Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Cell ; 150(3): 533-48, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22863007

ABSTRACT

Nephronophthisis-related ciliopathies (NPHP-RC) are degenerative recessive diseases that affect kidney, retina, and brain. Genetic defects in NPHP gene products that localize to cilia and centrosomes defined them as "ciliopathies." However, disease mechanisms remain poorly understood. Here, we identify by whole-exome resequencing, mutations of MRE11, ZNF423, and CEP164 as causing NPHP-RC. All three genes function within the DNA damage response (DDR) pathway. We demonstrate that, upon induced DNA damage, the NPHP-RC proteins ZNF423, CEP164, and NPHP10 colocalize to nuclear foci positive for TIP60, known to activate ATM at sites of DNA damage. We show that knockdown of CEP164 or ZNF423 causes sensitivity to DNA damaging agents and that cep164 knockdown in zebrafish results in dysregulated DDR and an NPHP-RC phenotype. Our findings link degenerative diseases of the kidney and retina, disorders of increasing prevalence, to mechanisms of DDR.


Subject(s)
DNA Damage , DNA-Binding Proteins/metabolism , Exome , Kidney Diseases, Cystic/genetics , Microtubule Proteins/metabolism , Animals , Cilia/metabolism , Gene Knockdown Techniques , Genes, Recessive , Humans , MRE11 Homologue Protein , Mice , Proteins , Signal Transduction , Zebrafish/embryology , Zebrafish/metabolism
2.
J Biol Chem ; 298(9): 102328, 2022 09.
Article in English | MEDLINE | ID: mdl-35933013

ABSTRACT

Within the intestine, the human G protein-coupled receptor (GPCR) GPR35 is involved in oncogenic signaling, bacterial infections, and inflammatory bowel disease. GPR35 is known to be expressed as two distinct isoforms that differ only in the length of their extracellular N-termini by 31 amino acids, but detailed insights into their functional differences are lacking. Through gene expression analysis in immune and gastrointestinal cells, we show that these isoforms emerge from distinct promoter usage and alternative splicing. Additionally, we employed optical assays in living cells to thoroughly profile both GPR35 isoforms for constitutive and ligand-induced activation and signaling of 10 different heterotrimeric G proteins, ligand-induced arrestin recruitment, and receptor internalization. Our results reveal that the extended N-terminus of the long isoform limits G protein activation yet elevates receptor-ß-arrestin interaction. To better understand the structural basis for this bias, we examined structural models of GPR35 and conducted experiments with mutants of both isoforms. We found that a proposed disulfide bridge between the N-terminus and extracellular loop 3, present in both isoforms, is crucial for constitutive G13 activation, while an additional cysteine contributed by the extended N-terminus of the long GPR35 isoform limits the extent of agonist-induced receptor-ß-arrestin2 interaction. The pharmacological profiles and mechanistic insights of our study provide clues for the future design of isoform-specific GPR35 ligands that selectively modulate GPR35-transducer interactions and allow for mechanism-based therapies against, for example, inflammatory bowel disease or bacterial infections of the gastrointestinal system.


Subject(s)
Receptors, G-Protein-Coupled , Allosteric Regulation , Cysteine/chemistry , Disulfides/chemistry , GTP-Binding Proteins/chemistry , Humans , Inflammatory Bowel Diseases/metabolism , Ligands , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , beta-Arrestins/metabolism
3.
Crit Rev Biochem Mol Biol ; 52(6): 614-637, 2017 12.
Article in English | MEDLINE | ID: mdl-28741966

ABSTRACT

Wnt signaling cascade has developed together with multicellularity to orchestrate the development and homeostasis of complex structures. Wnt pathway components - such as ß-catenin, Dishevelled (DVL), Lrp6, and Axin-- are often dedicated proteins that emerged in evolution together with the Wnt signaling cascade and are believed to function primarily in the Wnt cascade. It is interesting to see that in recent literature many of these proteins are connected with cellular functions that are more ancient and not limited to multicellular organisms - such as cell cycle regulation, centrosome biology, or cell division. In this review, we summarize the recent literature describing this crosstalk. Specifically, we attempt to find the answers to the following questions: Is the response to Wnt ligands regulated by the cell cycle? Is the centrosome and/or cilium required to activate the Wnt pathway? How do Wnt pathway components regulate the centrosomal cycle and cilia formation and function? We critically review the evidence that describes how these connections are regulated and how they help to integrate cell-to-cell communication with the cell and the centrosomal cycle in order to achieve a fine-tuned, physiological response.


