1
|
Shi JX, Wang ZY, Wang SW, Shen Q, Tan X. Exercise-mediated muscle-hypothalamus crosstalk: Improvement for cognitive dysfunction caused by disrupted circadian rhythm. Life Sci 2025; 373:123657. [PMID: 40306358 DOI: 10.1016/j.lfs.2025.123657] [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: 02/18/2025] [Revised: 04/14/2025] [Accepted: 04/20/2025] [Indexed: 05/02/2025]
Abstract
In contemporary societal evolution, the increasing disruption of the natural sleep-wake cycle, attributable to factors such as shift work and overexposure to artificial light, has been paralleled by a marked escalation in the incidence of cognitive impairments and the prevalence of neurodegenerative diseases. Current management strategies for cognitive impairments include pharmacological and non-pharmacological interventions. Pharmacological interventions for cognitive impairments typically involve medications to manage cognitive symptoms and improve neurological functions. However, these drugs show limited long-term efficacy in slowing disease progression and may cause side effects. Given the widespread occurrence of cognitive dysfunction, it is crucial to develop accessible non-pharmacological interventions. Physical activity and exercise have emerged as pivotal lifestyle determinants known to exert a modulatory effect on the risk profile for cognitive dysfunction caused by disrupted circadian rhythms. The skeletal muscle, a dynamic tissue, undergoes a profound morphological and metabolic reconfiguration in response to physical exertion, along with the secretion of myokines. Additionally, the hypothalamus, particularly the ventromedial nuclei, arcuate nuclei, and the suprachiasmatic nucleus, have crucial functions in regulating physical activity, influencing energy metabolism, and managing circadian cycles. Nevertheless, the communication between the hypothalamus and skeletal muscle during exercise is not fully understood. This narrative review integrates current knowledge on the interaction between the hypothalamus and skeletal muscle during exercise, emphasizing its neuroendocrine effects and potential therapeutic implications for alleviating cognitive dysfunction associated with disrupted circadian rhythms.
Collapse
Affiliation(s)
- Jun-Xiao Shi
- School of Basic Medical Science, Naval Medical University, Shanghai 200433, China
| | - Zi-Yuan Wang
- School of Basic Medical Science, Naval Medical University, Shanghai 200433, China
| | - Sheng-Wen Wang
- School of Basic Medical Science, Naval Medical University, Shanghai 200433, China
| | - Qi Shen
- Naval Medical Center, Naval Medical University (Second Military Medical University), Shanghai 200433, China.
| | - Xing Tan
- Naval Medical Center, Naval Medical University (Second Military Medical University), Shanghai 200433, China.
| |
Collapse
|
2
|
Gromova A, Cha B, Nguyen N, Garg D, Coscolluela C, Strickland LM, Luong D, Longo F, Sopher BL, ElMallah MK, La Spada AR. Neuromuscular junction transcriptome analysis of spinal and bulbar muscular atrophy mice implicates sarcomere gene expression and calcium flux dysregulation in disease pathogenesis. Hum Mol Genet 2025:ddaf074. [PMID: 40366765 DOI: 10.1093/hmg/ddaf074] [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: 11/06/2024] [Revised: 03/25/2025] [Accepted: 04/24/2025] [Indexed: 05/16/2025] Open
Abstract
X-linked Spinal and Bulbar Muscular Atrophy (SBMA) is a rare, late-onset neuromuscular disease caused by a CAG repeat expansion mutation in the androgen receptor (AR) gene. SBMA is characterized by progressive muscle atrophy of both neurogenic and myopathic etiologies. Previous work has established that mutant AR expression in skeletal muscle could be a significant contributor to neuromuscular decline, yet the mechanisms involved remain ill-defined. As AR is a nuclear hormone receptor transcription factor, we sought to define early changes in gene expression in skeletal muscle of pre-symptomatic SBMA mice, with a focus on transcriptional changes at the neuromuscular junction (NMJ). We describe loss of key NMJ-specific genes in synaptic muscle regions of pre-symptomatic SBMA mice, while extrasynaptic muscle features a coordinated loss of sarcomere genes that coincides with ectopic re-expression of certain NMJ genes. Furthermore, SBMA muscle prominently features dysregulated calcium flux, likely stemming from a compensatory response to early atrophy that greatly exacerbates over time. The SERCA activator CDN1163 conferred a mild rescue in function and muscle size in SBMA mice, while genetic deletion of the gene encoding Myf6/MRF4, a negative regulator of sarcomere gene expression and predicted AR interactor, did not ameliorate muscle atrophy. These studies suggest that modulation of calcium flux could be a promising pharmacological target in SBMA.
Collapse
Affiliation(s)
- Anastasia Gromova
- Department of Pathology and Laboratory Medicine, University of California Irvine, 1001 Health Sciences Road, Irvine, CA 92697, United States
- Muscle Biology and Disease Research Center, University of California Irvine, 1001 Health Sciences Road, Irvine, CA 92697, United States
| | - Byeonggu Cha
- Department of Pathology and Laboratory Medicine, University of California Irvine, 1001 Health Sciences Road, Irvine, CA 92697, United States
| | - Nhat Nguyen
- Department of Pathology and Laboratory Medicine, University of California Irvine, 1001 Health Sciences Road, Irvine, CA 92697, United States
| | - Diya Garg
- Department of Pathology and Laboratory Medicine, University of California Irvine, 1001 Health Sciences Road, Irvine, CA 92697, United States
| | - Connor Coscolluela
- Department of Pathology and Laboratory Medicine, University of California Irvine, 1001 Health Sciences Road, Irvine, CA 92697, United States
| | - Laura M Strickland
- Department of Neurosurgery, Duke University, 40 Duke Medicine Circle, Durham, NC 27710, United States
| | - David Luong
- Department of Pathology and Laboratory Medicine, University of California Irvine, 1001 Health Sciences Road, Irvine, CA 92697, United States
- Muscle Biology and Disease Research Center, University of California Irvine, 1001 Health Sciences Road, Irvine, CA 92697, United States
| | - Fabiana Longo
- Department of Pathology and Laboratory Medicine, University of California Irvine, 1001 Health Sciences Road, Irvine, CA 92697, United States
| | - Bryce L Sopher
- Department of Laboratory Medicine and Pathology, University of Washington Medical Center, 9750 3rd Ave NE, Seattle, WA, 98115, United States
| | - Mai K ElMallah
- Division of Pulmonary Medicine, Department of Pediatrics, Duke University, 2301 Erwin Road, Durham, NC 27710, United States
| | - Albert R La Spada
- Department of Pathology and Laboratory Medicine, University of California Irvine, 1001 Health Sciences Road, Irvine, CA 92697, United States
- Muscle Biology and Disease Research Center, University of California Irvine, 1001 Health Sciences Road, Irvine, CA 92697, United States
- Department of Neurosurgery, Duke University, 40 Duke Medicine Circle, Durham, NC 27710, United States
- Department of Neurology, University of California Irvine, 1001 Health Sciences Road, Irvine, CA 92697, United States
- Department of Biological Chemistry, University of California Irvine, 825 Health Sciences Road, Irvine, CA 92697, United States
- Department of Neurobiology and Behavior, University of California Irvine, 2205 McGaugh Hall, Irvine, CA 92697, United States
- UCI Center for Neurotherapeutics, University of California Irvine, 1001 Health Sciences Road, Irvine, CA 92697, United States
| |
Collapse
|
3
|
Bulgay C, Zorba E, Kazan HH, Bayraktar I, Uca M, Ergün MA, John G, Yusupov RA, Sultanov RI, Semenova EA, Larin AK, Kulemin NA, Generozov EV, Ahmetov II. BDNF coexpresses with MTOR and is associated with muscle fiber size, lean mass and power-related traits. Eur J Appl Physiol 2025:10.1007/s00421-025-05804-3. [PMID: 40299026 DOI: 10.1007/s00421-025-05804-3] [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: 02/16/2025] [Accepted: 04/18/2025] [Indexed: 04/30/2025]
Abstract
PURPOSE Recent research suggests a link between brain-derived neurotrophic factor (BDNF) and the mTOR signaling pathway, a key regulator of protein synthesis and muscle growth. However, it remains unclear whether BDNF influences muscle growth and power performance. Our study aimed to investigate the relationship between the expression of BDNF and MTOR genes in human skeletal muscle and examine the association between genetically predicted higher expression of the BDNF gene and muscle fiber size, lean mass, power performance, and power athlete status. METHODS The study involved 456,382 subjects, including 285 athletes, 112 physically active individuals with muscle fiber composition data, 291 sedentary individuals with gene expression data, 5451 controls, and 450,243 UK Biobank participants. The muscle fiber composition was evaluated using immunohistochemistry, while gene expression analysis was performed using RNA sequencing. BDNF genotyping was carried out using real-time PCR or microarrays. RESULTS We found that BDNF gene expression was positively associated with MTOR gene expression in the vastus lateralis (p < 0.0001). Furthermore, genetically predicted higher BDNF expression (i.e., carriage of the C allele of the rs6265 (Val66Met) BDNF polymorphism) was positively associated with the cross-sectional area of fast-twitch muscle fibers in athletes (p = 0.0069), appendicular lean mass (p = 2.6 × 10⁻⁷), personal best scores of power athletes (p = 0.029), and power athlete status (p = 0.0056). CONCLUSION Our study demonstrates a positive correlation between BDNF and MTOR gene expression in human skeletal muscle, with genetically predicted higher BDNF expression associated with greater muscle fiber size, lean mass, enhanced power performance, and power athlete status.
Collapse
Affiliation(s)
- Celal Bulgay
- Sports Science Faculty, Bingol University, Bingol, Türkiye
| | - Erdal Zorba
- Sport Sciences Faculty, Gazi University, Ankara, Türkiye
| | - Hasan H Kazan
- Department of Medical Biology, Gulhane Faculty of Medicine, University of Health Sciences, Ankara, Türkiye
| | - Işık Bayraktar
- Sports Science Faculty, Alanya Alaaddin Keykubat University, Alanya, Türkiye
| | - Merve Uca
- School of Physical Education and Sports, İstanbul Esenyurt University, Istanbul, Türkiye
| | - Mehmet A Ergün
- Department of Medical Genetics, Faculty of Medicine, Gazi University, Ankara, Türkiye
| | - George John
- Transform Specialist Medical Centre, Dubai, United Arab Emirates
| | - Rinat A Yusupov
- Department of Physical Culture and Sport, Kazan National Research Technical University Named After A.N. Tupolev-KAI, Kazan, Russia
| | - Rinat I Sultanov
- Department of Molecular Biology and Genetics, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Ekaterina A Semenova
- Department of Molecular Biology and Genetics, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
- Research Institute of Physical Culture and Sport, Volga Region State University of Physical Culture, Sport and Tourism, Kazan, Russia
| | - Andrey K Larin
- Department of Molecular Biology and Genetics, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Nikolay A Kulemin
- Department of Molecular Biology and Genetics, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Edward V Generozov
- Department of Molecular Biology and Genetics, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Ildus I Ahmetov
- Department of Molecular Biology and Genetics, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia.
- Sports Genetics Laboratory, St Petersburg Research Institute of Physical Culture, St Petersburg, Russia.
- Laboratory of Genetics of Aging and Longevity, Kazan State Medical University, Kazan, Russia.
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, UK.
| |
Collapse
|
4
|
Ham DJ, Semeraro M, Berger BM, McGowan TJ, Lin S, Maino E, Oliveri F, Rüegg MA. Muscle fiber Myc is dispensable for muscle growth and its forced expression severely perturbs homeostasis. Nat Commun 2025; 16:3190. [PMID: 40180928 PMCID: PMC11968879 DOI: 10.1038/s41467-025-58542-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 03/19/2025] [Indexed: 04/05/2025] Open
Abstract
The oncogenic transcription factor Myc stimulates many growth processes including cell cycle progression and ribosome biogenesis. Myc expression is low in adult skeletal muscle, but is upregulated upon growth stimuli. Furthermore, muscle fiber Myc overexpression recapitulates many aspects of growth-related gene expression, leading to the hypothesis that Myc mediates pro-growth responses to anabolic stimuli, such as exercise. Here, we test this hypothesis by examining mouse models in which Myc is specifically eliminated or overexpressed in skeletal muscle fibers or muscle stem cells (MuSC). While muscle fiber Myc expression increased during muscle growth and Myc expression in MuSCs was required for successful muscle regeneration, muscle fiber Myc expression was dispensable for post-natal, mechanical overload or PKBα/Akt1-induced muscle growth in mice. Similarly, constitutive Myc expression did not promote skeletal muscle hypertrophy, but instead impaired muscle fiber structure and function within days. These data question the role of Myc in skeletal muscle growth.
Collapse
Affiliation(s)
- Daniel J Ham
- Biozentrum, University of Basel, Basel, Switzerland.
| | | | - Bianca M Berger
- Biozentrum, University of Basel, Basel, Switzerland
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | | | - Shuo Lin
- Biozentrum, University of Basel, Basel, Switzerland
| | | | | | | |
Collapse
|
5
|
Orioli L, Thissen JP. Myokines as potential mediators of changes in glucose homeostasis and muscle mass after bariatric surgery. Front Endocrinol (Lausanne) 2025; 16:1554617. [PMID: 40171198 PMCID: PMC11958187 DOI: 10.3389/fendo.2025.1554617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 02/28/2025] [Indexed: 04/03/2025] Open
Abstract
Myokines are bioactive peptides released by skeletal muscle. Myokines exert auto-, para-, or endocrine effects, enabling them to regulate many aspects of metabolism in various tissues. However, the contribution of myokines to the dramatic changes in glucose homeostasis and muscle mass induced by bariatric surgery has not been established. Our review highlights that myokines such as brain-derived neurotrophic factor (BDNF), meteorin-like protein (Metrnl), secreted protein acidic and rich in cysteine (SPARC), apelin (APLN) and myostatin (MSTN) may mediate changes in glucose homeostasis and muscle mass after bariatric surgery. Our review also identifies myonectin as an interesting candidate for future studies, as this myokine may regulate lipid metabolism and muscle mass after bariatric surgery. These myokines may provide novel therapeutic targets and biomarkers for obesity, type 2 diabetes and sarcopenia.
Collapse
Affiliation(s)
- Laura Orioli
- Research Laboratory of Endocrinology, Diabetes, and Nutrition, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Jean-Paul Thissen
- Research Laboratory of Endocrinology, Diabetes, and Nutrition, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
6
|
Fancello I, Willett S, Castiglioni C, Amer S, Santoleri S, Bragg L, Galli F, Cossu G. TNAP expressing adventitial pericytes contribute to myogenesis during foetal development. Vascul Pharmacol 2025; 159:107489. [PMID: 40097085 DOI: 10.1016/j.vph.2025.107489] [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: 08/26/2024] [Revised: 02/27/2025] [Accepted: 03/14/2025] [Indexed: 03/19/2025]
Abstract
OBJECTIVE During growth and differentiation of skeletal muscle, cell types other than canonical myoblasts can be recruited to a myogenic fate. Among these, TNAP+ pericytes can differentiate into skeletal or smooth muscle cells during postnatal growth and contribute to muscle regeneration. However, their role in muscle development has not been investigated. This study aims to characterise pericyte fate choices during embryonic and foetal myogenesis, occurring in the second half of gestation. APPROACH AND RESULTS Using Cre-loxP lineage tracing with multiple reporters including the multifluorescent Confetti, we labelled TNAP+ precursors in vivo and assessed the smooth or skeletal muscle differentiation in their lineage at a perinatal stage. We found that TNAP+ cells contribute in vivo to skeletal and smooth muscle cells, as well as other pericytes, also during pre-natal muscle development. The resulting clones showed that such fate choices are likely to depend on distinct unipotent progenitors rather than multipotent progenitors. In addition, we isolated and differentiated in vitro foetal cells derived from TNAP+ precursors, which showed that they are not spontaneously myogenic unless co-cultured with other skeletal muscle cells. CONCLUSIONS This work extends our understanding of the differentiative potency of these non- canonical skeletal muscle progenitors during prenatal life, with a view to a future application of this knowledge to optimise cell therapies for muscle wasting disorders.
