Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Exp Cell Res ; 408(2): 112865, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34637763

ABSTRACT

Protein homeostasis (proteostasis) in multicellular organisms depends on the maintenance of force-bearing and force-generating cellular structures. Within myofibrillar Z-discs of striated muscle, isoforms of synaptopodin-2 (SYNPO2/myopodin) act as adapter proteins that are engaged in proteostasis of the actin-crosslinking protein filamin C (FLNc) under mechanical stress. SYNPO2 directly binds F-actin, FLNc and α-actinin and thus contributes to the architectural features of the actin cytoskeleton. By its association with autophagy mediating proteins, i.e. BAG3 and VPS18, SYNPO2 is also engaged in protein quality control and helps to target mechanical unfolded and damaged FLNc for degradation. Here we show that deficiency of all SYNPO2-isoforms in myotubes leads to decreased myofibrillar stability and deregulated autophagy under mechanical stress. In addition, isoform-specific proteostasis functions were revealed. The PDZ-domain containing variant SYNPO2b and the shorter, PDZ-less isoform SYNPO2e both localize to Z-discs. Yet, SYNPO2e is less stably associated with the Z-disc than SYNPO2b, and is dynamically transferred into FLNc-containing myofibrillar lesions under mechanical stress. SYNPO2e also recruits BAG3 into these lesions via interaction with the WW domain of BAG3. Our data provide evidence for a role of myofibrillar lesions as a transient quality control compartment essential to prevent and repair contraction-induced myofibril damage in muscle and indicate an important coordinating activity for SYNPO2 therein.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/genetics , Microfilament Proteins/genetics , Muscle, Skeletal/metabolism , Stress, Mechanical , Vesicular Transport Proteins/genetics , Actin Cytoskeleton/genetics , Actinin/genetics , Actins/genetics , Animals , Autophagy/genetics , Cell Line , Cytoskeleton/genetics , Humans , Mice , Muscle Fibers, Skeletal/metabolism , Muscle, Striated/metabolism , Myofibrils/genetics , Myofibrils/metabolism , PDZ Domains/genetics , Protein Isoforms/genetics , Synaptophysin/genetics
2.
Mol Biol Cell ; 32(20): ar9, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34379447

ABSTRACT

Basically, all mammalian tissues are constantly exposed to a variety of environmental mechanical signals. Depending on the signal strength, mechanics intervenes in a multitude of cellular processes and is thus capable of inducing simple cellular adaptations but also complex differentiation processes and even apoptosis. The underlying recognition typically depends on mechanosensitive proteins, which most often sense the mechanical signal for the induction of a cellular signaling cascade by changing their protein conformation. However, the fate of mechanosensors after mechanical stress application is still poorly understood, and it remains unclear whether protein degradation pathways affect the mechanosensitivity of cells. Here, we show that cyclic stretch induces autophagosome formation in a time-dependent manner. Formation depends on the cochaperone BAG family molecular chaperone regulator 3 (BAG3) and thus likely involves BAG3-mediated chaperone-assisted selective autophagy. Furthermore, we demonstrate that strain-induced cell reorientation is clearly delayed upon inhibition of autophagy, suggesting a bidirectional cross-talk between mechanotransduction and autophagic degradation. The strength of the observed delay depends on stable adhesion structures and stress fiber formation in a Ras homologue family member A (RhoA)-dependent manner.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Mechanoreceptors/metabolism , Animals , Apoptosis/physiology , Autophagosomes/metabolism , Autophagy/physiology , Biomechanical Phenomena , Cell Line , Fibroblasts/cytology , Fibroblasts/metabolism , Mechanoreceptors/cytology , Mechanotransduction, Cellular , Mice , Muscle, Smooth/cytology , Muscle, Smooth/metabolism , Proteolysis , Rats , Signal Transduction/physiology , Transcription Factors/metabolism
3.
EMBO Rep ; 22(8): e52507, 2021 08 04.
Article in English | MEDLINE | ID: mdl-34309183

ABSTRACT

Cell survival, tissue integrity and organismal health depend on the ability to maintain functional protein networks even under conditions that threaten protein integrity. Protection against such stress conditions involves the adaptation of folding and degradation machineries, which help to preserve the protein network by facilitating the refolding or disposal of damaged proteins. In multicellular organisms, cells are permanently exposed to stress resulting from mechanical forces. Yet, for long time mechanical stress was not recognized as a primary stressor that perturbs protein structure and threatens proteome integrity. The identification and characterization of protein folding and degradation systems, which handle force-unfolded proteins, marks a turning point in this regard. It has become apparent that mechanical stress protection operates during cell differentiation, adhesion and migration and is essential for maintaining tissues such as skeletal muscle, heart and kidney as well as the immune system. Here, we provide an overview of recent advances in our understanding of mechanical stress protection.