Subject(s)
Cell Cycle , Centrosome/metabolism , Wnt Signaling Pathway , Animals , Cell Communication , Cell Polarity , Humans
4.
Proc Natl Acad Sci U S A ; 113(33): 9304-9, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27486244

ABSTRACT

Dishevelled (DVL) is a key scaffolding protein and a branching point in Wnt signaling pathways. Here, we present conclusive evidence that DVL regulates the centrosomal cycle. We demonstrate that DVL dishevelled and axin (DIX) domain, but not DIX domain-mediated multimerization, is essential for DVL's centrosomal localization. DVL accumulates during the cell cycle and associates with NIMA-related kinase 2 (NEK2), which is able to phosphorylate DVL at a multitude of residues, as detected by a set of novel phospho-specific antibodies. This creates interfaces for efficient binding to CDK5 regulatory subunit-associated protein 2 (CDK5RAP2) and centrosomal Nek2-associated protein 1 (C-NAP1), two proteins of the centrosomal linker. Displacement of DVL from the centrosome and its release into the cytoplasm on NEK2 phosphorylation is coupled to the removal of linker proteins, an event necessary for centrosomal separation and proper formation of the mitotic spindle. Lack of DVL prevents NEK2-controlled dissolution of loose centrosomal linker and subsequent centrosomal separation. Increased DVL levels, in contrast, sequester centrosomal NEK2 and mimic monopolar spindle defects induced by a dominant negative version of this kinase. Our study thus uncovers molecular crosstalk between centrosome and Wnt signaling.


Subject(s)
Autoantigens/metabolism , Cell Cycle Proteins/metabolism , Centrosome/metabolism , Dishevelled Proteins/physiology , Intracellular Signaling Peptides and Proteins/metabolism , NIMA-Related Kinases/physiology , Nerve Tissue Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Phosphorylation , Wnt Signaling Pathway
5.
Physiol Genomics ; 50(12): 1071-1082, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30289747

ABSTRACT

Cancer-cachexia (CC) is a wasting condition directly responsible for 20-40% of cancer-related deaths. The mechanisms controlling development of CC-induced muscle wasting are not fully elucidated. Most investigations focus on the postcachectic state and do not examine progression of the condition. We recently demonstrated mitochondrial degenerations precede muscle wasting in time course progression of CC. However, the extent of muscle perturbations before wasting in CC is unknown. Therefore, we performed global gene expression analysis in CC-induced muscle wasting to enhance understanding of intramuscular perturbations across the development of CC. Lewis lung carcinoma (LLC) was injected into the hind-flank of C57BL6/J mice at 8 wk of age with tumor allowed to develop for 1, 2, 3, or 4 wk and compared with PBS-injected control. Muscle wasting was evident at 4 wk LLC. RNA sequencing of gastrocnemius muscle samples showed widespread alterations in LLC compared with PBS animals with largest differences seen in 4 wk LLC, suggesting extensive transcriptomic alterations concurrent to muscle wasting. Commonly altered pathways included: mitochondrial dysfunction and protein ubiquitination, along with other less studied processes in this condition regulating transcription/translation and cytoskeletal structure. Current findings present novel evidence of transcriptomic shifts and altered cellular pathways in CC-induced muscle wasting.


Subject(s)
Cachexia/genetics , Muscle Fibers, Skeletal/pathology , Muscular Atrophy/genetics , Transcriptome/genetics , Animals , Cachexia/pathology , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/pathology , Disease Progression , Gene Expression Profiling/methods , Mice , Mice, Inbred C57BL , Mitochondria/genetics , Mitochondria/pathology , Muscular Atrophy/pathology
6.
J Biol Chem ; 291(29): 15169-84, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27231350

ABSTRACT

Endurance and resistance exercise training induces specific and profound changes in the skeletal muscle transcriptome. Peroxisome proliferator-activated receptor γ coactivator-1 α (PGC-1α) coactivators are not only among the genes differentially induced by distinct training methods, but they also participate in the ensuing signaling cascades that allow skeletal muscle to adapt to each type of exercise. Although endurance training preferentially induces PGC-1α1 expression, resistance exercise activates the expression of PGC-1α2, -α3, and -α4. These three alternative PGC-1α isoforms lack the arginine/serine-rich (RS) and RNA recognition motifs characteristic of PGC-1α1. Discrete functions for PGC-1α1 and -α4 have been described, but the biological role of PGC-1α2 and -α3 remains elusive. Here we show that different PGC-1α variants can affect target gene splicing through diverse mechanisms, including alternative promoter usage. By analyzing the exon structure of the target transcripts for each PGC-1α isoform, we were able to identify a large number of previously unknown PGC-1α2 and -α3 target genes and pathways in skeletal muscle. In particular, PGC-1α2 seems to mediate a decrease in the levels of cholesterol synthesis genes. Our results suggest that the conservation of the N-terminal activation and repression domains (and not the RS/RNA recognition motif) is what determines the gene programs and splicing options modulated by each PGC-1α isoform. By using skeletal muscle-specific transgenic mice for PGC-1α1 and -α4, we could validate, in vivo, splicing events observed in in vitro studies. These results show that alternative PGC-1α variants can affect target gene expression both quantitatively and qualitatively and identify novel biological pathways under the control of this system of coactivators.


