1
|
Song G, Park J, Jung Y, Park WY, Park JY, Jung SJ, Kim B, Choi M, Kim SH, Choe SK, Kwak HJ, Lee J, Lee KY, Ahn KS, Um JY. Regulating Sirtuin 3-mediated mitochondrial dynamics through vanillic acid improves muscle atrophy in cancer-induced cachexia. Commun Biol 2025; 8:585. [PMID: 40204937 PMCID: PMC11982244 DOI: 10.1038/s42003-025-07770-0] [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: 02/06/2024] [Accepted: 02/18/2025] [Indexed: 04/11/2025] Open
Abstract
Cancer cachexia is a cancer-associated disease characterized by gradual body weight loss due to pathologic muscle and fat loss, but effective treatments are still lacking. Here, we investigate the possible effect of vanillic acid (VA), known for its antioxidant, anti-inflammatory, and anti-obesity effects, on mitochondria-mediated improvement of cancer cachexia. We utilized cachexia-like models using CT26 colon cancer and dexamethasone. VA improved representative parameters of cancer cachexia including body weight loss and increased serum intereukin-6 levels. VA also attenuated muscle loss in the tibialis anterior and gastrocnemius muscles, inhibited proteolytic markers including muscle RING-finger protein-1 (MURF1) and muscle atrophy F-box (MAFbx) and improved mitochondrial function through alteration of sirtuins 3 (SIRT3) and mitofusin 1 (MFN1). Importantly, silencing the SIRT3 gene abolished the effect of VA, indicating that SIRT3 is important in the mechanism of action of VA. Overall, we suggest using VA as a novel therapeutic agent that can fundamentally treat and recover muscle atrophy in cancer cachexia patients.
Collapse
Affiliation(s)
- Gahee Song
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
- Kyung Hee Institute of Convergence Korean Medicine, Kyung Hee University, 02447, Seoul, Korea
| | - Jinbong Park
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
- Kyung Hee Institute of Convergence Korean Medicine, Kyung Hee University, 02447, Seoul, Korea
| | - Yunu Jung
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Woo Yong Park
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Ja Yeon Park
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Se Jin Jung
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Beomsu Kim
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Minji Choi
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Sang Hee Kim
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Seong-Kyu Choe
- Department of Microbiology, Wonkwang University School of Medicine, Iksan, 54538, Republic of Korea
| | - Hyun Jeong Kwak
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul, 02707, Republic of Korea
| | - Junhee Lee
- Department of Sasang Constitutional Medicine, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Kil Yeon Lee
- Department of Surgery, College of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Jae-Young Um
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea.
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea.
- Kyung Hee Institute of Convergence Korean Medicine, Kyung Hee University, 02447, Seoul, Korea.
| |
Collapse
|
2
|
Libramento ZP, Tichy L, Parry TL. Muscle wasting in cancer cachexia: Mechanisms and the role of exercise. Exp Physiol 2025. [PMID: 40159295 DOI: 10.1113/ep092544] [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: 01/02/2025] [Accepted: 03/13/2025] [Indexed: 04/02/2025]
Abstract
Cancer cachexia (CC) is a multifactorial disease marked by a severe and progressive loss of lean muscle mass and characterized further by inflammation and a negative energy/protein balance, ultimately leading to muscle atrophy and loss of muscle tissue. As a result, patients experiencing cachexia have reduced muscle function and thus less independence and a lower quality of life. CC progresses through stages of increasing severity: pre-cachexia, cachexia and refractory cachexia. Two proposed underlying mechanisms that drive cancer-induced muscle wasting are the autophagy-lysosome and ubiquitin-proteasome systems. An increase in autophagic flux and proteolytic activity leads to atrophy of both cardiac and skeletal muscle, ultimately mediated by tumour or immune-secreted inflammatory cytokines. These pathways occur at a basal level to maintain cellular homeostasis; therefore, it is the overactivation of the pathways that leads to muscle atrophy. Recent evidence demonstrates the ability of aerobic and resistance training to restore these pathways to their basal levels. The mechanism is not yet understood, and more research is needed to determine exactly how exercise influences each pathway. However, exercise has great promise as a therapeutic strategy for CC because of the evidence for it preserving muscle mass and function, and attenuating protein degradative pathways. The extent to which exercise affects the ubiquitin-proteasome and autophagy-lysosome systems is determined by the frequency, intensity and duration of the exercise protocol. As such, an ideal exercise prescription is lacking for individuals with CC.
Collapse
Affiliation(s)
- Zoe P Libramento
- Department of Kinesiology, University of North Carolina Greensboro, Greensboro, North Carolina, USA
| | - Louisa Tichy
- Department of Kinesiology, University of North Carolina Greensboro, Greensboro, North Carolina, USA
| | - Traci L Parry
- Department of Kinesiology, University of North Carolina Greensboro, Greensboro, North Carolina, USA
| |
Collapse
|
3
|
Field JT, Chapman D, Hai Y, Ghavami S, West AR, Ozerklig B, Saleem A, Kline J, Mendelson AA, Kindrachuk J, Triggs-Raine B, Gordon JW. The mitophagy receptor BNIP3L/Nix coordinates nuclear calcium signaling to modulate the muscle phenotype. Autophagy 2025:1-12. [PMID: 40063005 DOI: 10.1080/15548627.2025.2476872] [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: 02/12/2024] [Revised: 02/22/2025] [Accepted: 03/04/2025] [Indexed: 03/25/2025] Open
Abstract
Mitochondrial quality control is critical in muscle to ensure contractile and metabolic function. BNIP3L/Nix is a BCL2 member, a mitophagy receptor, and has been implicated in muscle atrophy. Human genome-wide association studies (GWAS) suggest altered BNIP3L expression could predispose to mitochondrial disease. To investigate BNIP3L function, we generated a muscle-specific knockout model. bnip3l knockout mice displayed a ragged-red fiber phenotype, along with accumulation of mitochondria and endo/sarcoplasmic reticulum with altered morphology. Intriguingly, bnip3l knockout mice were more insulin sensitive with a corresponding increase in glycogen-rich muscle fibers. Kinome and gene expression analyses revealed that bnip3l knockout impairs NFAT and MSTN (myostatin) signaling, with alterations in muscle fiber-type and evidence of regeneration. Mechanistic experiments demonstrated that BNIP3L modulates mitophagy, along with reticulophagy leading to altered nuclear calcium signaling. Collectively, these observations identify novel roles for BNIP3L coordinating selective autophagy, oxidative gene expression, and signaling pathways that maintain the muscle phenotype.
Collapse
Affiliation(s)
- Jared T Field
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, Canada
| | - Donald Chapman
- Children's Hospital Research Institute of Manitoba, Winnipeg, Canada
| | - Yan Hai
- Children's Hospital Research Institute of Manitoba, Winnipeg, Canada
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, Canada
| | - Adrian R West
- Children's Hospital Research Institute of Manitoba, Winnipeg, Canada
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Berkay Ozerklig
- Children's Hospital Research Institute of Manitoba, Winnipeg, Canada
- Faculty of Kinesiology and Recreation Management, University of Manitoba, Winnipeg, Canada
| | - Ayesha Saleem
- Children's Hospital Research Institute of Manitoba, Winnipeg, Canada
- Faculty of Kinesiology and Recreation Management, University of Manitoba, Winnipeg, Canada
| | - Julia Kline
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Asher A Mendelson
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
- Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Jason Kindrachuk
- Children's Hospital Research Institute of Manitoba, Winnipeg, Canada
- Department of Medical Microbiology & Infectious Diseases, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Barbara Triggs-Raine
- Children's Hospital Research Institute of Manitoba, Winnipeg, Canada
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Joseph W Gordon
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, Canada
- Department of Pediatrics and Child Health, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
4
|
Santos MKR, Seghetto R, Hauptli L, Paiano D, da Silva AS, Benetti Filho V, Wagner G, de Oliveira Moraes P. Blended phytogenics as an alternative to growth-promoting antibiotics in newly weaned piglets. Trop Anim Health Prod 2024; 57:5. [PMID: 39710793 DOI: 10.1007/s11250-024-04225-2] [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: 06/10/2024] [Accepted: 10/29/2024] [Indexed: 12/24/2024]
Abstract
The research aimed to evaluate the effects of a commercial blend of phytogenic compounds on the digestibility, antioxidant system, intestinal microbiota, and performance of weaned piglets. Two experiments compared three treatments (diets): control, zinc bacitracin (300 g/t) and blended phytogenic compounds (400 g/t). The first experiment analised of digestibility of the dry matter, organic matter, crude protein, crude energy and metabolizable energy, in addition to blood parameters and gut microbiota in 15 piglets commercial cross-bred, weaned at 28 days of age, castrated males, weighing 9.40 ± 0.622 kg housed in metabolic cages. In the second experiment, performance was evaluated on 108 piglets commercial cross-bred, weaned at 26 days of age, females and castrated males, weighing 7.52 ± 0.356 kg housed in collective stalls with 1,5 m² (3 animals/stall). A completely randomized design was used. The data were subjected to analysis of variance, and the means compared by the Tukey test at 5% significance. There were no differences in piglet digestibility and performance. There was a reduction in the levels of the enzyme superoxide dismutase, lipid peroxidation, and haptoglobulin, and an increase in the levels of the non-protein thiol compound and IgA for the animals receiving the phytogenic compound when compared with the piglets of the other treatments (p < 0.05). A tendency in diversity was observed in the intestinal microbiota of piglets receiving the phytogenic compound in the feed (p = 0.054). Due to its important role in the antioxidant system and intestinal microbiota, it is suggested that the blend of phytogenic additives can replace antibiotics growth promoters in the diet of newly weaned piglets.
Collapse
Affiliation(s)
- Manoela Karolina Ribeiro Santos
- Centro de Ciências Agrárias-CCA, Universidade Federal de Santa Catarina, Rod. Admar Gonzaga, 1346 - Itacorubi, Florianopolis, SC, 88034-000, Brazil
| | - Ronaldo Seghetto
- Centro de Ciências Agrárias-CCA, Universidade Federal de Santa Catarina, Rod. Admar Gonzaga, 1346 - Itacorubi, Florianopolis, SC, 88034-000, Brazil
| | - Lucélia Hauptli
- Centro de Ciências Agrárias-CCA, Universidade Federal de Santa Catarina, Rod. Admar Gonzaga, 1346 - Itacorubi, Florianopolis, SC, 88034-000, Brazil
| | - Diovani Paiano
- Universidade do Estado de Santa Catarina, Beloni Trombeta Zanin 680E - Santo Antônio, Chapecó, 89815-630, SC, Brazil
| | - Aleksandro Schafer da Silva
- Universidade do Estado de Santa Catarina, Beloni Trombeta Zanin 680E - Santo Antônio, Chapecó, 89815-630, SC, Brazil
| | - Vilmar Benetti Filho
- Centro de Ciências Agrárias-CCA, Universidade Federal de Santa Catarina, Rod. Admar Gonzaga, 1346 - Itacorubi, Florianopolis, SC, 88034-000, Brazil
| | - Glauber Wagner
- Centro de Ciências Agrárias-CCA, Universidade Federal de Santa Catarina, Rod. Admar Gonzaga, 1346 - Itacorubi, Florianopolis, SC, 88034-000, Brazil
| | - Priscila de Oliveira Moraes
- Centro de Ciências Agrárias-CCA, Universidade Federal de Santa Catarina, Rod. Admar Gonzaga, 1346 - Itacorubi, Florianopolis, SC, 88034-000, Brazil.
| |
Collapse
|
5
|
Deng K, Pei M, Li B, Yang N, Wang Z, Wan X, Zhong Z, Yang Z, Chen Y. Signal pathways involved in contrast-induced acute kidney injury. Front Physiol 2024; 15:1490725. [PMID: 39655278 PMCID: PMC11625813 DOI: 10.3389/fphys.2024.1490725] [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: 09/03/2024] [Accepted: 11/12/2024] [Indexed: 12/12/2024] Open
Abstract
Contrast-induced acute kidney injury (CI-AKI) has emerged as a global public health concern, ranking as the third most prevalent cause of hospital-acquired acute kidney injury, which is related to adverse outcomes. However, its precise pathogenesis remains elusive. Consequently, researchers are dedicated to uncovering CI-AKI's pathophysiology and signaling pathways, including inflammation, oxidative stress, apoptosis, and ferroptosis, to improve prevention and treatment. This review thoroughly analyzes the signaling pathways and their interactions associated with CI-AKI, assesses the impact of various research models on pathway analysis, and explores more precise targeted treatment and prevention approaches. Aims to furnish a robust theoretical foundation for the molecular mechanisms underpinning clinical treatments.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yanling Chen
- Department of Pathophysiology, Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong, China
| |
Collapse
|
6
|
Gicquel T, Marchiano F, Reyes-Castellanos G, Audebert S, Camoin L, Habermann BH, Giannesini B, Carrier A. Integrative study of skeletal muscle mitochondrial dysfunction in a murine pancreatic cancer-induced cachexia model. eLife 2024; 13:RP93312. [PMID: 39422661 PMCID: PMC11488855 DOI: 10.7554/elife.93312] [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] [Indexed: 10/19/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), the most common pancreatic cancer, is a deadly cancer, often diagnosed late and resistant to current therapies. PDAC patients are frequently affected by cachexia characterized by muscle mass and strength loss (sarcopenia) contributing to patient frailty and poor therapeutic response. This study assesses the mechanisms underlying mitochondrial remodeling in the cachectic skeletal muscle, through an integrative exploration combining functional, morphological, and omics-based evaluation of gastrocnemius muscle from KIC genetically engineered mice developing autochthonous pancreatic tumor and cachexia. Cachectic PDAC KIC mice exhibit severe sarcopenia with loss of muscle mass and strength associated with reduced muscle fiber's size and induction of protein degradation processes. Mitochondria in PDAC atrophied muscles show reduced respiratory capacities and structural alterations, associated with deregulation of oxidative phosphorylation and mitochondrial dynamics pathways. Beyond the metabolic pathways known to be altered in sarcopenic muscle (carbohydrates, proteins, and redox), lipid and nucleic acid metabolisms are also affected. Although the number of mitochondria per cell is not altered, mitochondrial mass shows a twofold decrease and the mitochondrial DNA threefold, suggesting a defect in mitochondrial genome homeostasis. In conclusion, this work provides a framework to guide toward the most relevant targets in the clinic to limit PDAC-induced cachexia.
