1
|
Shahbaz SK, Mokhlesi A, Sadegh RK, Rahimi K, Jamialahmadi T, Butler AE, Kesharwani P, Sahebkar A. TLR/NLRP3 inflammasome signaling pathways as a main target in frailty, cachexia and sarcopenia. Tissue Cell 2025; 93:102723. [PMID: 39823704 DOI: 10.1016/j.tice.2025.102723] [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: 11/07/2024] [Revised: 12/28/2024] [Accepted: 01/03/2025] [Indexed: 01/20/2025]
Abstract
Mobility disability is a common condition affecting older adults, making walking and the performance of activities of daily living difficult. Frailty, cachexia and sarcopenia are related conditions that occur with advancing age and are characterized by a decline in muscle mass, strength, and functionality that negatively impacts health. Chronic low-grade inflammation is a significant factor in the onset and progression of these conditions. The toll-like receptors (TLRs) and the NLRP3 inflammasome are the pathways of signaling that regulate inflammation. These pathways can potentially be targeted therapeutically for frailty, cachexia and sarcopenia as research has shown that dysregulation of the TLR/NLRP3 inflammasome signaling pathways is linked to these conditions. Activation of TLRs with pathogen-associated molecular patterns (PAMPs or DAMPs) results in chronic inflammation and tissue damage by releasing pro-inflammatory cytokines. Additionally, NLRP3 inflammasome activation enhances the inflammatory response by promoting the production and release of interleukins (ILs), thus exacerbating the underlying inflammatory mechanisms. These pathways are activated in the advancement of disease in frail and sarcopenic individuals. Targeting these pathways may offer therapeutic options to reduce frailty, improve musculoskeletal resilience and prevent or reverse cachexia-associated muscle wasting. Modulating TLR/NLRP3 inflammasome pathways may also hold promise in slowing down the progression of sarcopenia, preserving muscle mass and enhancing overall functional ability in elderly people. The aim of this review is to investigate the signaling pathways of the TLR/NLRP3 inflammasome as a main target in frailty, cachexia and sarcopenia.
Collapse
Affiliation(s)
- Sanaz Keshavarz Shahbaz
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran; USERN Office, Qazvin University of Medical Sciences, Qazvin, Iran.
| | - Aida Mokhlesi
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran; Social Determinants of Health Research Center, Research Institute for Prevention of Non-communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran; USERN Office, Qazvin University of Medical Sciences, Qazvin, Iran; Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Roghaye Keshavarz Sadegh
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran; USERN Office, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Kimia Rahimi
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran; USERN Office, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Tannaz Jamialahmadi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Amirhossein Sahebkar
- Center for Global health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Chen J, Jia S, Xue X, Guo C, Dong K. Gut microbiota: a novel target for exercise-mediated regulation of NLRP3 inflammasome activation. Front Microbiol 2025; 15:1476908. [PMID: 39834360 PMCID: PMC11743191 DOI: 10.3389/fmicb.2024.1476908] [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: 08/07/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025] Open
Abstract
The NOD-like receptor family pyrin domain-containing 3 (NLRP3) is a key pattern recognition receptor in the innate immune system. Its overactivation leads to the production of pro-inflammatory cytokines, such as IL-1β and IL-18, which contribute to the development and progression of various diseases. In recent years, evidence has shown that gut microbiota plays an important role in regulating the activation of NLRP3 inflammasome. Variations in the function and composition of gut microbiota can directly or indirectly influence NLRP3 inflammasome activation by influencing bacterial components and gut microbiota metabolites. Additionally, exercise has been shown to effectively reduce NLRP3 inflammasome overactivation while promoting beneficial changes in gut microbiota. This suggests that gut microbiota may play a key role in mediating the effects of exercise on NLRP3 inflammasome regulation. This review explores the impact of exercise on gut microbiota and NLRP3 inflammasome activation, and examines the mechanisms through which gut microbiota mediates the anti-inflammatory effects of exercise, providing new avenues for research.
Collapse
Affiliation(s)
- Jun Chen
- School of Graduate of Wuhan Sports University, Wuhan, China
| | - Shaohui Jia
- School of Sports Medicine of Wuhan Sports University, Wuhan, China
| | - Xinxuan Xue
- School of Sports Training of Wuhan Sports University, Wuhan, China
| | - Chenggeng Guo
- School of Sports Training of Wuhan Sports University, Wuhan, China
| | - Kunwei Dong
- School of Art of Wuhan Sports University, Wuhan, China
| |
Collapse
|
3
|
Hejazian SM, Hejazian SS, Mostafavi SM, Hosseiniyan SM, Montazersaheb S, Ardalan M, Zununi Vahed S, Barzegari A. Targeting cellular senescence in kidney diseases and aging: A focus on mesenchymal stem cells and their paracrine factors. Cell Commun Signal 2024; 22:609. [PMID: 39696575 DOI: 10.1186/s12964-024-01968-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
Cellular senescence is a phenomenon distinguished by the halting of cellular division, typically triggered by DNA injury or numerous stress-inducing factors. Cellular senescence is implicated in various pathological and physiological processes and is a hallmark of aging. The presence of accumulated senescent cells, whether transiently (acute senescence) or persistently (chronic senescence) plays a dual role in various conditions such as natural kidney aging and different kidney disorders. Elevations in senescent cells and senescence-associated secretory phenotype (SASP) levels correlate with decreased kidney function, kidney ailments, and age-related conditions. Strategies involving senotherapeutic agents like senolytics, senomorphics, and senoinflammation have been devised to specifically target senescent cells. Mesenchymal stem cells (MSCs) and their secreted factors may also offer alternative approaches for anti-senescence interventions. The MSC-derived secretome compromises significant therapeutic benefits in kidney diseases by facilitating tissue repair via anti-inflammatory, anti-fibrosis, anti-apoptotic, and pro-angiogenesis effects, thereby improving kidney function and mitigating disease progression. Moreover, by promoting the clearance of senescent cells or modulating their secretory profiles, MSCs could potentially reverse some age-related declines in kidney function.This review article intends to shed light on the present discoveries concerning the role of cellular senescence in kidney aging and diseases. Furthermore, it outlines the role of senotherapeutics utilized to alleviate kidney damage and aging. It also highlights the possible impact of MSCs secretome on mitigating kidney injury and prolonging lifespan across various models of kidney diseases as a novel senotherapy.