Subject(s)
Alternative Splicing , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Animals , Cells, Cultured , Conserved Sequence , Exons , Gene Expression Regulation , Gene Regulatory Networks , Mice , Mice, Transgenic , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/chemistry , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Promoter Regions, Genetic , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Stability , Receptors, Steroid/chemistry , Receptors, Steroid/genetics , Receptors, Steroid/metabolism
7.
J Biol Chem ; 289(34): 23520-33, 2014 Aug 22.
Article in English | MEDLINE | ID: mdl-24993822

ABSTRACT

Dishevelled-3 (Dvl3), a key component of the Wnt signaling pathways, acts downstream of Frizzled (Fzd) receptors and gets heavily phosphorylated in response to pathway activation by Wnt ligands. Casein kinase 1ϵ (CK1ϵ) was identified as the major kinase responsible for Wnt-induced Dvl3 phosphorylation. Currently it is not clear which Dvl residues are phosphorylated and what is the consequence of individual phosphorylation events. In the present study we employed mass spectrometry to analyze in a comprehensive way the phosphorylation of human Dvl3 induced by CK1ϵ. Our analysis revealed >50 phosphorylation sites on Dvl3; only a minority of these sites was found dynamically induced after co-expression of CK1ϵ, and surprisingly, phosphorylation of one cluster of modified residues was down-regulated. Dynamically phosphorylated sites were analyzed functionally. Mutations within PDZ domain (S280A and S311A) reduced the ability of Dvl3 to activate TCF/LEF (T-cell factor/lymphoid enhancer factor)-driven transcription and induce secondary axis in Xenopus embryos. In contrast, mutations of clustered Ser/Thr in the Dvl3 C terminus prevented ability of CK1ϵ to induce electrophoretic mobility shift of Dvl3 and its even subcellular localization. Surprisingly, mobility shift and subcellular localization changes induced by Fzd5, a Wnt receptor, were in all these mutants indistinguishable from wild type Dvl3. In summary, our data on the molecular level (i) support previous the assumption that CK1ϵ acts via phosphorylation of distinct residues as the activator as well as the shut-off signal of Wnt/ß-catenin signaling and (ii) suggest that CK1ϵ acts on Dvl via different mechanism than Fzd5.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Casein Kinase 1 epsilon/metabolism , Frizzled Receptors/metabolism , Phosphoproteins/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Amino Acid Sequence , Animals , Chromatography, Liquid , Dishevelled Proteins , Electrophoretic Mobility Shift Assay , HEK293 Cells , Humans , Molecular Sequence Data , Phosphoproteins/chemistry , Phosphorylation , Protein Folding , Subcellular Fractions/metabolism , Tandem Mass Spectrometry , Transcription, Genetic , Xenopus Proteins , Xenopus laevis
8.
Nat Nanotechnol ; 19(2): 237-245, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37813939

ABSTRACT

Insulin binds the insulin receptor (IR) and regulates anabolic processes in target tissues. Impaired IR signalling is associated with multiple diseases, including diabetes, cancer and neurodegenerative disorders. IRs have been reported to form nanoclusters at the cell membrane in several cell types, even in the absence of insulin binding. Here we exploit the nanoscale spatial organization of the IR to achieve controlled multivalent receptor activation. To control insulin nanoscale spatial organization and valency, we developed rod-like insulin-DNA origami nanostructures carrying different numbers of insulin molecules with defined spacings. Increasing the insulin valency per nanostructure markedly extended the residence time of insulin-DNA origami nanostructures at the receptors. Both insulin valency and spacing affected the levels of IR activation in adipocytes. Moreover, the multivalent insulin design associated with the highest levels of IR activation also induced insulin-mediated transcriptional responses more effectively than the corresponding monovalent insulin nanostructures. In an in vivo zebrafish model of diabetes, treatment with multivalent-but not monovalent-insulin nanostructures elicited a reduction in glucose levels. Our results show that the control of insulin multivalency and spatial organization with nanoscale precision modulates the IR responses, independent of the insulin concentration. Therefore, we propose insulin nanoscale organization as a design parameter in developing new insulin therapies.


Subject(s)
DNA , Nanostructures , Receptor, Insulin , Animals , Diabetes Mellitus/drug therapy , DNA/chemistry , Insulin , Nanostructures/chemistry , Receptor, Insulin/drug effects , Receptor, Insulin/metabolism , Zebrafish
9.
Mol Metab ; 82: 101912, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38458566

ABSTRACT

OBJECTIVE: Skeletal muscle plasticity and remodeling are critical for adapting tissue function to use, disuse, and regeneration. The aim of this study was to identify genes and molecular pathways that regulate the transition from atrophy to compensatory hypertrophy or recovery from injury. Here, we have used a mouse model of hindlimb unloading and reloading, which causes skeletal muscle atrophy, and compensatory regeneration and hypertrophy, respectively. METHODS: We analyzed mouse skeletal muscle at the transition from hindlimb unloading to reloading for changes in transcriptome and extracellular fluid proteome. We then used qRT-PCR, immunohistochemistry, and bulk and single-cell RNA sequencing data to determine Mustn1 gene and protein expression, including changes in gene expression in mouse and human skeletal muscle with different challenges such as exercise and muscle injury. We generated Mustn1-deficient genetic mouse models and characterized them in vivo and ex vivo with regard to muscle function and whole-body metabolism. We isolated smooth muscle cells and functionally characterized them, and performed transcriptomics and proteomics analysis of skeletal muscle and aorta of Mustn1-deficient mice. RESULTS: We show that Mustn1 (Musculoskeletal embryonic nuclear protein 1, also known as Mustang) is highly expressed in skeletal muscle during the early stages of hindlimb reloading. Mustn1 expression is transiently elevated in mouse and human skeletal muscle in response to intense exercise, resistance exercise, or injury. We find that Mustn1 expression is highest in smooth muscle-rich tissues, followed by skeletal muscle fibers. Muscle from heterozygous Mustn1-deficient mice exhibit differences in gene expression related to extracellular matrix and cell adhesion, compared to wild-type littermates. Mustn1-deficient mice have normal muscle and aorta function and whole-body glucose metabolism. We show that Mustn1 is secreted from smooth muscle cells, and that it is present in arterioles of the muscle microvasculature and in muscle extracellular fluid, particularly during the hindlimb reloading phase. Proteomics analysis of muscle from Mustn1-deficient mice confirms differences in extracellular matrix composition, and female mice display higher collagen content after chemically induced muscle injury compared to wild-type littermates. CONCLUSIONS: We show that, in addition to its previously reported intracellular localization, Mustn1 is a microprotein secreted from smooth muscle cells into the muscle extracellular space. We explore its role in muscle ECM deposition and remodeling in homeostasis and upon muscle injury. The role of Mustn1 in fibrosis and immune infiltration upon muscle injury and dystrophies remains to be investigated, as does its potential for therapeutic interventions.


Subject(s)
Micropeptides , Muscle, Skeletal , Animals , Female , Humans , Mice , Extracellular Matrix/metabolism , Hypertrophy/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Myocytes, Smooth Muscle/metabolism
10.
bioRxiv ; 2023 Jun 13.
Article in English | MEDLINE | ID: mdl-37398033

ABSTRACT

Muscular atrophy is a mortality risk factor that happens with disuse, chronic disease, and aging. Recovery from atrophy requires changes in several cell types including muscle fibers, and satellite and immune cells. Here we show that Zfp697/ZNF697 is a damage-induced regulator of muscle regeneration, during which its expression is transiently elevated. Conversely, sustained Zfp697 expression in mouse muscle leads to a gene expression signature of chemokine secretion, immune cell recruitment, and extracellular matrix remodeling. Myofiber-specific Zfp697 ablation hinders the inflammatory and regenerative response to muscle injury, compromising functional recovery. We uncover Zfp697 as an essential interferon gamma mediator in muscle cells, interacting primarily with ncRNAs such as the pro-regenerative miR-206. In sum, we identify Zfp697 as an integrator of cell-cell communication necessary for tissue regeneration.