Collapse
Affiliation(s)
- Tristan Gicquel
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCMMarseilleFrance
- Nutrition And Cancer Research Network (NACRe Network)Jouy-en-JosasFrance
| | | | - Gabriela Reyes-Castellanos
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCMMarseilleFrance
- Nutrition And Cancer Research Network (NACRe Network)Jouy-en-JosasFrance
| | - Stephane Audebert
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCMMarseilleFrance
| | - Luc Camoin
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCMMarseilleFrance
| | | | | | - Alice Carrier
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCMMarseilleFrance
- Nutrition And Cancer Research Network (NACRe Network)Jouy-en-JosasFrance
| |
Collapse
|
7
|
Endo K, Ichinose M, Kobayashi E, Ueno T, Hirai N, Nakanishi Y, Kondo S, Yoshizaki T. Head and Neck Cancer and Sarcopenia: An Integrative Clinical and Functional Review. Cancers (Basel) 2024; 16:3460. [PMID: 39456555 PMCID: PMC11506384 DOI: 10.3390/cancers16203460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 09/26/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Sarcopenia is recognized as a crucial factor impacting the prognosis, treatment responses, and quality of life of HNC patients. This review discusses various mechanisms, including common etiological factors, such as aging, chronic inflammation, and metabolic dysregulation. Cancer-related factors, including tumor locations and treatment modalities, contribute to the development of sarcopenia. The clinical implications of sarcopenia in HNC patients extend beyond reduced muscle strength; it affects overall mobility, reduces quality of life, and increases the risk of falls and fractures. Sarcopenia serves as an independent predictor of postoperative complications, chemotherapy dose-limiting toxicity, and treatment outcomes, which affect therapy planning and perioperative management decisions. Methods to assess sarcopenia in HNC patients encompass various techniques. A sarcopenia assessment offers a potentially efficient and readily available tool for clinical practice. Interventions and management strategies for sarcopenia involve exercise interventions as a cornerstone; however, challenges arise due to patient-specific limitations during cancer treatment. A routine body composition analysis is proposed as a valuable addition to HNC patient management, with ongoing research required to refine preoperative exercise and nutrition programs for improved treatment outcomes and survival.
Collapse
Affiliation(s)
- Kazuhira Endo
- Division of Otolaryngology, Head & Neck Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8640, Japan; (M.I.); (E.K.); (T.U.); (N.H.); (Y.N.); (S.K.); (T.Y.)
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Wang Y, Zheng B, Zhang L, Zhang T, Zhao D, Sun Y, Xiao S, Zhang Y, Gong L, Wang W, Lu Q. Impact of sarcopenia and obesity on overall survival in patients with head and neck cancer receiving radiotherapy: A longitudinal study. Eur J Oncol Nurs 2024; 72:102679. [PMID: 39178752 DOI: 10.1016/j.ejon.2024.102679] [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: 01/02/2024] [Revised: 06/24/2024] [Accepted: 07/26/2024] [Indexed: 08/26/2024]
Abstract
PURPOSE To analyze the impact of sarcopenia and obesity on overall survival (OS) in patients with head and neck cancer (HNC) receiving radiotherapy (RT). METHODS This prospective longitudinal study recruited 494 patients using convenient sampling. Weight and body composition were assessed before RT (T1), and at the end of RT (T2) using bioelectrical impedance analysis (BIA). The appendicular skeletal mass index was used to define sarcopenia, while the body mass index and fat mass index were used to define obesity. Patient OS was followed and described using Kplan-Meier analysis. Cox proportional hazard regression was used to analyze influencing factors of OS. RESULTS The median follow-up time was 26.2 months (IQR: 18.4-34.4 months). Multivariable models indicated that sarcopenia/obesity type assessed at T1 was not significantly associated with OS. Multivariable models involving body composition at T2 showed that age (P < 0.001), tumor site (P = 0.003), tumor stage (P = 0.024), and sarcopenia/obesity type (P = 0.040) were significantly associated with OS, while sarcopenic patients without obesity at T2 had worse OS. CONCLUSIONS Patients with sarcopenia and no obesity at the end of RT might have worse OS. Healthcare professionals should enhance HNC patients' management during RT, helping them maintain a certain amount of muscle mass and fat mass to improve their survival.
Collapse
Affiliation(s)
- Yujie Wang
- Department of Nursing, Henan Provincial People's Hospital, #7 Weiwu Road, Jinshui District, Zhengzhou, Henan, 450003, China; Division of Medical & Surgical Nursing, School of Nursing, Peking University, #38 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Baomin Zheng
- Department of Radiation Oncology, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, #52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Lichuan Zhang
- Division of Medical & Surgical Nursing, School of Nursing, Peking University, #38 Xueyuan Road, Haidian District, Beijing, 100191, China; School of Nursing, Hebei University, #342 Yuhua East Road, LianChi District, Baoding, Hebei, 071000, China
| | - Tong Zhang
- Department of Nursing, Beijing Friendship Hospital, Capital Medical University, #95 Yongan Road, Xicheng District, Beijing, 100050, China
| | - Dan Zhao
- Department of Radiation Oncology, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, #52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Yan Sun
- Department of Radiation Oncology, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, #52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Shaowen Xiao
- Department of Radiation Oncology, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, #52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Yaru Zhang
- Department of Radiation Oncology, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, #52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Liqing Gong
- Department of Nutrition, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, #52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Weihu Wang
- Department of Radiation Oncology, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, #52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Qian Lu
- Division of Medical & Surgical Nursing, School of Nursing, Peking University, #38 Xueyuan Road, Haidian District, Beijing, 100191, China.
| |
Collapse
|
9
|
VanderVeen BN, Cardaci TD, Bullard BM, Madden M, Li J, Velazquez KT, Kubinak JL, Fan D, Murphy EA. Involvement of the gut microbiota in cancer cachexia. Am J Physiol Cell Physiol 2024; 327:C661-C670. [PMID: 38981609 PMCID: PMC11427007 DOI: 10.1152/ajpcell.00327.2024] [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: 05/16/2024] [Revised: 06/20/2024] [Accepted: 07/02/2024] [Indexed: 07/11/2024]
Abstract
Cancer cachexia, or the unintentional loss of body weight in patients with cancer, is a multiorgan and multifactorial syndrome with a complex and largely unknown etiology; however, metabolic dysfunction and inflammation remain hallmarks of cancer-associated wasting. Although cachexia manifests with muscle and adipose tissue loss, perturbations to the gastrointestinal tract may serve as the frontline for both impaired nutrient absorption and immune-activating gut dysbiosis. Investigations into the gut microbiota have exploded within the past two decades, demonstrating multiple gut-tissue axes; however, the link between adipose and skeletal muscle wasting and the gut microbiota with cancer is only beginning to be understood. Furthermore, the most used anticancer drugs (e.g. chemotherapy and immune checkpoint inhibitors) negatively impact gut homeostasis, potentially exacerbating wasting and contributing to poor patient outcomes and survival. In this review, we 1) highlight our current understanding of the microbial changes that occur with cachexia, 2) discuss how microbial changes may contribute to adipose and skeletal muscle wasting, and 3) outline study design considerations needed when examining the role of the microbiota in cancer-induced cachexia.
Collapse
Affiliation(s)
- Brandon N VanderVeen
- Department of Pathology, Microbiology and Immunology, School of Medicine Columbia, University of South Carolina, Columbia, South Carolina, United States
| | - Thomas D Cardaci
- Department of Pathology, Microbiology and Immunology, School of Medicine Columbia, University of South Carolina, Columbia, South Carolina, United States
| | - Brooke M Bullard
- Department of Pathology, Microbiology and Immunology, School of Medicine Columbia, University of South Carolina, Columbia, South Carolina, United States
| | - Michael Madden
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina, United States
| | - Jie Li
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina, United States
| | - Kandy T Velazquez
- Department of Pathology, Microbiology and Immunology, School of Medicine Columbia, University of South Carolina, Columbia, South Carolina, United States
| | - Jason L Kubinak
- Department of Pathology, Microbiology and Immunology, School of Medicine Columbia, University of South Carolina, Columbia, South Carolina, United States
| | - Daping Fan
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, United States
| | - E Angela Murphy
- Department of Pathology, Microbiology and Immunology, School of Medicine Columbia, University of South Carolina, Columbia, South Carolina, United States
| |
Collapse
|
10
|
Compton SLE, Heymsfield SB, Brown JC. Nutritional Mechanisms of Cancer Cachexia. Annu Rev Nutr 2024; 44:77-98. [PMID: 39207878 DOI: 10.1146/annurev-nutr-062122-015646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Cancer cachexia is a complex systemic wasting syndrome. Nutritional mechanisms that span energy intake, nutrient metabolism, body composition, and energy balance may be impacted by, and may contribute to, the development of cachexia. To date, clinical management of cachexia remains elusive. Leaning on discoveries and novel methodologies from other fields of research may bolster new breakthroughs that improve nutritional management and clinical outcomes. Characteristics that compare and contrast cachexia and obesity may reveal opportunities for cachexia research to adopt methodology from the well-established field of obesity research. This review outlines the known nutritional mechanisms and gaps in the knowledge surrounding cancer cachexia. In parallel, we present how obesity may be a different side of the same coin and how obesity research has tackled similar research questions. We present insights into how cachexia research may utilize nutritional methodology to expand our understanding of cachexia to improve definitions and clinical care in future directions for the field.
Collapse
Affiliation(s)
- Stephanie L E Compton
- Cancer Energetics Unit, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA;
| | - Steven B Heymsfield
- Metabolism and Body Composition Unit, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Justin C Brown
- Cancer Energetics Unit, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA;
| |
Collapse
|
11
|
Gao Y, Kim K, Vitrac H, Salazar RL, Gould BD, Soedkamp D, Spivia W, Raedschelders K, Dinh AQ, Guzman AG, Tan L, Azinas S, Taylor DJR, Schiffer W, McNavish D, Burks HB, Gottlieb RA, Lorenzi PL, Hanson BM, Van Eyk JE, Taegtmeyer H, Karlstaedt A. Autophagic signaling promotes systems-wide remodeling in skeletal muscle upon oncometabolic stress by D2-HG. Mol Metab 2024; 86:101969. [PMID: 38908793 PMCID: PMC11278897 DOI: 10.1016/j.molmet.2024.101969] [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: 05/20/2024] [Accepted: 06/11/2024] [Indexed: 06/24/2024] Open
Abstract
OBJECTIVES Cachexia is a metabolic disorder and comorbidity with cancer and heart failure. The syndrome impacts more than thirty million people worldwide, accounting for 20% of all cancer deaths. In acute myeloid leukemia, somatic mutations of the metabolic enzyme isocitrate dehydrogenase 1 and 2 cause the production of the oncometabolite D2-hydroxyglutarate (D2-HG). Increased production of D2-HG is associated with heart and skeletal muscle atrophy, but the mechanistic links between metabolic and proteomic remodeling remain poorly understood. Therefore, we assessed how oncometabolic stress by D2-HG activates autophagy and drives skeletal muscle loss. METHODS We quantified genomic, metabolomic, and proteomic changes in cultured skeletal muscle cells and mouse models of IDH-mutant leukemia using RNA sequencing, mass spectrometry, and computational modeling. RESULTS D2-HG impairs NADH redox homeostasis in myotubes. Increased NAD+ levels drive activation of nuclear deacetylase Sirt1, which causes deacetylation and activation of LC3, a key regulator of autophagy. Using LC3 mutants, we confirm that deacetylation of LC3 by Sirt1 shifts its distribution from the nucleus into the cytosol, where it can undergo lipidation at pre-autophagic membranes. Sirt1 silencing or p300 overexpression attenuated autophagy activation in myotubes. In vivo, we identified increased muscle atrophy and reduced grip strength in response to D2-HG in male vs. female mice. In male mice, glycolytic intermediates accumulated, and protein expression of oxidative phosphorylation machinery was reduced. In contrast, female animals upregulated the same proteins, attenuating the phenotype in vivo. Network modeling and machine learning algorithms allowed us to identify candidate proteins essential for regulating oncometabolic adaptation in mouse skeletal muscle. CONCLUSIONS Our multi-omics approach exposes new metabolic vulnerabilities in response to D2-HG in skeletal muscle and provides a conceptual framework for identifying therapeutic targets in cachexia.
Collapse
Affiliation(s)
- Yaqi Gao
- Department of Cardiology, Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Kyoungmin Kim
- Department of Cardiology, Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Heidi Vitrac
- Department of Biochemistry, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Bruker Daltonics, Billerica, MA, USA
| | - Rebecca L Salazar
- Department of Internal Medicine, Division of Cardiology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Benjamin D Gould
- Department of Internal Medicine, Division of Cardiology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Daniel Soedkamp
- Department of Cardiology, Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Weston Spivia
- Department of Cardiology, Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Koen Raedschelders
- Department of Cardiology, Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - An Q Dinh
- Center for Infectious Diseases, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Anna G Guzman
- Center for Stem Cell and Regeneration, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Lin Tan
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Stavros Azinas
- Department of Biochemistry, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Cell and Molecular Biology, Uppsala University, Sweden
| | - David J R Taylor
- Department of Cardiology, Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Walter Schiffer
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27101, USA
| | - Daniel McNavish
- Department of Internal Medicine, Division of Cardiology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Helen B Burks
- Department of Internal Medicine, Division of Cardiology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Roberta A Gottlieb
- Department of Cardiology, Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Philip L Lorenzi
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Blake M Hanson
- Center for Infectious Diseases, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jennifer E Van Eyk
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Heinrich Taegtmeyer
- Department of Biochemistry, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Anja Karlstaedt
- Department of Cardiology, Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
12
|
Homma ST, Wang X, Frere JJ, Gower AC, Zhou J, Lim JK, tenOever BR, Zhou L. Respiratory SARS-CoV-2 Infection Causes Skeletal Muscle Atrophy and Long-Lasting Energy Metabolism Suppression. Biomedicines 2024; 12:1443. [PMID: 39062017 PMCID: PMC11275164 DOI: 10.3390/biomedicines12071443] [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: 05/18/2024] [Revised: 06/19/2024] [Accepted: 06/22/2024] [Indexed: 07/28/2024] Open
Abstract
Muscle fatigue represents the most prevalent symptom of long-term COVID, with elusive pathogenic mechanisms. We performed a longitudinal study to characterize histopathological and transcriptional changes in skeletal muscle in a hamster model of respiratory SARS-CoV-2 infection and compared them with influenza A virus (IAV) and mock infections. Histopathological and bulk RNA sequencing analyses of leg muscles derived from infected animals at days 3, 30, and 60 post-infection showed no direct viral invasion but myofiber atrophy in the SARS-CoV-2 group, which was accompanied by persistent downregulation of the genes related to myofibers, ribosomal proteins, fatty acid β-oxidation, tricarboxylic acid cycle, and mitochondrial oxidative phosphorylation complexes. While both SARS-CoV-2 and IAV infections induced acute and transient type I and II interferon responses in muscle, only the SARS-CoV-2 infection upregulated TNF-α/NF-κB but not IL-6 signaling in muscle. Treatment of C2C12 myotubes, a skeletal muscle cell line, with combined IFN-γ and TNF-α but not with IFN-γ or TNF-α alone markedly impaired mitochondrial function. We conclude that a respiratory SARS-CoV-2 infection can cause myofiber atrophy and persistent energy metabolism suppression without direct viral invasion. The effects may be induced by the combined systemic interferon and TNF-α responses at the acute phase and may contribute to post-COVID-19 persistent muscle fatigue.