Collapse
Affiliation(s)
| | - Seyyed Sina Hejazian
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyyedeh Mina Mostafavi
- Ayatollah Taleghani Hospital, Research Development Unit, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Soheila Montazersaheb
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Abolfazl Barzegari
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
4
|
Zhang Y, Yu C, Li X. Kidney Aging and Chronic Kidney Disease. Int J Mol Sci 2024; 25:6585. [PMID: 38928291 PMCID: PMC11204319 DOI: 10.3390/ijms25126585] [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: 04/27/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
The process of aging inevitably leads to an increase in age-related comorbidities, including chronic kidney disease (CKD). In many aspects, CKD can be considered a state of accelerated and premature aging. Aging kidney and CKD have numerous common characteristic features, ranging from pathological presentation and clinical manifestation to underlying mechanisms. The shared mechanisms underlying the process of kidney aging and the development of CKD include the increase in cellular senescence, the decrease in autophagy, mitochondrial dysfunction, and the alterations of epigenetic regulation, suggesting the existence of potential therapeutic targets that are applicable to both conditions. In this review, we provide a comprehensive overview of the common characteristics between aging kidney and CKD, encompassing morphological changes, functional alterations, and recent advancements in understanding the underlying mechanisms. Moreover, we discuss potential therapeutic strategies for targeting senescent cells in both the aging process and CKD.
Collapse
Affiliation(s)
- Yingying Zhang
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Chen Yu
- Department of Nephrology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai 200092, China;
| | - Xiaogang Li
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
5
|
Cuccurullo SJ, Fleming TK, Petrosyan H, Hanley DF, Raghavan P. Mechanisms and benefits of cardiac rehabilitation in individuals with stroke: emerging role of its impact on improving cardiovascular and neurovascular health. Front Cardiovasc Med 2024; 11:1376616. [PMID: 38756753 PMCID: PMC11096558 DOI: 10.3389/fcvm.2024.1376616] [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: 01/25/2024] [Accepted: 04/17/2024] [Indexed: 05/18/2024] Open
Abstract
Human and animal studies have demonstrated the mechanisms and benefits of aerobic exercise for both cardiovascular and neurovascular health. Aerobic exercise induces neuroplasticity and neurophysiologic reorganization of brain networks, improves cerebral blood flow, and increases whole-body VO2peak (peak oxygen consumption). The effectiveness of a structured cardiac rehabilitation (CR) program is well established and a vital part of the continuum of care for people with cardiovascular disease. Individuals post stroke exhibit decreased cardiovascular capacity which impacts their neurologic recovery and extends disability. Stroke survivors share the same risk factors as patients with cardiac disease and can therefore benefit significantly from a comprehensive CR program in addition to neurorehabilitation to address their cardiovascular health. The inclusion of individuals with stroke into a CR program, with appropriate adaptations, can significantly improve their cardiovascular health, promote functional recovery, and reduce future cardiovascular and cerebrovascular events thereby reducing the economic burden of stroke.
Collapse
Affiliation(s)
- Sara J. Cuccurullo
- Department of Physical Medicine and Rehabilitation, JFK Johnson Rehabilitation Institute at Hackensack Meridian Health, Edison, NJ, United States
| | - Talya K. Fleming
- Department of Physical Medicine and Rehabilitation, JFK Johnson Rehabilitation Institute at Hackensack Meridian Health, Edison, NJ, United States
| | - Hayk Petrosyan
- Department of Physical Medicine and Rehabilitation, JFK Johnson Rehabilitation Institute at Hackensack Meridian Health, Edison, NJ, United States
| | - Daniel F. Hanley
- Brain Injury Outcomes, Johns Hopkins Medical Institutions, Baltimore, MD, United States
| | - Preeti Raghavan
- Department of Physical Medicine and Rehabilitation and Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
6
|
Troutman AD, Arroyo E, Sheridan EM, D'Amico DJ, Brandt PR, Hinrichs R, Chen X, Lim K, Avin KG. Skeletal muscle atrophy in clinical and preclinical models of chronic kidney disease: A systematic review and meta-analysis. J Cachexia Sarcopenia Muscle 2024; 15:21-35. [PMID: 38062879 PMCID: PMC10834351 DOI: 10.1002/jcsm.13400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 09/12/2023] [Accepted: 11/02/2023] [Indexed: 02/03/2024] Open
Abstract
Patients with chronic kidney disease (CKD) are often regarded as experiencing wasting of muscle mass and declining muscle strength and function, collectively termed sarcopenia. The extent of skeletal muscle wasting in clinical and preclinical CKD populations is unclear. We evaluated skeletal muscle atrophy in preclinical and clinical models of CKD, with multiple sub-analyses for muscle mass assessment methods, CKD severity, sex and across the different preclinical models of CKD. We performed a systematic literature review of clinical and preclinical studies that measured muscle mass/size using the following databases: Ovid Medline, Embase and Scopus. A random effects meta-analysis was utilized to determine standard mean difference (SMD; Hedges' g) between healthy and CKD. Heterogeneity was evaluated using the I2 statistic. Preclinical study quality was assessed via the Systematic Review Centre for Laboratory Animal Experimentation and clinical studies quality was assessed via the Newcastle-Ottawa Scale. This study was registered in PROSPERO (CRD42020180737) prior to initiation of the search. A total of 111 studies were included in this analysis using the following subgroups: 106 studies in the primary CKD analysis, 18 studies that accounted for diabetes and 7 kidney transplant studies. Significant atrophy was demonstrated in 78% of the preclinical studies and 49% of the clinical studies. The random effects model demonstrated a medium overall SMD (SMD = 0.58, 95% CI = 0.52-0.64) when combining clinical and preclinical studies, a medium SMD for the clinical population (SMD = 0.48, 95% CI = 0.42-0.55; all stages) and a large SMD for preclinical CKD (SMD = 0.95, 95% CI = 0.76-1.14). Further sub-analyses were performed based upon assessment methods, disease status and animal model. Muscle atrophy was reported in 49% of the clinical studies, paired with small mean differences. Preclinical studies reported significant atrophy in 78% of studies, with large mean differences. Across multiple clinical sub-analyses such as severity of CKD, dialysis modality and diabetes, a medium mean difference was found. Sub-analyses in both clinical and preclinical studies found a large mean difference for males and medium for females suggesting sex-specific implications. Muscle atrophy differences varied based upon assessment method for clinical and preclinical studies. Limitations in study design prevented conclusions to be made about the extent of muscle loss with disease progression, or the impact of dialysis. Future work would benefit from the use of standardized measurement methods and consistent clinical staging to improve our understanding of atrophy changes in CKD progression, and analysis of biological sex differences.
