1
|
Wu TX, Pang HZ, Liu XD, Liu L, Tang YF, Luo XF, Ran XK. Adiponectin alleviates inflammatory response in metabolic dysfunction-associated steatohepatitis by inhibiting NLRP3 inflammasome-mediated hepatocyte pyroptosis. Hepatobiliary Pancreat Dis Int 2025:S1499-3872(25)00061-X. [PMID: 40307114 DOI: 10.1016/j.hbpd.2025.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 04/10/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND Activation of NLRP3 (NOD-, LRR- and pyrin domain-containing protein 3) inflammasomes induced by pyroptosis is crucial in metabolic dysfunction-associated steatohepatitis (MASH) progression. Adiponectin possesses an anti-inflammatory role in various liver diseases. This study aimed to evaluate the effects of adiponectin on MASH. METHODS Adiponectin-mediated anti-inflammatory mechanisms, effects on pyroptosis-related proteins, and activation of NLRP3 inflammasomes were investigated using methionine-choline-deficient (MCD)-induced MASH murine model and in vitro models. The degree of MASH inflammation in liver tissue of C57BL/6J mice was assessed using histopathology. Enzyme-linked immunosorbent assay was performed to measure levels of inflammatory factors [interleukin-18 (IL-18), IL-1β, and tumor necrosis factor-α (TNF-α)] in mice serum and culture medium. Western blot and quantitative polymerase chain reaction were performed to analyze the expression of pyroptosis-related genes and proteins in liver tissues of mouse model and in vitro models. Macrophage recruitment in vitro was evaluated using co-culture of upper and lower chambers. RESULTS MASH developed in MCD diet mice [metabolic dysfunction-associated steatotic liver disease (MASLD) activity score = 6] but not in methionine-choline-sufficient (MCS) diet mice (MASLD activity score = 3). Compared to MCS-fed mice, MCD-fed mice showed increased serum levels of aspartate aminotransferase, IL-18, IL-1β, and TNF-α and higher MASLD activity score (P < 0.001). Adiponectin inhibited these increases (P < 0.05) and suppressed mRNA and protein levels of NLRP3, gasdermin-D (GSDMD), and GSDMD-N in liver tissues (P < 0.05). In vitro, lipopolysaccharide (LPS)/palmitic acid (PA) increased the levels of IL-18, IL-1β, and TNF-α, mRNA expressions of CASP1 and GSDMD, and production of CASP1, NLRP3, GSDMD, and GSDMD-N (P < 0.01). Adiponectin reduced the levels of these inflammatory factors and downregulated the mRNA expression and protein generation of pyroptosis-related markers (P < 0.05). HepG2 cells pretreated with LPS/PA recruited more J774A.1 cells (P < 0.001) and increased inflammatory factor secretion by J774A.1 cells (P < 0.001). Adiponectin inhibited this recruitment and reduced inflammatory factor secretion (P < 0.001). CONCLUSIONS Adiponectin inhibits hepatocyte pyroptosis by reducing the production and activation of NLRP3 inflammasomes, CASP1, and GSDMD, thus improving the inflammatory response in MASH and possibly delaying or reversing MASLD progression.
Collapse
Affiliation(s)
- Tie-Xiong Wu
- Graduate School, Guangxi University of Chinese Medicine, Nanning 530000, China; Department of Hepatobiliary, the Third Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou 350108, China
| | - Hua-Zhen Pang
- Department of Hepatology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning 530000, China; Department of Gastroenterology, Liuzhou Traditional Chinese Medical Hospital, Liuzhou 545001, China
| | - Xu-Dong Liu
- Graduate School, Guangxi University of Chinese Medicine, Nanning 530000, China; Department of Hepatology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning 530000, China.
| | - Li Liu
- Department of Hepatology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning 530000, China
| | - Yan-Fang Tang
- Department of Hepatology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning 530000, China
| | - Xue-Fei Luo
- Department of Hepatology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning 530000, China
| | - Xiao-Ke Ran
- Graduate School, Guangxi University of Chinese Medicine, Nanning 530000, China
| |
Collapse
|
2
|
Verlinden SF. The genetic advantage of healthy centenarians: unraveling the central role of NLRP3 in exceptional healthspan. FRONTIERS IN AGING 2024; 5:1452453. [PMID: 39301197 PMCID: PMC11410711 DOI: 10.3389/fragi.2024.1452453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/19/2024] [Indexed: 09/22/2024]
Abstract
Despite extensive research into extending human healthspan (HS) and compressing morbidity, the mechanisms underlying aging remain elusive. However, a better understanding of the genetic advantages responsible for the exceptional HS of healthy centenarians (HC), who live in good physical and mental health for one hundred or more years, could lead to innovative health-extending strategies. This review explores the role of NLRP3, a critical component of innate immunity that significantly impacts aging. It is activated by pathogen-associated signals and self-derived signals that increase with age, leading to low-grade inflammation implicated in age-related diseases. Furthermore, NLRP3 functions upstream in several molecular aging pathways, regulates cellular senescence, and may underlie the robust health observed in HC. By targeting NLRP3, mice exhibit a phenotype akin to that of HC, the HS of monkeys is extended, and aging symptoms are reversed in humans. Thus, targeting NLRP3 could offer a promising approach to extend HS. Additionally, a paradigm shift is proposed. Given that the HS of the broader population is 30 years shorter than that of HC, it is postulated that they suffer from a form of accelerated aging. The term 'auto-aging' is suggested to describe accelerated aging driven by NLRP3.
Collapse
|
3
|
Zou Y, Tang X, Yang S, Chen Z, Liu B, Zhou Z, Peng X, Tang C. New insights into the function of the NLRP3 inflammasome in sarcopenia: mechanism and therapeutic strategies. Metabolism 2024; 158:155972. [PMID: 38972476 DOI: 10.1016/j.metabol.2024.155972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 07/09/2024]
Abstract
Sarcopenia is one of the most common skeletal muscle disorders and is characterized by infirmity and disability. While extensive research has focused on elucidating the mechanisms underlying the progression of sarcopenia, further comprehensive insights into its pathogenesis are necessary to identify new preventive and therapeutic approaches. The involvement of inflammasomes in sarcopenia is widely recognized, with particular emphasis on the NLRP3 (NLR family pyrin domain containing 3) inflammasome. In this review, we aim to elucidate the underlying mechanisms of the NLRP3 inflammasome and its relevance in sarcopenia of various etiologies. Furthermore, we highlight interventions targeting the NLRP3 inflammasome in the context of sarcopenia and discuss the current limitations of our knowledge in this area.
Collapse
Affiliation(s)
- Yunyi Zou
- State Key Laboratory of Developmental Biology of Freshwater Fish, Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Xiangbin Tang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Siyuan Yang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Zhanglin Chen
- State Key Laboratory of Developmental Biology of Freshwater Fish, Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Bin Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Zuoqiong Zhou
- State Key Laboratory of Developmental Biology of Freshwater Fish, Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Xiyang Peng
- State Key Laboratory of Developmental Biology of Freshwater Fish, Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China.
| | - Changfa Tang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China.
| |
Collapse
|
4
|
Pan S, Wang Y, Zhang Y, Ma X, Peng J, Li F, Qian W, Zong J. The Relationship Between the Serum NLRP3 and Adiponectin Levels and Coronary Lesions in Patients with Unstable Angina with Type 2 Diabetes. Clin Interv Aging 2024; 19:1301-1308. [PMID: 39050520 PMCID: PMC11268749 DOI: 10.2147/cia.s467291] [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] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 07/10/2024] [Indexed: 07/27/2024] Open
Abstract
Objective To investigate the Levels of Nucleotide-binding, leucine-rich repeat and pyrin domain-containing protein 3 (NLRP3) and Adiponectin (APN) and their relationship with the severity of coronary artery disease in patients with Unstable Angina (UA) and Type 2 Diabetes (T2D). Methods Two hundred and thirty-one patients with UA were diagnosed by CAG in the Department of Cardiology of the Affiliated Hospital of Xuzhou Medical University from July 2022 to May 2023 were included, and 74 healthy subjects were included as the control group. The levels of NLRP3 and APN in each group were detected by ELISA and the Gensini score in each patient according to the results of CAG. The correlations between NLRP3, APN, and Gensini score were analyzed. According to whether complicated with T2D or not, we further analyze the effect of NLRP3 and APN levels of patients with UA and T2D on the severity of coronary artery stenosis. Results The levels of NLRP3 in UA with T2D group were the highest, followed by simple UA group, and the lowest in the control group, and the level of APN was the opposite. Spearman Correlation analysis showed that the level of NLRP3 was positively correlated with Gensini score (ρ1=0.688, P<0.05) and the level of APN was negatively associated with Gensini score (ρ2= -0.515, P<0.05). There was a negative correlation between NLRP3 and the level of APN (ρ3= -0.366, P<0.05). High NLRP3 and low APN levels are the risk factors for atherosclerosis. Conclusion The NLRP3 and APN were abnormally expressed in patients with UA complicated with T2D. With the aggravation of atherosclerosis, the level of NLRP3 increased and the level of APN decreased.
Collapse
Affiliation(s)
- Siyu Pan
- Institute of Cardiovascular Diseases, Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Yixiao Wang
- Institute of Cardiovascular Diseases, Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Yuchen Zhang
- Institute of Cardiovascular Diseases, Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Xiaoyu Ma
- Institute of Cardiovascular Diseases, Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Jingfeng Peng
- Institute of Cardiovascular Diseases, Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Fangfang Li
- Institute of Cardiovascular Diseases, Xuzhou Medical University, Xuzhou, People’s Republic of China
- Department of Cardiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Wenhao Qian
- Institute of Cardiovascular Diseases, Xuzhou Medical University, Xuzhou, People’s Republic of China
- Department of Cardiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Jing Zong
- Institute of Cardiovascular Diseases, Xuzhou Medical University, Xuzhou, People’s Republic of China
- Department of Cardiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, People’s Republic of China
| |
Collapse
|
5
|
Olie CS, O'Brien DP, Jones HB, Liang Z, Damianou A, Sur-Erdem I, Pinto-Fernández A, Raz V, Kessler BM. Deubiquitinases in muscle physiology and disorders. Biochem Soc Trans 2024; 52:1085-1098. [PMID: 38716888 PMCID: PMC11346448 DOI: 10.1042/bst20230562] [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: 11/14/2023] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 06/27/2024]
Abstract
In vivo, muscle and neuronal cells are post-mitotic, and their function is predominantly regulated by proteostasis, a multilayer molecular process that maintains a delicate balance of protein homeostasis. The ubiquitin-proteasome system (UPS) is a key regulator of proteostasis. A dysfunctional UPS is a hallmark of muscle ageing and is often impacted in neuromuscular disorders (NMDs). Malfunction of the UPS often results in aberrant protein accumulation which can lead to protein aggregation and/or mis-localization affecting its function. Deubiquitinating enzymes (DUBs) are key players in the UPS, controlling protein turnover and maintaining the free ubiquitin pool. Several mutations in DUB encoding genes are linked to human NMDs, such as ATXN3, OTUD7A, UCHL1 and USP14, whilst other NMDs are associated with dysregulation of DUB expression. USP5, USP9X and USP14 are implicated in synaptic transmission and remodeling at the neuromuscular junction. Mice lacking USP19 show increased maintenance of lean muscle mass. In this review, we highlight the involvement of DUBs in muscle physiology and NMDs, particularly in processes affecting muscle regeneration, degeneration and inflammation following muscle injury. DUBs have recently garnered much respect as promising drug targets, and their roles in muscle maturation, regeneration and degeneration may provide the framework for novel therapeutics to treat muscular disorders including NMDs, sarcopenia and cachexia.
Collapse
Affiliation(s)
- Cyriel S. Olie
- Department of Human Genetics, Leiden University Medical Centre, 2333ZC Leiden, The Netherlands
| | - Darragh P. O'Brien
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, U.K
| | - Hannah B.L. Jones
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, U.K
| | - Zhu Liang
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, U.K
- Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford OX3 7FZ, U.K
| | - Andreas Damianou
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, U.K
- Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford OX3 7FZ, U.K
| | - Ilknur Sur-Erdem
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, U.K
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford OX3 9DU, U.K
| | - Adán Pinto-Fernández
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, U.K
- Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford OX3 7FZ, U.K
| | - Vered Raz
- Department of Human Genetics, Leiden University Medical Centre, 2333ZC Leiden, The Netherlands
| | - Benedikt M. Kessler
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, U.K
- Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford OX3 7FZ, U.K
| |
Collapse
|
6
|
Gandhi S, Sweeney G, Perry CGR. Recent Advances in Pre-Clinical Development of Adiponectin Receptor Agonist Therapies for Duchenne Muscular Dystrophy. Biomedicines 2024; 12:1407. [PMID: 39061981 PMCID: PMC11274162 DOI: 10.3390/biomedicines12071407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/03/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is caused by genetic mutations in the cytoskeletal-sarcolemmal anchor protein dystrophin. Repeated cycles of sarcolemmal tearing and repair lead to a variety of secondary cellular and physiological stressors that are thought to contribute to weakness, atrophy, and fibrosis. Collectively, these stressors can contribute to a pro-inflammatory milieu in locomotor, cardiac, and respiratory muscles. Given the many unwanted side effects that accompany current anti-inflammatory steroid-based approaches for treating DMD (e.g., glucocorticoids), there is a need to develop new therapies that address inflammation and other cellular dysfunctions. Adiponectin receptor (AdipoR) agonists, which stimulate AdipoR1 and R2 isoforms on various cell types, have emerged as therapeutic candidates for DMD due to their anti-inflammatory, anti-fibrotic, and pro-myogenic properties in pre-clinical human and rodent DMD models. Although these molecules represent a new direction for therapeutic intervention, the mechanisms through which they elicit their beneficial effects are not yet fully understood, and DMD-specific data is limited. The overarching goal of this review is to investigate how adiponectin signaling may ameliorate pathology associated with dystrophin deficiency through inflammatory-dependent and -independent mechanisms and to determine if current data supports their future progression to clinical trials.