Collapse
Affiliation(s)
- I Fancello
- Division of Cell Matrix Biology & Regenerative Medicine, FBMH, University of Manchester, UK
| | - S Willett
- Division of Cell Matrix Biology & Regenerative Medicine, FBMH, University of Manchester, UK
| | - C Castiglioni
- Division of Cell Matrix Biology & Regenerative Medicine, FBMH, University of Manchester, UK
| | - S Amer
- Division of Cell Matrix Biology & Regenerative Medicine, FBMH, University of Manchester, UK
| | - S Santoleri
- Division of Cell Matrix Biology & Regenerative Medicine, FBMH, University of Manchester, UK
| | - L Bragg
- Division of Cell Matrix Biology & Regenerative Medicine, FBMH, University of Manchester, UK
| | - F Galli
- Division of Cell Matrix Biology & Regenerative Medicine, FBMH, University of Manchester, UK
| | - G Cossu
- Division of Cell Matrix Biology & Regenerative Medicine, FBMH, University of Manchester, UK; Institute of Experimental Neurology, Division of Neurosciences, Ospedale San Raffaele, Milan, Italy; Experimental and Clinical Research Center, Charité Medical Faculty, Max Delbrück Center Berlin, Germany.
| |
Collapse
|
7
|
Santoso J, Do SK, Verma R, Do AV, Hendricks E, Ichida JK, McCain ML. Human iPSC-Derived Motor Neuron Innervation Enhances the Differentiation of Muscle Bundles Engineered with Benchtop Fabrication Techniques. ACS Biomater Sci Eng 2025; 11:1731-1740. [PMID: 39973396 PMCID: PMC11897949 DOI: 10.1021/acsbiomaterials.4c02225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/21/2025]
Abstract
Engineered skeletal muscle tissues are critical tools for disease modeling, drug screening, and regenerative medicine, but are limited by insufficient maturation. Because innervation is a critical regulator of skeletal muscle development and regeneration in vivo, motor neurons are hypothesized to improve the maturity of engineered skeletal muscle tissues. However, the impact of motor neurons on muscle phenotype when added prior to the onset of muscle differentiation is not clearly established. In this study, benchtop fabrication equipment was used to facilely fabricate chambers for engineering three-dimensional (3D) skeletal muscles bundles and measuring their contractile performance. Primary chick myoblasts were embedded in an extracellular matrix hydrogel solution and differentiated into engineered muscle bundles, with or without the addition of human induced pluripotent stem cell (hiPSC)-derived motor neurons. Muscle bundles differentiated with motor neurons had neurites distributed throughout their volume and a higher myogenic index compared to muscle bundles without motor neurons. Innervated muscle bundles also generated significantly higher twitch and tetanus forces in response to electrical field stimulation after 1 and 2 weeks of differentiation compared to noninnervated muscle bundles cultured with or without neurotrophic factors. Noninnervated muscle bundles also experienced a decline in rise and fall times as the culture progressed, whereas innervated muscle bundles and noninnervated muscle bundles with neurotrophic factors maintained more consistent rise and fall times. Innervated muscle bundles also expressed the highest levels of the genes for slow myosin light chain 3 (MYL3) and myoglobin (MB), which are associated with slow twitch fibers. These data suggest that motor neuron innervation enhances the structural and functional development of engineered skeletal muscle constructs and maintains them in a more oxidative phenotype.
Collapse
Affiliation(s)
- Jeffrey
W. Santoso
- Alfred
E. Mann Department of Biomedical Engineering, USC Viterbi School of
Engineering, University of Southern California, Los Angeles, California 90089, United States
| | - Stephanie K. Do
- Alfred
E. Mann Department of Biomedical Engineering, USC Viterbi School of
Engineering, University of Southern California, Los Angeles, California 90089, United States
| | - Riya Verma
- Department
of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
of USC, University of Southern California, Los Angeles, California 90033, United States
| | - Alexander V. Do
- Alfred
E. Mann Department of Biomedical Engineering, USC Viterbi School of
Engineering, University of Southern California, Los Angeles, California 90089, United States
- Thomas
Jefferson High School for Science and Technology, Alexandria, Virginia 22312, United States
| | - Eric Hendricks
- Department
of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
of USC, University of Southern California, Los Angeles, California 90033, United States
| | - Justin K. Ichida
- Department
of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
of USC, University of Southern California, Los Angeles, California 90033, United States
| | - Megan L. McCain
- Alfred
E. Mann Department of Biomedical Engineering, USC Viterbi School of
Engineering, University of Southern California, Los Angeles, California 90089, United States
- Department
of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
of USC, University of Southern California, Los Angeles, California 90033, United States
| |
Collapse
|
8
|
Li J, Liu T, Xian M, Zhou K, Wei J. The Power of Exercise: Unlocking the Biological Mysteries of Peripheral-Central Crosstalk in Parkinson's Disease. J Adv Res 2025:S2090-1232(25)00143-2. [PMID: 40049515 DOI: 10.1016/j.jare.2025.03.001] [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: 07/23/2024] [Revised: 01/06/2025] [Accepted: 03/01/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Exercise is a widely recognized non-pharmacological treatment for Parkinson's Disease (PD). The bidirectional regulation between the brain and peripheral organs has emerged as a promising area of research, with the mechanisms by which exercise impacts PD closely linked to the interplay between peripheral signals and the central nervous system. AIM OF REVIEW This review aims to summarize the mechanisms by which exercise influences peripheral-central crosstalk to improve PD, discuss the molecular processes mediating these interactions, elucidate the pathways through which exercise may modulate PD pathophysiology, and identify directions for future research. KEY SCIENTIFIC CONCEPTS OF REVIEW This review examines how exercise-induced cytokine release promotes neuroprotection in PD. It discusses how exercise can stimulate cytokine secretion through various pathways, including the gut-brain, muscle-brain, liver-brain, adipose-brain, and bone-brain axes, thereby alleviating PD symptoms. Additionally, the potential contributions of the heart-brain, lung-brain, and spleen-brain axes, as well as multi-axis crosstalk-such as the brain-gut-muscle and brain-gut-bone axes-are explored in the context of exercise therapy. The study highlights the need for further research into peripheral-central crosstalk and outlines future directions to address challenges in clinical PD therapy.
Collapse
Affiliation(s)
- Jingwen Li
- Institute for Sports and Brain Health, School of Physical Education, Henan University, Kaifeng, Henan, 475004, China
| | - Tingting Liu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Meiyan Xian
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Ke Zhou
- Institute for Sports and Brain Health, School of Physical Education, Henan University, Kaifeng, Henan, 475004, China.
| | - Jianshe Wei
- Institute for Sports and Brain Health, School of Physical Education, Henan University, Kaifeng, Henan, 475004, China; Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China.
| |
Collapse
|
9
|
Zhang J, Kwan HLR, Chan CB, Lee CW. Localized release of muscle-generated BDNF regulates the initial formation of postsynaptic apparatus at neuromuscular synapses. Cell Death Differ 2025; 32:546-560. [PMID: 39511403 PMCID: PMC11893767 DOI: 10.1038/s41418-024-01404-4] [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: 07/12/2024] [Revised: 10/14/2024] [Accepted: 10/18/2024] [Indexed: 11/15/2024] Open
Abstract
Growing evidence indicates that brain-derived neurotrophic factor (BDNF) is produced in contracting skeletal muscles and is secreted as a myokine that plays an important role in muscle metabolism. However, the involvement of muscle-generated BDNF and the regulation of its vesicular trafficking, localization, proteolytic processing, and spatially restricted release during the development of vertebrate neuromuscular junctions (NMJs) remain largely unknown. In this study, we first reported that BDNF is spatially associated with the actin-rich core domain of podosome-like structures (PLSs) at topologically complex acetylcholine receptor (AChR) clusters in cultured Xenopus muscle cells. The release of spatially localized BDNF is tightly controlled by activity-regulated mechanisms in a calcium-dependent manner. Live-cell time-lapse imaging further showed that BDNF-containing vesicles are transported to and captured at PLSs in both aneural and synaptic AChR clusters for spatially restricted release. Functionally, BDNF knockdown or furin-mediated endoproteolytic activity inhibition significantly suppresses aneural AChR cluster formation, which in turn affects synaptic AChR clustering induced by nerve innervation or agrin-coated beads. Lastly, skeletal muscle-specific BDNF knockout (MBKO) mice exhibit structural defects in the formation of aneural AChR clusters and their subsequent recruitment to nerve-induced synaptic AChR clusters during the initial stages of NMJ development in vivo. Together, this study demonstrated the regulatory roles of PLSs in the intracellular trafficking, spatial localization, and activity-dependent release of BDNF in muscle cells and revealed the involvement of muscle-generated BDNF and its proteolytic conversion in regulating the initial formation of aneural and synaptic AChR clusters during early NMJ development in vitro and in vivo.
Collapse
Affiliation(s)
- Jinkai Zhang
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Hong Kong, China
| | - Hiu-Lam Rachel Kwan
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chi Bun Chan
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Hong Kong, China
| | - Chi Wai Lee
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Hong Kong, China.
- Golden Meditech Centre for NeuroRegeneration Sciences, Hong Kong Baptist University, Hong Kong, China.
| |
Collapse
|
10
|
Tomàs J, Cilleros-Mañé V, Just-Borràs L, Balanyà-Segura M, Polishchuk A, Nadal L, Tomàs M, Silvera-Simón C, Santafé MM, Lanuza MA. Brain-derived neurotrophic factor signaling in the neuromuscular junction during developmental axonal competition and synapse elimination. Neural Regen Res 2025; 20:394-401. [PMID: 38819042 PMCID: PMC11317955 DOI: 10.4103/1673-5374.391314] [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: 07/25/2023] [Revised: 10/25/2023] [Accepted: 11/16/2023] [Indexed: 06/01/2024] Open
Abstract
During the development of the nervous system, there is an overproduction of neurons and synapses. Hebbian competition between neighboring nerve endings and synapses performing different activity levels leads to their elimination or strengthening. We have extensively studied the involvement of the brain-derived neurotrophic factor-Tropomyosin-related kinase B receptor neurotrophic retrograde pathway, at the neuromuscular junction, in the axonal development and synapse elimination process versus the synapse consolidation. The purpose of this review is to describe the neurotrophic influence on developmental synapse elimination, in relation to other molecular pathways that we and others have found to regulate this process. In particular, we summarize our published results based on transmitter release analysis and axonal counts to show the different involvement of the presynaptic acetylcholine muscarinic autoreceptors, coupled to downstream serine-threonine protein kinases A and C (PKA and PKC) and voltage-gated calcium channels, at different nerve endings in developmental competition. The dynamic changes that occur simultaneously in several nerve terminals and synapses converge across a postsynaptic site, influence each other, and require careful studies to individualize the mechanisms of specific endings. We describe an activity-dependent balance (related to the extent of transmitter release) between the presynaptic muscarinic subtypes and the neurotrophin-mediated TrkB/p75NTR pathways that can influence the timing and fate of the competitive interactions between the different axon terminals. The downstream displacement of the PKA/PKC activity ratio to lower values, both in competing nerve terminals and at postsynaptic sites, plays a relevant role in controlling the elimination of supernumerary synapses. Finally, calcium entry through L- and P/Q- subtypes of voltage-gated calcium channels (both channels are present, together with the N-type channel in developing nerve terminals) contributes to reduce transmitter release and promote withdrawal of the most unfavorable nerve terminals during elimination (the weakest in acetylcholine release and those that have already become silent). The main findings contribute to a better understanding of punishment-rewarding interactions between nerve endings during development. Identifying the molecular targets and signaling pathways that allow synapse consolidation or withdrawal of synapses in different situations is important for potential therapies in neurodegenerative diseases.
Collapse
Affiliation(s)
- Josep Tomàs
- Unitat d’Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| | - Víctor Cilleros-Mañé
- Unitat d’Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| | - Laia Just-Borràs
- Unitat d’Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| | - Marta Balanyà-Segura
- Unitat d’Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| | - Aleksandra Polishchuk
- Unitat d’Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| | - Laura Nadal
- Unitat d’Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| | - Marta Tomàs
- Unitat d’Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| | - Carolina Silvera-Simón
- Unitat d’Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| | - Manel M. Santafé
- Unitat d’Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| | - Maria A. Lanuza
- Unitat d’Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| |
Collapse
|
11
|
Lissek T. Enhancement of physiology via adaptive transcription. Pflugers Arch 2025; 477:187-199. [PMID: 39482558 PMCID: PMC11761519 DOI: 10.1007/s00424-024-03037-5] [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: 07/03/2024] [Revised: 09/30/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024]
Abstract
The enhancement of complex physiological functions such as cognition and exercise performance in healthy individuals represents a challenging goal. Adaptive transcription programs that are naturally activated in animals to mediate cellular plasticity in response to stimulation can be leveraged to enhance physiological function above wild-type levels in young organisms and counteract complex functional decline in aging. In processes such as learning and memory and exercise-dependent muscle remodeling, a relatively small number of molecules such as certain stimulus-responsive transcription factors and immediate early genes coordinate widespread changes in cellular physiology. Adaptive transcription can be targeted by various methods including pharmaceutical compounds and gene transfer technologies. Important problems for leveraging adaptive transcription programs for physiological enhancement include a better understanding of their dynamical organization, more precise methods to influence the underlying molecular components, and the integration of adaptive transcription into multi-scale physiological enhancement concepts.
