Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Nucleic Acids Res ; 52(7): 4002-4020, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38321934

ABSTRACT

Poly(ADP-ribosylation) (PARylation) is a post-translational modification mediated by a subset of ADP-ribosyl transferases (ARTs). Although PARylation-inhibition based therapies are considered as an avenue to combat debilitating diseases such as cancer and myopathies, the role of this modification in physiological processes such as cell differentiation remains unclear. Here, we show that Tankyrase1 (TNKS1), a PARylating ART, plays a major role in myogenesis, a vital process known to drive muscle fiber formation and regeneration. Although all bona fide PARPs are expressed in muscle cells, experiments using siRNA-mediated knockdown or pharmacological inhibition show that TNKS1 is the enzyme responsible of catalyzing PARylation during myogenesis. Via this activity, TNKS1 controls the turnover of mRNAs encoding myogenic regulatory factors such as nucleophosmin (NPM) and myogenin. TNKS1 mediates these effects by targeting RNA-binding proteins such as Human Antigen R (HuR). HuR harbors a conserved TNKS-binding motif (TBM), the mutation of which not only prevents the association of HuR with TNKS1 and its PARylation, but also precludes HuR from regulating the turnover of NPM and myogenin mRNAs as well as from promoting myogenesis. Therefore, our data uncover a new role for TNKS1 as a key modulator of RBP-mediated post-transcriptional events required for vital processes such as myogenesis.


Subject(s)
Muscle Development , Muscle Fibers, Skeletal , Myogenin , RNA, Messenger , Tankyrases , Tankyrases/metabolism , Tankyrases/genetics , Humans , RNA, Messenger/metabolism , RNA, Messenger/genetics , Muscle Development/genetics , Animals , Muscle Fibers, Skeletal/metabolism , Mice , Myogenin/genetics , Myogenin/metabolism , Nucleophosmin , ELAV-Like Protein 1/metabolism , ELAV-Like Protein 1/genetics , RNA Stability/genetics , Poly ADP Ribosylation/genetics , Cell Line , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Cell Differentiation/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , HEK293 Cells
2.
EMBO Mol Med ; 13(7): e13591, 2021 07 07.
Article in English | MEDLINE | ID: mdl-34096686

ABSTRACT

Cachexia syndrome develops in patients with diseases such as cancer and sepsis and is characterized by progressive muscle wasting. While iNOS is one of the main effectors of cachexia, its mechanism of action and whether it could be targeted for therapy remains unexplored. Here, we show that iNOS knockout mice and mice treated with the clinically tested iNOS inhibitor GW274150 are protected against muscle wasting in models of both septic and cancer cachexia. We demonstrate that iNOS triggers muscle wasting by disrupting mitochondrial content, morphology, and energy production processes such as the TCA cycle and acylcarnitine transport. Notably, iNOS inhibits oxidative phosphorylation through impairment of complexes II and IV of the electron transport chain and reduces ATP production, leading to energetic stress, activation of AMPK, suppression of mTOR, and, ultimately, muscle atrophy. Importantly, all these effects were reversed by GW274150. Therefore, our data establish how iNOS induces muscle wasting under cachectic conditions and provide a proof of principle for the repurposing of iNOS inhibitors, such as GW274150 for the treatment of cachexia.


Subject(s)
Cachexia , Neoplasms , Animals , Humans , Mice , Mitochondria , Muscles , Muscular Atrophy
3.
Nat Commun ; 10(1): 4171, 2019 09 13.
Article in English | MEDLINE | ID: mdl-31519904

ABSTRACT

The master posttranscriptional regulator HuR promotes muscle fiber formation in cultured muscle cells. However, its impact on muscle physiology and function in vivo is still unclear. Here, we show that muscle-specific HuR knockout (muHuR-KO) mice have high exercise endurance that is associated with enhanced oxygen consumption and carbon dioxide production. muHuR-KO mice exhibit a significant increase in the proportion of oxidative type I fibers in several skeletal muscles. HuR mediates these effects by collaborating with the mRNA decay factor KSRP to destabilize the PGC-1α mRNA. The type I fiber-enriched phenotype of muHuR-KO mice protects against cancer cachexia-induced muscle loss. Therefore, our study uncovers that under normal conditions HuR modulates muscle fiber type specification by promoting the formation of glycolytic type II fibers. We also provide a proof-of-principle that HuR expression can be targeted therapeutically in skeletal muscles to combat cancer-induced muscle wasting.


Subject(s)
ELAV-Like Protein 1/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/etiology , Muscular Atrophy/metabolism , Neoplasms/complications , Animals , Cell Line , Cell Line, Tumor , Cross-Sectional Studies , ELAV-Like Protein 1/genetics , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Immunohistochemistry , Male , Mice , Mice, Knockout
4.
EMBO Mol Med ; 10(7)2018 07.
Article in English | MEDLINE | ID: mdl-29844217

ABSTRACT

Activation of AMPK has been associated with pro-atrophic signaling in muscle. However, AMPK also has anti-inflammatory effects, suggesting that in cachexia, a syndrome of inflammatory-driven muscle wasting, AMPK activation could be beneficial. Here we show that the AMPK agonist AICAR suppresses IFNγ/TNFα-induced atrophy, while the mitochondrial inhibitor metformin does not. IFNγ/TNFα impair mitochondrial oxidative respiration in myotubes and promote a metabolic shift to aerobic glycolysis, similarly to metformin. In contrast, AICAR partially restored metabolic function. The effects of AICAR were prevented by the AMPK inhibitor Compound C and were reproduced with A-769662, a specific AMPK activator. AICAR and A-769662 co-treatment was found to be synergistic, suggesting that the anti-cachectic effects of these drugs are mediated through AMPK activation. AICAR spared muscle mass in mouse models of cancer and LPS induced atrophy. Together, our findings suggest a dual function for AMPK during inflammation-driven atrophy, wherein it can play a protective role when activated exogenously early in disease progression, but may contribute to anabolic suppression and atrophy when activated later through mitochondrial dysfunction and subsequent metabolic stress.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Cachexia/prevention & control , Metformin/therapeutic use , Protein Kinases/metabolism , Ribonucleotides/therapeutic use , AMP-Activated Protein Kinase Kinases , Aminoimidazole Carboxamide/therapeutic use , Animals , Cachexia/etiology , Cell Line , Enzyme Activation , Inflammation/complications , Interferon-gamma/antagonists & inhibitors , Male , Mice, Inbred BALB C , Mitochondria/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/enzymology , Neoplasms, Experimental/pathology , Nitric Oxide Synthase Type II/metabolism , Protein Kinases/drug effects , Shock, Septic/chemically induced , Shock, Septic/complications , Tumor Necrosis Factor-alpha/antagonists & inhibitors
5.
EMBO Mol Med ; 9(5): 622-637, 2017 05.
Article in English | MEDLINE | ID: mdl-28264935

ABSTRACT

Cachexia is a debilitating syndrome characterized by involuntary muscle wasting that is triggered at the late stage of many cancers. While the multifactorial nature of this syndrome and the implication of cytokines such as IL-6, IFNγ, and TNFα is well established, we still do not know how various effector pathways collaborate together to trigger muscle atrophy. Here, we show that IFNγ/TNFα promotes the phosphorylation of STAT3 on Y705 residue in the cytoplasm of muscle fibers by activating JAK kinases. Unexpectedly, this effect occurs both in vitro and in vivo independently of IL-6, which is considered as one of the main triggers of STAT3-mediated muscle wasting. pY-STAT3 forms a complex with NF-κB that is rapidly imported to the nucleus where it is recruited to the promoter of the iNos gene to activate the iNOS/NO pathway, a well-known downstream effector of IFNγ/TNFα-induced muscle loss. Together, these findings show that STAT3 and NF-κB respond to the same upstream signal and cooperate to promote the expression of pro-cachectic genes, the identification of which could provide effective targets to combat this deadly syndrome.


Subject(s)
Interferon-gamma/immunology , Interleukin-6/immunology , Muscular Atrophy/immunology , NF-kappa B/immunology , STAT3 Transcription Factor/immunology , Tumor Necrosis Factor-alpha/immunology , Wasting Syndrome/immunology , Animals , Cell Line , Inflammation/immunology , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Muscles/immunology , Muscles/pathology , Muscular Atrophy/pathology , Protein Interaction Maps , Wasting Syndrome/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...