Collapse
Affiliation(s)
- Ashley D Troutman
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University Purdue University, Indianapolis, Indiana, USA
| | - Eliott Arroyo
- Department of Medicine, Division of Nephrology & Hypertension, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Elizabeth M Sheridan
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University Purdue University, Indianapolis, Indiana, USA
| | - Duncan J D'Amico
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University Purdue University, Indianapolis, Indiana, USA
| | - Peyton R Brandt
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University Purdue University, Indianapolis, Indiana, USA
| | - Rachel Hinrichs
- University Library, Indiana University-Purdue University Indianapolis, Indiana, USA
| | - Xiwei Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Indiana University Bloomington, Bloomington, Indiana, USA
| | - Kenneth Lim
- Department of Medicine, Division of Nephrology & Hypertension, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Keith G Avin
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University Purdue University, Indianapolis, Indiana, USA
- Department of Medicine, Division of Nephrology & Hypertension, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
7
|
Tian R, Chang L, Zhang Y, Zhang H. Development and validation of a nomogram model for predicting low muscle mass in patients undergoing hemodialysis. Ren Fail 2023; 45:2231097. [PMID: 37408481 PMCID: PMC10324438 DOI: 10.1080/0886022x.2023.2231097] [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: 02/09/2023] [Accepted: 06/24/2023] [Indexed: 07/07/2023] Open
Abstract
BACKGROUND Muscle mass is important in determining patients' nutritional status. However, measurement of muscle mass requires special equipment that is inconvenient for clinical use. We aimed to develop and validate a nomogram model for predicting low muscle mass in patients undergoing hemodialysis (HD). METHODS A total of 346 patients undergoing HD were enrolled and randomly divided into a 70% training set and a 30% validation set. The training set was used to develop the nomogram model, and the validation set was used to validate the developed model. The performance of the nomogram was assessed using the receiver operating characteristic (ROC) curve, a calibration curve, and the Hosmer-Lemeshow test. A decision curve analysis (DCA) was used to evaluate the clinical practicality of the nomogram model. RESULTS Age, sex, body mass index (BMI), handgrip strength (HGS), and gait speed (GS) were included in the nomogram for predicting low skeletal muscle mass index (LSMI). The diagnostic nomogram model exhibited good discrimination with an area under the ROC curve (AUC) of 0.906 (95% CI, 0.862-0.940) in the training set and 0.917 (95% CI, 0.846-0.962) in the validation set. The calibration analysis also showed excellent results. The nomogram demonstrated a high net benefit in the clinical decision curve for both sets. CONCLUSIONS The prediction model included age, sex, BMI, HGS, and GS, and it can successfully predict the presence of LSMI in patients undergoing HD. This nomogram provides an accurate visual tool for medical staff for prediction, early intervention, and graded management.
Collapse
Affiliation(s)
- Rongrong Tian
- Department of Blood Purification Centre, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Liyang Chang
- Department of Blood Purification Centre, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Ying Zhang
- Department of Science and Development, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Hongmei Zhang
- Department of Blood Purification Centre, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
8
|
Soto ME, Pérez-Torres I, Rubio-Ruiz ME, Cano-Martínez A, Manzano-Pech L, Guarner-Lans V. Frailty and the Interactions between Skeletal Muscle, Bone, and Adipose Tissue-Impact on Cardiovascular Disease and Possible Therapeutic Measures. Int J Mol Sci 2023; 24:ijms24054534. [PMID: 36901968 PMCID: PMC10003713 DOI: 10.3390/ijms24054534] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/18/2023] [Accepted: 02/24/2023] [Indexed: 03/02/2023] Open
Abstract
Frailty is a global health problem that impacts clinical practice. It is complex, having a physical and a cognitive component, and it is the result of many contributing factors. Frail patients have oxidative stress and elevated proinflammatory cytokines. Frailty impairs many systems and results in a reduced physiological reserve and increased vulnerability to stress. It is related to aging and to cardiovascular diseases (CVD). There are few studies on the genetic factors of frailty, but epigenetic clocks determine age and frailty. In contrast, there is genetic overlap of frailty with cardiovascular disease and its risk factors. Frailty is not yet considered a risk factor for CVD. It is accompanied by a loss and/or poor functioning of muscle mass, which depends on fiber protein content, resulting from the balance between protein breakdown and synthesis. Bone fragility is also implied, and there is a crosstalk between adipocytes, myocytes, and bone. The identification and assessment of frailty is difficult, without there being a standard instrument to identify or treat it. Measures to prevent its progression include exercises, as well as supplementing the diet with vitamin D and K, calcium, and testosterone. In conclusion, more research is needed to better understand frailty and to avoid complications in CVD.