Collapse
Affiliation(s)
- Thomas Lissek
- Interdisciplinary Center for Neurosciences, Heidelberg University, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany.
| |
Collapse
|
12
|
Brown AD, Marko AD, Marko DM, Baranowski BJ, Silvera S, Finch MS, Yang AJ, Dhaliwal R, Ryan CR, Roy BD, Fajardo VA, MacPherson REK. Brain-derived neurotrophic factor drives muscle adaptation similar to aerobic training in mice. FASEB J 2025; 39:e70321. [PMID: 39853792 PMCID: PMC11760663 DOI: 10.1096/fj.202402421r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/13/2024] [Accepted: 01/06/2025] [Indexed: 01/26/2025]
Abstract
This study, in vivo and in vitro, investigated the role of brain-derived neurotrophic factor (BDNF) in skeletal muscle adaptations to aerobic exercise. BDNF is a contraction-induced protein that may play a role in muscle adaptations to aerobic exercise. BDNF is involved in muscle repair, increased fat oxidation, and mitochondrial biogenesis, all of which are adaptations observed with aerobic training. The purpose of this study was two-pronged and investigated the skeletal muscle BDNF response to (1) acute and (2) chronic exercise in male C57BL/6J mice. It also examined if chronic BDNF treatment resulted in similar adaptations to chronic exercise. In aim 1, mice underwent a 2 hr. treadmill exercise bout. In aim 2, mice were assigned to one of four groups: (1) control (CON); (2) endurance training (ET; treadmill running 1 h/day, 5 days/wk); (3) BDNF (BDNF; 0.5 mg/kg·bw, 5 days/wk); (4) endurance training and BDNF (ET + BDNF) for 8 weeks. Results demonstrated that the soleus (SOL) had higher BDNF content compared with the extensor digitorum longus (EDL) and that SOL BDNF increased with acute exercise. After chronic exercise and BDNF treatment, treadmill testing to exhaustion demonstrated a main effect of BDNF and ET on increasing exercise capacity. In vitro contractile assessment of the EDL revealed BDNF treatment resulted in similar increases in the max rate of relaxation as ET. EDL force-frequency analysis showed ET + BDNF produced higher force than CON and BDNF, indicating an additive effect. BDNF increased EDL mitochondrial proteins, COXIV, and CS. These results demonstrate that BDNF contributes to muscle adaptations observed with ET.
Collapse
Affiliation(s)
- Alexander D. Brown
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | - Alexander D. Marko
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | - Daniel M. Marko
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | - Bradley J. Baranowski
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | - Sebastian Silvera
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | - Michael S. Finch
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | - Alex J. Yang
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | - Roopan Dhaliwal
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | - Chantal R. Ryan
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | - Brian D. Roy
- Department of KinesiologyFaculty of Applied Health Sciences, Brock UniversitySt. CatharinesOntarioCanada
| | - Val A. Fajardo
- Department of KinesiologyFaculty of Applied Health Sciences, Brock UniversitySt. CatharinesOntarioCanada
| | - Rebecca E. K. MacPherson
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
- Centre for NeuroscienceBrock UniversitySt. CatharinesOntarioCanada
| |
Collapse
|
13
|
Wang J, Jia D, Zhang Z, Wang D. Exerkines and Sarcopenia: Unveiling the Mechanism Behind Exercise-Induced Mitochondrial Homeostasis. Metabolites 2025; 15:59. [PMID: 39852400 PMCID: PMC11767263 DOI: 10.3390/metabo15010059] [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: 12/17/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 01/26/2025] Open
Abstract
Background/Objectives: Sarcopenia, characterized by the progressive loss of muscle mass and strength, is linked to physical disability, metabolic dysfunction, and an increased risk of mortality. Exercise therapy is currently acknowledged as a viable approach for addressing sarcopenia. Nevertheless, the molecular mechanisms behind exercise training or physical activity remain poorly understood. The disruption of mitochondrial homeostasis is implicated in the pathogenesis of sarcopenia. Exercise training effectively delays the onset of sarcopenia by significantly maintaining mitochondrial homeostasis, including promoting mitophagy, improving mitochondrial biogenesis, balancing mitochondrial dynamics, and maintaining mitochondrial redox. Exerkines (e.g., adipokines, myokines, hepatokines, and osteokines), signaling molecules released in response to exercise training, may potentially contribute to skeletal muscle metabolism through ameliorating mitochondrial homeostasis, reducing inflammation, and regulating protein synthesis as a defense against sarcopenia. Methods: In this review, we provide a detailed summary of exercise-induced exerkines and confer their benefit, with particular focus on their impact on mitochondrial homeostasis in the context of sarcopenia. Results: Exercise induces substantial adaptations in skeletal muscle, including increased muscle mass, improved muscle regeneration and hypertrophy, elevated hormone release, and enhanced mitochondrial function. An expanding body of research highlights that exerkines have the potential to regulate processes such as mitophagy, mitochondrial biogenesis, dynamics, autophagy, and redox balance. These mechanisms contribute to the maintenance of mitochondrial homeostasis, thereby supporting skeletal muscle metabolism and mitochondrial health. Conclusions: Through a comprehensive investigation of the molecular mechanisms within mitochondria, the context reveals new insights into the potential of exerkines as key exercise-protective sensors for combating sarcopenia.
Collapse
Affiliation(s)
- Jiayin Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (J.W.); (D.J.)
| | - Dandan Jia
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (J.W.); (D.J.)
| | - Zhiwang Zhang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (J.W.); (D.J.)
| | - Dan Wang
- School of Athletic Performance, Shanghai University of Sport, Shanghai 200438, China
| |
Collapse
|
14
|
Santoso JW, Do SK, Verma R, Do AV, Hendricks E, Ichida JK, McCain ML. Human iPSC-derived motor neuron innervation enhances the differentiation of muscle bundles engineered with benchtop fabrication techniques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.02.626391. [PMID: 39677637 PMCID: PMC11642770 DOI: 10.1101/2024.12.02.626391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Engineered skeletal muscle tissues are critical tools for disease modeling, drug screening, and regenerative medicine, but are limited by insufficient maturation. Because innervation is a critical regulator of skeletal muscle development and regeneration in vivo, motor neurons are hypothesized to improve the maturity of engineered skeletal muscle tissues. Although motor neurons have been added to pre-engineered muscle constructs, the impact of motor neurons added prior to the onset of muscle differentiation has not been evaluated. In this study, benchtop fabrication equipment was used to facilely fabricate chambers for engineering 3-dimensional (3-D) skeletal muscles bundles and measuring their contractile performance. Primary chick myoblasts were embedded in an extracellular matrix hydrogel solution and differentiated into engineered muscle bundles, with or without the addition of human induced pluripotent stem cell (hiPSC)-derived motor neurons. Muscle bundles differentiated with motor neurons had neurites distributed throughout their volume and a higher myogenic index compared to muscle bundles without motor neurons. Innervated muscle bundles also generated significantly higher twitch and tetanus forces in response to electrical field stimulation after one and two weeks of differentiation compared to non-innervated muscle bundles cultured with or without neurotrophic factors. Non-innervated muscle bundles also experienced a decline in rise and fall times as the culture progressed, whereas innervated muscle bundles and non-innervated muscle bundles with neurotrophic factors maintained more consistent rise and fall times. Innervated muscle bundles also expressed the highest levels of the genes for slow myosin light chain 3 (MYL3) and myoglobin (MB), which are associated with slow twitch fibers. These data suggest that motor neuron innervation enhances the structural and functional development of engineered skeletal muscle constructs and maintains them in a more oxidative phenotype.
Collapse
Affiliation(s)
- Jeffrey W. Santoso
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Stephanie K. Do
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Riya Verma
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90033, USA
| | - Alexander V. Do
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA
- Thomas Jefferson Highschool for Science and Technology, Alexandria, VA 22312, USA
| | - Eric Hendricks
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90033, USA
| | - Justin K. Ichida
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90033, USA
| | - Megan L. McCain
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
15
|
Kostka M, Morys J, Małecki A, Nowacka-Chmielewska M. Muscle-brain crosstalk mediated by exercise-induced myokines - insights from experimental studies. Front Physiol 2024; 15:1488375. [PMID: 39687518 PMCID: PMC11647023 DOI: 10.3389/fphys.2024.1488375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Over the past couple of decades, it has become apparent that skeletal muscles might be engaged in endocrine signaling, mostly as a result of exercise or physical activity in general. The importance of this phenomenon is currently studied in terms of the impact that exercise- or physical activity -induced signaling factors have, in the interaction of the "muscle-brain crosstalk." So far, skeletal muscle-derived myokines were demonstrated to intercede in the connection between muscles and a plethora of various organs such as adipose tissue, liver, or pancreas. However, the exact mechanism of muscle-brain communication is yet to be determined. It is speculated that, in particular, brain-derived neurotrophic factor (BDNF), irisin, cathepsin B (CTSB), interleukin 6 (IL-6), and insulin-like growth factor-1 (IGF-1) partake in this crosstalk by promoting neuronal proliferation and synaptic plasticity, also resulting in improved cognition and ameliorated behavioral alterations. Researchers suggest that myokines might act directly on the brain parenchyma via crossing the blood-brain barrier (BBB). The following article reviews the information available regarding rodent studies on main myokines determined to cross the BBB, specifically addressing the association between exercise-induced myokine release and central nervous system (CNS) impairments. Although the hypothesis of skeletal muscles being critical sources of myokines seems promising, it should not be forgotten that the origin of these factors might vary, depending on the cell types engaged in their synthesis. Limited amount of research providing information on alterations in myokines expression in various organs at the same time, results in taking them only as circumstantial evidence on the way to determine the actual involvement of skeletal muscles in the overall state of homeostasis. The following article reviews the information available regarding rodent studies on main myokines determined to cross the BBB, specifically addressing the association between exercise-induced myokine release and CNS impairments.
Collapse
Affiliation(s)
| | | | | | - Marta Nowacka-Chmielewska
- Laboratory of Molecular Biology, Institute of Physiotherapy and Health Sciences, Academy of Physical Education, Katowice, Poland
| |
Collapse
|
16
|
Pang BPS, Iu ECY, Hang M, Chan WS, Tse MCL, Yeung CTY, Wang M, Siu PMF, Lee CW, Ye K, So H, Chan CB. Deficiency of muscle-generated brain-derived neurotrophic factor causes inflammatory myopathy through reactive oxygen species-mediated necroptosis and pyroptosis. Redox Biol 2024; 78:103418. [PMID: 39531828 PMCID: PMC11602578 DOI: 10.1016/j.redox.2024.103418] [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: 09/12/2024] [Revised: 10/21/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024] Open
Abstract
Idiopathic inflammatory myopathy (commonly known as myositis) is a group of immune-related diseases characterized by muscle damage, weakness, and fatigue with unknown causes. Although overactivated innate immunity is a widely believed cause of myositis onset, the mechanism that provokes and maintains a high immune response in myositis patients is still unclear. This study aims to test if brain-derived neurotrophic factor (BDNF) deficiency per se is sufficient to cause myositis and determine its underlying mechanism. We found that ablating BDNF production in skeletal muscle is sufficient to trigger myositis development in mice. Muscle-specific Bdnf knockout (MBKO) mice displayed extensive myocyte necrosis, mononuclear cell infiltration, and myophagocytosis. In association with these damages, elevated production of pro-inflammatory cytokines such as interleukin (IL) 23, IL-1β, IL-18, and tumor necrosis factor α (TNFα) was found in the muscle of MBKO mice. Disruption of sarcolemma integrity was also detected in MBKO mice, which is a result of necroptosis executioner Mixed lineage kinase domain-like protein (MLKL) and pyroptosis executioner Gasdermin D (GSDMD) activation. Mechanistically, diminishing BDNF synthesis in myotubes enhances the accumulation of mitochondrial reactive oxygen species (mtROS), which sensitizes the cells towards TNFα-induced receptor-interacting protein kinase (RIPs) activation and promotes the formation of NLR family pyrin domain containing 3 (NLRP3)-containing inflammasome. BDNF deficiency-induced cell death could be alleviated by scavenging mtROS, suppressing the activity of GSDMD, or inhibiting receptor-interacting kinase 3 (RIP3). Similarly, supplementation of BDNF mimetics, suppression of RIP3 activity, increasing the intramyocellular antioxidant, or enhancing mitophagy ameliorated the myopathies of MBKO mice and improved their muscle strength. Together, our study demonstrates that insufficient BDNF production in mouse muscle causes the development of pathological features of myositis via enhancing oxidative stress, necroptosis, and pyroptosis in myofibers.
Collapse
Affiliation(s)
- Brian Pak Shing Pang
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Elsie Chit Yu Iu
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Miaojia Hang
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Wing Suen Chan
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Margaret Chui Ling Tse
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Connie Tsz Ying Yeung
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Mingfu Wang
- Shenzhen Key Laboratory of Food Nutrition and Health, Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, Hong Kong Special Administrative Region
| | - Parco Ming Fai Siu
- Division of Kinesiology, School of Public Health, The University of Hong Kong, Pok Fu Lam, Hong Kong Special Administrative Region
| | - Chi Wai Lee
- Department of Biology, Hong Kong Baptist University, Hong Kong Special Administrative Region
| | - Keqiang Ye
- Faculty of Life and Health Sciences, and Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Hong Kong Special Administrative Region
| | - Ho So
- Department of Medicine & Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Chi Bun Chan
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Hong Kong Special Administrative Region; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong Special Administrative Region.
| |
Collapse
|
17
|
Pei L, Yao Z, Liang D, Yang K, Tao L. Mitochondria in skeletal system-related diseases. Biomed Pharmacother 2024; 181:117505. [PMID: 39499974 DOI: 10.1016/j.biopha.2024.117505] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 09/16/2024] [Accepted: 09/25/2024] [Indexed: 12/21/2024] Open
Abstract
Skeletal system-related diseases, such as osteoporosis, arthritis, osteosarcoma and sarcopenia, are becoming major public health concerns. These diseases are characterized by insidious progression, which seriously threatens patients' health and quality of life. Early diagnosis and prevention in high-risk populations can effectively prevent the deterioration of these patients. Mitochondria are essential organelles for maintaining the physiological activity of the skeletal system. Mitochondrial functions include contributing to the energy supply, modulating the Ca2+ concentration, maintaining redox balance and resisting the inflammatory response. They participate in the regulation of cellular behaviors and the responses of osteoblasts, osteoclasts, chondrocytes and myocytes to external stimuli. In this review, we describe the pathogenesis of skeletal system diseases, focusing on mitochondrial function. In addition to osteosarcoma, a characteristic of which is active mitochondrial metabolism, mitochondrial damage occurs during the development of other diseases. Impairment of mitochondria leads to an imbalance in osteogenesis and osteoclastogenesis in osteoporosis, cartilage degeneration and inflammatory infiltration in arthritis, and muscle atrophy and excitationcontraction coupling blockade in sarcopenia. Overactive mitochondrial metabolism promotes the proliferation and migration of osteosarcoma cells. The copy number of mitochondrial DNA and mitochondria-derived peptides can be potential biomarkers for the diagnosis of these disorders. High-risk factor detection combined with mitochondrial component detection contributes to the early detection of these diseases. Targeted mitochondrial intervention is an effective method for treating these patients. We analyzed skeletal system-related diseases from the perspective of mitochondria and provided new insights for their diagnosis, prevention and treatment by demonstrating the relationship between mitochondria and the skeletal system.