11.
J Biol Chem ; 286(12): 10396-410, 2011 Mar 25.
Article in English | MEDLINE | ID: mdl-21285348

ABSTRACT

Dishevelled (Dvl) is a key component in the Wnt/ß-catenin signaling pathway. Dvl can multimerize to form dynamic protein aggregates, which are required for the activation of downstream signaling. Upon pathway activation by Wnts, Dvl becomes phosphorylated to yield phosphorylated and shifted (PS) Dvl. Both activation of Dvl in Wnt/ß-catenin signaling and Wnt-induced PS-Dvl formation are dependent on casein kinase 1 (CK1) δ/ε activity. However, the overexpression of CK1 was shown to dissolve Dvl aggregates, and endogenous PS-Dvl forms irrespective of whether or not the activating Wnt triggers the Wnt/ß-catenin pathway. Using a combination of gain-of-function, loss-of-function, and domain mapping approaches, we attempted to solve this discrepancy regarding the role of CK1ε in Dvl biology. We analyzed mutual interaction of CK1δ/ε and two other Dvl kinases, CK2 and PAR1, in the Wnt/ß-catenin pathway. We show that CK2 acts as a constitutive kinase whose activity is required for the further action of CK1ε. Furthermore, we demonstrate that the two consequences of CK1ε phosphorylation are separated both spatially and functionally; first, CK1ε-mediated induction of TCF/LEF-driven transcription (associated with dynamic recruitment of Axin1) is mediated via a PDZ-proline-rich region of Dvl. Second, CK1ε-mediated formation of PS-Dvl is mediated by the Dvl3 C terminus. Furthermore, we demonstrate with several methods that PS-Dvl has decreased ability to polymerize with other Dvls and could, thus, act as the inactive signaling intermediate. We propose a multistep and multikinase model for Dvl activation in the Wnt/ß-catenin pathway that uncovers a built-in de-activation mechanism that is triggered by activating phosphorylation of Dvl by CK1δ/ε.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Phosphoproteins/metabolism , Signal Transduction/physiology , Wnt Proteins/metabolism , beta Catenin/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Casein Kinase 1 epsilon/genetics , Casein Kinase 1 epsilon/metabolism , Casein Kinase II/genetics , Casein Kinase II/metabolism , Casein Kinase Idelta/genetics , Casein Kinase Idelta/metabolism , Dishevelled Proteins , HEK293 Cells , Humans , Mice , Peptide Mapping , Phosphoproteins/genetics , Phosphorylation/physiology , Receptor, PAR-1/genetics , Receptor, PAR-1/metabolism , Wnt Proteins/genetics , beta Catenin/genetics
12.
Sci Rep ; 11(1): 6453, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33742064

ABSTRACT

This study examined acute molecular responses to concurrent exercise involving different muscles. Eight men participated in a randomized crossover-trial with two sessions, one where they performed interval cycling followed by upper body resistance exercise (ER-Arm), and one with upper body resistance exercise only (R-Arm). Biopsies were taken from the triceps prior to and immediately, 90- and 180-min following exercise. Immediately after resistance exercise, the elevation in S6K1 activity was smaller and the 4E-BP1:eIF4E interaction greater in ER-Arm, but this acute attenuation disappeared during recovery. The protein synthetic rate in triceps was greater following exercise than at rest, with no difference between trials. The level of PGC-1α1 mRNA increased to greater extent in ER-Arm than R-Arm after 90 min of recovery, as was PGC-1α4 mRNA after both 90 and 180 min. Levels of MuRF-1 mRNA was unchanged in R-Arm, but elevated during recovery in ER-Arm, whereas MAFbx mRNA levels increased slightly in both trials. RNA sequencing in a subgroup of subjects revealed 862 differently expressed genes with ER-Arm versus R-Arm during recovery. These findings suggest that leg cycling prior to arm resistance exercise causes systemic changes that potentiate induction of specific genes in the triceps, without compromising the anabolic response.


Subject(s)
Arm/physiology , High-Intensity Interval Training/methods , Leg/physiology , Muscle, Skeletal/metabolism , Resistance Training/methods , Adaptor Proteins, Signal Transducing/metabolism , Adult , Cell Cycle Proteins/metabolism , Eukaryotic Initiation Factor-4E/metabolism , Humans , Male , Muscle Proteins/metabolism , Muscle, Skeletal/physiology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism
13.
Metabolites ; 11(8)2021 Aug 03.
Article in English | MEDLINE | ID: mdl-34436450

ABSTRACT

The kynurenine pathway of tryptophan (TRP) degradation (KP) generates metabolites with effects on metabolism, immunity, and mental health. Endurance exercise training can change KP metabolites by changing the levels of KP enzymes in skeletal muscle. This leads to a metabolite pattern that favors energy expenditure and an anti-inflammatory immune cell profile and reduces neurotoxic metabolites. Here, we aimed to understand if TRP supplementation in untrained vs. trained subjects affects KP metabolite levels and biological effects. Our data show that chronic TRP supplementation in mice increases all KP metabolites in circulation, and that exercise reduces the neurotoxic branch of the pathway. However, in addition to increasing wheel running, we did not observe other effects of TRP supplementation on training adaptations, energy metabolism or behavior in mice. A similar increase in KP metabolites was seen in trained vs. untrained human volunteers that took a TRP drink while performing a bout of aerobic exercise. With this acute TRP administration, TRP and KYN were higher in the trained vs. the untrained group. Considering the many biological effects of the KP, which can lead to beneficial or deleterious effects to health, our data encourage future studies of the crosstalk between TRP supplementation and physical exercise.