Collapse
Affiliation(s)
- María Elena Soto
- Department of Endocrinology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - Israel Pérez-Torres
- Department of Cardiovascular Biomedicine, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - María Esther Rubio-Ruiz
- Department of Physiology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - Agustina Cano-Martínez
- Department of Physiology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - Linaloe Manzano-Pech
- Department of Cardiovascular Biomedicine, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - Verónica Guarner-Lans
- Department of Physiology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
- Correspondence:
| |
Collapse
|
9
|
Anderson EM, Kim K, Fazzone BJ, Harland KC, Hu Q, Salyers Z, Palzkill VR, Cort TA, Kunz EM, Martin AJ, Neal D, O’Malley KA, Berceli SA, Ryan TE, Scali ST. Influences of renal insufficiency and ischemia on mitochondrial bioenergetics and limb dysfunction in a novel murine iliac arteriovenous fistula model. JVS Vasc Sci 2022; 3:345-362. [PMID: 36439698 PMCID: PMC9692039 DOI: 10.1016/j.jvssci.2022.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 10/06/2022] [Indexed: 11/05/2022] Open
Abstract
Objective Hand disability after hemodialysis access surgery has been common yet has remained poorly understood. Arteriovenous fistula (AVF) hemodynamic perturbations have not reliably correlated with the observed measures of hand function. Chronic kidney disease (CKD) is known to precipitate myopathy; however, the interactive influences of renal insufficiency and ischemia on limb outcomes have remained unknown. We hypothesized that CKD would contribute to access-related hand dysfunction via altered mitochondrial bioenergetics. Using a novel murine AVF model, we sought to characterize the skeletal muscle outcomes in mice with and without renal insufficiency. Methods Male, 8-week-old C57BL/6J mice were fed either an adenine-supplemented diet to induce renal insufficiency (CKD) or a casein-based control chow (CON). After 2 weeks of dietary intervention, the mice were randomly assigned to undergo iliac AVF surgery (n = 12/group) or a sham operation (n = 5/group). Measurements of aortoiliac hemodynamics, hindlimb perfusion, and hindlimb motor function were collected for 2 weeks. The mice were sacrificed on postoperative day 14 to assess skeletal muscle histopathologic features and mitochondrial function. To assess the late outcome trends, 20 additional mice had undergone CKD induction and sham (n = 5) or AVF (n = 15) surgery and followed up for 6 weeks postoperatively before sacrifice. Results The adenine-fed mice had had a significantly reduced glomerular filtration rate and elevated blood urea nitrogen, confirming the presence of CKD. The sham mice had a 100% survival rate and AVF cohorts an 82.1% survival rate with an 84.4% AVF patency rate. The aorta and inferior vena cava velocity measurements and the vessel diameter had increased after AVF creation (P < .0001 vs sham). The AVF groups had had a 78.4% deficit in paw perfusion compared with the contralateral limb after surgery (P < .0001 vs sham). Mitochondrial function was influenced by the presence of CKD. The respiratory capacity of the CKD-sham mice (8443 ± 1509 pmol/s/mg at maximal energy demand) was impaired compared with that of the CON-sham mice (12,870 ± 1203 pmol/s/mg; P = .0001). However, this difference was muted after AVF creation (CKD-AVF, 4478 ± 3685 pmol/s/mg; CON-AVF, 5407 ± 3582 pmol/s/mg; P = .198). The AVF cohorts had had impairments in grip strength (vs sham; P < .0001) and gait (vs sham; P = .012). However, the presence of CKD did not significantly alter the measurements of gross muscle function. The paw perfusion deficits had persisted 6 weeks postoperatively for the AVF mice (P < .0001 vs sham); however, the myopathy had resolved (grip strength, P = .092 vs sham; mitochondrial respiration, P = .108 vs sham). Conclusions CKD and AVF-induced distal limb ischemia both impaired skeletal muscle mitochondrial function. Renal insufficiency was associated with a baseline myopathy that was exacerbated by the acute ischemic injury resulting from AVF creation. However, ischemia was the primary driver of the observed phenotype of gross motor impairment. This model reliably reproduced the local and systemic influences that contribute to access-related hand dysfunction and provides a platform for further mechanistic and therapeutic investigation.