Collapse
Affiliation(s)
- Liang Pei
- Department of Pediatrics, Shengjing Hospital of China Medical University, China
| | - Zhuo Yao
- Department of Orthopedics, First Hospital of China Medical University, No.155 Nanjing North Street, Shenyang, China
| | - Dong Liang
- Department of Orthopedics, First Hospital of China Medical University, No.155 Nanjing North Street, Shenyang, China
| | - Keda Yang
- Department of Orthopedics, First Hospital of China Medical University, No.155 Nanjing North Street, Shenyang, China..
| | - Lin Tao
- Department of Orthopedics, First Hospital of China Medical University, No.155 Nanjing North Street, Shenyang, China..
| |
Collapse
|
18
|
Honda M, Inoue R, Nishiyama K, Ueda T, Komuro A, Amano H, Sugisawa R, Dash S, Shirakawa J, Okada H. Vgll2 as an integrative regulator of mitochondrial function and contractility specific to skeletal muscle. J Cell Physiol 2024; 239:e31436. [PMID: 39286968 DOI: 10.1002/jcp.31436] [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: 05/13/2024] [Revised: 07/26/2024] [Accepted: 09/05/2024] [Indexed: 09/19/2024]
Abstract
During skeletal muscle adaptation to physiological or pathophysiological signals, contractile apparatus and mitochondrial function are coordinated to alter muscle fiber type. Although recent studies have identified various factors involved in modifying contractile proteins and mitochondrial function, the molecular mechanisms coordinating contractile and metabolic functions during muscle fiber transition are not fully understood. Using a gene-deficient mouse approach, our previous studies uncovered that vestigial-like family member 2 (Vgll2), a skeletal muscle-specific transcription cofactor activated by exercise, is essential for fast-to-slow adaptation of skeletal muscle. The current study provides evidence that Vgll2 plays a role in increasing muscle mitochondrial mass and oxidative capacity. Transgenic Vgll2 overexpression in mice altered muscle fiber composition toward the slow type and enhanced exercise endurance, which contradicted the outcomes observed with Vgll2 deficiency. Vgll2 expression was positively correlated with the expression of genes related to mitochondrial function in skeletal muscle, mitochondrial DNA content, and protein abundance of oxidative phosphorylation complexes. Additionally, Vgll2 overexpression significantly increased the maximal respiration of isolated muscle fibers and enhanced the suppressive effects of endurance training on weight gain. Notably, no additional alteration in expression of myosin heavy chain genes was observed after exercise, suggesting that Vgll2 plays a direct role in regulating mitochondrial function, independent of its effect on contractile components. The observed increase in exercise endurance and metabolic efficiency may be attributed to the acute upregulation of genes promoting fatty acid utilization as a direct consequence of Vgll2 activation facilitated by endurance exercise. Thus, the current study establishes that Vgll2 is an integrative regulator of mitochondrial function and contractility in skeletal muscle.
Collapse
Affiliation(s)
- Masahiko Honda
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Ryota Inoue
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Gunma, Japan
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama, Kanagawa, Japan
| | - Kuniyuki Nishiyama
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Gunma, Japan
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama, Kanagawa, Japan
- Department of Pediatrics, Graduate School of Medicine, Yokohama City University, Yokohma, Kanagawa, Japan
| | - Takeshi Ueda
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
- Graduate School of Medical Sciences, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Akiyoshi Komuro
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Hisayuki Amano
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Ryoichi Sugisawa
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Suman Dash
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
- Graduate School of Medical Sciences, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Jun Shirakawa
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Gunma, Japan
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama, Kanagawa, Japan
| | - Hitoshi Okada
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
- Graduate School of Medical Sciences, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
- Antiaging Center, Kindai University, Higashi-Osaka, Osaka, Japan
| |
Collapse
|
19
|
Tosolini AP, Abatecola F, Negro S, Sleigh JN, Schiavo G. The node of Ranvier influences the in vivo axonal transport of mitochondria and signaling endosomes. iScience 2024; 27:111158. [PMID: 39524336 PMCID: PMC11544082 DOI: 10.1016/j.isci.2024.111158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/15/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Efficient long-range axonal transport is essential for maintaining neuronal function, and perturbations in this process underlie severe neurological diseases. Nodes of Ranvier (NoR) are short, specialized unmyelinated axonal domains with a unique molecular and structural composition. Currently, it remains unresolved how the distinct molecular structures of the NoR impact axonal transport dynamics. Using intravital time-lapse microscopy of sciatic nerves in live, anesthetized mice, we reveal (1) similar morphologies of the NoR in fast and slow motor axons, (2) signaling endosomes and mitochondria accumulate specifically at the distal node, and (3) unique axonal transport profiles of signaling endosomes and mitochondria transiting through the NoR. Collectively, these findings provide important insights into the fundamental physiology of peripheral nerve axons, motor neuron subtypes, and diverse organelle dynamics at the NoR. Furthermore, this work has relevance for several pathologies affecting peripheral nerves and the NoR.
Collapse
Affiliation(s)
- Andrew P. Tosolini
- Department of Neuromuscular Diseases and UCL Queen Square Motor Neuron Disease Centre, Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD 4067, Australia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4067, Australia
| | - Federico Abatecola
- Department of Neuromuscular Diseases and UCL Queen Square Motor Neuron Disease Centre, Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Samuele Negro
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
- U.O.C. Clinica Neurologica, Azienda Ospedale, University of Padua, 35128 Padua, Italy
| | - James N. Sleigh
- Department of Neuromuscular Diseases and UCL Queen Square Motor Neuron Disease Centre, Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- UK Dementia Research Institute, University College London, London WC1E 6BT, UK
| | - Giampietro Schiavo
- Department of Neuromuscular Diseases and UCL Queen Square Motor Neuron Disease Centre, Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- UK Dementia Research Institute, University College London, London WC1E 6BT, UK
| |
Collapse
|
20
|
Hu T, Furuichi Y, Manabe Y, Yamada K, Katakura K, Aoki Y, Tang K, Sakai T, Fujii NL. Myokine BDNF highly expressed in Type I fibers inhibits the differentiation of myotubes into Type II fibers. Mol Biol Rep 2024; 51:1143. [PMID: 39531063 PMCID: PMC11557626 DOI: 10.1007/s11033-024-10044-3] [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: 08/20/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Myofibers are broadly classified as slow-twitch (Type I) and fast-twitch (Type II) fibers. These two types of myofibers coexist within the same skeletal muscle tissue, determining the contractile and metabolic properties of skeletal muscle tissue by fiber type distribution. METHODS AND RESULTS By examining each fiber type separately, we confirmed that brain-derived neurotrophic factor (BDNF) gene is highly expressed in Type I fibers. When exposed to BDNF, primary myotubes exhibited reduced expression of Myosin Heavy Chain (MyHC) II, a marker protein characteristic of Type II fibers. BDNF overexpression in regenerating muscle tissue led to a decrease in the distribution of Type IIA fibers. CONCLUSIONS We suggest that BDNF highly expressed in Type I fibers downregulates MyHC II expression in myotubes, eventually inhibiting Type IIA fiber generation.
Collapse
Affiliation(s)
- Teng Hu
- Department of Health Promotion Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo, 192-0397, Japan
| | - Yasuro Furuichi
- Department of Health Promotion Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo, 192-0397, Japan
| | - Yasuko Manabe
- Department of Health Promotion Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo, 192-0397, Japan
| | - Kenichiro Yamada
- Department of Health Promotion Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo, 192-0397, Japan
| | - Kengo Katakura
- Department of Health Promotion Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo, 192-0397, Japan
| | - Yuna Aoki
- Department of Health Promotion Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo, 192-0397, Japan
| | - Kun Tang
- Department of Health Promotion Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo, 192-0397, Japan
| | - Takaomi Sakai
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo, 192-0397, Japan
| | - Nobuharu L Fujii
- Department of Health Promotion Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo, 192-0397, Japan.
| |
Collapse
|
21
|
Martins LA, Schiavo A, Paz LV, Xavier LL, Mestriner RG. Neural underpinnings of fine motor skills under stress and anxiety: A review. Physiol Behav 2024; 282:114593. [PMID: 38782244 DOI: 10.1016/j.physbeh.2024.114593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 05/25/2024]
Abstract
This review offers a comprehensive examination of how stress and anxiety affect motor behavior, particularly focusing on fine motor skills and gait adaptability. We explore the role of several neurochemicals, including brain-derived neurotrophic factor (BDNF) and dopamine, in modulating neural plasticity and motor control under these affective states. The review highlights the importance of developing therapeutic strategies that enhance motor performance by leveraging the interactions between key neurochemicals. Additionally, we investigate the complex interplay between emotional-cognitive states and sensorimotor behaviors, showing how stress and anxiety disrupt neural integration, leading to impairments in skilled movements and negatively impacting quality of life. Synthesizing evidence from human and rodent studies, we provide a detailed understanding of the relationships among stress, anxiety, and motor behavior. Our findings reveal neurophysiological pathways, behavioral outcomes, and potential therapeutic targets, emphasizing the intricate connections between neurobiological mechanisms, environmental factors, and motor performance.
Collapse
Affiliation(s)
- Lucas Athaydes Martins
- Pontifical Catholic University of Rio Grande do Sul (PUCRS). Graduate Program in Biomedical Gerontology, Av. Ipiranga, 6681, Porto Alegre, Brazil; Pontifical Catholic University of Rio Grande do Sul (PUCRS). Neuroscience, Motor Behavior, and Rehabilitation Research Group (NECORE-CNPq), Av. Ipiranga, 6681, Porto Alegre, Brazil
| | - Aniuska Schiavo
- Pontifical Catholic University of Rio Grande do Sul (PUCRS). Graduate Program in Biomedical Gerontology, Av. Ipiranga, 6681, Porto Alegre, Brazil; Pontifical Catholic University of Rio Grande do Sul (PUCRS). Neuroscience, Motor Behavior, and Rehabilitation Research Group (NECORE-CNPq), Av. Ipiranga, 6681, Porto Alegre, Brazil
| | - Lisiê Valéria Paz
- Pontifical Catholic University of Rio Grande do Sul (PUCRS). Graduate Program in Cellular and Molecular Biology, Av. Ipiranga, 6681, Porto Alegre, Brazil
| | - Léder Leal Xavier
- Pontifical Catholic University of Rio Grande do Sul (PUCRS). Neuroscience, Motor Behavior, and Rehabilitation Research Group (NECORE-CNPq), Av. Ipiranga, 6681, Porto Alegre, Brazil; Pontifical Catholic University of Rio Grande do Sul (PUCRS). Graduate Program in Cellular and Molecular Biology, Av. Ipiranga, 6681, Porto Alegre, Brazil
| | - Régis Gemerasca Mestriner
- Pontifical Catholic University of Rio Grande do Sul (PUCRS). Graduate Program in Biomedical Gerontology, Av. Ipiranga, 6681, Porto Alegre, Brazil; Pontifical Catholic University of Rio Grande do Sul (PUCRS). Neuroscience, Motor Behavior, and Rehabilitation Research Group (NECORE-CNPq), Av. Ipiranga, 6681, Porto Alegre, Brazil; Pontifical Catholic University of Rio Grande do Sul (PUCRS). Graduate Program in Cellular and Molecular Biology, Av. Ipiranga, 6681, Porto Alegre, Brazil.
| |
Collapse
|
22
|
Chen ZT, Weng ZX, Lin JD, Meng ZX. Myokines: metabolic regulation in obesity and type 2 diabetes. LIFE METABOLISM 2024; 3:loae006. [PMID: 39872377 PMCID: PMC11749576 DOI: 10.1093/lifemeta/loae006] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/23/2024] [Accepted: 02/29/2024] [Indexed: 01/30/2025]
Abstract
Skeletal muscle plays a vital role in the regulation of systemic metabolism, partly through its secretion of endocrine factors which are collectively known as myokines. Altered myokine levels are associated with metabolic diseases, such as type 2 diabetes (T2D). The significance of interorgan crosstalk, particularly through myokines, has emerged as a fundamental aspect of nutrient and energy homeostasis. However, a comprehensive understanding of myokine biology in the setting of obesity and T2D remains a major challenge. In this review, we discuss the regulation and biological functions of key myokines that have been extensively studied during the past two decades, namely interleukin 6 (IL-6), irisin, myostatin (MSTN), growth differentiation factor 11 (GDF11), fibroblast growth factor 21 (FGF21), apelin, brain-derived neurotrophic factor (BDNF), meteorin-like (Metrnl), secreted protein acidic and rich in cysteine (SPARC), β-aminoisobutyric acid (BAIBA), Musclin, and Dickkopf 3 (Dkk3). Related to these, we detail the role of exercise in myokine expression and secretion together with their contributions to metabolic physiology and disease. Despite significant advancements in myokine research, many myokines remain challenging to measure accurately and investigate thoroughly. Hence, new research techniques and detection methods should be developed and rigorously tested. Therefore, developing a comprehensive perspective on myokine biology is crucial, as this will likely offer new insights into the pathophysiological mechanisms underlying obesity and T2D and may reveal novel targets for therapeutic interventions.
Collapse
Affiliation(s)
- Zhi-Tian Chen
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Zhejiang University-University of Edinburgh Institute (ZJE), School of Medicine, Zhejiang University, Haining, Zhejiang 314400, China
| | - Zhi-Xuan Weng
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jiandie D Lin
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, United States
| | - Zhuo-Xian Meng
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Department of Geriatrics, Affiliated Hangzhou First People’s Hospital, Hangzhou, Zhejiang 310006, China
| |
Collapse
|
23
|
Brandenburg JE, Fogarty MJ, Zhan WZ, Kopper LA, Sieck GC. Postnatal survival of phrenic motor neurons is promoted by BDNF/TrkB.FL signaling. J Appl Physiol (1985) 2024; 136:1113-1121. [PMID: 38511211 PMCID: PMC11368516 DOI: 10.1152/japplphysiol.00911.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 03/22/2024] Open
Abstract
The number of motor neurons (MNs) declines precipitously during the final trimester before birth. Thereafter, the number of MNs remains relatively stable, with their connections to skeletal muscle dependent on neurotrophins, including brain-derived neurotrophic factor (BDNF) signaling through its high-affinity full-length tropomyosin-related kinase receptor subtype B (TrkB.FL) receptor. As a genetic knockout of BDNF leads to extensive MN loss and postnatal death within 1-2 days after birth, we tested the hypothesis that postnatal inhibition of BDNF/TrkB.FL signaling is important for postnatal phrenic MN (PhMN) survival. In the present study, we used a 1NMPP1-sensitive TrkBF616A mutant mouse to evaluate the effects of inhibition of TrkB kinase activity on phrenic MN (PhMN) numbers and diaphragm muscle (DIAm) fiber cross-sectional area (CSA). Pups were exposed to 1NMPP1 or vehicle (DMSO) from birth to 21 days old (weaning) via the mother's ingestion in the drinking water. Following weaning, the right phrenic nerve was exposed in the neck and the proximal end dipped in a rhodamine solution to retrogradely label PhMNs. After 24 h, the cervical spinal cord and DIAm were excised. Labeled PhMNs were imaged using confocal microscopy, whereas DIAm strips were frozen at ∼1.5× resting length, cryosectioned, and stained with hematoxylin and eosin to assess CSA. We observed an ∼34% reduction in PhMN numbers and increased primary dendrite numbers in 1NMPP1-treated TrkBF616A mice. The distribution of PhMN size (somal surface area) DIAm fiber cross-sectional areas did not differ. We conclude that survival of PhMNs during early postnatal development is sensitive to BDNF/TrkB.FL signaling.NEW & NOTEWORTHY During early postnatal development, BDNF/TrkB signaling promotes PhMN survival. Inhibition of BDNF/TrkB signaling in early postnatal development does not impact PhMN size. Inhibition of BDNF/TrkB signaling in early postnatal development does not impact the number or CSA of DIAm fibers.