Collapse
Affiliation(s)
- Shivam Gandhi
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, ON M3J 1P3, Canada;
| | - Gary Sweeney
- Department of Biology and Muscle Health Research Centre, York University, Toronto, ON M3J 1P3, Canada;
| | - Christopher G. R. Perry
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, ON M3J 1P3, Canada;
| |
Collapse
|
7
|
Livshits G, Kalinkovich A. Restoration of epigenetic impairment in the skeletal muscle and chronic inflammation resolution as a therapeutic approach in sarcopenia. Ageing Res Rev 2024; 96:102267. [PMID: 38462046 DOI: 10.1016/j.arr.2024.102267] [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: 11/20/2023] [Revised: 02/17/2024] [Accepted: 03/06/2024] [Indexed: 03/12/2024]
Abstract
Sarcopenia is an age-associated loss of skeletal muscle mass, strength, and function, accompanied by severe adverse health outcomes, such as falls and fractures, functional decline, high health costs, and mortality. Hence, its prevention and treatment have become increasingly urgent. However, despite the wide prevalence and extensive research on sarcopenia, no FDA-approved disease-modifying drugs exist. This is probably due to a poor understanding of the mechanisms underlying its pathophysiology. Recent evidence demonstrate that sarcopenia development is characterized by two key elements: (i) epigenetic dysregulation of multiple molecular pathways associated with sarcopenia pathogenesis, such as protein remodeling, insulin resistance, mitochondria impairments, and (ii) the creation of a systemic, chronic, low-grade inflammation (SCLGI). In this review, we focus on the epigenetic regulators that have been implicated in skeletal muscle deterioration, their individual roles, and possible crosstalk. We also discuss epidrugs, which are the pharmaceuticals with the potential to restore the epigenetic mechanisms deregulated in sarcopenia. In addition, we discuss the mechanisms underlying failed SCLGI resolution in sarcopenia and the potential application of pro-resolving molecules, comprising specialized pro-resolving mediators (SPMs) and their stable mimetics and receptor agonists. These compounds, as well as epidrugs, reveal beneficial effects in preclinical studies related to sarcopenia. Based on these encouraging observations, we propose the combination of epidrugs with SCLI-resolving agents as a new therapeutic approach for sarcopenia that can effectively attenuate of its manifestations.
Collapse
Affiliation(s)
- Gregory Livshits
- Department of Morphological Sciences, Adelson School of Medicine, Ariel University, Ariel 4077625, Israel; Department of Anatomy and Anthropology, Faculty of Medical and Health Sciences, School of Medicine, Tel-Aviv University, Tel-Aviv 6905126, Israel.
| | - Alexander Kalinkovich
- Department of Anatomy and Anthropology, Faculty of Medical and Health Sciences, School of Medicine, Tel-Aviv University, Tel-Aviv 6905126, Israel
| |
Collapse
|
8
|
Slavin MB, Khemraj P, Hood DA. Exercise, mitochondrial dysfunction and inflammasomes in skeletal muscle. Biomed J 2024; 47:100636. [PMID: 37499756 PMCID: PMC10828562 DOI: 10.1016/j.bj.2023.100636] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/18/2023] [Accepted: 07/20/2023] [Indexed: 07/29/2023] Open
Abstract
In the broad field of inflammation, skeletal muscle is a tissue that is understudied. Yet it represents about 40% of body mass in non-obese individuals and is therefore of fundamental importance for whole body metabolism and health. This article provides an overview of the unique features of skeletal muscle tissue, as well as its adaptability to exercise. This ability to adapt, particularly with respect to mitochondrial content and function, confers a level of metabolic "protection" against energy consuming events, and adds a measure of quality control that determines the phenotypic response to stress. Thus, we describe the particular role of mitochondria in promoting inflammasome activation in skeletal muscle, contributing to muscle wasting and dysfunction in aging, disuse and metabolic disease. We will then discuss how exercise training can be anti-inflammatory, mitigating the chronic inflammation that is observed in these conditions, potentially through improvements in mitochondrial quality and function.
Collapse
Affiliation(s)
- Mikhaela B Slavin
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, ON, M3J 1P3, Canada
| | - Priyanka Khemraj
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, ON, M3J 1P3, Canada
| | - David A Hood
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, ON, M3J 1P3, Canada.
| |
Collapse
|
9
|
Antuña E, Potes Y, Baena-Huerta FJ, Cachán-Vega C, Menéndez-Coto N, Álvarez Darriba E, Fernández-Fernández M, Burgos Bencosme N, Bermúdez M, López Álvarez EM, Gutiérrez-Rodríguez J, Boga JA, Caballero B, Vega-Naredo I, Coto-Montes A, Garcia-Gonzalez C. NLRP3 Contributes to Sarcopenia Associated to Dependency Recapitulating Inflammatory-Associated Muscle Degeneration. Int J Mol Sci 2024; 25:1439. [PMID: 38338718 PMCID: PMC10855188 DOI: 10.3390/ijms25031439] [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: 12/21/2023] [Revised: 01/17/2024] [Accepted: 01/21/2024] [Indexed: 02/12/2024] Open
Abstract
Sarcopenia, a complex and debilitating condition characterized by progressive deterioration of skeletal muscle, is the primary cause of age-associated disability and significantly impacts healthspan in elderly patients. Despite its prevalence among the aging population, the underlying molecular mechanisms are still under investigation. The NLRP3 inflammasome is crucial in the innate immune response and has a significant impact on diseases related to inflammation and aging. Here, we investigated the expression of the NLRP3 inflammasome pathway and pro-inflammatory cytokines in skeletal muscle and peripheral blood of dependent and independent patients who underwent hip surgery. Patients were categorized into independent and dependent individuals based on their Barthel Index. The expression of NLRP3 inflammasome components was significantly upregulated in sarcopenic muscle from dependent patients, accompanied by higher levels of Caspase-1, IL-1β and IL-6. Among older dependent individuals with sarcopenia, there was a significant increase in the MYH3/MYH2 ratio, indicating a transcriptional shift in expression from mature to developmental myosin isoforms. Creatine kinase levels and senescence markers were also higher in dependent patients, altogether resembling dystrophic diseases and indicating muscle degeneration. In summary, we present evidence for the involvement of the NLRP3/ASC/NEK7/Caspase-1 inflammasome pathway with activation of pro-inflammatory SASP in the outcome of sarcopenia in the elderly.
Collapse
Affiliation(s)
- Eduardo Antuña
- Research Group OSKAR, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Department of Morphology and Cell Biology, University of Oviedo, 33006 Oviedo, Spain
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33006 Oviedo, Spain
| | - Yaiza Potes
- Research Group OSKAR, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Department of Morphology and Cell Biology, University of Oviedo, 33006 Oviedo, Spain
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33006 Oviedo, Spain
| | | | - Cristina Cachán-Vega
- Research Group OSKAR, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Department of Morphology and Cell Biology, University of Oviedo, 33006 Oviedo, Spain
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33006 Oviedo, Spain
| | - Nerea Menéndez-Coto
- Department of Morphology and Cell Biology, University of Oviedo, 33006 Oviedo, Spain
| | | | | | | | - Manuel Bermúdez
- Research Group OSKAR, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Geriatric Service, Monte Naranco Hospital, 33012 Oviedo, Spain
| | - Eva María López Álvarez
- Research Group OSKAR, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Geriatric Service, Monte Naranco Hospital, 33012 Oviedo, Spain
| | - José Gutiérrez-Rodríguez
- Research Group OSKAR, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Geriatric Service, Monte Naranco Hospital, 33012 Oviedo, Spain
| | - José Antonio Boga
- Grupo de Investigación Microbiología Traslacional, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Servicio de Microbiología, Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Spain
| | - Beatriz Caballero
- Research Group OSKAR, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Department of Morphology and Cell Biology, University of Oviedo, 33006 Oviedo, Spain
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33006 Oviedo, Spain
| | - Ignacio Vega-Naredo
- Research Group OSKAR, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Department of Morphology and Cell Biology, University of Oviedo, 33006 Oviedo, Spain
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33006 Oviedo, Spain
| | - Ana Coto-Montes
- Research Group OSKAR, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Department of Morphology and Cell Biology, University of Oviedo, 33006 Oviedo, Spain
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33006 Oviedo, Spain
| | - Claudia Garcia-Gonzalez
- Research Group OSKAR, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Department of Morphology and Cell Biology, University of Oviedo, 33006 Oviedo, Spain
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33006 Oviedo, Spain
| |
Collapse
|
10
|
Granata S, La Russa D, Stallone G, Perri A, Zaza G. Inflammasome pathway in kidney transplantation. Front Med (Lausanne) 2023; 10:1303110. [PMID: 38020086 PMCID: PMC10663322 DOI: 10.3389/fmed.2023.1303110] [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: 09/27/2023] [Accepted: 10/25/2023] [Indexed: 12/01/2023] Open
Abstract
Kidney transplantation is the best available renal replacement therapy for patients with end-stage kidney disease and is associated with better quality of life and patient survival compared with dialysis. However, despite the significant technical and pharmaceutical advances in this field, kidney transplant recipients are still characterized by reduced long-term graft survival. In fact, almost half of the patients lose their allograft after 15-20 years. Most of the conditions leading to graft loss are triggered by the activation of a large immune-inflammatory machinery. In this context, several inflammatory markers have been identified, and the deregulation of the inflammasome (NLRP3, NLRP1, NLRC4, AIM2), a multiprotein complex activated by either whole pathogens (including fungi, bacteria, and viruses) or host-derived molecules, seems to play a pivotal pathogenetic role. However, the biological mechanisms leading to inflammasome activation in patients developing post-transplant complications (including, ischemia-reperfusion injury, rejections, infections) are still largely unrecognized, and only a few research reports, reviewed in this manuscript, have addressed the association between abnormal activation of this pathway and the onset/development of major clinical effects. Finally, the regulation of the inflammasome machinery could represent in future a valuable therapeutic target in kidney transplantation.
Collapse
Affiliation(s)
- Simona Granata
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Daniele La Russa
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Giovanni Stallone
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Anna Perri
- Department of Experimental and Clinical Medicine, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Gianluigi Zaza
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| |
Collapse
|
11
|
Sunthankar SD, George-Durrett K, Crum K, Slaughter JC, Kasten J, Raucci FJ, Markham LW, Soslow JH. Comprehensive cardiac magnetic resonance T1, T2, and extracellular volume mapping to define Duchenne cardiomyopathy. J Cardiovasc Magn Reson 2023; 25:44. [PMID: 37517994 PMCID: PMC10388519 DOI: 10.1186/s12968-023-00951-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 06/30/2023] [Indexed: 08/01/2023] Open
Abstract
BACKGROUND Cardiomyopathy is the leading cause of death in Duchenne muscular dystrophy (DMD). Cardiac magnetic resonance (CMR) parametric mapping sequences offer insights into disease pathophysiology. We propose a novel approach by leveraging T2 mapping in conjunction with T1 and extracellular volume (ECV) mapping to perform a virtual myocardial biopsy. While previous work has attempted to describe myocardial changes in DMD, our inclusion of T2 mapping enables comprehensive categorization of myocardial tissue characteristics of fibrosis, edema, and fat to better understand the pathological composition of the myocardium with disease progression. METHODS DMD patients (n = 49; median: 12 years-old) underwent CMR, including T1, T2, and ECV. Categories were defined as normal, isolated high T1 (normal ECV, high T1, normal T2), fibrosis (high ECV, normal or high T1, normal T2), edema (normal or high ECV, normal or high T1, high T2), fat (normal ECV, low T1, high T2) or fibrofatty (high ECV, low T1, high T2). RESULTS Median left ventricular ejection fraction (LVEF) was 59% with 27% having LVEF < 55%. Those with normal LVEF and no late gadolinium enhancement (37%) were younger in age (10.5 ± 2.6 vs. 15.0 ± 4.3 years-old, p < 0.001). Native T1 was elevated in at least one slice in 82% of patients. Those with high T2 at any slice (27%) were older (p = 0.005) and had lower LVEF (p = 0.005) compared with subjects with normal T2 (73%). The most common myocardial characterization was fibrosis (43%) followed by isolated high T1 (24%). Of the 13 with high T2, ten were categorized as edema, two as fibrofatty, and one as fat. CONCLUSION CMR parametric mapping sequences offer insights into Duchenne cardiomyopathy pathophysiology, which should drive development of therapeutic interventions aimed at these targets. Myocardial fibrosis is common in DMD. Patients with elevated T2 were older and had lower LVEF. Though fat infiltration was present, the majority of subjects with elevated T2 met criteria for myocardial edema.
Collapse
Affiliation(s)
- Sudeep D Sunthankar
- Thomas P. Graham Jr Division of Pediatric Cardiology, Department of Pediatrics, Vanderbilt University Medical Center, Monroe Carell Jr Children's Hospital at Vanderbilt, 2220 Children's Way, Suite 5230, TN, 37232, Nashville, USA.
| | - Kristen George-Durrett
- Thomas P. Graham Jr Division of Pediatric Cardiology, Department of Pediatrics, Vanderbilt University Medical Center, Monroe Carell Jr Children's Hospital at Vanderbilt, 2220 Children's Way, Suite 5230, TN, 37232, Nashville, USA
| | - Kimberly Crum
- Thomas P. Graham Jr Division of Pediatric Cardiology, Department of Pediatrics, Vanderbilt University Medical Center, Monroe Carell Jr Children's Hospital at Vanderbilt, 2220 Children's Way, Suite 5230, TN, 37232, Nashville, USA
| | - James C Slaughter
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jennifer Kasten
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Frank J Raucci
- Division of Pediatric Cardiology, Department of Pediatrics, Children's Hospital of Richmond at Virginia Commonwealth University Health System, Richmond, VA, 23219, USA
| | - Larry W Markham
- Division of Cardiology, Department of Pediatrics, Riley Hospital for Children at Indiana University Health, Indianapolis, IN, 46202, USA
| | - Jonathan H Soslow
- Thomas P. Graham Jr Division of Pediatric Cardiology, Department of Pediatrics, Vanderbilt University Medical Center, Monroe Carell Jr Children's Hospital at Vanderbilt, 2220 Children's Way, Suite 5230, TN, 37232, Nashville, USA
| |
Collapse
|
12
|
Kummer K, Bertram I, Zechel S, Hoffmann DB, Schmidt J. Inflammasome in Skeletal Muscle: NLRP3 Is an Inflammatory Cell Stress Component in Inclusion Body Myositis. Int J Mol Sci 2023; 24:10675. [PMID: 37445853 DOI: 10.3390/ijms241310675] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/30/2023] [Accepted: 06/04/2023] [Indexed: 07/15/2023] Open
Abstract
Inclusion body myositis (IBM) is a chronic, mostly treatment-resistant, inflammatory myopathy with a pathology that centers around specific interactions between inflammation and protein accumulation. The study aimed to identify the inflammasome as a key event in the complex network of pathomechanisms. Regulation of the inflammasome was assessed in a well-established pro-inflammatory cell culture model using human myoblasts and primary human myotubes. By quantitative PCR, western blot and immunocytochemistry, inflammasome markers including NLRP3 were assessed in muscle cells exposed to the cytokines IL-1β and IFN-γ. The data were corroborated by analysis of muscle biopsies from patients with IBM compared to other myositis subtypes. In the cell culture model of IBM, the NLRP3 inflammasome was significantly overexpressed, as evidenced by western blot (p = 0.03) and quantitative PCR (p < 0.01). Target genes that play a role in inflammasome assembly, T-cell migration, and MHC-I expression (p = 0.009) were highly co-upregulated. NLRP3 was significantly overexpressed in muscle biopsies from IBM samples compared to disease controls (p = 0.049), including other inflammatory myopathies. Due to the extraordinary features of the pathogenesis and the pronounced upregulation of NLRP3 in IBM, the inflammasome could serve as a key molecule that drives the inflammatory cascade as well as protein accumulation in the muscle. These data can be useful for future therapeutic developments.