Collapse
Affiliation(s)
- Erik M. Anderson
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, FL
- Malcolm Randall Veterans Affairs Medical Center, Gainesville, FL
| | - Kyoungrae Kim
- Department of Applied Physiology and Kinesiology, Center for Exercise Science, University of Florida, Gainesville, FL
| | - Brian J. Fazzone
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, FL
- Malcolm Randall Veterans Affairs Medical Center, Gainesville, FL
| | - Kenneth C. Harland
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, FL
- Malcolm Randall Veterans Affairs Medical Center, Gainesville, FL
| | - Qiongyao Hu
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, FL
- Malcolm Randall Veterans Affairs Medical Center, Gainesville, FL
| | - Zach Salyers
- Department of Applied Physiology and Kinesiology, Center for Exercise Science, University of Florida, Gainesville, FL
| | - Victoria R. Palzkill
- Department of Applied Physiology and Kinesiology, Center for Exercise Science, University of Florida, Gainesville, FL
| | - Tomas A. Cort
- Department of Applied Physiology and Kinesiology, Center for Exercise Science, University of Florida, Gainesville, FL
| | - Eric M. Kunz
- Department of Applied Physiology and Kinesiology, Center for Exercise Science, University of Florida, Gainesville, FL
| | - Andrew J. Martin
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, FL
- Malcolm Randall Veterans Affairs Medical Center, Gainesville, FL
| | - Dan Neal
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, FL
| | - Kerri A. O’Malley
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, FL
- Malcolm Randall Veterans Affairs Medical Center, Gainesville, FL
| | - Scott A. Berceli
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, FL
- Malcolm Randall Veterans Affairs Medical Center, Gainesville, FL
| | - Terence E. Ryan
- Department of Applied Physiology and Kinesiology, Center for Exercise Science, University of Florida, Gainesville, FL
| | - Salvatore T. Scali
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, FL
- Malcolm Randall Veterans Affairs Medical Center, Gainesville, FL
| |
Collapse
|
10
|
Galli F, Bonomini M, Bartolini D, Zatini L, Reboldi G, Marcantonini G, Gentile G, Sirolli V, Di Pietro N. Vitamin E (Alpha-Tocopherol) Metabolism and Nutrition in Chronic Kidney Disease. Antioxidants (Basel) 2022; 11:989. [PMID: 35624853 PMCID: PMC9137556 DOI: 10.3390/antiox11050989] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/07/2022] [Accepted: 05/10/2022] [Indexed: 01/27/2023] Open
Abstract
Vitamin E (alpha-tocopherol) is an essential micronutrient and fat-soluble antioxidant with proposed role in protecting tissues from uncontrolled lipid peroxidation. This vitamin has also important protein function and gene modulation effects. The metabolism of vitamin E depends on hepatic binding proteins that selectively retain food alpha-tocopherol for incorporation into nascent VLDL and tissue distribution together with esterified cholesterol and triglycerides. Chronic kidney disease (CKD) is a condition of oxidative stress and increased lipid peroxidation, that are associated with alterations of alpha-tocopherol metabolism and function. Specific changes have been reported for the levels of its enzymatic metabolites, including both short-chain and long-chain metabolites, the latter being endowed with regulatory functions on enzymatic and gene expression processes important for the metabolism of lipids and xenobiotics detoxification, as well as for the control of immune and inflammatory processes. Vitamin E therapy has been investigated in CKD using both oral vitamin E protocols and vitamin E-coated hemodialyzers, showing promising results in the secondary prevention of cardiovascular disease, as well as of immune and hematological complications. These therapeutic approaches are reviewed in the present article, together with a narrative excursus on the main findings indicating CKD as a condition of relative deficiency and impaired metabolism of vitamin E.
Collapse
Affiliation(s)
- Francesco Galli
- Department of Pharmaceutical Science, University of Perugia, 06126 Perugia, Italy; (D.B.); (L.Z.); (G.M.)
| | - Mario Bonomini
- Department of Medicine and Aging, G. d’Annunzio University Chieti-Pescara, 66100 Chieti, Italy; (M.B.); (V.S.)
| | - Desirée Bartolini
- Department of Pharmaceutical Science, University of Perugia, 06126 Perugia, Italy; (D.B.); (L.Z.); (G.M.)
| | - Linda Zatini
- Department of Pharmaceutical Science, University of Perugia, 06126 Perugia, Italy; (D.B.); (L.Z.); (G.M.)
| | - Gianpaolo Reboldi
- Department of Medicine and Surgery, Centro di Ricerca Clinica e Traslazionale, CERICLET, University of Perugia, 06126 Perugia, Italy;
| | - Giada Marcantonini
- Department of Pharmaceutical Science, University of Perugia, 06126 Perugia, Italy; (D.B.); (L.Z.); (G.M.)
| | - Giorgio Gentile
- Royal Cornwall Hospitals, NHS Trust, Cornwall, Truro TR1 3LJ, UK;
- Department of Nephrology, University of Exeter Medical School, Exeter EX1 2HZ, UK
| | - Vittorio Sirolli
- Department of Medicine and Aging, G. d’Annunzio University Chieti-Pescara, 66100 Chieti, Italy; (M.B.); (V.S.)
| | - Natalia Di Pietro
- Department of Medical, Oral and Biotechnological Sciences, Center for Advanced Studies and Technology-CAST, G. d’Annunzio University Chieti-Pescara, 66100 Chieti, Italy;
| |
Collapse
|
11
|
Fluorofenidone Inhibits UUO/IRI-Induced Renal Fibrosis by Reducing Mitochondrial Damage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2453617. [PMID: 35355864 PMCID: PMC8958071 DOI: 10.1155/2022/2453617] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 01/21/2022] [Accepted: 02/01/2022] [Indexed: 12/11/2022]
Abstract
Objective Mitochondrial damage contributes to extracellular matrix (ECM) deposition and renal fibrosis. In this study, we aimed (1) to investigate whether fluorofenidone (AKF-PD) can attenuate mitochondrial damage in two renal fibrosis models: unilateral ureteral obstruction (UUO) and renal ischemia-reperfusion injury (IRI), and (2) to explore the underlying mechanism. Method Mitochondrial damage and renal lesions were analyzed in the UUO and IRI models. Mitochondrial energy metabolism, mitochondrial biogenesis, and oxidative stress were measured to assess the effect of AKF-PD on mitochondrial damage and to explore the underlying mechanism. In addition, HK-2 cells were stimulated with TGF-β with and without AKF-PD. The mitochondrial morphology, mtROS, ATP contents, and redox-related proteins were then examined. Results In both UUO and IRI models, AKF-PD relieved renal fibrosis, maintained mitochondrial structure, and increased mitochondrial DNA copy numbers. The protection was associated with (1) sustaining mitochondrial energy metabolism, evident by elevations of tricarboxylic acid (TCA) cycle enzymes and mitochondrial respiratory chain complexes; (2) improving mitochondrial biogenesis with increases of TFAM, NRF1, PGC-1α, and SIRT1; and (3) reducing mitochondrial oxidative stress likely via regulating SOD2, SIRT3, and NOX4 expressions. In HK-2 cells treated with TGF-β, AKF-PD protected mitochondria along with improving mitochondrial morphology, enhancing ATP production, reducing mtROS, and regulating SOD2, SIRT3, and NOX4 expression. Conclusion We demonstrate that AKF-PD inhibited renal fibrosis at least in part via protecting mitochondria from damages developed in the UUO and IRI models. The mitochondrial protection was associated with sustaining mitochondrial energy metabolism, improving mitochondrial biogenesis, and reducing mitochondrial oxidative stress. This research verified the protective effect of AKF-PD on mitochondria in the UUO and IRI models and elaborated the underlying mechanism.