Collapse
Affiliation(s)
- Joline E Brandenburg
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, United States
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Matthew J Fogarty
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| | - Wen-Zhi Zhan
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| | - Leo A Kopper
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| | - Gary C Sieck
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| |
Collapse
|
24
|
Benucci S, Ruiz A, Franchini M, Ruggiero L, Zoppi D, Sitsapesan R, Lindsay C, Pelczar P, Pietrangelo L, Protasi F, Treves S, Zorzato F. A novel, patient-derived RyR1 mutation impairs muscle function and calcium homeostasis in mice. J Gen Physiol 2024; 156:e202313486. [PMID: 38445312 PMCID: PMC10911087 DOI: 10.1085/jgp.202313486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/20/2023] [Accepted: 02/09/2024] [Indexed: 03/07/2024] Open
Abstract
RYR1 is the most commonly mutated gene associated with congenital myopathies, a group of early-onset neuromuscular conditions of variable severity. The functional effects of a number of dominant RYR1 mutations have been established; however, for recessive mutations, these effects may depend on multiple factors, such as the formation of a hypomorphic allele, or on whether they are homozygous or compound heterozygous. Here, we functionally characterize a new transgenic mouse model knocked-in for mutations identified in a severely affected child born preterm and presenting limited limb movement. The child carried the homozygous c.14928C>G RYR1 mutation, resulting in the p.F4976L substitution. In vivo and ex vivo assays revealed that homozygous mice fatigued sooner and their muscles generated significantly less force compared with their WT or heterozygous littermates. Electron microscopy, biochemical, and physiological analyses showed that muscles from RyR1 p.F4976L homozygous mice have the following properties: (1) contain fewer calcium release units and show areas of myofibrillar degeneration, (2) contain less RyR1 protein, (3) fibers show smaller electrically evoked calcium transients, and (4) their SR has smaller calcium stores. In addition, single-channel recordings indicate that RyR1 p.F4976L exhibits higher Po in the presence of 100 μM [Ca2+]. Our mouse model partly recapitulates the clinical picture of the homozygous human patient and provides significant insight into the functional impact of this mutation. These results will help understand the pathology of patients with similar RYR1 mutations.
Collapse
Affiliation(s)
- Sofia Benucci
- Departments of Biomedicine and Neurology, Basel University Hospital, Basel, Switzerland
| | - Alexis Ruiz
- Departments of Biomedicine and Neurology, Basel University Hospital, Basel, Switzerland
| | - Martina Franchini
- Departments of Biomedicine and Neurology, Basel University Hospital, Basel, Switzerland
| | - Lucia Ruggiero
- Dipartimento di Neuroscienze, Scienze Riproduttive ed Odontostomatologiche, Università degli Studi di Napoli Federico II, Napoli, Italy
| | - Dario Zoppi
- Dipartimento di Neuroscienze, Scienze Riproduttive ed Odontostomatologiche, Università degli Studi di Napoli Federico II, Napoli, Italy
| | | | - Chris Lindsay
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Pawel Pelczar
- Center for Transgenic Models, University of Basel, Basel, Switzerland
| | - Laura Pietrangelo
- DMSI, Department of Medicine and Aging Sciences and CAST, Center for Advanced Studies and Technology, University G. d’Annunzio of Chieti-Pescara, Chieti, Italy
| | - Feliciano Protasi
- DMSI, Department of Medicine and Aging Sciences and CAST, Center for Advanced Studies and Technology, University G. d’Annunzio of Chieti-Pescara, Chieti, Italy
| | - Susan Treves
- Departments of Biomedicine and Neurology, Basel University Hospital, Basel, Switzerland
- Department of Life Science and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Francesco Zorzato
- Departments of Biomedicine and Neurology, Basel University Hospital, Basel, Switzerland
- Department of Life Science and Biotechnology, University of Ferrara, Ferrara, Italy
| |
Collapse
|
25
|
Chan WS, Ng CF, Pang BPS, Hang M, Tse MCL, Iu ECY, Ooi XC, Yang X, Kim JK, Lee CW, Chan CB. Exercise-induced BDNF promotes PPARδ-dependent reprogramming of lipid metabolism in skeletal muscle during exercise recovery. Sci Signal 2024; 17:eadh2783. [PMID: 38502732 PMCID: PMC11022078 DOI: 10.1126/scisignal.adh2783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 02/29/2024] [Indexed: 03/21/2024]
Abstract
Post-exercise recovery is essential to resolve metabolic perturbations and promote long-term cellular remodeling in response to exercise. Here, we report that muscle-generated brain-derived neurotrophic factor (BDNF) elicits post-exercise recovery and metabolic reprogramming in skeletal muscle. BDNF increased the post-exercise expression of the gene encoding PPARδ (peroxisome proliferator-activated receptor δ), a transcription factor that is a master regulator of lipid metabolism. After exercise, mice with muscle-specific Bdnf knockout (MBKO) exhibited impairments in PPARδ-regulated metabolic gene expression, decreased intramuscular lipid content, reduced β-oxidation, and dysregulated mitochondrial dynamics. Moreover, MBKO mice required a longer period to recover from a bout of exercise and did not show increases in exercise-induced endurance capacity. Feeding naïve mice with the bioavailable BDNF mimetic 7,8-dihydroxyflavone resulted in effects that mimicked exercise-induced adaptations, including improved exercise capacity. Together, our findings reveal that BDNF is an essential myokine for exercise-induced metabolic recovery and remodeling in skeletal muscle.
Collapse
Affiliation(s)
- Wing Suen Chan
- School of Biological Sciences, the University of Hong Kong, 5N10 Kadoorie Biological Sciences Building, Pokfulam Road, Hong Kong, China
| | - Chun Fai Ng
- School of Biological Sciences, the University of Hong Kong, 5N10 Kadoorie Biological Sciences Building, Pokfulam Road, Hong Kong, China
| | - Brian Pak Shing Pang
- School of Biological Sciences, the University of Hong Kong, 5N10 Kadoorie Biological Sciences Building, Pokfulam Road, Hong Kong, China
| | - Miaojia Hang
- School of Biological Sciences, the University of Hong Kong, 5N10 Kadoorie Biological Sciences Building, Pokfulam Road, Hong Kong, China
| | - Margaret Chui Ling Tse
- School of Biological Sciences, the University of Hong Kong, 5N10 Kadoorie Biological Sciences Building, Pokfulam Road, Hong Kong, China
| | - Elsie Chit Yu Iu
- School of Biological Sciences, the University of Hong Kong, 5N10 Kadoorie Biological Sciences Building, Pokfulam Road, Hong Kong, China
| | - Xin Ci Ooi
- School of Biological Sciences, the University of Hong Kong, 5N10 Kadoorie Biological Sciences Building, Pokfulam Road, Hong Kong, China
| | - Xiuying Yang
- Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica of Peking Union Medical College, Beijing 101399, China
| | - Jason K. Kim
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | - Chi Wai Lee
- Department of Biology, Hong Kong Baptist University, Hong Kong, China
| | - Chi Bun Chan
- School of Biological Sciences, the University of Hong Kong, 5N10 Kadoorie Biological Sciences Building, Pokfulam Road, Hong Kong, China
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
26
|
Edman S, Horwath O, Van der Stede T, Blackwood SJ, Moberg I, Strömlind H, Nordström F, Ekblom M, Katz A, Apró W, Moberg M. Pro-Brain-Derived Neurotrophic Factor (BDNF), but Not Mature BDNF, Is Expressed in Human Skeletal Muscle: Implications for Exercise-Induced Neuroplasticity. FUNCTION 2024; 5:zqae005. [PMID: 38706964 PMCID: PMC11065112 DOI: 10.1093/function/zqae005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/15/2023] [Accepted: 01/24/2024] [Indexed: 05/07/2024] Open
Abstract
Exercise promotes brain plasticity partly by stimulating increases in mature brain-derived neurotrophic factor (mBDNF), but the role of the pro-BDNF isoform in the regulation of BDNF metabolism in humans is unknown. We quantified the expression of pro-BDNF and mBDNF in human skeletal muscle and plasma at rest, after acute exercise (+/- lactate infusion), and after fasting. Pro-BDNF and mBDNF were analyzed with immunoblotting, enzyme-linked immunosorbent assay, immunohistochemistry, and quantitative polymerase chain reaction. Pro-BDNF was consistently and clearly detected in skeletal muscle (40-250 pg mg-1 dry muscle), whereas mBDNF was not. All methods showed a 4-fold greater pro-BDNF expression in type I muscle fibers compared to type II fibers. Exercise resulted in elevated plasma levels of mBDNF (55%) and pro-BDNF (20%), as well as muscle levels of pro-BDNF (∼10%, all P < 0.05). Lactate infusion during exercise induced a significantly greater increase in plasma mBDNF (115%, P < 0.05) compared to control (saline infusion), with no effect on pro-BDNF levels in plasma or muscle. A 3-day fast resulted in a small increase in plasma pro-BDNF (∼10%, P < 0.05), with no effect on mBDNF. Pro-BDNF is highly expressed in human skeletal muscle, particularly in type I fibers, and is increased after exercise. While exercising with higher lactate augmented levels of plasma mBDNF, exercise-mediated increases in circulating mBDNF likely derive partly from release and cleavage of pro-BDNF from skeletal muscle, and partly from neural and other tissues. These findings have implications for preclinical and clinical work related to a wide range of neurological disorders such as Alzheimer's, clinical depression, and amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Sebastian Edman
- Åstrand Laboratory, Department of Physiology, Nutrition and Biomechanics, Swedish School of Sport and Health Sciences, Stockholm 114 33, Sweden
- Department of Women’s and Children’s Health, Karolinska Institute, Stockholm 171 77, Sweden
| | - Oscar Horwath
- Åstrand Laboratory, Department of Physiology, Nutrition and Biomechanics, Swedish School of Sport and Health Sciences, Stockholm 114 33, Sweden
| | - Thibaux Van der Stede
- Department of Movement and Sport Sciences, Ghent University, Ghent 9000, Belgium
- The August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 1172, Denmark
| | - Sarah Joan Blackwood
- Åstrand Laboratory, Department of Physiology, Nutrition and Biomechanics, Swedish School of Sport and Health Sciences, Stockholm 114 33, Sweden
| | - Isabel Moberg
- Åstrand Laboratory, Department of Physiology, Nutrition and Biomechanics, Swedish School of Sport and Health Sciences, Stockholm 114 33, Sweden
| | - Henrik Strömlind
- Åstrand Laboratory, Department of Physiology, Nutrition and Biomechanics, Swedish School of Sport and Health Sciences, Stockholm 114 33, Sweden
| | - Fabian Nordström
- Åstrand Laboratory, Department of Physiology, Nutrition and Biomechanics, Swedish School of Sport and Health Sciences, Stockholm 114 33, Sweden
| | - Maria Ekblom
- Department of Physical Activity and Health, Swedish School of Sport and Health Sciences, Stockholm 114 33, Sweden
- Department of Neuroscience, Karolinska Institute, Stockholm 171 77, Sweden
| | - Abram Katz
- Åstrand Laboratory, Department of Physiology, Nutrition and Biomechanics, Swedish School of Sport and Health Sciences, Stockholm 114 33, Sweden
| | - William Apró
- Åstrand Laboratory, Department of Physiology, Nutrition and Biomechanics, Swedish School of Sport and Health Sciences, Stockholm 114 33, Sweden
- Department of Clinical Science, Intervention and Technology, Karolinska Institute, Stockholm 171 77, Sweden
| | - Marcus Moberg
- Åstrand Laboratory, Department of Physiology, Nutrition and Biomechanics, Swedish School of Sport and Health Sciences, Stockholm 114 33, Sweden
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm 171 77, Sweden
| |
Collapse
|
27
|
Li HH, Livneh H, Huang HL, Wang YH, Lu MC, Chen WJ, Tsai TY. Integrating Chinese Herbal Medicine into Conventional Care Was Related to Lower Risk of Sarcopenia Among Rheumatid Arthritis Patients: A Retrospective, Population-Based Study. J Multidiscip Healthc 2023; 16:3117-3127. [PMID: 37901596 PMCID: PMC10612505 DOI: 10.2147/jmdh.s428948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/28/2023] [Indexed: 10/31/2023] Open
Abstract
Objective Sarcopenia is a frequently observed comorbidity of rheumatoid arthritis (RA) due to the chronic activation of the innate immune system. Accumulating evidence has indicated that Chinese herbal medicine (CHM) safely suppresses proinflammatory pathways and controls inflammation-associated disease, but its effect in reducing the risk of developing sarcopenia among RA subjects has not been established. We conducted a population-level cohort study to compare the sarcopenia risk in patients with RA who use or do not use CHM. Methods Using claims from a nationwide insurance database, we recruited patients with newly diagnosed RA and without sarcopenia between 2002 and 2010. Propensity score matching was applied to randomly select sets of CHM users and non-CHM users to compare the sarcopenia risk until the end of 2013. The risk of new-onset sarcopenia was assessed using the Cox proportional hazards model. Results As compared to non-CHM users, those receiving CHM treatment had a lower incidence of sarcopenia (7.69 vs 9.83 per 1000 person-years). CHM was correlated with a decreased chance of sarcopenia after controlling for potential covariates. Notably, use of CHM for more than two years may diminish the risk of getting sarcopenia by about 47% when taken as prescribed. Prescriptions of several herbal formulae may benefit the reduction of sarcopenia risk, such as Yan-Hu-Suo, Bei-Mu, Da-Huang, Huang Qin, Ping-Wei-San (PWS), Shu-Jing-Huo-Xue-Tang (SJHXT) and Chuan-Xiong-Cha-Tiao-San (CXCTS). Conclusion This study produced new evidence as it is the first to show that the longer duration of CHM use was correlated to reduced risk of sarcopenia in a dose-dependent manner, implying that CHM treatment could be embraced as a routine care strategy for preventing sarcopenia.
Collapse
Affiliation(s)
- Hsin-Hua Li
- Department of Chinese Medicine, Dalin Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation, Chiayi, 62247, Taiwan
| | - Hanoch Livneh
- Rehabilitation Counseling Program, Portland State University, Portland, OR, 97207-0751, USA
| | - Hua-Lung Huang
- Department of Rehabilitation, Dalin Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
| | - Yu-Han Wang
- Center of Sports Medicine, Dalin Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation, Chiayi, 62247, Taiwan
| | - Ming-Chi Lu
- Division of Allergy, Immunology and Rheumatology, Dalin Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation, Chiayi, 62247, Taiwan
- School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan
| | - Wei-Jen Chen
- Department of Chinese Medicine, Dalin Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation, Chiayi, 62247, Taiwan
- Center of Sports Medicine, Dalin Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation, Chiayi, 62247, Taiwan
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien, 97004, Taiwan
- Graduate Institute of Sports Science, National Taiwan Sport University, Taoyuan, 33301, Taiwan
| | - Tzung-Yi Tsai
- Department of Medical Research, Dalin Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation, Chiayi, 62247, Taiwan
- Department of Nursing, Tzu Chi University of Science and Technology, Hualien, 97004, Taiwan
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan, 70428, Taiwan
| |
Collapse
|
28
|
Swain M, Soman SK, Tapia K, Dagda RY, Dagda RK. Brain-derived neurotrophic factor protects neurons by stimulating mitochondrial function through protein kinase A. J Neurochem 2023; 167:104-125. [PMID: 37688457 PMCID: PMC10543477 DOI: 10.1111/jnc.15945] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 07/28/2023] [Accepted: 08/01/2023] [Indexed: 09/10/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) stimulates dendrite outgrowth and synaptic plasticity by activating downstream protein kinase A (PKA) signaling. Recently, BDNF has been shown to modulate mitochondrial respiration in isolated brain mitochondria, suggesting that BDNF can modulate mitochondrial physiology. However, the molecular mechanisms by which BDNF stimulates mitochondrial function in neurons remain to be elucidated. In this study, we surmised that BDNF binds to the TrkB receptor and translocates to mitochondria to govern mitochondrial physiology in a PKA-dependent manner. Confocal microscopy and biochemical subcellular fractionation assays confirm the localization of the TrkB receptor in mitochondria. The translocation of the TrkB receptor to mitochondria was significantly enhanced upon treating primary cortical neurons with exogenous BDNF, leading to rapid PKA activation. Showing a direct role of BDNF in regulating mitochondrial structure/function, time-lapse confocal microscopy in primary cortical neurons showed that exogenous BDNF enhances mitochondrial fusion, anterograde mitochondrial trafficking, and mitochondrial content within dendrites, which led to increased basal and ATP-linked mitochondrial respiration and glycolysis as assessed by an XF24e metabolic analyzer. BDNF-mediated regulation of mitochondrial structure/function requires PKA activity as treating primary cortical neurons with a pharmacological inhibitor of PKA or transiently expressing constructs that target an inhibitor peptide of PKA (PKI) to the mitochondrion abrogated BDNF-mediated mitochondrial fusion and trafficking. Mechanistically, western/Phos-tag blots show that BDNF stimulates PKA-mediated phosphorylation of Drp1 and Miro-2 to promote mitochondrial fusion and elevate mitochondrial content in dendrites, respectively. Effects of BDNF on mitochondrial function were associated with increased resistance of neurons to oxidative stress and dendrite retraction induced by rotenone. Overall, this study revealed new mechanisms of BDNF-mediated neuroprotection, which entails enhancing mitochondrial health and function of neurons.
Collapse
Affiliation(s)
- Maryann Swain
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, 1664 North Virginia Street, Nevada, 89557, USA
| | - Smijin K. Soman
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, 1664 North Virginia Street, Nevada, 89557, USA
| | - Kylea Tapia
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, 1664 North Virginia Street, Nevada, 89557, USA
| | - Raul Y. Dagda
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, 1664 North Virginia Street, Nevada, 89557, USA
| | - Ruben K. Dagda
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, 1664 North Virginia Street, Nevada, 89557, USA
| |
Collapse
|
29
|
Stansberry WM, Pierchala BA. Neurotrophic factors in the physiology of motor neurons and their role in the pathobiology and therapeutic approach to amyotrophic lateral sclerosis. Front Mol Neurosci 2023; 16:1238453. [PMID: 37692101 PMCID: PMC10483118 DOI: 10.3389/fnmol.2023.1238453] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 08/11/2023] [Indexed: 09/12/2023] Open
Abstract
The discovery of the neurotrophins and their potent survival and trophic effects led to great enthusiasm about their therapeutic potential to rescue dying neurons in neurodegenerative diseases. The further discovery that brain-derived neurotrophic factor (BDNF), ciliary neurotrophic factor (CNTF) and glial cell line-derived neurotrophic factor (GDNF) had potent survival-promoting activity on motor neurons led to the proposal for their use in motor neuron diseases such as amyotrophic lateral sclerosis (ALS). In this review we synthesize the literature pertaining to the role of NGF, BDNF, CNTF and GDNF on the development and physiology of spinal motor neurons, as well as the preclinical studies that evaluated their potential for the treatment of ALS. Results from the clinical trials of these molecules will also be described and, with the aid of decades of hindsight, we will discuss what can reasonably be concluded and how this information can inform future clinical development of neurotrophic factors for ALS.
Collapse
Affiliation(s)
- Wesley M. Stansberry
- The Department of Anatomy, Cell Biology and Physiology, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Brian A. Pierchala
- The Department of Anatomy, Cell Biology and Physiology, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
30
|
Félix-Soriano E, Stanford KI. Exerkines and redox homeostasis. Redox Biol 2023; 63:102748. [PMID: 37247469 PMCID: PMC10236471 DOI: 10.1016/j.redox.2023.102748] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/12/2023] [Accepted: 05/12/2023] [Indexed: 05/31/2023] Open
Abstract
Exercise physiology has gained increasing interest due to its wide effects to promote health. Recent years have seen a growth in this research field also due to the finding of several circulating factors that mediate the effects of exercise. These factors, termed exerkines, are metabolites, growth factors, and cytokines secreted by main metabolic organs during exercise to regulate exercise systemic and tissue-specific effects. The metabolic effects of exerkines have been broadly explored and entail a promising target to modulate beneficial effects of exercise in health and disease. However, exerkines also have broad effects to modulate redox signaling and homeostasis in several cellular processes to improve stress response. Since redox biology is central to exercise physiology, this review summarizes current evidence for the cross-talk between redox biology and exerkines actions. The role of exerkines in redox biology entails a response to oxidative stress-induced pathological cues to improve health outcomes and to modulate exercise adaptations that integrate redox signaling.
Collapse
Affiliation(s)
- Elisa Félix-Soriano
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Kristin I Stanford
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA; Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
31
|
Zheng XQ, Huang J, Lin JL, Song CL. Pathophysiological mechanism of acute bone loss after fracture. J Adv Res 2023; 49:63-80. [PMID: 36115662 PMCID: PMC10334135 DOI: 10.1016/j.jare.2022.08.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 07/29/2022] [Accepted: 08/31/2022] [Indexed: 10/14/2022] Open
Abstract
BACKGROUND Acute bone loss after fracture is associated with various effects on the complete recovery process and a risk of secondary fractures among patients. Studies have reported similarities in pathophysiological mechanisms involved in acute bone loss after fractures and osteoporosis. However, given the silence nature of bone loss and bone metabolism complexities, the actual underlying pathophysiological mechanisms have yet to be fully elucidated. AIM OF REVIEW To elaborate the latest findings in basic research with a focus on acute bone loss after fracture. To briefly highlight potential therapeutic targets and current representative drugs. To arouse researchers' attention and discussion on acute bone loss after fracture. KEY SCIENTIFIC CONCEPTS OF REVIEW Bone loss after fracture is associated with immobilization, mechanical unloading, blood supply damage, sympathetic nerve regulation, and crosstalk between musculoskeletals among other factors. Current treatment strategies rely on regulation of osteoblasts and osteoclasts, therefore, there is a need to elucidate on the underlying mechanisms of acute bone loss after fractures to inform the development of efficacious and safe drugs. In addition, attention should be paid towards ensuring long-term skeletal health.
Collapse
Affiliation(s)
- Xuan-Qi Zheng
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Jie Huang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Jia-Liang Lin
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Chun-Li Song
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China; Beijing Key Laboratory of Spinal Disease Research, Beijing, China.
| |
Collapse
|
32
|
Furrer R, Hawley JA, Handschin C. The molecular athlete: exercise physiology from mechanisms to medals. Physiol Rev 2023; 103:1693-1787. [PMID: 36603158 PMCID: PMC10110736 DOI: 10.1152/physrev.00017.2022] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 12/12/2022] [Accepted: 12/19/2022] [Indexed: 01/07/2023] Open
Abstract
Human skeletal muscle demonstrates remarkable plasticity, adapting to numerous external stimuli including the habitual level of contractile loading. Accordingly, muscle function and exercise capacity encompass a broad spectrum, from inactive individuals with low levels of endurance and strength to elite athletes who produce prodigious performances underpinned by pleiotropic training-induced muscular adaptations. Our current understanding of the signal integration, interpretation, and output coordination of the cellular and molecular mechanisms that govern muscle plasticity across this continuum is incomplete. As such, training methods and their application to elite athletes largely rely on a "trial-and-error" approach, with the experience and practices of successful coaches and athletes often providing the bases for "post hoc" scientific enquiry and research. This review provides a synopsis of the morphological and functional changes along with the molecular mechanisms underlying exercise adaptation to endurance- and resistance-based training. These traits are placed in the context of innate genetic and interindividual differences in exercise capacity and performance, with special consideration given to aging athletes. Collectively, we provide a comprehensive overview of skeletal muscle plasticity in response to different modes of exercise and how such adaptations translate from "molecules to medals."
Collapse
Affiliation(s)
| | - John A Hawley
- Exercise and Nutrition Research Program, Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Victoria, Australia
| | | |
Collapse
|
33
|
Aby K, Antony R, Li Y. ProBDNF Upregulation in Murine Hind Limb Ischemia Reperfusion Injury: A Driver of Inflammation. BIOLOGY 2023; 12:903. [PMID: 37508336 PMCID: PMC10375988 DOI: 10.3390/biology12070903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/16/2023] [Accepted: 06/22/2023] [Indexed: 07/30/2023]
Abstract
Brain-derived neurotropic factor (BDNF) has been shown to be expressed in many nonneuronal tissues including skeletal muscle. Skeletal muscle BDNF has been studied regarding its function in metabolism and exercise; however, less is known about its role in skeletal muscle injury. The precursor to BDNF, proBDNF, has an unknown role in skeletal muscle. The levels of proBDNF, mature BDNF, and their receptors were compared in the skeletal muscle and brain tissues of C57BL/6J mice. Tourniquet-induced hind limb ischemia-reperfusion injury was used to assess the function of skeletal muscle-derived proBDNF in skeletal muscle injury. Skeletal muscle-specific knockout of BDNF and pharmacological inhibition of p75NTR, the proBDNF receptor, were used to determine the role of proBDNF-p75NTR signaling. We show for the first time that proBDNF is the predominantly expressed form of BDNF in skeletal muscle and that proBDNF is significantly upregulated in skeletal muscle following hind limb ischemia-reperfusion injury. Skeletal muscle-specific knockout of BDNF blunted the inflammatory response in the injured tissue and appears to be mediated by the proBDNF-p75NTR pathway, as shown by the pharmacological inhibition of p75NTR. These findings suggest that skeletal muscle proBDNF plays a critical role in driving the inflammatory response following skeletal muscle injury.
Collapse
Affiliation(s)
| | | | - Yifan Li
- Department of Basic Biomedical Science, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA; (K.A.); (R.A.)
| |
Collapse
|
34
|
Esvald EE, Tuvikene J, Kiir CS, Avarlaid A, Tamberg L, Sirp A, Shubina A, Cabrera-Cabrera F, Pihlak A, Koppel I, Palm K, Timmusk T. Revisiting the expression of BDNF and its receptors in mammalian development. Front Mol Neurosci 2023; 16:1182499. [PMID: 37426074 PMCID: PMC10325033 DOI: 10.3389/fnmol.2023.1182499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/22/2023] [Indexed: 07/11/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) promotes the survival and functioning of neurons in the central nervous system and contributes to proper functioning of many non-neural tissues. Although the regulation and role of BDNF have been extensively studied, a rigorous analysis of the expression dynamics of BDNF and its receptors TrkB and p75NTR is lacking. Here, we have analyzed more than 3,600 samples from 18 published RNA sequencing datasets, and used over 17,000 samples from GTEx, and ~ 180 samples from BrainSpan database, to describe the expression of BDNF in the developing mammalian neural and non-neural tissues. We show evolutionarily conserved dynamics and expression patterns of BDNF mRNA and non-conserved alternative 5' exon usage. Finally, we also show increasing BDNF protein levels during murine brain development and BDNF protein expression in several non-neural tissues. In parallel, we describe the spatiotemporal expression pattern of BDNF receptors TrkB and p75NTR in both murines and humans. Collectively, our in-depth analysis of the expression of BDNF and its receptors gives insight into the regulation and signaling of BDNF in the whole organism throughout life.
Collapse
Affiliation(s)
- Eli-Eelika Esvald
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
- Protobios LLC, Tallinn, Estonia
| | - Jürgen Tuvikene
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
- Protobios LLC, Tallinn, Estonia
- dxlabs LLC, Tallinn, Estonia
| | - Carl Sander Kiir
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Annela Avarlaid
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Laura Tamberg
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Alex Sirp
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Anastassia Shubina
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | | | | | - Indrek Koppel
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | | | - Tõnis Timmusk
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
- Protobios LLC, Tallinn, Estonia
| |
Collapse
|
35
|
Fogarty MJ, Dasgupta D, Khurram OU, Sieck GC. Chemogenetic inhibition of TrkB signalling reduces phrenic motor neuron survival and size. Mol Cell Neurosci 2023; 125:103847. [PMID: 36958643 PMCID: PMC10247511 DOI: 10.1016/j.mcn.2023.103847] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 03/07/2023] [Accepted: 03/16/2023] [Indexed: 03/25/2023] Open
Abstract
Brain derived neurotrophic factor (BDNF) signalling through its high-affinity tropomyosin receptor kinase B (TrkB) is known to have potent effects on motor neuron survival and morphology during development and in neurodegenerative diseases. Here, we employed a novel 1NMPP1 sensitive TrkBF616 rat model to evaluate the effect of 14 days inhibition of TrkB signalling on phrenic motor neurons (PhMNs). Adult female and male TrkBF616 rats were divided into 1NMPP1 or vehicle treated groups. Three days prior to treatment, PhMNs in both groups were initially labeled via intrapleural injection of Alexa-Fluor-647 cholera toxin B (CTB). After 11 days of treatment, retrograde axonal uptake/transport was assessed by secondary labeling of PhMNs by intrapleural injection of Alexa-Fluor-488 CTB. After 14 days of treatment, the spinal cord was excised 100 μm thick spinal sections containing PhMNs were imaged using two-channel confocal microscopy. TrkB inhibition reduced the total number of PhMNs by ∼16 %, reduced the mean PhMN somal surface areas by ∼25 %, impaired CTB uptake 2.5-fold and reduced the estimated PhMN dendritic surface area by ∼38 %. We conclude that inhibition of TrkB signalling alone in adult TrkBF616 rats is sufficient to lead to PhMN loss, morphological degeneration and deficits in retrograde axonal uptake/transport.
Collapse
Affiliation(s)
- Matthew J Fogarty
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Debanjali Dasgupta
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Obaid U Khurram
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Gary C Sieck
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
36
|
Sleigh JN, Villarroel-Campos D, Surana S, Wickenden T, Tong Y, Simkin RL, Vargas JNS, Rhymes ER, Tosolini AP, West SJ, Zhang Q, Yang XL, Schiavo G. Boosting peripheral BDNF rescues impaired in vivo axonal transport in CMT2D mice. JCI Insight 2023; 8:e157191. [PMID: 36928301 PMCID: PMC10243821 DOI: 10.1172/jci.insight.157191] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 03/15/2023] [Indexed: 03/18/2023] Open
Abstract
Gain-of-function mutations in the housekeeping gene GARS1, which lead to the expression of toxic versions of glycyl-tRNA synthetase (GlyRS), cause the selective motor and sensory pathology characterizing Charcot-Marie-Tooth disease (CMT). Aberrant interactions between GlyRS mutants and different proteins, including neurotrophin receptor tropomyosin receptor kinase receptor B (TrkB), underlie CMT type 2D (CMT2D); however, our pathomechanistic understanding of this untreatable peripheral neuropathy remains incomplete. Through intravital imaging of the sciatic nerve, we show that CMT2D mice displayed early and persistent disturbances in axonal transport of neurotrophin-containing signaling endosomes in vivo. We discovered that brain-derived neurotrophic factor (BDNF)/TrkB impairments correlated with transport disruption and overall CMT2D neuropathology and that inhibition of this pathway at the nerve-muscle interface perturbed endosome transport in wild-type axons. Accordingly, supplementation of muscles with BDNF, but not other neurotrophins, completely restored physiological axonal transport in neuropathic mice. Together, these findings suggest that selectively targeting muscles with BDNF-boosting therapies could represent a viable therapeutic strategy for CMT2D.
