Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 180
Filter
Add more filters

Publication year range
1.
FASEB J ; 34(2): 2126-2146, 2020 02.
Article in English | MEDLINE | ID: mdl-31909533

ABSTRACT

Bladder dysfunction is associated with the overexpression of the intermediate filament (IF) proteins desmin and vimentin in obstructed bladder smooth muscle (BSM). However, the mechanisms by which these proteins contribute to BSM dysfunction are not known. Previous studies have shown that desmin and vimentin directly participate in signal transduction. In this study, we hypothesized that BSM dysfunction associated with overexpression of desmin or vimentin is mediated via c-Jun N-terminal kinase (JNK). We employed a model of murine BSM tissue in which increased expression of desmin or vimentin was induced by adenoviral transduction to examine the sufficiency of increased IF protein expression to reduce BSM contraction. Murine BSM strips overexpressing desmin or vimentin generated less force in response to KCl and carbachol relative to the levels in control murine BSM strips, an effect associated with increased JNK2 phosphorylation and reduced myosin light chain (MLC20 ) phosphorylation. Furthermore, desmin and vimentin overexpressions did not alter BSM contractility and MLC20 phosphorylation in strips isolated from JNK2 knockout mice. Pharmacological JNK2 inhibition produced results qualitatively similar to those caused by JNK2 knockout. These findings suggest that inhibition of JNK2 may improve diminished BSM contractility associated with obstructive bladder disease.


Subject(s)
Desmin/biosynthesis , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 9/metabolism , Muscle Contraction , Muscle, Smooth/metabolism , Urinary Bladder/metabolism , Vimentin/biosynthesis , Animals , Desmin/genetics , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 9/genetics , Muscle, Smooth/cytology , Urinary Bladder/cytology , Vimentin/genetics
2.
Scand J Immunol ; 87(4): e12645, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29446486

ABSTRACT

Studies show that the Th17/IL-17A axis plays an important role in the pathogenesis of kidney diseases. Previously, we also showed that IL-17A may play a role in the pathogenesis of primary nephrotic syndrome; however, the underlying mechanism(s) is unclear. The aim of this study was to explore the molecular mechanism of IL-17A-inducing podocyte injury in vitro. In this study, the NLRP3 inflammasome activation and the morphology of podocytes were detected by Western blot and immunofluorescence. The results showed that podocytes persistently expressed IL-17A receptor and that NLRP3 inflammasome in these cells was activated upon exposure to IL-17A. Also, activity of caspase-1 and secretion of IL-1ß increased in the presence of IL-17A. In addition, IL-17A disrupted podocyte morphology by decreasing expression of podocin and increasing expression of desmin. Blockade of intracellular ROS or inhibition of caspase-1 prevented activation of the NLRP3 inflammasome, thereby restoring podocyte morphology. Taken together, the results suggest that IL-17A induces podocyte injury by activating the NLRP3 inflammasome and IL-1ß secretion and contributes to disruption of the kidney's filtration system.


Subject(s)
Acute Kidney Injury/pathology , Caspase 1/metabolism , Interleukin-17/immunology , Interleukin-1beta/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Podocytes/pathology , Animals , Caspase Inhibitors/pharmacology , Cell Line , Desmin/biosynthesis , Glomerular Filtration Rate/physiology , Humans , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins/biosynthesis , Membrane Proteins/biosynthesis , Mice , Nephrotic Syndrome/pathology , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Th17 Cells/immunology
3.
Pediatr Surg Int ; 34(2): 217-225, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29043445

ABSTRACT

PURPOSE: Intermediate filaments (IFs) are a part of the cytoskeleton that extend throughout the cytoplasm of all cells and function in the maintenance of cell-shape by bearing tension and serving as structural components of the nuclear lamina. In normal intestine, IFs provide a tissue-specific three-dimensional scaffolding with unique context-dependent organizational features. The purpose of this study was to evaluate the role of IFs during intestinal adaptation in a rat model of short bowel syndrome (SBS). MATERIALS AND METHODS: Male rats were divided into two groups: Sham rats underwent bowel transection and SBS rats underwent a 75% bowel resection. Parameters of intestinal adaptation, enterocyte proliferation and apoptosis were determined 2 weeks after operation. Illumina's Digital Gene Expression (DGE) analysis was used to determine the cytoskeleton-related gene expression profiling. IF-related genes and protein expression were determined using real-time PCR, Western blotting and immunohistochemistry. RESULTS: Massive small bowel resection resulted in a significant increase in enterocyte proliferation and concomitant increase in cell apoptosis. From the total number of 20,000 probes, 16 cytoskeleton-related genes were investigated. Between these genes, only myosin and tubulin levels were upregulated in SBS compared to sham animals. Between IF-related genes, desmin, vimentin and lamin levels were down-regulated and keratin and neurofilament remain unchanged. The levels of TGF-ß, vimentin and desmin gene and protein were down-regulated in resected rats (vs sham animals). CONCLUSIONS: Two weeks following massive bowel resection in rats, the accelerated cell turnover was accompanied by a stimulated microfilaments and microtubules, and by inhibited intermediate filaments. Resistance to cell compression rather that maintenance of cell-shape by bearing tension are responsible for contraction, motility and postmitotic cell separation in a late stage of intestinal adaptation.


