulated kinase (ERK) inhibitors, we established that the JNK and P38 MAPK pathways were involved in the MKP-5-mediated apoptosis, dysfunction, and autophagic inhibition observed in islet β cells in response to lipotoxicity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Weiqun Yan
- Correspondence: (W.Y.); (P.J.); Tel.: +86-431-8561-9289 (P.J.)
| | - Ping Jiao
- Correspondence: (W.Y.); (P.J.); Tel.: +86-431-8561-9289 (P.J.)
| |
Collapse
|
29
|
Hyperactive Innate Immunity Causes Degeneration of Dopamine Neurons upon Altering Activity of Cdk5. Cell Rep 2020; 26:131-144.e4. [PMID: 30605670 PMCID: PMC6442473 DOI: 10.1016/j.celrep.2018.12.025] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 10/10/2018] [Accepted: 12/05/2018] [Indexed: 11/23/2022] Open
Abstract
Innate immunity is central to the pathophysiology of neurodegenerative disorders, but it remains unclear why immunity is altered in the disease state and whether changes in immunity are a cause or a consequence of neuronal dysfunction. Here, we identify a molecular pathway that links innate immunity to age-dependent loss of dopaminergic neurons in Drosophila. We find, first, that altering the expression of the activating subunit of the Cdk5 protein kinase (Cdk5α) causes severe disruption of autophagy. Second, this disruption of autophagy is both necessary and sufficient to cause the hyperactivation of innate immunity, particularly expression of anti-microbial peptides. Finally, it is the upregulation of immunity that induces the age-dependent death of dopaminergic neurons. Given the dysregulation of Cdk5 and innate immunity in human neurodegeneration and the conserved role of the kinase in the regulation of autophagy, this sequence is likely to have direct application to the chain of events in human neurodegenerative disease. How can one disentangle the many pathologies of neurodegeneration from one another and from normal aging? Shukla et al. show that a mutation in Drosophila kills neurons by impairing autophagy, which in turn stimulates neurotoxic levels of innate immunity, and this acts synergistically with a parallel pathway that accelerates aging.
Collapse
|
30
|
Chen W, Liang J, Fu Y, Jin Y, Yan R, Chi J, Liu W, Liu Y, Yin X. Cardioprotection of cortistatin against isoproterenol-induced myocardial injury in rats. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:309. [PMID: 32355753 PMCID: PMC7186754 DOI: 10.21037/atm.2020.02.93] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background The present study was designed to examine whether cortistatin (CORT) could protect rats from myocardial injury induced by subcutaneously injecting isoproterenol (ISO) and to clarify the possible mechanisms. Methods Male Sprague-Dawley (SD) rats were placed at random into four groups: the control group, the ISO group, the ISO + CORT 25 µg/(kg·d) group, and the ISO + CORT 50 µg/(kg·d) group. Rat models of myocardial injury were established with the subcutaneous (s.c.) injections of 85 mg/kg ISO for 2 days. In the ISO+ CORT 25 µg/(kg·d) group and ISO+ CORT 50 µg/(kg·d) group, rats were given s.c. injections of CORT 25 µg/(kg·d) and CORT 50 µg/(kg·d) on the day before ISO, 3 days, respectively. Serum malondialdehyde (MDA) content, lactate dehydrogenase (LDH) activity, and creatine kinase isoenzyme (CK-MB) activity were measured by corresponding test kits. Western blot was applied to evaluate the expression of endoplasmic reticulum stress-related protein glucose regulatory protein 78 (GRP78), enhancer-binding protein homologous protein (CHOP), cysteinyl aspartate specific proteinase-12 (caspase-12), LC3-II, Beclin-1, and p62 in the rat myocardium. Results CORT alleviated the increased enzyme activities of serum LDH and CK-MB, and content of MDA (a typical marker of lipid peroxidation) in rats induced by ISO. CORT also prevented pathological myocardial injury in rats induced by ISO. Moreover, CORT attenuated the increased protein levels of GRP78, CHOP, and caspase-12, and reduced the increase of LC3-II, LC3-II/I, Beclin-1, and p62 in rats induced by ISO. Conclusions These data demonstrate that CORT can attenuate ISO-induced acute myocardial injury in rats likely by reducing lipid peroxidation, and inhibiting endoplasmic reticulum stress and autophagy. This supports CORT as a potentially being a new target for preventing and treating myocardial injury and its related disease.
Collapse
Affiliation(s)
- Wenjia Chen
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Juan Liang
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510120, China
| | - Yu Fu
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Yuanyuan Jin
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Runan Yan
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Jinyu Chi
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Wenxiu Liu
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Yue Liu
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Xinhua Yin
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| |
Collapse
|
31
|
Wang W, Zhang F, Yan X, Tan Q. Wnt7a regulates high autophagic and inflammatory response of epidermis in high-glucose environment. Burns 2020; 46:121-127. [DOI: 10.1016/j.burns.2019.07.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 06/23/2019] [Accepted: 07/18/2019] [Indexed: 12/23/2022]
|
32
|
Bai Y, Bao X, Jiang G, Ge D, He W, Zhao D, Zhang Y, Dong R, Hua J, Yang N, Mo F, Gao S. Jiang Tang Xiao Ke Granule Protects Hepatic Tissue of Diabetic Mice Through Modulation of Insulin and Ras Signaling - A Bioinformatics Analysis of MicroRNAs and mRNAs Network. Front Pharmacol 2020; 11:173. [PMID: 32210802 PMCID: PMC7067923 DOI: 10.3389/fphar.2020.00173] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 02/07/2020] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE To investigate the impact of JTXK granule on the miRNA expression profiles in hepatic tissue of diabetic mice, and to explore the molecular targets and associated signaling pathways of JTXK granule in its anti-diabetic effect. METHODS Eight mice were randomly selected as normal group fed with chow diet. Then high fat diet was used to induce diabetic model, and the mice were subsequently divided into JTXK-treated group (J group, n = 6) and model group (M group, n = 6). After 8 weeks' intervention we examined the fasting blood glucose and observed the histopathologic changes in hepatic tissue between these two groups. Next we screened the differentially expressed miRNAs between the two groups using microRNA sequencing analysis. Finally, miRNA target gene prediction, GO and KEGG analysis were applied to explore the function of DEMs. RESULTS The blood glucose level in J group was significantly lower than M group (P < 0.05). The results from H&E staining showed that the arrangement and structure of hepatocytes from J group were basically normal with fewer ballooning degeneration and less inflammatory cell infiltration. Furthermore, a total of 33 significantly differentiated miRNAs were detected in comparison between the two groups (| log2(fold change) | >0.3, P < 0.05). MiRNA-mRNA analysis showed that mmu-miR-30a-5p, mmu-miR-23b-5p, mmu-miR-199a-5p, mmu-miR-425-5p, and mmu-miR-214-3p are closely related to inflammatory response, histological changes and insulin signal transduction in liver. In addition, KEGG analysis showed that the DEMs were closely related to Ras and insulin signaling pathway. CONCLUSION JTXK granule exerts anti-diabetic effect in hepatic tissue of diabetic mice by modulating miRNAs and mRNAs network.
Collapse
Affiliation(s)
- Ying Bai
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xueli Bao
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Guangjian Jiang
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Dongyu Ge
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Weipeng He
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Dandan Zhao
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yi Zhang
- College of City Management, Beijing Open University, Beijing, China
| | - Ruijuan Dong
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jing Hua
- Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Nan Yang
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Fangfang Mo
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Fangfang Mo,
| | - Sihua Gao
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Sihua Gao,
| |
Collapse
|
33
|
Shin JH, Cho DH. TMP21 regulates autophagy by modulating ROS production and mTOR activation. Biochem Biophys Res Commun 2019; 518:746-751. [PMID: 31472964 DOI: 10.1016/j.bbrc.2019.08.125] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 08/23/2019] [Indexed: 02/06/2023]
Abstract
Autophagy is a catabolic cellular response to stress that has been liked to various human diseases. However, the precise involvement of autophagy in health and disease remains unclear. To explore the molecular mechanisms of autophagy, we investigated the effect of TMP21. We found that the down-regulation of TMP21 induced autophagy in SH-SY5Y cells. In addition, the enhanced autophagy observed upon TMP21 depletion was almost completely blocked in ATG5 knockout (KO) or ATG7-KO HeLa cells. Silencing of TMP21 in SH-SY5Y cells also increased the production of cellular reactive oxygen species (ROS). Accordingly, treatment with the ROS scavenger NAC suppressed autophagy activation as well as ROS production in TMP21-depleted cells. In addition, the inhibition of mTOR by treatment with Torin1 was mitigated in TMP21 overexpressing cells compared with that in control cells. Taken together, these results indicated that TMP21 could regulate autophagy by modulating ROS production and mTOR activation.
Collapse
Affiliation(s)
- Ji Hyun Shin
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 0XY, UK
| | - Dong-Hyung Cho
- School of Life Sciences, Kyungpook National University, Daegu, 41566, South Korea.
| |
Collapse
|
34
|
Xu Y, Li Y, Liu X, Pan Y, Sun Z, Xue Y, Wang T, Dou H, Hou Y. SPIONs enhances IL-10-producing macrophages to relieve sepsis via Cav1-Notch1/HES1-mediated autophagy. Int J Nanomedicine 2019; 14:6779-6797. [PMID: 31692534 PMCID: PMC6711564 DOI: 10.2147/ijn.s215055] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 07/28/2019] [Indexed: 12/12/2022] Open
Abstract
Background Sepsis is a life-threatening condition caused by dysregulated host responses to infection. Macrophages, which recognize microbial infections through identification of bacterial markers such as lipopolysaccharide (LPS), are crucial to the pathogenesis of sepsis-associated liver injury. However, the understanding of the SPIONs-mediated modulation of macrophage responses in LPS-induced sepsis and liver injury is limited. Materials and methods Superparamagnetic iron oxide nanoparticles (SPIONs) of γ-Fe2O3 nanoparticles were prepared, and their morphology and magnetic properties were characterized. Results Using a murine model of LPS-induced sepsis and liver injury, we found that SPIONs alleviated LPS-induced sepsis, preventing infiltration of inflammatory cells into the liver. SPIONs also increased the level of interleukin-10 (IL-10) in liver macrophages, while SPIONs’s effect on LPS-induced sepsis was abrogated in IL-10-/- mice, indicating that the protective effect of SPIONs is dependent on IL-10+ macrophages. Moreover, SPIONs activated macrophage autophagy to increase IL-10 production, which was markedly attenuated by autophagy inhibition. Furthermore, SPIONs upregulated the expression of Caveolin-1 (Cav1) in macrophages, which plays a role in cellular uptake of metallic nanoparticles. Interestingly, activation of Cav1 and Notch1/HES1 signaling was involved in SPIONs-induced autophagy in both RAW 264.7 cells and bone marrow-derived macrophages (BMDMs). Our data reveal a novel mechanism for SPIONs -induced autophagy in macrophages, which occurs through activation of the Cav1-Notch1/HES1 signaling pathway, which promotes the production of IL-10 in macrophages, leading to inhibition of inflammation in LPS-induced sepsis and liver injury. Conclusion Our results suggest that SPIONs may represent a potential therapeutic agent for the treatment of sepsis and sepsis-induced liver injury.
Collapse
Affiliation(s)
- Yujun Xu
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, People's Republic of China
| | - Yi Li
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, People's Republic of China
| | - Xinghan Liu
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, People's Republic of China
| | - Yuchen Pan
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, People's Republic of China
| | - Zhiheng Sun
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, People's Republic of China
| | - Yaxian Xue
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, People's Republic of China
| | - Tingting Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, People's Republic of China.,Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, People's Republic of China
| | - Huan Dou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, People's Republic of China.,Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, People's Republic of China
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, People's Republic of China.,Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, People's Republic of China
| |
Collapse
|
35
|
Hong P, Gu RN, Li FX, Xiong XX, Liang WB, You ZJ, Zhang HF. NLRP3 inflammasome as a potential treatment in ischemic stroke concomitant with diabetes. J Neuroinflammation 2019; 16:121. [PMID: 31174550 PMCID: PMC6554993 DOI: 10.1186/s12974-019-1498-0] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 05/02/2019] [Indexed: 02/07/2023] Open
Abstract
The NLRP3 (nucleotide-binding oligomerization domain-like receptor [NLR] family pyrin domain-containing 3) inflammasome is a member of the NLR family of innate immune cell sensors. These are crucial regulators of cytokine secretions, which promote ischemic cell death and insulin resistance. This review summarizes recent progress regarding the NLRP3 inflammasome as a potential treatment for ischemic stroke in patients with diabetes, two complicated diseases that often occur together. Stroke worsens glucose metabolism abnormalities, and the outcomes after stroke are more serious for diabetic patients compared with those without diabetes. Inflammation contributes to organ injury after ischemic stroke and diabetes. Recent research has focused on inhibiting the activation of inflammasomes and thus reducing the maturation of proinflammatory cytokines such as interleukin (IL)-1β and IL-18. Studies suggest that inhibition of NLRP3 prevents or alleviates both ischemic stroke and diabetes. Targeting against the assembly and activity of the NLRP3 inflammasome is a potential and novel therapy for inflammasome-associated diseases, including ischemic stroke concomitant with diabetes.