14.
Cell Metab ; 33(11): 2215-2230.e8, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34592133

ABSTRACT

Endurance exercise promotes skeletal muscle vascularization, oxidative metabolism, fiber-type switching, and neuromuscular junction integrity. Importantly, the metabolic and contractile properties of the muscle fiber must be coupled to the identity of the innervating motor neuron (MN). Here, we show that muscle-derived neurturin (NRTN) acts on muscle fibers and MNs to couple their characteristics. Using a muscle-specific NRTN transgenic mouse (HSA-NRTN) and RNA sequencing of MN somas, we observed that retrograde NRTN signaling promotes a shift toward a slow MN identity. In muscle, NRTN increased capillary density and oxidative capacity and induced a transcriptional reprograming favoring fatty acid metabolism over glycolysis. This combination of effects on muscle and MNs makes HSA-NRTN mice lean with remarkable exercise performance and motor coordination. Interestingly, HSA-NRTN mice largely recapitulate the phenotype of mice with muscle-specific expression of its upstream regulator PGC-1ɑ1. This work identifies NRTN as a myokine that couples muscle oxidative capacity to slow MN identity.


Subject(s)
Motor Neurons , Neurturin , Animals , Mice , Mice, Transgenic , Motor Neurons/metabolism , Muscle, Skeletal/metabolism , Neurturin/genetics , Neurturin/metabolism , Neurturin/pharmacology , Oxidative Stress
15.
Front Endocrinol (Lausanne) ; 11: 591476, 2020.
Article in English | MEDLINE | ID: mdl-33193103

ABSTRACT

Increasing exercise capacity promotes healthy aging and is strongly associated with lower mortality rates. In this study, we analyzed skeletal muscle transcriptomics coupled to exercise performance in humans and rats to dissect the inherent and response components of aerobic exercise capacity. Using rat models selected for intrinsic and acquired aerobic capacity, we determined that the high aerobic capacity muscle transcriptome is associated with pathways for tissue oxygenation and vascularization. Conversely, the low capacity muscle transcriptome indicated immune response and metabolic dysfunction. Low response to training was associated with an inflammatory signature and revealed a potential link to circadian rhythm. Next, we applied bioinformatics tools to predict potential secreted factors (myokines). The predicted secretome profile for exercise capacity highlighted circulatory factors involved in lipid metabolism and the exercise response secretome was associated with extracellular matrix remodelling. Lastly, we utilized human muscle mitochondrial respiration and transcriptomics data to explore molecular mediators of exercise capacity and response across species. Human transcriptome comparison highlighted epigenetic mechanisms linked to exercise capacity and the damage repair for response. Overall, our findings from this cross-species transcriptome analysis of exercise capacity and response establish a foundation for future studies on the mechanisms that link exercise and health.


Subject(s)
Circadian Rhythm , Exercise Tolerance , Gene Expression Regulation , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Physical Conditioning, Animal , Transcriptome , Animals , Humans , Inflammation/genetics , Male , Muscle Proteins/genetics , Rats
16.
Nat Commun ; 10(1): 2767, 2019 06 24.
Article in English | MEDLINE | ID: mdl-31235694

ABSTRACT

The coactivator PGC-1α1 is activated by exercise training in skeletal muscle and promotes fatigue-resistance. In exercised muscle, PGC-1α1 enhances the expression of kynurenine aminotransferases (Kats), which convert kynurenine into kynurenic acid. This reduces kynurenine-associated neurotoxicity and generates glutamate as a byproduct. Here, we show that PGC-1α1 elevates aspartate and glutamate levels and increases the expression of glycolysis and malate-aspartate shuttle (MAS) genes. These interconnected processes improve energy utilization and transfer fuel-derived electrons to mitochondrial respiration. This PGC-1α1-dependent mechanism allows trained muscle to use kynurenine metabolism to increase the bioenergetic efficiency of glucose oxidation. Kat inhibition with carbidopa impairs aspartate biosynthesis, mitochondrial respiration, and reduces exercise performance and muscle force in mice. Our findings show that PGC-1α1 activates the MAS in skeletal muscle, supported by kynurenine catabolism, as part of the adaptations to endurance exercise. This crosstalk between kynurenine metabolism and the MAS may have important physiological and clinical implications.