Subject(s)
Digestive System Surgical Procedures , Gene Expression Regulation , Intermediate Filaments/genetics , RNA/genetics , Short Bowel Syndrome/genetics , Animals , Apoptosis , Blotting, Western , Cell Proliferation , Desmin/biosynthesis , Desmin/genetics , Disease Models, Animal , Enterocytes/metabolism , Enterocytes/pathology , Immunohistochemistry , Intestine, Small/metabolism , Intestine, Small/pathology , Intestine, Small/surgery , Keratins/biosynthesis , Keratins/genetics , Lamins/biosynthesis , Lamins/genetics , Male , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Short Bowel Syndrome/metabolism , Short Bowel Syndrome/surgery , Vimentin/biosynthesis , Vimentin/genetics
4.
Am J Dermatopathol ; 39(7): 528-533, 2017 Jul.
Article in English | MEDLINE | ID: mdl-27655122

ABSTRACT

BACKGROUND: Osteogenic differentiation is rarely seen in melanomas, when it occurs it is mainly in acral lesions. METHODS: We report a case of an osteogenic melanoma in a 49-year-old woman who presented with a pigmented lesion in the subungueal region of her left hallux. The lesion was ulcerated and infiltrated until the deep dermis without bone involvement. RESULTS: The tumor was composed of pleomorphic atypical epithelioid and fusiform cells disposed in nests or cords, with vesicular nuclei and prominent central nucleoli. Focal lentiginous proliferation of large atypical melanocytes was present along the dermoepidermal junction. Areas of osteoid matrix focally mineralized were disposed in trabeculae, and there were islands of neoplastic cells. Immunohistochemistry revealed strong expression of S-100 protein and, unexpectedly, of desmin. Focal expression of Melan-A, microphthalmia transcription factor, and HMB-45 is also revealed. Mutations in BRAF and NRAS genes were not present. The patient was submitted to an amputation of the left hallux with negative sentinel lymph node. CONCLUSION: The importance of recognizing osteogenic melanoma is based on difficulties for histologic recognition and its differentials diagnosis.


Subject(s)
Biomarkers, Tumor/analysis , Desmin/biosynthesis , Melanoma/pathology , Nail Diseases/pathology , Skin Neoplasms/pathology , Desmin/analysis , Female , Humans , Melanoma/diagnosis , Middle Aged , Nail Diseases/diagnosis , Osteogenesis , Skin Neoplasms/diagnosis
5.
Pak J Pharm Sci ; 30(3(Special)): 1041-1045, 2017 May.
Article in English | MEDLINE | ID: mdl-28671078

ABSTRACT

In this paper, the author study on the effect of drug treatment on sports injury, and makes a comparative analysis of drug effects. In sports, the incidence of various types of injuries is increasing, especially in muscle injury. In the experiment, we compared the effects of three different drugs on the treatment and relief of muscle loss. After 3 weeks, the average optical density of desmin in muscle fiber positive region have decreased, as xiaotong plaster (0.4708±0.0126), votalin (0.5124±0.0264) and placebo (0.3856±0.0312). It has a certain effect to promote the repair and regeneration of desmin expression by drugs. Through the analysis of the effect of drug intervention on sports injury repair, we can effectively improve the therapeutic effect of sports injury.


Subject(s)
Diclofenac/analogs & derivatives , Diethylamines/therapeutic use , Fatigue/drug therapy , Resistance Training/adverse effects , Animals , Desmin/biosynthesis , Diclofenac/therapeutic use , Drugs, Chinese Herbal/administration & dosage , Male , Muscle Fibers, Fast-Twitch/metabolism , Rabbits , Time Factors
6.
Exp Cell Res ; 330(2): 300-310, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25304103

