1
|
Reid RM, Turkmen S, Cleveland BM, Biga PR. Direct actions of growth hormone in rainbow trout, Oncorhynchus mykiss, skeletal muscle cells in vitro. Comp Biochem Physiol A Mol Integr Physiol 2024; 297:111725. [PMID: 39122107 DOI: 10.1016/j.cbpa.2024.111725] [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: 06/08/2024] [Revised: 08/06/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
The growth hormone (GH)-insulin-like growth factor-1 (IGF-1) system regulates skeletal muscle growth and function. GH has a major function of targeting the liver to regulate IGF-1 production and release, and IGF-1 mediates the primary anabolic action of GH on growth. However, skeletal muscle is a target tissue of GH as evidenced by dynamic GH receptor expression, but it is unclear if GH elicits any direct actions on extrahepatic tissues as it is difficult to distinguish the effects of IGF-1 from GH. Fish growth regulation is complex compared to mammals, as genome duplication events have resulted in multiple isoforms of GHs, GHRs, IGFs, and IGFRs expressed in most fish tissues. This study investigated the potential for GH direct actions on fish skeletal muscle using an in vitro system, where rainbow trout myogenic precursor cells (MPCs) were cultured in normal and serum-deprived media, to mimic in vivo fasting conditions. Fasting reduces IGF-1 signaling in the muscle, which is critical for disentangling the roles of GH from IGF-1. The direct effects of GH were analyzed by measuring changes in myogenic proliferation and differentiation genes, as well as genes regulating muscle growth and proteolysis. This study provides the first in-depth analysis of the direct actions of GH on serum-deprived fish muscle cells in vitro. Data suggest that GH induces the expression of markers for proliferation and muscle growth in the presence of serum, but all observed GH action was blocked in serum-deprived conditions. Additionally, serum deprivation alone reduced the expression of several proliferation and differentiation markers, while increasing growth and proteolysis markers. Results also demonstrate dynamic gene expression response in the presence of GH and a JAK inhibitor in serum-provided but not serum-deprived conditions. These data provide a better understanding of GH signaling in relation to serum in trout muscle cells in vitro.
Collapse
Affiliation(s)
- Ross M Reid
- Department of Biology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Serhat Turkmen
- Department of Cell Development and Integrative Biology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Beth M Cleveland
- National Center for Cool and Cold Water Aquaculture, Agricultural Research Service (ARS-USDA), Kearneysville, WV 25430, USA
| | - Peggy R Biga
- Department of Biology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
2
|
Dou L, Liu C, Su R, Corazzin M, Jin Z, Yang Z, Hu G, Zhang M, Sun L, Zhao L, Jin Y, Su L. Effects of dietary arginine supplementation on muscle structure, meat characteristics and lipid oxidation products in lambs and its potential mechanisms of action. Meat Sci 2024; 216:109581. [PMID: 38970933 DOI: 10.1016/j.meatsci.2024.109581] [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: 01/22/2024] [Revised: 05/18/2024] [Accepted: 06/25/2024] [Indexed: 07/08/2024]
Abstract
This study aimed to assess the effect of dietary arginine supplementation on muscle structure and meat characteristics of lambs also considering lipid oxidation products and to contribute to reveal its mechanisms of action using tandem mass tagging (TMT) proteomics. Eighteen lambs were allocated to two dietary treatment groups: control diet or control diet with the addition of 1% L-arginine. The results revealed that dietary arginine supplementation increased muscle fibre diameter and cross-sectional area (P < 0.05), which was attributable to protein deposition, as evidenced by increased RNA content, RNA/DNA ratio, inhibition of apoptotic enzyme activity, and alterations in the IGF-1/Akt signaling pathway (P < 0.05). In addition, dietary arginine elevated pH24h, a* values, and IMF content, decreased shear force value and backfat thickness (P < 0.05), as well as decreased the formation of lipid oxidation products involved in meat flavor including hexanal, heptanal, octanal, nonanal and 1-octen-3-ol by increasing the antioxidant capacity of the muscle (P < 0.05). The proteomics results suggested that seven enrichment pathways may be potential mechanisms by which arginine affected the muscle structure and meat characteristics of lambs. In summary, arginine supplementation in lamb diets provides a safe and effective way to improve meat quality, and antioxidant capacity of muscle of lamb.
Collapse
Affiliation(s)
- Lu Dou
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; Integrative Research Base of Beef and Lamb Processing Technology, Ministry of Agriculture and Rural Affairs of the people's Republic of China, Hohhot 010018, China
| | - Chang Liu
- Inner Mongolia Vocational College of Chemical Engineering, Hohhot 010018, China
| | - Rina Su
- Inner Mongolia Vocational College of Chemical Engineering, Hohhot 010018, China
| | - Mirco Corazzin
- Department of Agricultural, Food, Environmental and Animal Sciences, University of Udine, 33100, Udine, Italy
| | - Zhimin Jin
- Inner Mongolia Autonomous Region Administration of Market Supervision Evaluation & Inspection Center, Hohhot 010018, China
| | - Zhihao Yang
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; Integrative Research Base of Beef and Lamb Processing Technology, Ministry of Agriculture and Rural Affairs of the people's Republic of China, Hohhot 010018, China
| | - Guanhua Hu
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; Integrative Research Base of Beef and Lamb Processing Technology, Ministry of Agriculture and Rural Affairs of the people's Republic of China, Hohhot 010018, China
| | - Min Zhang
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; Integrative Research Base of Beef and Lamb Processing Technology, Ministry of Agriculture and Rural Affairs of the people's Republic of China, Hohhot 010018, China
| | - Lina Sun
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; Integrative Research Base of Beef and Lamb Processing Technology, Ministry of Agriculture and Rural Affairs of the people's Republic of China, Hohhot 010018, China
| | - Lihua Zhao
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; Integrative Research Base of Beef and Lamb Processing Technology, Ministry of Agriculture and Rural Affairs of the people's Republic of China, Hohhot 010018, China
| | - Ye Jin
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; Integrative Research Base of Beef and Lamb Processing Technology, Ministry of Agriculture and Rural Affairs of the people's Republic of China, Hohhot 010018, China
| | - Lin Su
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; Integrative Research Base of Beef and Lamb Processing Technology, Ministry of Agriculture and Rural Affairs of the people's Republic of China, Hohhot 010018, China.
| |
Collapse
|
3
|
Park Y, Lee I, Lee MJ, Park H, Jung GS, Kim N, Im W, Kim H, Lee JH, Cho S, Choi YS. Particulate matter exposure induces adverse effects on endometrium and fertility via aberrant inflammatory and apoptotic pathways in vitro and in vivo. CHEMOSPHERE 2024; 361:142466. [PMID: 38810796 DOI: 10.1016/j.chemosphere.2024.142466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/27/2024] [Accepted: 05/26/2024] [Indexed: 05/31/2024]
Abstract
This study aimed to evaluate the adverse effects of particulate matter (PM) exposure on endometrial cells and fertility and to identify possible underlying mechanisms. Thirteen women (aged 15-52 years) were included in this study. Enrolled patients underwent laparoscopic surgery at Gangnam Severance Hospital between 1 January and 31 December 2021. For in vivo experiments, 36 female and nine male C57BL/6 mice were randomly divided into control(vehicle), low-dose(10 mg/kg/d), and high-dose exposure groups(20 mg/kg/d). PM was inhaled nasally for four weeks and natural mating was performed. NIST® SRM® 1648a was used for PM exposure. qRT-PCR, western blotting and Masson's trichrome staining were performed. PM treatment in human endometrial stromal cells induced inflammation with significant upregulation of IL-1β, p-NF-kB, and p-c-Jun compared to those of controls. Additionally, PM treatment significantly increased apoptosis in human endometrial stromal cells by downregulating p-AKT and upregulating p-p53/p53, Cas-3, BAX/Bcl-2, p-AMPK, and p-ERK. After PM treatment, the relative expression of IL-1β, IL-6, TNF-α, p-NF-κB, p-c-Jun, and p-Nrf2/Nrf2 significantly increased in murine endometrium compared to those of the controls. Expression of apoptotic proteins p53, p27, and Cas-3, was also significantly elevated in murine endometrium of the PM exposure group compared to that of the controls. A significant increase in expression of procollagen Ⅰ, and Masson's trichrome staining scores in the murine endometrium was noted after PM treatment. PM treatment significantly decreased ERα expression. After natural mating, all 3 female mice in the control group gave birth to 25 offspring (mean 8.1), whereas in the low-dose PM treatment group, two of three female mice gave birth to nine offspring (mean 4.5). No pregnant mice or offspring was present in the high-dose PM treatment group. PM exposure induces adverse effects on the endometrium through aberrant activation of inflammatory and apoptotic pathways and is associated with detrimental effects on murine fertility.
Collapse
Affiliation(s)
- Yunjeong Park
- Department of Obstetrics and Gynecology, Guro Hospital, Korea University College of Medicine, Seoul, 08308, Republic of Korea
| | - Inha Lee
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 06229, Republic of Korea; Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Min Jung Lee
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 06229, Republic of Korea; Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Hyemin Park
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 06229, Republic of Korea
| | - Gee Soo Jung
- Department of Medical Device Engineering and Management, Yonsei University College of Medicine, Seoul, 06229, Republic of Korea
| | - Nara Kim
- Department of Medical Device Engineering and Management, Yonsei University College of Medicine, Seoul, 06229, Republic of Korea
| | - Wooseok Im
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 06229, Republic of Korea; Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Heeyon Kim
- Department of Obstetrics and Gynecology, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jae Hoon Lee
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 06229, Republic of Korea; Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - SiHyun Cho
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 06229, Republic of Korea; Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| | - Young Sik Choi
- Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea; Department of Obstetrics and Gynecology, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| |
Collapse
|
4
|
Lan XQ, Deng CJ, Wang QQ, Zhao LM, Jiao BW, Xiang Y. The role of TGF-β signaling in muscle atrophy, sarcopenia and cancer cachexia. Gen Comp Endocrinol 2024; 353:114513. [PMID: 38604437 DOI: 10.1016/j.ygcen.2024.114513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/24/2024] [Accepted: 04/03/2024] [Indexed: 04/13/2024]
Abstract
Skeletal muscle, comprising a significant proportion (40 to 50 percent) of total body weight in humans, plays a critical role in maintaining normal physiological conditions. Muscle atrophy occurs when the rate of protein degradation exceeds protein synthesis. Sarcopenia refers to age-related muscle atrophy, while cachexia represents a more complex form of muscle wasting associated with various diseases such as cancer, heart failure, and AIDS. Recent research has highlighted the involvement of signaling pathways, including IGF1-Akt-mTOR, MuRF1-MAFbx, and FOXO, in regulating the delicate balance between muscle protein synthesis and breakdown. Myostatin, a member of the TGF-β superfamily, negatively regulates muscle growth and promotes muscle atrophy by activating Smad2 and Smad3. It also interacts with other signaling pathways in cachexia and sarcopenia. Inhibition of myostatin has emerged as a promising therapeutic approach for sarcopenia and cachexia. Additionally, other TGF-β family members, such as TGF-β1, activin A, and GDF11, have been implicated in the regulation of skeletal muscle mass. Furthermore, myostatin cooperates with these family members to impair muscle differentiation and contribute to muscle loss. This review provides an overview of the significance of myostatin and other TGF-β signaling pathway members in muscular dystrophy, sarcopenia, and cachexia. It also discusses potential novel therapeutic strategies targeting myostatin and TGF-β signaling for the treatment of muscle atrophy.
Collapse
Affiliation(s)
- Xin-Qiang Lan
- Metabolic Control and Aging Group, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Cheng-Jie Deng
- Department of Biochemistry and Molecular Biology, Faculty of Basic Medical Science, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Qi-Quan Wang
- Metabolic Control and Aging Group, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Li-Min Zhao
- Senescence and Cancer Group, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Bao-Wei Jiao
- National Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
| | - Yang Xiang
- Metabolic Control and Aging Group, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, Nanchang 330031, Jiangxi, China.
| |
Collapse
|
5
|
Artigas-Arias M, Curi R, Marzuca-Nassr GN. Myogenic microRNAs as Therapeutic Targets for Skeletal Muscle Mass Wasting in Breast Cancer Models. Int J Mol Sci 2024; 25:6714. [PMID: 38928418 PMCID: PMC11204047 DOI: 10.3390/ijms25126714] [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: 05/02/2024] [Revised: 05/31/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Breast cancer is the type of cancer with the highest prevalence in women worldwide. Skeletal muscle atrophy is an important prognostic factor in women diagnosed with breast cancer. This atrophy stems from disrupted skeletal muscle homeostasis, triggered by diminished anabolic signalling and heightened inflammatory conditions, culminating in an upregulation of skeletal muscle proteolysis gene expression. The importance of delving into research on modulators of skeletal muscle atrophy, such as microRNAs (miRNAs), which play a crucial role in regulating cellular signalling pathways involved in skeletal muscle protein synthesis and degradation, has been recognised. This holds true for conditions of homeostasis as well as pathologies like cancer. However, the determination of specific miRNAs that modulate skeletal muscle atrophy in breast cancer conditions has not yet been explored. In this narrative review, we aim to identify miRNAs that could directly or indirectly influence skeletal muscle atrophy in breast cancer models to gain an updated perspective on potential therapeutic targets that could be modulated through resistance exercise training, aiming to mitigate the loss of skeletal muscle mass in breast cancer patients.
Collapse
Affiliation(s)
- Macarena Artigas-Arias
- Programa de Doctorado en Ciencias Mención Biología Celular y Molecular Aplicada, Universidad de La Frontera, Temuco 4811230, Chile;
| | - Rui Curi
- Interdisciplinary Post-graduate Program in Health Sciences, Universidade Cruzeiro do Sul, São Paulo 01506-000, Brazil;
| | - Gabriel Nasri Marzuca-Nassr
- Departamento de Ciencias de la Rehabilitación, Facultad de Medicina, Universidad de La Frontera, Temuco 4811230, Chile
| |
Collapse
|
6
|
Hawley AL, Baum JI. Nutrition as the foundation for successful aging: a focus on dietary protein and omega-3 polyunsaturated fatty acids. Nutr Rev 2024; 82:389-406. [PMID: 37319363 DOI: 10.1093/nutrit/nuad061] [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: 06/17/2023] Open
Abstract
Skeletal muscle plays a critical role throughout the aging process. People living with sarcopenia, a progressive and generalized loss of skeletal muscle mass and function, often experience diminished quality of life, which can be attributed to a long period of decline and disability. Therefore, it is important to identify modifiable factors that preserve skeletal muscle and promote successful aging (SA). In this review, SA was defined as (1) low cardiometabolic risk, (2) preservation of physical function, and (3) positive state of wellbeing, with nutrition as an integral component. Several studies identify nutrition, specifically high-quality protein (eg, containing all essential amino acids), and long-chain omega-3 polyunsaturated fatty acids (n-3 PUFAs), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA), as positive regulators of SA. Recently, an additive anabolic effect of protein and n-3 PUFAs has been identified in skeletal muscle of older adults. Evidence further suggests that the additive effect of protein and n-3 PUFAs may project beyond skeletal muscle anabolism and promote SA. The key mechanism(s) behind the enhanced effects of intake of protein and n-3 PUFAs needs to be defined. The first objective of this review is to evaluate skeletal muscle as a driver of cardiometabolic health, physical function, and wellbeing to promote SA. The second objective is to examine observational and interventional evidence of protein and n-3 PUFAs on skeletal muscle to promote SA. The final objective is to propose mechanisms by which combined optimal intake of high-quality protein and n-3 PUFAs likely play a key role in SA. Current evidence suggests that increased intake of protein above the Recommended Dietary Allowance and n-3 PUFAs above the Dietary Guidelines for Americans recommendations for late middle-aged and older adults is required to maintain skeletal muscle mass and to promote SA, potentially through the mechanistical target of rapamycin complex 1 (mTORC1).