Collapse
Affiliation(s)
- Karsten Kummer
- Department of Neurology and Pain Treatment, Neuromuscular Center, Center for Translational Medicine, Immanuel Klinik Rüdersdorf, University Hospital of the Brandenburg Medical School, 15562 Rüdersdorf bei Berlin, Germany
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School, 15562 Rüdersdorf bei Berlin, Germany
- Department of Neurology, Neuromuscular Center, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Imke Bertram
- Department of Neurology, Neuromuscular Center, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Sabrina Zechel
- Department of Neuropathology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Daniel B Hoffmann
- Department of Trauma, Orthopaedic and Plastic Surgery, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Jens Schmidt
- Department of Neurology and Pain Treatment, Neuromuscular Center, Center for Translational Medicine, Immanuel Klinik Rüdersdorf, University Hospital of the Brandenburg Medical School, 15562 Rüdersdorf bei Berlin, Germany
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School, 15562 Rüdersdorf bei Berlin, Germany
- Department of Neurology, Neuromuscular Center, University Medical Center Göttingen, 37075 Göttingen, Germany
| |
Collapse
|
13
|
Esposito P, Verzola D, Saio M, Picciotto D, Frascio M, Laudon A, Zanetti V, Brunori G, Garibotto G, Viazzi F. The Contribution of Muscle Innate Immunity to Uremic Cachexia. Nutrients 2023; 15:2832. [PMID: 37447158 PMCID: PMC10343562 DOI: 10.3390/nu15132832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/15/2023] [Accepted: 06/16/2023] [Indexed: 07/15/2023] Open
Abstract
Protein energy wasting (PEW) is a common complication both in chronic kidney disease (CKD) and end-stage kidney disease (ESKD). Of note, PEW is one of the stronger predictors of morbidity and mortality in this patient population. The pathogenesis of PEW involves several mechanisms, including anorexia, insulin resistance, acidosis and low-grade inflammation. In addition, "sterile" muscle inflammation contributes to PEW at an advanced CKD stage. Both immune and resident muscle cells can activate innate immunity; thus, they have critical roles in triggering "sterile" tissue inflammation. Toll-like receptor 4 (TLR4) can detect endogenous danger-associated molecular patterns generated or retained in blood in uremia and induce a sterile muscle inflammatory response via NF-κB in myocytes. In addition, TLR4, though the activation of the NLRP3 inflammasome, links the sensing of metabolic uremic stress in muscle to the activation of pro-inflammatory cascades, which lead to the production of IL-1β and IL-18. Finally, uremia-induced accelerated cell senescence is associated with a secretory phenotype that favors fibrosis in muscle. Targeting these innate immune pathways could lead to novel therapies for CKD-related PEW.
Collapse
Affiliation(s)
- Pasquale Esposito
- Division of Nephrology, Dialysis and Transplantation, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (P.E.); (M.S.); (D.P.); (V.Z.); (F.V.)
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy;
| | - Daniela Verzola
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy;
| | - Michela Saio
- Division of Nephrology, Dialysis and Transplantation, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (P.E.); (M.S.); (D.P.); (V.Z.); (F.V.)
| | - Daniela Picciotto
- Division of Nephrology, Dialysis and Transplantation, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (P.E.); (M.S.); (D.P.); (V.Z.); (F.V.)
| | - Marco Frascio
- Division of Surgery, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
- Department of Surgical Sciences and Integrated Diagnostics, University of Genova, 16132 Genova, Italy
| | - Alessandro Laudon
- Division of Nephrology, Ospedale Santa Chiara, 38122 Trento, Italy; (A.L.); (G.B.)
| | - Valentina Zanetti
- Division of Nephrology, Dialysis and Transplantation, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (P.E.); (M.S.); (D.P.); (V.Z.); (F.V.)
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy;
| | - Giuliano Brunori
- Division of Nephrology, Ospedale Santa Chiara, 38122 Trento, Italy; (A.L.); (G.B.)
| | - Giacomo Garibotto
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy;
| | - Francesca Viazzi
- Division of Nephrology, Dialysis and Transplantation, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (P.E.); (M.S.); (D.P.); (V.Z.); (F.V.)
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy;
| |
Collapse
|
14
|
Zabłocki K, Górecki DC. The Role of P2X7 Purinoceptors in the Pathogenesis and Treatment of Muscular Dystrophies. Int J Mol Sci 2023; 24:ijms24119434. [PMID: 37298386 DOI: 10.3390/ijms24119434] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Muscular dystrophies are inherited neuromuscular diseases, resulting in progressive disability and often affecting life expectancy. The most severe, common types are Duchenne muscular dystrophy (DMD) and Limb-girdle sarcoglycanopathy, which cause advancing muscle weakness and wasting. These diseases share a common pathomechanism where, due to the loss of the anchoring dystrophin (DMD, dystrophinopathy) or due to mutations in sarcoglycan-encoding genes (LGMDR3 to LGMDR6), the α-sarcoglycan ecto-ATPase activity is lost. This disturbs important purinergic signaling: An acute muscle injury causes the release of large quantities of ATP, which acts as a damage-associated molecular pattern (DAMP). DAMPs trigger inflammation that clears dead tissues and initiates regeneration that eventually restores normal muscle function. However, in DMD and LGMD, the loss of ecto-ATPase activity, that normally curtails this extracellular ATP (eATP)-evoked stimulation, causes exceedingly high eATP levels. Thus, in dystrophic muscles, the acute inflammation becomes chronic and damaging. The very high eATP over-activates P2X7 purinoceptors, not only maintaining the inflammation but also tuning the potentially compensatory P2X7 up-regulation in dystrophic muscle cells into a cell-damaging mechanism exacerbating the pathology. Thus, the P2X7 receptor in dystrophic muscles is a specific therapeutic target. Accordingly, the P2X7 blockade alleviated dystrophic damage in mouse models of dystrophinopathy and sarcoglycanopathy. Therefore, the existing P2X7 blockers should be considered for the treatment of these highly debilitating diseases. This review aims to present the current understanding of the eATP-P2X7 purinoceptor axis in the pathogenesis and treatment of muscular dystrophies.
Collapse
Affiliation(s)
- Krzysztof Zabłocki
- Laboratory of Cellular Metabolism, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Dariusz C Górecki
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DT, UK
| |
Collapse
|
15
|
Onódi Z, Szabó PL, Kucsera D, Pokreisz P, Dostal C, Hilber K, Oudit GY, Podesser BK, Ferdinandy P, Varga ZV, Kiss A. Inflammasome Activity in the Skeletal Muscle and Heart of Rodent Models for Duchenne Muscular Dystrophy. Int J Mol Sci 2023; 24:8497. [PMID: 37239853 PMCID: PMC10218525 DOI: 10.3390/ijms24108497] [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: 03/25/2023] [Revised: 05/02/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is characterized by wasting of muscles that leads to difficulty moving and premature death, mainly from heart failure. Glucocorticoids are applied in the management of the disease, supporting the hypothesis that inflammation may be driver as well as target. However, the inflammatory mechanisms during progression of cardiac and skeletal muscle dysfunction are still not well characterized. Our objective was to characterize the inflammasomes in myocardial and skeletal muscle in rodent models of DMD. Gastrocnemius and heart samples were collected from mdx mice and DMDmdx rats (3 and 9-10 months). Inflammasome sensors and effectors were assessed by immunoblotting. Histology was used to assess leukocyte infiltration and fibrosis. In gastrocnemius, a tendency towards elevation of gasdermin D irrespective of the age of the animal was observed. The adaptor protein was elevated in the mdx mouse skeletal muscle and heart. Increased cleavage of the cytokines was observed in the skeletal muscle of the DMDmdx rats. Sensor or cytokine expression was not changed in the tissue samples of the mdx mice. In conclusion, inflammatory responses are distinct between the skeletal muscle and heart in relevant models of DMD. Inflammation tends to decrease over time, supporting the clinical observations that the efficacy of anti-inflammatory therapies might be more prominent in the early stage.
Collapse
Affiliation(s)
- Zsófia Onódi
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (Z.O.)
- HCEMM-SE Cardiometabolic Immunology Research Group, Semmelweis University, 1085 Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, 1085 Budapest, Hungary
| | - Petra Lujza Szabó
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Dániel Kucsera
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (Z.O.)
- HCEMM-SE Cardiometabolic Immunology Research Group, Semmelweis University, 1085 Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, 1085 Budapest, Hungary
| | - Péter Pokreisz
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Christopher Dostal
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Karlheinz Hilber
- Department of Neurophysiology & Neuropharmacology, Center for Physiology & Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Gavin Y. Oudit
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Bruno K. Podesser
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (Z.O.)
- Pharmahungary Group, 6728 Szeged, Hungary
| | - Zoltán V. Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (Z.O.)
- HCEMM-SE Cardiometabolic Immunology Research Group, Semmelweis University, 1085 Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, 1085 Budapest, Hungary
| | - Attila Kiss
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
16
|
Gόrecki DC, Rumney RMH. The P2X7 purinoceptor in pathogenesis and treatment of dystrophino- and sarcoglycanopathies. Curr Opin Pharmacol 2023; 69:102357. [PMID: 36842388 DOI: 10.1016/j.coph.2023.102357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/16/2022] [Accepted: 12/29/2022] [Indexed: 02/26/2023]
Abstract
Dystrophinopathy and sarcoglycanopathies are incurable diseases caused by mutations in the genes encoding dystrophin or members of the dystrophin associated protein complex (DAPC). Restoration of the missing dystrophin or sarcoglycans via genetic approaches is complicated by the downsides of personalised medicines and immune responses against re-expressed proteins. Thus, the targeting of disease mechanisms downstream from the mutant protein has a strong translational potential. Acute muscle damage causes release of large quantities of ATP, which activates P2X7 purinoceptors, resulting in inflammation that clears dead tissues and triggers regeneration. However, in dystrophic muscles, loss of α-sarcoglycan ecto-ATPase activity further elevates extracellular ATP (eATP) levels, exacerbating the pathology. Moreover, seemingly compensatory P2X7 upregulation in dystrophic muscle cells, combined with high eATP leads to further damage. Accordingly, P2X7 blockade alleviated dystrophic damage in mouse models of both dystrophinopathy and sarcoglycanopathy. Existing P2X7 blockers could be re-purposed for the treatment of these highly debilitating diseases.
Collapse
Affiliation(s)
- Dariusz C Gόrecki
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, UK.
| | - Robin M H Rumney
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, UK
| |
Collapse
|
17
|
Kalkan H, Pagano E, Paris D, Panza E, Cuozzo M, Moriello C, Piscitelli F, Abolghasemi A, Gazzerro E, Silvestri C, Capasso R, Motta A, Russo R, Di Marzo V, Iannotti FA. Targeting gut dysbiosis against inflammation and impaired autophagy in Duchenne muscular dystrophy. EMBO Mol Med 2023; 15:e16225. [PMID: 36594243 PMCID: PMC9994484 DOI: 10.15252/emmm.202216225] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 01/04/2023] Open
Abstract
Nothing is known about the potential implication of gut microbiota in skeletal muscle disorders. Here, we provide evidence that fecal microbiota composition along with circulating levels of short-chain fatty acids (SCFAs) and related metabolites are altered in the mdx mouse model of Duchenne muscular dystrophy (DMD) compared with healthy controls. Supplementation with sodium butyrate (NaB) in mdx mice rescued muscle strength and autophagy, and prevented inflammation associated with excessive endocannabinoid signaling at CB1 receptors to the same extent as deflazacort (DFZ), the standard palliative care for DMD. In LPS-stimulated C2C12 myoblasts, NaB reduces inflammation, promotes autophagy, and prevents dysregulation of microRNAs targeting the endocannabinoid CB1 receptor gene, in a manner depending on the activation of GPR109A and PPARγ receptors. In sum, we propose a novel disease-modifying approach in DMD that may have benefits also in other muscular dystrophies.
Collapse
Affiliation(s)
- Hilal Kalkan
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, Italy
| | - Ester Pagano
- Department of Pharmacy, University Federico II of Naples, Italy
| | - Debora Paris
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, Italy
| | | | | | - Claudia Moriello
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, Italy
| | - Fabiana Piscitelli
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, Italy
| | - Armita Abolghasemi
- Institut Universitaire de Cardiologie et de Pneumologie de Québec and Institut Sur la Nutrition et Les Aliments Fonctionnels, Centre NUTRISS, Université Laval, Quebec City, QC, Canada
| | - Elisabetta Gazzerro
- Unit of Muscle Research, Experimental and Clinical Research Center Charité Universitätsmedizin and Max Delbrück Research Center, Berlin, Germany
| | - Cristoforo Silvestri
- Institut Universitaire de Cardiologie et de Pneumologie de Québec and Institut Sur la Nutrition et Les Aliments Fonctionnels, Centre NUTRISS, Université Laval, Quebec City, QC, Canada
| | - Raffaele Capasso
- Department of Agricultural Sciences, University of Naples Federico II, Portici, Italy
| | - Andrea Motta
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, Italy
| | - Roberto Russo
- Department of Pharmacy, University Federico II of Naples, Italy
| | - Vincenzo Di Marzo
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, Italy
- Institut Universitaire de Cardiologie et de Pneumologie de Québec and Institut Sur la Nutrition et Les Aliments Fonctionnels, Centre NUTRISS, Université Laval, Quebec City, QC, Canada
| | - Fabio Arturo Iannotti
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, Italy
| |
Collapse
|
18
|
Mahon N, Glennon JC. The Bi-directional Relationship Between Sleep and Inflammation in Muscular Dystrophies: A Narrative Review. Neurosci Biobehav Rev 2023; 150:105116. [PMID: 36870583 DOI: 10.1016/j.neubiorev.2023.105116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 01/31/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023]
Abstract
Muscular dystrophies vary in presentation and severity, but are associated with profound disability in many people. Although characterised by muscle weakness and wasting, there is also a very high prevalence of sleep problems and disorders which have significant impacts on quality of life in these individuals. There are no curative therapies for muscular dystrophies, with the only options for patients being supportive therapies to aid with symptoms. Therefore, there is an urgent need for new therapeutic targets and a greater understanding of pathogenesis. Inflammation and altered immunity are factors which have prominent roles in some muscular dystrophies and emerging roles in others such as type 1 myotonic dystrophy, signifying a link to pathogenesis. Interestingly, there is also a strong link between inflammation/immunity and sleep. In this review, we will explore this link in the context of muscular dystrophies and how it may influence potential therapeutic targets and interventions.