ABSTRACT

Pancreatic stellate cells (PSCs) play a key role in the dense desmoplastic stroma associated with pancreatic ductal adenocarcinoma. Studies on human PSCs have been minimal due to difficulty in maintaining primary PSC in culture. We have generated the first conditionally immortalized human non-tumor (NPSC) and tumor-derived (TPSC) pancreatic stellate cells via transformation with the temperature-sensitive SV40 large T antigen and human telomerase (hTERT). These cells proliferate at 33°C. After transfer to 37°C, the SV40LT is switched off and the cells regain their primary PSC phenotype and growth characteristics. NPSC contained cytoplasmic vitamin A-storing lipid droplets, while both NPSC and TPSC expressed the characteristic markers αSMA, vimentin, desmin and GFAP. Proteome array analysis revealed that of the 55 evaluated proteins, 27 (49%) were upregulated ≥3-fold in TPSC compared to NPSC, including uPA, pentraxin-3, endoglin and endothelin-1. Two insulin-like growth factor binding proteins (IGFBPs) were inversely expressed. Although discordant IGFBP-2 and IGFBP-3 levels, IGF-I was found to stimulate proliferation of both NPSC and TPSC. Both basal and IGF-I stimulated motility was significantly enhanced in TPSC compared to NPSC. In conclusion, these cells provide a unique resource that will facilitate further study of the active stroma compartment associated with pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Insulin-Like Growth Factor I/pharmacology , Pancreatic Ducts/pathology , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/metabolism , Antigens, Polyomavirus Transforming/genetics , Cell Culture Techniques , Cell Cycle/physiology , Cell Movement , Cell Proliferation , Desmin/biosynthesis , Glial Fibrillary Acidic Protein/biosynthesis , Humans , Insulin-Like Growth Factor Binding Protein 2/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor I/metabolism , Neoplasm Invasiveness/pathology , Primary Cell Culture , Smad Proteins/biosynthesis , Telomerase/genetics , Tumor Cells, Cultured , Vimentin/biosynthesis
7.
J Heart Valve Dis ; 25(1): 104-111, 2016 01.
Article in English | MEDLINE | ID: mdl-27989094

ABSTRACT

BACKGROUND: Prosthetic valve dysfunction due to pannus formation is a rare but serious complication. Currently, limited data are available concerning the pathogenesis and immunohistochemical properties of pannus. The study aim was to investigate the morphological, histopathological and immunohistochemical characteristics of pannus formation in patients with prosthetic valve dysfunction. METHODS: A total of 35 patients (10 males, 25 females; mean age 44 ± 16 years) who had undergone re-do valve surgery due to prosthetic valve obstruction was enrolled in the study. Immunohistochemical studies were aimed at evaluating the expression of alphasmooth muscle actin (α-SMA) and desmin in myofibroblasts and smooth muscle cells; epithelial membrane antigen (EMA) in epithelial cells; and CD34, Factor VIII and vascular endothelial growth factor (VEGF) in endothelial cells. Matrix metalloproteinases (MMPs) -2 and -9, and transforming growth factor-beta (TGF-ß) were used to demonstrate cytokine release from macrophages, leukocytes, fibroblasts and myofibroblasts. RESULTS: Pannus appeared as a tough and thick tissue hyperplasia which began from outside the suture ring in the periannular region and extended to the inflow and outflow surfaces of the prosthetic valves. Histopathological analysis showed the pannus tissue to consist of chronic inflammatory cells (lymphocytes, plasma cells, macrophages and foreign body giant cells), spindle cells such as myofibroblasts, capillary blood vessels and endothelial cells laying down the lumens. Calcification was present in the pannus tissue of 19 explanted prostheses. Immunohistochemical studies revealed positive α-SMA expression in all patients, whereas 60.5% of patients were positive for desmin, 50% for EMA, 42.1% for VEGF, 39.5% for TBF-ß, 42.1% for MMP-2, 86.8% for CD34, and 97.4% for Factor VIII. MMP-9 was negative in all patients. CONCLUSIONS: Pannus tissue appears to be formed as the result of a neointimal response in periannular regions of prosthetic valves that consist of periannular tissue migration, myofibroblast and extracellular matrix proliferation with vascular components. It is a chronic active process in which mediators such as TGF-ß, VEGF and MMP-2 play roles in both matrix formation and degradation.


Subject(s)
Heart Valve Diseases , Heart Valve Prosthesis Implantation , Heart Valve Prosthesis/adverse effects , Neointima/pathology , Actins/biosynthesis , Adult , Aged , Antigens, CD34/metabolism , Desmin/biosynthesis , Factor VIII/metabolism , Female , Fibroblasts/metabolism , Heart Valve Diseases/surgery , Humans , Macrophages/metabolism , Male , Matrix Metalloproteinases/metabolism , Middle Aged , Mucin-1/metabolism , Neointima/metabolism , Reoperation , Treatment Outcome , Vascular Endothelial Growth Factor A/metabolism
8.
Aviakosm Ekolog Med ; 50(1): 28-34, 2016.
Article in Russian | MEDLINE | ID: mdl-27344854

ABSTRACT

The signaling processes initiating proteolytic events in m. soleus of humans during short-term exposure in the non-weight bearing conditions were analyzed. Dry immersion (DI) was used to induce weight deprivation over 3 days. Western blotting was used to define the IRS-1 content, total and phosphorylated neuronal NO-synthase (nNOS), AMP-activated protein kinase (AMPK) that control the anabolic and catabolic pathways, and concentrations of cytoskeletal protein desmin and Ca²âº-activated protease calpin. Already on day-3 of DI calpain-dependent proteolysis manifests itself by reductions in both the total content and level of nNOS phosphorilation. Moreover, AMPK phosphorilation was decreased drastically.