Collapse
|
12
|
Soto ME, Pérez-Torres I, Rubio-Ruiz ME, Manzano-Pech L, Guarner-Lans V. Interconnection between Cardiac Cachexia and Heart Failure—Protective Role of Cardiac Obesity. Cells 2022; 11:cells11061039. [PMID: 35326490 PMCID: PMC8946995 DOI: 10.3390/cells11061039] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/25/2022] [Accepted: 03/16/2022] [Indexed: 02/01/2023] Open
Abstract
Cachexia may be caused by congestive heart failure, and it is then called cardiac cachexia, which leads to increased morbidity and mortality. Cardiac cachexia also worsens skeletal muscle degradation. Cardiac cachexia is the loss of edema-free muscle mass with or without affecting fat tissue. It is mainly caused by a loss of balance between protein synthesis and degradation, or it may result from intestinal malabsorption. The loss of balance in protein synthesis and degradation may be the consequence of altered endocrine mediators such as insulin, insulin-like growth factor 1, leptin, ghrelin, melanocortin, growth hormone and neuropeptide Y. In contrast to many other health problems, fat accumulation in the heart is protective in this condition. Fat in the heart can be divided into epicardial, myocardial and cardiac steatosis. In this review, we describe and discuss these topics, pointing out the interconnection between heart failure and cardiac cachexia and the protective role of cardiac obesity. We also set the basis for possible screening methods that may allow for a timely diagnosis of cardiac cachexia, since there is still no cure for this condition. Several therapeutic procedures are discussed including exercise, nutritional proposals, myostatin antibodies, ghrelin, anabolic steroids, anti-inflammatory substances, beta-adrenergic agonists, medroxyprogesterone acetate, megestrol acetate, cannabinoids, statins, thalidomide, proteasome inhibitors and pentoxifylline. However, to this date, there is no cure for cachexia.
Collapse
Affiliation(s)
- María Elena Soto
- Department of Immunology, Instituto Nacional de Cardiología “Ignacio Chávez”, México City 14080, Mexico;
| | - Israel Pérez-Torres
- Department of Cardiovascular Biomedicine, Instituto Nacional de Cardiología “Ignacio Chávez”, México City 14080, Mexico; (I.P.-T.); (L.M.-P.)
| | - María Esther Rubio-Ruiz
- Department of Physiology, Instituto Nacional de Cardiología “Ignacio Chávez”, México City 14080, Mexico;
| | - Linaloe Manzano-Pech
- Department of Cardiovascular Biomedicine, Instituto Nacional de Cardiología “Ignacio Chávez”, México City 14080, Mexico; (I.P.-T.); (L.M.-P.)
| | - Verónica Guarner-Lans
- Department of Physiology, Instituto Nacional de Cardiología “Ignacio Chávez”, México City 14080, Mexico;
- Correspondence:
| |
Collapse
|
13
|
Li J, Xie Q, Liu L, Cheng Y, Han Y, Chen X, Lin J, Li Z, Liu H, Zhang X, Chen H, Peng J, Shen A. Swimming Attenuates Muscle Wasting and Mediates Multiple Signaling Pathways and Metabolites in CT-26 Bearing Mice. Front Mol Biosci 2022; 8:812681. [PMID: 35127824 PMCID: PMC8811507 DOI: 10.3389/fmolb.2021.812681] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/06/2021] [Indexed: 11/13/2022] Open
Abstract
Objectives: To investigate the effects of swimming on cancer induced muscle wasting and explore its underlying mechanism in CT-26 bearing mice.Methods: BALB/c mice (n = 16) injected with CT-26 cells were divided into two groups, including Tumor group (n = 8) and Swimming group (n = 8). Another 8 un-injected mice were set as Control group. Mice in Swimming group were subjected to physical training for swimming twice per day for 30 min intervals and 6 days per week for a total of 4 weeks. The tumor volume was monitored every 3 days and tumor weight was measured at the end of experiment. The changes of muscle function, pathological and cell apoptosis of quadriceps muscles were further assessed, and its underlying mechanisms were further explored using multiple biological technologies.Results: Swimming obviously alleviated tumor volume and weight in CT-26 bearing mice. Moreover, swimming attenuated the decrease of muscle tension, autonomic activities, and increase of muscle atrophy, pathological ultrastructure, as well as cell apoptosis of quadriceps muscles in CT-26 bearing mice. Furthermore, swimming significantly down-regulated the protein expression of NF-κB, p-NF-κB, TNF-α, IL-1β, IL-6 and Bax, while up-regulated the expression of Bcl-2. Further differential expressed metabolites (DEMs) analysis identified a total of 76 (in anion mode) and 330 (in cationic mode) DEMs in quadriceps muscles of CT-26 bearing mice after swimming, including taurochenodeoxycholic acid, taurocholic acid, ascorbic acid and eicosapentaenoic acid.Conclusion: Swimming attenuates tumor growth and muscle wasting, and by suppressing the activation of NF-κB signaling pathway mediated inflammation, reducing the level of Bax medicated cell apoptosis, as well as modulating multiple metabolites might be the importantly underlying mechanisms.