Collapse
Affiliation(s)
- Pu Hong
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Ruo-Nan Gu
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Feng-Xian Li
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Xiao-Xing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Wen-Bin Liang
- Cardiac Electrophysiology Lab, University of Ottawa Heart Institute, Ottawa, Ontario, K1Y 4 W7, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, K1Y 4 W7, Canada
| | - Zhi-Jian You
- Department of Anesthesiology, Shenzhen SAMII Medical Center, Shenzhen, Guangdong, People's Republic of China.
| | - Hong-Fei Zhang
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|
36
|
Moura J, Madureira P, Leal EC, Fonseca AC, Carvalho E. Immune aging in diabetes and its implications in wound healing. Clin Immunol 2019; 200:43-54. [PMID: 30735729 PMCID: PMC7322932 DOI: 10.1016/j.clim.2019.02.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 02/04/2019] [Accepted: 02/04/2019] [Indexed: 02/06/2023]
Abstract
Immune systems have evolved to recognize and eliminate pathogens and damaged cells. In humans, it is estimated to recognize 109 epitopes and natural selection ensures that clonally expanded cells replace unstimulated cells and overall immune cell numbers remain stationary. But, with age, it faces continuous repertoire restriction and concomitant accumulation of primed cells. Changes shaping the aging immune system have bitter consequences because, as inflammatory responses gain intensity and duration, tissue-damaging immunity and inflammatory disease arise. During inflammation, the glycolytic flux cannot cope with increasing ATP demands, limiting the immune response's extent. In diabetes, higher glucose availability stretches the glycolytic limit, dysregulating proteostasis and increasing T-cell expansion. Long-term hyperglycemia exerts an accumulating effect, leading to higher inflammatory cytokine levels and increased cytotoxic mediator secretion upon infection, a phenomenon known as diabetic chronic inflammation. Here we review the etiology of diabetic chronic inflammation and its consequences on wound healing.
Collapse
Affiliation(s)
- J Moura
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; INEB - Instituto Nacional de Engenharia Biomédica, University of Porto, Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.
| | - P Madureira
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal; IBMC - Instituto de Biologia Celular e Molecular, University of Porto, Porto, Portugal; Immunethep, Biocant Park, Cantanhede, Portugal
| | - E C Leal
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - A C Fonseca
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - E Carvalho
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Instituto de Investigação Interdisciplinar, University of Coimbra, Coimbra, Portugal; Department of Geriatrics, University of Arkansas for Medical Sciences and Arkansas Children's Research Institute, Little Rock, AR, United States
| |
Collapse
|
37
|
Das LM, Binko AM, Traylor ZP, Peng H, Lu KQ. Vitamin D improves sunburns by increasing autophagy in M2 macrophages. Autophagy 2019; 15:813-826. [PMID: 30661440 PMCID: PMC6526871 DOI: 10.1080/15548627.2019.1569298] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Cutaneous inflammation from UV radiation exposure causes epidermal damage, cellular infiltration, and secretion of pro-inflammatory mediators that exacerbate tissue destruction. Recovery is mediated chiefly by anti-inflammatory M2 macrophages that suppress inflammation and augment epidermal regeneration. Vitamin D enables anti-inflammation to promote tissue repair in response to injury. Since vitamin D enhances cellular macroautophagy/autophagy, we investigated the role of autophagy in vitamin D protection of UV-mediated sunburn and inflammation. Using a UV-mediated acute skin injury mouse model, we demonstrate that a single dose of vitamin D resolves injury with sustained inhibition of inflammatory cytokines associated with enhanced autophagy in myeloid anti-inflammatory M2 macs. Increased MAP1LC3B/LC3 expression corroborated with complete autolysosome formation detected by electron microscopy and correlated with degradation of SQSTM1/p62 in the skin following vitamin D treatment. Specifically, pharmacological inhibition of autophagy increased UV-induced apoptosis, suppressed M2 macs recruitment, and prevented vitamin D downregulation of Tnf and Mmp9 in the skin. Furthermore, selective deletion of autophagy in myeloid cells of atg7 cKO mice abrogated vitamin D-mediated protection and recapitulated UV-induced inflammation. Mechanistically, vitamin D signaling activated M2-autophagy regulators Klf4, Pparg, and Arg1. Lastly, analysis of UV-exposed human skin biopsies detected a similar increase in macrophage autophagy following vitamin D intervention, identifying an essential role for autophagy in vitamin D-mediated protection of skin from UV damage. Abbreviations: ARG1: arginase 1; ATG7 cKO: autophagy related 7 conditional knockout; HPF: high powered field; KLF4: Kruppel like factor 4; MAP1LC3B/LC3: microtubule-associated protein 1 light chain 3 beta; macs: macrophage; 3-MA: 3-methyladenine; MMP9: matrix metallopeptidase 9; NOS2: nitric oxide synthase 2, inducible; PPARG: peroxisome proliferator activated receptor gamma; SQSTM1/p62: sequestosome 1; TNF: tumor necrosis factor; UV: ultraviolet; VD: vitamin D, 25-hydroxy vitamin D3; 1,25-VD: 1, 25-dihydroxy vitamin D3
Collapse
Affiliation(s)
- Lopa M Das
- a Department of Dermatology , Case Western Reserve University School of Medicine , Cleveland , OH , USA
| | - Amy M Binko
- a Department of Dermatology , Case Western Reserve University School of Medicine , Cleveland , OH , USA
| | - Zachary P Traylor
- a Department of Dermatology , Case Western Reserve University School of Medicine , Cleveland , OH , USA
| | - Han Peng
- b Department of Dermatology , Northwestern University , Chicago , IL , USA
| | - Kurt Q Lu
- a Department of Dermatology , Case Western Reserve University School of Medicine , Cleveland , OH , USA.,b Department of Dermatology , Northwestern University , Chicago , IL , USA.,c Department of Dermatology , University Hospitals Cleveland Medical Center , Cleveland , OH , USA
| |
Collapse
|
38
|
Chang SL, Huang W, Han H, Sariyer IK. Binge-Like Exposure to Ethanol Enhances Morphine's Anti-nociception in B6 Mice. Front Psychiatry 2019; 9:756. [PMID: 30723430 PMCID: PMC6349749 DOI: 10.3389/fpsyt.2018.00756] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 12/20/2018] [Indexed: 01/01/2023] Open
Abstract
Elevation of the blood ethanol concentration (BEC) to > 80 mg/dL (17.4 mM) after binge drinking enhances inflammation in brain and neuroimmune signaling pathways. Morphine abuse is frequently linked to excessive drinking. Morphine exerts its actions mainly via the seven transmembrane G-protein-coupled mu opioid receptors (MORs). Opioid use disorders (OUDs) include combination of opioids with alcohol, leading to opioid overdose-related deaths. We hypothesized that binge drinking potentiates onset and progression of OUD. Using C57BL/6J (B6) mice, we first characterized time-dependent inflammatory gene expression change as molecular markers using qRT-PCR within 24 h after binge-like exposure to high-dose, high-concentration ethanol (EtOH). The mice were given one injection of EtOH (5 g/kg, 42% v/v, i.g.) and sacrificed at 2.5 h, 5 h, 7.5 h, or 24 h later. Inflammatory cytokines interleukin (IL)-1β, IL-6, and IL-18 were elevated in both the striatum (STr) and the nucleus accumbens (NAc) of the mice. We then investigated the expression profile of MOR in the STr at 2 min, 5 h, or 24 h after the first EtOH injection and at 24 h and 48 h after the third injection. This binge-like exposure to EtOH upregulated MOR expression in the STr and NAc, an effect that could enhance morphine's anti-nociception. Therefore, we examined the impact of binge-like exposure to EtOH on morphine's anti-nociception at the behavioral level. The mice were treated with or without 3-d binge-like exposure to EtOH, and the anti-nociceptive changes were evaluated using the hot-plate test 24 h after the final (3rd) EtOH injection with or without a cumulative subcutaneous dose (0, 0.1, 0.3, 1.0, and 3.0 mg/kg) of morphine at intervals of 30 min. The response curve of the mice given EtOH was shifted to the left, showing enhanced latency to response to morphine up to 3 mg/kg. Furthermore, co-treatment with the MOR antagonist naltrexone blocked morphine's anti-nociception in animals given either EtOH or saline. This confirms that MOR is involved in binge-like exposure to EtOH-induced changes in morphine's anti-nociception. Our results suggest that EtOH enhanced latency to analgesic response to morphine, and such effect might initiate the onset and progression of OUDs.
Collapse
Affiliation(s)
- Sulie L Chang
- Institute of NeuroImmune Pharmacology, South Orange, NJ, United States
- Department of Biological Sciences, Seton Hall University, South Orange, NJ, United States
| | - Wenfei Huang
- Institute of NeuroImmune Pharmacology, South Orange, NJ, United States
- Department of Biological Sciences, Seton Hall University, South Orange, NJ, United States
| | - Haijun Han
- Institute of NeuroImmune Pharmacology, South Orange, NJ, United States
| | - Ilker K Sariyer
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
39
|
Shi GJ, Li Y, Cao QH, Wu HX, Tang XY, Gao XH, Yu JQ, Chen Z, Yang Y. In vitro and in vivo evidence that quercetin protects against diabetes and its complications: A systematic review of the literature. Biomed Pharmacother 2019; 109:1085-1099. [DOI: 10.1016/j.biopha.2018.10.130] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 10/21/2018] [Accepted: 10/21/2018] [Indexed: 12/14/2022] Open
|
40
|
Yin J, Ren W, Chen S, Li Y, Han H, Gao J, Liu G, Wu X, Li T, Woo Kim S, Yin Y. Metabolic Regulation of Methionine Restriction in Diabetes. Mol Nutr Food Res 2018; 62:e1700951. [PMID: 29603632 DOI: 10.1002/mnfr.201700951] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 01/25/2018] [Indexed: 12/16/2022]
Abstract
Although the effects of dietary methionine restriction have been investigated in the physiology of aging and diseases related to oxidative stress, the relationship between methionine restriction (MR) and the development of metabolic disorders has not been explored extensively. This review summarizes studies of the possible involvement of dietary methionine restriction in improving insulin resistance, glucose homeostasis, oxidative stress, lipid metabolism, the pentose phosphate pathway (PPP), and inflammation, with an emphasis on the fibroblast growth factor 21 and protein phosphatase 2A signals and autophagy in diabetes. Diets deficient in methionine may be a useful nutritional strategy in patients with diabetes.
Collapse
Affiliation(s)
- Jie Yin
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China.,Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China.,University of Chinese Academy of Sciences, Beijing, PR, China
| | - Wenkai Ren
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China.,Jiangsu Co-Innovation Center for Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product, Safety of Ministry of Education of China, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Shuai Chen
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China.,University of Chinese Academy of Sciences, Beijing, PR, China
| | - Yuying Li
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China.,University of Chinese Academy of Sciences, Beijing, PR, China
| | - Hui Han
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China.,University of Chinese Academy of Sciences, Beijing, PR, China
| | - Jing Gao
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China.,University of Chinese Academy of Sciences, Beijing, PR, China
| | - Gang Liu
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| | - Xin Wu
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China.,Hunan Co-Innovation Center of Animal Production Safety, Changsha, PR, China
| | - Tiejun Li
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China.,Hunan Co-Innovation Center of Animal Production Safety, Changsha, PR, China
| | - Sung Woo Kim
- Department of Animal Science, North Carolina State University, Raleigh, NC, USA
| | - Yulong Yin
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China.,Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China.,Hunan Co-Innovation Center of Animal Production Safety, Changsha, PR, China
| |
Collapse
|
41
|
Sokolova M, Sahraoui A, Høyem M, Øgaard J, Lien E, Aukrust P, Yndestad A, Ranheim T, Scholz H. NLRP3 inflammasome mediates oxidative stress-induced pancreatic islet dysfunction. Am J Physiol Endocrinol Metab 2018; 315:E912-E923. [PMID: 30016155 DOI: 10.1152/ajpendo.00461.2017] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Inflammasomes are multiprotein inflammatory platforms that induce caspase-1 activation and subsequently interleukin (IL)-1β and IL-18 processing. The NLRP3 inflammasome is activated by different forms of oxidative stress, and, based on the central role of IL-1β in the destruction of pancreatic islets, it could be related to the development of diabetes. We therefore investigated responses in wild-type C57Bl/6 (WT) mice, NLRP3-/- mice, and mice deficient in apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC) after exposing islets to short-term hypoxia or alloxan-induced islet damage. NLRP3-deficient islets compared with WT islets had preserved function ex vivo and were protected against hypoxia-induced cell death. Furthermore, NLRP3 and ASC-deficient mice were protected against oxidative stress-induced diabetes caused by repetitive low-dose alloxan administration, and this was associated with reduced β-cell death and reduced macrophage infiltration. This suggests that the beneficial effect of NLRP3 inflammasome deficiency on oxidative stress-mediated β-cell damage could involve reduced macrophage infiltration and activation. To support the role of macrophage activation in alloxan-induced diabetes, we injected WT mice with liposomal clodronate, which causes macrophage depletion before induction of a diabetic phenotype by alloxan treatment, resulting in improved glucose homeostasis in WT mice. We show here that the NLRP3 inflammasome acts as a mediator of hypoxia and oxidative stress in insulin-producing cells, suggesting that inhibition of the NLRP3 inflammasome could have beneficial effects on β-cell preservation.
Collapse
Affiliation(s)
- Marina Sokolova
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Norway
- Institute of Clinical Medicine, University of Oslo , Oslo , Norway
- K.G. Jebsen Inflammation Research Center, University of Oslo , Oslo , Norway
| | - Afaf Sahraoui
- Institute of Clinical Medicine, University of Oslo , Oslo , Norway
- Institute for Surgical Research and Section for Transplantation Surgery, Oslo University Hospital , Oslo , Norway
| | - Merete Høyem
- Institute for Surgical Research and Section for Transplantation Surgery, Oslo University Hospital , Oslo , Norway
| | - Jonas Øgaard
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Norway
| | - Egil Lien
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Norway
- Institute of Clinical Medicine, University of Oslo , Oslo , Norway
- K.G. Jebsen Inflammation Research Center, University of Oslo , Oslo , Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital , Oslo , Norway
| | - Arne Yndestad
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Norway
- Institute of Clinical Medicine, University of Oslo , Oslo , Norway
- K.G. Jebsen Inflammation Research Center, University of Oslo , Oslo , Norway
| | - Trine Ranheim
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Norway
- Institute of Clinical Medicine, University of Oslo , Oslo , Norway
- K.G. Jebsen Inflammation Research Center, University of Oslo , Oslo , Norway
| | - Hanne Scholz
- Institute of Clinical Medicine, University of Oslo , Oslo , Norway
- Institute for Surgical Research and Section for Transplantation Surgery, Oslo University Hospital , Oslo , Norway
| |
Collapse
|
42
|
Kazemi A, Sadri M, Houshmand M, Yazdi N, Zarif MN, Anjam-Najmedini A, Tavakoli R, Ojaghi M, Ajami M, Ajami M, Atashi A. The anticancer effects of pharmacological inhibition of autophagy in acute erythroid leukemia cells. Anticancer Drugs 2018; 29:944-955. [DOI: 10.1097/cad.0000000000000668] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
43
|
Kosacka J, Nowicki M, Paeschke S, Baum P, Blüher M, Klöting N. Up-regulated autophagy: as a protective factor in adipose tissue of WOKW rats with metabolic syndrome. Diabetol Metab Syndr 2018; 10:13. [PMID: 29507613 PMCID: PMC5834836 DOI: 10.1186/s13098-018-0317-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 02/26/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Wistar Ottawa Karlsburg W (RT1u) rats (WOKW) are a model of the metabolic syndrome (MetS). Adipose tissue (AT) and peripheral nerves of WOKW rats exhibit up-regulated autophagy and inflammation corresponding with decreased apoptosis rate. The aim of this study was to characterize AT in WOKW rats in relation to autophagic activity. METHODS mRNA and protein expression of adiponectin, pro-inflammatory and pro-apoptotic markers including MCP1, TNFα, cleaved caspase-3 and RNF157, a new candidate gene regulated through autophagy, were analyzed in adipocytes isolated from visceral and subcutaneous AT of 5-month old WOKW rats with MetS and LEW.1W controls in response to pharmacological inhibition of autophagy. Immunohistochemistry was performed to detect adiponectin and RNF157 protein in cultured adipocytes. RESULTS Inhibition of autophagy by LY294002 was associated with a fourfold up-regulation of adiponectin expression and a decrease of RNF157 protein and pro-inflammatory markers-MCP-1 and TNFα predominantly in visceral adipocytes of obese WOKW rats compared to LEW.1W rats. Moreover, inhibition of autophagic activity correlates with an activation of cleaved caspase-3 apoptotic signaling pathway. CONCLUSIONS Up-regulated autophagy in obese WOKW rats contributes to the regulation of visceral AT function and involves an altered balance between pro-inflammatory and protective adipokine expression. Our data suggest that activation of AT autophagy protects against adipocyte apoptosis at least under conditions of obesity related MetS in WOKW rats.
Collapse
Affiliation(s)
- J. Kosacka
- Department of Neurology, University of Leipzig, Liebigstraße 20, 04103 Leipzig, Germany
- Department of Medicine, University of Leipzig, Liebigstraße 21, 04103 Leipzig, Germany
| | - M. Nowicki
- Institute of Anatomy, University of Leipzig, Oststraße 25, 04317 Leipzig, Germany
| | - S. Paeschke
- Institute of Anatomy, University of Leipzig, Oststraße 25, 04317 Leipzig, Germany
| | - P. Baum
- Department of Neurology, University of Leipzig, Liebigstraße 20, 04103 Leipzig, Germany
| | - M. Blüher
- Department of Medicine, University of Leipzig, Liebigstraße 21, 04103 Leipzig, Germany
| | - N. Klöting
- Department of Medicine, University of Leipzig, Liebigstraße 21, 04103 Leipzig, Germany
- Integrated Research and Treatment Center (IFB) Adiposity Diseases, Liebigstraße 19-21, 04103 Leipzig, Germany
| |
Collapse
|
44
|
Zheng W, Zhou J, Song S, Kong W, Xia W, Chen L, Zeng T. Dipeptidyl-Peptidase 4 Inhibitor Sitagliptin Ameliorates Hepatic Insulin Resistance by Modulating Inflammation and Autophagy in ob/ob Mice. Int J Endocrinol 2018; 2018:8309723. [PMID: 30123267 PMCID: PMC6079465 DOI: 10.1155/2018/8309723] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 06/12/2018] [Accepted: 06/19/2018] [Indexed: 02/06/2023] Open
Abstract
Obesity and type 2 diabetes are the most common metabolic diseases globally. They are associated with inflammation, oxidative stress, autophagy, and insulin resistance. Sitagliptin, a dipeptidyl-peptidase 4 inhibitor, has been reported to show multiple biological activities beyond the antidiabetic property. This study was aimed at investigating the effect of sitagliptin on hepatic steatosis, insulin resistance, inflammation, and autophagy and exploring the underlying molecular mechanism. In the current study, ob/ob mice, a mouse model of genetic obesity and diabetes, were administered via gavage with sitagliptin 50 mg/kg daily for 4 weeks. Changes in glycolipid metabolism, inflammatory responses, and autophagy in the liver were evaluated. Body weight gain, lipid metabolic disorder, and hepatic steatosis as well as systemic and hepatic insulin sensitivity in ob/ob mice were significantly attenuated after sitagliptin treatment. Furthermore, sitagliptin decreased inflammatory responses by regulating macrophage M1/M2 polarization and inhibiting the activities of NF-κB and JNK. Moreover, sitagliptin increased the levels of phosphorylation of AMPK and decreased those of mTOR. This study indicates that sitagliptin significantly ameliorates the development of hepatic steatosis and insulin resistance in ob/ob mice by inhibiting inflammatory responses and activating autophagy via AMPK/mTOR signaling pathway.
Collapse
Affiliation(s)
- Wenbin Zheng
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jing Zhou
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, USA
| | - Shasha Song
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wen Kong
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wenfang Xia
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lulu Chen
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tianshu Zeng
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
45
|
Laiglesia LM, Lorente-Cebrián S, López-Yoldi M, Lanas R, Sáinz N, Martínez JA, Moreno-Aliaga MJ. Maresin 1 inhibits TNF-alpha-induced lipolysis and autophagy in 3T3-L1 adipocytes. J Cell Physiol 2017; 233:2238-2246. [PMID: 28703289 DOI: 10.1002/jcp.26096] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Accepted: 07/11/2017] [Indexed: 12/30/2022]
Abstract
Obesity is associated with high levels of pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α), which promotes inflammation in adipose tissue. The omega-3 PUFAs, and their derived lipid mediators, such as Maresin 1 (MaR1) have anti-inflammatory effects on adipose tissue. This study aimed to analyze if MaR1 may counteract alterations induced by TNF-α on lipolysis and autophagy in mature 3T3-L1 adipocytes. Our data revealed that MaR1 (1-100 nM) inhibited the TNF-α-induced glycerol release after 48 hr, which may be related to MaR1 ability of preventing the decrease in lipid droplet-coating protein perilipin and G0/G1 Switch 2 protein expression. MaR1 also reversed the decrease in total hormone sensitive lipase (total HSL), and the ratio of phosphoHSL at Ser-565/total HSL, while preventing the increased ratio of phosphoHSL at Ser-660/total HSL and phosphorylation of extracellular signal-regulated kinase 1/2 induced by TNF-α. Moreover, MaR1 counteracted the cytokine-induced decrease of p62 protein, a key autophagy indicator, and also prevented the induction of LC3II/LC3I, an important autophagosome formation marker. Current data suggest that MaR1 may ameliorate TNF-α-induced alterations on lipolysis and autophagy in adipocytes. This may also contribute to the beneficial actions of MaR1 on adipose tissue and insulin sensitivity in obesity.
Collapse
Affiliation(s)
- Laura M Laiglesia
- Department Nutrition Food Science Physiology, University of Navarra, Pamplona, Spain.,Centre for Nutrition Research, University of Navarra, Pamplona, Spain
| | - Silvia Lorente-Cebrián
- Department Nutrition Food Science Physiology, University of Navarra, Pamplona, Spain.,Centre for Nutrition Research, University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Miguel López-Yoldi
- Department Nutrition Food Science Physiology, University of Navarra, Pamplona, Spain.,Centre for Nutrition Research, University of Navarra, Pamplona, Spain
| | - Raquel Lanas
- Department Nutrition Food Science Physiology, University of Navarra, Pamplona, Spain
| | - Neira Sáinz
- Centre for Nutrition Research, University of Navarra, Pamplona, Spain
| | - Jose Alfredo Martínez
- Department Nutrition Food Science Physiology, University of Navarra, Pamplona, Spain.,Centre for Nutrition Research, University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain.,CIBERobn, Physiopathology of Obesity and Nutrition, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Maria J Moreno-Aliaga
- Department Nutrition Food Science Physiology, University of Navarra, Pamplona, Spain.,Centre for Nutrition Research, University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain.,CIBERobn, Physiopathology of Obesity and Nutrition, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
46
|
Yang JS, Lu CC, Kuo SC, Hsu YM, Tsai SC, Chen SY, Chen YT, Lin YJ, Huang YC, Chen CJ, Lin WD, Liao WL, Lin WY, Liu YH, Sheu JC, Tsai FJ. Autophagy and its link to type II diabetes mellitus. Biomedicine (Taipei) 2017; 7:8. [PMID: 28612706 PMCID: PMC5479440 DOI: 10.1051/bmdcn/2017070201] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 05/02/2017] [Indexed: 02/06/2023] Open
Abstract
Autophagy, a double-edged sword for cell survival, is the research object on 2016 Nobel Prize in Physiology or Medicine. Autophagy is a molecular mechanism for maintaining cellular physiology and promoting survival. Defects in autophagy lead to the etiology of many diseases, including diabetes mellitus (DM), cancer, neurodegeneration, infection disease and aging. DM is a metabolic and chronic disorder and has a higher prevalence in the world as well as in Taiwan. The character of diabetes mellitus is hyperglycemia resulting from defects in insulin secretion, insulin action, or both. Type 2 diabetes mellitus (T2DM) is characterized by insulin resistance and failure of producing insulin on pancreatic beta cells. In T2DM, autophagy is not only providing nutrients to maintain cellular energy during fasting, but also removes damaged organelles, lipids and miss-folded proteins. In addition, autophagy plays an important role in pancreatic beta cell dysfunction and insulin resistance. In this review, we summarize the roles of autophagy in T2DM.
Collapse
Affiliation(s)
- Jai-Sing Yang
-
Department of Medical Research, China Medical University Hospital, China Medical University Taichung
404 Taiwan
| | - Chi-Cheng Lu
-
Department of Medical Research, China Medical University Hospital, China Medical University Taichung
404 Taiwan
| | - Sheng-Chu Kuo
-
School of Pharmacy, China Medical University Taichung
404 Taiwan
| | - Yuan-Man Hsu
-
Department of Biological Science and Technology, China Medical University Taichung
404 Taiwan
| | - Shih-Chang Tsai
-
Department of Biological Science and Technology, China Medical University Taichung
404 Taiwan
| | - Shih-Yin Chen
-
Genetics Center, Department of Medical Research, China Medical University Hospital Taichung
404 Taiwan
-
School of Chinese Medicine, China Medical University Taichung
404 Taiwan
| | - Yng-Tay Chen
-
Genetics Center, Department of Medical Research, China Medical University Hospital Taichung
404 Taiwan
-
School of Chinese Medicine, China Medical University Taichung
404 Taiwan
| | - Ying-Ju Lin
-
Genetics Center, Department of Medical Research, China Medical University Hospital Taichung
404 Taiwan
-
School of Chinese Medicine, China Medical University Taichung
404 Taiwan
| | - Yu-Chuen Huang
-
Genetics Center, Department of Medical Research, China Medical University Hospital Taichung
404 Taiwan
-
School of Chinese Medicine, China Medical University Taichung
404 Taiwan
| | - Chao-Jung Chen
-
Genetics Center, Department of Medical Research, China Medical University Hospital Taichung
404 Taiwan
-
School of Chinese Medicine, China Medical University Taichung
404 Taiwan
| | - Wei-De Lin
-
Genetics Center, Department of Medical Research, China Medical University Hospital Taichung
404 Taiwan
-
School of Chinese Medicine, China Medical University Taichung
404 Taiwan
| | - Wen-Lin Liao
-
Genetics Center, Department of Medical Research, China Medical University Hospital Taichung
404 Taiwan
-
School of Chinese Medicine, China Medical University Taichung
404 Taiwan
| | - Wei-Yong Lin
-
Genetics Center, Department of Medical Research, China Medical University Hospital Taichung
404 Taiwan
-
School of Chinese Medicine, China Medical University Taichung
404 Taiwan
| | - Yu-Huei Liu
-
Genetics Center, Department of Medical Research, China Medical University Hospital Taichung
404 Taiwan
-
School of Chinese Medicine, China Medical University Taichung
404 Taiwan
| | - Jinn-Chyuan Sheu
-
Institute of Biomedical Sciences, National Sun Yat-sen University Kaohsiung
804 Taiwan
| | - Fuu-Jen Tsai
-
Genetics Center, Department of Medical Research, China Medical University Hospital Taichung
404 Taiwan
-
School of Chinese Medicine, China Medical University Taichung
404 Taiwan
-
Department of Medical Genetics, China Medical University Hospital, China Medical University Taichung
404 Taiwan
| |
Collapse
|
47
|
De Nunzio C, Giglio S, Stoppacciaro A, Gacci M, Cirombella R, Luciani E, Tubaro A, Vecchione A. Autophagy deactivation is associated with severe prostatic inflammation in patients with lower urinary tract symptoms and benign prostatic hyperplasia. Oncotarget 2017; 8:50904-50910. [PMID: 28881614 PMCID: PMC5584215 DOI: 10.18632/oncotarget.15144] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 01/11/2017] [Indexed: 11/25/2022] Open
Abstract
Autophagy is a conserved evolutionary process that allows cells to maintain macromolecular synthesis and energy homeostasis during starvation and stressful conditions. We prospectively evaluated the relationship between autophagy and prostatic inflammation in a series of transurethral prostatic resection samples. Inflammatory infiltrates were defined according to the standardized classification of chronic prostatitis of the National Institute of Health. The inflammatory score (IS score) was calculated. High IS score was defined as ≥7. Each sample was stained for anti-LC3B and for anti-P62/SQSTM1 and scored. High p62 or LC3B percentage was defined as >25%, whereas low was defined as <25% of cells with dots. We analyzed 94 specimens. Overall, 18/94 (19%) showed no sign of prostatic inflammation, whereas 76/94 (81%) presented inflammatory infiltrates. Inflammation was mild in 61/76 (80%), moderate/severe in 15/76 (20%). Patients with high p62 percentage were 62/94 (66%) while 32 (34%) showed low p62 percentage. Patients with high LC3B percentage were 37/94 (39%) while 57(61%) showed low LC3B percentage. Overall 42/94 (44%) patients presented a high p62 percentage and concomitant a low LC3B percentage. IS score was significantly higher in patients with a with high p62 percentage (median IS 7 (6/8) vs 5 (3/7); p= 0.04) and in patients with a low LC3B percentage (median IS 7 (6/8) vs 5 (3/7); p= 0.004) when compared to patients with a low p62 percentage or a high LC3B percentage respectively. On multivariate analysis, p62 (OR: 10.1, 95%CI: 2.6-38.6; p= 0,001) and LC3B expression (OR: 0.319; 95%CI: 0.112-0.907; p= 0.032) were independent predictors of a high IS. Here we present the first evidence of autophagy deregulation in prostatic inflammation. These results raise many questions about the mechanisms mediating the autophagy dysfunction and the links to prostatic inflammation that need to be addressed.
Collapse
Affiliation(s)
- Cosimo De Nunzio
- Urology Units, Department of Clinical and Molecular Medicine, Ospedale Sant'Andrea, Sapienza University, Rome, Italy
| | - Simona Giglio
- Surgical Pathology Units, Department of Clinical and Molecular Medicine, Ospedale Sant'Andrea, Sapienza University, Rome, Italy
| | - Antonella Stoppacciaro
- Surgical Pathology Units, Department of Clinical and Molecular Medicine, Ospedale Sant'Andrea, Sapienza University, Rome, Italy
| | - Mauro Gacci
- Department of Urology, Careggi Hospital, Firenze, Italy
| | - Roberto Cirombella
- Surgical Pathology Units, Department of Clinical and Molecular Medicine, Ospedale Sant'Andrea, Sapienza University, Rome, Italy
| | - Emidio Luciani
- Surgical Pathology Units, Department of Clinical and Molecular Medicine, Ospedale Sant'Andrea, Sapienza University, Rome, Italy
| | - Andrea Tubaro
- Urology Units, Department of Clinical and Molecular Medicine, Ospedale Sant'Andrea, Sapienza University, Rome, Italy
| | - Andrea Vecchione
- Surgical Pathology Units, Department of Clinical and Molecular Medicine, Ospedale Sant'Andrea, Sapienza University, Rome, Italy.,Department of Cancer Biology and Genetics/CCC, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
48
|
Yin JJ, Xie G, Zhang N, Li Y. Inhibiting autophagy promotes endoplasmic reticulum stress and the ROS‑induced nod‑like receptor 3‑dependent proinflammatory response in HepG2 cells. Mol Med Rep 2016; 14:3999-4007. [PMID: 27600251 DOI: 10.3892/mmr.2016.5708] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 08/09/2016] [Indexed: 11/06/2022] Open
Abstract
Inflammation and endoplasmic reticulum (ER) stress are key contributors to insulin resistance and metabolic disease, and interleukin (IL)‑1β is involved in insulin resistance. The present study aimed to investigated the role of autophagy in LPS‑induced ER stress and inflammation, which may provide evidence for controlling metabolic disease associated with inflammation. Lipopolysaccharide (LPS) induced the activation of ER stress and the nod‑like receptor 3‑dependent expression of IL‑1β and caspase‑1, as shown by western blotting, which contributed to HepG2 cell death. This also involved the generation of mitochondrial reactive oxygen species and the autophagy signaling response, which are derived from the ER stress pathway. The percentage of apoptotic cells was measured by flow cytometry with fluorescein isothiocyanate/propidium iodide staining. Reactive oxygen species formation was detected by flow cytometry using the peroxide sensitive fluorescent probe 2',7'‑dichlorofluorescin diacetate. Autophagy activation was measured by western blotting and confirmed using transmission electron microscopy. Furthermore, inhibiting autophagy promoted ER stress and the proinflammatory response in addition to cell death. These findings provide insights into the protective role of autophagy in LPS‑induced cell death and ER stress, and further identified the association of autophagy, ER stress and inflammation in HepG2 cells.
Collapse
Affiliation(s)
- Jia-Jing Yin
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Guangying Xie
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Ning Zhang
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yanbo Li
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
49
|
Blocking Nuclear Factor-Kappa B Protects against Diet-Induced Hepatic Steatosis and Insulin Resistance in Mice. PLoS One 2016; 11:e0149677. [PMID: 26930600 PMCID: PMC4773172 DOI: 10.1371/journal.pone.0149677] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 02/02/2016] [Indexed: 02/06/2023] Open
Abstract
Inflammation critically contributes to the development of various metabolic diseases. However, the effects of inhibiting inflammatory signaling on hepatic steatosis and insulin resistance, as well as the underlying mechanisms remain obscure. In the current study, male C57BL/6J mice were fed a chow diet or high-fat diet (HFD) for 8 weeks. HFD-fed mice were respectively treated with p65 siRNA, non-silence control siRNA or vehicle every 4th day for the last 4 weeks. Vehicle-treated (HF) and non-silence siRNA-treated (HFNS) mice displayed overt inflammation, hepatic steatosis and insulin resistance compared with chow-diet-fed (NC) mice. Upon treatment with NF-κB p65 siRNA, HFD-fed (HFPS) mice were protected from hepatic steatosis and insulin resistance. Furthermore, Atg7 and Beclin1 expressions and p-AMPK were increased while p-mTOR was decreased in livers of HFPS mice in relative to HF and HFNS mice. These results suggest a crosslink between NF-κB signaling pathway and liver AMPK/mTOR/autophagy axis in the context of hepatic steatosis and insulin resistance.
Collapse
|
50
|
Kim Y, Wang W, Okla M, Kang I, Moreau R, Chung S. Suppression of NLRP3 inflammasome by γ-tocotrienol ameliorates type 2 diabetes. J Lipid Res 2015; 57:66-76. [PMID: 26628639 DOI: 10.1194/jlr.m062828] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Indexed: 12/14/2022] Open
Abstract
The Nod-like receptor 3 (NLRP3) inflammasome is an intracellular sensor that sets off the innate immune system in response to microbial-derived and endogenous metabolic danger signals. We previously reported that γ-tocotrienol (γT3) attenuated adipose tissue inflammation and insulin resistance in diet-induced obesity, but the underlying mechanism remained elusive. Here, we investigated the effects of γT3 on NLRP3 inflammasome activation and attendant consequences on type 2 diabetes. γT3 repressed inflammasome activation, caspase-1 cleavage, and interleukin (IL) 1β secretion in murine macrophages, implicating the inhibition of NLRP3 inflammasome in the anti-inflammatory and antipyroptotic properties of γT3. Furthermore, supplementation of leptin-receptor KO mice with γT3 attenuated immune cell infiltration into adipose tissue, decreased circulating IL-18 levels, preserved pancreatic β-cells, and improved insulin sensitivity. Mechanistically, γT3 regulated the NLRP3 inflammasome via a two-pronged mechanism: 1) the induction of A20/TNF-α interacting protein 3 leading to the inhibition of the TNF receptor-associated factor 6/nuclear factor κB pathway and 2) the activation of AMP-activated protein kinase/autophagy axis leading to the attenuation of caspase-1 cleavage. Collectively, we demonstrated, for the first time, that γT3 inhibits the NLRP3 inflammasome thereby delaying the progression of type 2 diabetes. This study also provides an insight into the novel therapeutic values of γT3 for treating NLRP3 inflammasome-associated chronic diseases.
Collapse
Affiliation(s)
- Yongeun Kim
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583
| | - Wei Wang
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583
| | - Meshail Okla
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583
| | - Inhae Kang
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583
| | - Regis Moreau
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583
| | - Soonkyu Chung
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583
| |
Collapse
|