Subject(s)
AMP-Activated Protein Kinases/biosynthesis , Muscle, Skeletal/metabolism , Nitric Oxide Synthase Type I/biosynthesis , Proteolysis , Calpain/biosynthesis , Desmin/biosynthesis , Humans , Immersion , Insulin Receptor Substrate Proteins/biosynthesis , Metabolism/genetics , Muscle, Skeletal/physiology
9.
Cytotherapy ; 17(9): 1213-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26276004

ABSTRACT

BACKGROUND AIMS: Bone marrow-derived mesenchymal stromal cells (BMSCs) are a promising therapeutic option for treating Duchenne muscular dystrophy (DMD). Myogenic differentiation occurs in the skeletal muscle of the mdx mouse (a mouse model of DMD) after BMSC transplantation. The transcription factor bone morphogenic protein 4 (BMP4) plays a crucial role in growth regulation, differentiation and survival of many cell types, including BMSCs. We treated BMSCs with BMP4 or the BMP antagonist noggin to examine the effects of BMP signaling on the myogenic potential of BMSCs in mdx mice. METHODS: We added BMP4 or noggin to cultured BMSCs under myogenic differentiation conditions. We then injected BMP4- or noggin-treated BMSCs into the muscles of mdx mice to determine their myogenic potential. RESULTS: We found that the expression levels of desmin and myosin heavy chain decreased after treating BMSCs with BMP4, whereas the expression levels of phosphorylated Smad, a downstream target of BMP4, were higher in these BMSCs than in the controls. Mdx mouse muscles injected with BMSCs pretreated with BMP4 showed decreased dystrophin expression and increased phosphorylated Smad levels compared with muscles injected with non-treated BMSCs. The opposite effects were seen after pretreatment with noggin, as expected. CONCLUSIONS: Our results identified BMP/Smad signaling as an essential negative regulator of promyogenic BMSC activity; inhibition of this pathway improved the efficiency of BMSC myogenic differentiation, which suggests that this pathway might serve as a target to regulate BMSC function for better myogenic differentiation during treatment of DMD and degenerative skeletal muscle diseases.


Subject(s)
Bone Morphogenetic Protein 4/pharmacology , Carrier Proteins/pharmacology , Cell- and Tissue-Based Therapy/methods , Mesenchymal Stem Cell Transplantation , Muscle Development/drug effects , Animals , Bone Marrow/metabolism , Bone Marrow Cells/cytology , Cell Differentiation , Desmin/biosynthesis , Disease Models, Animal , Dystrophin/metabolism , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred mdx , Muscle, Skeletal/cytology , Muscular Dystrophy, Duchenne/therapy , Myosin Heavy Chains/biosynthesis , Phosphorylation , Signal Transduction/drug effects , Smad Proteins/biosynthesis , Smad Proteins/metabolism
10.
Kidney Blood Press Res ; 40(4): 344-54, 2015.
Article in English | MEDLINE | ID: mdl-26160272

ABSTRACT

BACKGROUND/AIMS: Primary glomerulonephritis (PGN) is the most common reason inducing end stage renal disease in China, however, its pathogenesis remains unclear. The present study was designed to test the hypothesis that the formation and activation of NLRP3 (Nod-like receptor family pyrin domain containing 3) inflammasomes is an important initiating mechanism resulting in PGN. METHODS: Serum samples and frozen sections were collected from 38 cases with PGN, and renal tissues were obtained from 22 of them. NLRP3 inflammasomes were detected by RT-PCR and immunofluoresence methods. The relationship between NLRP3 and clinical/pathologic indexes was analyzed. RESULTS: RT-PCR analyses demonstrated that the mRNA levels of NLRP3 and caspase-1 genes were elevated significantly in renal tissues of PGN patients compared to those from normal pericarcinoma tissues. Moreover, the increased level of NLRP3 mRNA was correlative with a decrease in nephrin mRNA level and an increase in desmin mRNA level, which indicates that NLRP3 participates in podocyte injury in PGN patients. Immunofluorescence analysis also showed the protein expressions of NLRP3 and caspase-1 were increased in the glomeruli of PGN patients. Neverthless, there was no obvious regularity was presented in further subgroup analysis according to pathological types. In addition, increased NLRP3 was associated with the deterioration of renal function and glomerulosclerosis. IL-1ß, a product of NLRP3 inflammasome activation, had a significant correlation with proteinuria. CONCLUSIONS: The formation and activation of NLRP3 inflammasomes in podocytes has been importantly implicated in the development of PGN-associated glomerular injury.


