High-Temperature Stress Induces Autophagy in Rainbow Trout Skeletal Muscle

Ectothermic animals, such as teleosts, have increasingly been exposed to stressful high-temperature events due to global warming. Currently, the effects of thermal stress on skeletal muscle, a key tissue for fish growth, are unknown. This study examined the impact of high-temperature stress on the skeletal muscle transcriptome of rainbow trout (Oncorhynchus mykiss) in control (15 °C) and high-temperature (20 °C) conditions. Additionally, we examined the plasmatic levels of cortisol, glucose, and creatine kinase activity, and examined oxidative damage and autophagy activation in skeletal muscle. High-temperature stress induced significant increases in cortisol and glucose plasmatic levels. Nevertheless, no changes were observed in creatine kinase activity in plasma and skeletal muscle oxidation. Skeletal muscle RNA was isolated and sequenced using the HiSeq Illumina platform. A total of 383,796,290 reads were mapped onto the reference rainbow trout genome. The transcriptomic analysis showed that 293 genes were upregulated in the high-temperature group, mainly associated with autophagosome assembly, amino acid transport, and the glutamine metabolic process. On the other hand, 119 genes were downregulated in the high-temperature group, mainly associated with digestion, proteolysis, and the muscle contraction process. In addition, RT-qPCR of differentially expressed representative genes and Western blot analysis of LC3-II/LC3-I levels confirmed skeletal muscle autophagy induced by high temperature. This study sheds light on intriguing facets of the adaptive response of rainbow trout skeletal muscle to high-temperature stress and provides significant insights into the physiology of autophagy in teleosts.

[1]  T. Ventura,et al.  Histological and transcriptomic analysis of muscular atrophy associated with depleted flesh pigmentation in Atlantic salmon (Salmo salar) exposed to elevated seawater temperatures , 2023, Scientific Reports.

[2]  K. Palikaras,et al.  The Role of Mitophagy in Skeletal Muscle Damage and Regeneration , 2023, Cells.

[3]  M. Sojka,et al.  How Climate Change Affects River and Lake Water Temperature in Central-West Poland—A Case Study of the Warta River Catchment , 2023, Atmosphere.

[4]  Hongfu Zhang,et al.  Role of Mitophagy in Regulating Intestinal Oxidative Damage , 2023, Antioxidants.

[5]  J. Aedo,et al.  Role of glucocorticoid and mineralocorticoid receptors in rainbow trout (Oncorhynchus mykiss) skeletal muscle: A transcriptomic perspective of cortisol action , 2023, Frontiers in Physiology.

[6]  Hong-Wen Tang,et al.  The Importance of mTORC1-Autophagy Axis for Skeletal Muscle Diseases , 2022, International journal of molecular sciences.

[7]  Jinfeng Shi,et al.  High temperature induced metabolic reprogramming and lipid remodeling in a high-altitude fish species, Triplophysa bleekeri , 2022, Frontiers in Marine Science.

[8]  P. Dettleff,et al.  High-Temperature Stress Effect on the Red Cusk-Eel (Geypterus chilensis) Liver: Transcriptional Modulation and Oxidative Stress Damage , 2022, Biology.

[9]  Peifeng Li,et al.  Autophagy regulation in teleost fish: A double-edged sword , 2022, Aquaculture.

[10]  Yushi Chen,et al.  Autophagy in muscle regeneration: potential therapies for myopathies , 2022, Journal of cachexia, sarcopenia and muscle.

[11]  Muntasir Murshed,et al.  A review of the global climate change impacts, adaptation, and sustainable mitigation measures , 2022, Environmental Science and Pollution Research.

[12]  P. Tatsapong,et al.  Effects of long-term exposure to high temperature on growth performance, chemical composition, hematological and histological changes, and physiological responses in hybrid catfish [♂Clarias gariepinus (Burchell, 1822) ×♀C. macrocephalus (Günther, 1864)]. , 2022, Journal of thermal biology.

[13]  A. Rivas-Aravena,et al.  RNA-seq analysis reveals the dynamic regulation of proteasomal and autophagic degradation systems of rainbow trout (Oncorhynchus mykiss) skeletal muscle challenged with infectious pancreatic necrosis virus (IPNV) , 2022, Aquaculture.

[14]  P. Currie,et al.  Dynamics of muscle growth and regeneration: Lessons from the teleost. , 2021, Experimental cell research.

[15]  E. Jewett,et al.  An Overview of Ocean Climate Change Indicators: Sea Surface Temperature, Ocean Heat Content, Ocean pH, Dissolved Oxygen Concentration, Arctic Sea Ice Extent, Thickness and Volume, Sea Level and Strength of the AMOC (Atlantic Meridional Overturning Circulation) , 2021, Frontiers in Marine Science.

[16]  Lan Zheng,et al.  Identification of Potentially Related Genes and Mechanisms Involved in Skeletal Muscle Atrophy Induced by Excessive Exercise in Zebrafish , 2021, Biology.

[17]  X. Ju,et al.  Effect of heat stress on growth and production performance of livestock and poultry: Mechanism to prevention. , 2021, Journal of thermal biology.

[18]  Yonten Chophel Global Warming and Climate Change (GWCC) Realities , 2021, Global Warming and Climate Change [Working Title].

[19]  O. Carnevali,et al.  Effects of Di-Isononyl Phthalate (DiNP) on Follicular Atresia in Zebrafish Ovary , 2021, Frontiers in Endocrinology.

[20]  R. Srikuea,et al.  Autophagy-lysosomal signaling responses to heat stress in tenotomy-induced rat skeletal muscle atrophy. , 2021, Life sciences.

[21]  Jinmiao Zha,et al.  Carbamazepine at environmentally relevant concentrations caused DNA damage and apoptosis in the liver of Chinese rare minnows (Gobiocypris rarus) by the Ras/Raf/ERK/p53 signaling pathway. , 2020, Environmental pollution.

[22]  B. Sadoul,et al.  Temperature increase and its effects on fish stress physiology in the context of global warming. , 2020, Journal of fish biology.

[23]  P. Dettleff,et al.  Physiological and molecular responses to thermal stress in red cusk-eel (Genypterus chilensis) juveniles reveals atrophy and oxidative damage in skeletal muscle. , 2020, Journal of thermal biology.

[24]  Z. Qin,et al.  Escherichia coli induced ferroptosis in red blood cells of grass carp (Ctenopharyngodon idella). , 2020, Fish & shellfish immunology.

[25]  Zhenyan Dai,et al.  Exposure to waterborne cadmium induce oxidative stress, autophagy and mitochondrial dysfunction in the liver of Procypris merus. , 2020, Ecotoxicology and environmental safety.

[26]  Hanhua Cheng,et al.  Srag Regulates Autophagy via Integrating into a Preexisting Autophagy Pathway in Testis , 2020, Molecular biology and evolution.

[27]  T. Choi,et al.  Roles of Autophagy in Oxidative Stress , 2020, International journal of molecular sciences.

[28]  A. Zachariou,et al.  Energetic, antioxidant, inflammatory and cell death responses in the red muscle of thermally stressed Sparus aurata , 2020, Journal of Comparative Physiology B.

[29]  Feng Xie,et al.  Bnip3 in mitophagy: novel insights and potential therapeutic target for diseases of secondary mitochondrial dysfunction. , 2020, Clinica chimica acta; international journal of clinical chemistry.

[30]  R. Valentine,et al.  Acute Heat Exposure Alters Autophagy Signaling in C2C12 Myotubes , 2020, Frontiers in Physiology.

[31]  C. Meneses,et al.  Early transcriptomic responses associated with the membrane-initiated action of cortisol in the skeletal muscle of rainbow trout (Oncorhynchus mykiss). , 2019, Physiological genomics.

[32]  Bin Wang,et al.  Historical change of El Niño properties sheds light on future changes of extreme El Niño , 2019, Proceedings of the National Academy of Sciences.

[33]  Hafiz M.N. Iqbal,et al.  A Comprehensive Review of Autophagy and Its Various Roles in Infectious, Non-Infectious, and Lifestyle Diseases: Current Knowledge and Prospects for Disease Prevention, Novel Drug Design, and Therapy , 2019, Cells.

[34]  A. Rivas-Aravena,et al.  Transcriptomic response of rainbow trout (Oncorhynchus mykiss) skeletal muscle to Flavobacterium psychrophilum. , 2019, Comparative biochemistry and physiology. Part D, Genomics & proteomics.

[35]  H. Wen,et al.  Identification of mapk gene family in Lateolabrax maculatus and their expression profiles in response to hypoxia and salinity challenges. , 2019, Gene.

[36]  A. Meijer,et al.  Macrophages target Salmonella by Lc3-associated phagocytosis in a systemic infection model , 2019, Autophagy.

[37]  P. Orozco‐terWengel,et al.  Contrasting effects of acute and chronic stress on the transcriptome, epigenome, and immune response of Atlantic salmon , 2018, bioRxiv.

[38]  L. Baumgard,et al.  Short-term heat stress results in increased apoptotic signaling and autophagy in oxidative skeletal muscle in Sus scrofa. , 2018, Journal of thermal biology.

[39]  E. Reddy,et al.  JNK-signaling: A multiplexing hub in programmed cell death , 2017, Genes & cancer.

[40]  A. Ballabio,et al.  Molecular definitions of autophagy and related processes , 2017, The EMBO journal.

[41]  M. I. Borella,et al.  Involution processes of follicular atresia and post-ovulatory complex in a characid fish ovary: a study of apoptosis and autophagy pathways , 2017, Journal of Molecular Histology.

[42]  H. Donninger,et al.  Ras signaling through RASSF proteins. , 2016, Seminars in cell & developmental biology.

[43]  M. Vijayan,et al.  Mechanisms of cortisol action in fish hepatocytes. , 2016, Comparative biochemistry and physiology. Part B, Biochemistry & molecular biology.

[44]  B. Salin,et al.  Looking at the metabolic consequences of the colchicine-based in vivo autophagic flux assay , 2016, Autophagy.

[45]  H. Silva,et al.  mRNA-seq reveals skeletal muscle atrophy in response to handling stress in a marine teleost, the red cusk-eel (Genypterus chilensis) , 2015, BMC Genomics.

[46]  L. Mercado,et al.  Transcriptional dynamics of immune, growth and stress related genes in skeletal muscle of the fine flounder (Paralichthys adpersus) during different nutritional statuses. , 2015, Developmental and comparative immunology.

[47]  Daisuke Hoshino,et al.  Daily heat stress treatment rescues denervation‐activated mitochondrial clearance and atrophy in skeletal muscle , 2015, The Journal of physiology.

[48]  N. P. Sahu,et al.  Biochemical and Physiological Stress Responses to Heat Shock and Their Recovery in Labeo rohita Fingerlings , 2015, Proceedings of the National Academy of Sciences, India Section B: Biological Sciences.

[49]  D. Klionsky,et al.  Transcriptional regulation of ATG9 by the Pho23-Rpd3 complex modulates the frequency of autophagosome formation , 2014, Autophagy.

[50]  E. Jaimes,et al.  Mitochondria and Reactive Oxygen Species: Physiology and Pathophysiology , 2013, International journal of molecular sciences.

[51]  Kevin W Eliceiri,et al.  NIH Image to ImageJ: 25 years of image analysis , 2012, Nature Methods.

[52]  I. Johnston,et al.  Growth and the regulation of myotomal muscle mass in teleost fish , 2011, Journal of Experimental Biology.

[53]  K. Gilmour,et al.  Chronic social stress impairs thermal tolerance in the rainbow trout (Oncorhynchus mykiss) , 2011, Journal of Experimental Biology.

[54]  N. W. Pankhurst,et al.  The endocrinology of stress in fish: an environmental perspective. , 2011, General and comparative endocrinology.

[55]  V. Beneš,et al.  The MIQE guidelines: minimum information for publication of quantitative real-time PCR experiments. , 2009, Clinical chemistry.

[56]  S. C. Johnson,et al.  The stress and metabolic responses of juvenile Atlantic cod Gadus morhua L. to an acute thermal challenge , 2008 .

[57]  G. Iwama,et al.  The effects of cortisol on heat shock protein 70 levels in two fish species. , 2001, General and comparative endocrinology.

[58]  B. Björnsson,et al.  Chronic stress inhibits growth and induces proteolytic mechanisms through two different nonoverlapping pathways in the skeletal muscle of a teleost fish. , 2018, American journal of physiology. Regulatory, integrative and comparative physiology.

[59]  Mathilakath M. Vijayan,et al.  Stress and Growth , 2016 .

[60]  K. Nislow,et al.  Thermal onset of cellular and endocrine stress responses correspond to ecological limits in brook trout, an iconic cold-water fish. , 2015, Conservation physiology.