Subject(s)
Protein Folding , Proteostasis , Cell Survival , Proteome/metabolism , Stress, Mechanical
4.
Nat Commun ; 12(1): 3575, 2021 06 11.
Article in English | MEDLINE | ID: mdl-34117258

ABSTRACT

An amino acid exchange (P209L) in the HSPB8 binding site of the human co-chaperone BAG3 gives rise to severe childhood cardiomyopathy. To phenocopy the disease in mice and gain insight into its mechanisms, we generated humanized transgenic mouse models. Expression of human BAG3P209L-eGFP in mice caused Z-disc disintegration and formation of protein aggregates. This was accompanied by massive fibrosis resulting in early-onset restrictive cardiomyopathy with increased mortality as observed in patients. RNA-Seq and proteomics revealed changes in the protein quality control system and increased autophagy in hearts from hBAG3P209L-eGFP mice. The mutation renders hBAG3P209L less soluble in vivo and induces protein aggregation, but does not abrogate hBAG3 binding properties. In conclusion, we report a mouse model mimicking the human disease. Our data suggest that the disease mechanism is due to accumulation of hBAG3P209L and mouse Bag3, causing sequestering of components of the protein quality control system and autophagy machinery leading to sarcomere disruption.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cardiomyopathy, Restrictive/genetics , Cardiomyopathy, Restrictive/metabolism , Animals , Autophagy , Binding Sites , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cardiomyopathies/therapy , Cardiomyopathy, Restrictive/therapy , Child , Disease Models, Animal , Gene Expression Regulation , Genetic Therapy , Heart , Heat-Shock Proteins , Humans , Mice , Mice, Transgenic , Molecular Chaperones/metabolism , Mutation , Protein Binding , Proteomics , Sarcomeres/metabolism
5.
Am J Physiol Cell Physiol ; 319(2): C300-C312, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32520607

ABSTRACT

Skeletal muscle is a target of contraction-induced loading (CiL), leading to protein unfolding or cellular perturbations, respectively. While cytoskeletal desmin is responsible for ongoing structural stabilization, in the immediate response to CiL, alpha-crystallin B (CRYAB) is phosphorylated at serine 59 (pCRYABS59) by P38, acutely protecting the cytoskeleton. To reveal adaptation and deadaptation of these myofibrillar subsystems to CiL, we examined CRYAB, P38, and desmin regulation following resistance exercise at diverse time points of a chronic training period. Mechanosensitive JNK phosphorylation (pJNKT183/Y185) was determined to indicate the presence of mechanical components in CiL. Within 6 wk, subjects performed 13 resistance exercise bouts at the 8-12 repetition maximum, followed by 10 days detraining and a final 14th bout. Biopsies were taken at baseline and after the 1st, 3rd, 7th, 10th, 13th, and 14th bout. To assess whether potential desensitization to CiL can be mitigated, one group trained with progressive and a second with constant loading. As no group differences were found, all subjects were combined for statistics. Total and phosphorylated P38 was not regulated over the time course. pCRYABS59 and pJNKT183/Y185 strongly increased following the unaccustomed first bout. This exercise-induced pCRYABS59/pJNKT183/Y185 increase disappeared with the 10th until 13th bout. As response to the detraining period, the 14th bout led to a renewed increase in pCRYABS59. Desmin content followed pCRYABS59 inversely, i.e., was up- when pCRYABS59 was downregulated and vice versa. In conclusion, the pCRYABS59 response indicates increase and decrease in resistance to CiL, in which a reinforced desmin network could play an essential role by structurally stabilizing the cells.


Subject(s)
Adaptation, Physiological/genetics , Desmin/genetics , Muscle, Skeletal/metabolism , alpha-Crystallin B Chain/genetics , Adult , Cytoskeleton/genetics , Cytoskeleton/metabolism , Desmin/metabolism , Gene Expression Regulation/genetics , Humans , Male , Muscle Contraction/physiology , Muscle, Skeletal/physiology , Phosphorylation/genetics , Resistance Training/adverse effects , Young Adult , alpha-Crystallin B Chain/metabolism
6.
J Am Soc Nephrol ; 31(3): 544-559, 2020 03.
Article in English | MEDLINE | ID: mdl-32047005

ABSTRACT

BACKGROUND: Understanding podocyte-specific responses to injury at a systems level is difficult because injury leads to podocyte loss or an increase of extracellular matrix, altering glomerular cellular composition. Finding a window into early podocyte injury might help identify molecular pathways involved in the podocyte stress response. METHODS: We developed an approach to apply proteome analysis to very small samples of purified podocyte fractions. To examine podocytes in early disease states in FSGS mouse models, we used podocyte fractions isolated from individual mice after chemical induction of glomerular disease (with Doxorubicin or LPS). We also applied single-glomerular proteome analysis to tissue from patients with FSGS. RESULTS: Transcriptome and proteome analysis of glomeruli from patients with FSGS revealed an underrepresentation of podocyte-specific genes and proteins in late-stage disease. Proteome analysis of purified podocyte fractions from FSGS mouse models showed an early stress response that includes perturbations of metabolic, mechanical, and proteostasis proteins. Additional analysis revealed a high correlation between the amount of proteinuria and expression levels of the mechanosensor protein Filamin-B. Increased expression of Filamin-B in podocytes in biopsy samples from patients with FSGS, in single glomeruli from proteinuric rats, and in podocytes undergoing mechanical stress suggests that this protein has a role in detrimental stress responses. In Drosophila, nephrocytes with reduced filamin homolog Cher displayed altered filtration capacity, but exhibited no change in slit diaphragm structure. CONCLUSIONS: We identified conserved mechanisms of the podocyte stress response through ultrasensitive proteome analysis of human glomerular FSGS tissue and purified native mouse podocytes during early disease stages. This approach enables systematic comparisons of large-scale proteomics data and phenotype-to-protein correlation.


Subject(s)
Filamins/genetics , Gene Expression Regulation , Glomerulosclerosis, Focal Segmental/pathology , Proteomics/methods , Stress, Physiological/genetics , Animals , Cells, Cultured , Disease Models, Animal , Glomerulosclerosis, Focal Segmental/genetics , Humans , Mice , Podocytes/metabolism , Proteinuria/genetics , Proteinuria/physiopathology , Random Allocation , Rats
7.
Biochim Biophys Acta Mol Cell Res ; 1866(10): 1556-1566, 2019 10.
Article in English | MEDLINE | ID: mdl-31326538

ABSTRACT

Chaperone-assisted selective autophagy (CASA) initiated by the cochaperone Bcl2-associated athanogene 3 (BAG3) represents an important mechanism for the disposal of misfolded and damaged proteins in mammalian cells. Under mechanical stress, the cochaperone cooperates with the small heat shock protein HSPB8 and the cytoskeleton-associated protein SYNPO2 to degrade force-unfolded forms of the actin-crosslinking protein filamin. This is essential for muscle maintenance in flies, fish, mice and men. Here, we identify the serine/threonine protein kinase 38 (STK38), which is part of the Hippo signaling network, as a novel interactor of BAG3. STK38 was previously shown to facilitate cytoskeleton assembly and to promote mitophagy as well as starvation and detachment induced autophagy. Significantly, our study reveals that STK38 exerts an inhibitory activity on BAG3-mediated autophagy. Inhibition relies on a disruption of the functional interplay of BAG3 with HSPB8 and SYNPO2 upon binding of STK38 to the cochaperone. Of note, STK38 attenuates CASA independently of its kinase activity, whereas previously established regulatory functions of STK38 involve target phosphorylation. The ability to exert different modes of regulation on central protein homeostasis (proteostasis) machineries apparently allows STK38 to coordinate the execution of diverse macroautophagy pathways and to balance cytoskeleton assembly and degradation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Autophagy/physiology , Protein Serine-Threonine Kinases/metabolism , Proteostasis/physiology , Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/genetics , Cytoskeleton/metabolism , Filamins/metabolism , HEK293 Cells , HeLa Cells , Heat-Shock Proteins/metabolism , Humans , Microfilament Proteins , Mitophagy , Molecular Chaperones/metabolism , Protein Binding , Proteomics , Signal Transduction , Stress, Mechanical
8.
Autophagy ; 15(7): 1199-1213, 2019 07.
Article in English | MEDLINE | ID: mdl-30744518