Subject(s)
Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/metabolism , Inflammasomes/genetics , Adult , Caspase 1/metabolism , China , Desmin/biosynthesis , Desmin/genetics , Female , Glomerulosclerosis, Focal Segmental/pathology , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Kidney/chemistry , Kidney/enzymology , Kidney/pathology , Kidney Function Tests , Male , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Middle Aged , NLR Family, Pyrin Domain-Containing 3 Protein , Podocytes/pathology , Proteinuria/genetics , Proteinuria/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Retrospective Studies
11.
Histochem Cell Biol ; 141(1): 1-16, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24091796

ABSTRACT

Desmin, the muscle-specific intermediate filament, is involved in myofibrillar myopathies, dilated cardiomyopathy and muscle wasting. Desmin is the target of posttranslational modifications (PTMs) such as phosphorylation, ADP-ribosylation and ubiquitylation as well as nonenzymatic modifications such as glycation, oxidation and nitration. Several PTM target residues and their corresponding modifying enzymes have been discovered in human and nonhuman desmin. The major effect of phosphorylation and ADP-ribosylation is the disassembly of desmin filaments, while ubiquitylation of desmin leads to its degradation. The regulation of the desmin filament network by phosphorylation and ADP-ribosylation was found to be implicated in several major biological processes such as myogenesis, myoblast fusion, muscle contraction, muscle atrophy, cell division and possibly desmin interactions with its binding partners. Phosphorylation of desmin is also implicated in many forms of desmin-related myopathies (desminopathies). In this review, we summarize the findings on desmin PTMs and their implication in biological processes and pathologies, and discuss the current knowledge on the regulation of the desmin network by PTMs. We conclude that the desmin filament network can be seen as an intricate scaffold for muscle cell structure and biological processes and that its dynamics can be affected by PTMs. There are now precise tools to investigate PTMs and visualize cellular structures that have been underexploited in the study of desminopathies. Future studies should focus on these aspects.


Subject(s)
Cardiomyopathies/genetics , Desmin/genetics , Muscular Diseases/genetics , Muscular Dystrophies/genetics , Protein Processing, Post-Translational/genetics , Animals , Chickens , Cricetinae , Desmin/biosynthesis , Humans , Intermediate Filaments , Mice , Muscle Cells/cytology , Muscle Cells/metabolism , Muscle Contraction , Muscles/cytology
12.
J Muscle Res Cell Motil ; 35(5-6): 249-57, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25380573

ABSTRACT

Research on skeletal muscles suffers from a lack of appropriate human models to study muscle formation and regeneration on the regulatory level of single cells. This hampers both basic understanding and the development of new therapeutic approaches. The use of imaging multicolour flow cytometry and myogenic stem cells can help fill this void by allowing researchers to visualize and quantify the reaction of individual cultured cells to bioactives or other physiological impulses. As proof of concept, we subjected human CD56+ satellite cells to reference bioactives follistatin and Malva sylvestris extracts and then used imaging multicolor flow cytometry to visualize the stepwise activation of myogenic factors MyoD and myogenin in individual cells. This approach enabled us to evaluate the potency of these bioactives to stimulate muscle commitment. To validate this method, we used multi-photon confocal microscopy to confirm the potential of bioactives to stimulate muscle differentiation and expression of desmin. Imaging multicolor flow cytometry revealed statistically significant differences between treated and untreated groups of myogenic progenitors and we propose the utilization of this concept as an integral part of future muscle research strategies.


Subject(s)
Muscle, Skeletal/cytology , Satellite Cells, Skeletal Muscle/cytology , Stem Cells/cytology , CD56 Antigen/metabolism , Cell Differentiation/physiology , Cells, Cultured , Desmin/biosynthesis , Flow Cytometry/methods , Follistatin/pharmacology , Humans , Immunohistochemistry , Malva/chemistry , Microscopy, Confocal , Muscle Proteins/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , MyoD Protein/metabolism , Myogenin/metabolism , Plant Extracts/pharmacology , Satellite Cells, Skeletal Muscle/drug effects , Satellite Cells, Skeletal Muscle/metabolism , Single-Cell Analysis/methods , Stem Cells/drug effects , Stem Cells/metabolism
13.
Exp Physiol ; 99(5): 802-15, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24443353

ABSTRACT

A mutant non-inhibiting plasminogen activator inhibitor type 1 (PAI-1), termed PAI-1R, which reduces endogenous PAI-1 activity, has been shown to inhibit albuminuria and reduce glomerulosclerosis in experimental diabetes. The mechanism of the reduction of albuminuria is unclear. This study sought to determine whether the administration of PAI-1R protected podocytes from injury directly, thereby reducing albuminuria in the db/db mouse, a model of type 2 diabetes. Untreated uninephrectomized db/db mice developed significant mesangial matrix expansion and albuminuria at week 22 of age, associated with segmental podocyte foot-process effacement, reduction of renal nephrin, podocin and zonula occludin-1 production and induction of renal desmin and B7-1 generation. In contrast, treatment with PAI-1R at 0.5 mg (kg body weight)(-1) i.p., twice daily from week 20 to 22, reduced glomerular matrix accumulation, fibronectin and collagen production and albuminuria by 36, 62, 65 and 31%, respectively (P < 0.05), without affecting blood glucose level or body weight. Podocyte morphology and protein markers were also significantly attenuated by PAI-1R administration. Importantly, recombinant PAI-1 downregulated nephrin and zonula occludin-1 but increased desmin and B7-1 mRNA expression and protein production by podocytes in vitro, similar to the effects of transforming growth factor-ß1. These observations provide evidence that PAI-1, in a manner similar to transforming growth factor-ß1, directly induces podocyte injury, particularly in the setting of diabetes, where elevated PAI-1 may contribute to the progression of albuminuria. Reducing the increased PAI-1 activity by administration of PAI-1R, in fact, reduces podocyte injury, thereby reducing albuminuria. Therefore, PAI-1R provides an additional therapeutic effect in slowing the progression of diabetic nephropathy via the protection of podocytes.


Subject(s)
Diabetic Nephropathies/prevention & control , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/pharmacology , Podocytes/drug effects , Albuminuria/prevention & control , Animals , B7-1 Antigen/biosynthesis , Cells, Cultured , Desmin/biosynthesis , Diabetes Mellitus, Experimental/physiopathology , Disease Progression , Humans , Male , Mice , Mutation , Podocytes/metabolism , Zonula Occludens-1 Protein/biosynthesis
14.
Biofizika ; 59(5): 983-9, 2014.
Article in Russian | MEDLINE | ID: mdl-25730983

ABSTRACT

The aim of this study was to determine the transversal stiffness of the cortical cytoskeleton and the cytoskeletal protein desmin content in the left ventricle cardiomyocytes, fibers of the mouse soleus and tibialis anterior muscle after a 30-day space flight on board the "BION-M1" biosatellite (Russia, 2013). The dissection was made after 13-16.5 h after landing. The transversal stiffness was measured in relaxed and calcium activated state by, atomic force microscopy. The desmin content was estimated by western blotting, and the expression level of desmin-coding gene was detected using real-time PCR. The results indicate that, the transversal stiffness of the left ventricle cardiomyocytes and fibers of the soleus muscle in relaxed and activated states did not differ from the control. The transversal stiffness of the tibialis muscle fibers in relaxed and activated state was increased in the mice group after space flight. At the same time, in all types of studied tissues the desmin content and the expression level of desmin-coding gene did not differ from the control level.


Subject(s)
Desmin/biosynthesis , Heart Ventricles/metabolism , Muscle Fibers, Skeletal/metabolism , Myocytes, Cardiac/metabolism , Spacecraft , Weightlessness/adverse effects , Animals , Gene Expression Regulation , Heart Ventricles/pathology , Mice , Muscle Fibers, Skeletal/pathology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/pathology , Time Factors
15.
Am J Physiol Renal Physiol ; 304(7): F1000-8, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23389451

ABSTRACT

Deoxycorticosterone salt (DOC-salt) hypertension-induced renal damage is enhanced in α-calcitonin gene-related peptide (α-CGRP) knockout (KO) compared with wild-type (WT) mice. However, since the α-CGRP KO mice have a 15-20 mmHg higher baseline mean arterial pressure (MAP) than WT mice, they also have a higher MAP than WT mice throughout the course of DOC-salt hypertension. To determine the mechanism by which the absence of α-CGRP enhances hypertension-induced renal damage, DOC-salt hypertension was induced in telemetry probe implanted α-CGRP KO and WT mice. To equalize the blood pressure (BP) to that of DOC-salt WT mice, an additional group of DOC-salt α-CGRP KO mice was given 0.025% hydralazine to drink. The DOC-salt protocol increased the final MAP in α-CGRP KO mice to 155 ± 6 mmHg and in WT mice to 140 ± 5 mmHg. The MAP of the hydralazine-treated DOC-salt α-CGRP KO mice was 139 ± 6 mmHg. Urinary excretion of microalbumin and isoprostane, a marker for oxidative stress, was increased, and creatinine clearance was decreased in DOC-salt α-CGRP KO compared with DOC-salt WT mice. Equalization of the MAP in DOC-salt α-CGRP KO to that of DOC-salt WT mice did not significantly improve these parameters. Renal macrophage infiltration; desmin, a marker of podocyte damage; and the inflammatory cytokines TNF-α and IFN-γ and the chemokines monocyte chemoattractant protein-1 (MCP-1) and macrophage inflammatory protein-1α (MIP-1α) were increased in DOC-salt α-CGRP KO mice and were not reduced by hydralazine treatment. However, BP equalization did improve the renal histopathological damage, as determined by light microscopy. Therefore, in DOC-salt hypertension in mice, the mechanism(s) of the renal protective effects of α-CGRP are both BP independent and BP dependent.


Subject(s)
Calcitonin Gene-Related Peptide/physiology , Hypertension/physiopathology , Kidney/physiology , Animals , Arterial Pressure/drug effects , Calcitonin Gene-Related Peptide/genetics , Chemokines/metabolism , Cytokines/metabolism , Desmin/biosynthesis , Desoxycorticosterone , Hydralazine/pharmacology , Hypertension/chemically induced , Kidney/pathology , Macrophages/immunology , Mice , Mice, Knockout
16.
Am J Physiol Gastrointest Liver Physiol ; 304(9): G823-34, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23429582

ABSTRACT

Recently platelet-derived growth factor-α-positive cells (PDGFRα(+) cells), previously called "fibroblast-like" cells, have been described in the muscle layers of the gastrointestinal tract. These cells form networks and are involved in purinergic motor neurotransduction. Examination of colon from mice with enhanced green fluorescent protein (eGFP) driven from the endogenous Pdgfra (PDGFRα-eGFP mice) revealed a unique population of PDGFRα(+) cells in the mucosal layer of colon. We investigated the phenotype and potential role of these cells, which have not been characterized previously. Expression of PDGFRα and several additional proteins was surveyed in human and murine colonic mucosae by immunolabeling; PDGFRα(+) cells in colonic mucosa were isolated from PDGFRα-eGFP mice, and the gene expression profile was analyzed by quantitative polymerase chain reaction. We found for the first time that PDGFRα was expressed in subepithelial cells (subepithelial PDGFRα(+) cells) forming a pericryptal sheath from the base to the tip of crypts. These cells were in close proximity to the basolateral surface of epithelial cells and distinct from subepithelial myofibroblasts, which were identified by expression of α-smooth muscle actin and smooth muscle myosin. PDGFRα(+) cells also lay in close proximity to varicose processes of nerve fibers. Mouse subepithelial PDGFRα(+) cells expressed Toll-like receptor genes, purinergic receptor genes, 5-hydroxytryptamine (5-HT) 4 receptor gene, and hedgehog signaling genes. Subepithelial PDGFRα(+) cells occupy an important niche in the lamina propria and may function in transduction of sensory and immune signals and in the maintenance of mucosal homeostasis.


Subject(s)
Colon/cytology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Animals , Colon/metabolism , Desmin/biosynthesis , Humans , Intestinal Mucosa/cytology , Mice , Myofibroblasts/metabolism , Myosins/metabolism , Vimentin/biosynthesis
17.
Exp Mol Pathol ; 94(1): 79-83, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23099313

ABSTRACT

OBJECTIVE: This study investigates the effects of noradrenaline (NA) on cytoskeletal protein expression of vascular smooth muscle cells (VSMCs). METHODS: VSMCs were isolated from rat aortic tissue and cultured. The cultured VSMCs were divided into 4 experimental groups: (1) control group, (2) NA treatment group, (3) starvation group, and (4) NA treatment+starvation group. The expression of cytoskeletal protein (smooth muscle α-actin, ß-tubulin and desmin) was evaluated by (i) Coomassie blue staining, (ii) immunofluorescent staining, and (iii) RT-PCR and Western Blot. RESULTS: NA treatment significantly downregulated the expression of SM α-actin, ß-tubulin and desmin (P<0.05). The serum starvation did not affect the expression of cytoskeletal protein (SM α-actin, ß-tubulin and desmin), but when the cells were treated with the combination of NA and serum starvation, the expression of SM α-actin, ß-tubulin and desmin were down-regulated than those of the serum starvation group (P<0.05). CONCLUSION: These results suggested that NA might play a key role in regulating the cytoskeletal protein expression of VSMCs.


Subject(s)
Cytoskeletal Proteins/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Norepinephrine/pharmacology , Actins/biosynthesis , Actins/genetics , Animals , Aorta/metabolism , Cell Proliferation , Cells, Cultured , Cytoskeletal Proteins/genetics , Desmin/biosynthesis , Desmin/genetics , Down-Regulation , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/drug effects , Norepinephrine/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Starvation , Tubulin/biosynthesis , Tubulin/genetics , Vasoconstriction
18.
Heart Vessels ; 28(1): 101-13, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22241736

ABSTRACT

The aim of this work is to study cytoskeletal impairment during the development of ouabain-induced ventricular hypertrophy. Male Sprague-Dawley rats were treated with either ouabain or saline. Systolic blood pressure (SBP) was recorded weekly. At the end of the 3rd and 6th week, the rats were killed and cardiac mass index were measured. Hematoxylin-eosin and Sirius red staining were carried out and cardiac ultrastructure were studied using transmission electron microscopy. The mRNA level of Profilin-1, Desmin, PCNA, TGF-ß(1) and ET-1 in the left ventricle were measured using real-time quantitative PCR while their protein levels were examined by Western blot or immunohistochemistry. After 3 weeks, there was no significant difference in the mean SBP, cardiac mass index, mRNA and protein expression of PCNA, TGF-ß(1) and ET-1 between the two groups. However, ouabain-treated rats showed disorganized cardiac cytoskeleton with abnormal expression of Profilin-1 and Desmin. After 6 weeks, the cardiac mass index remained the same in the two groups while PCNA, TGF-ß(1), and ET-1 have been upregulated in ouabain-treated rats. The cardiac cytoskeletal impairment was more severe in ouabain-treated rats with further changes of Profilin-1 and Desmin. Cytoskeletal abnormality is an ultra-early change during ouabain-induced ventricular hypertrophy, before the release of hypertrophic factors. Therapy for prevention of ouabain-induced hypertrophy should start at the early stage by preventing the cytoskeleton from disorganization.


