Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
J Biol Chem ; 290(40): 24592-603, 2015 Oct 02.
Article in English | MEDLINE | ID: mdl-26306047

ABSTRACT

Cell membrane repair is an important aspect of physiology, and disruption of this process can result in pathophysiology in a number of different tissues, including wound healing, chronic ulcer and scarring. We have previously identified a novel tripartite motif family protein, MG53, as an essential component of the cell membrane repair machinery. Here we report the functional role of MG53 in the modulation of wound healing and scarring. Although MG53 is absent from keratinocytes and fibroblasts, remarkable defects in skin architecture and collagen overproduction are observed in mg53(-/-) mice, and these animals display delayed wound healing and abnormal scarring. Recombinant human MG53 (rhMG53) protein, encapsulated in a hydrogel formulation, facilitates wound healing and prevents scarring in rodent models of dermal injuries. An in vitro study shows that rhMG53 protects against acute injury to keratinocytes and facilitates the migration of fibroblasts in response to scratch wounding. During fibrotic remodeling, rhMG53 interferes with TGF-ß-dependent activation of myofibroblast differentiation. The resulting down-regulation of α smooth muscle actin and extracellular matrix proteins contributes to reduced scarring. Overall, these studies establish a trifunctional role for MG53 as a facilitator of rapid injury repair, a mediator of cell migration, and a modulator of myofibroblast differentiation during wound healing. Targeting the functional interaction between MG53 and TGF-ß signaling may present a potentially effective means for promoting scarless wound healing.


Subject(s)
Carrier Proteins/physiology , Cell Membrane/metabolism , Muscle Proteins/physiology , Vesicular Transport Proteins/physiology , Wound Healing/physiology , 3T3 Cells , Actins/metabolism , Animals , Cell Differentiation , Cell Movement , Cicatrix/pathology , Collagen Type I/metabolism , Fibroblasts/cytology , Fibronectins/metabolism , Fibrosis/pathology , Gene Expression Regulation , Humans , Hydrogels/chemistry , Keratinocytes/metabolism , Membrane Proteins , Mice , Muscle, Smooth/metabolism , Myofibroblasts/metabolism , Rabbits , Rats , Rats, Sprague-Dawley , Recombinant Proteins/metabolism , Skin/pathology , Tripartite Motif Proteins
2.
Opt Express ; 22(10): 12160-76, 2014 May 19.
Article in English | MEDLINE | ID: mdl-24921337

ABSTRACT

One key factor that limits resolution of single-molecule superresolution microscopy relates to the localization accuracy of the activated emitters, which is usually deteriorated by two factors. One originates from the background noise due to out-of-focus signals, sample auto-fluorescence, and camera acquisition noise; and the other is due to the low photon count of emitters at a single frame. With fast acquisition rate, the activated emitters can last multiple frames before they transiently switch off or permanently bleach. Effectively incorporating the temporal information of these emitters is critical to improve the spatial resolution. However, majority of the existing reconstruction algorithms locate the emitters frame by frame, discarding or underusing the temporal information. Here we present a new image reconstruction algorithm based on tracklets, short trajectories of the same objects. We improve the localization accuracy by associating the same emitters from multiple frames to form tracklets and by aggregating signals to enhance the signal to noise ratio. We also introduce a weighted mean-shift algorithm (WMS) to automatically detect the number of modes (emitters) in overlapping regions of tracklets so that not only well-separated single emitters but also individual emitters within multi-emitter groups can be identified and tracked. In combination with a maximum likelihood estimator method (MLE), we are able to resolve low to medium density of overlapping emitters with improved localization accuracy. We evaluate the performance of our method with both synthetic and experimental data, and show that the tracklet-based reconstruction is superior in localization accuracy, particularly for weak signals embedded in a strong background. Using this method, for the first time, we resolve the transverse tubule structure of the mammalian skeletal muscle.


Subject(s)
Algorithms , Image Processing, Computer-Assisted , Muscle, Skeletal/cytology , Nanotechnology/methods , Photons , Animals , Cells, Cultured , Signal-To-Noise Ratio
3.
Redox Biol ; 54: 102357, 2022 08.
Article in English | MEDLINE | ID: mdl-35679798

ABSTRACT

Ischemic injury to the heart induces mitochondrial dysfunction due to increasing oxidative stress. MG53, also known as TRIM72, is highly expressed in striated muscle, is secreted as a myokine after exercise, and is essential for repairing damaged plasma membrane of many tissues by interacting with the membrane lipid phosphatidylserine (PS). We hypothesized MG53 could preserve mitochondrial integrity after an ischemic event by binding to the mitochondrial-specific lipid, cardiolipin (CL), for mitochondria protection to prevent mitophagy. Fluorescent imaging and Western blotting experiments showed recombinant human MG53 (rhMG53) translocated to the mitochondria after ischemic injury in vivo and in vitro. Fluorescent imaging indicated rhMG53 treatment reduced superoxide generation in ex vivo and in vitro models. Lipid-binding assay indicated MG53 binds to CL. Transfecting cardiomyocytes with the mitochondria-targeted mt-mKeima showed inhibition of mitophagy after MG53 treatment. Overall, we show that rhMG53 treatment may preserve cardiac function by preserving mitochondria in cardiomyocytes. These findings suggest MG53's interactions with mitochondria could be an attractive avenue for developing MG53 as a targeted protein therapy for cardioprotection.