Collapse
Affiliation(s)
- Sachiko T. Homma
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Xingyu Wang
- Department of Neurology, Hospital for Special Surgery, New York, NY 10021, USA
| | - Justin J. Frere
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Adam C. Gower
- Clinical and Translational Science Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Jingsong Zhou
- College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX 76010, USA
| | - Jean K. Lim
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Benjamin R. tenOever
- Department of Microbiology, Grossman School of Medicine, New York University, New York, NY 10016, USA
| | - Lan Zhou
- Department of Neurology, Hospital for Special Surgery, New York, NY 10021, USA
| |
Collapse
|
13
|
Wu Y, Yao X, Shi X, Xu Z, Ren J, Shi M, Li M, Liu J, Du X. Myeloma extracellular vesicle-derived RAGE increases inflammatory responses and myotube atrophy in multiple myeloma through activation of the TLR4/NF-κB p65 pathway. Apoptosis 2024; 29:849-864. [PMID: 38117373 DOI: 10.1007/s10495-023-01920-7] [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] [Accepted: 11/06/2023] [Indexed: 12/21/2023]
Abstract
Sarcopenia manifests as muscle atrophy and loss that is complicated with malignancy. This study explored the mechanism of extracellular vesicles (EVs) in multiple myeloma (MM) with sarcopenia. SP2/0 conditioned medium (CM) was collected to isolate SP2/0-EVs. C2C12 cells were incubated with SP2/0 CM or SP2/0-EVs. ROS, TNF-α, IL-6, MuRF1 and MyHC levels were detected by DCF-DA fluorescent probe, ELISA, and Western blot. GW4869 was used to inhibit EV secretion in SP2/0 to confirm its effect on muscle atrophy. Serum was collected from MM patients with or without sarcopenia to detect RAGE mRNA expression. SP2/0 cells were transfected with RAGE siRNA and C2C12 cells were treated with the isolated si-RAGE-EVs or/and TLR4 agonist. SP2/0 tumor-bearing mouse model was established. Healthy mice and SP2/0-tumor bearing mice were treated with SP2/0-EVs or si-RAGE-EVs. SP2/0 CM or SP2/0-EVs stimulated ROS, inflammatory responses, and myotube atrophy in C2C12 cells. GW4869 blocked EV secretion and the effects of SP2/0 CM. RAGE mRNA expression in serum EVs was increased in MM&Sarcopenia patients and RAGE knockdown in SP2/0-EVs partially nullified SP2/0-EVs' effects. SP2/0-EVs activated the TLR4/NF-κB p65 pathway by translocating RAGE. SP2/0-EVs-derived RAGE elevated ROS production, inflammation, and myotube atrophy in C2C12 cells and caused muscle loss in SP2/0 tumor-bearing mice by activating the TLR4/NF-κB p65 pathway. SP2/0-EVs partially recapitulated muscle loss in healthy mice. SP2/0-EVs-derived RAGE increased ROS production, inflammation, and myotube atrophy in MM through TLR4/NF-κB p65 pathway activation.
Collapse
Affiliation(s)
- Yue Wu
- Department of Orthopedics, Beijing Chao-Yang Hospital, Beijing, China
| | - Xingchen Yao
- Department of Orthopedics, Beijing Chao-Yang Hospital, Beijing, China
| | - Xiangjun Shi
- Department of Orthopedics, Beijing Chao-Yang Hospital, Beijing, China
| | - Ziyu Xu
- Department of Orthopedics, Beijing Chao-Yang Hospital, Beijing, China
| | - Jie Ren
- Department of Orthopedics, Beijing Chao-Yang Hospital, Beijing, China
| | - Ming Shi
- Department of Orthopedics, Beijing Chao-Yang Hospital, Beijing, China
| | - Meng Li
- Department of Orthopedics, Beijing Chao-Yang Hospital, Beijing, China
| | - Junpeng Liu
- Department of Orthopedics, Beijing Chao-Yang Hospital, Beijing, China
| | - Xinru Du
- Department of Orthopedics, Beijing Chao-Yang Hospital, Beijing, China.
- Beijing Chao-Yang Hospital, No.8 Gongti South Rd, Chaoyang District, 100020, Beijing, China.
| |
Collapse
|
14
|
Calvez V, Becherucci G, Covello C, Piccirilli G, Mignini I, Esposto G, Laterza L, Ainora ME, Scaldaferri F, Gasbarrini A, Zocco MA. Navigating the Intersection: Sarcopenia and Sarcopenic Obesity in Inflammatory Bowel Disease. Biomedicines 2024; 12:1218. [PMID: 38927425 PMCID: PMC11200968 DOI: 10.3390/biomedicines12061218] [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: 03/25/2024] [Revised: 05/19/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Inflammatory bowel diseases (IBDs) are intricate systemic conditions that can extend beyond the gastrointestinal tract through both direct and indirect mechanisms. Sarcopenia, characterized by a reduction in muscle mass and strength, often emerges as a consequence of the clinical course of IBDs. Indeed, sarcopenia exhibits a high prevalence in Crohn's disease (52%) and ulcerative colitis (37%). While computed tomography and magnetic resonance imaging remain gold-standard methods for assessing muscle mass, ultrasound is gaining traction as a reliable, cost-effective, and widely available diagnostic method. Muscle strength serves as a key indicator of muscle function, with grip strength test emerging nowadays as the most reliable assessment method. In IBDs, sarcopenia may arise from factors such as inflammation, malnutrition, and gut dysbiosis, leading to the formulation of the 'gut-muscle axis' hypothesis. This condition determines an increased need for surgery with poorer post-surgical outcomes and a reduced response to biological treatments. Sarcopenia and its consequences lead to reduced quality of life (QoL), in addition to the already impaired QoL. Of emerging concern is sarcopenic obesity in IBDs, a challenging condition whose pathogenesis and management are still poorly understood. Resistance exercise and nutritional interventions, particularly those aimed at augmenting protein intake, have demonstrated efficacy in addressing sarcopenia in IBDs. Furthermore, anti-TNF biological therapies showed interesting outcomes in managing this condition. This review seeks to furnish a comprehensive overview of sarcopenia in IBDs, elucidating diagnostic methodologies, pathophysiological mechanisms, and clinical implications and management. Attention will also be paid to sarcopenic obesity, exploring the pathophysiology and possible treatment modalities of this condition.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Maria Assunta Zocco
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Catholic University of Rome, 00168 Rome, Italy; (V.C.); (G.B.); (C.C.); (G.P.); (I.M.); (G.E.); (L.L.); (M.E.A.); (F.S.); (A.G.)
| |
Collapse
|
15
|
Langer HT, Rohm M, Goncalves MD, Sylow L. AMPK as a mediator of tissue preservation: time for a shift in dogma? Nat Rev Endocrinol 2024:10.1038/s41574-024-00992-y. [PMID: 38760482 DOI: 10.1038/s41574-024-00992-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/19/2024] [Indexed: 05/19/2024]
Abstract
Ground-breaking discoveries have established 5'-AMP-activated protein kinase (AMPK) as a central sensor of metabolic stress in cells and tissues. AMPK is activated through cellular starvation, exercise and drugs by either directly or indirectly affecting the intracellular AMP (or ADP) to ATP ratio. In turn, AMPK regulates multiple processes of cell metabolism, such as the maintenance of cellular ATP levels, via the regulation of fatty acid oxidation, glucose uptake, glycolysis, autophagy, mitochondrial biogenesis and degradation, and insulin sensitivity. Moreover, AMPK inhibits anabolic processes, such as lipogenesis and protein synthesis. These findings support the notion that AMPK is a crucial regulator of cell catabolism. However, studies have revealed that AMPK's role in cell homeostasis might not be as unidirectional as originally thought. This Review explores emerging evidence for AMPK as a promoter of cell survival and an enhancer of anabolic capacity in skeletal muscle and adipose tissue during catabolic crises. We discuss AMPK-activating interventions for tissue preservation during tissue wasting in cancer-associated cachexia and explore the clinical potential of AMPK activation in wasting conditions. Overall, we provide arguments that call for a shift in the current dogma of AMPK as a mere regulator of cell catabolism, concluding that AMPK has an unexpected role in tissue preservation.
Collapse
Affiliation(s)
- Henning Tim Langer
- Division of Endocrinology, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riβ, Germany.
| | - Maria Rohm
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Marcus DaSilva Goncalves
- Division of Endocrinology, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Lykke Sylow
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
16
|
Lei T, Rui Y, Xiaoshuang Z, Jinglan Z, Jihong Z. Mitochondria transcription and cancer. Cell Death Discov 2024; 10:168. [PMID: 38589371 PMCID: PMC11001877 DOI: 10.1038/s41420-024-01926-3] [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: 12/14/2023] [Revised: 03/14/2024] [Accepted: 03/20/2024] [Indexed: 04/10/2024] Open
Abstract
Mitochondria are major organelles involved in several processes related to energy supply, metabolism, and cell proliferation. The mitochondria function is transcriptionally regulated by mitochondria DNA (mtDNA), which encodes the key proteins in the electron transport chain that is indispensable for oxidative phosphorylation (OXPHOS). Mitochondrial transcriptional abnormalities are closely related to a variety of human diseases, such as cardiovascular diseases, and diabetes. The mitochondria transcription is regulated by the mtDNA, mitochondrial RNA polymerase (POLRMT), two transcription factors (TFAM and TF2BM), one transcription elongation (TEFM), and one known transcription termination factor (mTERFs). Dysregulation of these factors directly leads to altered expression of mtDNA in tumor cells, resulting in cellular metabolic reprogramming and mitochondrial dysfunction. This dysregulation plays a role in modulating tumor progression. Therefore, understanding the role of mitochondrial transcription in cancer can have implications for cancer diagnosis, prognosis, and treatment. Targeting mitochondrial transcription or related pathways may provide potential therapeutic strategies for cancer treatment. Additionally, assessing mitochondrial transcriptional profiles or biomarkers in cancer cells or patient samples may offer diagnostic or prognostic information.
Collapse
Affiliation(s)
- Tang Lei
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Yu Rui
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Zhou Xiaoshuang
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Zhang Jinglan
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Zhang Jihong
- Medical School, Kunming University of Science and Technology, Kunming, China.
- Yunnan Province Clinical Research Center for Hematologic Disease, Kunming, China.
| |
Collapse
|
17
|
Yu X, Ren P, Yang R, Yue H, Tang Q, Xue C. Astaxanthin Ameliorates Skeletal Muscle Atrophy in Mice With Cancer Cachexia. Nutr Cancer 2024; 76:529-542. [PMID: 38567899 DOI: 10.1080/01635581.2024.2335584] [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: 12/26/2023] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 06/06/2024]
Abstract
Astaxanthin (AST) is a natural marine carotenoid with a variety of biological activities. This study aimed to demonstrate the possible mechanisms by which AST improves skeletal muscle atrophy in cancer cachexia. In this study, the effects of different doses of AST (30 mg/kg b.w., 60 mg/kg b.w. and 120 mg/kg b.w.) on skeletal muscle functions were explored in mice with cancer cachexia. The results showed that AST (30, 60 and 120 mg/kg b.w.) could effectively protect cachexia mice from body weight and skeletal muscle loss. AST dose-dependently ameliorated the decrease in myofibres cross-sectional area and increased the expression of myosin heavy chain (MHC). AST treatment decreased both the serum and muscle level of IL-6 but not TNF-α in C26 tumor-bearing cachexia mice. Moreover, AST alleviated skeletal muscle atrophy by decreasing the expression of two muscle-specific E3 ligases MAFBx and MuRF-1. AST improved mitochondrial function by downregulating the levels of muscle Fis1, LC3B and Bax, upregulating the levels of muscle Mfn2 and Bcl-2. In conclusion, our study show that AST might be expected to be a nutritional supplement for cancer cachexia patients.
Collapse
Affiliation(s)
- Xinyue Yu
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Shandong, Qingdao, China
| | - Pengfei Ren
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Shandong, Qingdao, China
| | - Ruzhen Yang
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Shandong, Qingdao, China
| | - Han Yue
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Shandong, Qingdao, China
| | - Qingjuan Tang
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Shandong, Qingdao, China
| | - Changhu Xue
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Shandong, Qingdao, China
- Laboratory of Marine Drugs and Biological Products, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
18
|
Hesketh SJ. Advancing cancer cachexia diagnosis with -omics technology and exercise as molecular medicine. SPORTS MEDICINE AND HEALTH SCIENCE 2024; 6:1-15. [PMID: 38463663 PMCID: PMC10918365 DOI: 10.1016/j.smhs.2024.01.006] [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: 09/21/2023] [Revised: 01/15/2024] [Accepted: 01/20/2024] [Indexed: 03/12/2024] Open
Abstract
Muscle atrophy exacerbates disease outcomes and increases mortality, whereas the preservation of skeletal muscle mass and function play pivotal roles in ensuring long-term health and overall quality-of-life. Muscle atrophy represents a significant clinical challenge, involving the continued loss of muscle mass and strength, which frequently accompany the development of numerous types of cancer. Cancer cachexia is a highly prevalent multifactorial syndrome, and although cachexia is one of the main causes of cancer-related deaths, there are still no approved management strategies for the disease. The etiology of this condition is based on the upregulation of systemic inflammation factors and catabolic stimuli, resulting in the inhibition of protein synthesis and enhancement of protein degradation. Numerous necessary cellular processes are disrupted by cachectic pathology, which mediate intracellular signalling pathways resulting in the net loss of muscle and organelles. However, the exact underpinning molecular mechanisms of how these changes are orchestrated are incompletely understood. Much work is still required, but structured exercise has the capacity to counteract numerous detrimental effects linked to cancer cachexia. Primarily through the stimulation of muscle protein synthesis, enhancement of mitochondrial function, and the release of myokines. As a result, muscle mass and strength increase, leading to improved mobility, and quality-of-life. This review summarises existing knowledge of the complex molecular networks that regulate cancer cachexia and exercise, highlighting the molecular interplay between the two for potential therapeutic intervention. Finally, the utility of mass spectrometry-based proteomics is considered as a way of establishing early diagnostic biomarkers of cachectic patients.
Collapse
|
19
|
Cardaci TD, VanderVeen BN, Bullard BM, McDonald SJ, Unger CA, Enos RT, Fan D, Velázquez KT, Frizzell N, Spangenburg EE, Murphy EA. Obesity worsens mitochondrial quality control and does not protect against skeletal muscle wasting in murine cancer cachexia. J Cachexia Sarcopenia Muscle 2024; 15:124-137. [PMID: 38062911 PMCID: PMC10834333 DOI: 10.1002/jcsm.13391] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 09/15/2023] [Accepted: 11/02/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND More than 650 million people are obese (BMI > 30) worldwide, which increases their risk for several metabolic diseases and cancer. While cachexia and obesity are at opposite ends of the weight spectrum, leading many to suggest a protective effect of obesity against cachexia, mechanistic support for obesity's benefit is lacking. Given that obesity and cachexia are both accompanied by metabolic dysregulation, we sought to investigate the impact of obesity on skeletal muscle mass loss and mitochondrial dysfunction in murine cancer cachexia. METHODS Male C57BL/6 mice were given a purified high fat or standard diet for 16 weeks before being implanted with 106 Lewis lung carcinoma (LLC) cells. Mice were monitored for 25 days, and hindlimb muscles were collected for cachexia indices and mitochondrial assessment via western blotting, high-resolution respirometry and transmission electron microscopy (TEM). RESULTS Obese LLC mice experienced significant tumour-free body weight loss similar to lean (-12.8% vs. -11.8%, P = 0.0001) but had reduced survival (33.3% vs. 6.67%, χ2 = 10.04, P = 0.0182). Obese LLC mice had reduced muscle weights (-24%, P < 0.0354) and mCSA (-16%, P = 0.0004) with similar activation of muscle p65 (P = 0.0337), and p38 (P = 0.0008). ADP-dependent coupled respiration was reduced in both Obese and Obese LLC muscle (-30%, P = 0.0072) consistent with reductions in volitional cage activity (-39%, P < 0.0001) and grip strength (-41%, P < 0.0001). TEM revealed stepwise reductions in intermyofibrillar and subsarcolemmal mitochondrial size with Obese (IMF: -37%, P = 0.0009; SS: -21%, P = 0.0101) and LLC (IMF: -40%, P = 0.0019; SS: -27%, P = 0.0383) mice. Obese LLC mice had increased pAMPK (T172; P = 0.0103) and reduced FIS1 (P = 0.0029) and DRP1 (P < 0.0001) mitochondrial fission proteins, which were each unchanged in Lean LLC. Further, mitochondrial TEM analysis revealed that Obese LLC mice had an accumulation of damaged and dysfunctional mitochondria (IMF: 357%, P = 0.0395; SS: 138%, P = 0.0174) in concert with an accumulation of p62 (P = 0.0328) suggesting impaired autophagy and clearance of damaged mitochondria. Moreover, we observed increases in electron lucent vacuoles only in Obese LLC muscle (IMF: 421%, P = 0.0260; SS: 392%, P = 0.0192), further supporting an accumulation of damaged materials that cannot be properly cleared in the obese cachectic muscle. CONCLUSIONS Taken together, these results demonstrate that obesity is not protective against cachexia and suggest exacerbated impairments to mitochondrial function and quality control with a particular disruption in the removal of damaged mitochondria. Our findings highlight the need for consideration of the severity of obesity and pre-existing metabolic conditions when determining the impact of weight status on cancer-induced cachexia and functional mitochondrial deficits.