Collapse
Affiliation(s)
- Aubree L Hawley
- School of Human and Environmental Sciences, University of Arkansas, Fayetteville, AR, USA
| | - Jamie I Baum
- Center for Human Nutrition, Department of Food Science, University of Arkansas System Division of Agriculture, Fayetteville, AR, USA
| |
Collapse
|
7
|
Zou J, Niu K, Lu T, Kan J, Cheng H, Xu L. The Multifunction of TRIM26: From Immune Regulation to Oncology. Protein Pept Lett 2024; 31:424-436. [PMID: 38956921 PMCID: PMC11475100 DOI: 10.2174/0109298665311516240621114519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/24/2024] [Accepted: 05/29/2024] [Indexed: 07/04/2024]
Abstract
Ubiquitination, a crucial post-translational modification, plays a role in nearly all physiological processes. Its functional execution depends on a series of catalytic reactions involving numerous proteases. TRIM26, a protein belonging to the TRIM family, exhibits E3 ubiquitin ligase activity because of its RING structural domain, and is present in diverse cell lineages. Over the last few decades, TRIM26 has been documented to engage in numerous physiological and pathological processes as a controller, demonstrating a diverse array of biological roles. Despite the growing research interest in TRIM26, there has been limited attention given to examining the protein's structure and function in existing reviews. This review begins with a concise overview of the composition and positioning of TRIM26 and then proceeds to examine its roles in immune response, viral invasion, and inflammatory processes. Simultaneously, we demonstrate the contribution of TRIM26 to the progression of various diseases, encompassing numerous malignancies and neurologic conditions. Finally, we have investigated the potential areas for future research on TRIM26.
Collapse
Affiliation(s)
- Jialai Zou
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| | - Kaiyi Niu
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| | - Tao Lu
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| | - Jianxun Kan
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| | - Hao Cheng
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| | - Lijian Xu
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| |
Collapse
|
8
|
Hosoi T, Yakabe M, Hashimoto S, Akishita M, Ogawa S. The roles of sex hormones in the pathophysiology of age-related sarcopenia and frailty. Reprod Med Biol 2024; 23:e12569. [PMID: 38476959 PMCID: PMC10927916 DOI: 10.1002/rmb2.12569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 02/13/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
Background Sarcopenia is an age-related condition characterized by a progressive and systemic decline in skeletal muscle mass, quality, and strength. The incidence of sarcopenia contains sex-specific aspects, indicating the contribution of sex hormones to its pathophysiology. This review focuses on changing trends in sarcopenia, discusses alterations in definitions and diagnostic criteria, and emphasizes the association between sarcopenia and sex hormones. Methods A literature search was performed on PubMed for related articles published between 1997 and December 2023 using appropriate keywords. Main Findings Results Advances in research have emphasized the significance of muscle quality and strength over muscle mass, resulting in new diagnostic criteria for sarcopenia. Androgens demonstrated anabolic effects on skeletal muscles and played a significant role in the pathophysiology of sarcopenia. In clinical settings, androgen replacement therapy has exhibited certain positive outcomes for treating sarcopenia, despite concerns about potential side effects. Conversely, estrogen is involved in skeletal muscle maintenance, but the detailed mechanisms remain unclear. Moreover, results regarding the clinical application of estrogen replacement therapy for treating sarcopenia remained inconsistent. Conclusion The elucidation of molecular mechanisms that involve sex hormones is eagerly awaited for novel therapeutic interventions for sarcopenia.
Collapse
Affiliation(s)
- Tatsuya Hosoi
- Department of Geriatric Medicine, Graduate School of MedicineThe University of TokyoBunkyo‐ku, TokyoJapan
| | - Mitsutaka Yakabe
- Department of Geriatric Medicine, Graduate School of MedicineThe University of TokyoBunkyo‐ku, TokyoJapan
| | - Seiji Hashimoto
- Department of Geriatric Medicine, Graduate School of MedicineThe University of TokyoBunkyo‐ku, TokyoJapan
| | - Masahiro Akishita
- Department of Geriatric Medicine, Graduate School of MedicineThe University of TokyoBunkyo‐ku, TokyoJapan
| | - Sumito Ogawa
- Department of Geriatric Medicine, Graduate School of MedicineThe University of TokyoBunkyo‐ku, TokyoJapan
| |
Collapse
|
9
|
Gellhaus B, Böker KO, Schilling AF, Saul D. Therapeutic Consequences of Targeting the IGF-1/PI3K/AKT/FOXO3 Axis in Sarcopenia: A Narrative Review. Cells 2023; 12:2787. [PMID: 38132107 PMCID: PMC10741475 DOI: 10.3390/cells12242787] [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: 11/06/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
The high prevalence of sarcopenia in an aging population has an underestimated impact on quality of life by increasing the risk of falls and subsequent hospitalization. Unfortunately, the application of the major established key therapeutic-physical activity-is challenging in the immobile and injured sarcopenic patient. Consequently, novel therapeutic directions are needed. The transcription factor Forkhead-Box-Protein O3 (FOXO3) may be an option, as it and its targets have been observed to be more highly expressed in sarcopenic muscle. In such catabolic situations, Foxo3 induces the expression of two muscle specific ubiquitin ligases (Atrogin-1 and Murf-1) via the PI3K/AKT pathway. In this review, we particularly evaluate the potential of Foxo3-targeted gene therapy. Foxo3 knockdown has been shown to lead to increased muscle cross sectional area, through both the AKT-dependent and -independent pathways and the reduced impact on the two major downstream targets Atrogin-1 and Murf-1. Moreover, a Foxo3 reduction suppresses apoptosis, activates satellite cells, and initiates their differentiation into muscle cells. While this indicates a critical role in muscle regeneration, this mechanism might exhaust the stem cell pool, limiting its clinical applicability. As systemic Foxo3 knockdown has also been associated with risks of inflammation and cancer progression, a muscle-specific approach would be necessary. In this review, we summarize the current knowledge on Foxo3 and conceptualize a specific and targeted therapy that may circumvent the drawbacks of systemic Foxo3 knockdown. This approach presumably would limit the side effects and enable an activity-independent positive impact on skeletal muscle.
Collapse
Affiliation(s)
- Benjamin Gellhaus
- Department of Trauma, Orthopedics and Reconstructive Surgery, Georg-August University of Goettingen, 37075 Goettingen, Germany; (B.G.); (K.O.B.); (A.F.S.)
| | - Kai O. Böker
- Department of Trauma, Orthopedics and Reconstructive Surgery, Georg-August University of Goettingen, 37075 Goettingen, Germany; (B.G.); (K.O.B.); (A.F.S.)
| | - Arndt F. Schilling
- Department of Trauma, Orthopedics and Reconstructive Surgery, Georg-August University of Goettingen, 37075 Goettingen, Germany; (B.G.); (K.O.B.); (A.F.S.)
| | - Dominik Saul
- Department of Trauma, Orthopedics and Reconstructive Surgery, Georg-August University of Goettingen, 37075 Goettingen, Germany; (B.G.); (K.O.B.); (A.F.S.)
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72072 Tuebingen, Germany
- Division of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
10
|
Teyssier JR, Cozannet P, Greene E, Dridi S, Rochell SJ. Influence of different heat stress models on nutrient digestibility and markers of stress, inflammation, lipid, and protein metabolism in broilers. Poult Sci 2023; 102:103048. [PMID: 37797358 PMCID: PMC10613759 DOI: 10.1016/j.psj.2023.103048] [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: 05/16/2023] [Revised: 08/03/2023] [Accepted: 08/16/2023] [Indexed: 10/07/2023] Open
Abstract
This experiment determined the effects of different HS models and pair-feeding (PF) on nutrient digestibility and markers of stress, inflammation, and metabolism in broilers. Birds (720 total) were allocated into 12 environmentally controlled chambers and reared under thermoneutral conditions until 20 d. Until 41 d birds were exposed to 4 treatments, including: thermoneutral at 24°C (TN-al), daily cyclic HS (12 h at 24 and 12 h at 35°C; cyHS), constant HS at 35°C (coHS), and PF birds maintained at 24°C and fed to equalize FI with coHS birds (TN-coPF). At d 41, ileal digesta were collected to determine nutrient apparent ileal digestibility (AID). Blood, liver, and breast tissues were collected from 8 birds per treatment to determine the mRNA expression of stress, inflammation, and metabolism markers. An additional 8 TN-al birds were sampled after acute HS exposure at 35°C for 4 h (aHS), and 8 cyHS birds were sampled either right before or 4 h after HS initiation. Data were analyzed by 1-way ANOVA and means were separated using Tukey's HSD test. Compared with TN-al birds, AID of nitrogen and ether extract were reduced in coHS birds, and both cyHS and coHS reduced (P < 0.05) AID of total essential amino acids. TNFα and SOD2 expression were increased (P < 0.05) under aHS, coHS, and TN-coPF conditions. IL6 and HSP70 were increased (P < 0.05) under coHS and aHS, respectively. Expression of lipogenic enzymes ACCα and FASN were reduced by coHS and TN-coPF, while coHS increased the lipolytic enzyme ATGL (P < 0.05). IGF1 was lowered in coHS birds, and p70S6K and MyoG were reduced under coHS and TN-coPF (P < 0.05). Interestingly, MuRF1 and MAFbx were increased (P < 0.05) under coHS only. Overall, these results indicate that coHS has a greater impact on nutrient digestibility and metabolism than aHS and cyHS. Interestingly, increased protein degradation during HS appears to be mostly driven by HS per se and not the reduced FI.
Collapse
Affiliation(s)
- J R Teyssier
- Center of Excellence for Poultry Science, University of Arkansas System Division of Agriculture, Fayetteville, AR 72701, USA
| | - P Cozannet
- Adisseo France S.A.S., Center of Expertise in Research and Nutrition, 03600 Malicorne, France
| | - E Greene
- Center of Excellence for Poultry Science, University of Arkansas System Division of Agriculture, Fayetteville, AR 72701, USA
| | - S Dridi
- Center of Excellence for Poultry Science, University of Arkansas System Division of Agriculture, Fayetteville, AR 72701, USA
| | - S J Rochell
- Department of Poultry Science, Auburn University, Auburn, AL 36849, USA.
| |
Collapse
|
11
|
Wang Z, Liang W, Yan D, Tian H, Dong B, Zhao W, Chang G, Chen G. Identification of genes related to growth traits from transcriptome profiles of duck breast muscle tissue. Anim Biotechnol 2023; 34:1239-1246. [PMID: 34965198 DOI: 10.1080/10495398.2021.2018333] [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: 10/19/2022]
Abstract
The growth and development of duck skeletal muscle is an important economic trait that is genetically regulated. The internal mechanism underlying the regulation of skeletal muscle growth and development in ducks remains unclear. The purpose of this study was to identify candidate genes related to the growth of duck skeletal muscle. RNA-sequencing technology was used to compare the transcriptome of duck breast muscles in an F2 population with the high breast muscle rate (HB) and the low breast muscle rate (LB). A total of 14,522 genes were confirmed to be expressed in the breast muscle, and 173 differentially expressed genes (DEGs) were identified between the HB and LB groups. Functional analysis showed that these DEGs were mainly involved in biological processes and pathways of fat metabolism and muscle growth, especially the FABP3 and MYL4 involved in the PPAR signaling pathway and cardiac muscle contraction pathway. These findings deepened our understanding of the molecular mechanisms involved in muscle growth in ducks and provided a theoretical basis for improving duck production and breeding of ducks.
Collapse
Affiliation(s)
- Zhixiu Wang
- Key Laboratory of Animal Genetics and Breeding and Molecular Design of Jiangsu Province, Yangzhou University, Yangzhou, People's Republic of China
| | - Wenshuang Liang
- Key Laboratory of Animal Genetics and Breeding and Molecular Design of Jiangsu Province, Yangzhou University, Yangzhou, People's Republic of China
| | - Dan Yan
- Key Laboratory of Animal Genetics and Breeding and Molecular Design of Jiangsu Province, Yangzhou University, Yangzhou, People's Republic of China
| | - Huiyue Tian
- Key Laboratory of Animal Genetics and Breeding and Molecular Design of Jiangsu Province, Yangzhou University, Yangzhou, People's Republic of China
| | - Bingqiang Dong
- Key Laboratory of Animal Genetics and Breeding and Molecular Design of Jiangsu Province, Yangzhou University, Yangzhou, People's Republic of China
| | - Wenming Zhao
- Key Laboratory of Animal Genetics and Breeding and Molecular Design of Jiangsu Province, Yangzhou University, Yangzhou, People's Republic of China
| | - Guobin Chang
- Key Laboratory of Animal Genetics and Breeding and Molecular Design of Jiangsu Province, Yangzhou University, Yangzhou, People's Republic of China
| | - Guohong Chen
- Key Laboratory of Animal Genetics and Breeding and Molecular Design of Jiangsu Province, Yangzhou University, Yangzhou, People's Republic of China
| |
Collapse
|
12
|
Gartling G, Nakamura R, Sayce L, Zimmerman Z, Slater A, Wilson A, Bing R, Branski RC, Rousseau B. Acute In Vitro and In Vivo Effects of Dexamethasone in the Vocal Folds: a Pilot Study. Laryngoscope 2023; 133:2264-2270. [PMID: 36317801 PMCID: PMC10149570 DOI: 10.1002/lary.30461] [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: 07/19/2022] [Revised: 09/29/2022] [Accepted: 10/06/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVES/HYPOTHESIS Glucocorticoids (GC)s are commonly employed to treat vocal fold (VF) pathologies. However, VF atrophy has been associated with intracordal GC injections. Dexamethasone-induced skeletal muscle atrophy is well-documented in other tissues and believed to be mediated by increased muscle proteolysis via upregulation of Muscle Ring Finger (MuRF)-1 and Atrogin-1. Mechanisms of dexamethasone-mediated VF atrophy have not been described. This pilot study employed in vitro and in vivo models to investigate the effects of dexamethasone on VF epithelium, thyroarytenoid (TA) muscle, and TA-derived myoblasts. We hypothesized that dexamethasone will increase atrophy-associated gene expression in TA muscle and myoblasts and decrease TA muscle fiber size and epithelial thickness. STUDY DESIGN In vitro, pre-clinical. METHODS TA myoblasts were isolated from a female Sprague-Dawley rat and treated with 1 μM dexamethasone for 24-h. In vivo, 15 New Zealand white rabbits were randomly assigned to three treatment groups: (1) bilateral intracordal injection of 40 μL dexamethasone (10 mg/ml; n = 5), (2) volume-matched saline (n = 5), and (3) untreated controls (n = 5). Larynges were harvested 7-days post-injection. Across in vivo and in vitro experimentation, MuRF-1 and Atrogin-1 mRNA expression were measured via RT-qPCR. TA muscle fiber cross-sectional area (CSA) and epithelial thickness were also quantified in vivo. RESULTS Dexamethasone increased MuRF-1 gene expression in TA myoblasts. Dexamethasone injection, however, did not alter atrophy-associated gene expression, TA CSA, or epithelial thickness in vivo. CONCLUSION Dexamethasone increased atrogene expression in TA myoblasts, providing foundational insight into GC induced atrophic gene transcription. Repeated dexamethasone injections may be required to elicit atrophy in vivo. LEVEL OF EVIDENCE NA Laryngoscope, 133:2264-2270, 2023.
Collapse
Affiliation(s)
- Gary Gartling
- Department of Rehabilitation Medicine, NYU Grossman School of Medicine, New York, NY
- Department of Communication Science and Disorders, School of Health and Rehabilitation Sciences, University of Pittsburgh, Pittsburgh, PA
| | - Ryosuke Nakamura
- Department of Rehabilitation Medicine, NYU Grossman School of Medicine, New York, NY
| | - Lea Sayce
- Department of Communication Science and Disorders, School of Health and Rehabilitation Sciences, University of Pittsburgh, Pittsburgh, PA
| | - Zachary Zimmerman
- Department of Communication Science and Disorders, School of Health and Rehabilitation Sciences, University of Pittsburgh, Pittsburgh, PA
| | - Alysha Slater
- Department of Communication Science and Disorders, School of Health and Rehabilitation Sciences, University of Pittsburgh, Pittsburgh, PA
| | - Azure Wilson
- Department of Communication Science and Disorders, School of Health and Rehabilitation Sciences, University of Pittsburgh, Pittsburgh, PA
| | - Renjie Bing
- Department of Rehabilitation Medicine, NYU Grossman School of Medicine, New York, NY
| | - Ryan C. Branski
- Department of Rehabilitation Medicine, NYU Grossman School of Medicine, New York, NY
- Department of Otolaryngology-Head and Neck Surgery, NYU Grossman School of Medicine, New York, NY
| | - Bernard Rousseau
- Department of Communication Science and Disorders, School of Health and Rehabilitation Sciences, University of Pittsburgh, Pittsburgh, PA
- Doisy College of Health Sciences, Saint Louis University, St. Louis, MO
| |
Collapse
|
13
|
Gellhaus B, Böker KO, Gsaenger M, Rodenwaldt E, Hüser MA, Schilling AF, Saul D. Foxo3 Knockdown Mediates Decline of Myod1 and Myog Reducing Myoblast Conversion to Myotubes. Cells 2023; 12:2167. [PMID: 37681900 PMCID: PMC10486649 DOI: 10.3390/cells12172167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 08/18/2023] [Accepted: 08/24/2023] [Indexed: 09/09/2023] Open
Abstract
Sarcopenia has a high prevalence among the aging population. Sarcopenia is of tremendous socioeconomic importance because it can lead to falls and hospitalization, subsequently increasing healthcare costs while limiting quality of life. In sarcopenic muscle fibers, the E3 ubiquitin ligase F-Box Protein 32 (Fbxo32) is expressed at substantially higher levels, driving ubiquitin-proteasomal muscle protein degradation. As one of the key regulators of muscular equilibrium, the transcription factor Forkhead Box O3 (FOXO3) can increase the expression of Fbxo32, making it a possible target for the regulation of this detrimental pathway. To test this hypothesis, murine C2C12 myoblasts were transduced with AAVs carrying a plasmid for four specific siRNAs against Foxo3. Successfully transduced myoblasts were selected via FACS cell sorting to establish single clone cell lines. Sorted myoblasts were further differentiated into myotubes and stained for myosin heavy chain (MHC) by immunofluorescence. The resulting area was calculated. Myotube contractions were induced by electrical stimulation and quantified. We found an increased Foxo3 expression in satellite cells in human skeletal muscle and an age-related increase in Foxo3 expression in older mice in silico. We established an in vitro AAV-mediated FOXO3 knockdown on protein level. Surprisingly, the myotubes with FOXO3 knockdown displayed a smaller myotube size and a lower number of nuclei per myotube compared to the control myotubes (AAV-transduced with a functionless control plasmid). During differentiation, a lower level of FOXO3 reduced the expression Fbxo32 within the first three days. Moreover, the expression of Myod1 and Myog via ATM and Tp53 was reduced. Functionally, the Foxo3 knockdown myotubes showed a higher contraction duration and time to peak. Early Foxo3 knockdown seems to terminate the initiation of differentiation due to lack of Myod1 expression, and mediates the inhibition of Myog. Subsequently, the myotube size is reduced and the excitability to electrical stimulation is altered.
Collapse
Affiliation(s)
- Benjamin Gellhaus
- Department of Trauma Surgery, Orthopedics and Plastic Surgery, University Medical Center Goettingen, Robert-Koch-Str. 40, 37099 Göttingen, Germany; (B.G.); (K.O.B.); (E.R.); (A.F.S.)
| | - Kai O. Böker
- Department of Trauma Surgery, Orthopedics and Plastic Surgery, University Medical Center Goettingen, Robert-Koch-Str. 40, 37099 Göttingen, Germany; (B.G.); (K.O.B.); (E.R.); (A.F.S.)
| | - Marlene Gsaenger
- Department of Trauma Surgery, Orthopedics and Plastic Surgery, University Medical Center Goettingen, Robert-Koch-Str. 40, 37099 Göttingen, Germany; (B.G.); (K.O.B.); (E.R.); (A.F.S.)
| | - Eyck Rodenwaldt
- Department of Trauma Surgery, Orthopedics and Plastic Surgery, University Medical Center Goettingen, Robert-Koch-Str. 40, 37099 Göttingen, Germany; (B.G.); (K.O.B.); (E.R.); (A.F.S.)
| | - Marc A. Hüser
- Department of Otorhinolaryngology, Head and Neck Surgery, University Medical Center Goettingen, Robert-Koch-Str. 40, 37099 Göttingen, Germany;
| | - Arndt F. Schilling
- Department of Trauma Surgery, Orthopedics and Plastic Surgery, University Medical Center Goettingen, Robert-Koch-Str. 40, 37099 Göttingen, Germany; (B.G.); (K.O.B.); (E.R.); (A.F.S.)
| | - Dominik Saul
- Department of Trauma Surgery, Orthopedics and Plastic Surgery, University Medical Center Goettingen, Robert-Koch-Str. 40, 37099 Göttingen, Germany; (B.G.); (K.O.B.); (E.R.); (A.F.S.)
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tübingen, BG Trauma Center Tübingen, 72076 Tübingen, Germany
- Kogod Center on Aging and Division of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
14
|
Wan Y, Piao L, Xu S, Meng X, Huang Z, Inoue A, Wang H, Yue X, Jin X, Nan Y, Shi GP, Murohara T, Umegaki H, Kuzuya M, Cheng XW. Cathepsin S activity controls chronic stress-induced muscle atrophy and dysfunction in mice. Cell Mol Life Sci 2023; 80:254. [PMID: 37589754 PMCID: PMC10435624 DOI: 10.1007/s00018-023-04888-4] [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: 01/28/2023] [Revised: 06/06/2023] [Accepted: 07/17/2023] [Indexed: 08/18/2023]
Abstract
Exposure to chronic psychological stress (CPS) is an intractable risk factor for inflammatory and metabolic diseases. Lysosomal cysteinyl cathepsins play an important role in human pathobiology. Given that cathepsin S (CTSS) is upregulated in the stressed vascular and adipose tissues, we investigated whether CTSS participates in chronic stress-induced skeletal muscle mass loss and dysfunction, with a special focus on muscle protein metabolic imbalance and apoptosis. Eight-week-old male wildtype (CTSS+/+) and CTSS-knockout (CTSS-/-) mice were randomly assigned to non-stress and variable-stress groups. CTSS+/+ stressed mice showed significant losses of muscle mass, dysfunction, and fiber area, plus significant mitochondrial damage. In this setting, stressed muscle in CTSS+/+ mice presented harmful alterations in the levels of insulin receptor substrate 2 protein content (IRS-2), phospho-phosphatidylinositol 3-kinase, phospho-protein kinase B, and phospho-mammalian target of rapamycin, forkhead box-1, muscle RING-finger protein-1 protein, mitochondrial biogenesis-related peroxisome proliferator-activated receptor-γ coactivator-α, and apoptosis-related B-cell lymphoma 2 and cleaved caspase-3; these alterations were prevented by CTSS deletion. Pharmacological CTSS inhibition mimics its genetic deficiency-mediated muscle benefits. In C2C12 cells, CTSS silencing prevented stressed serum- and oxidative stress-induced IRS-2 protein reduction, loss of the myotube myosin heavy chain content, and apoptosis accompanied by a rectification of investigated molecular harmful changes; these changes were accelerated by CTSS overexpression. These findings demonstrated that CTSS plays a role in IRS-2-related protein anabolism and catabolism and cell apoptosis in stress-induced muscle wasting, suggesting a novel therapeutic strategy for the control of chronic stress-related muscle disease in mice under our experimental conditions by regulating CTSS activity.
Collapse
Affiliation(s)
- Ying Wan
- Department of Cardiology and Hypertension, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
| | - Limei Piao
- Department of Cardiology and Hypertension, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China.
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China.
| | - Shengnan Xu
- Department of Cardiology and Hypertension, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
| | - Xiangkun Meng
- Department of Vascular Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Zhe Huang
- Department of Neurology, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Aiko Inoue
- Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, Aichiken, 4660855, Japan
| | - Hailong Wang
- Department of Cardiology and Hypertension, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
| | - Xueling Yue
- Department of Cardiology and Hypertension, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
| | - Xueying Jin
- Department of Cardiology and Hypertension, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
| | - Yongshan Nan
- Department of Anesthesiology, Yanbian University Hospital, 1327 Juzijie, Yanji, 133000, Jilin, People's Republic of China.
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi-ken, 466-8550, Japan
| | - Hiroyuki Umegaki
- Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, Aichiken, 4660855, Japan
- Department of Community Healthcare and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi-ken, 466-8550, Japan
| | - Masafumi Kuzuya
- Department of Community Healthcare and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi-ken, 466-8550, Japan
- Meitetsu Hospital, Nagoya, Aichi, 451-8511, Japan
| | - Xian Wu Cheng
- Department of Cardiology and Hypertension, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China.
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China.
| |
Collapse
|
15
|
Zhao X, Wang G, Han H, Zhou Y, Feng J, Zhang M. Effects of Atmospheric Ammonia on Skeletal Muscle Growth in Broilers. Animals (Basel) 2023; 13:1926. [PMID: 37370436 DOI: 10.3390/ani13121926] [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: 04/04/2023] [Revised: 05/16/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
Ammonia, one of the most polluted gases in poultry houses, has always been an urgent problem to solve. Exposure to ammonia can threaten the respiratory tract, induce inflammation, and decrease growth performance. To date, there are few studies investigating the effects of ammonia on skeletal muscle growth. In this experiment, a total of 144 broilers were randomly divided into two groups, and 0 ppm and 35 ppm atmospheric ammonia were administered in the chambers. The trial lasted for 21 days. The breast muscle, thigh muscle, dressed weight, and serum biochemical indexes were measured. The skeletal muscle fibre morphology was observed using light microscopy, and the expressions of genes associated with skeletal muscle development and myosin heavy chain genes were assessed. After 7 days of ammonia exposure, the broilers' weight in the ammonia group decreased. On the 21st day of the experiment, in the ammonia group, the breast muscle weight, thigh muscle weight, and dressed weight decreased, the blood urea nitrogen content increased, skeletal muscle fibre diameter shortened, the expression of myostatin increased, and the expression of myosin heavy chain-FWM and myosin heavy chain-FRM decreased significantly. This article suggests that 35 ppm atmospheric ammonia seriously affects the skeletal muscle gain rate of broilers, and the myostatin pathway could be a potential regulation of the growth rate of muscle fibre under ammonia exposure.
Collapse
Affiliation(s)
- Xin Zhao
- State Key Laboratory of Animals Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Guangju Wang
- State Key Laboratory of Animals Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Hongyu Han
- State Key Laboratory of Animals Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Ying Zhou
- State Key Laboratory of Animals Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Jinghai Feng
- State Key Laboratory of Animals Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Minhong Zhang
- State Key Laboratory of Animals Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
16
|
Bersin TV, Cordova KL, Saenger EK, Journey ML, Beckman BR, Lema SC. Nutritional status affects Igf1 regulation of skeletal muscle myogenesis, myostatin, and myofibrillar protein degradation pathways in gopher rockfish (Sebastes carnatus). Mol Cell Endocrinol 2023; 573:111951. [PMID: 37169322 DOI: 10.1016/j.mce.2023.111951] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 05/13/2023]
Abstract
Insulin-like growth factor-1 (Igf1) regulates skeletal muscle growth in fishes by increasing protein synthesis and promoting muscle hypertrophy. When fish experience periods of insufficient food intake, they undergo slower muscle growth or even muscle wasting, and those changes emerge in part from nutritional modulation of Igf1 signaling. Here, we examined how food deprivation (fasting) modulates Igf1 regulation of liver and skeletal muscle gene expression in gopher rockfish (Sebastes carnatus), a nearshore rockfish of importance for commercial and recreational fisheries in the northeastern Pacific Ocean, to understand how food limitation impacts Igf regulation of muscle growth pathways. Rockfish were either fed or fasted for 14 d, after which a subset of fish from each group was treated with recombinant Igf1 from sea bream (Sparus aurata). Fish that were fasted lost body mass and had lower body condition, reduced hepatosomatic index, and lower plasma Igf1 concentrations, as well as a decreased abundance of igf1 gene transcripts in the liver, increased hepatic mRNAs for Igf binding proteins igfbp1a, igfbp1b, and igfbp3a, and decreased mRNA abundances for igfbp2b and a putative Igf acid labile subunit (igfals) gene. In skeletal muscle, fasted fish showed a reduced abundance of intramuscular igf1 mRNAs but elevated gene transcripts encoding Igf1 receptors A (igf1ra) and B (igf1rb), which also showed downregulation by Igf1. Fasting increased skeletal muscle mRNAs for myogenin and myostatin1, as well as ubiquitin ligase F-box only protein 32 (fbxo32) and muscle RING-finger protein-1 (murf1) genes involved in muscle atrophy, while concurrently downregulating mRNAs for myoblast determination protein 2 (myod2), myostatin2, and myogenic factors 5 (myf5) and 6 (myf6 encoding Mrf4). Treatment with Igf1 downregulated muscle myostatin1 and fbxo32 under both feeding conditions, but showed feeding-dependent effects on murf1, myf5, and myf6/Mrf4 gene expression indicating that Igf1 effects on muscle growth and atrophy pathways is contingent on recent food consumption experience.
Collapse
Affiliation(s)
- Theresa V Bersin
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University, San Luis Obispo, CA, 93407, USA
| | - Kasey L Cordova
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University, San Luis Obispo, CA, 93407, USA
| | - E Kate Saenger
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University, San Luis Obispo, CA, 93407, USA
| | - Meredith L Journey
- Lynker Technology, 202 Church St SE #536, Leesburg, VA, 20175, USA; Under Contract to Environmental and Fisheries Sciences Division, Northwest Fisheries Science Center, National Marine Fisheries Service, National Oceanic and Atmospheric Administration, Seattle, WA, 98112, USA
| | - Brian R Beckman
- Environmental and Fisheries Sciences Division, Northwest Fisheries Science Center, National Marine Fisheries Service, National Oceanic and Atmospheric Administration, Seattle, WA, 98112, USA
| | - Sean C Lema
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University, San Luis Obispo, CA, 93407, USA.
| |
Collapse
|
17
|
Rutin Prevents Dexamethasone-Induced Muscle Loss in C2C12 Myotube and Mouse Model by Controlling FOXO3-Dependent Signaling. Antioxidants (Basel) 2023; 12:antiox12030639. [PMID: 36978887 PMCID: PMC10045290 DOI: 10.3390/antiox12030639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 02/22/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
One of the causes of sarcopenia is that homeostasis between anabolism and catabolism breaks down due to muscle metabolism changes. Rutin has shown antioxidant and anti-inflammatory effects in various diseases, but there are few studies on the effect on muscle loss with aging. The effect of rutin on muscle loss was evaluated using dexamethasone-induced muscle loss C2C12 myoblast and mouse model. In the group treated with dexamethasone, the muscle weight of gastrocnemius (GA), tibialis anterior (TA), and extensor digitorum longus (EDL) in the mouse model were significantly decreased (p < 0.0001 in GA, p < 0.0001 in TA, and p < 0.001 in EDL) but recovered (p < 0.01 in GA, p < 0.0001 in TA, and p < 0.01 in EDL) when treated with rutin. MAFbx, MuRF1, and FOXO3 protein expression of C2C12 myoblast were significantly increased (p < 0.01 in MAFbx, p < 0.01 in MuRF1, and p < 0.01 in FOXO3) when treated with dexamethasone, but it was recovered (p < 0.01 in MAFbx, p < 0.01 in MuRF1, and p < 0.01 in FOXO3) when rutin was treated. In addition, MAFbx and FOXO3 protein expression in GA of mouse model was significantly increased (p < 0.0001 in MAFbx and p < 0.001 in FOXO3) when treated with dexamethasone, but it was also recovered (p < 0.01 in MAFbx and p < 0.001 in FOXO3) when rutin was treated. The present study shows that rutin blocks the FOXO3/MAFbx and FOXO3/MuRf1 pathways to prevent protein catabolism. Therefore, rutin could be a potential agent for muscle loss such as sarcopenia through the blocking ubiquitin-proteasome pathway associated with catabolic protein degradation.
Collapse
|
18
|
Maier MC, Nankervis S, Wallace ME, Develyn T, Myers MA. Dexamethasone leads to Zn 2+ accumulation and increased unbound Zn 2+ in C2C12 muscle and 3T3-L1 adipose cells. J Cell Biochem 2023; 124:409-420. [PMID: 36716229 DOI: 10.1002/jcb.30376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 01/08/2023] [Accepted: 01/15/2023] [Indexed: 01/31/2023]
Abstract
Skeletal muscle atrophy is associated with increases in circulating glucocorticoid levels and insulin resistance. Zinc accumulates in atrophic muscle, but the relationship between atrophy, insulin resistance, and Zn2+ homeostasis remains unclear. In this study, the effect of the glucocorticoid dexamethasone (DEX) on insulin and Zn2+ homeostasis was explored. Treatment of differentiated C2C12 skeletal myotubes and 3T3-L1 adipocytes with DEX significantly increased mRNA expression of the metal-binding proteins Mt1 and 2 and altered energy storage as shown by the increased size of lipid droplets in 3T3-L1 cells. In C2C12 cells the total cellular Zn2+ was higher after DEX treatment, and in both C2C12 and 3T3-L1 adipocytes, free unbound Zn2+ was increased. Insulin treatment led to a gradual increase in free Zn2+ in C2C12 cells, and no significant change in DEX-treated cells such that concentrations were similar 10 min after insulin treatment. These data demonstrate that DEX disturbs Zn2+ homeostasis in muscle and fat cells. Further study of the molecular pathways involved to identify novel therapeutic targets for treatment of skeletal muscle atrophy is warranted.
Collapse
Affiliation(s)
- Michelle C Maier
- Health Innovation and Transformation Centre, Federation University Australia, Mt Helen, Victoria, Australia
| | - Scott Nankervis
- Biomedical Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Mt Helen, Victoria, Australia
| | - Morgan E Wallace
- Biomedical Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Mt Helen, Victoria, Australia
| | - Tamekha Develyn
- Fiona Elsey Cancer Research Centre, Ballarat, Victoria, Australia
| | - Mark A Myers
- Health Innovation and Transformation Centre, Federation University Australia, Mt Helen, Victoria, Australia
- Biomedical Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Mt Helen, Victoria, Australia
| |
Collapse
|
19
|
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: 9.0] [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
|
20
|
Lee SM, Jeong EG, Jeong YI, Rha SH, Kim SE, An WS. Omega-3 fatty acid and menaquinone-7 combination are helpful for aortic calcification prevention, reducing osteoclast area of bone and Fox0 expression of muscle in uremic rats. Ren Fail 2022; 44:1873-1885. [PMID: 36632744 PMCID: PMC9848285 DOI: 10.1080/0886022x.2022.2142140] [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] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Osteopenia, sarcopenia, and vascular calcification (VC) are prevalent in patients with chronic kidney disease and often coexist. In the absence of proven therapies, it is necessary to develop therapeutic or preventive nutrients supplementation for osteopenia, sarcopenia, and VC. The present study investigated the effect of omega-3 fatty acid (FA) and menaquinone-7 (MK-7) on osteopenia, sarcopenia, and VC in adenine and low-protein diet-induced uremic rats. METHODS Thirty-two male Sprague-Dawley rats were fed diets containing 0.75% adenine and 2.5% protein for three weeks. Rats were randomly divided into four groups that were fed diets containing 2.5% protein for four weeks: adenine control (0.9% saline), omega-3 FA (300 mg/kg/day), MK-7 (50 µg/kg/day), and omega-3 FA/MK-7. Von Kossa staining for aortic calcification assessment was performed. Osteoclast surface/bone surface ratio (OcS/BS) of bone and muscle fiber were analyzed using hematoxylin and eosin staining. Osteoprotegerin (OPG) immunohistochemical staining was done in the aorta and bone. Molecules related with sarcopenia were analyzed using western blotting. RESULTS Compared to the normal control, OcS/BS and aortic calcification, and OPG staining in the aorta and bone were significantly increased in the adenine controls. OPG staining and aortic calcification progressed the least in the group supplemented with both omega-3 FA/MK-7. In the adenine controls, the regular arrangement of muscle fiber was severely disrupted, and inflammatory cell infiltration was more prominent. These findings were reduced after combined supplementation with omega-3 FA/MK-7. Furthermore, decreased mammalian target of rapamycin and increased Forkhead box protein 1 expression was significantly restored by combined supplementation. CONCLUSIONS Combined nutrients supplementation with omega-3 FA and MK-7 may be helpful for aortic VC prevention, reducing osteoclast activation and improving sarcopenia-related molecules in adenine and low-protein diet induced uremic rats.
Collapse
Affiliation(s)
- Su Mi Lee
- Department of Internal Medicine, College of Medicine, Dong-A University, Busan, Republic of Korea
| | - Eu Gene Jeong
- Department of Internal Medicine, College of Medicine, Dong-A University, Busan, Republic of Korea
| | - Yu In Jeong
- Department of Internal Medicine, College of Medicine, Dong-A University, Busan, Republic of Korea
| | - Seo Hee Rha
- Department of Pathology, College of Medicine, Dong-A University, Busan, Republic of Korea
| | - Seong Eun Kim
- Department of Internal Medicine, College of Medicine, Dong-A University, Busan, Republic of Korea
| | - Won Suk An
- Department of Internal Medicine, College of Medicine, Dong-A University, Busan, Republic of Korea,CONTACT Won Suk An Department of Internal Medicine, Dong-A University, 3Ga-1, Dongdaesin-Dong, Seo-Gu, Busan, 602-715, Republic of Korea
| |
Collapse
|
21
|
Comparative Transcriptomic Analysis of mRNAs, miRNAs and lncRNAs in the Longissimus dorsi Muscles between Fat-Type and Lean-Type Pigs. Biomolecules 2022; 12:biom12091294. [PMID: 36139132 PMCID: PMC9496231 DOI: 10.3390/biom12091294] [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: 06/26/2022] [Revised: 09/05/2022] [Accepted: 09/09/2022] [Indexed: 11/24/2022] Open
Abstract
In pigs, meat quality and production are two important traits affecting the pig industry and human health. Compared to lean-type pigs, fat-type pigs contain higher intramuscular fat (IMF) contents, better taste and nutritional value. To uncover genetic factors controlling differences related to IMF in pig muscle, we performed RNA-seq analysis on the transcriptomes of the Longissimus dorsi (LD) muscle of Laiwu pigs (LW, fat-type pigs) and commercial Duroc × Landrace × Yorkshire pigs (DLY, lean-type pigs) at 150 d to compare the expression profiles of mRNA, miRNA and lncRNA. A total of 225 mRNAs, 12 miRNAs and 57 lncRNAs were found to be differentially expressed at the criteria of |log2(foldchange)| > 1 and q < 0.05. The mRNA expression of LDHB was significantly higher in the LD muscle of LW compared to DLY pigs with log2(foldchange) being 9.66. Using protein interaction prediction method, we identified more interactions of estrogen-related receptor alpha (ESRRA) associated with upregulated mRNAs, whereas versican (VCAN) and proenkephalin (PENK) were associated with downregulated mRNAs in LW pigs. Integrated analysis on differentially expressed (DE) mRNAs and miRNAs in the LD muscle between LW and DLY pigs revealed two network modules: between five upregulated mRNA genes (GALNT15, FKBP5, PPARGC1A, LOC110258214 and LOC110258215) and six downregulated miRNA genes (ssc-let-7a, ssc-miR190-3p, ssc-miR356-5p, ssc-miR573-5p, ssc-miR204-5p and ssc-miR-10383), and between three downregulated DE mRNA genes (IFRD1, LOC110258600 and LOC102158401) and six upregulated DE miRNA genes (ssc-miR1379-3p, ssc-miR1379-5p, ssc-miR397-5p, ssc-miR1358-5p, ssc-miR299-5p and ssc-miR1156-5p) in LW pigs. Based on the mRNA and ncRNA binding site targeting database, we constructed a regulatory network with miRNA as the center and mRNA and lncRNA as the target genes, including GALNT15/ssc-let-7a/LOC100523888, IFRD1/ssc-miR1379-5p/CD99, etc., forming a ceRNA network in the LD muscles that are differentially expressed between LW and DLY pigs. Collectively, these data may provide resources for further investigation of molecular mechanisms underlying differences in meat traits between lean- and fat-type pigs.
Collapse
|
22
|
Transforming Growth Factor-Beta Signaling in Cancer-Induced Cachexia: From Molecular Pathways to the Clinics. Cells 2022; 11:cells11172671. [PMID: 36078078 PMCID: PMC9454487 DOI: 10.3390/cells11172671] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 02/06/2023] Open
Abstract
Cachexia is a metabolic syndrome consisting of massive loss of muscle mass and function that has a severe impact on the quality of life and survival of cancer patients. Up to 20% of lung cancer patients and up to 80% of pancreatic cancer patients are diagnosed with cachexia, leading to death in 20% of them. The main drivers of cachexia are cytokines such as interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), macrophage inhibitory cytokine 1 (MIC-1/GDF15) and transforming growth factor-beta (TGF-β). Besides its double-edged role as a tumor suppressor and activator, TGF-β causes muscle loss through myostatin-based signaling, involved in the reduction in protein synthesis and enhanced protein degradation. Additionally, TGF-β induces inhibin and activin, causing weight loss and muscle depletion, while MIC-1/GDF15, a member of the TGF-β superfamily, leads to anorexia and so, indirectly, to muscle wasting, acting on the hypothalamus center. Against this background, the blockade of TGF-β is tested as a potential mechanism to revert cachexia, and antibodies against TGF-β reduced weight and muscle loss in murine models of pancreatic cancer. This article reviews the role of the TGF-β pathway and to a minor extent of other molecules including microRNA in cancer onset and progression with a special focus on their involvement in cachexia, to enlighten whether TGF-β and such other players could be potential targets for therapy.
Collapse
|
23
|
Elbialy ZI, Gamal S, Al-Hawary II, Shukry M, Salah AS, Aboshosha AA, Assar DH. Exploring the impacts of different fasting and refeeding regimes on Nile tilapia (Oreochromis niloticus L.): growth performance, histopathological study, and expression levels of some muscle growth-related genes. FISH PHYSIOLOGY AND BIOCHEMISTRY 2022; 48:973-989. [PMID: 35781858 PMCID: PMC9385825 DOI: 10.1007/s10695-022-01094-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/24/2022] [Indexed: 06/15/2023]
Abstract
The current study investigated how different fasting and refeeding regimes would impact Nile tilapia growth performance, histopathological examination, and gene expression of myostatin, myogenin, GH, IGF-1, and NPYa. Nile tilapia fish (n = 120) were randomly allocated into four groups, including the control group fed on a basal diet for 6 weeks (F6), group A starved for 1 week and then refed for 5 weeks (S1F5), group B starved for 2 weeks and then refed for 4 weeks (S2F4), while group C starved for 4 weeks and then refed for 2 weeks (S4F2). Fasting provoked a decrease in body weight coincided with more extended starvation periods. Also, it induced muscle and liver histological alterations; the severity was correlated with the length of fasting periods. Gene expression levels of GH, MSTN, MYOG, and NPYa were significantly increased, while IGF1 was markedly depressed in fasted fish compared to the control group. Interestingly, refeeding after well-planned short fasting period (S1F5) modulated the histopathological alterations. To some extent, these changes were restored after refeeding. Restored IGF-I and opposing fasting expression profiles of the genes mentioned above thus recovered weights almost like the control group and achieved satisfactory growth compensation. Conversely, refeeding following more extended fasting periods failed to restore body weight. In conclusion, refeeding after fasting can induce a compensatory response. Still, the restoration capacity is dependent on the length of fasting and refeeding periods through exhibiting differential morphological structure and expressions pattern for muscle and growth-related genes.
Collapse
Affiliation(s)
- Zizy I. Elbialy
- Fish Processing and Biotechnology Department, Faculty of Aquatic and Fisheries Sciences, Kafrelsheikh University, Kafrelsheikh, 33516 Egypt
| | - Shrouk Gamal
- Fish Processing and Biotechnology Department, Faculty of Aquatic and Fisheries Sciences, Kafrelsheikh University, Kafrelsheikh, 33516 Egypt
| | - Ibrahim I. Al-Hawary
- Fish Processing and Biotechnology Department, Faculty of Aquatic and Fisheries Sciences, Kafrelsheikh University, Kafrelsheikh, 33516 Egypt
| | - Mustafa Shukry
- Department of Physiology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, 33516 Egypt
| | - Abdallah S. Salah
- Department of Aquaculture, Faculty of Aquatic and Fisheries Sciences, Kafrelsheikh University, Kafrelsheikh, 33516 Egypt
- Institute of Aquaculture, Faculty of Natural Sciences, University of Stirling, Stirling, FK9 4LA UK
| | - Ali A. Aboshosha
- Department of Genetics, Faculty of Agriculture, Kafrelsheikh University, Kafrelsheikh, 33516 Egypt
| | - Doaa H. Assar
- Clinical Pathology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, 33516 Egypt
| |
Collapse
|
24
|
β-Sitosterol Attenuates Dexamethasone-Induced Muscle Atrophy via Regulating FoxO1-Dependent Signaling in C2C12 Cell and Mice Model. Nutrients 2022; 14:nu14142894. [PMID: 35889851 PMCID: PMC9315776 DOI: 10.3390/nu14142894] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/24/2022] [Accepted: 07/04/2022] [Indexed: 12/10/2022] Open
Abstract
Sarcopenia refers to a decline in muscle mass and strength with age, causing significant impairment in the ability to carry out normal daily functions and increased risk of falls and fractures, eventually leading to loss of independence. Maintaining protein homeostasis is an important factor in preventing muscle loss, and the decrease in muscle mass is caused by an imbalance between anabolism and catabolism of muscle proteins. Although β-sitosterol has various effects such as anti-inflammatory, protective effect against nonalcoholic fatty liver disease (NAFLD), antioxidant, and antidiabetic activity, the mechanism of β-sitosterol effect on the catabolic pathway was not well known. β-sitosterol was assessed in vitro and in vivo using a dexamethasone-induced muscle atrophy mice model and C2C12 myoblasts. β-sitosterol protected mice from dexamethasone-induced muscle mass loss. The thickness of gastrocnemius muscle myofibers was increased in dexamethasone with the β-sitosterol treatment group (DS). Grip strength and creatine kinase (CK) activity were also recovered when β-sitosterol was treated. The muscle loss inhibitory efficacy of β-sitosterol in dexamethasone-induced muscle atrophy in C2C12 myotube was also verified in C2C12 myoblast. β-sitosterol also recovered the width of myotubes. The protein expression of muscle atrophy F-box (MAFbx) was increased in dexamethasone-treated animal models and C2C12 myoblast, but it was reduced when β-sitosterol was treated. MuRF1 also showed similar results to MAFbx in the mRNA level of C2C12 myotubes. In addition, in the gastrocnemius and tibialis anterior muscles of mouse models, Forkhead Box O1 (FoxO1) protein was increased in the dexamethasone-treated group (Dexa) compared with the control group and reduced in the DS group. Therefore, β-sitosterol would be a potential treatment agent for aging sarcopenia.
Collapse
|
25
|
Non-alcoholic fatty liver disease-related fibrosis and sarcopenia: An altered liver-muscle crosstalk leading to increased mortality risk. Ageing Res Rev 2022; 80:101696. [PMID: 35843589 DOI: 10.1016/j.arr.2022.101696] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/11/2022] [Accepted: 07/13/2022] [Indexed: 11/22/2022]
Abstract
In the last few decades, the loss of skeletal muscle mass and function, known as sarcopenia, has significantly increased in prevalence, becoming a major global public health concern. On the other hand, the prevalence of non-alcoholic fatty liver disease (NAFLD) has also reached pandemic proportions, constituting the leading cause of hepatic fibrosis worldwide. Remarkably, while sarcopenia and NAFLD-related fibrosis are independently associated with all-cause mortality, the combination of both conditions entails a greater risk for all-cause and cardiac-specific mortality. Interestingly, both sarcopenia and NAFLD-related fibrosis share common pathophysiological pathways, including insulin resistance, chronic inflammation, hyperammonemia, alterations in the regulation of myokines, sex hormones and growth hormone/insulin-like growth factor-1 signaling, which may explain reciprocal connections between these two disorders. Additional contributing factors, such as the gut microbiome, may also play a role in this relationship. In skeletal muscle, phosphatidylinositol 3-kinase/Akt and myostatin signaling are the central anabolic and catabolic pathways, respectively, and the imbalance between them can lead to muscle wasting in patients with NAFLD-related fibrosis. In this review, we summarize the bidirectional influence between NAFLD-related fibrosis and sarcopenia, highlighting the main potential mechanisms involved in this complex crosstalk, and we discuss the synergistic effects of both conditions in overall and cardiovascular mortality.
Collapse
|
26
|
Prevention of Loss of Muscle Mass and Function in Older Adults during COVID-19 Lockdown: Potential Role of Dietary Essential Amino Acids. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19138090. [PMID: 35805748 PMCID: PMC9265941 DOI: 10.3390/ijerph19138090] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 02/04/2023]
Abstract
As the COVID-19 pandemic became a global emergency, social distancing, quarantine, and limitations in outdoor activities have resulted in an environment of enforced physical inactivity (EPI). A prolonged period of EPI in older individuals accelerates the deterioration of skeletal muscle health, including loss of muscle mass and function, commonly referred to as sarcopenia. Sarcopenia is associated with an increased likelihood of the progression of diabetes, obesity, and/or depression. Well-known approaches to mitigate the symptoms of sarcopenia include participation in resistance exercise training and/or intake of balanced essential amino acids (EAAs) and high-quality (i.e., containing high EEAs) protein. As the pandemic situation discourages physical exercise, nutritional approaches, especially dietary EAA intake, could be a good alternative for counteracting against EPI-promoted loss of muscle mass and function. Therefore, in the present review, we cover (1) the impact of EPI-induced muscle loss and function on health, (2) the therapeutic potential of dietary EAAs for muscle health (e.g., muscle mass and function) in the EPI condition in comparison with protein sources, and finally (3) practical guidelines of dietary EAA intake for optimal anabolic response in EPI.
Collapse
|
27
|
Ho JQ, Abramowitz MK. Clinical Consequences of Metabolic Acidosis-Muscle. Adv Chronic Kidney Dis 2022; 29:395-405. [PMID: 36175077 DOI: 10.1053/j.ackd.2022.04.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 04/10/2022] [Accepted: 04/25/2022] [Indexed: 01/25/2023]
Abstract
Metabolic acidosis is common in people with chronic kidney disease and can contribute to functional decline, morbidity, and mortality. One avenue through which metabolic acidosis can result in these adverse clinical outcomes is by negatively impacting skeletal muscle; this can occur through several pathways. First, metabolic acidosis promotes protein degradation and impairs protein synthesis, which lead to muscle breakdown. Second, metabolic acidosis hinders mitochondrial function, which decreases oxidative phosphorylation and reduces energy production. Third, metabolic acidosis directly limits muscle contraction. The purpose of this review is to examine the specific mechanisms of each pathway through which metabolic acidosis affects muscle, the impact of metabolic acidosis on physical function, and the effect of treating metabolic acidosis on functional outcomes.
Collapse
Affiliation(s)
- Jim Q Ho
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Matthew K Abramowitz
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY; Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY; Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY; Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY.
| |
Collapse
|
28
|
Abstract
Sarcopenia is common in aging and in patients with heart failure (HF) who may experience worse outcomes. Patients with muscle wasting are more likely to experience falls and can have serious complications when undergoing cardiac procedures. While intensive nutritional support and exercise rehabilitation can help reverse some of these changes, they are often under-prescribed in a timely manner, and we have limited insights into who would benefit. Mechanistic links between gut microbial metabolites (GMM) have been identified and may contribute to adverse clinical outcomes in patients with cardio-renal diseases and aging. This review will examine the emerging evidence for the influence of the gut microbiome-derived metabolites and notable signaling pathways involved in both sarcopenia and HF, especially those linked to dietary intake and mitochondrial metabolism. This provides a unique opportunity to gain mechanistic and clinical insights into developing novel therapeutic strategies that target these GMM pathways or through tailored nutritional modulation to prevent progressive muscle wasting in elderly patients with heart failure.
Collapse
Affiliation(s)
- Chia-Feng Liu
- Center for Microbiome and Human Health, Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland, OH 44195, USA
| | - W H Wilson Tang
- Center for Microbiome and Human Health, Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland, OH 44195, USA.,Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
29
|
Saneyasu T, Ogasawara K, Fujiwara Y, Honda K, Kamisoyama H. Atrogin-1 knockdown inhibits the autophagy-lysosome system in mammalian and avian myotubes. Comp Biochem Physiol A Mol Integr Physiol 2022; 271:111262. [PMID: 35750158 DOI: 10.1016/j.cbpa.2022.111262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/07/2022] [Accepted: 06/17/2022] [Indexed: 10/17/2022]
Abstract
Atrogin-1 plays an important role in ubiquitin-proteasome proteolysis in vertebrate skeletal muscles. Recently, atrogin-1 has been shown to be involved in the autophagy-lysosome system, another proteolytic system, in the murine and fish hearts and skeletal muscles. With the aim to elucidate the effect of atrogin-1 on the autophagy-lysosome system in mammalian and avian skeletal muscles, this study has examined the effects of atrogin-1 knockdown on autophagy-lysosome-related proteins in C2C12 and chicken embryonic myotubes. Using the levels of microtubule-associated protein light chain 3 (LC3)-II protein, it was confirmed that atrogin-1 knockdown blocked the autophagic flux in both the myotubes. In addition, atrogin-1 knockdown in C2C12 myotubes significantly decreased the level of autophagy-related gene (ATG)12-ATG5 conjugate, which is supposedly necessary for the fusion of autophagosomes and lysosomes. Atrogin-1 knockdown also resulted in downregulation of forkhead box O3, a transcription factor for ATG12. These data suggest that atrogin-1 is essential for the normal autophagy-lysosome system in the striated muscles of vertebrates.
Collapse
Affiliation(s)
- Takaoki Saneyasu
- Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan.
| | - Kazuki Ogasawara
- Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan
| | - Yuki Fujiwara
- Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan
| | - Kazuhisa Honda
- Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan
| | - Hiroshi Kamisoyama
- Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan
| |
Collapse
|
30
|
Nishimura Y, Chunthorng-Orn J, Lord S, Musa I, Dawson P, Holm L, Lai YC. Ubiquitin E3 ligase Atrogin-1 protein is regulated via the rapamycin-sensitive mTOR-S6K1 signaling pathway in C2C12 muscle cells. Am J Physiol Cell Physiol 2022; 323:C215-C225. [PMID: 35704697 DOI: 10.1152/ajpcell.00384.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Atrogin-1 and MuRF1 are highly expressed in multiple conditions of skeletal muscle atrophy. The PI3K/Akt/FoxO signaling pathway is well known to regulate Atrogin-1 and MuRF1 gene expressions. However, Akt activation also activates the mammalian target of rapamycin complex 1 (mTORC1) which induces skeletal muscle hypertrophy. Whether mTORC1-dependent signaling has a role in regulating Atrogin-1 and/or MuRF1 gene and protein expression is currently unclear. In this study, we showed that activation of insulin-mediated Akt signaling suppresses both Atrogin-1 and MuRF1 protein contents and that inhibition of Akt increases both Atrogin-1 and MuRF1 protein contents in C2C12 myotubes. Interestingly, inhibition of mTORC1 using a specific mTORC1 inhibitor, rapamycin, increased Atrogin-1, but not MuRF1, protein content. Furthermore, activation of AMP-activated protein kinase (AMPK), a negative regulator of the mTORC1 signaling pathway, also showed distinct time-dependent changes between Atrogin-1 and MuRF1 protein contents, suggesting differential regulatory mechanisms between Atrogin-1 and MuRF1 protein content. To further explore the downstream of mTORC1 signaling, we employed a specific S6K1 inhibitor, PF-4708671. We found that Atrogin-1 protein content was dose-dependently increased with PF-4708671 treatment, whereas MuRF1 protein content was decreased at 50 μM of PF-4708671 treatment. However, MuRF1 protein content was unexpectedly increased when treated with PF-4708671 for a longer period. Overall, our results indicate that Atrogin-1 and MuRF1 protein contents are regulated by different mechanisms, the downstream of Akt, and that Atrogin-1 protein content can be regulated by rapamycin-sensitive mTOR-S6K1 dependent signaling pathway.
Collapse
Affiliation(s)
- Yusuke Nishimura
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Jitpisute Chunthorng-Orn
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Samuel Lord
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Ibrahim Musa
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Peter Dawson
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom.,MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, United Kingdom
| | - Lars Holm
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Yu-Chiang Lai
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom.,MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, United Kingdom.,Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
31
|
Park SM, Kim J, Baek S, Jeon JY, Lee SJ, Kang SY, Yoo MY, Yoon HJ, Kwon SH, Lim K, Oh SJ, Kim BS, Lee KP, Moon BS. Feasibility of 18F-Fluorocholine PET for Evaluating Skeletal Muscle Atrophy in a Starved Rat Model. Diagnostics (Basel) 2022; 12:diagnostics12051274. [PMID: 35626428 PMCID: PMC9141294 DOI: 10.3390/diagnostics12051274] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 05/04/2022] [Accepted: 05/19/2022] [Indexed: 12/25/2022] Open
Abstract
Imaging techniques for diagnosing muscle atrophy and sarcopenia remain insufficient, although various advanced diagnostic methods have been established. We explored the feasibility of 18F-fluorocholine (18F-FCH) positron emission tomography/computed tomography (PET/CT) for evaluating skeletal muscle atrophy, as an imaging technique that tracks choline level changes in muscles. Cell uptake in L6 cells by 18F-FCH was performed in a complete medium containing serum (untreated group, UN) and a serum-free medium (starved group, ST). Small-animal-dedicated PET/CT imaging with 18F-FCH was examined in in-vivo models with rats that were starved for 2 days to cause muscle atrophy. After the hind limbs were dissected, starvation-induced in-vivo models were anatomically confirmed by reverse-transcription polymerase chain reaction to evaluate the expression levels of the atrophy markers muscle RING-finger protein-1 (MuRF-1) and atrogin-1. 18F-FCH uptake was lower in the starvation-induced cells than in the untreated group, and in-vivo PET uptake also revealed a similar tendency (the average standardized uptake value (SUVmean) = 0.26 ± 0.06 versus 0.37 ± 0.07, respectively). Furthermore, the expression levels of MuRF-1 and atrogin-1 mRNA were significantly increased in the starvation-induced muscle atrophy of rats compared to the untreated group. 18F-FCH PET/CT may be a promising tool for diagnosing skeletal muscle atrophy.
Collapse
Affiliation(s)
- Sun Mi Park
- Department of Nuclear Medicine, College of Medicine, Ewha Womans University Seoul Hospital, Ewha Womans University, Seoul 07804, Korea; (S.M.P.); (S.Y.K.)
| | - Jisu Kim
- Physical Activity and Performance Institute, Konkuk University, Seoul 05029, Korea; (J.K.); (K.L.)
| | - Suji Baek
- Research and Development Center, UMUST R&D Corporation, Seoul 01411, Korea;
| | - Joo-Yeong Jeon
- Seoul Center, Korean Basic Science Institute, Seoul 02841, Korea; (J.-Y.J.); (S.H.K.)
| | - Sang Ju Lee
- Department of Nuclear Medicine, College of Medicine, Asan Medical Center, University of Ulsan, Seoul 05505, Korea; (S.J.L.); (S.J.O.)
| | - Seo Young Kang
- Department of Nuclear Medicine, College of Medicine, Ewha Womans University Seoul Hospital, Ewha Womans University, Seoul 07804, Korea; (S.M.P.); (S.Y.K.)
| | - Min Young Yoo
- Department of Nuclear Medicine, College of Medicine, Ewha Womans University Mokdong Hospital, Ewha Womans University, Seoul 07985, Korea; (M.Y.Y.); (H.-J.Y.)
| | - Hai-Jeon Yoon
- Department of Nuclear Medicine, College of Medicine, Ewha Womans University Mokdong Hospital, Ewha Womans University, Seoul 07985, Korea; (M.Y.Y.); (H.-J.Y.)
| | - Seung Hae Kwon
- Seoul Center, Korean Basic Science Institute, Seoul 02841, Korea; (J.-Y.J.); (S.H.K.)
| | - Kiwon Lim
- Physical Activity and Performance Institute, Konkuk University, Seoul 05029, Korea; (J.K.); (K.L.)
| | - Seung Jun Oh
- Department of Nuclear Medicine, College of Medicine, Asan Medical Center, University of Ulsan, Seoul 05505, Korea; (S.J.L.); (S.J.O.)
| | - Bom Sahn Kim
- Department of Nuclear Medicine, College of Medicine, Ewha Womans University Seoul Hospital, Ewha Womans University, Seoul 07804, Korea; (S.M.P.); (S.Y.K.)
- Correspondence: (B.S.K.); (K.P.L.); (B.S.M.)
| | - Kang Pa Lee
- Department of Nuclear Medicine, College of Medicine, Ewha Womans University Seoul Hospital, Ewha Womans University, Seoul 07804, Korea; (S.M.P.); (S.Y.K.)
- Research and Development Center, UMUST R&D Corporation, Seoul 01411, Korea;
- Correspondence: (B.S.K.); (K.P.L.); (B.S.M.)
| | - Byung Seok Moon
- Department of Nuclear Medicine, College of Medicine, Ewha Womans University Seoul Hospital, Ewha Womans University, Seoul 07804, Korea; (S.M.P.); (S.Y.K.)
- Correspondence: (B.S.K.); (K.P.L.); (B.S.M.)
| |
Collapse
|
32
|
Re Cecconi AD, Barone M, Forti M, Lunardi M, Cagnotto A, Salmona M, Olivari D, Zentilin L, Resovi A, Persichitti P, Belotti D, Palo F, Takakura N, Kidoya H, Piccirillo R. Apelin Resistance Contributes to Muscle Loss during Cancer Cachexia in Mice. Cancers (Basel) 2022; 14:cancers14071814. [PMID: 35406586 PMCID: PMC8997437 DOI: 10.3390/cancers14071814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/15/2022] [Accepted: 03/29/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Cancer cachexia is a highly debilitating syndrome involving severe body weight loss. Worldwide around 9–14.5 million cancer patients suffer from cachexia every year and many of them die because of cachexia. Our study aimed to assess the possible role of apelin against muscle loss during cancer growth given its beneficial effects against muscle atrophy during aging. We found apelin exhibiting advantageous effects against atrophy in in vitro models, but not in in vivo models, where we unraveled undesirable apelin resistance that may nullify apelin-based therapy for cancer cachexia. Abstract Cancer cachexia consists of dramatic body weight loss with rapid muscle depletion due to imbalanced protein homeostasis. We found that the mRNA levels of apelin decrease in muscles from cachectic hepatoma-bearing rats and three mouse models of cachexia. Furthermore, apelin expression inversely correlates with MuRF1 in muscle biopsies from cancer patients. To shed light on the possible role of apelin in cachexia in vivo, we generated apelin 13 carrying all the last 13 amino acids of apelin in D isomers, ultimately extending plasma stability. Notably, apelin D-peptides alter cAMP-based signaling in vitro as the L-peptides, supporting receptor binding. In vitro apelin 13 protects myotube diameter from dexamethasone-induced atrophy, restrains rates of degradation of long-lived proteins and MuRF1 expression, but fails to protect mice from atrophy. D-apelin 13 given intraperitoneally for 13 days in colon adenocarcinoma C26-bearing mice does not reduce catabolic pathways in muscles, as it does in vitro. Puzzlingly, the levels of circulating apelin seemingly deriving from cachexia-inducing tumors, increase in murine plasma during cachexia. Muscle electroporation of a plasmid expressing its receptor APJ, unlike apelin, preserves myofiber area from C26-induced atrophy, supporting apelin resistance in vivo. Altogether, we believe that during cachexia apelin resistance occurs, contributing to muscle wasting and nullifying any possible peptide-based treatment.
Collapse
Affiliation(s)
- Andrea David Re Cecconi
- Department of Neurosciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy; (A.D.R.C.); (M.B.); (M.F.); (M.L.); (D.O.); (F.P.)
| | - Mara Barone
- Department of Neurosciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy; (A.D.R.C.); (M.B.); (M.F.); (M.L.); (D.O.); (F.P.)
| | - Mara Forti
- Department of Neurosciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy; (A.D.R.C.); (M.B.); (M.F.); (M.L.); (D.O.); (F.P.)
| | - Martina Lunardi
- Department of Neurosciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy; (A.D.R.C.); (M.B.); (M.F.); (M.L.); (D.O.); (F.P.)
| | - Alfredo Cagnotto
- Molecular Biochemistry and Pharmacology Department, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy; (A.C.); (M.S.)
| | - Mario Salmona
- Molecular Biochemistry and Pharmacology Department, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy; (A.C.); (M.S.)
| | - Davide Olivari
- Department of Neurosciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy; (A.D.R.C.); (M.B.); (M.F.); (M.L.); (D.O.); (F.P.)
| | - Lorena Zentilin
- Molecular Medicine, International Centre for Genetic Engineering and Biotechnology, Via Padriciano 99, 34149 Trieste, Italy;
| | - Andrea Resovi
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Stezzano 87, 24126 Bergamo, Italy; (A.R.); (P.P.); (D.B.)
| | - Perla Persichitti
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Stezzano 87, 24126 Bergamo, Italy; (A.R.); (P.P.); (D.B.)
| | - Dorina Belotti
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Stezzano 87, 24126 Bergamo, Italy; (A.R.); (P.P.); (D.B.)
| | - Federica Palo
- Department of Neurosciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy; (A.D.R.C.); (M.B.); (M.F.); (M.L.); (D.O.); (F.P.)
| | - Nobuyuki Takakura
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita-shi, Osaka 565-0871, Japan;
| | - Hiroyasu Kidoya
- Department of Integrative Vascular Biology, Faculty of Medical Sciences, University of Fukui, 23-3 Matsuoka-Shimoaizuki, Eiheiji, Yoshida, Fukui 910-1193, Japan;
| | - Rosanna Piccirillo
- Department of Neurosciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy; (A.D.R.C.); (M.B.); (M.F.); (M.L.); (D.O.); (F.P.)
- Correspondence: ; Tel.: +39-02-39014371
| |
Collapse
|
33
|
Meng X, Huang Z, Inoue A, Wang H, Wan Y, Yue X, Xu S, Jin X, Shi GP, Kuzuya M, Cheng XW. Cathepsin K activity controls cachexia-induced muscle atrophy via the modulation of IRS1 ubiquitination. J Cachexia Sarcopenia Muscle 2022; 13:1197-1209. [PMID: 35098692 PMCID: PMC8978007 DOI: 10.1002/jcsm.12919] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 11/22/2021] [Accepted: 12/14/2021] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Cachexia is a complicated metabolic disorder that is characterize by progressive atrophy of skeletal muscle. Cathepsin K (CTSK) is a widely expressed cysteine protease that has garnered attention because of its enzymatic and non-enzymatic functions in signalling in various pathological conditions. Here, we examined whether CTSK participates in cancer-induced skeletal muscle loss and dysfunction, focusing on protein metabolic imbalance. METHODS Male 9-week-old wild-type (CTSK+/+ , n = 10) and CTSK-knockout (CTSK-/- , n = 10) mice were injected subcutaneously with Lewis lung carcinoma cells (LLC; 5 × 105 ) or saline, respectively. The mice were then subjected to muscle mass and muscle function measurements. HE staining, immunostaining, quantitative polymerase chain reaction, enzyme-linked immunosorbent assay, and western blotting were used to explore the CTSK expression and IRS1/Akt pathway in the gastrocnemius muscle at various time points. In vitro measurements included CTSK expression, IRS1/Akt pathway-related target molecule expressions, and the diameter of C2C12 myotubes with or without LLC-conditioned medium (LCM). An IRS1 ubiquitin assay, and truncation, co-immunoprecipitation, and co-localization experiments were also performed. RESULTS CTSK+/+ cachectic animals exhibited loss of skeletal muscle mass (muscle weight loss of 15%, n = 10, P < 0.01), muscle dysfunction (grip strength loss > 15%, n = 10, P < 0.01), and fibre area (average area reduction > 30%, n = 5, P < 0.01). Compared with that of non-cachectic CTSK+/+ mice, the skeletal muscle of cachectic CTSK+/+ mice exhibited greater degradation of insulin receptor substrate 1 (IRS1, P < 0.01). In this setting, cachectic muscles exhibited decreases in the phosphorylation levels of protein kinase B (Akt308 , P < 0.01; Akt473 , P < 0.05) and anabolic-related proteins (the mammalian target of rapamycin, P < 0.01) and increased levels of catabolism-related proteins (muscle RING-finger protein-1, P < 0.01; MAFbx1, P < 0.01) in CTSK+/+ mice (n = 3). Although there was no difference in LLC tumour growth (n = 10, P = 0.44), CTSK deletion mitigated the IRS1 degradation, loss of the skeletal muscle mass (n = 10, P < 0.01), and dysfunction (n = 10, P < 0.01). In vitro, CTSK silencing prevented the IRS1 ubiquitination and loss of the myotube myosin heavy chain content (P < 0.01) induced by LCM, and these changes were accelerated by CTSK overexpression even without LCM. Immunoprecipitation showed that CTSK selectively acted on IRS1 in the region of amino acids 268 to 574. The results of co-transfection of IRS1-N-FLAG or IRS1-C-FLAG with CTSK suggested that CTSK selectively cleaves IRS1 and causes ubiquitination-related degradation of IRS1. CONCLUSIONS These results demonstrate that CTSK plays a novel role in IRS1 ubiquitination in LLC-induced muscle wasting, and suggest that CTSK could be an effective therapeutic target for cancer-related cachexia.
Collapse
Affiliation(s)
- Xiangkun Meng
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Zhe Huang
- Department of Cardiology and Hypertension, Yanbian University Hospital, Yanji, China.,Department of Human Cord Stem Cell Therapy, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Aiko Inoue
- Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hailong Wang
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ying Wan
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Xueling Yue
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shengnan Xu
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Xueying Jin
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Masafumi Kuzuya
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Xian Wu Cheng
- Department of Cardiology and Hypertension, Yanbian University Hospital, Yanji, China.,Department of Human Cord Stem Cell Therapy, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| |
Collapse
|
34
|
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: 2.5] [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
|
35
|
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: 10] [Impact Index Per Article: 5.0] [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
|
36
|
Murata K, Namisaki T, Fujimoto Y, Takeda S, Enomoto M, Takaya H, Tsuji Y, Shibamoto A, Suzuki J, Kubo T, Iwai S, Tomooka F, Tanaka M, Kaneko M, Asada S, Koizumi A, Yorioka N, Matsuda T, Ozutsumi T, Ishida K, Ogawa H, Takagi H, Fujinaga Y, Furukawa M, Sawada Y, Nishimura N, Kitagawa K, Sato S, Kaji K, Inoue T, Asada K, Kawaratani H, Moriya K, Akahane T, Mitoro A, Yoshiji H. Clinical Significance of Serum Zinc Levels on the Development of Sarcopenia in Cirrhotic Patients. CANCER DIAGNOSIS & PROGNOSIS 2022; 2:184-193. [PMID: 35399181 PMCID: PMC8962814 DOI: 10.21873/cdp.10093] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/12/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND/AIM Sarcopenia increases the mortality in patients with cirrhosis. Approximately 60% of zinc is accumulated in skeletal muscle. We aimed to determine the role of subclinical zinc deficiency on sarcopenia development in patients with cirrhosis. PATIENTS AND METHODS We enrolled 151 patients with cirrhosis and divided them into the group with normal serum zinc levels (Group N: 80-130 μg/dl; n=38) and group with subclinical zinc deficiency (Group D: <80 μg/dl; n=113). The risk factors for sarcopenia were then investigated. RESULTS Group D had more sarcopenia cases than Group N (31.0% vs. 13.2%). In group D, HGS exhibited a weakly positive but significant correlation with serum zinc levels (R=0.287, p=0.00212), serum zinc levels negatively correlated with both ammonia and myostatin levels (R=-0.254, p=0.0078; R=-0.33, p<0.01), and low zinc levels were independently associated with sarcopenia development. CONCLUSION Patients with cirrhosis showing subclinical zinc deficiency have a significantly higher risk of developing sarcopenia.
Collapse
Affiliation(s)
- Koji Murata
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Tadashi Namisaki
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Yuki Fujimoto
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Soichi Takeda
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Masahide Enomoto
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Hiroaki Takaya
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Yuki Tsuji
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Akihiko Shibamoto
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Junya Suzuki
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Takahiro Kubo
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Satoshi Iwai
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Fumimasa Tomooka
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Misako Tanaka
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Miki Kaneko
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Shohei Asada
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Aritoshi Koizumi
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Nobuyuki Yorioka
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Takuya Matsuda
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Takahiro Ozutsumi
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Koji Ishida
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Hiroyuki Ogawa
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Hirotetsu Takagi
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Yukihisa Fujinaga
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Masanori Furukawa
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Yasuhiko Sawada
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Norihisa Nishimura
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Koh Kitagawa
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Shinya Sato
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Kosuke Kaji
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Takashi Inoue
- Institute for Clinical and Translational Science, Nara Medical University Hospital, Kashihara, Japan
| | - Kiyoshi Asada
- Institute for Clinical and Translational Science, Nara Medical University Hospital, Kashihara, Japan
| | - Hideto Kawaratani
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Kei Moriya
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Takemi Akahane
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Akira Mitoro
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| | - Hitoshi Yoshiji
- Department of Gastroenterology of Nara Medical University, Kashihara, Japan
| |
Collapse
|
37
|
Dalab A, Ali A, Althnaian T, Alkhodair K, Al-Ramadan S. Molecular and ultrastructural investigations of the effect of thermal manipulation during embryogenesis on pectoral and thigh muscles growth factors in broilers. J APPL POULTRY RES 2022. [DOI: 10.1016/j.japr.2021.100188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
38
|
Shimamoto S, Nakashima K, Nishikoba N, Kohrogi R, Ohtsuka A, Fujimura S, Ijiri D. Suppression of FoxO1 mRNA by β 2 -adrenoceptor-cAMP signaling through miR-374b-5p and miR-7a-1-3p in C2C12 myotubes. FEBS Open Bio 2022; 12:627-637. [PMID: 35038382 PMCID: PMC8886325 DOI: 10.1002/2211-5463.13368] [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: 11/08/2021] [Revised: 12/21/2021] [Accepted: 01/13/2022] [Indexed: 11/09/2022] Open
Abstract
β2 -Adrenoceptor (β2 -AR) signaling decreases the transcriptional activity of forkhead box O (FoxO), but the underlying mechanisms remain incompletely understood. Here, we investigated how β2 -AR signaling regulates the protein abundance of FoxO and its transcriptional activity in skeletal muscle. We observed that stimulation of β2 -AR with its selective agonist, clenbuterol, rapidly decreased FoxO1 mRNA expression, and this was accompanied by a decrease in either FoxO1 protein level or FoxO transcriptional activity. We subsequently observed that miR-374b-5p and miR-7a-1-3p were rapidly upregulated in response to β2 -AR stimulation. Transfection with mimics of either miRNA successfully decreased FoxO1 mRNA. Moreover, because β2 -AR stimulation increased cAMP concentration, pretreatment with an adenylyl cyclase inhibitor canceled out these effects of β2 -AR stimulation. These results suggest that β2 -AR stimulation results in rapid upregulation of miR-374b-5p and miR-7a-1-3p in myotubes, which in turn results in a decrease in FoxO1 mRNA expression via the β2 -AR-cAMP signaling pathway.
Collapse
Affiliation(s)
- Saki Shimamoto
- Department of Agricultural Sciences and Natural Resources, Kagoshima University, 1-21-24 Korimoto, Kagoshima, 890-0065, Japan.,The United Graduate School of Agricultural Sciences, Kagoshima University, 1-21-24 Korimoto, Kagoshima, 890-0065, Japan.,Graduate School of Science and Technology, Niigata University, 8050 Ikarashi 2-nocho, Niigata, 950-2181, Japan
| | - Kazuki Nakashima
- Division of Meat Animal and Poultry Research, Institute of Livestock and Grassland Science, NARO, Ikenodai, Tsukuba, 305-0901, Japan
| | - Nao Nishikoba
- Department of Agricultural Sciences and Natural Resources, Kagoshima University, 1-21-24 Korimoto, Kagoshima, 890-0065, Japan
| | - Rukana Kohrogi
- Department of Agricultural Sciences and Natural Resources, Kagoshima University, 1-21-24 Korimoto, Kagoshima, 890-0065, Japan
| | - Akira Ohtsuka
- Department of Agricultural Sciences and Natural Resources, Kagoshima University, 1-21-24 Korimoto, Kagoshima, 890-0065, Japan.,The United Graduate School of Agricultural Sciences, Kagoshima University, 1-21-24 Korimoto, Kagoshima, 890-0065, Japan
| | - Shinobu Fujimura
- Graduate School of Science and Technology, Niigata University, 8050 Ikarashi 2-nocho, Niigata, 950-2181, Japan
| | - Daichi Ijiri
- Department of Agricultural Sciences and Natural Resources, Kagoshima University, 1-21-24 Korimoto, Kagoshima, 890-0065, Japan.,The United Graduate School of Agricultural Sciences, Kagoshima University, 1-21-24 Korimoto, Kagoshima, 890-0065, Japan
| |
Collapse
|
39
|
Kwon D, Kim C, Woo YK, Hwang JK. Inhibitory Effects of Chrysanthemum ( Chrysanthemum morifolium Ramat.) Extract and Its Active Compound Isochlorogenic Acid A on Sarcopenia. Prev Nutr Food Sci 2021; 26:408-416. [PMID: 35047437 PMCID: PMC8747960 DOI: 10.3746/pnf.2021.26.4.408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/19/2021] [Accepted: 08/25/2021] [Indexed: 12/17/2022] Open
Abstract
Sarcopenia, age-related muscle atrophy, weakening muscle strength, and exercise capacity, generally accompany imbalances in protein metabolism. Chrysanthemum morifolium Ramat. extract (CME) and its active compound, isochlo-rogenic acid A (IcA), have been reported to have anti-oxidative, anti-diabetic, and neuroprotective effects. However, the roles of CME and IcA in the regulation of muscle protein turnover-related signaling pathways to attenuate sarcopenia have not been explored. In this study, we investigated CME and IcA based regulation of protein turnover in synthesizing muscle in vitro and in vivo. At the molecular level, CME and IcA promoted phosphorylation of PI3K/Akt and mTOR pathways, which stimulate synthesis of muscle proteins, and suppressed FoxO3a and E3 ubiquitin ligases during protein degrada-tion. In vivo, CME and IcA increased grip strength, exercise capacity, muscle mass and volume, and cross-sectional area of myofibers in middle-aged C57BL/6J mice. These results suggest that CME and IcA may have roles as functional food supplements for delaying sarcopenia by enhancing muscle mass recovery and function.
Collapse
Affiliation(s)
- Dowan Kwon
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Changhee Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Yu Kyong Woo
- Graduate Program in Bioindustrial Engineering, Yonsei University, Seoul 03722, Korea
| | - Jae-Kwan Hwang
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea.,Graduate Program in Bioindustrial Engineering, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
40
|
Multiscale-Engineered Muscle Constructs: PEG Hydrogel Micro-Patterning on an Electrospun PCL Mat Functionalized with Gold Nanoparticles. Int J Mol Sci 2021; 23:ijms23010260. [PMID: 35008686 PMCID: PMC8745500 DOI: 10.3390/ijms23010260] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/10/2021] [Accepted: 12/20/2021] [Indexed: 12/25/2022] Open
Abstract
The development of new, viable, and functional engineered tissue is a complex and challenging task. Skeletal muscle constructs have specific requirements as cells are sensitive to the stiffness, geometry of the materials, and biological micro-environment. The aim of this study was thus to design and characterize a multi-scale scaffold and to evaluate it regarding the differentiation process of C2C12 skeletal myoblasts. The significance of the work lies in the microfabrication of lines of polyethylene glycol, on poly(ε-caprolactone) nanofiber sheets obtained using the electrospinning process, coated or not with gold nanoparticles to act as a potential substrate for electrical stimulation. The differentiation of C2C12 cells was studied over a period of seven days and quantified through both expression of specific genes, and analysis of the myotubes’ alignment and length using confocal microscopy. We demonstrated that our multiscale bio-construct presented tunable mechanical properties and supported the different stages skeletal muscle, as well as improving the parallel orientation of the myotubes with a variation of less than 15°. These scaffolds showed the ability of sustained myogenic differentiation by enhancing the organization of reconstructed skeletal muscle. Moreover, they may be suitable for applications in mechanical and electrical stimulation to mimic the muscle’s physiological functions.
Collapse
|
41
|
Yeh CC, Liu HM, Lee MC, Leu YL, Chiang WH, Chang HH, Lee TY. Phytochemical‑rich herbal formula ATG‑125 protects against sucrose‑induced gastrocnemius muscle atrophy by rescuing Akt signaling and improving mitochondrial dysfunction in young adult mice. Mol Med Rep 2021; 25:57. [PMID: 34913071 PMCID: PMC8711025 DOI: 10.3892/mmr.2021.12572] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/18/2021] [Indexed: 11/06/2022] Open
Abstract
The antioxidant capability of herbal remedies has attracted widespread attention, but their molecular mechanisms in a muscle atrophy model have not been explored. The aim of the present study was to compare the bioactivity of sucrose challenged mice following treatment with ATG‑125. Here, through a combination of transcriptomic and biomedical analysis, herbal formula ATG‑125, a phytochemical‑rich formula, was identified as a protective factor against muscle atrophy in sucrose challenged mice. Gene ontology (GO) identified differentially expressed genes that were primarily enriched in the 'negative regulation of proteolysis', 'cellular amino acid metabolic process', 'lipoprotein particle' and 'cell cycle', all of which were associated with the ATG‑125‑mediated prevention of muscle atrophy, particularly with regard to mitochondrial biogenesis. In skeletal muscle, a set of mitochondrial‑related genes, including angiopoietin‑like 4, nicotinamide riboside kinase 2 (Nmrk2), pyruvate dehydrogenase lipoamide kinase isozyme 4, Asc‑type amino acid transporter 1 and mitochondrial uncoupling protein 3 (Ucp3) were markedly upregulated following ATG‑125 intervention. An increase in Nmrk2 and Ucp3 expression were noted after ATG‑125 treatment, in parallel with upregulation of the 'nicotinate and nicotinamide metabolism' pathway, as determined using the Kyoto Encyclopedia of Genes and Genomes (KEGG). Furthermore, KEGG pathway analysis revealed the downregulation of 'complement and coagulation cascades', 'cholesterol metabolism', 'biosynthesis of amino acids' and 'PPAR signaling pathway', which were associated with the downregulation of serine (or cysteine) peptidase inhibitor clade A member (Serpina)3, Serpina1b, Serpina1d, Serpina1e, apolipoprotein (Apo)a1 and Apoa2, all of which were cardiovascular and diabetes‑associated risk factors and were regulated by ATG‑125. In addition, ATG‑125 treatment resulted in downregulated mRNA expression levels of ATPase sarcoplasmic/endoplasmic reticulum Ca2+ transporting 2, troponin‑I1, troponin‑C1 and troponin‑T1 in young adult gastrocnemius muscle compared with the sucrose group. Nuclear factor‑κB‑hypoxia inducible factor‑1α‑TGFβ receptor type‑II‑vascular endothelial growth factor staining indicated that ATG‑125 decreased sucrose‑induced chronic inflammation. ATG‑125 was sufficient to prevent muscle atrophy, and this protective effect may be mediated through upregulation of AKT phosphorylation, upregulating the insulin growth factor‑1R‑insulin receptor substrate‑PI3K‑AKT pathway, which in turn resulted in a forkhead box O‑dependent decrease in protein degradation pathways, including regulation of atrogin1 and E3 ubiquitin‑protein ligase TRIM63. Peroxisome‑proliferator activated receptor γ coactivator 1α (PGC1α) was decreased in young adult mice challenged with sucrose. ATG‑125 treatment significantly increased PGC1α and significantly increased UCP‑1,2,3 expression levels, which suggested ATG‑125 poised the mitochondria for uncoupling of respiration. This effect is consistent with the increased SIRT1 levels and may explain an increase in mitochondria biogenesis. Taken together, the present study showed that ATG‑125, as an integrator of protein synthesis and degradative pathways, prevented muscle wasting.
Collapse
Affiliation(s)
- Ching-Chuan Yeh
- Graduate Institute of Traditional Chinese Medicine, School of Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan, R.O.C
| | - Hsuan-Miao Liu
- Graduate Institute of Traditional Chinese Medicine, School of Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan, R.O.C
| | - Ming-Chung Lee
- Brion Research Institute of Taiwan, New Taipei City 23143, Taiwan, R.O.C
| | - Yann-Lii Leu
- Graduate Institute of Nature Products, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan, R.O.C
| | - Wei-Han Chiang
- Department of Rehabilitation, Cheng‑Hsin General Hospital, Taipei 11283, Taiwan, R.O.C
| | - Hen-Hong Chang
- Graduate Institute of Integrated Medicine, China Medical University, Taichung 40402, Taiwan, R.O.C
| | - Tzung-Yan Lee
- Graduate Institute of Traditional Chinese Medicine, School of Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan, R.O.C
| |
Collapse
|
42
|
Xia WG, Huang ZH, Chen W, Fouad AM, Abouelezz KFM, Li KC, Huang XB, Wang S, Ruan D, Zhang YN, Zheng CT. Effects of maternal and progeny dietary selenium supplementation on growth performance and antioxidant capacity in ducklings. Poult Sci 2021; 101:101574. [PMID: 34852313 PMCID: PMC8639456 DOI: 10.1016/j.psj.2021.101574] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/03/2021] [Accepted: 09/13/2021] [Indexed: 12/11/2022] Open
Abstract
This study evaluated the effects of selenium (Se) supplementation in maternal and offspring diets on performance and antioxidant capacity of ducklings aged from 0 to 2 wk. A total of 144 female Longyan duck breeders aged 22-wk were allotted into 2 treatments and fed a control diet or a 0.16 mg Se/kg supplemented diet. At 40-wk, 120 offspring from each treatment were divided into 2 groups, with 6 replicates of 10 birds. Using a 2 × 2 factorial design, ducklings from each maternal dietary treatment were assigned to a control diet or a 0.16 mg Se/kg supplemented diet from hatch to 2-wk. Compared with Se-deficient diet, maternal diet supplemented with 0.16 mg Se/kg increased the BW of hatchlings (P < 0.01). There were interactions between maternal and progeny diet with 0.16 mg Se/kg in BW of ducklings aged 2 wk and BW gain (BWG) as ducklings from maternal Se/progeny none treatment had the lightest BW and BWG (P < 0.01). Maternal diet with 0.16 mg Se/kg decreased plasma concentration of uric acid and insulin-like growth factor 1 (P < 0.01), and progeny diet supplemented with 0.16 mg Se/kg increased the activities of glutathione peroxidase 3 (GPx3) in plasma and glutathione peroxidase 1 in erythrocyte (P < 0.01). Maternal diet with 0.16 mg Se/kg increased (P < 0.05) the hepatic activity of total superoxide dismutase (T-SOD). Progeny diet supplemented with 0.16 mg Se/kg increased (P < 0.01) hepatic activity of GPx3 and decreased (P < 0.01) the hepatic concentration of malondialdehyde. Interactions were detected between maternal and progeny diet with 0.16 mg Se/kg in hepatic activity of T-SOD and maternal and progeny diet supplemented with Se displayed the highest hepatic activity of T-SOD (P < 0.05). Overall, Se supplementation in the diet of duck breeders and offspring increased the antioxidant capacity of ducklings. Maternal Se supplementation increased the BW of hatchlings, whereas maternal and progeny dietary Se supplementation did not affect the BWG of ducklings aged from 0 to 2 wk. Se supplementation with additional 0.16 mg/kg in the diet of duck breeders and offspring displayed beneficial effects particularly on the antioxidant capacity in ducklings.
Collapse
Affiliation(s)
- W G Xia
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, P. R. China
| | - Z H Huang
- College of Animal Science, South China Agricultural University, Guangzhou 510640, P. R. China
| | - W Chen
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, P. R. China
| | - A M Fouad
- Department of Animal Production, Faculty of Agriculture, Cairo University, Giza 12613, Egypt
| | - K F M Abouelezz
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, P. R. China; Department of Poultry Production, Faculty of Agriculture, Assiut University, Assiut 71526, Egypt
| | - K C Li
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, P. R. China
| | - X B Huang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, P. R. China
| | - S Wang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, P. R. China
| | - D Ruan
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, P. R. China
| | - Y N Zhang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, P. R. China
| | - C T Zheng
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, P. R. China.
| |
Collapse
|
43
|
Shahsavari M, Mohammadabadi M, Khezri A, Asadi Fozi M, Babenko O, Kalashnyk O, Oleshko V, Tkachenko S. Correlation between insulin-like growth factor 1 gene expression and fennel ( Foeniculum vulgare) seed powder consumption in muscle of sheep. Anim Biotechnol 2021:1-11. [PMID: 34783639 DOI: 10.1080/10495398.2021.2000997] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
It has been shown that addition of fennel in the diets of domestic animals has positive and beneficial effects on growth and meat production traits. Thus, the purpose of current study was to investigate the effect of adding fennel in the ration on growth characteristics and on insulin-like growth factor 1 (IGF1) gene expression in muscle tissue of Kermani lamb. Feeding of animals were performed with three levels of fennel including zero, 10 and 20 g/kg dry matter (DM) for 90 days. After slaughter, small pieces of tissues were removed and rapidly transferred to a nitrogen tank. Then, total RNA extracting and the Real-Time PCR reaction was performed. Results showed that as the level of fennel in the diet increases the amount of IGF1 gene expression also increases significantly in humeral muscle and femur (leg) muscle tissues (p < 0.05). In animals fed with fennel, femur muscle weight, back muscle weight, lean meat weight, final weight, warm carcass weight and live daily gain were greater than in animals fed with diet without fennel (p < 0.05). According to the findings of this investigation, it can be concluded that fennel, by creating positive effects on IGF1 gene expression can be used to improve muscle structure.
Collapse
Affiliation(s)
- Mojtaba Shahsavari
- Faculty of Agriculture, Department of Animal Science, Shahid Bahonar University of Kerman, Kerman, Iran
| | | | - Amin Khezri
- Faculty of Agriculture, Department of Animal Science, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Masoud Asadi Fozi
- Faculty of Agriculture, Department of Animal Science, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Olena Babenko
- Department of Animal Science, Bila Tserkva National Agrarian University, Bila Tserkva, Ukraine
| | - Oleksandr Kalashnyk
- Department of Animal Science, Sumy National Agrarian University, Sumy, Ukraine
| | - Valentyna Oleshko
- Department of Animal Science, Bila Tserkva National Agrarian University, Bila Tserkva, Ukraine
| | - Serhii Tkachenko
- Department of Animal Science, Bila Tserkva National Agrarian University, Bila Tserkva, Ukraine
| |
Collapse
|
44
|
Shokrollahi B, Fazli A, Morammazi S, Saadati N, Ahmad HI, Hassan FU. Cysteamine administration in lambs grazing on mountain pastures: Effects on the body weight, antioxidant capacity, thyroid hormones and growth hormone secretion. Vet Med Sci 2021; 8:328-335. [PMID: 34587370 PMCID: PMC8788981 DOI: 10.1002/vms3.644] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
This study aimed to evaluate the effects of intravenous injection of cysteamine (CS) on body weight (BW), growth hormone (GH), thyroid hormones (TH) secretion, and antioxidant status of growing lambs grazing on mountain pastures. Fifteen lambs (3-4 months of age) were randomly allocated into three experimental groups which received different dosages of CS: 0, 20, and 50 mg/kg BW-1 . The CS was injected on the 1st, 10th, and 20th days of the experiment to the lambs through the jugular vein. Assessment of plasma concentration of GH and TH hormones was carried out at days 0 (a day before the start of CS injections), 15, and 30 of the experiment. The antioxidant enzymes were measured at the end of the experiment. Lambs were weighed at days 0, 10, 20, and 30 of the experiment. The results showed that treatment and time affected the BW, GH, triiodothyronine (T3 ), and tetraiodothyronine (T4 ) secretion. The intravenous injection of CS increased the BW of growing lambs (p < 0.01) and increased the plasma concentration of GH, T3, and T4 (p < 0.01). The treatment also enhanced glutathione peroxidase (GSH-Px; p < 0.05) and reduced malondialdehyde concentrations (MDA; p < 0.01). Total antioxidant capacity (T-AOC) level reduced in CS-1 treatment compared to GC and CS-2 treatments (p < 0.01). The levels of superoxide dismutase (SOD) and catalase (CAT) were not affected by CS. In conclusion, intravenous injection of CS improved BW, GH, and TH concentrations and antioxidant capacity in growing lambs grazing on mountain pastures.
Collapse
Affiliation(s)
- Borhan Shokrollahi
- Department of Animal Science, Faculty of Agriculture, Sanandaj Branch, Islamic Azad University, Sanandaj, Iran
| | - Abdullah Fazli
- Department of Animal Science, Faculty of Agriculture, Sanandaj Branch, Islamic Azad University, Sanandaj, Iran
| | - Salim Morammazi
- Department of Animal Science, Faculty of Agricultural and Natural Resources, Persian Gulf University, Bushehr, Iran
| | - Nazila Saadati
- Department of Biology, Faculty of Basic Sciences, Kurdistan University, Sanandaj, Iran
| | - Hafiz Ishfaq Ahmad
- Department of Animal Breeding and Genetics, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Faiz-Ul Hassan
- Department of Animal Breeding and Genetics, University of Agriculture, Faisalabad, Pakistan
| |
Collapse
|
45
|
Haberecht-Müller S, Krüger E, Fielitz J. Out of Control: The Role of the Ubiquitin Proteasome System in Skeletal Muscle during Inflammation. Biomolecules 2021; 11:biom11091327. [PMID: 34572540 PMCID: PMC8468834 DOI: 10.3390/biom11091327] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 02/07/2023] Open
Abstract
The majority of critically ill intensive care unit (ICU) patients with severe sepsis develop ICU-acquired weakness (ICUAW) characterized by loss of muscle mass, reduction in myofiber size and decreased muscle strength leading to persisting physical impairment. This phenotype results from a dysregulated protein homeostasis with increased protein degradation and decreased protein synthesis, eventually causing a decrease in muscle structural proteins. The ubiquitin proteasome system (UPS) is the predominant protein-degrading system in muscle that is activated during diverse muscle atrophy conditions, e.g., inflammation. The specificity of UPS-mediated protein degradation is assured by E3 ubiquitin ligases, such as atrogin-1 and MuRF1, which target structural and contractile proteins, proteins involved in energy metabolism and transcription factors for UPS-dependent degradation. Although the regulation of activity and function of E3 ubiquitin ligases in inflammation-induced muscle atrophy is well perceived, the contribution of the proteasome to muscle atrophy during inflammation is still elusive. During inflammation, a shift from standard- to immunoproteasome was described; however, to which extent this contributes to muscle wasting and whether this changes targeting of specific muscular proteins is not well described. This review summarizes the function of the main proinflammatory cytokines and acute phase response proteins and their signaling pathways in inflammation-induced muscle atrophy with a focus on UPS-mediated protein degradation in muscle during sepsis. The regulation and target-specificity of the main E3 ubiquitin ligases in muscle atrophy and their mode of action on myofibrillar proteins will be reported. The function of the standard- and immunoproteasome in inflammation-induced muscle atrophy will be described and the effects of proteasome-inhibitors as treatment strategies will be discussed.
Collapse
Affiliation(s)
- Stefanie Haberecht-Müller
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, 17475 Greifswald, Germany;
| | - Elke Krüger
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, 17475 Greifswald, Germany;
- Correspondence: (E.K.); (J.F.)
| | - Jens Fielitz
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, 17475 Greifswald, Germany
- Department of Internal Medicine B, Cardiology, University Medicine Greifswald, 17475 Greifswald, Germany
- Correspondence: (E.K.); (J.F.)
| |
Collapse
|
46
|
Liu M, Li C, Tang H, Gong M, Yue Z, Zhao M, Liu L, Li F. Dietary lysine supplementation improves growth performance and skeletal muscle development in rabbits fed a low protein diet. J Anim Physiol Anim Nutr (Berl) 2021; 106:1118-1129. [PMID: 34496098 DOI: 10.1111/jpn.13632] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/17/2021] [Indexed: 12/20/2022]
Abstract
The purpose of this study was to investigate the effects on growth of Lysine (Lys) supplementation in a low protein diet. We also investigated the gene or protein expression related to skeletal muscle development and intestinal amino acid transporters, and determined the major signalling associated with Lys-regulating skeletal muscle development. 1000 healthy, weights averaging 938.6 ± 6.54 g weaned rabbits were randomly divided into five groups (five replicates in each group and 40 rabbits in each replicate). These groups consisted of the normal protein group (NP group, consuming a diet containing 16.27% protein), the low protein group (LP group, 14.15%-14.19% protein) and the LP group with an addition of 0.15%, 0.3% or 0.45% Lys. The trial included 7 d of pre-feeding and 28 d of exposure to the treatment. Compared with NP diet and LP diet, LP+0.3% Lys group improved growth performance (p < 0.05), full-bore weight and half-bore weight of rabbits (p < 0.05). The LP+0.3% Lys group also resulted in a decrease in the excretion of faecal nitrogen and urinary nitrogen (FN; UN; p < 0.05), and an increase in nitrogen utilisation rate (NUR; p < 0.05). LP diet increased the mRNA expression of MSTN and WWP1, and decreased the mRNA expression of IGF1 (p < 0.05). LP diet decreased the protein expression of P-P70S6K1, P-4EBP1 and P-S6 (p < 0.05). LP+0.3% Lys group attenuated the effects of LP diet on the expression of MSTN, WWP1, IGF1, P-P70S6K1, P-4EBP1 and P-S6 (p < 0.05). LP+0.3% Lys group resulted in an increase in mRNA expression of MyoD and protein expression of P-mTOR relative to the NP and LP groups (p < 0.05). In summary, the addition of Lys to a LP diet provides a theoretical basis for the popularisation and application of Lys in rabbit production.
Collapse
Affiliation(s)
- Mengqi Liu
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science, Shandong Agricultural University, Taian, Shandong, China
| | - Chenyang Li
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science, Shandong Agricultural University, Taian, Shandong, China
| | - Haojia Tang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science, Shandong Agricultural University, Taian, Shandong, China
| | - Maohua Gong
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science, Shandong Agricultural University, Taian, Shandong, China
| | - Zhengkai Yue
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science, Shandong Agricultural University, Taian, Shandong, China
| | - Man Zhao
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science, Shandong Agricultural University, Taian, Shandong, China
| | - Lei Liu
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science, Shandong Agricultural University, Taian, Shandong, China
| | - Fuchang Li
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science, Shandong Agricultural University, Taian, Shandong, China
| |
Collapse
|
47
|
Yamauchi Y, Ferdousi F, Fukumitsu S, Isoda H. Maslinic Acid Attenuates Denervation-Induced Loss of Skeletal Muscle Mass and Strength. Nutrients 2021; 13:nu13092950. [PMID: 34578826 PMCID: PMC8468537 DOI: 10.3390/nu13092950] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/23/2021] [Accepted: 08/23/2021] [Indexed: 11/16/2022] Open
Abstract
Maslinic acid (MA) is a pentacyclic triterpene abundant in olive peels. MA reportedly increases skeletal muscle mass and strength in older adults; however, the underlying mechanism is unknown. This study aimed to investigate the effects of MA on denervated muscle atrophy and strength and to explore the underlying molecular mechanism. Mice were fed either a control diet or a 0.27% MA diet. One week after intervention, the sciatic nerves of both legs were cut to induce muscle atrophy. Mice were examined 14 days after denervation. MA prevented the denervation-induced reduction in gastrocnemius muscle mass and skeletal muscle strength. Microarray gene expression profiling in gastrocnemius muscle demonstrated several potential mechanisms for muscle maintenance. Gene set enrichment analysis (GSEA) revealed different enriched biological processes, such as myogenesis, PI3/AKT/mTOR signaling, TNFα signaling via NF-κB, and TGF-β signaling in MA-treated mice. In addition, qPCR data showed that MA induced Igf1 expression and suppressed the expressions of Atrogin-1, Murf1 and Tgfb. Altogether, our results suggest the potential of MA as a new therapeutic and preventive dietary ingredient for muscular atrophy and strength.
Collapse
Affiliation(s)
- Yuki Yamauchi
- Tsukuba Life Science Innovation Program (T-LSI), University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Japan; (Y.Y.); (S.F.)
- Central Research Laboratory Innovation Center, Nippn Corporation, 5-1-3 Midorigaoka, Atsugi 243-0041, Japan
| | - Farhana Ferdousi
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8572, Japan;
- AIST-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), University of Tsukuba, Tsukuba 305-8572, Japan
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba 305-8575, Japan
| | - Satoshi Fukumitsu
- Tsukuba Life Science Innovation Program (T-LSI), University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Japan; (Y.Y.); (S.F.)
- Central Research Laboratory Innovation Center, Nippn Corporation, 5-1-3 Midorigaoka, Atsugi 243-0041, Japan
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8572, Japan;
| | - Hiroko Isoda
- Tsukuba Life Science Innovation Program (T-LSI), University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Japan; (Y.Y.); (S.F.)
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8572, Japan;
- AIST-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), University of Tsukuba, Tsukuba 305-8572, Japan
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba 305-8575, Japan
- R&D Center for Tailor-Made QOL, University of Tsukuba, Tsukuba 305-8550, Japan
- Correspondence: ; Tel.: +81-298-53-5775
| |
Collapse
|
48
|
Spradlin RA, Vassilakos G, Matheny MK, Jones NC, Goldman JL, Lei H, Barton ER. Deletion of muscle Igf1 exacerbates disuse atrophy weakness in mice. J Appl Physiol (1985) 2021; 131:881-894. [PMID: 34292789 DOI: 10.1152/japplphysiol.00090.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Muscle atrophy occurs as a result of prolonged periods of reduced mechanical stimulation associated with injury or disease. The growth hormone/insulin-like growth factor-1 (GH/IGF-1) axis and load sensing pathways can both aid in recovery from disuse through their shared downstream signaling, but their relative contributions to these processes are not fully understood. The goal of this study was to determine whether reduced muscle IGF-1 altered the response to disuse and reloading. Adult male mice with inducible muscle-specific IGF-1 deletion (MID) induced 1 wk before suspension and age-matched controls (CON) were subjected to hindlimb suspension and reloading. Analysis of muscle force, morphology, gene expression, signaling, and tissue weights was performed in nonsuspended (NS) mice, and those suspended for 7 days or reloaded following suspension for 3, 7, and 14 days. MID mice displayed diminished IGF-1 protein levels and muscle atrophy before suspension. Muscles from suspended CON mice displayed a similar extent of atrophy and depletion of IGF-1, yet combined loss of load and IGF-1 was not additive with respect to muscle mass. In contrast, soleus force generation capacity was diminished to the greatest extent when both suspension and IGF-1 deletion occurred. Recovery of mass, force, and gene expression patterns following suspension were similar in CON and MID mice, even though IGF-1 levels increased only in muscles from CON mice. Diminished strength in disuse atrophy is exacerbated with the loss of muscle IGF-1 production, whereas recovery of mass and strength upon reloading can occur even IGF-1 is low.NEW & NOTEWORTHY A mouse model with skeletal muscle-specific inducible deletion of Igf1 was used to address the importance of this growth factor for the consequences of disuse atrophy. Rapid and equivalent loss of IGF-I and mass occurred with deletion or disuse. Decrements in strength were most severe with combined loss of load and IGF-1. Return of mass and strength upon reloading was independent of IGF-1.
Collapse
Affiliation(s)
- Ray A Spradlin
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida
| | - Georgios Vassilakos
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida
| | - Michael K Matheny
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, Florida
| | - Nathan C Jones
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida
| | - Jessica L Goldman
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida
| | - Hanqin Lei
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida
| | - Elisabeth R Barton
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida
| |
Collapse
|
49
|
Lin YA, Li YR, Chang YC, Hsu MC, Chen ST. Activation of IGF-1 pathway and suppression of atrophy related genes are involved in Epimedium extract (icariin) promoted C2C12 myotube hypertrophy. Sci Rep 2021; 11:10790. [PMID: 34031457 PMCID: PMC8144409 DOI: 10.1038/s41598-021-89039-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 04/12/2021] [Indexed: 02/07/2023] Open
Abstract
The regenerative effect of Epimedium and its major bioactive flavonoid icariin (ICA) have been documented in traditional medicine, but their effect on sarcopenia has not been evaluated. The aim of this study was to investigate the effects of Epimedium extract (EE) on skeletal muscle as represented by differentiated C2C12 cells. Here we demonstrated that EE and ICA stimulated C2C12 myotube hypertrophy by activating several, including IGF-1 signal pathways. C2C12 myotube hypertrophy was demonstrated by enlarged myotube and increased myosin heavy chains (MyHCs). In similar to IGF-1, EE/ICA activated key components of the IGF-1 signal pathway, including IGF-1 receptor. Pre-treatment with IGF-1 signal pathway specific inhibitors such as picropodophyllin, LY294002, and rapamycin attenuated EE induced myotube hypertrophy and MyHC isoform overexpression. In a different way, EE induced MHyC-S overexpression can be blocked by AMPK, but not by mTOR inhibitor. On the level of transcription, EE suppressed myostatin and MRF4 expression, but did not suppress atrogenes MAFbx and MuRF1 like IGF-1 did. Differential regulation of MyHC isoform and atrogenes is probably due to inequivalent AKT and AMPK phosphorylation induced by EE and IGF-1. These findings suggest that EE/ICA stimulates pathways partially overlapping with IGF-1 signaling pathway to promote myotube hypertrophy.
Collapse
Affiliation(s)
- Yi-An Lin
- Department of Sports Medicine, Kaohsiung Medical University, Kaohsiung City, Taiwan.,Division of Endocrinology and Metabolism, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan City, Taiwan.,Graduate Institute of Athletics and Coaching Science, National Taiwan Sport University, Taoyuan City, Taiwan
| | - Yan-Rong Li
- Division of Endocrinology and Metabolism, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan City, Taiwan
| | - Yi-Ching Chang
- Division of Endocrinology and Metabolism, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan City, Taiwan
| | - Mei-Chich Hsu
- Department of Sports Medicine, Kaohsiung Medical University, Kaohsiung City, Taiwan.
| | - Szu-Tah Chen
- Division of Endocrinology and Metabolism, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan City, Taiwan.
| |
Collapse
|
50
|
Memme JM, Slavin M, Moradi N, Hood DA. Mitochondrial Bioenergetics and Turnover during Chronic Muscle Disuse. Int J Mol Sci 2021; 22:ijms22105179. [PMID: 34068411 PMCID: PMC8153634 DOI: 10.3390/ijms22105179] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 12/15/2022] Open
Abstract
Periods of muscle disuse promote marked mitochondrial alterations that contribute to the impaired metabolic health and degree of atrophy in the muscle. Thus, understanding the molecular underpinnings of muscle mitochondrial decline with prolonged inactivity is of considerable interest. There are translational applications to patients subjected to limb immobilization following injury, illness-induced bed rest, neuropathies, and even microgravity. Studies in these patients, as well as on various pre-clinical rodent models have elucidated the pathways involved in mitochondrial quality control, such as mitochondrial biogenesis, mitophagy, fission and fusion, and the corresponding mitochondrial derangements that underlie the muscle atrophy that ensues from inactivity. Defective organelles display altered respiratory function concurrent with increased accumulation of reactive oxygen species, which exacerbate myofiber atrophy via degradative pathways. The preservation of muscle quality and function is critical for maintaining mobility throughout the lifespan, and for the prevention of inactivity-related diseases. Exercise training is effective in preserving muscle mass by promoting favourable mitochondrial adaptations that offset the mitochondrial dysfunction, which contributes to the declines in muscle and whole-body metabolic health. This highlights the need for further investigation of the mechanisms in which mitochondria contribute to disuse-induced atrophy, as well as the specific molecular targets that can be exploited therapeutically.
Collapse
Affiliation(s)
| | | | | | - David A. Hood
- Correspondence: ; Tel.: +1-(416)-736-2100 (ext. 66640)
| |
Collapse
|