Subject(s)
Carrier Proteins , Myocytes, Cardiac , Carrier Proteins/metabolism , Humans , Ischemia/metabolism , Lipids , Mitochondria/metabolism , Myocytes, Cardiac/metabolism , Oxidative Stress , Reperfusion
4.
Nat Commun ; 11(1): 3624, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32681036

ABSTRACT

TRIM family proteins play integral roles in the innate immune response to virus infection. MG53 (TRIM72) is essential for cell membrane repair and is believed to be a muscle-specific TRIM protein. Here we show human macrophages express MG53, and MG53 protein expression is reduced following virus infection. Knockdown of MG53 in macrophages leads to increases in type I interferon (IFN) upon infection. MG53 knockout mice infected with influenza virus show comparable influenza virus titres to wild type mice, but display increased morbidity accompanied by more accumulation of CD45+ cells and elevation of IFNß in the lung. We find that MG53 knockdown results in activation of NFκB signalling, which is linked to an increase in intracellular calcium oscillation mediated by ryanodine receptor (RyR). MG53 inhibits IFNß induction in an RyR-dependent manner. This study establishes MG53 as a new target for control of virus-induced morbidity and tissue injury.


Subject(s)
Influenza, Human/immunology , Interferon-beta/metabolism , Membrane Proteins/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Tripartite Motif Proteins/metabolism , Animals , Calcium Signaling/immunology , Cell Line, Tumor , Disease Models, Animal , Gene Knockdown Techniques , Gene Knockout Techniques , Humans , Immunity, Innate , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/virology , Interferon-beta/immunology , Macrophages/immunology , Macrophages/metabolism , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , NF-kappa B/metabolism , RNA, Small Interfering , Signal Transduction/immunology , Tripartite Motif Proteins/genetics
5.
Methods Mol Biol ; 1854: 35-43, 2019.
Article in English | MEDLINE | ID: mdl-28842895

ABSTRACT

Muscle wasting or cachexia is commonly associated with aging and many diseases such as cancer, infection, autoimmune disorders, and trauma. Decrease in muscle mass, or muscle atrophy, is often caused by dysfunction of protein proteolytic systems, such as lysosomes, which regulate protein turnover and homeostasis. Lysosomes contain many hydrolases and proteases and, thus, represent the major organelle that control protein turnover. Recently, lysosomes have emerged as a signaling hub to integrate cellular functions of nutrient sensing and metabolism, autophagy, phagocytosis, and endocytosis, which are all related to tissue homeostasis. In this chapter, we describe the protocol used to measure lysosomal proteinase (cathepsins) activity in the skeletal muscle. A better understanding of lysosomal function in muscle homeostasis is critical in developing new therapeutic approaches to prevent muscle wasting.


Subject(s)
Cathepsins/analysis , Lysosomes/enzymology , Muscle, Skeletal/cytology , Animals , Autophagy , Fluorescent Dyes/chemistry , Homeostasis , Mice , Microscopy, Fluorescence , Muscle, Skeletal/enzymology , Signal Transduction
6.
J Am Heart Assoc ; 8(4): e009960, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30741589

ABSTRACT

Background The aortic valve of the heart experiences constant mechanical stress under physiological conditions. Maladaptive valve injury responses contribute to the development of valvular heart disease. Here, we test the hypothesis that MG 53 (mitsugumin 53), an essential cell membrane repair protein, can protect valvular cells from injury and fibrocalcific remodeling processes associated with valvular heart disease. Methods and Results We found that MG 53 is expressed in pig and human patient aortic valves and observed aortic valve disease in aged Mg53-/- mice. Aortic valves of Mg53-/- mice showed compromised cell membrane integrity. In vitro studies demonstrated that recombinant human MG 53 protein protects primary valve interstitial cells from mechanical injury and that, in addition to mediating membrane repair, recombinant human MG 53 can enter valve interstitial cells and suppress transforming growth factor-ß-dependent activation of fibrocalcific signaling. Conclusions Together, our data characterize valve interstitial cell membrane repair as a novel mechanism of protection against valvular remodeling and assess potential in vivo roles of MG 53 in preventing valvular heart disease.


Subject(s)
Aortic Valve Stenosis/metabolism , Aortic Valve/metabolism , Calcinosis/metabolism , Tripartite Motif Proteins/biosynthesis , Ventricular Remodeling , Animals , Aortic Valve/pathology , Aortic Valve Stenosis/diagnosis , Aortic Valve Stenosis/physiopathology , Biomarkers/metabolism , Blotting, Western , Calcinosis/diagnosis , Calcinosis/physiopathology , Cells, Cultured , Disease Models, Animal , Echocardiography , Humans , Immunohistochemistry , Male , Mice , Signal Transduction , Stress, Mechanical , Swine
7.
Nat Commun ; 10(1): 4659, 2019 10 11.
Article in English | MEDLINE | ID: mdl-31604915

ABSTRACT

MG53 is a muscle-specific TRIM-family protein that presides over the cell membrane repair response. Here, we show that MG53 present in blood circulation acts as a myokine to facilitate tissue injury-repair and regeneration. Transgenic mice with sustained elevation of MG53 in the bloodstream (tPA-MG53) have a healthier and longer life-span when compared with littermate wild type mice. The tPA-MG53 mice show normal glucose handling and insulin signaling in skeletal muscle, and sustained elevation of MG53 in the bloodstream does not have a deleterious impact on db/db mice. More importantly, the tPA-MG53 mice display remarkable dermal wound healing capacity, enhanced muscle performance, and improved injury-repair and regeneration. Recombinant human MG53 protein protects against eccentric contraction-induced acute and chronic muscle injury in mice. Our findings highlight the myokine function of MG53 in tissue protection and present MG53 as an attractive biological reagent for regenerative medicine without interference with glucose handling in the body.


Subject(s)
Membrane Proteins/physiology , Wound Healing , Animals , Calcium/metabolism , Glucose/metabolism , Glucose Tolerance Test , Insulin/metabolism , Membrane Proteins/blood , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Skeletal/metabolism , Regeneration/genetics , Systems Biology
8.
Nutrients ; 10(1)2017 Dec 24.
Article in English | MEDLINE | ID: mdl-29295546

ABSTRACT

Wound care is a major healthcare expenditure. Treatment of burns, surgical and trauma wounds, diabetic lower limb ulcers and skin wounds is a major medical challenge with current therapies largely focused on supportive care measures. Successful wound repair requires a series of tightly coordinated steps including coagulation, inflammation, angiogenesis, new tissue formation and extracellular matrix remodelling. Zinc is an essential trace element (micronutrient) which plays important roles in human physiology. Zinc is a cofactor for many metalloenzymes required for cell membrane repair, cell proliferation, growth and immune system function. The pathological effects of zinc deficiency include the occurrence of skin lesions, growth retardation, impaired immune function and compromised would healing. Here, we discuss investigations on the cellular and molecular mechanisms of zinc in modulating the wound healing process. Knowledge gained from this body of research will help to translate these findings into future clinical management of wound healing.


Subject(s)
Wound Healing , Zinc/metabolism , Animals , Antioxidants/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Humans , Oxidative Stress , Signal Transduction , Time Factors , Tripartite Motif Proteins/metabolism , Zinc/deficiency , Zinc/immunology
9.
Front Biosci (Landmark Ed) ; 22(1): 48-65, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27814601

ABSTRACT

Autophagy, a form of lysosomal degradation capable of eliminating dysfunctional proteins and organelles, is a cellular process associated with homeostasis. Autophagy functions in cell survival by breaking down proteins and organelles and recycling them to meet metabolic demands. However, aberrant up regulation of autophagy can function as an alternative to apoptosis. The duality of autophagy, and its regulation over cell survival/death, intimately links it with human disease. Non-coding RNAs regulate mRNA levels and elicit diverse effects on mammalian protein expression. The most studied non-coding RNAs to-date are microRNAs (miRNA). MicroRNAs function in post-transcriptional regulation, causing profound changes in protein levels, and affect many biological processes and diseases. The role and regulation of autophagy, whether it is beneficial or harmful, is a controversial topic in cardiovascular disease. A number of recent studies have identified miRNAs that target autophagy-related proteins and influence the development, progression, or treatment of cardiovascular disease. Understanding the mechanisms by which these miRNAs work can provide promising insight and potential progress towards the development of therapeutic treatments in cardiovascular disease.


Subject(s)
Autophagy/genetics , Cardiovascular Diseases/genetics , Cardiovascular Diseases/pathology , MicroRNAs/genetics , Animals , Autophagy/physiology , Cardiovascular Diseases/physiopathology , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/pathology , Diabetic Cardiomyopathies/physiopathology , Humans , MicroRNAs/metabolism , Models, Cardiovascular , Myocardial Ischemia/genetics , Myocardial Ischemia/pathology , Myocardial Ischemia/physiopathology , Ventricular Remodeling/genetics , Ventricular Remodeling/physiology
SELECTION OF CITATIONS
SEARCH DETAIL