Collapse
Affiliation(s)
- Niamh Mahon
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Jeffrey C Glennon
- School of Medicine, University College Dublin, Dublin, Ireland; UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| |
Collapse
|
19
|
TRIM72 Alleviates Muscle Inflammation in mdx Mice via Promoting Mitophagy-Mediated NLRP3 Inflammasome Inactivation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:8408574. [PMID: 36713032 PMCID: PMC9876702 DOI: 10.1155/2023/8408574] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/15/2022] [Accepted: 01/05/2023] [Indexed: 01/19/2023]
Abstract
Chronic muscle inflammation exacerbates the pathogenesis of Duchenne muscular dystrophy (DMD), which is characterized by progressive muscle degeneration and weakness. NLRP3 (nucleotide-binding domain and leucine-rich repeat pyrin domain containing 3) inflammasome plays a key role in the inflammatory process, and its abnormal activation leads to a variety of inflammatory or immune diseases. TRIM72 (MG53) is a protective myokine for tissue repair and regeneration. However, little is known about the potential impact of TRIM72 in the crosstalk between mitophagy and inflammatory process of DMD. Here, 10-week-old male mdx mice were injected intramuscularly with adeno-associated virus (AAV-TRIM72) to overexpress TRIM72 protein for 6 weeks. Then, skeletal muscle samples were collected, and relevant parameters were measured by histopathological analysis and molecular biology techniques. C2C12 cell line was transfected with lentivirus (LV-TRIM72) to overexpress or siRNA (si-TRIM72) to suppress the TRIM72 expression for the following experiment. Our data firstly showed that the TRIM72 expression was decreased in skeletal muscles of mdx mice. Then, we observed the increased NLRP3 inflammasome and impaired mitophagy in mdx mice compared with wild type mice. In mdx mice, administration of AAV-TRIM72 alleviated the accumulation of NLRP3 inflammasome and the consequent IL-18 and IL1β maturation by inducing autophagy, while this protective effect was reversed by chloroquine. Mitochondrial reactive oxygen species (mtROS), as a recognized activator for NLRP3 inflammasome, was attenuated by TRIM72 through the induction of mitophagy in C2C12 cells. Additionally, we proposed that the TRIM72 overexpression might promote mitophagy through both the early stage by PI3K-AKT pathway and the late stage by autolysosome fusion. In conclusion, the current study suggests that TRIM72 prevents DMD inflammation via decreasing NLRP3 inflammasomes and enhancing mitophagy. Collectively, our study provides insight into TRIM72 as a promising target for therapeutic intervention for DMD.
Collapse
|
20
|
Bencze M. Mechanisms of Myofibre Death in Muscular Dystrophies: The Emergence of the Regulated Forms of Necrosis in Myology. Int J Mol Sci 2022; 24:ijms24010362. [PMID: 36613804 PMCID: PMC9820579 DOI: 10.3390/ijms24010362] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/28/2022] Open
Abstract
Myofibre necrosis is a central pathogenic process in muscular dystrophies (MD). As post-lesional regeneration cannot fully compensate for chronic myofibre loss, interstitial tissue accumulates and impairs muscle function. Muscle regeneration has been extensively studied over the last decades, however, the pathway(s) controlling muscle necrosis remains largely unknown. The recent discovery of several regulated cell death (RCD) pathways with necrotic morphology challenged the dogma of necrosis as an uncontrolled process, opening interesting perspectives for many degenerative disorders. In this review, we focus on how cell death affects myofibres in MDs, integrating the latest research in the cell death field, with specific emphasis on Duchenne muscular dystrophy, the best-known and most common hereditary MD. The role of regulated forms of necrosis in myology is still in its infancy but there is increasing evidence that necroptosis, a genetically programmed form of necrosis, is involved in muscle degenerating disorders. The existence of apoptosis in myofibre demise will be questioned, while other forms of non-apoptotic RCDs may also have a role in myonecrosis, illustrating the complexity and possibly the heterogeneity of the cell death pathways in muscle degenerating conditions.
Collapse
Affiliation(s)
- Maximilien Bencze
- “Biology of the Neuromuscular System” Team, Institut Mondor de Recherche Biomédicale (IMRB), University Paris-Est Créteil, INSERM, U955 IMRB, 94010 Créteil, France;
- École Nationale Vétérinaire d’Alfort, IMRB, 94700 Maisons-Alfort, France
| |
Collapse
|
21
|
Tyagi SC, Pushpakumar S, Sen U, Mokshagundam SPL, Kalra DK, Saad MA, Singh M. COVID-19 Mimics Pulmonary Dysfunction in Muscular Dystrophy as a Post-Acute Syndrome in Patients. Int J Mol Sci 2022; 24:ijms24010287. [PMID: 36613731 PMCID: PMC9820572 DOI: 10.3390/ijms24010287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/12/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Although progressive wasting and weakness of respiratory muscles are the prominent hallmarks of Duchenne muscular dystrophy (DMD) and long-COVID (also referred as the post-acute sequelae of COVID-19 syndrome); however, the underlying mechanism(s) leading to respiratory failure in both conditions remain unclear. We put together the latest relevant literature to further understand the plausible mechanism(s) behind diaphragm malfunctioning in COVID-19 and DMD conditions. Previously, we have shown the role of matrix metalloproteinase-9 (MMP9) in skeletal muscle fibrosis via a substantial increase in the levels of tumor necrosis factor-α (TNF-α) employing a DMD mouse model that was crossed-bred with MMP9-knockout (MMP9-KO or MMP9-/-) strain. Interestingly, recent observations from clinical studies show a robust increase in neopterin (NPT) levels during COVID-19 which is often observed in patients having DMD. What seems to be common in both (DMD and COVID-19) is the involvement of neopterin (NPT). We know that NPT is generated by activated white blood cells (WBCs) especially the M1 macrophages in response to inducible nitric oxide synthase (iNOS), tetrahydrobiopterin (BH4), and tetrahydrofolate (FH4) pathways, i.e., folate one-carbon metabolism (FOCM) in conjunction with epigenetics underpinning as an immune surveillance protection. Studies from our laboratory, and others researching DMD and the genetically engineered humanized (hACE2) mice that were administered with the spike protein (SP) of SARS-CoV-2 revealed an increase in the levels of NPT, TNF-α, HDAC, IL-1β, CD147, and MMP9 in the lung tissue of the animals that were subsequently accompanied by fibrosis of the diaphragm depicting a decreased oscillation phenotype. Therefore, it is of interest to understand how regulatory processes such as epigenetics involvement affect DNMT, HDAC, MTHFS, and iNOS that help generate NPT in the long-COVID patients.
Collapse
Affiliation(s)
- Suresh C. Tyagi
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Sathnur Pushpakumar
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Utpal Sen
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Sri Prakash L. Mokshagundam
- Division of Endocrinology, Metabolism and Diabetes and Robley Rex VA Medical Center, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Dinesh K. Kalra
- Division of Cardiovascular Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Mohamed A. Saad
- Division of Pulmonary, Critical Care and Sleep Disorders Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Mahavir Singh
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
- Correspondence: or
| |
Collapse
|
22
|
Dubuisson N, Versele R, Planchon C, Selvais CM, Noel L, Abou-Samra M, Davis-López de Carrizosa MA. Histological Methods to Assess Skeletal Muscle Degeneration and Regeneration in Duchenne Muscular Dystrophy. Int J Mol Sci 2022; 23:16080. [PMID: 36555721 PMCID: PMC9786356 DOI: 10.3390/ijms232416080] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/09/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive disease caused by the loss of function of the protein dystrophin. This protein contributes to the stabilisation of striated cells during contraction, as it anchors the cytoskeleton with components of the extracellular matrix through the dystrophin-associated protein complex (DAPC). Moreover, absence of the functional protein affects the expression and function of proteins within the DAPC, leading to molecular events responsible for myofibre damage, muscle weakening, disability and, eventually, premature death. Presently, there is no cure for DMD, but different treatments help manage some of the symptoms. Advances in genetic and exon-skipping therapies are the most promising intervention, the safety and efficiency of which are tested in animal models. In addition to in vivo functional tests, ex vivo molecular evaluation aids assess to what extent the therapy has contributed to the regenerative process. In this regard, the later advances in microscopy and image acquisition systems and the current expansion of antibodies for immunohistological evaluation together with the development of different spectrum fluorescent dyes have made histology a crucial tool. Nevertheless, the complexity of the molecular events that take place in dystrophic muscles, together with the rise of a multitude of markers for each of the phases of the process, makes the histological assessment a challenging task. Therefore, here, we summarise and explain the rationale behind different histological techniques used in the literature to assess degeneration and regeneration in the field of dystrophinopathies, focusing especially on those related to DMD.
Collapse
Affiliation(s)
- Nicolas Dubuisson
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium
- Neuromuscular Reference Center, Cliniques Universitaires Saint-Luc (CUSL), Avenue Hippocrate 10, 1200 Brussels, Belgium
| | - Romain Versele
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium
| | - Chloé Planchon
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium
| | - Camille M. Selvais
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium
| | - Laurence Noel
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium
| | - Michel Abou-Samra
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium
| | - María A. Davis-López de Carrizosa
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012 Seville, Spain
| |
Collapse
|
23
|
Analysis of Long Noncoding RNAs-Related Regulatory Mechanisms in Duchenne Muscular Dystrophy Using a Disease-Related lncRNA-mRNA Pathway Network. Genet Res (Camb) 2022; 2022:8548804. [PMID: 36619896 PMCID: PMC9771664 DOI: 10.1155/2022/8548804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 10/25/2022] [Accepted: 12/05/2022] [Indexed: 12/16/2022] Open
Abstract
Objective This study aimed to investigate the molecular regulatory mechanisms underpinning Duchenne muscular dystrophy (DMD). Methods Using microarray data, differentially expressed long noncoding RNAs (DELs) and DMD-related differentially expressed mRNAs (DEMs) were screened based on the comparative toxicogenomics database, using a cutoff of |log2 fold change| > 1 and false discovery rate (FDR) < 0.05. Then, protein-protein interaction (PPI), coexpression network of lncRNA-mRNA, and DMD-related lncRNA-mRNA pathway networks were constructed, and functional analyses of the genes in the network were performed. Finally, the proportions of immune cells infiltrating the muscle tissues in DMD were analyzed, and the correlation between the immune cells and expression of the DELs/DEMs was studied. Results A total of 46 DELs and 313 DMD-related DEMs were identified. The PPI network revealed STAT1, VEGFA, and CCL2 to be the top three hub genes. The DMD-related lncRNA-mRNA pathway network comprising two pathways, nine DELs, and nine DMD-related DEMs showed that PYCARD, RIPK2, and CASP1 were significantly enriched in the NOD-like receptor signaling pathway, whereas MAP2K2, LUM, RPS6, PDCD4, TWIST1, and HIF1A were significantly enriched with proteoglycans in cancers. The nine DELs in this network were DBET, MBNL1-AS1, MIR29B2CHG, CCDC18-AS1, FAM111A-DT, GAS5, LINC01290, ATP2B1-AS1, and PSMB8-AS1. Conclusion The nine DMD-related DEMs and DELs identified in this study may play important roles in the occurrence and progression of DMD through the two pathways of the NOD-like receptor signaling pathway and proteoglycans in cancers.
Collapse
|
24
|
Dubuisson N, Davis-López de Carrizosa MA, Versele R, Selvais CM, Noel L, Van den Bergh PYD, Brichard SM, Abou-Samra M. Inhibiting the inflammasome with MCC950 counteracts muscle pyroptosis and improves Duchenne muscular dystrophy. Front Immunol 2022; 13:1049076. [PMID: 36569900 PMCID: PMC9770793 DOI: 10.3389/fimmu.2022.1049076] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is the most common inherited human myopathy. Typically, the secondary process involving severe inflammation and necrosis exacerbate disease progression. Previously, we reported that the NLRP3 inflammasome complex plays a crucial role in this disorder. Moreover, pyroptosis, a form of programmed necrotic cell death, is triggered by NLRP3 via gasdermin D (GSDMD). So far, pyroptosis has never been described either in healthy muscle or in dystrophic muscle. The aim of this study was to unravel the role of NLRP3 inflammasome in DMD and explore a potentially promising treatment with MCC950 that selectively inhibits NLRP3. Methods Four-week-old mdx mice (n=6 per group) were orally treated for 2 months with MCC950 (mdx-T), a highly potent, specific, small-molecule inhibitor of NLRP3, and compared with untreated (mdx) and wild-type (WT) mice. In vivo functional tests were carried out to measure the global force and endurance of mice. Ex vivo biochemical and molecular analyses were performed to evaluate the pathophysiology of the skeletal muscle. Finally, in vitro tests were conducted on primary cultures of DMD human myotubes. Results After MCC950 treatment, mdx mice exhibited a significant reduction of inflammation, macrophage infiltration and oxidative stress (-20 to -65%, P<0.05 vs untreated mdx). Mdx-T mice displayed considerably less myonecrosis (-54%, P<0.05 vs mdx) and fibrosis (-75%, P<0.01 vs mdx). Moreover, a more mature myofibre phenotype, characterized by larger-sized fibres and higher expression of mature myosin heavy chains 1 and 7 was observed. Mdx-T also exhibited enhanced force and resistance to fatigue (+20 to 60%, P<0.05 or less). These beneficial effects resulted from MCC950 inhibition of both active caspase-1 (-46%, P=0.075) and cleaved gasdermin D (N-GSDMD) (-42% in medium-sized-fibres, P<0.001). Finally, the anti-inflammatory action and the anti-pyroptotic effect of MCC950 were also recapitulated in DMD human myotubes. Conclusion Specific inhibition of the NLRP3 inflammasome can significantly attenuate the dystrophic phenotype. A novel finding of this study is the overactivation of GSDMD, which is hampered by MCC950. This ultimately leads to less inflammation and pyroptosis and to a better muscle maturation and function. Targeting NLRP3 might lead to an effective therapeutic approach for a better management of DMD.
Collapse
Affiliation(s)
- Nicolas Dubuisson
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Brussels, Belgium,Neuromuscular Reference Center, Department of Neurology, Cliniques Universitaires Saint-Luc, Brussels, Belgium,*Correspondence: Nicolas Dubuisson,
| | - María A. Davis-López de Carrizosa
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Brussels, Belgium,Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain
| | - Romain Versele
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Camille M. Selvais
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Laurence Noel
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - P. Y. D. Van den Bergh
- Neuromuscular Reference Center, Department of Neurology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Sonia M. Brichard
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Michel Abou-Samra
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
25
|
Eggelbusch M, Shi A, Broeksma BC, Vázquez-Cruz M, Soares MN, de Wit GMJ, Everts B, Jaspers RT, Wüst RCI. The NLRP3 inflammasome contributes to inflammation-induced morphological and metabolic alterations in skeletal muscle. J Cachexia Sarcopenia Muscle 2022; 13:3048-3061. [PMID: 35978267 PMCID: PMC9745466 DOI: 10.1002/jcsm.13062] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 06/09/2022] [Accepted: 07/04/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Systemic inflammation is associated with skeletal muscle atrophy and metabolic dysfunction. Although the nucleotide-binding oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3) inflammasome contributes to cytokine production in immune cells, its role in skeletal muscle is poorly understood. Here, we studied the link between inflammation, NLRP3, muscle morphology, and metabolism in in vitro cultured C2C12 myotubes, independent of immune cell involvement. METHODS Differentiated C2C12 myotubes were treated with lipopolysaccharide (LPS; 0, 10, and 100-200 ng/mL) to induce activation of the NLRP3 inflammasome with and without MCC950, a pharmacological inhibitor of NLRP3-induced IL-1β production. We assessed markers of the NLRP3 inflammasome, cell diameter, reactive oxygen species, and mitochondrial function. RESULTS NLRP3 gene expression and protein concentrations increased in a time-dependent and dose-dependent manner. Intracellular IL-1β concentration significantly increased (P < 0.0001), but significantly less with MCC950 (P = 0.03), suggestive of moderate activation of the NLRP3 inflammasome in cultured myotubes upon LPS stimulation. LPS suppressed myotube growth after 24 h (P = 0.03), and myotubes remained smaller up to 72 h (P = 0.0009). Exposure of myotubes to IL-1β caused similar alterations in cell morphology, and MCC950 mitigated these LPS-induced differences in cell diameter. NLRP3 appeared to co-localize with mitochondria, more so upon exposure to LPS. Mitochondrial reactive oxygen species were higher after LPS (P = 0.03), but not after addition of MCC950. Myotubes had higher glycolytic rates, and mitochondria were more fragmented upon LPS exposure, which was not altered by MCC950 supplementation. CONCLUSIONS LPS-induced activation of the NLRP3 inflammasome in cultured myotubes contributes to morphological and metabolic alterations, likely due to its mitochondrial association.