ABSTRACT

A major cellular catabolic pathway in neurons is macroautophagy/autophagy, through which misfolded or aggregation-prone proteins are sequestered into autophagosomes that fuse with lysosomes, and are degraded. MAPT (microtubule-associated protein tau) is one of the protein clients of autophagy. Given that accumulation of hyperphosphorylated MAPT contributes to the pathogenesis of Alzheimer disease and other tauopathies, decreasing endogenous MAPT levels has been shown to be beneficial to neuronal health in models of these diseases. A previous study demonstrated that the HSPA/HSP70 co-chaperone BAG3 (BCL2-associated athanogene 3) facilitates endogenous MAPT clearance through autophagy. These findings prompted us to further investigate the mechanisms underlying BAG3-mediated autophagy in the degradation of endogenous MAPT. Here we demonstrate for the first time that BAG3 plays an important role in autophagic flux in the neurites of mature neurons (20-24 days in vitro [DIV]) through interaction with the post-synaptic cytoskeleton protein SYNPO (synaptopodin). Loss of either BAG3 or SYNPO impeded the fusion of autophagosomes and lysosomes predominantly in the post-synaptic compartment. A block of autophagy leads to accumulation of the autophagic receptor protein SQSTM1/p62 (sequestosome 1) as well as MAPT phosphorylated at Ser262 (p-Ser262). Furthermore, p-Ser262 appears to accumulate in autophagosomes at post-synaptic densities. Overall these data provide evidence of a novel role for the co-chaperone BAG3 in synapses. In cooperation with SYNPO, it functions as part of a surveillance complex that facilitates the autophagic clearance of MAPT p-Ser262, and possibly other MAPT species at the post-synapse. This appears to be crucial for the maintenance of a healthy, functional synapse.Abbreviations: aa: amino acids; ACTB: actin beta; BafA1: bafilomycin A1; BAG3: BCL2 associated athanogene 3; CQ chloroquine; CTSL: cathepsin L; DIV: days in vitro; DLG4/PSD95: discs large MAGUK scaffold protein 4; HSPA/HSP70: heat shock protein family A (Hsp70); MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MAP2: microtubule associated protein 2; MAPT: microtubule associated protein tau; p-Ser262: MAPT phosphorylated at serine 262; p-Ser396/404: MAPT phosphorylated at serines 396 and 404; p-Thr231: MAPT phosphorylated at threonine 231; PBS: phosphate buffered saline; PK: proteinase K; scr: scrambled; shRNA: short hairpin RNA; SQSTM1/p62 sequestosome 1; SYN1: synapsin I; SYNPO synaptopodin; SYNPO2/myopodin: synaptopodin 2; VPS: vacuolar protein sorting.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Autophagy/genetics , Microfilament Proteins/metabolism , Neurons/metabolism , tau Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis Regulatory Proteins/genetics , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagy/drug effects , HeLa Cells , Humans , Lysosomes/metabolism , Microfilament Proteins/genetics , Neurites/metabolism , Neurons/drug effects , Neurons/enzymology , Phosphorylation , Post-Synaptic Density/metabolism , Proteolysis , Rats , Rats, Sprague-Dawley , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , Synapses/metabolism , WW Domains , tau Proteins/chemistry
9.
Trends Cell Biol ; 28(7): 512-522, 2018 07.
Article in English | MEDLINE | ID: mdl-29704981

ABSTRACT

Conjugation of ubiquitin onto proteins generates a degradation signal or exerts degradation-independent regulatory functions. Ubiquitylation is governed by the antagonistic action of ubiquitin ligases and deubiquitylating enzymes (DUBs). Several recent publications illustrate a balanced interplay of ligases and DUBs at signaling hubs that are central to longevity and protein homeostasis (proteostasis). In addition, stress-induced alterations of ubiquitin conjugation are emerging as key events that drive aging and contribute to the pathology of age-related diseases. This physiological role of dynamic ubiquitylation further extends its well-known function in protein regulation and quality control at the cellular level. Recent work thus significantly advances our understanding of the aging process both at the molecular and organismal level.


Subject(s)
Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Humans , Proteolysis , Signal Transduction , Ubiquitination
10.
Nat Cell Biol ; 19(10): 1142-1144, 2017 Sep 29.
Article in English | MEDLINE | ID: mdl-28960199

ABSTRACT

It is generally accepted that protein function depends on a defined 3D structure, with unfolding and aggregation dealing a final blow to functionality. A study now shows that the regulated exposure of an unstructured region in yeast pyruvate kinase triggers reversible aggregation to preserve protein function under stress.

11.
J Cell Sci ; 130(17): 2781-2788, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28808089

ABSTRACT

Cellular and organismal survival depend on the ability to maintain the proteome, even under conditions that threaten protein integrity. BCL2-associated athanogene 3 (BAG3) is essential for protein homeostasis (proteostasis) in stressed cells. Owing to its multi-domain structure, it engages in diverse processes that are crucial for proteome maintenance. BAG3 promotes the activity of molecular chaperones, sequesters and concentrates misfolded proteins, initiates autophagic disposal, and balances transcription, translation and degradation. In this Cell Science at a Glance article and the accompanying poster, we discuss the functions of this multi-functional proteostasis tool with a focus on mechanical stress protection and describe the importance of BAG3 for human physiology and pathophysiology.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Proteostasis , Animals , Autophagy , Cytoskeleton/metabolism , Filamins/metabolism , Humans , Molecular Chaperones/metabolism
12.
Cell ; 169(3): 470-482.e13, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28431247

ABSTRACT

Aging is attended by a progressive decline in protein homeostasis (proteostasis), aggravating the risk for protein aggregation diseases. To understand the coordination between proteome imbalance and longevity, we addressed the mechanistic role of the quality-control ubiquitin ligase CHIP, which is a key regulator of proteostasis. We observed that CHIP deficiency leads to increased levels of the insulin receptor (INSR) and reduced lifespan of worms and flies. The membrane-bound INSR regulates the insulin and IGF1 signaling (IIS) pathway and thereby defines metabolism and aging. INSR is a direct target of CHIP, which triggers receptor monoubiquitylation and endocytic-lysosomal turnover to promote longevity. However, upon proteotoxic stress conditions and during aging, CHIP is recruited toward disposal of misfolded proteins, reducing its capacity to degrade the INSR. Our study indicates a competitive relationship between proteostasis and longevity regulation through CHIP-assisted proteolysis, providing a mechanistic concept for understanding the impact of proteome imbalance on aging.


Subject(s)
Aging , Antigens, CD/metabolism , Receptor, Insulin/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Caenorhabditis elegans , Drosophila melanogaster , Endocytosis , Humans , Longevity , Lysosomes/metabolism , Proteolysis , Proteome , Signal Transduction , Somatomedins , Ubiquitination
13.
Biochim Biophys Acta Mol Cell Res ; 1864(1): 62-75, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27756573

ABSTRACT

The cochaperone BAG3 is a central protein homeostasis factor in mechanically strained mammalian cells. It mediates the degradation of unfolded and damaged forms of the actin-crosslinker filamin through chaperone-assisted selective autophagy (CASA). In addition, BAG3 stimulates filamin transcription in order to compensate autophagic disposal and to maintain the actin cytoskeleton under strain. Here we demonstrate that BAG3 coordinates protein synthesis and autophagy through spatial regulation of the mammalian target of rapamycin complex 1 (mTORC1). The cochaperone utilizes its WW domain to contact a proline-rich motif in the tuberous sclerosis protein TSC1 that functions as an mTORC1 inhibitor in association with TSC2. Interaction with BAG3 results in a recruitment of TSC complexes to actin stress fibers, where the complexes act on a subpopulation of mTOR-positive vesicles associated with the cytoskeleton. Local inhibition of mTORC1 is essential to initiate autophagy at sites of filamin unfolding and damage. At the same time, BAG3-mediated sequestration of TSC1/TSC2 relieves mTORC1 inhibition in the remaining cytoplasm, which stimulates protein translation. In human muscle, an exercise-induced association of TSC1 with the cytoskeleton coincides with mTORC1 activation in the cytoplasm. The spatial regulation of mTORC1 exerted by BAG3 apparently provides the basis for a simultaneous induction of autophagy and protein synthesis to maintain the proteome under mechanical strain.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/genetics , Autophagy/genetics , Multiprotein Complexes/genetics , Muscle, Skeletal/metabolism , Myocytes, Smooth Muscle/metabolism , Stress, Mechanical , TOR Serine-Threonine Kinases/genetics , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins/metabolism , Biomechanical Phenomena , Cell Line , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Filamins/genetics , Filamins/metabolism , Gene Expression , Gene Expression Regulation , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes/metabolism , Muscle, Skeletal/cytology , Myocytes, Smooth Muscle/ultrastructure , Protein Binding , Protein Biosynthesis , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
14.
Curr Biol ; 26(15): R703-R705, 2016 08 08.
Article in English | MEDLINE | ID: mdl-27505239

ABSTRACT

Recent publications illustrate an extensive crosstalk between the actin cytoskeleton and autophagy, a program for self-digestion. Actin polymerization provides a pushing force for organelle shaping and trafficking during autophagy, but the cytoskeleton is also targeted by autophagy under mechanical strain.


Subject(s)
Actin Cytoskeleton , Autophagy , Actins , Cell Movement , Cytoskeleton , Microtubules
15.
Autophagy ; 11(3): 538-46, 2015.
Article in English | MEDLINE | ID: mdl-25714469

ABSTRACT

Chaperone-assisted selective autophagy (CASA) is a tension-induced degradation pathway essential for muscle maintenance. Impairment of CASA causes childhood muscle dystrophy and cardiomyopathy. However, the importance of CASA for muscle function in healthy individuals has remained elusive so far. Here we describe the impact of strength training on CASA in a group of healthy and moderately trained men. We show that strenuous resistance exercise causes an acute induction of CASA in affected muscles to degrade mechanically damaged cytoskeleton proteins. Moreover, repeated resistance exercise during 4 wk of training led to an increased expression of CASA components. In human skeletal muscle, CASA apparently acts as a central adaptation mechanism that responds to acute physical exercise and to repeated mechanical stimulation.


Subject(s)
Adaptation, Physiological , Autophagy/physiology , Molecular Chaperones/physiology , Quadriceps Muscle/physiology , Adaptor Proteins, Signal Transducing/metabolism , Adult , Apoptosis Regulatory Proteins/metabolism , Biopsy , Creatine Kinase/metabolism , Cytoskeleton/metabolism , Filamins/metabolism , Humans , Male , Microfilament Proteins/metabolism , Microscopy, Electron , Muscle Contraction , Quadriceps Muscle/pathology , Quadriceps Muscle/ultrastructure , Young Adult
16.
Cell Mol Neurobiol ; 34(8): 1123-30, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25078755

ABSTRACT

Genetic studies have linked the evolutionary novel, anthropoid primate-specific gene locus G72/G30 in the etiology of schizophrenia and other psychiatric disorders. However, the function of the protein encoded by this locus, LG72, is currently controversially discussed. Some studies have suggested that LG72 binds to and regulates the activity of the peroxisomal enzyme D-amino-acid-oxidase, while others proposed an alternative role of this protein due to its mitochondrial location in vitro. Studies with transgenic mice expressing LG72 further suggested that high levels of LG72 lead to an impairment of mitochondrial functions with a concomitant increase in reactive oxygen species production. In the present study, we now performed extensive interaction analyses and identified the mitochondrial methionine-R-sulfoxide reductase B2 (MSRB2) as a specific interaction partner of LG72. MSRB2 belongs to the MSR protein family and functions in mitochondrial oxidative stress defense. Based on our results, we propose that LG72 is involved in the regulation of mitochondrial oxidative stress.


Subject(s)
Carrier Proteins/metabolism , Methionine Sulfoxide Reductases/metabolism , Mitochondria/metabolism , Transcription Factors/metabolism , Animals , COS Cells , Chlorocebus aethiops , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins , Mice , Microfilament Proteins , Protein Binding
17.
Mol Biol Cell ; 25(15): 2260-71, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24899640

ABSTRACT

Molecular chaperones play key roles during growth, development, and stress survival. The ability to induce chaperone expression enables cells to cope with the accumulation of nonnative proteins under stress and complete developmental processes with an increased requirement for chaperone assistance. Here we generate and analyze transgenic mice that lack the cochaperone HSPBP1, a nucleotide-exchange factor of HSP70 proteins and inhibitor of chaperone-assisted protein degradation. Male HSPBP1(-/-) mice are sterile because of impaired meiosis and massive apoptosis of spermatocytes. HSPBP1 deficiency in testes strongly reduces the expression of the inducible, antiapoptotic HSP70 family members HSPA1L and HSPA2, the latter of which is essential for synaptonemal complex disassembly during meiosis. We demonstrate that HSPBP1 affects chaperone expression at a posttranslational level by inhibiting the ubiquitylation and proteasomal degradation of inducible HSP70 proteins. We further provide evidence that the cochaperone BAG2 contributes to HSP70 stabilization in tissues other than testes. Our findings reveal that chaperone expression is determined not only by regulated transcription, but also by controlled degradation, with degradation-inhibiting cochaperones exerting essential prosurvival functions.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Spermatogenesis , Ubiquitination , Animals , Gene Expression , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , HeLa Cells , Humans , Infertility, Male/genetics , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Proteolysis , Testis/metabolism , Testis/pathology
18.
Commun Integr Biol ; 6(4): e24925, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23986815

ABSTRACT

Maintaining the dynamic proteome of a living cell in the face of an ever-changing environment depends on a fine-tuned balance of protein synthesis and protein degradation. Molecular chaperones exert key functions during protein homeostasis (proteostasis). They associate with nonnative client proteins following synthesis or damage and facilitate client sorting and folding. When client proteins are terminally misfolded, chaperones cooperate with protein degradation systems to dispose of such clients. This dual proteostasis activity of chaperones is essential for maintaining cell function under normal growth conditions and becomes even more important under stress conditions such as heat and oxidative stress. The recent identification of chaperone-assisted selective autophagy (CASA) as a tension-induced autophagy pathway highlights the critical role of molecular chaperones in mechanically strained cells and tissues. The CASA complex, assembled by the cochaperone BAG3, coordinates protein degradation and protein synthesis in response to mechanical force. Here we describe the composition and function of this chaperone complex in mammals and discuss its relevance for tissue homeostasis and the regulation of cell adhesion, migration and proliferation. We provide a unifying concept for the function of BAG3, which integrates its involvement in muscle maintenance, tumor formation and virus infection.

19.
Autophagy ; 9(6): 920-2, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23518596

ABSTRACT

Impairment of autophagy in patients and animal models severely affects mechanically strained tissues such as skeletal muscle, heart, lung and kidney, leading for example to muscle dystrophy, cardiomyopathy and renal injury. However, the reason for this high reliance on autophagy remained largely elusive. Recent work in our lab now provides a possible explanation. We identified chaperone-assisted selective autophagy (CASA) as a tension-induced autophagy pathway essential for mechanotransduction in mammalian cells.


Subject(s)
Autophagy , Molecular Chaperones/metabolism , Stress, Physiological , Animals , Humans , Mice , Models, Biological , Signal Transduction
20.
Curr Biol ; 23(5): 430-5, 2013 Mar 04.
Article in English | MEDLINE | ID: mdl-23434281

ABSTRACT

Mechanical tension is an ever-present physiological stimulus essential for the development and homeostasis of locomotory, cardiovascular, respiratory, and urogenital systems. Tension sensing contributes to stem cell differentiation, immune cell recruitment, and tumorigenesis. Yet, how mechanical signals are transduced inside cells remains poorly understood. Here, we identify chaperone-assisted selective autophagy (CASA) as a tension-induced autophagy pathway essential for mechanotransduction in muscle and immune cells. The CASA complex, comprised of the molecular chaperones Hsc70 and HspB8 and the cochaperone BAG3, senses the mechanical unfolding of the actin-crosslinking protein filamin. Together with the chaperone-associated ubiquitin ligase CHIP, the complex initiates the ubiquitin-dependent autophagic sorting of damaged filamin to lysosomes for degradation. Autophagosome formation during CASA depends on an interaction of BAG3 with synaptopodin-2 (SYNPO2). This interaction is mediated by the BAG3 WW domain and facilitates cooperation with an autophagosome membrane fusion complex. BAG3 also utilizes its WW domain to engage in YAP/TAZ signaling. Via this pathway, BAG3 stimulates filamin transcription to maintain actin anchoring and crosslinking under mechanical tension. By integrating tension sensing, autophagosome formation, and transcription regulation during mechanotransduction, the CASA machinery ensures tissue homeostasis and regulates fundamental cellular processes such as adhesion, migration, and proliferation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Autophagy , Mechanotransduction, Cellular , Molecular Chaperones/metabolism , Acyltransferases , Animals , Apoptosis Regulatory Proteins , Humans , Jurkat Cells , Male , Mice , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Rats , Stress, Mechanical , Transcription Factors/metabolism , YAP-Signaling Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...