Collapse
Affiliation(s)
- Thomas D Cardaci
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Brandon N VanderVeen
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Brooke M Bullard
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Sierra J McDonald
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Christian A Unger
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Reilly T Enos
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Daping Fan
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Kandy T Velázquez
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Norma Frizzell
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Espen E Spangenburg
- Department of Physiology, East Carolina University, Greenville, North Carolina, USA
- Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| | - E Angela Murphy
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| |
Collapse
|
20
|
Nunthanawanich P, Wichansawakun S, Luangjinda C, Hudthagosol C. Effectiveness of Web Applications on Improving Nutritional Status of Patients with Colorectal Cancer. Nutrients 2024; 16:408. [PMID: 38337693 PMCID: PMC10857273 DOI: 10.3390/nu16030408] [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: 11/10/2023] [Revised: 01/11/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
The most common cancer in Thailand is colorectal cancer (CRC). A lack of knowledge and misleading information from social media have contributed to cancer deaths from malnutrition. A web application is a tool that provides easy access to scientific nutritional information via an online platform. In this study, our goal was to compare the nutritional status of CRC patients using different nutrition-based educational tools with nutrition counseling, namely the Nutrition Educational Prototype based on Smartphone Web Applications (NEPSA) and standard hospital leaflets. Anthropometric and biochemical analyses and a dietary assessment, especially calories and protein, were measured during three visits. This study finally included 28 CRC patients who were undergoing chemotherapy and malnutrition with a body mass index (BMI) of <20 kg/m2. Thirteen participants received NEPSA while the remaining fifteen participants received a standard hospital leaflet. The results showed that NEPSAs improved nutritional outcomes by encouraging weight gain, increasing BMI, hemoglobin, hematocrit, and albumin levels, and consuming more calories and protein. NEPSA should be implemented to enhance the nutrition outcomes from anthropometric, biochemical, and dietary perspectives from nutrition advice among CRC patients. There could be positive impacts at the national level regarding equal accessibility to Thailand's nutrition information.
Collapse
Affiliation(s)
- Pornpimon Nunthanawanich
- Doctor of Public Health (International Program), Faculty of Public Health, Mahidol University, Bangkok 10400, Thailand
| | - Sanit Wichansawakun
- Division of Clinical Nutrition, Department of Internal Medicine, Faculty of Medicine, Thammasat University, Pathum Thani 12120, Thailand;
| | - Cholrit Luangjinda
- Research and Development Office, Thailand Institute of Occupational Safety and Health, Ministry of Labor, Bangkok 10170, Thailand
| | - Chatrapa Hudthagosol
- Department of Nutrition, Faculty of Public Health, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
21
|
López-Gómez JJ, Bachiller BR, de Luis Roman D. Management of disease-related malnutrition: a real-world experience with a novel concentrated high-protein energy-dense oral nutritional supplement. Postgrad Med 2024; 136:52-59. [PMID: 38251982 DOI: 10.1080/00325481.2024.2307869] [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: 11/23/2023] [Accepted: 01/17/2024] [Indexed: 01/23/2024]
Abstract
OBJECTIVE Despite the availability of a wide range of oral nutritional supplements (ONS) offerings, individuals with malnutrition are still struggling to meet their nutritional targets. A new concentrated and high-protein energy-dense ONS (≥2.1 kcal/mL;32 g protein/200 mL) with high-quality protein (60% whey protein) has emerged as a pivotal formula to reach the patient's energy-protein requirements, enhance compliance, and maximize stimulation of muscle protein synthesis, key factors driving better nutritional, functional, and clinical outcomes. The purpose of this article is to provide our clinical experience using this new nutritionally concentrated ONS as a therapeutic strategy for patients with DRM. METHODS Three clinical cases have been examined using new assessment procedures and a new form of nutritional therapy, and their impact on the nutritional and functional outcomes in patients with moderate-to-severe DRM. RESULTS A tailored individualized nutritional interventions improved anthropometric, biochemical, and functional outcomes (Case 1,2, and 3) assessed using hand grip strength, bioimpedance and muscle ultrasound, and as well as good gastrointestinal tolerance (Case 1) and compliance to the ONS in patients with DRM (Case 1,2,3). CONCLUSION The use of this novel high-protein energy-dense formula with high-quality protein source (≥2.1 kcal/mL; 32 g protein/200 mL; 60% whey protein) overcome common practical challenges in the medical nutrition therapy of patients with DRM, either because these patients require a highly concentrated formulation to meet nutritional requirements due to loss of appetite, lack of interest in food, and high caloric-protein needs due to disease, and a large quantity and quality of protein to optimize muscle recovery due to sarcopenia, common in patients with moderate-severe malnutrition.
Collapse
Affiliation(s)
- Juan J López-Gómez
- Centro de Investigación de Endocrinología y Nutrición, Facultad de Medicina Valladolid, Servicio Endocrinología y Nutrición Hospital Clínico Universitario, Valladolid, Spain
| | - Beatriz Ramos Bachiller
- Centro de Investigación de Endocrinología y Nutrición, Facultad de Medicina Valladolid, Servicio Endocrinología y Nutrición Hospital Clínico Universitario, Valladolid, Spain
| | - Daniel de Luis Roman
- Centro de Investigación de Endocrinología y Nutrición, Facultad de Medicina Valladolid, Servicio Endocrinología y Nutrición Hospital Clínico Universitario, Valladolid, Spain
| |
Collapse
|
22
|
Papadopetraki A, Giannopoulos A, Maridaki M, Zagouri F, Droufakou S, Koutsilieris M, Philippou A. The Role of Exercise in Cancer-Related Sarcopenia and Sarcopenic Obesity. Cancers (Basel) 2023; 15:5856. [PMID: 38136400 PMCID: PMC10741686 DOI: 10.3390/cancers15245856] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/03/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
One of the most common adverse effects of cancer and its therapeutic strategies is sarcopenia, a condition which is characterised by excess muscle wasting and muscle strength loss due to the disrupted muscle homeostasis. Moreover, cancer-related sarcopenia may be combined with the increased deposition of fat mass, a syndrome called cancer-associated sarcopenic obesity. Both clinical conditions have significant clinical importance and can predict disease progression and survival. A growing body of evidence supports the claim that physical exercise is a safe and effective complementary therapy for oncology patients which can limit the cancer- and its treatment-related muscle catabolism and promote the maintenance of muscle mass. Moreover, even after the onset of sarcopenia, exercise interventions can counterbalance the muscle mass loss and improve the clinical appearance and quality of life of cancer patients. The aim of this narrative review was to describe the various pathophysiological mechanisms, such as protein synthesis, mitochondrial function, inflammatory response, and the hypothalamic-pituitary-adrenal axis, which are regulated by exercise and contribute to the management of sarcopenia and sarcopenic obesity. Moreover, myokines, factors produced by and released from exercising muscles, are being discussed as they appear to play an important role in mediating the beneficial effects of exercise against sarcopenia.
Collapse
Affiliation(s)
- Argyro Papadopetraki
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece; (A.P.)
| | - Antonios Giannopoulos
- Section of Sports Medicine, Department of Community Medicine & Rehabilitation, Umeå University, 901 87 Umeå, Sweden;
- National Centre for Sport and Exercise Medicine (NCSEM), School of Sport, Exercise and Health Sciences, Loughborough University, Leicestershire LE11 3TU, UK
| | - Maria Maridaki
- Faculty of Physical Education and Sport Science, National and Kapodistrian University of Athens, 172 37 Dafne, Greece
| | - Flora Zagouri
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, National and Kapodistrian University of Athens, 115 28 Athens, Greece
| | | | - Michael Koutsilieris
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece; (A.P.)
| | - Anastassios Philippou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece; (A.P.)
| |
Collapse
|
23
|
Zhou J, Li M, Yu Z, Li C, Zhou L, Zhou X. Protective effect of Qingluotongbi formula against Tripterygium wilfordii induced liver injury in mice by improving fatty acid β-oxidation and mitochondrial biosynthesis. PHARMACEUTICAL BIOLOGY 2023; 61:80-88. [PMID: 36541729 PMCID: PMC9788700 DOI: 10.1080/13880209.2022.2157842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/21/2022] [Accepted: 12/07/2022] [Indexed: 06/17/2023]
Abstract
CONTEXT Qingluotongbi formula (QLT) is a Chinese medicine compound consisting of Tripterygium wilfordii Hook. f. (Celastraceae, TW), Panax notoginseng (Burkill) F.H.Chen (Araliaceae, PN), Rehmannia glutinosa (Gaertn.) DC. (Orobanchaceae, RG), Sinomenium acutum (Thunb.) Rehder & E.H. Wilson (Menispermaceae, SA), and Bombyx mori L. (Bombycidae, BM). OBJECTIVE This study investigated the protective effect and possible mechanism of QLT against TW-induced liver injury in mice. MATERIALS AND METHODS To establish the model of TW-induced liver injury in mice, C57BL/6J mice were randomly divided into 4 groups: control group, low-dose TW group, middle-dose TW group, and high-dose TW group. To observe the effects of QLT and its individual ingredients against TW-induced liver injury, C57BL/6J mice were randomly divided into 7 groups: control group, TW group, QLT group, PN group, RG group, SA group, BM group.After administration for 7 days, C57BL/6J mice were tested for biochemical indicators and liver pathological changes. Then, we evaluated the mitochondrial function and analysed the gene and protein expression related to the peroxisome proliferator-activated receptor alpha (PPARα)/peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) pathway by quantitative real-time PCR (qRT-PCR) and Western blotting. RESULTS Compared with the control group (0.30 ± 0.35), TW significantly increased mice liver histological score (L, 0.95 ± 1.14; M, 1.25 ± 1.16; H, 4.00 ± 1.13). QLT and its ingredients significantly improved the pathology scores (CON, 0.63 ± 0.74; TW, 4.19 ± 1.53; QLT, 1.56 ± 0.62; PN, 1.94 ± 0.68; RG, 2.75 ± 1.39; SA, 4.13 ± 0.99; BM, 4.13 ± 0.99). Western blot and qRT-PCR analysis revealed that QLT and its ingredients reversed TW-induced suppression of PPARα/PGC1-α pathway.Discussion and conclusions: These findings provide valuable information for compound compatibility studies and TW clinical applications.
Collapse
Affiliation(s)
- Jie Zhou
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ming Li
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Zhichao Yu
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Changqing Li
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lingling Zhou
- Jiangsu Provincial Key Laboratory of Pharmacology and Safety Evaluation of Material Medical, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xueping Zhou
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
24
|
Zhang Y, Zhao Y, Rong J, Liu K, Zhan Y, Chai Y, Ning J, Yuan W. A bibliometric analysis of inflammation in sarcopenia from 2007 to 2022. Exp Gerontol 2023; 183:112316. [PMID: 37862732 DOI: 10.1016/j.exger.2023.112316] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/13/2023] [Accepted: 10/17/2023] [Indexed: 10/22/2023]
Abstract
OBJECTIVE In recent years, the impact of inflammation regulation on the progression of sarcopenia has garnered significant attention in research. However, there has been a lack of bibliometric analysis on the literature pertaining to inflammation in sarcopenia. This study was designed for the purpose of exploring the current research trends in this field as well as general situations and hot spots through bibliometric analysis. METHODS Searches were performed on the Web of Science Core Collection for articles related to inflammation in sarcopenia from 2007 to 2022, and selected in compliance with PRISMA guidelines. A variety of data were analyzed and visualized using VOSviewer and CiteSpace, including countries, institutions, authors, keywords, journals, and publications. RESULTS 1833 articles were obtained in the last 16 years in all. The number of publications and citations increased from 2007 to 2022, with a notable rise occurring after 2016. Based on the results, the United States, the University of Melbourne, Nutrients, and Marzetti Emanuele were the most productive countries, institutions, journals, and authors, respectively. The primary keywords were oxidative stress and insulin resistance, and the burst detection analysis of keywords found that there is a possibility that future research will focus on "Inflammatory Bowel Disease". CONCLUSION This is the first bibliometric analysis of inflammation in sarcopenia. The interaction between oxidative stress, insulin resistance and inflammation in sarcopenia is regarded as the current research priorities. As sarcopenia becomes more prevalent, a focus will be placed on determining the molecular mechanisms and therapeutic targets for regulating inflammation to intervene in sarcopenia.
Collapse
Affiliation(s)
- Yujie Zhang
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ye Zhao
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiaqi Rong
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Kaoqiang Liu
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yunfan Zhan
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yongli Chai
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiawei Ning
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wei'an Yuan
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
25
|
Wickramasinghe HKJP, Stepanchenko N, Oconitrillo MJ, Goetz BM, Abeyta MA, Gorden PJ, Baumgard LH, Appuhamy JADRN. Effects of a phytogenic feed additive on weaned dairy heifer calves subjected to a diurnal heat stress bout. J Dairy Sci 2023; 106:6114-6127. [PMID: 37479578 DOI: 10.3168/jds.2022-22856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 03/17/2023] [Indexed: 07/23/2023]
Abstract
The study objective was to evaluate the effects of a phytogenic feed additive (PFA) on dry matter intake (DMI), average daily gain (ADG), inflammation, and oxidative stress markers of heifer calves exposed to a heat stress bout in the summer. A total of18 Holstein and 4 Jersey heifer calves (192 ± 5 kg of body weight at 162 ± 16 d of age) housed in indoor stalls were assigned to 1 of 2 dietary treatments (n = 11; 9 Holstein and 2 Jersey): (1) a basal total mixed ration (CTL), and (2) CTL top-dressed with 0.25 g/d of PFA. Following 7 d of acclimation, baseline measurements were made over 7 d under regular summer conditions [average temperature-humidity index (THI) = 79 from 0900 to 2000 h, and 75 from 2000 to 0900 h]. Calves were then subjected to a 7-d cyclic heat stress bout (HS) by turning on barn heaters and increasing the barn temperature to 33.0°C only during the daytime (the average THI = 85 from 0900 to 2000 h). The study continued for an extra 4-d period after HS ended (post-HS). The HS increased rectal temperature, skin temperature, and respiration rate from the baseline by 1.0°C, 4.0°C, and 49 breaths/min, respectively. The drinking water intake increased by 32% in response to HS, and calves continued to consume more water (44%) than the baseline consumption even after HS ended. The treatment × time interactions were not significant for feed intake, ADG, partial pressure of O2 in the blood, and blood concentrations of inflammation markers such as haptoglobin and lipopolysaccharide binding protein (LBP), and antioxidant markers such as protein carbonyl and thiobarbituric acid (TBARS). The PFA tended to increase daytime DMI (0.24 kg/d) compared with CTL throughout the experiment but did not affect ADG, which decreased from 1.12 kg/d to 0.26 kg/d in response to HS. Both DMI (13%) and ADG (85%) increased during post-HS relative to baseline, indicating compensatory performances that were not affected by the PFA. Serum haptoglobin and plasma LBP concentrations of PFA calves were 44% and 38% lower than that of CTL calves across all time points. The PFA decreased O2 pressure and tended to decrease protein carbonyl concentration in the blood across all time points. The PFA tended to decrease TBARS concentration on the first day of HS and increase and decrease the ratio of reduced to oxidized glutathione in the blood during the baseline and post-HS periods, respectively. Despite the lack of growth improvements, feeding PFA seems to increase O2 levels in the blood and alleviate oxidative stress and inflammation of heifer calve exposed to diurnal heat waves (~7 d) in the summer.