Collapse
Affiliation(s)
- Moritz Eggelbusch
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands.,Department of Nutrition and Dietetics, Amsterdam UMC location VUmc, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands.,Faculty of Sports and Nutrition, Center of Expertise Urban Vitality, Amsterdam University of Applied Sciences, Amsterdam, The Netherlands
| | - Andi Shi
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands.,Department of Oral and Maxillofacial Surgery/Pathology, Amsterdam UMC and Academic Center for Dentistry Amsterdam, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands.,Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Bonnie C Broeksma
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Mariana Vázquez-Cruz
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Madu N Soares
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Gerard M J de Wit
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Bart Everts
- Department of Parasitology, Leiden University Medical Center, University of Leiden, Leiden, The Netherlands
| | - Richard T Jaspers
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands.,Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Rob C I Wüst
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| |
Collapse
|
26
|
Liu Y, Wang D, Li T, Yang F, Li Z, Bai X, Wang Y. The role of NLRP3 inflammasome in inflammation-related skeletal muscle atrophy. Front Immunol 2022; 13:1035709. [PMID: 36405697 PMCID: PMC9668849 DOI: 10.3389/fimmu.2022.1035709] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 10/13/2022] [Indexed: 04/04/2024] Open
Abstract
Skeletal muscle atrophy is a common complication in survivors of sepsis, which affects the respiratory and motor functions of patients, thus severely impacting their quality of life and long-term survival. Although several advances have been made in investigations on the pathogenetic mechanism of sepsis-induced skeletal muscle atrophy, the underlying mechanisms remain unclear. Findings from recent studies suggest that the nucleotide-binding and oligomerisation domain (NOD)-like receptor family pyrin domain containing 3 (NLRP3) inflammasome, a regulator of inflammation, may be crucial in the development of skeletal muscle atrophy. NLRP3 inhibitors contribute to the inhibition of catabolic processes, skeletal muscle atrophy and cachexia-induced inflammation. Here, we review the mechanisms by which NLRP3 mediates these responses and analyse how NLRP3 affects muscle wasting during inflammation.
Collapse
Affiliation(s)
- Yukun Liu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dongfang Wang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tianyu Li
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan Yang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhanfei Li
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangjun Bai
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuchang Wang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
27
|
Zuo Y, Xiao T, Qiu X, Liu Z, Zhang S, Zhou N. Adiponectin reduces apoptosis of diabetic cardiomyocytes by regulating miR-711/TLR4 axis. Diabetol Metab Syndr 2022; 14:131. [PMID: 36114541 PMCID: PMC9479314 DOI: 10.1186/s13098-022-00904-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 09/02/2022] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE To investigate the regulation of adiponectin/miR-711 on TLR4/NF-κB-mediated inflammatory response and diabetic cardiomyocyte apoptosis. METHODS Diabetes models were established using rats and H9c2 cardiomyocytes. qRT-PCR was used to detect adiponectin, miR-711, and TLR4. MTT, β-galactosidase staining, and flow cytometry were utilized to assess cell viability, senescence, and apoptosis, respectively. The colorimetric method was used to measure caspase-3 activity, DCFH-DA probes to detect ROS, and western blotting to determine the protein levels of Bax, Bcl-2, TLR4, and p-NF-κB p65. ELISA was performed to measure the levels of adiponectin, ICAM-1, MCP-1, and IL-1β. Dual-luciferase reporter system examined the targeting relationship between miR-711 and TLR4. H&E and TUNEL staining revealed myocardial structure and apoptosis, respectively. RESULTS Adiponectin and miR-711 were underexpressed and TLR4/NF-κB signaling pathway was activated in high glucose-treated H9c2 cells. High glucose treatment reduced viability, provoked inflammatory response, and accelerated senescence and apoptosis in H9c2 cells. miR-711 could bind TLR4 mRNA and inactivate TLR4/NF-κB signaling. Adiponectin treatment increased miR-711 expression and blocked TLR4/NF-κB signaling. Adiponectin/miR-711 reduced myocardial inflammation and apoptosis in diabetic rats. CONCLUSION Adiponectin inhibits inflammation and alleviates high glucose-induced cardiomyocyte apoptosis by blocking TLR4/NF-κB signaling pathway through miR-711.
Collapse
Affiliation(s)
- Yu Zuo
- Department of the Pre-Hospital First-Aid, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Tao Xiao
- Nursing Department, The Third Xiangya Hospital of Central South University, No. 138, Tongzipo Road, Yuelu District, Changsha, Hunan, 410013, People's Republic of China.
| | - Xiangdong Qiu
- Department of the Pre-Hospital First-Aid, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Zuoliang Liu
- Intensive Care Unit, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Shengnan Zhang
- Department of the Pre-Hospital First-Aid, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Na Zhou
- Department of Anesthesiology, Hunan Aerospace Hospital, Changsha, Hunan, 410205, People's Republic of China
| |
Collapse
|
28
|
Feng X, Xiao J, Bai L. Role of adiponectin in osteoarthritis. Front Cell Dev Biol 2022; 10:992764. [PMID: 36158216 PMCID: PMC9492855 DOI: 10.3389/fcell.2022.992764] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/17/2022] [Indexed: 11/20/2022] Open
Abstract
Osteoarthritis (OA) is a widespread and most common joint disease which leads to social cost increasing accompany with aging population. Surgery is often the final treatment option. The major progression of OA includes cartilage degradation caused by chondrocytes metabolism imbalance. So, the molecular mechanisms of action in chondrocytes may provide insights into treatment methods for OA. Adiponectin is an adipokine with many biological functions in the cell metabolism. Numerous studies have illustrated that adiponectin has diverse biological effects, such as inhibition of cell apoptosis. It regulates various functions in different organs, including muscle, adipose tissue, brain, and bone, and regulates skeletal homeostasis. However, the relationship between adiponectin and cell death in the progression of OA needs further investigation. We elaborate the structure and function and the effect of adiponectin and state the correlation and intersection between adiponectin, autophagy, inflammation, and OA. From the perspective of oxidative stress, apoptosis, pyroptosis, and autophagy, we discuss the possible association between adiponectin, chondrocyte metabolism, and inflammatory factor efforts in OA. What’s more, we summarize the possible treatment methods, including the use of adiponectin as a drug target, and highlight the potential future mechanistic research. In this review, we summarize the molecular pathways and mechanisms of action of adiponectin in chondrocyte inflammation and death and the pathogenesis of OA. We also review the research on adiponectin as a target for treating OA. These studies provide a novel perspective to explore more effective treatment options considering the complex interrelationship between inflammation and metabolism in OA.
Collapse
Affiliation(s)
- Xinyuan Feng
- Department of Orthopedic Surgery, Shengjing Hospital, China Medical University, Shenyang, China
| | - Jiaying Xiao
- Department of Internal Medicine Integrated Ward 2, Shengjing Hospital, China Medical University, Shenyang, China
| | - Lunhao Bai
- Department of Orthopedic Surgery, Shengjing Hospital, China Medical University, Shenyang, China
- *Correspondence: Lunhao Bai,
| |
Collapse
|
29
|
Cao Z, Yang F, Lin Y, Shan J, Cao H, Zhang C, Zhuang Y, Xing C, Hu G. Selenium Antagonizes Cadmium-Induced Inflammation and Oxidative Stress via Suppressing the Interplay between NLRP3 Inflammasome and HMGB1/NF-κB Pathway in Duck Hepatocytes. Int J Mol Sci 2022; 23:ijms23116252. [PMID: 35682929 PMCID: PMC9181349 DOI: 10.3390/ijms23116252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 05/29/2022] [Accepted: 05/31/2022] [Indexed: 12/14/2022] Open
Abstract
Cadmium (Cd) is a toxic heavy metal that can accumulate in the liver of animals, damaging liver function. Inflammation and oxidative stress are considered primary causes of Cd-induced liver damage. Selenium (Se) is an antioxidant and can resist the detrimental impacts of Cd on the liver. To elucidate the antagonism of Se on Cd against hepatocyte injury and its mechanism, duck embryo hepatocytes were treated with Cd (4 μM) and/or Se (0.4 μM) for 24 h. Then, the hepatocyte viability, oxidative stress and inflammatory status were assessed. The findings manifested that the accumulation of reactive oxygen species (ROS) and the levels of pro-inflammatory factors were elevated in the Cd group. Simultaneously, immunofluorescence staining revealed that the interaction between NOD-like receptor pyran domain containing 3 (NLRP3) and apoptosis-associated speck-like protein (ASC) was enhanced, the movement of high-mobility group box 1 (HMGB1) from nucleus to cytoplasm was increased and the inflammatory response was further amplified. Nevertheless, the addition of Se relieved the above-mentioned effects, thereby alleviating cellular oxidative stress and inflammation. Collectively, the results suggested that Se could mitigate Cd-stimulated oxidative stress and inflammation in hepatocytes, which might be correlated with the NLRP3 inflammasome and HMGB1/nuclear factor-κB (NF-κB) signaling pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Chenghong Xing
- Correspondence: (C.X.); (G.H.); Tel.: +86-18770046182 (C.X.); +86-13807089905 (G.H.)
| | - Guoliang Hu
- Correspondence: (C.X.); (G.H.); Tel.: +86-18770046182 (C.X.); +86-13807089905 (G.H.)
| |
Collapse
|
30
|
Schultz TI, Raucci FJ, Salloum FN. Cardiovascular Disease in Duchenne Muscular Dystrophy. JACC Basic Transl Sci 2022; 7:608-625. [PMID: 35818510 PMCID: PMC9270569 DOI: 10.1016/j.jacbts.2021.11.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 11/06/2021] [Indexed: 12/11/2022]
Abstract
Cardiomyopathy is the leading cause of death in patients with DMD. DMD has no cure, and there is no current consensus for treatment of DMD cardiomyopathy. This review discusses therapeutic strategies to potentially reduce or prevent cardiac dysfunction in DMD patients. Additional studies are needed to firmly establish optimal treatment modalities for DMD cardiomyopathy.
Duchenne muscular dystrophy (DMD) is a devastating disease affecting approximately 1 in every 3,500 male births worldwide. Multiple mutations in the dystrophin gene have been implicated as underlying causes of DMD. However, there remains no cure for patients with DMD, and cardiomyopathy has become the most common cause of death in the affected population. Extensive research is under way investigating molecular mechanisms that highlight potential therapeutic targets for the development of pharmacotherapy for DMD cardiomyopathy. In this paper, the authors perform a literature review reporting on recent ongoing efforts to identify novel therapeutic strategies to reduce, prevent, or reverse progression of cardiac dysfunction in DMD.
Collapse
|
31
|
González-Jamett A, Vásquez W, Cifuentes-Riveros G, Martínez-Pando R, Sáez JC, Cárdenas AM. Oxidative Stress, Inflammation and Connexin Hemichannels in Muscular Dystrophies. Biomedicines 2022; 10:biomedicines10020507. [PMID: 35203715 PMCID: PMC8962419 DOI: 10.3390/biomedicines10020507] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/13/2022] [Accepted: 02/15/2022] [Indexed: 12/16/2022] Open
Abstract
Muscular dystrophies (MDs) are a heterogeneous group of congenital neuromuscular disorders whose clinical signs include myalgia, skeletal muscle weakness, hypotonia, and atrophy that leads to progressive muscle disability and loss of ambulation. MDs can also affect cardiac and respiratory muscles, impairing life-expectancy. MDs in clude Duchenne muscular dystrophy, Emery-Dreifuss muscular dystrophy, facioscapulohumeral muscular dystrophy and limb-girdle muscular dystrophy. These and other MDs are caused by mutations in genes that encode proteins responsible for the structure and function of skeletal muscles, such as components of the dystrophin-glycoprotein-complex that connect the sarcomeric-actin with the extracellular matrix, allowing contractile force transmission and providing stability during muscle contraction. Consequently, in dystrophic conditions in which such proteins are affected, muscle integrity is disrupted, leading to local inflammatory responses, oxidative stress, Ca2+-dyshomeostasis and muscle degeneration. In this scenario, dysregulation of connexin hemichannels seem to be an early disruptor of the homeostasis that further plays a relevant role in these processes. The interaction between all these elements constitutes a positive feedback loop that contributes to the worsening of the diseases. Thus, we discuss here the interplay between inflammation, oxidative stress and connexin hemichannels in the progression of MDs and their potential as therapeutic targets.
Collapse
Affiliation(s)
- Arlek González-Jamett
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (W.V.); (J.C.S.)
- Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso 2360102, Chile; (G.C.-R.); (R.M.-P.)
- Correspondence: (A.G.-J.); (A.M.C.)
| | - Walter Vásquez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (W.V.); (J.C.S.)
| | - Gabriela Cifuentes-Riveros
- Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso 2360102, Chile; (G.C.-R.); (R.M.-P.)
| | - Rafaela Martínez-Pando
- Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso 2360102, Chile; (G.C.-R.); (R.M.-P.)
| | - Juan C. Sáez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (W.V.); (J.C.S.)
| | - Ana M. Cárdenas
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (W.V.); (J.C.S.)
- Correspondence: (A.G.-J.); (A.M.C.)
| |
Collapse
|
32
|
Dubuisson N, Versele R, Davis-López de Carrizosa MA, Selvais CM, Brichard SM, Abou-Samra M. Walking down Skeletal Muscle Lane: From Inflammasome to Disease. Cells 2021; 10:cells10113023. [PMID: 34831246 PMCID: PMC8616386 DOI: 10.3390/cells10113023] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 10/29/2021] [Accepted: 11/02/2021] [Indexed: 12/14/2022] Open
Abstract
Over the last decade, innate immune system receptors and sensors called inflammasomes have been identified to play key pathological roles in the development and progression of numerous diseases. Among them, the nucleotide-binding oligomerization domain (NOD-), leucine-rich repeat (LRR-) and pyrin domain-containing protein 3 (NLRP3) inflammasome is probably the best characterized. To date, NLRP3 has been extensively studied in the heart, where its effects and actions have been broadly documented in numerous cardiovascular diseases. However, little is still known about NLRP3 implications in muscle disorders affecting non-cardiac muscles. In this review, we summarize and present the current knowledge regarding the function of NLRP3 in diseased skeletal muscle, and discuss the potential therapeutic options targeting the NLRP3 inflammasome in muscle disorders.
Collapse
Affiliation(s)
- Nicolas Dubuisson
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain, 1200 Brussels, Belgium; (R.V.); (M.A.D.-L.d.C.); (C.M.S.); (S.M.B.); (M.A.-S.)
- Neuromuscular Reference Center, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
- Correspondence:
| | - Romain Versele
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain, 1200 Brussels, Belgium; (R.V.); (M.A.D.-L.d.C.); (C.M.S.); (S.M.B.); (M.A.-S.)
| | - María A. Davis-López de Carrizosa
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain, 1200 Brussels, Belgium; (R.V.); (M.A.D.-L.d.C.); (C.M.S.); (S.M.B.); (M.A.-S.)
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012 Seville, Spain
| | - Camille M. Selvais
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain, 1200 Brussels, Belgium; (R.V.); (M.A.D.-L.d.C.); (C.M.S.); (S.M.B.); (M.A.-S.)
| | - Sonia M. Brichard
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain, 1200 Brussels, Belgium; (R.V.); (M.A.D.-L.d.C.); (C.M.S.); (S.M.B.); (M.A.-S.)
| | - Michel Abou-Samra
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain, 1200 Brussels, Belgium; (R.V.); (M.A.D.-L.d.C.); (C.M.S.); (S.M.B.); (M.A.-S.)
| |
Collapse
|
33
|
Américo-Da-Silva L, Aguilera J, Quinteros-Waltemath O, Sánchez-Aguilera P, Russell J, Cadagan C, Meneses-Valdés R, Sánchez G, Estrada M, Jorquera G, Barrientos G, Llanos P. Activation of the NLRP3 Inflammasome Increases the IL-1β Level and Decreases GLUT4 Translocation in Skeletal Muscle during Insulin Resistance. Int J Mol Sci 2021; 22:10212. [PMID: 34638553 PMCID: PMC8508423 DOI: 10.3390/ijms221910212] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 12/16/2022] Open
Abstract
Low-grade chronic inflammation plays a pivotal role in the pathogenesis of insulin resistance (IR), and skeletal muscle has a central role in this condition. NLRP3 inflammasome activation pathways promote low-grade chronic inflammation in several tissues. However, a direct link between IR and NLRP3 inflammasome activation in skeletal muscle has not been reported. Here, we evaluated the NLRP3 inflammasome components and their role in GLUT4 translocation impairment in skeletal muscle during IR. Male C57BL/6J mice were fed with a normal control diet (NCD) or high-fat diet (HFD) for 8 weeks. The protein levels of NLRP3, ASC, caspase-1, gasdermin-D (GSDMD), and interleukin (IL)-1β were measured in both homogenized and isolated fibers from the flexor digitorum brevis (FDB) or soleus muscle. GLUT4 translocation was determined through GLUT4myc-eGFP electroporation of the FBD muscle. Our results, obtained using immunofluorescence, showed that adult skeletal muscle expresses the inflammasome components. In the FDB and soleus muscles, homogenates from HFD-fed mice, we found increased protein levels of NLRP3 and ASC, higher activation of caspase-1, and elevated IL-1β in its mature form, compared to NCD-fed mice. Moreover, GSDMD, a protein that mediates IL-1β secretion, was found to be increased in HFD-fed-mice muscles. Interestingly, MCC950, a specific pharmacological NLRP3 inflammasome inhibitor, promoted GLUT4 translocation in fibers isolated from the FDB muscle of NCD- and HFD-fed mice. In conclusion, we found increased NLRP3 inflammasome components in adult skeletal muscle of obese insulin-resistant animals, which might contribute to the low-grade chronic metabolic inflammation of skeletal muscle and IR development.
Collapse
Affiliation(s)
- Luan Américo-Da-Silva
- Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Santiago 8380544, Chile; (L.A.-D.-S.); (J.A.); (O.Q.-W.); (C.C.)
| | - Javiera Aguilera
- Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Santiago 8380544, Chile; (L.A.-D.-S.); (J.A.); (O.Q.-W.); (C.C.)
| | - Oscar Quinteros-Waltemath
- Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Santiago 8380544, Chile; (L.A.-D.-S.); (J.A.); (O.Q.-W.); (C.C.)
| | - Pablo Sánchez-Aguilera
- Centro de Estudios en Ejercicio, Metabolismo y Cáncer, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (P.S.-A.); (R.M.-V.)
| | - Javier Russell
- Escuela de Pedagogía en Educación Física, Facultad de Educación, Universidad Autónoma de Chile, Santiago 8900000, Chile;
| | - Cynthia Cadagan
- Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Santiago 8380544, Chile; (L.A.-D.-S.); (J.A.); (O.Q.-W.); (C.C.)
| | - Roberto Meneses-Valdés
- Centro de Estudios en Ejercicio, Metabolismo y Cáncer, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (P.S.-A.); (R.M.-V.)
| | - Gina Sánchez
- Programa de Fisiopatología, ICBM, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile;
| | - Manuel Estrada
- Programa de Fisiología y Biofísica, ICBM, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile;
| | - Gonzalo Jorquera
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile;
| | - Genaro Barrientos
- Centro de Estudios en Ejercicio, Metabolismo y Cáncer, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (P.S.-A.); (R.M.-V.)
- Programa de Fisiología y Biofísica, ICBM, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile;
| | - Paola Llanos
- Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Santiago 8380544, Chile; (L.A.-D.-S.); (J.A.); (O.Q.-W.); (C.C.)
- Centro de Estudios en Ejercicio, Metabolismo y Cáncer, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (P.S.-A.); (R.M.-V.)
| |
Collapse
|
34
|
She Y, Li C, Jiang T, Lei S, Zhou S, Shi H, Chen R. Knockdown of CNN3 Impairs Myoblast Proliferation, Differentiation, and Protein Synthesis via the mTOR Pathway. Front Physiol 2021; 12:659272. [PMID: 34305633 PMCID: PMC8295729 DOI: 10.3389/fphys.2021.659272] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 05/20/2021] [Indexed: 11/22/2022] Open
Abstract
Background Myogenesis is a complex process that requires optimal outside–in substrate–cell signaling. Calponin 3 (CNN3) plays an important role in regulating myogenic differentiation and muscle regeneration; however, the precise function of CNN3 in myogenesis regulation remains poorly understood. Here, we investigated the role of CNN3 in a knockdown model in the mouse muscle cell line C2C12. Methods Myoblast proliferation, migration, differentiation, fusion, and protein synthesis were examined in CNN3 knockdown C2C12 mouse muscle cells. Involvement of the mTOR pathway in CNN3 signaling was explored by treating cells with the mTOR activator MHY1485. The regulatory mechanisms of CNN3 in myogenesis were further examined by RNA sequencing and subsequent gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and gene set enrichment analysis (GSEA). Results During proliferation, CNN3 knockdown caused a decrease in cell proliferation and migration. During differentiation, CNN3 knockdown inhibited myogenic differentiation, fusion, and protein synthesis in C2C12 cells via the AKT/mTOR and AMPK/mTOR pathways; this effect was reversed by MHY1485 treatment. Finally, KEGG and GSEA indicated that the NOD-like receptor signaling pathway is affected in CNN3 knockdown cell lines. Conclusion CNN3 may promote C2C12 cell growth by regulating AKT/mTOR and AMPK/mTOR signaling. The KEGG and GSEA indicated that inhibiting CNN3 may activate several pathways, including the NOD-like receptor pathway and pathways involved in necroptosis, apoptosis, and inflammation.
Collapse
Affiliation(s)
- Yanling She
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Cheng Li
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Ting Jiang
- Department of Radiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Si Lei
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Shanyao Zhou
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Huacai Shi
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Rui Chen
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute, Guangdong Second Provincial General Hospital, Guangzhou, China
| |
Collapse
|
35
|
The Interplay of Mitophagy and Inflammation in Duchenne Muscular Dystrophy. Life (Basel) 2021; 11:life11070648. [PMID: 34357020 PMCID: PMC8307817 DOI: 10.3390/life11070648] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/30/2021] [Accepted: 07/02/2021] [Indexed: 12/11/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked neuromuscular disease caused by a pathogenic disruption of the DYSTROPHIN gene that results in non-functional dystrophin protein. DMD patients experience loss of ambulation, cardiac arrhythmia, metabolic syndrome, and respiratory failure. At the molecular level, the lack of dystrophin in the muscle results in myofiber death, fibrotic infiltration, and mitochondrial dysfunction. There is no cure for DMD, although dystrophin-replacement gene therapies and exon-skipping approaches are being pursued in clinical trials. Mitochondrial dysfunction is one of the first cellular changes seen in DMD myofibers, occurring prior to muscle disease onset and progresses with disease severity. This is seen by reduced mitochondrial function, abnormal mitochondrial morphology and impaired mitophagy (degradation of damaged mitochondria). Dysfunctional mitochondria release high levels of reactive oxygen species (ROS), which can activate pro-inflammatory pathways such as IL-1β and IL-6. Impaired mitophagy in DMD results in increased inflammation and further aggravates disease pathology, evidenced by increased muscle damage and increased fibrosis. This review will focus on the critical interplay between mitophagy and inflammation in Duchenne muscular dystrophy as a pathological mechanism, as well as describe both candidate and established therapeutic targets that regulate these pathways.
Collapse
|
36
|
Lou Y, Miao J, Li F, Ding J, Wang L. Maternal smoking during pregnancy aggravated muscle phenotype in FHL1 -/y offspring mice similar to congenital clubfoot through P2RX7-mediated pyroptosis. Toxicol Lett 2021; 345:54-60. [PMID: 33872746 DOI: 10.1016/j.toxlet.2021.04.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 04/13/2021] [Accepted: 04/14/2021] [Indexed: 01/08/2023]
Abstract
Congenital clubfoot (CCF) is a common birth defect. Maternal smoking during pregnancy increases the risk of CCF. In previous research, we found muscle phenotypes similar to CCF in four and a half LIM domain protein 1 (FHLI) offspring mice (FHL1-/y). However, the role of P2RX7-mediated pyroptosis in the effect of cigarette smoke (CS) on the skeletal muscle of FHL1-/y mice during pregnancy is unclear. In the present study, pregnant mice at 11 days of gestation were exposed to CS and male offspring of wild-type (WT) and FHL1-/y mice were divided into four groups (Control-WT, Control-KO, CS-WT, CS-KO). The histomorphology of lower limb muscles was examined using hematoxylin and eosin (H&E) staining. P2RX7, indicators of pyroptosis (NLRP3, ASC, cleaved-caspase 1, IL-1β), and cytoskeletal proteins (MYBPC2, LDB3) were also detected using immunoblotting. CS exposure during pregnancy aggravated the muscle phenotype similar to CCF in FHL1-/y offspring mice. FHL1 gene knockout (KO) or CS exposure during pregnancy each activated the expression of P2RX7, cell pyroptosis-related proteins (NLRP3, ASC, cleaved-caspase 1, IL-1β), a muscle injury marker (MYOD1), and cytoskeletal proteins (MYBPC2, LDB3); these two factors had an additive effect. The results showed maternal smoking during pregnancy aggravated muscle phenotype similar to CCF in FHL1-/y offspring mice through P2RX7-mediated pyroptosis.
Collapse
Affiliation(s)
- Yi Lou
- Department of Medical Genetics, China Medical University, Shenyang, China
| | - Jianing Miao
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, Shenyang, 110004, China; Medical Research Center of Shengjing Hospital, China Medical University, Shenyang, 110004, China; Key Laboratory of Research and Application of Animal Model for Environmental and Metabolic Diseases, Liaoning Province, China.
| | - Fang Li
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, Shenyang, 110004, China; Medical Research Center of Shengjing Hospital, China Medical University, Shenyang, 110004, China; Key Laboratory of Research and Application of Animal Model for Environmental and Metabolic Diseases, Liaoning Province, China
| | - Jingjing Ding
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, Shenyang, 110004, China; Medical Research Center of Shengjing Hospital, China Medical University, Shenyang, 110004, China; Key Laboratory of Research and Application of Animal Model for Environmental and Metabolic Diseases, Liaoning Province, China
| | - Lili Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, Shenyang, 110004, China; Medical Research Center of Shengjing Hospital, China Medical University, Shenyang, 110004, China; Key Laboratory of Research and Application of Animal Model for Environmental and Metabolic Diseases, Liaoning Province, China.
| |
Collapse
|
37
|
Péladeau C, Sandhu JK. Aberrant NLRP3 Inflammasome Activation Ignites the Fire of Inflammation in Neuromuscular Diseases. Int J Mol Sci 2021; 22:ijms22116068. [PMID: 34199845 PMCID: PMC8200055 DOI: 10.3390/ijms22116068] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 05/30/2021] [Accepted: 06/01/2021] [Indexed: 12/24/2022] Open
Abstract
Inflammasomes are molecular hubs that are assembled and activated by a host in response to various microbial and non-microbial stimuli and play a pivotal role in maintaining tissue homeostasis. The NLRP3 is a highly promiscuous inflammasome that is activated by a wide variety of sterile triggers, including misfolded protein aggregates, and drives chronic inflammation via caspase-1-mediated proteolytic cleavage and secretion of proinflammatory cytokines, interleukin-1β and interleukin-18. These cytokines further amplify inflammatory responses by activating various signaling cascades, leading to the recruitment of immune cells and overproduction of proinflammatory cytokines and chemokines, resulting in a vicious cycle of chronic inflammation and tissue damage. Neuromuscular diseases are a heterogeneous group of muscle disorders that involve injury or dysfunction of peripheral nerves, neuromuscular junctions and muscles. A growing body of evidence suggests that dysregulation, impairment or aberrant NLRP3 inflammasome signaling leads to the initiation and exacerbation of pathological processes associated with neuromuscular diseases. In this review, we summarize the available knowledge about the NLRP3 inflammasome in neuromuscular diseases that affect the peripheral nervous system and amyotrophic lateral sclerosis, which affects the central nervous system. In addition, we also examine whether therapeutic targeting of the NLRP3 inflammasome components is a viable approach to alleviating the detrimental phenotype of neuromuscular diseases and improving clinical outcomes.
Collapse
Affiliation(s)
- Christine Péladeau
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON K1A 0R6, Canada;
| | - Jagdeep K. Sandhu
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON K1A 0R6, Canada;
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Correspondence: ; Tel.: +1-613-993-5304
| |
Collapse
|
38
|
Chen Y, Ma Y, Feng JJ, Wang YH, Li TF, Nurmi K, Eklund KK, Wen JG. Histamine H 3 Receptor Signaling Regulates the NLRP3 Inflammasome Activation in C2C12 Myocyte During Myogenic Differentiation. Front Pharmacol 2021; 12:599393. [PMID: 34135750 PMCID: PMC8202077 DOI: 10.3389/fphar.2021.599393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 05/05/2021] [Indexed: 11/13/2022] Open
Abstract
NLRP3 inflammasome has been implicated in impaired post-injury muscle healing and in muscle atrophy. Histamine receptors play an important role in inflammation, but the role of histamine H3 receptor (H3R) in myocyte regeneration and in the regulation of NLRP3 inflammasome is not known. We studied the effects of H3R signaling on C2C12 myocyte viability, apoptosis, and tumor necrosis factor alpha (TNFα)-induced NLRP3 inflammasome activation during striated myogenic differentiation at three time points (days 0, 3, and 6). Expression of Nlrp3, interleukin-1β (IL-1β), and myogenesis markers were determined. TNFα reduced overall viability of C2C12 cells, and exposure to TNFα induced apoptosis of cells at D6. Activation of H3R had no effect on viability or apoptosis, whereas inhibition of H3R increased TNFα-induced apoptosis. Stimulation of C2C12 cells with TNFα increased Nlrp3 mRNA expression at D3 and D6. Moreover, TNFα reduced the expression of myogenesis markers MyoD1, Myogenin, and Myosin-2 at D3 and D6. H3R attenuated TNFα-induced expression of Nlrp3 and further inhibited the myogenesis marker expression; while H3R -blockage enhanced the proinflammatory effects of TNFα and increased the myogenesis marker expression. TNFα-induced secretion of mature IL-1β was dependent on the activation of the NLRP3 inflammasome, as shown by the reduced secretion of mature IL-1β upon treatment of the cells with the small molecule inhibitor of the NLRP3 inflammasome (MCC950). The activation of H3R reduced TNFα-induced IL-1β secretion, while the H3R blockage had an opposite effect. In conclusion, the modulation of H3R activity regulates the effects of TNFα on C2C12 myocyte differentiation and TNFα-induced activation of NLRP3 inflammasome. Thus, H3R signaling may represent a novel target for limiting postinjury muscle inflammation and muscle atrophy.
Collapse
Affiliation(s)
- Yan Chen
- Urodynamic Center and Department of Urology, Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Helsinki Rheumatic Diseases and Inflammation Research Group, Clinicum Helsinki University, Helsinki, Finland.,Translational Immunology Research Program, University of Helsinki, Helsinki University Clinicum, Helsinki, Finland
| | - Yuan Ma
- Urodynamic Center and Department of Urology, Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jin Jin Feng
- Urodynamic Center and Department of Urology, Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi He Wang
- Urodynamic Center and Department of Urology, Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tian Fang Li
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Katariina Nurmi
- Helsinki Rheumatic Diseases and Inflammation Research Group, Clinicum Helsinki University, Helsinki, Finland.,Translational Immunology Research Program, University of Helsinki, Helsinki University Clinicum, Helsinki, Finland
| | - Kari K Eklund
- Helsinki Rheumatic Diseases and Inflammation Research Group, Clinicum Helsinki University, Helsinki, Finland.,Translational Immunology Research Program, University of Helsinki, Helsinki University Clinicum, Helsinki, Finland.,Department of Medicine, Division of Rheumatology, Helsinki University Central Hospital, and Orton Orthopedic Hospital, Helsinki, Finland
| | - Jian Guo Wen
- Urodynamic Center and Department of Urology, Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
39
|
Yang FM, Fan D, Yang XQ, Zhu FH, Shao MJ, Li Q, Liu YT, Lin ZM, Cao SQ, Tang W, He SJ, Zuo JP. The artemisinin analog SM934 alleviates dry eye disease in rodent models by regulating TLR4/NF-κB/NLRP3 signaling. Acta Pharmacol Sin 2021; 42:593-603. [PMID: 32747720 PMCID: PMC8114933 DOI: 10.1038/s41401-020-0484-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 07/15/2020] [Indexed: 01/22/2023]
Abstract
Dry eye disease (DED) is a multifactorial disorder of the tears and ocular surface characterized by manifestations of dryness and irritation. Although the pathogenesis is not fully illuminated, it is recognized that inflammation has a prominent role in the development and deterioration of DED. β-aminoarteether maleate (SM934) is a water-soluble artemisinin derivative with anti-inflammatory and immunosuppressive activities. In this study, we established scopolamine hydrobromide (SCOP)-induced rodent model as well as benzalkonium chloride (BAC)-induced rat model to investigate the therapeutic potential of SM934 for DED. We showed that topical application of SM934 (0.1%, 0.5%) significantly increased tear secretion, maintained the number of conjunctival goblet cells, reduced corneal damage, and decreased the levels of inflammatory mediators (TNF-α, IL-6, IL-10, or IL-1β) in conjunctiva in SCOP-induced and BAC-induced DED models. Moreover, SM934 treatment reduced the accumulation of TLR4-expressing macrophages in conjunctiva, and suppressed the expression of inflammasome components, i.e., myeloid differentiation factor88 (MyD88), Nod-like receptor protein 3 (NLRP3), apoptosis-associated speck-like protein containing CARD (ASC), and cleaved caspase 1. In LPS-treated RAW 264.7 cells, we demonstrated that pretreatment with SM934 (10 μM) impeded the upregulation of TLR4 and downstream NF-κB/NLRP3 signaling proteins. Collectively, artemisinin analog SM934 exerts therapeutic benefits on DED by simultaneously reserving the structural integrity of ocular surface and preventing the corneal and conjunctival inflammation, suggested a further application of SM934 in ophthalmic therapy, especially for DED.
Collapse
Affiliation(s)
- Fang-Ming Yang
- Laboratory of Immunology and Virology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Di Fan
- Laboratory of Immunology and Virology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiao-Qian Yang
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Feng-Hua Zhu
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Mei-Juan Shao
- Laboratory of Immunology and Virology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Qian Li
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yu-Ting Liu
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ze-Min Lin
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Shi-Qi Cao
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wei Tang
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shi-Jun He
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Jian-Ping Zuo
- Laboratory of Immunology and Virology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
40
|
Childers GM, Perry CA, Blachut B, Martin N, Bortner CD, Sieber S, Li JL, Fessler MB, Harry GJ. Assessing the Association of Mitochondrial Function and Inflammasome Activation in Murine Macrophages Exposed to Select Mitotoxic Tri-Organotin Compounds. ENVIRONMENTAL HEALTH PERSPECTIVES 2021; 129:47015. [PMID: 33929904 PMCID: PMC8086801 DOI: 10.1289/ehp8314] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
BACKGROUND Mitochondrial function is implicated as a target of environmental toxicants and found in disease or injury models, contributing to acute and chronic inflammation. One mechanism by which mitochondrial damage can propagate inflammation is via activation of the nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) family, pyrin domain-containing receptor (NLRP)3 inflammasome, a protein complex that processes mature interleukin (IL)-1β. IL-1β plays an important role in the innate immune response and dysregulation is associated with autoinflammatory disorders. OBJECTIVE The objective was to evaluate whether mitochondrial toxicants recruit inflammasome activation and IL-1β processing. METHOD Murine macrophages (RAW 264.7) exposed to tri-organotins (triethyltin bromide (TETBr), trimethyltin hydroxide (TMTOH), triphenyltin hydroxide (TPTOH), bis(tributyltin)oxide) [Bis(TBT)Ox] were examined for pro-inflammatory cytokine induction. TMTOH and TETBr were examined in RAW 264.7 and bone marrow-derived macrophages for mitochondrial bioenergetics, reactive oxygen species (ROS) production, and inflammasome activation via visualization of aggregate formation, caspase-1 flow cytometry, IL-1β enzyme-linked immunosorbent assay and Western blots, and microRNA (miRNA) and mRNA arrays. RESULTS TETBr and TMTOH induced inflammasome aggregate formation and IL-1β release in lipopolysaccharide (LPS)-primed macrophages. Mitochondrial bioenergetics and mitochondrial ROS were suppressed. Il1a and Il1b induction with LPS or LPS+ATP challenge was diminished. Differential miRNA and mRNA profiles were observed. Lower miR-151-3p targeted cyclic adenosine monophosphate (cAMP)-mediated and AMP-activated protein kinase signaling pathways; higher miR-6909-5p, miR-7044-5p, and miR-7686-5p targeted Wnt beta-catenin signaling, retinoic acid receptor activation, apoptosis, signal transducer and activator of transcription 3, IL-22, IL-12, and IL-10 signaling. Functional enrichment analysis identified apoptosis and cell survival canonical pathways. CONCLUSION Select mitotoxic tri-organotins disrupted murine macrophage transcriptional response to LPS, yet triggered inflammasome activation. The differential response pattern suggested unique functional changes in the inflammatory response that may translate to suppressed host defense or prolong inflammation. We posit a framework to examine immune cell effects of environmental mitotoxic compounds for adverse health outcomes. https://doi.org/10.1289/EHP8314.
Collapse
Affiliation(s)
- Gabrielle M. Childers
- Molecular Toxicology Branch, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina, USA
| | - Caroline A. Perry
- Molecular Toxicology Branch, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina, USA
| | - Barbara Blachut
- Molecular Toxicology Branch, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina, USA
| | - Negin Martin
- Laboratory of Neurobiology, NIEHS, NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Carl D. Bortner
- Signal Transduction Laboratory, NIEHS, NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Stella Sieber
- Molecular Genomics Core Laboratory, NIEHS, NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Jian-Liang Li
- Integrative Bioinformatics Support Group, NIEHS, NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Michael B. Fessler
- Immunity, Inflammation, and Disease Laboratory, NIEHS, NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - G. Jean Harry
- Molecular Toxicology Branch, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina, USA
| |
Collapse
|
41
|
Implication of the NLRP3 Inflammasome in Bovine Age-Related Sarcopenia. Int J Mol Sci 2021; 22:ijms22073609. [PMID: 33808510 PMCID: PMC8036417 DOI: 10.3390/ijms22073609] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/18/2021] [Accepted: 03/29/2021] [Indexed: 12/20/2022] Open
Abstract
Sarcopenia is defined as the age-related loss of skeletal muscle mass, quality, and strength. The pathophysiological mechanisms underlying sarcopenia are still not completely understood. The aim of this work was to evaluate, for the first time, the expression of NLRP3 inflammasome in bovine skeletal muscle in order to investigate the hypothesis that inflammasome activation may trigger and sustain a pro-inflammatory environment leading to sarcopenia. Samples of skeletal muscle were collected from 60 cattle belonging to three age-based groups. Morphologic, immunohistochemical and molecular analysis were performed to assess the presence of age-related pathologic changes and chronic inflammation, the expression of NLRP3 inflammasome and to determine the levels of interleukin-1β, interleukin-18 and tumor necrosis factor alpha in muscle tissue. Our results revealed the presence of morphologic sarcopenia hallmark, chronic lymphocytic inflammation and a type II fibers-selective NLRP3 expression associated to a significant decreased number of immunolabeled-fibers in aged animals. Moreover, we found a statistically significant age-related increase of pro-inflammatory cytokines such as interleukin-1β and interleukin-18 suggesting the activation of NLRP3 inflammasome. Taken together, our data suggest that NLRP3 inflammasome components may be normally expressed in skeletal muscle, but its priming and activation during aging may contribute to enhance a pro-inflammatory environment altering normal muscular anabolism and metabolism.
Collapse
|
42
|
Jorquera G, Russell J, Monsalves-Álvarez M, Cruz G, Valladares-Ide D, Basualto-Alarcón C, Barrientos G, Estrada M, Llanos P. NLRP3 Inflammasome: Potential Role in Obesity Related Low-Grade Inflammation and Insulin Resistance in Skeletal Muscle. Int J Mol Sci 2021; 22:ijms22063254. [PMID: 33806797 PMCID: PMC8005007 DOI: 10.3390/ijms22063254] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/07/2021] [Accepted: 03/10/2021] [Indexed: 02/07/2023] Open
Abstract
Among multiple mechanisms, low-grade inflammation is critical for the development of insulin resistance as a feature of type 2 diabetes. The nucleotide-binding oligomerization domain-like receptor family (NOD-like) pyrin domain containing 3 (NLRP3) inflammasome has been linked to the development of insulin resistance in various tissues; however, its role in the development of insulin resistance in the skeletal muscle has not been explored in depth. Currently, there is limited evidence that supports the pathological role of NLRP3 inflammasome activation in glucose handling in the skeletal muscle of obese individuals. Here, we have centered our focus on insulin signaling in skeletal muscle, which is the main site of postprandial glucose disposal in humans. We discuss the current evidence showing that the NLRP3 inflammasome disturbs glucose homeostasis. We also review how NLRP3-associated interleukin and its gasdermin D-mediated efflux could affect insulin-dependent intracellular pathways. Finally, we address pharmacological NLRP3 inhibitors that may have a therapeutical use in obesity-related metabolic alterations.
Collapse
Affiliation(s)
- Gonzalo Jorquera
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (G.J.); (G.C.)
| | - Javier Russell
- Escuela de Pedagogía en Educación Física, Facultad de Educación, Universidad Autónoma de Chile, Santiago 8900000, Chile;
| | - Matías Monsalves-Álvarez
- Instituto de Ciencias de la Salud, Universidad de O’Higgins, Rancagua 2820000, Chile; (M.M.-Á.); (D.V.-I.)
| | - Gonzalo Cruz
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (G.J.); (G.C.)
| | - Denisse Valladares-Ide
- Instituto de Ciencias de la Salud, Universidad de O’Higgins, Rancagua 2820000, Chile; (M.M.-Á.); (D.V.-I.)
| | - Carla Basualto-Alarcón
- Departamento de Ciencias de la Salud, Universidad de Aysén, Coyhaique 5951537, Chile;
- Departamento de Anatomía y Medicina Legal, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Genaro Barrientos
- Programa de Fisiología y Biofísica, ICBM, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (G.B.); (M.E.)
- Centro de Estudios en Ejercicio, Metabolismo y Cáncer, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Manuel Estrada
- Programa de Fisiología y Biofísica, ICBM, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (G.B.); (M.E.)
| | - Paola Llanos
- Centro de Estudios en Ejercicio, Metabolismo y Cáncer, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
- Facultad de Odontología, Instituto de Investigación en Ciencias Odontológicas, Universidad de Chile, Santiago 8380544, Chile
- Correspondence: ; Tel.: +56-229-781-727
| |
Collapse
|
43
|
Inflammasome in ALS Skeletal Muscle: NLRP3 as a Potential Biomarker. Int J Mol Sci 2021; 22:ijms22052523. [PMID: 33802349 PMCID: PMC7959138 DOI: 10.3390/ijms22052523] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/19/2021] [Accepted: 02/26/2021] [Indexed: 02/07/2023] Open
Abstract
Since NLRP3 inflammasome plays a pivotal role in several neurodegenerative disorders, we hypothesized that levels of inflammasome components could help in diagnosis or prognosis of amyotrophic lateral sclerosis (ALS). Gene and protein expression was assayed by RT-PCR and Western blot. Spearman's correlation coefficient was used to determine the linear correlation of transcriptional expression levels with longevity throughout disease progression in mice models. Kaplan-Meier analysis was performed to evaluate MCC950 effects (NLRP3 inhibitor) on lifespan of SOD1G93A mice. The results showed significant alterations in NLRP3 inflammasome gene and protein levels in the skeletal muscle of SOD1G93A mice. Spearman's correlation coefficient revealed a positive association between Nlrp3 transcriptional levels in skeletal muscle and longevity of SOD1G93A mice (r = 0.506; p = 0.027). Accordingly, NLRP3 inactivation with MCC950 decreased the lifespan of mice. Furthermore, NLRP3 mRNA levels were significantly elevated in the blood of ALS patients compared to healthy controls (p = 0.03). In conclusion, NLRP3 could be involved in skeletal muscle pathogenesis of ALS, either through inflammasome or independently, and may play a dual role during disease progression. NLRP3 gene expression levels could be used as a biomarker to improve diagnosis and prognosis in skeletal muscle from animal models and also to support diagnosis in clinical practice with the blood of ALS patients.
Collapse
|
44
|
Recent insights on modulation of inflammasomes by adipokines: a critical event for the pathogenesis of obesity and metabolism-associated diseases. Arch Pharm Res 2020; 43:997-1016. [PMID: 33078304 DOI: 10.1007/s12272-020-01274-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/13/2020] [Indexed: 12/17/2022]
Abstract
Aberrant production of adipokines, a group of adipocytes-derived hormones, is considered one of the most important pathological characteristics of obesity. In individuals with obesity, beneficial adipokines, such as adiponectin are downregulated, whereas leptin and other pro-inflammatory adipokines are highly upregulated. Hence, the imbalance in levels of these adipokines is thought to promote the development of obesity-linked complications. However, the mechanisms by which adipokines contribute to the pathogenesis of various diseases have not been clearly understood. Inflammasomes represent key signaling platform that triggers the inflammatory and immune responses through the processing of the interleukin family of pro-inflammatory cytokines in a caspase-1-dependent manner. Beyond their traditional function as a component of the innate immune system, inflammasomes have been recently integrated into the pathological process of multiple metabolism- and obesity-related disorders such as cardiovascular diseases, diabetes, fatty liver disease, and cancer. Interestingly, emerging evidence also highlights the role of adipokines in the modulation of inflammasomes activation, making it a promising mechanism underlying distinct biological actions of adipokines in diseases driven by inflammation and metabolic disorders. In this review, we summarize the effects of adipokines, in particular adiponectin, leptin, visfatin and apelin, on inflammasomes activation and their implications in the pathophysiology of obesity-linked complications.
Collapse
|
45
|
Lau YS, Zhao L, Zhang C, Li H, Han R. Genetic disruption of the inflammasome adaptor ASC has minimal impact on the pathogenesis of Duchenne muscular dystrophy in mdx mice. Life Sci 2020; 257:118069. [PMID: 32659370 PMCID: PMC7484256 DOI: 10.1016/j.lfs.2020.118069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 06/23/2020] [Accepted: 07/06/2020] [Indexed: 12/24/2022]
Abstract
AIM Up-regulation of inflammasome proteins was reported in dystrophin-deficient muscles. However, it remains to be determined whether inflammasome activation plays a role in the pathogenesis of Duchenne muscular dystrophy. This study was therefore set out to investigate whether genetic disruption of the inflammasome pathway impacts the disease progression in mdx mice. MAIN METHODS Mice deficient in both dystrophin and ASC (encoded by Pycard [PYD And CARD Domain Containing]) were generated. The impact of ASC deficiency on muscular dystrophy of mdx mice were assessed by measurements of serum cytokines, Western blot, real-time PCR and histopathological staining. KEY FINDINGS The pro-inflammatory cytokines such as TNF-α, IL-6, KC/GRO and IL-10 were markedly increased in the sera of 8-week-old mdx mice compared to WT. Western blotting showed that P2X7, caspase-1, ASC and IL-18 were upregulated. Disruption of ASC and dystrophin expression in the mdx/ASC-/- mice was verified by Western blot analysis. Histopathological analysis did not find significant alterations in the muscular dystrophy phenotype in mdx/ASC-/- mice as compared to mdx mice. SIGNIFICANCE Taken together, our results show that disruption of the central adaptor ASC of the inflammasome is insufficient to alleviate muscular dystrophy phenotype in mdx mice.
Collapse
Affiliation(s)
- Yeh Siang Lau
- Department of Surgery, Davis Heart and Lung Research Institute, Biomedical Sciences Graduate Program, Biophysics Graduate Program, The Ohio State University Wexner Medical Center, Columbus, OH 43210, United States
| | - Lixia Zhao
- Department of Surgery, Davis Heart and Lung Research Institute, Biomedical Sciences Graduate Program, Biophysics Graduate Program, The Ohio State University Wexner Medical Center, Columbus, OH 43210, United States
| | - Chen Zhang
- Department of Surgery, Davis Heart and Lung Research Institute, Biomedical Sciences Graduate Program, Biophysics Graduate Program, The Ohio State University Wexner Medical Center, Columbus, OH 43210, United States
| | - Haiwen Li
- Department of Surgery, Davis Heart and Lung Research Institute, Biomedical Sciences Graduate Program, Biophysics Graduate Program, The Ohio State University Wexner Medical Center, Columbus, OH 43210, United States
| | - Renzhi Han
- Department of Surgery, Davis Heart and Lung Research Institute, Biomedical Sciences Graduate Program, Biophysics Graduate Program, The Ohio State University Wexner Medical Center, Columbus, OH 43210, United States.
| |
Collapse
|
46
|
Liu Y, Bi X, Zhang Y, Wang Y, Ding W. Mitochondrial dysfunction/NLRP3 inflammasome axis contributes to angiotensin II-induced skeletal muscle wasting via PPAR-γ. J Transl Med 2020; 100:712-726. [PMID: 31857693 DOI: 10.1038/s41374-019-0355-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 10/19/2019] [Accepted: 11/04/2019] [Indexed: 12/15/2022] Open
Abstract
Angiotensin II (Ang II) levels are elevated in patients with chronic kidney disease or heart failure, and directly causes skeletal muscle wasting in rodents, but the molecular mechanisms of Ang II-induced skeletal muscle wasting and its potential as a therapeutic target are unknown. We investigated the NLR family pyrin domain containing 3 (NLRP3) inflammasome-mediated muscle atrophy response to Ang II in C2C12 myotubes and Nlrp3 knockout mice. We also assessed the mitochondrial dysfunction (MtD)/NLRP3 inflammasome axis in Ang II-induced C2C12 myotubes. Finally, we examined whether a peroxisome proliferator-activated receptor-γ (PPAR-γ) agonist could attenuate skeletal muscle wasting by targeting the MtD/NLRP3 inflammasome axis in vitro and in vivo. We demonstrated that Ang II increased NLRP3 inflammasome activation in cultured C2C12 myotubes dose dependently. Nlrp3 knockdown or Nlrp3-/- mice were protected from the imbalance of protein synthesis and degradation. Exposure of C2C12 to Ang II increased mitochondrial ROS (mtROS) generation, accompanied by MtD. Remarkably, the mitochondrial-targeted antioxidant not only decreased mtROS and MtD, it also significantly inhibited NLRP3 inflammasome activation and restored skeletal muscle atrophy. Finally, the PPAR-γ agonist protected against Ang II-induced muscle wasting by preventing MtD, oxidative stress, and NLRP3 inflammasome activation in vitro and in vivo. This work suggests a potential role of MtD/NLRP3 inflammasome pathway in the pathogenesis of Ang II-induced skeletal muscle wasting, and targeting the PPAR-γ/MtD/NLRP3 inflammasome axis may provide a therapeutic approach for muscle wasting.
Collapse
Affiliation(s)
- Yuqing Liu
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, 639 zhizaoju Road, 200011, Shanghai, China
| | - Xiao Bi
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, 639 zhizaoju Road, 200011, Shanghai, China
| | - Yumei Zhang
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, 639 zhizaoju Road, 200011, Shanghai, China
| | - Yingdeng Wang
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, 639 zhizaoju Road, 200011, Shanghai, China
| | - Wei Ding
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, 639 zhizaoju Road, 200011, Shanghai, China.
| |
Collapse
|
47
|
Abou-Samra M, Selvais CM, Dubuisson N, Brichard SM. Adiponectin and Its Mimics on Skeletal Muscle: Insulin Sensitizers, Fat Burners, Exercise Mimickers, Muscling Pills … or Everything Together? Int J Mol Sci 2020; 21:ijms21072620. [PMID: 32283840 PMCID: PMC7178193 DOI: 10.3390/ijms21072620] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/01/2020] [Accepted: 04/03/2020] [Indexed: 02/07/2023] Open
Abstract
Adiponectin (ApN) is a hormone abundantly secreted by adipocytes and it is known to be tightly linked to the metabolic syndrome. It promotes insulin-sensitizing, fat-burning, and anti-atherosclerotic actions, thereby effectively counteracting several metabolic disorders, including type 2 diabetes, obesity, and cardiovascular diseases. ApN is also known today to possess powerful anti-inflammatory/oxidative and pro-myogenic effects on skeletal muscles exposed to acute or chronic inflammation and injury, mainly through AdipoR1 (ApN specific muscle receptor) and AMP-activated protein kinase (AMPK) pathway, but also via T-cadherin. In this review, we will report all the beneficial and protective properties that ApN can exert, specifically on the skeletal muscle as a target tissue. We will highlight its effects and mechanisms of action, first in healthy skeletal muscle including exercised muscle, and second in diseased muscle from a variety of pathological conditions. In the end, we will go over some of AdipoRs agonists that can be easily produced and administered, and which can greatly mimic ApN. These interesting and newly identified molecules could pave the way towards future therapeutic approaches to potentially prevent or combat not only skeletal muscle disorders but also a plethora of other diseases with sterile inflammation or metabolic dysfunction.
Collapse
|
48
|
Light exercise without lactate elevation induces ischemic tolerance through the modulation of microRNA in the gerbil hippocampus. Brain Res 2020; 1732:146710. [PMID: 32035888 DOI: 10.1016/j.brainres.2020.146710] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 01/31/2020] [Accepted: 02/05/2020] [Indexed: 12/11/2022]
Abstract
Previously we studied the possible neuroprotective effects of ischemia-resistant exercise in a gerbil model of transient whole-brain ischemia and evaluated the histology, expression of specific proteins, and brain function under different conditions. The present study investigated the neuroprotective effects of light exercise, without lactate elevation, in a gerbil model of ischemia/reperfusion injury. Transient whole-brain ischemia was induced by occlusion of the bilateral common carotid arteries for 5 min. A group of animals was subjected to treadmill exercise before ischemia induction. Hippocampal neuronal damage and miRNA expression, as well as behavioral deficits and plasma lactate levels, were evaluated. Light exercise suppressed hippocampal neuron loss and preserved short-term memory. Moreover, 14 miRNAs (mmu-miR-211-3p, -327, -451b, -711, -3070-3p, -3070-2-3p, -3097-5p, -3620-5p, -6240, -6916-5p, -6944-5p, 7083-5p, -7085-5p, and -7674-5p) were upregulated and 6 miRNAs (mmu-miR-148b-3p, -152-3p, -181c-5p, -299b-5p, -455-3p, and -664-3p) were downregulated due to ischemia. However, the expression of these miRNAs remained unchanged when animals performed light exercise before the ischemic event. Differentially expressed miRNAs regulate multiple biological processes such as inflammation, metabolism, and cell death. These findings suggest that light exercise reduces neuronal death and behavioral deficits after transient ischemia by regulating hippocampal miRNAs.
Collapse
|
49
|
Qu XY, Zhang YM, Tao LN, Gao H, Zhai JH, Sun JM, Song YQ, Zhang SX. XingNaoJing injections protect against cerebral ischemia/reperfusion injury and alleviate blood-brain barrier disruption in rats, through an underlying mechanism of NLRP3 inflammasomes suppression. Chin J Nat Med 2020; 17:498-505. [PMID: 31514981 DOI: 10.1016/s1875-5364(19)30071-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Indexed: 12/15/2022]
Abstract
The aim of this study was to explore the neuroprotective effect and mechanism of XingNaoJing injections (XNJ) on cerebral ischemia injury and blood-brain barrier (BBB) disruption. Middle cerebral artery occlusion (MCAO) method was applicated to establish the model of cerebral ischemia/reperfusion (I/R) injury in rats. BBB permeability after I/R injury was assessed with the leaking amount of Evans Blue and the expression of occludin and ZO-1. The expression of NOD-like receptor family, pyrin domain containing (NLRP3) was checked to explore the inhibition of inflammation by XNJ. The results showed that XNJ could significantly increase the survival percent, decrease the infarct area and ameliorate neurological deficits and brain damage after I/R injury. Leaking amount of Evans Blue was reduced by XNJ, and the expression of tight junction protein, occludin and ZO-1 was also up-regulated by XNJ, which showed a role of protection on BBB disruption. The expression of NLRP3 was inhibited after exposure of XNJ, which was associated with inhibition of the inflammatory response. In summary, XNJ could suppress NLRP3 inflammasomes and improve BBB disruption and brain damage in rats after cerebral I/R injury, which provided a beneficial insight to further explore XNJ.
Collapse
Affiliation(s)
- Xiao-Yu Qu
- Department of Pharmacy, the First Hospital of Jilin University, Changchun 130021, China
| | - Yue-Ming Zhang
- Department of Pharmacy, the First Hospital of Jilin University, Changchun 130021, China
| | - Li-Na Tao
- Department of Pharmacy, the First Hospital of Jilin University, Changchun 130021, China
| | - Huan Gao
- Department of Pharmacy, the First Hospital of Jilin University, Changchun 130021, China
| | - Jing-Hui Zhai
- Department of Pharmacy, the First Hospital of Jilin University, Changchun 130021, China
| | - Jing-Meng Sun
- Department of Pharmacy, the First Hospital of Jilin University, Changchun 130021, China
| | - Yan-Qing Song
- Department of Pharmacy, the First Hospital of Jilin University, Changchun 130021, China.
| | - Si-Xi Zhang
- Department of Pharmacy, the First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
50
|
Zamani P, Oskuee RK, Atkin SL, Navashenaq JG, Sahebkar A. MicroRNAs as important regulators of the NLRP3 inflammasome. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2020; 150:50-61. [PMID: 31100298 DOI: 10.1016/j.pbiomolbio.2019.05.004] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 05/13/2019] [Indexed: 12/28/2022]
Abstract
Inflammasomes are a group of cytosolic multi-protein signaling complexes that regulate maturation of the interleukin (IL)-1 family cytokines IL-1β and IL-18 through activation of inflammatory caspase-1. The NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome is the best characterized and consists of several key components that are assembled and activated in response to different endogenous and exogenous signals. The NLRP3 inflammasome is common to a number of human inflammatory diseases and its targeting may lead to novel anti-inflammatory therapy. NLRP3 inflammasome activation is tightly regulated by different mechanisms especially post-transcriptional modulation via microRNAs (miRNA). MicroRNAs are small endogenous noncoding RNAs that are 21-23 nucleotides in length and control the expression of various genes through binding to the 3'-untranslated regions of the respective mRNA and subsequent post-transcriptional regulation. MicroRNAs have recently been recognized as crucial regulators of the NLRP3 inflammasome. In this review, we summarize the current understanding of the role of miRNAs in the regulation of NLRP3 inflammasome complexes and their impact on the pathogenesis of inflammatory disease processes.
Collapse
Affiliation(s)
- Parvin Zamani
- Nanotechnology Research Center, Student Research Committee, Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Kazemi Oskuee
- Targeted Drug Delivery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | | | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|