Collapse
Affiliation(s)
- James N. Sleigh
- Department of Neuromuscular Diseases and UCL Queen Square Motor Neuron Disease Centre, UCL Queen Square Institute of Neurology, and
- UK Dementia Research Institute, University College London (UCL), London, United Kingdom
| | - David Villarroel-Campos
- Department of Neuromuscular Diseases and UCL Queen Square Motor Neuron Disease Centre, UCL Queen Square Institute of Neurology, and
| | - Sunaina Surana
- Department of Neuromuscular Diseases and UCL Queen Square Motor Neuron Disease Centre, UCL Queen Square Institute of Neurology, and
- UK Dementia Research Institute, University College London (UCL), London, United Kingdom
| | - Tahmina Wickenden
- Department of Neuromuscular Diseases and UCL Queen Square Motor Neuron Disease Centre, UCL Queen Square Institute of Neurology, and
| | - Yao Tong
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Rebecca L. Simkin
- Department of Neuromuscular Diseases and UCL Queen Square Motor Neuron Disease Centre, UCL Queen Square Institute of Neurology, and
| | - Jose Norberto S. Vargas
- Department of Neuromuscular Diseases and UCL Queen Square Motor Neuron Disease Centre, UCL Queen Square Institute of Neurology, and
| | - Elena R. Rhymes
- Department of Neuromuscular Diseases and UCL Queen Square Motor Neuron Disease Centre, UCL Queen Square Institute of Neurology, and
| | - Andrew P. Tosolini
- Department of Neuromuscular Diseases and UCL Queen Square Motor Neuron Disease Centre, UCL Queen Square Institute of Neurology, and
| | | | - Qian Zhang
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Xiang-Lei Yang
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Giampietro Schiavo
- Department of Neuromuscular Diseases and UCL Queen Square Motor Neuron Disease Centre, UCL Queen Square Institute of Neurology, and
- UK Dementia Research Institute, University College London (UCL), London, United Kingdom
| |
Collapse
|
37
|
Park MJ, Choi KM. Interplay of skeletal muscle and adipose tissue: sarcopenic obesity. Metabolism 2023; 144:155577. [PMID: 37127228 DOI: 10.1016/j.metabol.2023.155577] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 04/19/2023] [Accepted: 04/21/2023] [Indexed: 05/03/2023]
Abstract
Sarcopenic obesity is becoming a global health concern, owing to the rising older population, causing cardiometabolic morbidity and mortality. Loss of muscle exceeding normal age-related changes has been revealed to be associated with obesity, aggravating each other through complex interactions. Physiological regeneration and proliferation of muscle tissue are achieved through harmonious processes of regulated inflammation, autophagy, muscle satellite cell proliferation, and signaling molecule function. Adipokines and myokines are signaling molecules from adipose tissue and muscle, respectively, that exert autocrine, paracrine, and endocrine effects on fat and muscle tissues. These signaling molecules interact with each other to regulate metabolic homeostasis. However, excessive adiposity creates pro-inflammatory conditions, leading to metabolic disorders and the disorganization of systemic homeostasis. Therefore, obesity impedes muscle tissue regeneration and induces the loss of muscle mass and function. Numerous studies have attempted to demonstrate the pathophysiological interaction between sarcopenia and obesity, but the interwoven matrix of the relationship between myokines and adipokines has made it difficult for researchers to understand them. This review briefly describes updated information about the crosstalk between muscle and adipose tissue.
Collapse
Affiliation(s)
- Min Jeong Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Kyung Mook Choi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
38
|
Grzelak N, Krutki P, Bączyk M, Kaczmarek D, Mrówczyński W. Influence of altered serum and muscle concentrations of BDNF on electrophysiological properties of spinal motoneurons in wild-type and BDNF-knockout rats. Sci Rep 2023; 13:4571. [PMID: 36941445 PMCID: PMC10027728 DOI: 10.1038/s41598-023-31703-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 03/16/2023] [Indexed: 03/23/2023] Open
Abstract
The purpose of this study was to determine whether altered serum and/or muscle concentrations of brain-derived neurotrophic factor (BDNF) can modify the electrophysiological properties of spinal motoneurons (MNs). This study was conducted in wild-type and Bdnf heterozygous knockout rats (HET, SD-BDNF). Rats were divided into four groups: control, knockout, control trained, and knockout trained. The latter two groups underwent moderate-intensity endurance training to increase BDNF levels in serum and/or hindlimb muscles. BDNF and other neurotrophic factors (NFs), including glial cell-derived neurotrophic factor (GDNF), neurotrophin-3 (NT-3), nerve growth factor (NGF), and neurotrophin-4 (NT-4) were assessed in serum and three hindlimb muscles: the tibialis anterior (TA), medial gastrocnemius (MG), and soleus (Sol). The concentrations of tropomyosin kinase receptor B (Trk-B), interleukin-15 (IL-15), and myoglobin (MYO/MB) were also evaluated in these muscles. The electrophysiological properties of lumbar MNs were studied in vivo using whole-cell current-clamp recordings. Bdnf knockout rats had reduced levels of all studied NFs in serum but not in hindlimb muscles. Interestingly, decreased serum NF levels did not influence the electrophysiological properties of spinal MNs. Additionally, endurance training did not change the serum concentrations of any of the NFs tested but significantly increased BDNF and GDNF levels in the TA and MG muscles in both trained groups. Furthermore, the excitability of fast MNs was reduced in both groups of trained rats. Thus, changes in muscle (but not serum) concentrations of BDNF and GDNF may be critical factors that modify the excitability of spinal MNs after intense physical activity.
Collapse
Affiliation(s)
- Norbert Grzelak
- Department of Neurobiology, Poznań University of Physical Education, 27/39 Królowej Jadwigi St., 61-871, Poznań, Poland
| | - Piotr Krutki
- Department of Neurobiology, Poznań University of Physical Education, 27/39 Królowej Jadwigi St., 61-871, Poznań, Poland
| | - Marcin Bączyk
- Department of Neurobiology, Poznań University of Physical Education, 27/39 Królowej Jadwigi St., 61-871, Poznań, Poland
| | - Dominik Kaczmarek
- Department of Physiology and Biochemistry, Poznań University of Physical Education, Poznań, Poland
| | - Włodzimierz Mrówczyński
- Department of Neurobiology, Poznań University of Physical Education, 27/39 Królowej Jadwigi St., 61-871, Poznań, Poland.
| |
Collapse
|
39
|
Zhang L, Lv J, Wang C, Ren Y, Yong M. Myokine, a key cytokine for physical exercise to alleviate sarcopenic obesity. Mol Biol Rep 2023; 50:2723-2734. [PMID: 36571655 DOI: 10.1007/s11033-022-07821-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 06/29/2022] [Accepted: 07/24/2022] [Indexed: 12/27/2022]
Abstract
Skeletal muscle has a robust endocrine function as a powerful organ and can secrete and release cytokines or polypeptides known as myokines. These myokines have significant regulatory effects on signal transduction in skeletal muscle and the metabolism of peripheral tissues and organs and exert biological effects via autocrine, paracrine, or endocrine forms. Obesity and aging cause myokine secretion dysregulation, and hastening sarcopenic obesity (SO) development. Exercise is currently an excellent intervention and prevention method for SO. Meanwhile, exercise impacts many organs and tissues. These organs and tissues will produce various myokines in response to movement and metabolism throughout the body to govern muscle differentiation, growth, and remodeling. According to accumulating data, exercise can increase the release of myokines from diverse tissues into the blood and postpone the SO onset and progression by influencing protein metabolism, inflammation, mitochondrial quality control, and other mechanisms.
Collapse
Affiliation(s)
- Lei Zhang
- Physical Education and Sport Science, Soochow University, Suzhou, Jiangsu Province, China
| | - Junjie Lv
- Department of Sport, Physical Education and Health, Hong Kong Baptist University, Hong Kong, China
| | - Cenyi Wang
- Physical Education and Sport Science, Soochow University, Suzhou, Jiangsu Province, China
| | - Yuanyuan Ren
- Physical Education and Sport Science, Soochow University, Suzhou, Jiangsu Province, China.
| | - Ming Yong
- Physical Education and Sport Science, Soochow University, Suzhou, Jiangsu Province, China.
| |
Collapse
|
40
|
The Preventive Effect of Specific Collagen Peptides against Dexamethasone-Induced Muscle Atrophy in Mice. Molecules 2023; 28:molecules28041950. [PMID: 36838938 PMCID: PMC9960993 DOI: 10.3390/molecules28041950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/13/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
Muscle atrophy, also known as muscle wasting, is the thinning of muscle mass due to muscle disuse, aging, or diseases such as cancer or neurological problems. Muscle atrophy is closely related to the quality of life and has high morbidity and mortality. However, therapeutic options for muscle atrophy are limited, so studies to develop therapeutic agents for muscle loss are always required. For this study, we investigated how orally administered specific collagen peptides (CP) affect muscle atrophy and elucidated its molecular mechanism using an in vivo model. We treated mice with dexamethasone (DEX) to induce a muscular atrophy phenotype and then administered CP (0.25 and 0.5 g/kg) for four weeks. In a microcomputed tomography analysis, CP (0.5 g/kg) intake significantly increased the volume of calf muscles in mice with DEX-induced muscle atrophy. In addition, the administration of CP (0.25 and 0.5 g/kg) restored the weight of the gluteus maximus and the fiber cross-sectional area (CSA) of the pectoralis major and calf muscles, which were reduced by DEX. CP significantly inhibited the mRNA expression of myostatin and the phosphorylation of Smad2, but it did not affect TGF-β, BDNF, or FNDC5 gene expression. In addition, AKT/mTOR, a central pathway for muscle protein synthesis and related to myostatin signaling, was enhanced in the groups that were administered CP. Finally, CP decreased serum albumin levels and increased TNF-α gene expression. Collectively, our in vivo results demonstrate that CP can alleviate muscle wasting through a multitude of mechanisms. Therefore, we propose CP as a supplement or treatment to prevent muscle atrophy.
Collapse
|
41
|
Moosaie F, Mohammadi S, Saghazadeh A, Dehghani Firouzabadi F, Rezaei N. Brain-derived neurotrophic factor in diabetes mellitus: A systematic review and meta-analysis. PLoS One 2023; 18:e0268816. [PMID: 36787304 PMCID: PMC9928073 DOI: 10.1371/journal.pone.0268816] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 03/10/2022] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND Brain-derived neurotrophic factor (BDNF) is a neurotrophic factor expressed in several tissues, including the brain, gut, and pancreas. Activation of the BDNF/TrkB/CREB reduces hepatic gluconeogenesis, induces hepatic insulin signal transduction, and protects against pancreatic beta-cell loss in diabetes mellitus (DM). Several studies have investigated the possible association between BDNF and DM and its complications, but the results have been conflicting. AIM In the present study, we aimed at systematically reviewing the literature on the serum and plasma levels of BDNF in DM and its subgroups such as T2DM, DM patients with depression, and patients with retinopathy. METHODS A comprehensive search was conducted in PubMed, Scopus, and Web of Science. We identified 28 eligible studies and calculated the standardized mean difference (SMD) of outcomes as an effect measure. RESULTS The meta-analysis included 2734 patients with DM and 6004 controls. Serum BDNF levels were significantly lower in patients with DM vs. controls (SMD = -1.00, P<0.001). Plasma BDNF levels were not different in patients with DM compared with controls. When conducting subgroup analysis, serum BDNF levels were lower among patients with T2DM (SMD = -1.26, P<0.001), DM and depression (SMD = -1.69, P<0.001), and patients with diabetic retinopathy (DR) vs. controls (SMD = -1.03, P = 0.01). CONCLUSIONS Serum BDNF levels were lower in patients with DM, T2DM, DM with depression, and DM and DR than the controls. Our findings are in line with the hypothesis that decreased BDNF levels might impair glucose metabolism and contribute to the pathogenesis of DM and its complications.
Collapse
Affiliation(s)
- Fatemeh Moosaie
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran University of Medical Sciences, Tehran, Iran
| | - Soheil Mohammadi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran University of Medical Sciences, Tehran, Iran
| | - Amene Saghazadeh
- Research Center for Immunodeficiencies, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- MetaCognition Interest Group (MCIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Fatemeh Dehghani Firouzabadi
- Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- MetaCognition Interest Group (MCIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
42
|
Sirago G, Pellegrino MA, Bottinelli R, Franchi MV, Narici MV. Loss of neuromuscular junction integrity and muscle atrophy in skeletal muscle disuse. Ageing Res Rev 2023; 83:101810. [PMID: 36471545 DOI: 10.1016/j.arr.2022.101810] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 11/25/2022] [Accepted: 11/25/2022] [Indexed: 11/27/2022]
Abstract
Physical inactivity (PI) is a major risk factor of chronic diseases. A major aspect of PI is loss of muscle mass and strength. The latter phenomenon significantly impacts daily life and represent a major issue for global health. Understandably, skeletal muscle itself has been the major focus of studies aimed at understanding the mechanisms underlying loss of mass and strength. Relatively lesser attention has been given to the contribution of alterations in somatomotor control, despite the fact that these changes can start very early and can occur at multiple levels, from the cortex down to the neuromuscular junction (NMJ). It is well known that exposure to chronic inactivity or immobilization causes a disproportionate loss of force compared to muscle mass, i.e. a loss of specific or intrinsic whole muscle force. The latter phenomenon may be partially explained by the loss of specific force of individual muscle fibres, but several other players are very likely to contribute to such detrimental phenomenon. Irrespective of the length of the disuse period, the loss of force is, in fact, more than two-fold greater than that of muscle size. It is very likely that somatomotor alterations may contribute to this loss in intrinsic muscle force. Here we review evidence that alterations of one component of somatomotor control, namely the neuromuscular junction, occur in disuse. We also discuss some of the novel players in NMJ stability (e.g., homer, bassoon, pannexin) and the importance of new established and emerging molecular markers of neurodegenerative processes in humans such as agrin, neural-cell adhesion molecule and light-chain neurofilaments.
Collapse
Affiliation(s)
- Giuseppe Sirago
- Department of Biomedical Sciences, University of Padova, Padova 35131, Italy.
| | - Maria A Pellegrino
- Department of Molecular Medicine, University of Pavia, Pavia 27100, Italy
| | - Roberto Bottinelli
- Department of Molecular Medicine, University of Pavia, Pavia 27100, Italy; IRCCS Mondino Foundation, Pavia 27100, Italy
| | - Martino V Franchi
- Department of Biomedical Sciences, University of Padova, Padova 35131, Italy
| | - Marco V Narici
- Department of Biomedical Sciences, University of Padova, Padova 35131, Italy; CIR-MYO Myology Center, University of Padova, Padova 35131, Italy.
| |
Collapse
|
43
|
Galindo C, Nguyen VT, Hill B, Sims N, Heck A, Negron M, Lusk C. Brain-derived neurotrophic factor rs6265 (Val66Met) single nucleotide polymorphism as a master modifier of human pathophysiology. Neural Regen Res 2023. [PMID: 35799516 PMCID: PMC9241394 DOI: 10.4103/1673-5374.343894] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Brain-derived neurotrophic factor is the most prevalent member of the nerve growth factor family. Since its discovery in 1978, this enigmatic molecule has spawned more than 27,000 publications, most of which are focused on neurological disorders. Brain-derived neurotrophic factor is indispensable during embryogenesis and postnatally for the normal development and function of both the central and peripheral nervous systems. It is becoming increasingly clear, however, that brain-derived neurotrophic factor likewise plays crucial roles in a variety of other biological functions independently of sympathetic or parasympathetic involvement. Brain-derived neurotrophic factor is also increasingly recognized as a sophisticated environmental sensor and master coordinator of whole organismal physiology. To that point, we recently found that a common nonsynonymous (Val66→Met) single nucleotide polymorphism in the brain-derived neurotrophic factor gene (rs6265) not only substantially alters basal cardiac transcriptomics in mice but subtly influences heart gene expression and function differentially in males and females. In addition to a short description of recent results from associative neuropsychiatric studies, this review provides an eclectic assortment of research reports that support a modulatory role for rs6265 including and beyond the central nervous system.
Collapse
|
44
|
Lucon-Xiccato T, Tomain M, D’Aniello S, Bertolucci C. bdnf loss affects activity, sociability, and anxiety-like behaviour in zebrafish. Behav Brain Res 2023; 436:114115. [DOI: 10.1016/j.bbr.2022.114115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 08/18/2022] [Accepted: 09/11/2022] [Indexed: 10/14/2022]
|
45
|
Just-Borràs L, Cilleros-Mañé V, Polishchuk A, Balanyà-Segura M, Tomàs M, Garcia N, Tomàs J, Lanuza MA. TrkB signaling is correlated with muscular fatigue resistance and less vulnerability to neurodegeneration. Front Mol Neurosci 2022; 15:1069940. [PMID: 36618825 PMCID: PMC9813967 DOI: 10.3389/fnmol.2022.1069940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022] Open
Abstract
At the neuromuscular junction (NMJ), motor neurons and myocytes maintain a bidirectional communication that guarantees adequate functionality. Thus, motor neurons' firing pattern, which is influenced by retrograde muscle-derived neurotrophic factors, modulates myocyte contractibility. Myocytes can be fast-twitch fibers and become easily fatigued or slow-twitch fibers and resistant to fatigue. Extraocular muscles (EOM) show mixed properties that guarantee fast contraction speed and resistance to fatigue and the degeneration caused by Amyotrophic lateral sclerosis (ALS) disease. The TrkB signaling is an activity-dependent pathway implicated in the NMJ well-functioning. Therefore, it could mediate the differences between fast and slow myocytes' resistance to fatigue. The present study elucidates a specific protein expression profile concerning the TrkB signaling that correlates with higher resistance to fatigue and better neuroprotective capacity through time. The results unveil that Extra-ocular muscles (EOM) express lower levels of NT-4 that extend TrkB signaling, differential PKC expression, and a higher abundance of phosphorylated synaptic proteins that correlate with continuous neurotransmission requirements. Furthermore, common molecular features between EOM and slow soleus muscles including higher neurotrophic consumption and classic and novel PKC isoforms balance correlate with better preservation of these two muscles in ALS. Altogether, higher resistance of Soleus and EOM to fatigue and ALS seems to be associated with specific protein levels concerning the TrkB neurotrophic signaling.
Collapse
|
46
|
Promoter-Adjacent DNA Hypermethylation Can Downmodulate Gene Expression: TBX15 in the Muscle Lineage. EPIGENOMES 2022; 6:epigenomes6040043. [PMID: 36547252 PMCID: PMC9778270 DOI: 10.3390/epigenomes6040043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/01/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
TBX15, which encodes a differentiation-related transcription factor, displays promoter-adjacent DNA hypermethylation in myoblasts and skeletal muscle (psoas) that is absent from non-expressing cells in other lineages. By whole-genome bisulfite sequencing (WGBS) and enzymatic methyl-seq (EM-seq), these hypermethylated regions were found to border both sides of a constitutively unmethylated promoter. To understand the functionality of this DNA hypermethylation, we cloned the differentially methylated sequences (DMRs) in CpG-free reporter vectors and tested them for promoter or enhancer activity upon transient transfection. These cloned regions exhibited strong promoter activity and, when placed upstream of a weak promoter, strong enhancer activity specifically in myoblast host cells. In vitro CpG methylation targeted to the DMR sequences in the plasmids resulted in 86−100% loss of promoter or enhancer activity, depending on the insert sequence. These results as well as chromatin epigenetic and transcription profiles for this gene in various cell types support the hypothesis that DNA hypermethylation immediately upstream and downstream of the unmethylated promoter region suppresses enhancer/extended promoter activity, thereby downmodulating, but not silencing, expression in myoblasts and certain kinds of skeletal muscle. This promoter-border hypermethylation was not found in cell types with a silent TBX15 gene, and these cells, instead, exhibit repressive chromatin in and around the promoter. TBX18, TBX2, TBX3 and TBX1 display TBX15-like hypermethylated DMRs at their promoter borders and preferential expression in myoblasts. Therefore, promoter-adjacent DNA hypermethylation for downmodulating transcription to prevent overexpression may be used more frequently for transcription regulation than currently appreciated.
Collapse
|
47
|
Takasawa S, Shobatake R, Itaya‐Hironaka A, Makino M, Uchiyama T, Sakuramoto‐Tsuchida S, Takeda Y, Ota H, Yamauchi A. Upregulation of IL-8, osteonectin, and myonectin mRNAs by intermittent hypoxia via OCT1- and NRF2-mediated mechanisms in skeletal muscle cells. J Cell Mol Med 2022; 26:6019-6031. [PMID: 36457269 PMCID: PMC9753449 DOI: 10.1111/jcmm.17618] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 10/21/2022] [Accepted: 11/02/2022] [Indexed: 12/04/2022] Open
Abstract
Sleep apnoea syndrome is characterized by recurrent episodes of oxygen desaturation and reoxygenation (intermittent hypoxia [IH]) and is a risk factor for insulin resistance/Type 2 diabetes. The induction of insulin resistance in skeletal muscle is a key phenomenon to develop diabetes. However, the mechanisms linking IH stress and insulin resistance remain elusive. We exposed human RD and mouse C2C12 muscle cells to normoxia or IH and measured their mRNA levels by real-time RT-PCR. We found that IH significantly increased the mRNA and protein levels of muscle-derived insulin resistance-factors (myokines) such as IL-8, osteonectin (ON), and myonectin (MN) in muscle cells. We further analysed the IH-induced expression mechanisms of IL-8, ON, and MN genes in muscle cells. Deletion analyses of the human myokine promoter(s) revealed that the regions -152 to -151 in IL-8, -105 to -99 in ON, and - 3741 to -3738 in MN promoters were responsible for the activation by IH in RD cells. The promoters contain consensus transcription factor binding sequences for OCT1 in IL-8 and MN promoters, and for NRF2 in ON promoter, respectively. The introduction of siRNA for OCT1 abolished the IH-induced expression(s) of IL-8 and MN and siRNA for NRF2 abolished the IH-induced expression of ON.
Collapse
Affiliation(s)
- Shin Takasawa
- Department of BiochemistryNara Medical UniversityNaraJapan
| | - Ryogo Shobatake
- Department of BiochemistryNara Medical UniversityNaraJapan,Department of NeurologyNara Medical UniversityNaraJapan,Department of NeurologyNara City HospitalNaraJapan
| | | | - Mai Makino
- Department of BiochemistryNara Medical UniversityNaraJapan
| | - Tomoko Uchiyama
- Department of BiochemistryNara Medical UniversityNaraJapan,Department of Diagnostic PathologyNara Medical UniversityNaraJapan
| | | | | | - Hiroyo Ota
- Department of BiochemistryNara Medical UniversityNaraJapan,Department of Respiratory MedicineNara Medical UniversityNaraJapan
| | - Akiyo Yamauchi
- Department of BiochemistryNara Medical UniversityNaraJapan
| |
Collapse
|
48
|
BDNF Spinal Overexpression after Spinal Cord Injury Partially Protects Soleus Neuromuscular Junction from Disintegration, Increasing VAChT and AChE Transcripts in Soleus but Not Tibialis Anterior Motoneurons. Biomedicines 2022; 10:biomedicines10112851. [DOI: 10.3390/biomedicines10112851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/20/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
After spinal cord transection (SCT) the interaction between motoneurons (MNs) and muscle is impaired, due to reorganization of the spinal network after a loss of supraspinal inputs. Rats subjected to SCT, treated with intraspinal injection of a AAV-BDNF (brain-derived neurotrophic factor) construct, partially regained the ability to walk. The central effects of this treatment have been identified, but its impact at the neuromuscular junction (NMJ) has not been characterized. Here, we compared the ability of NMJ pre- and postsynaptic machinery in the ankle extensor (Sol) and flexor (TA) muscles to respond to intraspinal AAV-BDNF after SCT. The gene expression of cholinergic molecules (VAChT, ChAT, AChE, nAChR, mAChR) was investigated in tracer-identified, microdissected MN perikarya, and in muscle fibers with the use of qPCR. In the NMJs, a distribution of VAChT, nAChR and Schwann cells was studied by immunofluorescence, and of synaptic vesicles and membrane active zones by electron microscopy. We showed partial protection of the Sol NMJs from disintegration, and upregulation of the VAChT and AChE transcripts in the Sol, but not the TA MNs after spinal enrichment with BDNF. We propose that the observed discrepancy in response to BDNF treatment is an effect of difference in the TrkB expression setting BDNF responsiveness, and of BDNF demands in Sol and TA muscles.
Collapse
|
49
|
Minniti G, Pescinini-Salzedas LM, Minniti GADS, Laurindo LF, Barbalho SM, Vargas Sinatora R, Sloan LA, Haber RSDA, Araújo AC, Quesada K, Haber JFDS, Bechara MD, Sloan KP. Organokines, Sarcopenia, and Metabolic Repercussions: The Vicious Cycle and the Interplay with Exercise. Int J Mol Sci 2022; 23:13452. [PMID: 36362238 PMCID: PMC9655425 DOI: 10.3390/ijms232113452] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 10/26/2022] [Accepted: 10/30/2022] [Indexed: 11/06/2022] Open
Abstract
Sarcopenia is a disease that becomes more prevalent as the population ages, since it is directly linked to the process of senility, which courses with muscle atrophy and loss of muscle strength. Over time, sarcopenia is linked to obesity, being known as sarcopenic obesity, and leads to other metabolic changes. At the molecular level, organokines act on different tissues and can improve or harm sarcopenia. It all depends on their production process, which is associated with factors such as physical exercise, the aging process, and metabolic diseases. Because of the seriousness of these repercussions, the aim of this literature review is to conduct a review on the relationship between organokines, sarcopenia, diabetes, and other metabolic repercussions, as well the role of physical exercise. To build this review, PubMed-Medline, Embase, and COCHRANE databases were searched, and only studies written in English were included. It was observed that myokines, adipokines, hepatokines, and osteokines had direct impacts on the pathophysiology of sarcopenia and its metabolic repercussions. Therefore, knowing how organokines act is very important to know their impacts on age, disease prevention, and how they can be related to the prevention of muscle loss.
Collapse
Affiliation(s)
- Giulia Minniti
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marilia 17525-902, SP, Brazil
| | | | | | - Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marilia 17525-902, SP, Brazil
| | - Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marilia 17525-902, SP, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marilia 17525-902, SP, Brazil
- School of Food and Technology of Marilia (FATEC), Marilia 17506-000, SP, Brazil
| | - Renata Vargas Sinatora
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marilia 17525-902, SP, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marilia 17525-902, SP, Brazil
| | - Lance Alan Sloan
- Texas Institute for Kidney and Endocrine Disorders, Lufkin, TX 75904, USA
- Department of Internal Medicine, University of Texas Medical Branch-Galveston, Galveston, TX 75904, USA
| | - Rafael Santos de Argollo Haber
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marilia 17525-902, SP, Brazil
| | - Adriano Cressoni Araújo
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marilia 17525-902, SP, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marilia 17525-902, SP, Brazil
| | - Karina Quesada
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marilia 17525-902, SP, Brazil
| | - Jesselina F. dos Santos Haber
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marilia 17525-902, SP, Brazil
| | - Marcelo Dib Bechara
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marilia 17525-902, SP, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marilia 17525-902, SP, Brazil
| | | |
Collapse
|
50
|
Kuhnen G, Guedes Russomanno T, Murgia M, Pillon NJ, Schönfelder M, Wackerhage H. Genes Whose Gain or Loss of Function Changes Type 1, 2A, 2X, or 2B Muscle Fibre Proportions in Mice—A Systematic Review. Int J Mol Sci 2022; 23:ijms232112933. [PMID: 36361732 PMCID: PMC9658117 DOI: 10.3390/ijms232112933] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/16/2022] [Accepted: 10/18/2022] [Indexed: 11/25/2022] Open
Abstract
Adult skeletal muscle fibres are classified as type 1, 2A, 2X, and 2B. These classifications are based on the expression of the dominant myosin heavy chain isoform. Muscle fibre-specific gene expression and proportions of muscle fibre types change during development and in response to exercise, chronic electrical stimulation, or inactivity. To identify genes whose gain or loss-of-function alters type 1, 2A, 2X, or 2B muscle fibre proportions in mice, we conducted a systematic review of transgenic mouse studies. The systematic review was conducted in accordance with the 2009 PRISMA guidelines and the PICO framework. We identified 25 “muscle fibre genes” (Akirin1, Bdkrb2, Bdnf, Camk4, Ccnd3, Cpt1a, Epas1, Esrrg, Foxj3, Foxo1, Il15, Mapk12, Mstn, Myod1, Ncor1, Nfatc1, Nol3, Ppargc1a, Ppargc1b, Sirt1, Sirt3, Thra, Thrb, Trib3, and Vgll2) whose gain or loss-of-function significantly changes type 1, 2A, 2X or 2B muscle fibre proportions in mice. The fact that 15 of the 25 muscle fibre genes are transcriptional regulators suggests that muscle fibre-specific gene expression is primarily regulated transcriptionally. A reanalysis of existing datasets revealed that the expression of Ppargc1a and Vgll2 increases and Mstn decreases after exercise, respectively. This suggests that these genes help to regulate the muscle fibre adaptation to exercise. Finally, there are many known DNA sequence variants of muscle fibre genes. It seems likely that such DNA sequence variants contribute to the large variation of muscle fibre type proportions in the human population.
Collapse
Affiliation(s)
- Gabryela Kuhnen
- Department of Sports and Health Sciences, Technical University of Munich, 80809 Munich, Germany
| | - Tiago Guedes Russomanno
- Department of Sports and Health Sciences, Technical University of Munich, 80809 Munich, Germany
| | - Marta Murgia
- Max Planck Institute, Martinsried, 82152 Munich, Germany
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi, 58/B, 35131 Padua, Italy
| | - Nicolas J Pillon
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Martin Schönfelder
- Department of Sports and Health Sciences, Technical University of Munich, 80809 Munich, Germany
| | - Henning Wackerhage
- Department of Sports and Health Sciences, Technical University of Munich, 80809 Munich, Germany
| |
Collapse
|