Collapse
Affiliation(s)
| | - N Stepanchenko
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | - M J Oconitrillo
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | - B M Goetz
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | - M A Abeyta
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | - P J Gorden
- Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA 50011
| | - L H Baumgard
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | | |
Collapse
|
26
|
Wesolowski LT, Simons JL, Semanchik PL, Othman MA, Kim JH, Lawler JM, Kamal KY, White-Springer SH. The Impact of SRT2104 on Skeletal Muscle Mitochondrial Function, Redox Biology, and Loss of Muscle Mass in Hindlimb Unloaded Rats. Int J Mol Sci 2023; 24:11135. [PMID: 37446313 DOI: 10.3390/ijms241311135] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/01/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Mechanical unloading during microgravity causes skeletal muscle atrophy and impairs mitochondrial energetics. The elevated production of reactive oxygen species (ROS) by mitochondria and Nox2, coupled with impairment of stress protection (e.g., SIRT1, antioxidant enzymes), contribute to atrophy. We tested the hypothesis that the SIRT1 activator, SRT2104 would rescue unloading-induced mitochondrial dysfunction. Mitochondrial function in rat gastrocnemius and soleus muscles were evaluated under three conditions (10 days): ambulatory control (CON), hindlimb unloaded (HU), and hindlimb-unloaded-treated with SRT2104 (SIRT). Oxidative phosphorylation, electron transfer capacities, H2O2 production, and oxidative and antioxidant enzymes were quantified using high-resolution respirometry and colorimetry. In the gastrocnemius, (1) integrative (per mg tissue) proton LEAK was lesser in SIRT than in HU or CON; (2) intrinsic (relative to citrate synthase) maximal noncoupled electron transfer capacity (ECI+II) was lesser, while complex I-supported oxidative phosphorylation to ECI+II was greater in HU than CON; (3) the contribution of LEAK to ECI+II was greatest, but cytochrome c oxidase activity was lowest in HU. In both muscles, H2O2 production and concentration was greatest in SIRT, as was gastrocnemius superoxide dismutase activity. In the soleus, H2O2 concentration was greater in HU compared to CON. These results indicate that SRT2104 preserves mitochondrial function in unloaded skeletal muscle, suggesting its potential to support healthy muscle cells in microgravity by promoting necessary energy production in mitochondria.
Collapse
Affiliation(s)
- Lauren T Wesolowski
- Department of Animal Science, College of Agriculture and Life Science, Texas A&M University and Texas A&M AgriLife Research, College Station, TX 77843, USA
| | - Jessica L Simons
- Department of Animal Science, College of Agriculture and Life Science, Texas A&M University and Texas A&M AgriLife Research, College Station, TX 77843, USA
| | - Pier L Semanchik
- Department of Animal Science, College of Agriculture and Life Science, Texas A&M University and Texas A&M AgriLife Research, College Station, TX 77843, USA
| | - Mariam A Othman
- Department of Kinesiology & Sport Management, School of Education and Human Development, Texas A&M University, College Station, TX 77843, USA
| | - Joo-Hyun Kim
- Department of Kinesiology & Sport Management, School of Education and Human Development, Texas A&M University, College Station, TX 77843, USA
| | - John M Lawler
- Department of Kinesiology & Sport Management, School of Education and Human Development, Texas A&M University, College Station, TX 77843, USA
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Khaled Y Kamal
- Department of Kinesiology & Sport Management, School of Education and Human Development, Texas A&M University, College Station, TX 77843, USA
| | - Sarah H White-Springer
- Department of Animal Science, College of Agriculture and Life Science, Texas A&M University and Texas A&M AgriLife Research, College Station, TX 77843, USA
| |
Collapse
|
27
|
Boran T, Zengin OS, Seker Z, Akyildiz AG, Oztas E, Özhan G. Ripretinib induced skeletal muscle toxicity through mitochondrial impairment in C2C12 myotubes. Toxicology 2023; 489:153489. [PMID: 36933644 DOI: 10.1016/j.tox.2023.153489] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 03/03/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023]
Abstract
Ripretinib is a multikinase inhibitor drug approved in 2020 by the FDA and in 2021 by EMA for use in the treatment of advanced gastrointestinal stromal tumors (GIST) which have not adequately responded to previous treatments with kinase inhibitors. The most common side effects of the drug are myalgia and fatigue, which likely causes interruption of the treatment or reduction of the dose. Skeletal muscle cells highly depend on ATP to perform their functions and mitochondrial damage may play a role in skeletal muscle toxicity induced by kinase inhibitors. However, the molecular mechanism has not been clearly identified in the literature yet. In this study, it has been aimed to elucidate the role of mitochondria in the toxic effect of ripretinib on skeletal muscle using the mouse C2C12 myoblast-derived myotubes. The myotubes were exposed to ripretinib at the range of 1-20 μM concentrations for 24 h. To determine the potential role of mitochondrial impairment in ripretinib-induced skeletal muscle toxicity, intracellular ATP level, mitochondrial membrane potential (MMP), mitochondrial ROS production (mtROS), mitochondrial DNA (mtDNA) copy number, and mitochondrial mass were examined after ripretinib treatment. Furthermore, changes in PGC 1α/NRF 1/NRF 2 expression levels that play a role in mitochondrial biogenesis and mitophagy were investigated. Additionally, the mitochondrial electron transport chain (ETC) enzyme activities were evaluated. Lastly, a molecular docking study was done to see ripretinib's possible interaction with DNA polymerase gamma (POLG) which is important for DNA replication in the mitochondria. According to the findings, ripretinib decreases the ATP level and mtDNA copy number, induces loss of MMP, and reduces mitochondrial mass. The activities of the ETC complexes were inhibited with ripretinib exposure which is in line with the observed ATP depletion and MMP loss. The molecular docking study revealed that ripretinib has inhibitory potential against POLG which supports the observed inhibition of mtDNA. The expression of PGC 1α was reduced in the nuclear fraction indicating that PGC-1α was not activated since the NRF 1 expression was reduced and NRF 2 level did not show significant change. Consequently, mtROS production increased in all treatment groups and mitophagy-related gene expressions and Parkin protein expression level were up-regulated at high doses. In conclusion, mitochondrial damage/loss can be one of the underlying causes of ripretinib-induced skeletal muscle toxicity. However, further studies are needed to confirm the results in vivo.
Collapse
Affiliation(s)
- Tugce Boran
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34116 Istanbul, Turkey; Istanbul University-Cerrahpaşa, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34500 Istanbul, Turkey
| | - Ozge Sultan Zengin
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34116 Istanbul, Turkey; Institute of Graduate Studies in Health Sciences, Istanbul University, 34116 Istanbul, Turkey
| | - Zehra Seker
- Institute of Graduate Studies in Health Sciences, Istanbul University, 34116 Istanbul, Turkey; Bezmialem Vakif University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34093 Istanbul, Turkey
| | - Aysenur Gunaydin Akyildiz
- Bezmialem Vakif University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34093 Istanbul, Turkey
| | - Ezgi Oztas
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34116 Istanbul, Turkey
| | - Gül Özhan
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34116 Istanbul, Turkey.
| |
Collapse
|
28
|
Shi H, Qi H, Xie D, Zhuang J, Qi H, Dai Y, Wu J. Inhibition of ACSF2 protects against renal ischemia/reperfusion injury via mediating mitophagy in proximal tubular cells. Free Radic Biol Med 2023; 198:68-82. [PMID: 36764625 DOI: 10.1016/j.freeradbiomed.2023.02.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/27/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023]
Abstract
Acute kidney injury (AKI) is a prevalent clinical condition caused by sepsis and ischemia reperfusion (IR) injury. The principal driver of IR-induced AKI involves renal tubular structural changes triggered by the impairment of function in renal tubular cells. The target gene, Acyl-CoA Synthetase Family Member 2 (ACSF2), was retrieved from the GEO database based on high specific expression in renal tubular cells and location in mitochondria. Here, we substantiate that ACSF2 is specifically localized in the mitochondria of the renal tubular epithelium. Functionally silencing ACSF2 in HK2 cells enhanced hypoxia-reoxygenation (HR)-induced mitophagy, restored mitochondrial function and decreased the production of mitochondrial superoxide. Our study demonstrated that these effects were reversed by silencing Bcl-2 19-kDa interacting protein 3 (BNIP3), a receptor regulating mitophagy. In vivo, ACSF2 knockdown significantly enhanced IR-induced mitophagy and improved renal function in mice with IR injury. Conversely, BNIP3 knockdown inhibited mitophagy and exacerbated renal damage in ACSF2-knockdown mice with IR injury. In conclusion, our study demonstrated that inhibition of ACSF2 enhances mitophagy, restoring mitochondrial function and protects against IR-induced AKI, providing a new target and potential strategy for therapy.
Collapse
Affiliation(s)
- Haoyu Shi
- Department of Urology, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
| | - Hao Qi
- Department of Urology, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
| | - Dongdong Xie
- Department of Urology, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
| | - Jiayi Zhuang
- Department of Urology, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
| | - Huiyue Qi
- Department of Urology, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
| | - Yingbo Dai
- Department of Urology, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China.
| | - Jiaqing Wu
- Department of Urology, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China.
| |
Collapse
|
29
|
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
|
30
|
Unveiling the Role of the Proton Gateway, Uncoupling Proteins (UCPs), in Cancer Cachexia. Cancers (Basel) 2023; 15:cancers15051407. [PMID: 36900198 PMCID: PMC10000250 DOI: 10.3390/cancers15051407] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/30/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Uncoupling proteins (UCPs) are identified as carriers of proton ions between the mitochondrial inner membrane and the mitochondrial matrix. ATP is mainly generated through oxidative phosphorylation in mitochondria. The proton gradient is generated across the inner mitochondrial membrane and the mitochondrial matrix, which facilitates a smooth transfer of electrons across ETC complexes. Until now, it was thought that the role of UCPs was to break the electron transport chain and thereby inhibit the synthesis of ATP. UCPs allow protons to pass from the inner mitochondrial membrane to the mitochondrial matrix and decrease the proton gradient across the membrane, which results in decreased ATP synthesis and increased production of heat by mitochondria. In recent years, the role of UCPs in other physiological processes has been deciphered. In this review, we first highlighted the different types of UCPs and their precise location across the body. Second, we summarized the role of UCPs in different diseases, mainly metabolic disorders such as obesity and diabetes, cardiovascular complications, cancer, wasting syndrome, neurodegenerative diseases, and kidney complications. Based on our findings, we conclude that UCPs play a major role in maintaining energy homeostasis, mitochondrial functions, ROS production, and apoptosis. Finally, our findings reveal that mitochondrial uncoupling by UCPs may treat many diseases, and extensive clinical studies are required to meet the unmet need of certain diseases.
Collapse
|
31
|
Su L, Zhang J, Gomez H, Kellum JA, Peng Z. Mitochondria ROS and mitophagy in acute kidney injury. Autophagy 2023; 19:401-414. [PMID: 35678504 PMCID: PMC9851232 DOI: 10.1080/15548627.2022.2084862] [Citation(s) in RCA: 336] [Impact Index Per Article: 168.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 05/24/2022] [Accepted: 05/26/2022] [Indexed: 01/22/2023] Open
Abstract
Mitophagy is an essential mitochondrial quality control mechanism that eliminates damaged mitochondria and the production of reactive oxygen species (ROS). The relationship between mitochondria oxidative stress, ROS production and mitophagy are intimately interwoven, and these processes are all involved in various pathological conditions of acute kidney injury (AKI). The elimination of damaged mitochondria through mitophagy in mammals is a complicated process which involves several pathways. Furthermore, the interplay between mitophagy and different types of cell death, such as apoptosis, pyroptosis and ferroptosis in kidney injury is unclear. Here we will review recent advances in our understanding of the relationship between ROS and mitophagy, the different mitophagy pathways, the relationship between mitophagy and cell death, and the relevance of these processes in the pathogenesis of AKI.Abbreviations: AKI: acute kidney injury; AMBRA1: autophagy and beclin 1 regulator 1; ATP: adenosine triphosphate; BAK1: BCL2 antagonist/killer 1; BAX: BCL2 associated X, apoptosis regulator; BCL2: BCL2 apoptosis regulator; BECN1: beclin 1; BH3: BCL2 homology domain 3; BNIP3: BCL2 interacting protein 3; BNIP3L/NIX: BCL2 interacting protein 3 like; CASP1: caspase 1; CAT: catalase; CCCP: carbonyl cyanide m-chlorophenylhydrazone; CI-AKI: contrast-induced acute kidney injury; CISD1: CDGSH iron sulfur domain 1; CL: cardiolipin; CNP: 2',3'-cyclic nucleotide 3'-phosphodiesterase; DNM1L/DRP1: dynamin 1 like; E3: enzyme 3; ETC: electron transport chain; FA: folic acid; FUNDC1: FUN14 domain containing 1; G3P: glycerol-3-phosphate; G6PD: glucose-6-phosphate dehydrogenase; GPX: glutathione peroxidase; GSH: glutathione; GSK3B: glycogen synthase kinase 3 beta; GSR: glutathione-disulfide reductase; HIF1A: hypoxia inducible factor 1 subunit alpha; HUWE1: HECT, UBA and WWE domain containing 1; IL1B: interleukin 1 beta; IMM: inner mitochondrial membrane; IPC: ischemic preconditioning; IRI: ischemia-reperfusion injury; LIR: LC3-interacting region; LPS: lipopolysaccharide; MA: malate-aspartate; MPT: mitochondrial permeability transition; MUL1: mitochondrial E3 ubiquitin protein ligase 1; mtROS: mitochondrial ROS; NLR: NOD-like receptor; NLRP3: NLR family pyrin domain containing 3; NOX: NADPH oxidase; OGD-R: oxygen-glucose deprivation-reperfusion; OMM: outer mitochondrial membrane; OPA1: OPA1 mitochondrial dynamin like GTPase; OXPHOS: oxidative phosphorylation; PARL: presenilin associated rhomboid like; PINK1: PTEN induced kinase 1; PLSCR3: phospholipid scramblase 3; PMP: peptidase, mitochondrial processing; PRDX: peroxiredoxin; PRKN: parkin RBR E3 ubiquitin protein ligase; RPTC: rat proximal tubular cells; ROS: reactive oxygen species; SLC7A11/xCT: solute carrier family 7 member 11; SOD: superoxide dismutase; SOR: superoxide reductase; SQSTM1/p62: sequestosome 1; TCA: tricarboxylic acid; TIMM: translocase of inner mitochondrial membrane; TOMM: translocase of outer mitochondrial membrane; TXN: thioredoxin; VDAC: voltage dependent anion channel; VCP: valosin containing protein.
Collapse
Affiliation(s)
- Lianjiu Su
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan430071, China
- Branch, Center for Cancer Research, National Cancer Institute, National Institutes of HealthNeuro-Oncology, Bethesda, Maryland, USA
| | - Jiahao Zhang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan430071, China
| | - Hernando Gomez
- Center of Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, USA
| | - John A Kellum
- Center of Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, USA
| | - Zhiyong Peng
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan430071, China
- Center of Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, USA
| |
Collapse
|
32
|
The complex pathophysiology of cardiac cachexia: A review of current pathophysiology and implications for clinical practice. Am J Med Sci 2023; 365:9-18. [PMID: 36055378 DOI: 10.1016/j.amjms.2022.08.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 04/09/2022] [Accepted: 08/24/2022] [Indexed: 01/04/2023]
Abstract
Cardiac cachexia is a muscle wasting process that often develops in those with chronic heart failure resulting in weight loss, low levels of physical activity, reduced quality of life, and is associated with a poor prognosis. The pathology of cardiac cachexia is complex with new evidence emerging that implicates several body systems. This review describes the pathophysiology associated with cardiac cachexia and addresses: 1) hormonal changes- neurohormonal abnormalities and metabolic hormone imbalance; 2) mechanisms of muscle wasting in cardiac cachexia, and the integral mechanisms between changed hormones due to cardiac cachexia and muscle wasting processes, and 3) associated abnormalities of gastrointestinal system that contribute to cardiac cachexia. These pleiotropic mechanisms demonstrate the intricate interplay between the affected systems and account for why cardiac cachexia is difficult to manage clinically. This review summarises current pathophysiology of cardiac cachexia and highlights symptoms of cardiac cachexia, implications for clinical practice and research gaps.
Collapse
|
33
|
VanderVeen BN, Cardaci TD, McDonald SJ, Madero SS, Unger CA, Bullard BM, Enos RT, Velázquez KT, Kubinak JL, Fan D, Murphy EA. Obesity reduced survival with 5-fluorouracil and did not protect against chemotherapy-induced cachexia or immune cell cytotoxicity in mice. Cancer Biol Ther 2022; 23:1-15. [PMID: 35968771 PMCID: PMC9377261 DOI: 10.1080/15384047.2022.2108306] [Citation(s) in RCA: 9] [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: 05/16/2022] [Revised: 06/28/2022] [Accepted: 07/24/2022] [Indexed: 01/12/2023] Open
Abstract
Fluorouracil/5-flourouracil (5FU) is a first-line chemotherapy drug for many cancer types; however, its associated toxicities contribute to poor quality of life and reduced dose intensities negatively impacting patient prognosis. While obesity remains a critical risk factor for most cancers, our understanding regarding how obesity may impact chemotherapy's toxicities is extremely limited. C56BL/6 mice were given high fat (Obese) or standard diets (Lean) for 4 months and then subjected to three cycles of 5FU (5d-40 mg/kg Lean Mass, 9d rest) or PBS vehicle control. Shockingly, only 60% of Obese survived 3 cycles compared to 100% of Lean, and Obese lost significantly more body weight. Dihydropyrimidine dehydrogenase (DPD), the enzyme responsible for 5FU catabolism, was reduced in obese livers. Total white blood cells, neutrophils, and lymphocytes were reduced in Obese 5FU compared to Lean 5FU and PBS controls. While adipocyte size was not affected by 5FU in Obese, skeletal muscle mass and myofibrillar cross section area were decreased following 5FU in Lean and Obese. Although adipose tissue inflammatory gene expression was not impacted by 5FU, distinct perturbations to skeletal muscle inflammatory gene expression and immune cell populations (CD45+ Immune cells, CD45+CD11b+CD68+ macrophages and CD45+CD11b+Ly6clo/int macrophage/monocytes) were observed in Obese only. Our evidence suggests that obesity induced liver pathologies and reduced DPD exacerbated 5FU toxicities. While obesity has been suggested to protect against cancer/chemotherapy-induced cachexia and other toxicities, our results demonstrate that obese mice are not protected, but rather show evidence of increased susceptibility to 5FU-induced cytotoxicity even when dosed for relative lean mass.
Collapse
Affiliation(s)
- Brandon N. VanderVeen
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| | - Thomas D. Cardaci
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| | - Sierra J. McDonald
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| | - Sarah S. Madero
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| | - Christian A. Unger
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| | - Brooke M. Bullard
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| | - Reilly T. Enos
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| | - Kandy T. Velázquez
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| | - Jason L. Kubinak
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| | - Daping Fan
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| | - E. Angela Murphy
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| |
Collapse
|
34
|
VanderVeen BN, Cardaci TD, Cunningham P, McDonald SJ, Bullard BM, Fan D, Murphy EA, Velázquez KT. Quercetin Improved Muscle Mass and Mitochondrial Content in a Murine Model of Cancer and Chemotherapy-Induced Cachexia. Nutrients 2022; 15:102. [PMID: 36615760 PMCID: PMC9823918 DOI: 10.3390/nu15010102] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/19/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
A cachexia diagnosis is associated with a doubling in hospital stay and increased healthcare cost for cancer patients and most cachectic patients do not survive treatment. Unfortunately, complexity in treating cachexia is amplified by both the underlying malignancy and the anti-cancer therapy which can independently promote cachexia. Quercetin, an organic polyphenolic flavonoid, has demonstrated anti-inflammatory and antioxidant properties with promise in protecting against cancer and chemotherapy-induced dysfunction; however, whether quercetin is efficacious in maintaining muscle mass in tumor-bearing animals receiving chemotherapy has not been investigated. C26 tumor-bearing mice were given 5-fluorouracil (5FU; 30 mg/kg of lean mass i.p.) concomitant with quercetin (Quer; 50 mg/kg of body weight via oral gavage) or vehicle. Both C26 + 5FU and C26 + 5FU + Quer had similar body weight loss; however, muscle mass and cross-sectional area was greater in C26 + 5FU + Quer compared to C26 + 5FU. Additionally, C26 + 5FU + Quer had a greater number and larger intermyofibrillar mitochondria with increased relative protein expression of mitochondrial complexes V, III, and II as well as cytochrome c expression. C26 + 5FU + Quer also had increased MFN1 and reduced FIS1 relative protein expression without apparent benefits to muscle inflammatory signaling. Our data suggest that quercetin protected against cancer and chemotherapy-induced muscle mass loss through improving mitochondrial homeostatic balance.
Collapse
Affiliation(s)
- Brandon N. VanderVeen
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
- AcePre, LLC, Columbia, SC 29209, USA
| | - Thomas D. Cardaci
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
| | - Patrice Cunningham
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
| | - Sierra J. McDonald
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
| | - Brooke M. Bullard
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
| | - Daping Fan
- AcePre, LLC, Columbia, SC 29209, USA
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
| | - E. Angela Murphy
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
- AcePre, LLC, Columbia, SC 29209, USA
| | - Kandy T. Velázquez
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
| |
Collapse
|
35
|
Murphy BT, Mackrill JJ, O'Halloran KD. Impact of cancer cachexia on respiratory muscle function and the therapeutic potential of exercise. J Physiol 2022; 600:4979-5004. [PMID: 36251564 PMCID: PMC10091733 DOI: 10.1113/jp283569] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/09/2022] [Indexed: 01/05/2023] Open
Abstract
Cancer cachexia is defined as a multi-factorial syndrome characterised by an ongoing loss of skeletal muscle mass and progressive functional impairment, estimated to affect 50-80% of patients and responsible for 20% of cancer deaths. Elevations in the morbidity and mortality rates of cachectic cancer patients has been linked to respiratory failure due to atrophy and dysfunction of the ventilatory muscles. Despite this, there is a distinct scarcity of research investigating the structural and functional condition of the respiratory musculature in cancer, with the majority of studies exclusively focusing on limb muscle. Treatment strategies are largely ineffective in mitigating the cachectic state. It is now widely accepted that an efficacious intervention will likely combine elements of pharmacology, nutrition and exercise. However, of these approaches, exercise has received comparatively little attention. Therefore, it is unlikely to be implemented optimally, whether in isolation or combination. In consideration of these limitations, the current review describes the mechanistic basis of cancer cachexia and subsequently explores the available respiratory- and exercise-focused literature within this context. The molecular basis of cachexia is thoroughly reviewed. The pivotal role of inflammatory mediators is described. Unravelling the mechanisms of exercise-induced support of muscle via antioxidant and anti-inflammatory effects in addition to promoting efficient energy metabolism via increased mitochondrial biogenesis, mitochondrial function and muscle glucose uptake provide avenues for interventional studies. Currently available pre-clinical mouse models including novel transgenic animals provide a platform for the development of multi-modal therapeutic strategies to protect respiratory muscles in people with cancer.
Collapse
Affiliation(s)
- Ben T. Murphy
- Department of PhysiologySchool of MedicineCollege of Medicine and HealthUniversity College CorkCorkIreland
| | - John J. Mackrill
- Department of PhysiologySchool of MedicineCollege of Medicine and HealthUniversity College CorkCorkIreland
| | - Ken D. O'Halloran
- Department of PhysiologySchool of MedicineCollege of Medicine and HealthUniversity College CorkCorkIreland
| |
Collapse
|
36
|
Wijaya YT, Setiawan T, Sari IN, Park K, Lee CH, Cho KW, Lee YK, Lim JY, Yoon JK, Lee SH, Kwon HY. Ginsenoside Rd ameliorates muscle wasting by suppressing the signal transducer and activator of transcription 3 pathway. J Cachexia Sarcopenia Muscle 2022; 13:3149-3162. [PMID: 36127129 PMCID: PMC9745546 DOI: 10.1002/jcsm.13084] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/18/2022] [Accepted: 08/14/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The effects of some drugs, aging, cancers, and other diseases can cause muscle wasting. Currently, there are no effective drugs for treating muscle wasting. In this study, the effects of ginsenoside Rd (GRd) on muscle wasting were studied. METHODS Tumour necrosis factor-alpha (TNF-α)/interferon-gamma (IFN-γ)-induced myotube atrophy in mouse C2C12 and human skeletal myoblasts (HSkM) was evaluated based on cell thickness. Atrophy-related signalling, reactive oxygen species (ROS) level, mitochondrial membrane potential, and mitochondrial number were assessed. GRd (10 mg/kg body weight) was orally administered to aged mice (23-24 months old) and tumour-bearing (Lewis lung carcinoma [LLC1] or CT26) mice for 5 weeks and 16 days, respectively. Body weight, grip strength, inverted hanging time, and muscle weight were assessed. Histological analysis was also performed to assess the effects of GRd. The evolutionary chemical binding similarity (ECBS) approach, molecular docking, Biacore assay, and signal transducer and activator of transcription (STAT) 3 reporter assay were used to identify targets of GRd. RESULTS GRd significantly induced hypertrophy in the C2C12 and HSkM myotubes (average diameter 50.8 ± 2.6% and 49.9% ± 3.7% higher at 100 nM, vs. control, P ≤ 0.001). GRd treatment ameliorated aging- and cancer-induced (LLC1 or CT26) muscle atrophy in mice, which was evidenced by significant increases in grip strength, hanging time, muscle mass, and muscle tissue cross-sectional area (1.3-fold to 4.6-fold, vs. vehicle, P ≤ 0.05; P ≤ 0.01; P ≤ 0.001). STAT3 was found to be a possible target of GRd by the ECBS approach and molecular docking assay. Validation of direct interaction between GRd and STAT3 was confirmed through Biacore analysis. GRd also inhibited STAT3 phosphorylation and STAT3 reporter activity, which led to the inhibition of STAT3 nuclear translocation and the suppression of downstream targets of STAT3, such as atrogin-1, muscle-specific RING finger protein (MuRF-1), and myostatin (MSTN) (29.0 ± 11.2% to 84.3 ± 30.5%, vs. vehicle, P ≤ 0.05; P ≤ 0.01; P ≤ 0.001). Additionally, GRd scavenged ROS (91.7 ± 1.4% reduction at 1 nM, vs. vehicle, P ≤ 0.001), inhibited TNF-α-induced dysregulation of ROS level, and improved mitochondrial integrity (P ≤ 0.05; P ≤ 0.01; P ≤ 0.001). CONCLUSIONS GRd ameliorates aging- and cancer-induced muscle wasting. Our findings suggest that GRd may be a novel therapeutic agent or adjuvant for reversing muscle wasting.
Collapse
Affiliation(s)
- Yoseph Toni Wijaya
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, Republic of Korea
| | - Tania Setiawan
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, Republic of Korea
| | - Ita Novita Sari
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Republic of Korea
| | - Keunwan Park
- Natural Product Informatics Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Chan Hee Lee
- Program of Material Science for Medicine and Pharmaceutics, Department of Biomedical Science, Hallym University, Chuncheon, Republic of Korea
| | - Kae Won Cho
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, Republic of Korea.,Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Republic of Korea
| | - Yun Kyung Lee
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, Republic of Korea.,Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Republic of Korea
| | - Jae-Young Lim
- Institute of Aging, Seoul National University, Seoul, Republic of Korea.,Department of Rehabilitation Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Jeong Kyo Yoon
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, Republic of Korea.,Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Republic of Korea
| | - Sae Hwan Lee
- Liver Clinic, Soonchunhyang University Cheonan Hospital, Cheonan, Republic of Korea
| | - Hyog Young Kwon
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, Republic of Korea.,Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Republic of Korea
| |
Collapse
|
37
|
Jo Y, Yeo MK, Dao T, Kwon J, Yi H, Ryu D. Machine learning-featured Secretogranin V is a circulating diagnostic biomarker for pancreatic adenocarcinomas associated with adipopenia. Front Oncol 2022; 12:942774. [PMID: 36059698 PMCID: PMC9428794 DOI: 10.3389/fonc.2022.942774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
Background Pancreatic cancer is one of the most fatal malignancies of the gastrointestinal cancer, with a challenging early diagnosis due to lack of distinctive symptoms and specific biomarkers. The exact etiology of pancreatic cancer is unknown, making the development of reliable biomarkers difficult. The accumulation of patient-derived omics data along with technological advances in artificial intelligence is giving way to a new era in the discovery of suitable biomarkers. Methods We performed machine learning (ML)-based modeling using four independent transcriptomic datasets, including GSE16515, GSE62165, GSE71729, and the pancreatic adenocarcinoma (PAC) dataset of the Cancer Genome Atlas. To find candidates for circulating biomarkers, we exported expression profiles of 1,703 genes encoding secretory proteins. Integrating three transcriptomic datasets into either a training or test set, ML-based modeling distinguishing PAC from normal was carried out. Another ML-model classifying long-lived and short-lived patients with PAC was also built to select prognosis-associated features. Finally, circulating level of SCG5 in the plasma was determined from the independent cohort (non-tumor = 25 and pancreatic cancer = 25). We also investigated the impact of SCG5 on adipocyte biology using recombinant protein. Results Three distinctive ML-classifiers selected 29-, 64- and 18-featured genes, recognizing the only common gene, SCG5. As per the prediction of ML-models, the SCG5 transcripts was significantly reduced in PAC and decreased further with the progression of the tumor, indicating its potential as a diagnostic as well as prognostic marker for PAC. External validation of SCG5 using plasma samples from patients with PAC confirmed that SCG5 was reduced significantly in patients with PAC when compared to controls. Interestingly, plasma SCG5 levels were correlated with the body mass index and age of donors, implying pancreas-originated SCG5 could regulate energy metabolism systemically. Additionally, analyses using publicly available Genotype-Tissue Expression datasets, including adipose tissue histology and pancreatic SCG5 expression, further validated the association between pancreatic SCG5 expression and the size of subcutaneous adipocytes in humans. However, we could not observe any definite effect of rSCG5 on the cultured adipocyte, in 2D in vitro culture. Conclusion Circulating SCG5, which may be associated with adipopenia, is a promising diagnostic biomarker for PAC.
Collapse
Affiliation(s)
- Yunju Jo
- Department of Molecular Cell Biology, Sungkyunkwan University (SKKU) School of Medicine, Suwon, South Korea
| | - Min-Kyung Yeo
- Department of Pathology, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Tam Dao
- Department of Molecular Cell Biology, Sungkyunkwan University (SKKU) School of Medicine, Suwon, South Korea
| | - Jeongho Kwon
- Department of Molecular Cell Biology, Sungkyunkwan University (SKKU) School of Medicine, Suwon, South Korea
| | - Hyon‐Seung Yi
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea
- Laboratory of Endocrinology and Immune System, Chungnam National University School of Medicine, Daejeon, South Korea
- *Correspondence: Hyon‐Seung Yi, ; Dongryeol Ryu,
| | - Dongryeol Ryu
- Department of Molecular Cell Biology, Sungkyunkwan University (SKKU) School of Medicine, Suwon, South Korea
- *Correspondence: Hyon‐Seung Yi, ; Dongryeol Ryu,
| |
Collapse
|
38
|
Malla J, Zahra A, Venugopal S, Selvamani TY, Shoukrie SI, Selvaraj R, Dhanoa RK, Hamouda RK, Mostafa J. What Role Do Inflammatory Cytokines Play in Cancer Cachexia? Cureus 2022; 14:e26798. [PMID: 35971351 PMCID: PMC9372379 DOI: 10.7759/cureus.26798] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 07/12/2022] [Indexed: 11/05/2022] Open
|
39
|
Rich MM, Housley SN, Nardelli P, Powers RK, Cope TC. Imbalanced Subthreshold Currents Following Sepsis and Chemotherapy: A Shared Mechanism Offering a New Therapeutic Target? Neuroscientist 2022; 28:103-120. [PMID: 33345706 PMCID: PMC8215085 DOI: 10.1177/1073858420981866] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Both sepsis and treatment of cancer with chemotherapy are known to cause neurologic dysfunction. The primary defects seen in both groups of patients are neuropathy and encephalopathy; the underlying mechanisms are poorly understood. Analysis of preclinical models of these disparate conditions reveal similar defects in ion channel function contributing to peripheral neuropathy. The defects in ion channel function extend to the central nervous system where lower motoneurons are affected. In motoneurons the defect involves ion channels responsible for subthreshold currents that convert steady depolarization into repetitive firing. The inability to correctly translate depolarization into steady, repetitive firing has profound effects on motor function, and could be an important contributor to weakness and fatigue experienced by both groups of patients. The possibility that disruption of function, either instead of, or in addition to neurodegeneration, may underlie weakness and fatigue leads to a novel approach to therapy. Activation of serotonin (5HT) receptors in a rat model of sepsis restores the normal balance of subthreshold currents and normal motoneuron firing. If an imbalance of subthreshold currents also occurs in other central nervous system neurons, it could contribute to encephalopathy. We hypothesize that pharmacologically restoring the proper balance of subthreshold currents might provide effective therapy for both neuropathy and encephalopathy in patients recovering from sepsis or treatment with chemotherapy.
Collapse
Affiliation(s)
- Mark M. Rich
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH, USA
| | - Stephen N. Housley
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA,Integrated Cancer Research Center, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Paul Nardelli
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Randall K. Powers
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Timothy C. Cope
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA,Integrated Cancer Research Center, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA,Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
40
|
Pin F, Huot JR, Bonetto A. The Mitochondria-Targeting Agent MitoQ Improves Muscle Atrophy, Weakness and Oxidative Metabolism in C26 Tumor-Bearing Mice. Front Cell Dev Biol 2022; 10:861622. [PMID: 35392166 PMCID: PMC8980422 DOI: 10.3389/fcell.2022.861622] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/07/2022] [Indexed: 01/06/2023] Open
Abstract
Cancer cachexia is a debilitating syndrome characterized by skeletal muscle wasting, weakness and fatigue. Several pathogenetic mechanisms can contribute to these muscle derangements. Mitochondrial alterations, altered metabolism and increased oxidative stress are known to promote muscle weakness and muscle catabolism. To the extent of improving cachexia, several drugs have been tested to stimulate mitochondrial function and normalize the redox balance. The aim of this study was to test the potential beneficial anti-cachectic effects of Mitoquinone Q (MitoQ), one of the most widely-used mitochondria-targeting antioxidant. Here we show that MitoQ administration (25 mg/kg in drinking water, daily) in vivo was able to improve body weight loss in Colon-26 (C26) bearers, without affecting tumor size. Consistently, the C26 hosts displayed ameliorated skeletal muscle and strength upon treatment with MitoQ. In line with improved skeletal muscle mass, the treatment with MitoQ was able to partially correct the expression of the E3 ubiquitin ligases Atrogin-1 and Murf1. Contrarily, the anabolic signaling was not improved by the treatment, as showed by unchanged AKT, mTOR and 4EBP1 phosphorylation. Assessment of gene expression showed altered levels of markers of mitochondrial biogenesis and homeostasis in the tumor hosts, although only Mitofusin-2 levels were significantly affected by the treatment. Interestingly, the levels of Pdk4 and CytB, genes involved in the regulation of mitochondrial function and metabolism, were also partially increased by MitoQ, in line with the modulation of hexokinase (HK), pyruvate dehydrogenase (PDH) and succinate dehydrogenase (SDH) enzymatic activities. The improvement of the oxidative metabolism was associated with reduced myosteatosis (i.e., intramuscular fat infiltration) in the C26 bearers receiving MitoQ, despite unchanged muscle LDL receptor expression, therefore suggesting that MitoQ could boost β-oxidation in the muscle tissue and promote a glycolytic-to-oxidative shift in muscle metabolism and fiber composition. Overall, our data identify MitoQ as an effective treatment to improve skeletal muscle mass and function in tumor hosts and further support studies aimed at testing the anti-cachectic properties of mitochondria-targeting antioxidants also in combination with routinely administered chemotherapy agents.
Collapse
Affiliation(s)
- Fabrizio Pin
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
- Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Joshua R. Huot
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Andrea Bonetto
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
- Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- *Correspondence: Andrea Bonetto,
| |
Collapse
|
41
|
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
|
42
|
Mangano GD, Fouani M, D’Amico D, Di Felice V, Barone R. Cancer-Related Cachexia: The Vicious Circle between Inflammatory Cytokines, Skeletal Muscle, Lipid Metabolism and the Possible Role of Physical Training. Int J Mol Sci 2022; 23:ijms23063004. [PMID: 35328423 PMCID: PMC8949960 DOI: 10.3390/ijms23063004] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/25/2022] [Accepted: 03/09/2022] [Indexed: 12/20/2022] Open
Abstract
Cachexia is a multifactorial and multi-organ syndrome that is a major cause of morbidity and mortality in late-stage chronic diseases. The main clinical features of cancer-related cachexia are chronic inflammation, wasting of skeletal muscle and adipose tissue, insulin resistance, anorexia, and impaired myogenesis. A multimodal treatment has been suggested to approach the multifactorial genesis of cachexia. In this context, physical exercise has been found to have a general effect on maintaining homeostasis in a healthy life, involving multiple organs and their metabolism. The purpose of this review is to present the evidence for the relationship between inflammatory cytokines, skeletal muscle, and fat metabolism and the potential role of exercise training in breaking the vicious circle of this impaired tissue cross-talk. Due to the wide-ranging effects of exercise training, from the body to the behavior and cognition of the individual, it seems to be able to improve the quality of life in this syndrome. Therefore, studying the molecular effects of physical exercise could provide important information about the interactions between organs and the systemic mediators involved in the overall homeostasis of the body.
Collapse
Affiliation(s)
- Giuseppe Donato Mangano
- Correspondence: (G.D.M.); (R.B.); Tel.: +39-09-1238-65823 (G.D.M.); +39-09-1238-65823 (R.B.)
| | | | | | | | - Rosario Barone
- Correspondence: (G.D.M.); (R.B.); Tel.: +39-09-1238-65823 (G.D.M.); +39-09-1238-65823 (R.B.)
| |
Collapse
|
43
|
Allan J, Buss LA, Draper N, Currie MJ. Exercise in People With Cancer: A Spotlight on Energy Regulation and Cachexia. Front Physiol 2022; 13:836804. [PMID: 35283780 PMCID: PMC8914107 DOI: 10.3389/fphys.2022.836804] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/07/2022] [Indexed: 12/26/2022] Open
Abstract
Exercise is increasingly becoming a standard of cancer care, with well-documented benefits for patients including improved mental wellbeing and reduced treatment-related side effects. However, important gaps in knowledge remain about how to optimise exercise prescription for people with cancer. Importantly, it remains unclear how exercise affects the progression of cancer cachexia (a wasting disease stemming from energy imbalance, and a common manifestation of advanced malignant disease), particularly once the condition has already developed. It was recently suggested that the anti-tumour effect of exercise might come from improved energetic capacity. Here, we highlight the possible effect of exercise on energetic capacity and energy regulation in the context of cancer, and how this might affect the progression of cancer cachexia. We suggest that due to the additional energy demand caused by the tumour and associated systemic inflammation, overreaching may occur more easily in people with cancer. Importantly, this could result in impaired anti-tumour immunity and/or the exacerbation of cancer cachexia. This highlights the importance of individualised exercise programs for people with cancer, with special consideration for the regulation of energy balance, ongoing monitoring and possible nutritional supplementation to support the increased energy demand caused by exercise.
Collapse
Affiliation(s)
- Jessica Allan
- School of Health Sciences, Health and Human Development, University of Canterbury, Christchurch, New Zealand
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Linda A. Buss
- Department of Medicine, Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
- *Correspondence: Linda A. Buss,
| | - Nick Draper
- School of Health Sciences, Health and Human Development, University of Canterbury, Christchurch, New Zealand
| | - Margaret J. Currie
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| |
Collapse
|
44
|
Mavropalias G, Sim M, Taaffe DR, Galvão DA, Spry N, Kraemer WJ, Häkkinen K, Newton RU. Exercise medicine for cancer cachexia: targeted exercise to counteract mechanisms and treatment side effects. J Cancer Res Clin Oncol 2022; 148:1389-1406. [PMID: 35088134 PMCID: PMC9114058 DOI: 10.1007/s00432-022-03927-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 01/13/2022] [Indexed: 12/27/2022]
Abstract
Purpose Cancer-induced muscle wasting (i.e., cancer cachexia, CC) is a common and devastating syndrome that results in the death of more than 1 in 5 patients. Although primarily a result of elevated inflammation, there are multiple mechanisms that complement and amplify one another. Research on the use of exercise to manage CC is still limited, while exercise for CC management has been recently discouraged. Moreover, there is a lack of understanding that exercise is not a single medicine, but mode, type, dosage, and timing (exercise prescription) have distinct health outcomes. The purpose of this review was to examine the effects of these modes and subtypes to identify the most optimal form and dosage of exercise therapy specific to each underlying mechanism of CC. Methods The relevant literatures from MEDLINE and Scopus databases were examined. Results Exercise can counteract the most prominent mechanisms and signs of CC including muscle wasting, increased protein turnover, systemic inflammation, reduced appetite and anorexia, increased energy expenditure and fat wasting, insulin resistance, metabolic dysregulation, gut dysbiosis, hypogonadism, impaired oxidative capacity, mitochondrial dysfunction, and cancer treatments side-effects. There are different modes of exercise, and each mode has different sub-types that induce vastly diverse changes when performed over multiple sessions. Choosing suboptimal exercise modes, types, or dosages can be counterproductive and could further contribute to the mechanisms of CC without impacting muscle growth. Conclusion Available evidence shows that patients with CC can safely undertake higher-intensity resistance exercise programs, and benefit from increases in body mass and muscle mass.
Collapse
Affiliation(s)
- Georgios Mavropalias
- Exercise Medicine Research Institute, Edith Cowan University, 270 Joondalup Drive, Joondalup, WA, 6027, Australia.
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia.
| | - Marc Sim
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
- Institute for Nutrition Research, Edith Cowan University, Joondalup, Australia
- Medical School, University of Western Australia, Perth, Australia
| | - Dennis R Taaffe
- Exercise Medicine Research Institute, Edith Cowan University, 270 Joondalup Drive, Joondalup, WA, 6027, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
| | - Daniel A Galvão
- Exercise Medicine Research Institute, Edith Cowan University, 270 Joondalup Drive, Joondalup, WA, 6027, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
| | - Nigel Spry
- Exercise Medicine Research Institute, Edith Cowan University, 270 Joondalup Drive, Joondalup, WA, 6027, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
| | - William J Kraemer
- Exercise Medicine Research Institute, Edith Cowan University, 270 Joondalup Drive, Joondalup, WA, 6027, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
- Department of Human Sciences, Ohio State University, Columbus, USA
| | - Keijo Häkkinen
- Neuromuscular Research Center, Faculty of Sport and Health Sciences, University of Jyväskylä, Jyvaskyla, Finland
| | - Robert U Newton
- Exercise Medicine Research Institute, Edith Cowan University, 270 Joondalup Drive, Joondalup, WA, 6027, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
| |
Collapse
|
45
|
Fairman CM, Lønbro S, Cardaci TD, VanderVeen BN, Nilsen TS, Murphy AE. Muscle wasting in cancer: opportunities and challenges for exercise in clinical cancer trials. JCSM RAPID COMMUNICATIONS 2022; 5:52-67. [PMID: 36118249 PMCID: PMC9481195 DOI: 10.1002/rco2.56] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
BACKGROUND Low muscle in cancer is associated with an increase in treatment-related toxicities and is a predictor of cancer-related and all-cause mortality. The mechanisms of cancer-related muscle loss are multifactorial, including anorexia, hypogonadism, anaemia, inflammation, malnutrition, and aberrations in skeletal muscle protein turnover and metabolism. METHODS In this narrative review, we summarise relevant literature to (i) review the factors influencing skeletal muscle mass regulation, (ii) provide an overview of how cancer/treatments negatively impact these, (iii) review factors beyond muscle signalling that can impact the ability to participate in and respond to an exercise intervention to counteract muscle loss in cancer, and (iv) provide perspectives on critical areas of future research. RESULTS Despite the well-known benefits of exercise, there remains a paucity of clinical evidence supporting the impact of exercise in cancer-related muscle loss. There are numerous challenges to reversing muscle loss with exercise in clinical cancer settings, ranging from the impact of cancer/treatments on the molecular regulation of muscle mass, to clinical challenges in responsiveness to an exercise intervention. For example, tumour-related/treatment-related factors (e.g. nausea, pain, anaemia, and neutropenia), presence of comorbidities (e.g. diabetes, arthritis, and chronic obstructive pulmonary disease), injuries, disease progression and bone metastases, concomitant medications (e.g., metformin), can negatively affect an individual's ability to exercise safely and limit subsequent adaptation. CONCLUSIONS This review identifies numerous gaps and oppportunities in the area of low muscle and muscle loss in cancer. Collaborative efforts between preclinical and clinical researchers are imperative to both understanding the mechanisms of atrophy, and develop appropriate therapeutic interventions.
Collapse
Affiliation(s)
- Ciaran M. Fairman
- Department of Exercise Science, University of South Carolina, Columbia, South Carolina 29033, USA
- Correspondence to: Ciaran Fairman, Department of Exercise Science, University of South Carolina, Columbia, SC 29033, USA.
| | - Simon Lønbro
- Department of Public Health, Section for Sports Science, Aarhus University, Aarhus, Denmark
| | - Thomas D. Cardaci
- Department of Exercise Science, University of South Carolina, Columbia, South Carolina 29033, USA
| | - Brandon N. VanderVeen
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, USA
| | - Tormod S. Nilsen
- Department of Physical Performance, Norwegian School of Sports Sciences, Oslo, Norway
| | - Angela E. Murphy
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, USA
| |
Collapse
|
46
|
Lim S, Deaver JW, Rosa-Caldwell ME, Haynie WS, Morena da Silva F, Cabrera AR, Schrems ER, Saling LW, Jansen LT, Dunlap KR, Wiggs MP, Washington TA, Greene NP. Development of metabolic and contractile alterations in development of cancer cachexia in female tumor-bearing mice. J Appl Physiol (1985) 2022; 132:58-72. [PMID: 34762526 PMCID: PMC8747017 DOI: 10.1152/japplphysiol.00660.2021] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/26/2021] [Accepted: 11/08/2021] [Indexed: 01/03/2023] Open
Abstract
Cancer cachexia (CC) results in impaired muscle function and quality of life and is the primary cause of death for ∼20%-30% of patients with cancer. We demonstrated mitochondrial degeneration as a precursor to CC in male mice; however, whether such alterations occur in females is currently unknown. The purpose of this study was to elucidate muscle alterations in CC development in female tumor-bearing mice. Sixty female C57BL/6J mice were injected with PBS or Lewis lung carcinoma at 8 wk of age, and tumors developed for 1, 2, 3, or 4 wk to assess the time course of cachectic development. In vivo muscle contractile function, protein fractional synthetic rate (FSR), protein turnover, and mitochondrial health were assessed. Three- and four-week tumor-bearing mice displayed a dichotomy in tumor growth and were reassigned to high tumor (HT) and low tumor (LT) groups. HT mice exhibited lower soleus, tibialis anterior, and fat weights than PBS mice. HT mice showed lower peak isometric torque and slower one-half relaxation time than PBS mice. HT mice had lower FSR than PBS mice, whereas E3 ubiquitin ligases were greater in HT than in other groups. Bnip3 (mitophagy) and pMitoTimer red puncta (mitochondrial degeneration) were greater in HT mice, whereas Pgc1α1 and Tfam (mitochondrial biogenesis) were lower in HT mice than in PBS mice. We demonstrate alterations in female tumor-bearing mice where HT exhibited greater protein degradation, impaired muscle contractility, and mitochondrial degeneration compared with other groups. Our data provide novel evidence for a distinct cachectic development in tumor-bearing female mice compared with previous male studies.NEW & NOTEWORTHY Our study demonstrates divergent tumor development and tissue wasting within 3- and 4-wk mice, where approximately half the mice developed large tumors and subsequent cachexia. Unlike previous male studies, where metabolic perturbations precede the onset of cachexia, females appear to exhibit protections from the metabolic perturbations and cachexia development. Our data provide novel evidence for divergent cachectic development in tumor-bearing female mice compared with previous male CC studies, suggesting different mechanisms of CC between sexes.
Collapse
Affiliation(s)
- Seongkyun Lim
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - J William Deaver
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - Megan E Rosa-Caldwell
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - Wesley S Haynie
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - Francielly Morena da Silva
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - Ana Regina Cabrera
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - Eleanor R Schrems
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - Landen W Saling
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - Lisa T Jansen
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - Kirsten R Dunlap
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - Michael P Wiggs
- Mooney Laboratory for Exercise, Nutrition, and Biochemistry, Department of Health, Human Performance and Recreation, Baylor University, Waco, Texas
| | - Tyrone A Washington
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - Nicholas P Greene
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| |
Collapse
|
47
|
The Atrophic Effect of 1,25(OH) 2 Vitamin D 3 (Calcitriol) on C2C12 Myotubes Depends on Oxidative Stress. Antioxidants (Basel) 2021; 10:antiox10121980. [PMID: 34943083 PMCID: PMC8750283 DOI: 10.3390/antiox10121980] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/30/2021] [Accepted: 12/07/2021] [Indexed: 11/17/2022] Open
Abstract
Dysfunctional mitochondrial metabolism has been linked to skeletal muscle loss in several physio-pathological states. Although it has been reported that vitamin D (VD) supports cellular redox homeostasis by maintaining normal mitochondrial functions, and VD deficiency often occurs in conditions associated with skeletal muscle loss, the efficacy of VD supplementation to overcome muscle wasting is debated. Investigations on the direct effects of VD metabolites on skeletal muscle using C2C12 myotubes have revealed an unexpected pro-atrophic activity of calcitriol (1,25VD), while its upstream metabolites cholecalciferol (VD3) and calcidiol (25VD) have anti-atrophic effects. Here, we investigated if the atrophic effects of 1,25VD on myotubes depend on its activity on mitochondrial metabolism. The impact of 1,25VD and its upstream metabolites VD3 and 25VD on mitochondria dynamics and the activity of C2C12 myotubes was evaluated by measuring mitochondrial content, architecture, metabolism, and reactive oxygen species (ROS) production. We found that 1,25VD induces atrophy through protein kinase C (PKC)-mediated ROS production, mainly of extramitochondrial origin. Consistent with this, cotreatment with the antioxidant N-acetylcysteine (NAC), but not with the mitochondria-specific antioxidant mitoTEMPO, was sufficient to blunt the atrophic activity of 1,25VD. In contrast, VD3 and 25VD have antioxidant properties, suggesting that the efficacy of VD supplementation might result from the balance between atrophic pro-oxidant (1,25VD) and protective antioxidant (VD3 and 25VD) metabolites.
Collapse
|
48
|
Watson EL, Baker LA, Wilkinson TJ, Gould DW, Xenophontos S, Graham-Brown M, Major RW, Ashford RU, Viana JL, Smith AC. Inflammation and physical dysfunction: responses to moderate intensity exercise in chronic kidney disease. Nephrol Dial Transplant 2021; 37:860-868. [PMID: 35090033 DOI: 10.1093/ndt/gfab333] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND People with chronic kidney disease (CKD) experience skeletal muscle wasting, reduced levels of physical function and performance, and chronic systemic inflammation. While it is known that a relationship exists between inflammation and muscle wasting, the association between inflammation and physical function or performance in CKD has not been well studied. Exercise has anti-inflammatory effects, but little is known regarding the effect of moderate intensity exercise. This study aimed to (i) compare systemic and intramuscular inflammation between CKD stage G3b-5 and non-CKD controls; (ii) establish whether a relationship exists between physical performance, exercise capacity and inflammation in CKD; (iii) determine changes in systemic and intramuscular inflammation following 12 weeks of exercise; and (iv) investigate whether improving inflammatory status via training contributes to improvements in physical performance and muscle mass. METHODS This is a secondary analysis of previously collected data. CKD patients stages G3b-5 (n = 84, n = 43 males) and non-CKD controls (n = 26, n = 17 males) underwent tests of physical performance, exercise capacity, muscle strength and muscle size. In addition, a subgroup of CKD participants underwent 12 weeks of exercise training, randomized to aerobic (AE, n = 21) or combined (CE, n = 20) training. Plasma and intramuscular inflammation and myostatin were measured at rest and following exercise. RESULTS Tumour necrosis factor-α was negatively associated with lower $^{^{^{.}}}{\rm V}$O2Peak (P = 0.01), Rectus femoris-cross sectional area (P = 0.002) and incremental shuttle walk test performance (P < 0.001). Interleukin-6 was negatively associated with sit-to-stand 60 performances (P = 0.006) and hand grip strength (P = 0.001). Unaccustomed exercise created an intramuscular inflammatory response that was attenuated following 12 weeks of training. Exercise training did not reduce systemic inflammation, but AE training did significantly reduce mature myostatin levels (P = 0.02). Changes in inflammation were not associated with changes in physical performance. CONCLUSIONS Systemic inflammation may contribute to reduced physical function in CKD. Twelve weeks of exercise training was unable to reduce the level of chronic systemic inflammation in these patients, but did reduce plasma myostatin concentrations. Further research is required to further investigate this.
Collapse
Affiliation(s)
- Emma L Watson
- Leicester Kidney Lifestyle Team, University of Leicester, Leicester, UK.,Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Luke A Baker
- Leicester Kidney Lifestyle Team, University of Leicester, Leicester, UK.,Department of Health Sciences, University of Leicester, Leicester, UK
| | - Tom J Wilkinson
- Leicester Kidney Lifestyle Team, University of Leicester, Leicester, UK.,Department of Health Sciences, University of Leicester, Leicester, UK.,Leicester Biomedical Research Centre, Leicester, UK
| | - Doug W Gould
- Leicester Kidney Lifestyle Team, University of Leicester, Leicester, UK
| | - Soteris Xenophontos
- Leicester Kidney Lifestyle Team, University of Leicester, Leicester, UK.,Department of Health Sciences, University of Leicester, Leicester, UK
| | - Matthew Graham-Brown
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK.,John Walls Renal Unit, Leicester General Hospital, Leicester, UK
| | - Rupert W Major
- Department of Health Sciences, University of Leicester, Leicester, UK.,John Walls Renal Unit, Leicester General Hospital, Leicester, UK
| | - Robert U Ashford
- Leicester Orthopaedics, University Hospitals of Leicester, Leicester, UK.,Department of Cancer Studies, University of Leicester, Leicester, UK
| | - Joao L Viana
- Research Center in Sports Sciences, Health Sciences and Human Development, CIDESD, University Institute of Maia, ISMAI, Portugal
| | - Alice C Smith
- Leicester Kidney Lifestyle Team, University of Leicester, Leicester, UK.,Department of Health Sciences, University of Leicester, Leicester, UK.,Leicester Biomedical Research Centre, Leicester, UK
| |
Collapse
|
49
|
Towards Drug Repurposing in Cancer Cachexia: Potential Targets and Candidates. Pharmaceuticals (Basel) 2021; 14:ph14111084. [PMID: 34832866 PMCID: PMC8618795 DOI: 10.3390/ph14111084] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/19/2021] [Accepted: 10/22/2021] [Indexed: 12/11/2022] Open
Abstract
As a multifactorial and multiorgan syndrome, cancer cachexia is associated with decreased tolerance to antitumor treatments and increased morbidity and mortality rates. The current approaches for the treatment of this syndrome are not always effective and well established. Drug repurposing or repositioning consists of the investigation of pharmacological components that are already available or in clinical trials for certain diseases and explores if they can be used for new indications. Its advantages comparing to de novo drugs development are the reduced amount of time spent and costs. In this paper, we selected drugs already available or in clinical trials for non-cachexia indications and that are related to the pathways and molecular components involved in the different phenotypes of cancer cachexia syndrome. Thus, we introduce known drugs as possible candidates for drug repurposing in the treatment of cancer-induced cachexia.
Collapse
|
50
|
Buss LA, Hock B, Merry TL, Ang AD, Robinson BA, Currie MJ, Dachs GU. Effect of immune modulation on the skeletal muscle mitochondrial exercise response: An exploratory study in mice with cancer. PLoS One 2021; 16:e0258831. [PMID: 34665826 PMCID: PMC8525738 DOI: 10.1371/journal.pone.0258831] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 10/06/2021] [Indexed: 11/18/2022] Open
Abstract
Cancer causes mitochondrial alterations in skeletal muscle, which may progress to muscle wasting and, ultimately, to cancer cachexia. Understanding how exercise adaptations are altered by cancer and cancer treatment is important for the effective design of exercise interventions aimed at improving cancer outcomes. We conducted an exploratory study to investigate how tumor burden and cancer immunotherapy treatment (anti-PD-1) modify the skeletal muscle mitochondrial response to exercise training in mice with transplantable tumors (B16-F10 melanoma and EO771 breast cancer). Mice remained sedentary or were provided with running wheels for ~19 days immediately following tumor implant while receiving no treatment (Untreated), isotype control antibody (IgG2a) or anti-PD-1. Exercise and anti-PD-1 did not alter the growth rate of either tumor type, either alone or in combination therapy. Untreated mice with B16-F10 tumors showed increases in most measured markers of skeletal muscle mitochondrial content following exercise training, as did anti-PD-1-treated mice, suggesting increased mitochondrial content following exercise training in these groups. However, mice with B16-F10 tumors receiving the isotype control antibody did not exhibit increased skeletal muscle mitochondrial content following exercise. In untreated mice with EO771 tumors, only citrate synthase activity and complex IV activity were increased following exercise. In contrast, IgG2a and anti-PD-1-treated groups both showed robust increases in most measured markers following exercise. These results indicate that in mice with B16-F10 tumors, IgG2a administration prevents exercise adaptation of skeletal muscle mitochondria, but adaptation remains intact in mice receiving anti-PD-1. In mice with EO771 tumors, both IgG2a and anti-PD-1-treated mice show robust skeletal muscle mitochondrial exercise responses, while untreated mice do not. Taken together, we postulate that immune modulation may enhance skeletal muscle mitochondrial response to exercise in tumor-bearing mice, and suggest this as an exciting new avenue for future research in exercise oncology.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Citrate (si)-Synthase/metabolism
- Electron Transport Complex IV/metabolism
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Immune Checkpoint Inhibitors/administration & dosage
- Immune Checkpoint Inhibitors/pharmacology
- Immunoglobulin G/administration & dosage
- Immunoglobulin G/pharmacology
- Immunotherapy
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/therapy
- Melanoma, Experimental/immunology
- Melanoma, Experimental/metabolism
- Melanoma, Experimental/therapy
- Mice
- Mitochondria, Muscle/metabolism
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Physical Conditioning, Animal/methods
- Random Allocation
- Treatment Outcome
Collapse
Affiliation(s)
- Linda A. Buss
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
- * E-mail:
| | - Barry Hock
- Hematology Research Group, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Troy L. Merry
- Discipline of Nutrition, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Abel D. Ang
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Bridget A. Robinson
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
- Canterbury Regional Cancer and Hematology Service, Canterbury District Health Board, Christchurch, New Zealand
| | - Margaret J. Currie
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Gabi U. Dachs
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| |
Collapse
|