Subject(s)
Cytoskeleton/drug effects , Hypertrophy, Left Ventricular/pathology , Myocardium/ultrastructure , Ouabain/toxicity , Animals , Blood Pressure , Cytoskeleton/ultrastructure , Desmin/biosynthesis , Desmin/genetics , Disease Models, Animal , Disease Progression , Gene Expression Regulation/drug effects , Hypertrophy, Left Ventricular/chemically induced , Hypertrophy, Left Ventricular/physiopathology , Male , Microscopy, Electron, Transmission , Myocardium/metabolism , Profilins/biosynthesis , Profilins/genetics , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction
19.
Clin Exp Nephrol ; 17(4): 515-24, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23268284

ABSTRACT

BACKGROUND: SHRSP.Z-Lepr(fa)/IzmDmcr (SHRSP fatty) rats create a new animal model of metabolic syndrome. However, the renoprotective effect of telmisartan therapy and its underlying mechanisms in SHRSP fatty rats remain unknown. We evaluate the effects of long-term telmisartan therapy on renal dysfunction, podocyte injury, inflammation, and transforming growth factor-ß1 (TGF-ß1)/Smad, epithelial-mesenchymal transition (EMT), mitogen-activated protein kinase (MAPK), Rho-kinase, and cell-cycle progression pathway in the renal cortex of SHRSP fatty rats. METHODS: Seven-week-old male SHRSP fatty rats were treated with vehicle, telmisartan, and hydralazine for 8 weeks. Age-matched male Wistar-Kyoto/Izumo rats served as a control group. RESULTS: Vehicle-treated SHRSP fatty rats developed proteinuria and renal dysfunction, which in the telmisartan group was less than the vehicle and hydralazine group without changing blood pressure. Glomerulosclerosis and interstitial fibrosis were impaired in SHRSP fatty rats, and the renal damage in the telmisartan group was less than the vehicle and hydralazine groups. Decreased expression of nephrin and podocin and increased desmin-positive area in SHRSP fatty rats were restored by telmisartan but not hydralazine. TGF-ß1/Smad, EMT marker, MAPK, Rho-kinase, and cell-cycle progression pathways were upregulated in SHRSP fatty rats, and these increased proteins in the telmisartan group were less than the vehicle and hydralazine group. Telmisartan administration resulted in significant suppression in tumor necrosis factor-α expression and nuclear factor-κB phosphorylation. CONCLUSION: Long-term telmisartan therapy may improve renal dysfunction, glomerulosclerosis, podocyte injury, and inflammation associated with EMT, TGF-ß/Smad, MAPK, Rho-kinase pathway in SHRSP fatty rats. Thus, telmisartan may have significant therapeutic potential for metabolic syndrome.


Subject(s)
Benzimidazoles/therapeutic use , Benzoates/therapeutic use , Kidney Diseases/prevention & control , Nephritis/drug therapy , Animals , Cell Cycle Proteins/biosynthesis , Desmin/biosynthesis , Epithelial-Mesenchymal Transition/drug effects , Hydralazine/therapeutic use , Male , Metabolic Syndrome/drug therapy , Nephritis/metabolism , Podocytes/drug effects , Podocytes/pathology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Signal Transduction/drug effects , Smad4 Protein/physiology , Telmisartan , Transforming Growth Factor beta1/metabolism , Tumor Necrosis Factor-alpha/metabolism
20.
Biochem Biophys Res Commun ; 428(2): 309-14, 2012 Nov 16.
Article in English | MEDLINE | ID: mdl-23085232

ABSTRACT

Matrix metalloproteinase-1 (MMP-1) is a member of the family of zinc-dependent endopeptidases that are capable of degrading extracellular matrix (ECM) and certain non-matrix proteins. It has been shown that MMP-1 can enhance muscle regeneration by improving the differentiation and migration of myoblasts. However, it is still not known whether MMP-1 can promote the myogenesis of bone marrow-derived mesenchymal stem cells (BMSCs). To address this question, we isolated BMSCs from C57BL/6J mice and investigated the effects of MMP-1 on their proliferation and myogenic differentiation. Our results showed that MMP-1 treatment, which had no cytotoxic effects on BMSCs, increased the mRNA and protein levels of MyoD and desmin in a dose-dependent manner, indicating that MMP-1 promoted myogenic differentiation of BMSCs in vitro. These results suggest that BMSCs may have a therapeutic potential for treating muscular disorders.


Subject(s)
Cell Differentiation/drug effects , Matrix Metalloproteinase 1/pharmacology , Mesenchymal Stem Cells/drug effects , Muscle Development/drug effects , Adipogenesis/drug effects , Animals , Bone Marrow Cells/cytology , Cell Separation , Cells, Cultured , Desmin/biosynthesis , Male , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred C57BL , MyoD Protein/biosynthesis , Osteogenesis/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL