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.
Cells ; 11(9)2022 04 22.
Article in English | MEDLINE | ID: mdl-35563723

ABSTRACT

Duchenne muscular dystrophy (DMD) is a common X-linked degenerative muscle disorder that involves mutations in the DMD gene that frequently reduce the expression of the dystrophin protein, compromising the structural integrity of the sarcolemmal membrane and leaving it vulnerable to injury during cycles of muscle contraction and relaxation. This results in an increased frequency of sarcolemma disruptions that can compromise the barrier function of the membrane and lead to death of the myocyte. Sarcolemmal membrane repair processes can potentially compensate for increased membrane disruptions in DMD myocytes. Previous studies demonstrated that TRIM72, a muscle-enriched tripartite motif (TRIM) family protein also known as mitsugumin 53 (MG53), is a component of the cell membrane repair machinery in striated muscle. To test the importance of membrane repair in striated muscle in compensating for the membrane fragility in DMD, we crossed TRIM72/MG53 knockout mice into the mdx mouse model of DMD. These double knockout (DKO) mice showed compromised sarcolemmal membrane integrity compared to mdx mice, as measured by immunoglobulin G staining and ex vivo muscle laser microscopy wounding assays. We also found a significant decrease in muscle ex vivo contractile function as compared to mdx mice at both 6 weeks and 1.5 years of age. As the DKO mice aged, they developed more extensive fibrosis in skeletal muscles compared to mdx. Our findings indicate that TRIM72/MG53-mediated membrane repair can partially compensate for the sarcolemmal fragility associated with DMD and that the loss of membrane repair results in increased pathology in the DKO mice.


Subject(s)
Muscular Dystrophy, Duchenne , Animals , Disease Models, Animal , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred mdx , Mice, Knockout , Muscle, Skeletal/metabolism , Sarcolemma/metabolism , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism
2.
J Clin Invest ; 130(8): 4440-4455, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32687067

ABSTRACT

Idiopathic inflammatory myopathies (IIM) involve chronic inflammation of skeletal muscle and subsequent muscle degeneration due to an uncontrolled autoimmune response; however, the mechanisms leading to pathogenesis are not well understood. A compromised sarcolemmal repair process could promote an aberrant exposure of intramuscular antigens with the subsequent initiation of an inflammatory response that contributes to IIM. Using an adoptive transfer mouse model of IIM, we show that sarcolemmal repair is significantly compromised in distal skeletal muscle in the absence of inflammation. We identified autoantibodies against TRIM72 (also known as MG53), a muscle-enriched membrane repair protein, in IIM patient sera and in our mouse model of IIM by ELISA. We found that patient sera with elevated levels of TRIM72 autoantibodies suppress sarcolemmal resealing in healthy skeletal muscle, and depletion of TRIM72 antibodies from these same serum samples rescues sarcolemmal repair capacity. Autoantibodies targeting TRIM72 lead to skeletal muscle fibers with compromised membrane barrier function, providing a continuous source of autoantigens to promote autoimmunity and further amplifying humoral responses. These findings reveal a potential pathogenic mechanism that acts as a feedback loop contributing to the progression of IIM.


Subject(s)
Autoantibodies/immunology , Autoimmune Diseases/immunology , Membrane Proteins/immunology , Muscle Fibers, Skeletal/immunology , Myositis/immunology , Sarcolemma/immunology , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Humans , Membrane Proteins/genetics , Mice , Mice, Knockout , Muscle Fibers, Skeletal/pathology , Myositis/genetics , Myositis/pathology , Rabbits , Sarcolemma/genetics , Sarcolemma/pathology
3.
PLoS One ; 15(4): e0231194, 2020.
Article in English | MEDLINE | ID: mdl-32271817

ABSTRACT

Various injuries to the neural tissues can cause irreversible damage to multiple functions of the nervous system ranging from motor control to cognitive function. The limited treatment options available for patients have led to extensive interest in studying the mechanisms of neuronal regeneration and recovery from injury. Since many neurons are terminally differentiated, by increasing cell survival following injury it may be possible to minimize the impact of these injuries and provide translational potential for treatment of neuronal diseases. While several cell types are known to survive injury through plasma membrane repair mechanisms, there has been little investigation of membrane repair in neurons and even fewer efforts to target membrane repair as a therapy in neurons. Studies from our laboratory group and others demonstrated that mitsugumin 53 (MG53), a muscle-enriched tripartite motif (TRIM) family protein also known as TRIM72, is an essential component of the cell membrane repair machinery in skeletal muscle. Interestingly, recombinant human MG53 (rhMG53) can be applied exogenously to increase membrane repair capacity both in vitro and in vivo. Increasing the membrane repair capacity of neurons could potentially minimize the death of these cells and affect the progression of various neuronal diseases. In this study we assess the therapeutic potential of rhMG53 to increase membrane repair in cultured neurons and in an in vivo mouse model of neurotrauma. We found that a robust repair response exists in various neuronal cells and that rhMG53 can increase neuronal membrane repair both in vitro and in vivo. These findings provide direct evidence of conserved membrane repair responses in neurons and that these repair mechanisms can be targeted as a potential therapeutic approach for neuronal injury.


Subject(s)
Nerve Regeneration , Sciatic Nerve/injuries , Sciatic Nerve/physiopathology , Wound Healing , Animals , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Crush Injuries/pathology , Crush Injuries/physiopathology , Disease Models, Animal , Humans , Membrane Proteins/metabolism , Membranes , Mice, Inbred C57BL , Nerve Regeneration/drug effects , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Recombinant Proteins/pharmacology , Sciatic Nerve/drug effects , Sciatic Nerve/pathology , Tripartite Motif Proteins/pharmacology , Wound Healing/drug effects
4.
Cell Rep ; 28(6): 1612-1622.e4, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31390573

ABSTRACT

Cachexia is a wasting syndrome characterized by pronounced skeletal muscle loss. In cancer, cachexia is associated with increased morbidity and mortality and decreased treatment tolerance. Although advances have been made in understanding the mechanisms of cachexia, translating these advances to the clinic has been challenging. One reason for this shortcoming may be the current animal models, which fail to fully recapitulate the etiology of human cancer-induced tissue wasting. Because pancreatic ductal adenocarcinoma (PDA) presents with a high incidence of cachexia, we engineered a mouse model of PDA that we named KPP. KPP mice, similar to PDA patients, progressively lose skeletal and adipose mass as a consequence of their tumors. In addition, KPP muscles exhibit a similar gene ontology as cachectic patients. We envision that the KPP model will be a useful resource for advancing our mechanistic understanding and ability to treat cancer cachexia.


Subject(s)
Cachexia/etiology , Disease Models, Animal , Pancreatic Neoplasms/complications , Animals , Cachexia/genetics , Cachexia/metabolism , Disease Progression , Female , Gene Ontology , Heterografts , Humans , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Neoplasm Transplantation , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , RNA-Seq , Transcriptome , Pancreatic Neoplasms
5.
Physiology (Bethesda) ; 30(6): 438-48, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26525343

ABSTRACT

Plasma membrane repair is a conserved cellular response mediating active resealing of membrane disruptions to maintain homeostasis and prevent cell death and progression of multiple diseases. Cell membrane repair repurposes mechanisms from various cellular functions, including vesicle trafficking, exocytosis, and endocytosis, to mend the broken membrane. Recent studies increased our understanding of membrane repair by establishing the molecular machinery contributing to membrane resealing. Here, we review some of the key proteins linked to cell membrane repair.


Subject(s)
Cell Membrane/metabolism , Membrane Proteins/metabolism , Regeneration , Animals , Cell Membrane/pathology , Homeostasis , Humans , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...