Collapse
Affiliation(s)
- Jiapeng Li
- The Department of Physical Education, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- School of Physical Education and Sport Science, Fujian Normal University, Fuzhou, China
| | - Qiurong Xie
- Academy of Integrative Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, China
| | - Liya Liu
- Academy of Integrative Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, China
| | - Ying Cheng
- Academy of Integrative Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, China
| | - Yuying Han
- Academy of Integrative Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, China
| | - Xiaoping Chen
- Academy of Integrative Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, China
| | - Jia Lin
- Rehabilitation Industry Institute, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Zuanfang Li
- Academy of Integrative Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, China
| | - Huixin Liu
- Academy of Integrative Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, China
| | - Xiuli Zhang
- Academy of Integrative Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, China
| | - Haichun Chen
- School of Physical Education and Sport Science, Fujian Normal University, Fuzhou, China
- Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou, China
| | - Jun Peng
- Academy of Integrative Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, China
- *Correspondence: Jun Peng, ; Aling Shen,
| | - Aling Shen
- Academy of Integrative Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, China
- *Correspondence: Jun Peng, ; Aling Shen,
| |
Collapse
|
14
|
Morrisson MJ, Bi F, Yang K, Cady SL, Hartwich TMP, Cerchia AP, Li Z, Kim J, Irwin ML, Yang-Hartwich Y. Effect of exercise on peritoneal microenvironment and progression of ovarian cancer. Am J Cancer Res 2021; 11:5045-5062. [PMID: 34765311 PMCID: PMC8569339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 06/12/2021] [Indexed: 06/13/2023] Open
Abstract
Ovarian cancer is one of the deadliest gynecological malignancies and lacks treatments that do not significantly impact patient health-related quality of life. Exercise has been associated with reduced cancer risk and improved clinical outcomes; however the underlying molecular mechanisms are unknown. In this study, we utilized a treadmill-running exercise model to investigate the effects of exercise on high-grade serous ovarian carcinoma (HGSOC) progression and chemotherapy outcomes. We found that treadmill-running suppressed peritoneal colonization of tumors in a syngeneic mouse ovarian cancer model. Acute exercise stimulated the production of CCL2 and IL-15 in the peritoneal microenvironment while downregulating CCL22, VEGF, and CCL12. Using a co-culture model, we demonstrated the role of CCL2 in mediating the activity of peritoneal cells to inhibit cancer cell viability. We showed that the activation of M1 macrophages may contribute to the exercise-induced changes in the peritoneal microenvironment. We identified that chronic exercise modulates gene expression of intraperitoneal fat tissues related to lipid formation, thermogenesis, browning, and inflammation, which can contribute to inhibiting the colonization of metastatic ovarian cancer. Treadmill running also lowered blood urea nitrogen levels and reduced incidence of neutropenia and thrombocytopenia during chemotherapy in a mouse model, suggesting the potential beneficial effects of exercise in improving chemotherapy outcomes. Our data provided new insights into the acute and chronic effects of physical activity on ovarian cancer at the molecular and in vivo levels.
Collapse
Affiliation(s)
- Madeline J Morrisson
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of MedicineNew Haven, CT 06510, USA
| | - Fangfang Bi
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of MedicineNew Haven, CT 06510, USA
- Sheng Jing Hospital of China Medical UniversityShenyang 110004, Liaoning, China
| | - Kevin Yang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of MedicineNew Haven, CT 06510, USA
| | - Sarah L Cady
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of MedicineNew Haven, CT 06510, USA
| | - Tobias MP Hartwich
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of MedicineNew Haven, CT 06510, USA
| | - Alexandra P Cerchia
- Department of Biology and Environmental Science, University of New HavenWest Haven, CT 06516, USA
| | - Zhigui Li
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of MedicineNew Haven, CT 06510, USA
| | - Jaeyeon Kim
- Department of Biochemistry and Molecular Biology, Indiana University School of MedicineIndianapolis, IN 46202, USA
- Melvin & Bren Simon Cancer Center, Indiana University School of MedicineIndianapolis, IN 46202, USA
| | - Melinda L Irwin
- Yale School of Public HealthNew Haven, CT 06510, USA
- Yale Cancer CenterNew Haven, CT 06510, USA
| | - Yang Yang-Hartwich
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of MedicineNew Haven, CT 06510, USA
- Yale Cancer CenterNew Haven, CT 06510, USA
| |
Collapse
|
15
|
Zhang T, Ding S, Wang R. Research Progress of Mitochondrial Mechanism in NLRP3 Inflammasome Activation and Exercise Regulation of NLRP3 Inflammasome. Int J Mol Sci 2021; 22:ijms221910866. [PMID: 34639204 PMCID: PMC8509472 DOI: 10.3390/ijms221910866] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 12/13/2022] Open
Abstract
NLRP3 is an important pattern recognition receptor in the innate immune system, and its activation induces a large number of pro-inflammatory cytokines, IL-1β and IL-18 which are involved in the development of various diseases. In recent years, it has been suggested that mitochondria are the platform for NLRP3 inflammasome activation. Additionally, exercise is considered as an important intervention strategy to mediate the innate immune responses. Generally, chronic moderate-intensity endurance training, resistance training and high-intensity interval training inhibit NLRP3 inflammasome activation in response to various pathological factors. In contrast, acute exercise activates NLRP3 inflammasome. However, the mechanisms by which exercise regulates NLRP3 inflammasome activation are largely unclear. Therefore, the mechanism of NLRP3 inflammasome activation is discussed mainly from the perspective of mitochondria in this review. Moreover, the effect and potential mechanism of exercise on NLRP3 inflammasome are explored, hoping to provide new target for relevant research.
Collapse
Affiliation(s)
- Tan Zhang
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China;
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai 200438, China
| | - Shuzhe Ding
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention, Ministry of Education, East China Normal University, Shanghai 200241, China
- Correspondence: (S.D.); (R.W.)
| | - Ru Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China;
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai 200438, China
- Correspondence: (S.D.); (R.W.)
| |
Collapse
|
16
|
Xu J, Zhou L, Liu Y. Cellular Senescence in Kidney Fibrosis: Pathologic Significance and Therapeutic Strategies. Front Pharmacol 2020; 11:601325. [PMID: 33362554 PMCID: PMC7759549 DOI: 10.3389/fphar.2020.601325] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/21/2020] [Indexed: 01/10/2023] Open
Abstract
Age-related disorders such as chronic kidney disease (CKD) are increasingly prevalent globally and pose unprecedented challenges. In many aspects, CKD can be viewed as a state of accelerated and premature aging. Aging kidney and CKD share many common characteristic features with increased cellular senescence, a conserved program characterized by an irreversible cell cycle arrest with altered transcriptome and secretome. While developmental senescence and acute senescence may positively contribute to the fine-tuning of embryogenesis and injury repair, chronic senescence, when unresolved promptly, plays a crucial role in kidney fibrogenesis and CKD progression. Senescent cells elicit their fibrogenic actions primarily by secreting an assortment of inflammatory and profibrotic factors known as the senescence-associated secretory phenotype (SASP). Increasing evidence indicates that senescent cells could be a promising new target for therapeutic intervention known as senotherapy, which includes depleting senescent cells, modulating SASP and restoration of senescence inhibitors. In this review, we discuss current understanding of the role and mechanism of cellular senescence in kidney fibrosis. We also highlight potential options of targeting senescent cells for the treatment of CKD.
Collapse
Affiliation(s)
- Jie Xu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lili Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Youhua Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
17
|
Thome T, Kumar RA, Burke SK, Khattri RB, Salyers ZR, Kelley RC, Coleman MD, Christou DD, Hepple RT, Scali ST, Ferreira LF, Ryan TE. Impaired muscle mitochondrial energetics is associated with uremic metabolite accumulation in chronic kidney disease. JCI Insight 2020; 6:139826. [PMID: 33290279 PMCID: PMC7821598 DOI: 10.1172/jci.insight.139826] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 11/25/2020] [Indexed: 01/01/2023] Open
Abstract
Chronic kidney disease (CKD) causes progressive skeletal myopathy involving atrophy, weakness, and fatigue. Mitochondria have been thought to contribute to skeletal myopathy; however, the molecular mechanisms underlying muscle metabolism changes in CKD are unknown. We employed a comprehensive mitochondrial phenotyping platform to elucidate the mechanisms of skeletal muscle mitochondrial impairment in mice with adenine-induced CKD. CKD mice displayed significant reductions in mitochondrial oxidative phosphorylation (OXPHOS), which was strongly correlated with glomerular filtration rate, suggesting a link between kidney function and muscle mitochondrial health. Biochemical assays uncovered that OXPHOS dysfunction was driven by reduced activity of matrix dehydrogenases. Untargeted metabolomics analyses in skeletal muscle revealed a distinct metabolite profile in CKD muscle including accumulation of uremic toxins that strongly associated with the degree of mitochondrial impairment. Additional muscle phenotyping found CKD mice experienced muscle atrophy and increased muscle protein degradation, but only male CKD mice had lower maximal contractile force. CKD mice had morphological changes indicative of destabilization in the neuromuscular junction. This study provides the first comprehensive evaluation of mitochondrial health in murine CKD muscle to our knowledge and uncovers several unknown uremic metabolites that strongly associate with the degree of mitochondrial impairment.
Collapse
Affiliation(s)
- Trace Thome
- Department of Applied Physiology and Kinesiology, College of Health & Human Performance
| | - Ravi A Kumar
- Department of Applied Physiology and Kinesiology, College of Health & Human Performance
| | - Sarah K Burke
- Department of Physical Therapy, College of Public Health and Health Professions
| | - Ram B Khattri
- Department of Applied Physiology and Kinesiology, College of Health & Human Performance
| | - Zachary R Salyers
- Department of Applied Physiology and Kinesiology, College of Health & Human Performance
| | - Rachel C Kelley
- Department of Applied Physiology and Kinesiology, College of Health & Human Performance
| | - Madeline D Coleman
- Department of Applied Physiology and Kinesiology, College of Health & Human Performance
| | - Demetra D Christou
- Department of Applied Physiology and Kinesiology, College of Health & Human Performance.,Center for Exercise Science, College of Health & Human Performance, and
| | - Russell T Hepple
- Department of Physical Therapy, College of Public Health and Health Professions
| | - Salvatore T Scali
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, College of Medicine, University of Florida, Gainesville, Florida, USA.,Malcom Randall Veterans Affairs Medical Center, Gainesville, Florida, USA
| | - Leonardo F Ferreira
- Department of Applied Physiology and Kinesiology, College of Health & Human Performance.,Center for Exercise Science, College of Health & Human Performance, and
| | - Terence E Ryan
- Department of Applied Physiology and Kinesiology, College of Health & Human Performance.,Center for Exercise Science, College of Health & Human Performance, and
| |
Collapse
|