Subject(s)
Energy Metabolism/physiology , Fatigue/physiopathology , Kynurenine/metabolism , Muscle, Skeletal/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Adaptation, Physiological , Animals , Aspartate Aminotransferases/metabolism , Aspartic Acid/metabolism , Carbidopa/pharmacology , Cell Respiration/drug effects , Cell Respiration/physiology , Energy Metabolism/drug effects , Glycolysis/physiology , Malates/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism , Models, Animal , Muscle, Skeletal/physiopathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Physical Conditioning, Animal/physiology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Transaminases/antagonists & inhibitors , Transaminases/metabolism
17.
Skelet Muscle ; 9(1): 26, 2019 10 31.
Article in English | MEDLINE | ID: mdl-31666122

ABSTRACT

BACKGROUND: Skeletal muscle mass and strength are crucial determinants of health. Muscle mass loss is associated with weakness, fatigue, and insulin resistance. In fact, it is predicted that controlling muscle atrophy can reduce morbidity and mortality associated with diseases such as cancer cachexia and sarcopenia. METHODS: We analyzed gene expression data from muscle of mice or human patients with diverse muscle pathologies and identified LMCD1 as a gene strongly associated with skeletal muscle function. We transiently expressed or silenced LMCD1 in mouse gastrocnemius muscle or in mouse primary muscle cells and determined muscle/cell size, targeted gene expression, kinase activity with kinase arrays, protein immunoblotting, and protein synthesis levels. To evaluate force, calcium handling, and fatigue, we transduced the flexor digitorum brevis muscle with a LMCD1-expressing adenovirus and measured specific force and sarcoplasmic reticulum Ca2+ release in individual fibers. Finally, to explore the relationship between LMCD1 and calcineurin, we ectopically expressed Lmcd1 in the gastrocnemius muscle and treated those mice with cyclosporine A (calcineurin inhibitor). In addition, we used a luciferase reporter construct containing the myoregulin gene promoter to confirm the role of a LMCD1-calcineurin-myoregulin axis in skeletal muscle mass control and calcium handling. RESULTS: Here, we identify LIM and cysteine-rich domains 1 (LMCD1) as a positive regulator of muscle mass, that increases muscle protein synthesis and fiber size. LMCD1 expression in vivo was sufficient to increase specific force with lower requirement for calcium handling and to reduce muscle fatigue. Conversely, silencing LMCD1 expression impairs calcium handling and force, and induces muscle fatigue without overt atrophy. The actions of LMCD1 were dependent on calcineurin, as its inhibition using cyclosporine A reverted the observed hypertrophic phenotype. Finally, we determined that LMCD1 represses the expression of myoregulin, a known negative regulator of muscle performance. Interestingly, we observed that skeletal muscle LMCD1 expression is reduced in patients with skeletal muscle disease. CONCLUSIONS: Our gain- and loss-of-function studies show that LMCD1 controls protein synthesis, muscle fiber size, specific force, Ca2+ handling, and fatigue resistance. This work uncovers a novel role for LMCD1 in the regulation of skeletal muscle mass and function with potential therapeutic implications.


Subject(s)
Co-Repressor Proteins/genetics , Co-Repressor Proteins/physiology , LIM Domain Proteins/genetics , LIM Domain Proteins/physiology , Muscle, Skeletal/physiology , Animals , Calcineurin/physiology , Calcineurin Inhibitors/pharmacology , Calcium/metabolism , Cells, Cultured , Gene Expression Regulation , Humans , Hypertrophy/genetics , Hypertrophy/pathology , Hypertrophy/physiopathology , Mice , Mice, Inbred C57BL , Mice, SCID , Mice, Transgenic , Muscle Fibers, Skeletal/pathology , Muscle Fibers, Skeletal/physiology , Muscle Proteins/deficiency , Muscle Proteins/genetics , Muscle Proteins/physiology , Muscle Strength/genetics , Muscle Strength/physiology , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Diseases/genetics , Muscular Diseases/pathology , Muscular Diseases/physiopathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction
18.
Cell Metab ; 27(2): 378-392.e5, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29414686

ABSTRACT

The role of tryptophan-kynurenine metabolism in psychiatric disease is well established, but remains less explored in peripheral tissues. Exercise training activates kynurenine biotransformation in skeletal muscle, which protects from neuroinflammation and leads to peripheral kynurenic acid accumulation. Here we show that kynurenic acid increases energy utilization by activating G protein-coupled receptor Gpr35, which stimulates lipid metabolism, thermogenic, and anti-inflammatory gene expression in adipose tissue. This suppresses weight gain in animals fed a high-fat diet and improves glucose tolerance. Kynurenic acid and Gpr35 enhance Pgc-1α1 expression and cellular respiration, and increase the levels of Rgs14 in adipocytes, which leads to enhanced beta-adrenergic receptor signaling. Conversely, genetic deletion of Gpr35 causes progressive weight gain and glucose intolerance, and sensitizes to the effects of high-fat diets. Finally, exercise-induced adipose tissue browning is compromised in Gpr35 knockout animals. This work uncovers kynurenine metabolism as a pathway with therapeutic potential to control energy homeostasis.


Subject(s)
Adipose Tissue/metabolism , Adipose Tissue/pathology , Energy Metabolism , Homeostasis , Inflammation/metabolism , Inflammation/pathology , Kynurenic Acid/metabolism , Receptors, G-Protein-Coupled/metabolism , Adipocytes/metabolism , Adipose Tissue, Beige/metabolism , Adipose Tissue, White/metabolism , Adiposity , Animals , Body Weight/drug effects , Cells, Cultured , Diet, High-Fat , Epididymis/metabolism , Gene Expression Profiling , Gene Expression Regulation , Glucose/metabolism , Lymphocytes/metabolism , Male , Mice, Inbred C57BL , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Physical Conditioning, Animal , RGS Proteins/metabolism , Receptors, Adrenergic, beta/metabolism , Receptors, G-Protein-Coupled/deficiency , Subcutaneous Fat/metabolism , Transcription, Genetic
19.
Science ; 357(6349)2017 Jul 28.
Article in English | MEDLINE | ID: mdl-28751584

ABSTRACT

Kynurenine metabolites are generated by tryptophan catabolism and regulate biological processes that include host-microbiome signaling, immune cell response, and neuronal excitability. Enzymes of the kynurenine pathway are expressed in different tissues and cell types throughout the body and are regulated by cues, including nutritional and inflammatory signals. As a consequence of this systemic metabolic integration, peripheral inflammation can contribute to accumulation of kynurenine in the brain, which has been associated with depression and schizophrenia. Conversely, kynurenine accumulation can be suppressed by activating kynurenine clearance in exercised skeletal muscle. The effect of exercise training on depression through modulation of the kynurenine pathway highlights an important mechanism of interorgan cross-talk mediated by these metabolites. Here, we discuss peripheral mechanisms of tryptophan-kynurenine metabolism and their effects on inflammatory, metabolic, oncologic, and psychiatric disorders.


Subject(s)
Brain/metabolism , Depression/metabolism , Exercise , Inflammation/metabolism , Kynurenine/metabolism , Mental Disorders/metabolism , Tryptophan/metabolism , Gastrointestinal Microbiome , Gastrointestinal Tract/metabolism , Humans , Liver/metabolism , Mental Health , Muscle, Skeletal/metabolism , Neoplasms/metabolism , Signal Transduction
20.
Mol Cell Biol ; 31(1): 179-89, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20974802

ABSTRACT

LDL-related protein 6 (LRP6) is a coreceptor of WNTs and a key regulator of the WNT/ß-catenin pathway. Upon activation, LRP6 is phosphorylated within its intracellular PPPS/TP motifs. These phosphorylated motifs are required to recruit axin and to inhibit glycogen synthase kinase 3 (GSK3), two basic components of the ß-catenin destruction complex. On the basis of a kinome-wide small interfering RNA (siRNA) screen and confirmative biochemical analysis, we show that several proline-directed mitogen-activated protein kinases (MAPKs), such as p38, ERK1/2, and JNK1 are sufficient and required for the phosphorylation of PPPS/TP motifs of LRP6. External stimuli, which control the activity of MAPKs, such as phorbol esters and fibroblast growth factor 2 (FGF2) control the choice of the LRP6-PPPS/TP kinase and regulate the amplitude of LRP6 phosphorylation and WNT/ß-catenin-dependent transcription. Our findings suggest that cells not only recruit one dedicated LRP6 kinase but rather select their LRP6 kinase depending on cell type and the external stimulus. Moreover, direct phosphorylation of LRP6 by MAPKs provides a unique point for convergence between WNT/ß-catenin signaling and mitogenic pathways.


Subject(s)
LDL-Receptor Related Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism , Amino Acid Motifs , Animals , Cell Line , Humans , LDL-Receptor Related Proteins/chemistry , LDL-Receptor Related Proteins/genetics , Low Density Lipoprotein Receptor-Related Protein-6 , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/genetics , Phosphorylation , RNA, Small Interfering/genetics , Rats , Receptors, LDL/metabolism , Signal Transduction , Tumor Cells, Cultured , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL