首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 31 毫秒
1.
Myotubularin is a lipid phosphatase implicated in endosomal trafficking in vitro, but with an unknown function in vivo. Mutations in myotubularin cause myotubular myopathy, a devastating congenital myopathy with unclear pathogenesis and no current therapies. Myotubular myopathy was the first described of a growing list of conditions caused by mutations in proteins implicated in membrane trafficking. To advance the understanding of myotubularin function and disease pathogenesis, we have created a zebrafish model of myotubular myopathy using morpholino antisense technology. Zebrafish with reduced levels of myotubularin have significantly impaired motor function and obvious histopathologic changes in their muscle. These changes include abnormally shaped and positioned nuclei and myofiber hypotrophy. These findings are consistent with those observed in the human disease. We demonstrate for the first time that myotubularin functions to regulate PI3P levels in a vertebrate in vivo, and that homologous myotubularin-related proteins can functionally compensate for the loss of myotubularin. Finally, we identify abnormalities in the tubulo-reticular network in muscle from myotubularin zebrafish morphants and correlate these changes with abnormalities in T-tubule organization in biopsies from patients with myotubular myopathy. In all, we have generated a new model of myotubular myopathy and employed this model to uncover a novel function for myotubularin and a new pathomechanism for the human disease that may explain the weakness associated with the condition (defective excitation–contraction coupling). In addition, our findings of tubuloreticular abnormalities and defective excitation-contraction coupling mechanistically link myotubular myopathy with several other inherited muscle diseases, most notably those due to ryanodine receptor mutations. Based on our findings, we speculate that congenital myopathies, usually considered entities with similar clinical features but very disparate pathomechanisms, may at their root be disorders of calcium homeostasis.  相似文献   

2.
3.
Six years ago, DMM launched a subject collection called ‘Drosophila as a Disease Model’. This collection features Review-type articles and original research that highlight the power of Drosophila research in many aspects of human disease modeling. In the ensuing years, Drosophila research has further expanded to capitalize on genome editing, development of resources, and further interest in studying rare disease mechanisms. In the current issue of DMM, we again highlight the versatility, breadth, and scope of Drosophila research in human disease modeling and translational medicine. While many researchers have embraced the power of the fly, many more could still be encouraged to appreciate the strengths of Drosophila and how such research can integrate across species in a multi-pronged approach. Only when we truly acknowledge that all models contribute to our understanding of human biology, can we take advantage of the scope of current research endeavors.

Summary: This Editorial encourages us to embrace the power of the fly in studying human disease and highlights how Drosophila studies can be integrated with research in other species to further our understanding of human biology.

For over a century, scientists have used the fruit fly to learn about fundamental and evolutionarily conserved genetic and cellular processes. The pioneering work of Thomas Hunt Morgan and his students, in the early 20th century, proved that genes are located on chromosomes and led to the first chromosome linkage maps (Morgan, 1910). In the 1980s, Ed Lewis, Christiane Nüsslein-Volhard and Eric Wieschaus showed that individual genes could be mutated to cause characteristic embryonic patterning defects (Lewis, 1978; Nüsslein-Vollhard and Wieschaus, 1980). Their genetic studies allowed them to order genes within functional pathways through epistasis analyses. The genes they identified have counterparts across species and play key roles in development and disease from flies to humans. Indeed, much of the molecular circuitry for key signaling pathways, such as RAS, Notch, Hedgehog and Wnt, was elucidated in Drosophila (Ashton-Beaucage and Therrien, 2017; Bejsovec, 2018; Ingham, 2018; Salazar and Yamamoto, 2018). This rich history has established Drosophila as a powerful tool in biology, paving the way for further advances in basic and translational research.  相似文献   

4.
5.
6.
Summary: The launch of a new brand and website for DMM.Regular readers of Disease Models & Mechanisms (DMM) during 2015 will have noticed some changes to the journal. This is part of the gradual implementation of a new Company brand and migration to a new, better, web platform. This culminated in October with the launch of a new website for The Company of Biologists (www.biologists.com), and a brand new look and feel for the DMM website (dmm.biologists.org) and the articles it publishes (see Fig. 1). The new DMM website (and those of its sister journals Development, Journal of Cell Science, Journal of Experimental Biology and Biology Open) is the result of a mammoth project to ensure that users have an enhanced experience when visiting our pages. The site is easier to navigate and uncluttered, making it even quicker to search and find the content you need. We hope it looks good too. Open in a separate windowFig. 1.The Company of Biologists and DMM: supporting biologists, inspiring biology.Although our five journals are well known, fewer people are aware of the other areas of support that The Company of Biologists brings to the biological community. Our new brand will help us to increase the awareness of our work, and strengthen the links between our journals and charitable activities.  相似文献   

7.
8.
Destabilization of medial meniscus (DMM) model is an important tool for studying the pathophysiological roles of numerous arthritis associated molecules in the pathogenesis of osteoarthritis (OA) in vivo. However, the detailed, especially the visualized protocol for establishing this complicated model in mice, is not available. Herein we took advantage of wildtype and progranulin (PGRN)-/- mice as examples to introduce a protocol for inducing DMM model in mice, and compared the onset of OA following establishment of this surgically induced model. The operations performed on mice were either sham operation, which just opened joint capsule, or DMM operation, which cut the menisco-tibial ligament and caused destabilization of medial meniscus. Osteoarthritis severity was evaluated using histological assay (e.g. Safranin O staining), expressions of OA-associated genes, degradation of cartilage extracellular matrix molecules, and osteophyte formation. DMM operation successfully induced OA initiation and progression in both wildtype and PGRN-/- mice, and loss of PGNR growth factor led to a more severe OA phenotype in this surgically induced model.  相似文献   

9.
X-linked myotubular myopathy (XLMTM) is a congenital disorder caused by mutations of the myotubularin gene, MTM1. Myotubularin belongs to a large family of conserved lipid phosphatases that include both catalytically active and inactive myotubularin-related proteins (i.e., “MTMRs”). Biochemically, catalytically inactive MTMRs have been shown to form heteroligomers with active members within the myotubularin family through protein-protein interactions. However, the pathophysiological significance of catalytically inactive MTMRs remains unknown in muscle. By in vitro as well as in vivo studies, we have identified that catalytically inactive myotubularin-related protein 12 (MTMR12) binds to myotubularin in skeletal muscle. Knockdown of the mtmr12 gene in zebrafish resulted in skeletal muscle defects and impaired motor function. Analysis of mtmr12 morphant fish showed pathological changes with central nucleation, disorganized Triads, myofiber hypotrophy and whorled membrane structures similar to those seen in X-linked myotubular myopathy. Biochemical studies showed that deficiency of MTMR12 results in reduced levels of myotubularin protein in zebrafish and mammalian C2C12 cells. Loss of myotubularin also resulted in reduction of MTMR12 protein in C2C12 cells, mice and humans. Moreover, XLMTM mutations within the myotubularin interaction domain disrupted binding to MTMR12 in cell culture. Analysis of human XLMTM patient myotubes showed that mutations that disrupt the interaction between myotubularin and MTMR12 proteins result in reduction of both myotubularin and MTMR12. These studies strongly support the concept that interactions between myotubularin and MTMR12 are required for the stability of their functional protein complex in normal skeletal muscles. This work highlights an important physiological function of catalytically inactive phosphatases in the pathophysiology of myotubular myopathy and suggests a novel therapeutic approach through identification of drugs that could stabilize the myotubularin-MTMR12 complex and hence ameliorate this disorder.  相似文献   

10.
11.

Background

Non-invasive monitoring of disease progression in kidney disease is still a major challenge in clinical practice. In vivo near-infrared (NIR) imaging provides a new tool for studying disease mechanisms and non-invasive monitoring of disease development, even in deep organs. The LI-COR IRDye® 800CW RGD optical probe (RGD probe) is a NIR fluorophore, that can target integrin alpha v beta 3 (αvβ3) in tissues.

Objective

This study aims to monitor renal disease progression in an anti-glomerular basement membrane (GBM) nephritis mouse model.

Methods

Anti-GBM nephritis was induced in 129x1/svJ mice by anti-GBM serum challenge. The expression of integrin αvβ3 in the diseased kidney was examined by immunohistochemistry and quantitative polymerase chain reaction. The RGD probe and control fluorophores, the 800CW dye, and the BSA-conjugated 800CW dye, were administered into anti-GBM nephritic mice. LI-COR Pearl® Impulse imaging system was used for in vivo imaging; while ex vivo organ imaging was acquired using the MaestroTM imaging system.

Results

Kidney tissue from anti-GBM nephritic mice showed higher levels of integrin αvβ3 expression at both the protein and the mRNA level compared to normal mice. The RGD probe allowed in vivo renal imaging and the fluorescent signal could be specifically captured in the diseased kidneys up to 14 days, reflecting longitudinal changes in renal function.

Conclusion

The infrared RGD molecular probe that tracks integrin expression can be successfully used to monitor renal disease progression following immune-mediated nephritis.  相似文献   

12.

Introduction

Recent studies have revealed that rapamycin activates autophagy in human chondrocytes preventing the development of osteoarthritis (OA) like changes in vitro, while the systemic injection of rapamycin reduces the severity of experimental osteoarthritis in a murine model of OA in vivo. Since the systemic use of rapamycin is associated with numerous side effects, the goal of the current study was to examine the beneficial effect of local intra-articular injection of rapamycin in a murine model of OA and to elucidate the mechanism of action of rapamycin on articular cartilage.

Methods

Destabilization of the medial meniscus (DMM) was performed on 10-week-old male mice to induce OA. Intra-articular injections of 10 μl of rapamycin (10 μM) were administered twice weekly for 8 weeks. Articular cartilage damage was analyzed by histology using a semi-quantitative scoring system at 8 and 12 weeks after surgery. Mammalian target of rapamycin (mTOR), light chain 3 (LC3), vascular endothelial growth factor (VEGF), collagen, type X alpha 1 (COL10A1), and matrix metallopeptidase 13 (MMP13) expressions were analyzed by immunohistochemistry. VEGF, COL10A1, and MMP13 expressions were further examined via quantitative RT-PCR (qPCR).

Results

Intra-articular injection of rapamycin significantly reduced the severity of articular cartilage degradation at 8 and 12 weeks after DMM surgery. A reduction in mTOR expression and the activation of LC3 (an autophagy marker) in the chondrocytes was observed in the rapamycin treated mice. Rapamycin treatment also reduced VEGF, COL10A1, and MMP13 expressions at 8 and 12 weeks after DMM surgery.

Conclusion

These results demonstrate that the intra-articular injection of rapamycin could reduce mTOR expression, leading to a delay in articular cartilage degradation in our OA murine model. Our observations suggest that local intra-articular injection of rapamycin could represent a potential therapeutic approach to prevent OA.  相似文献   

13.
14.
Urinary biomarkers may offer a more sensitive and less invasive means to monitor kidney disease than traditional blood chemistry biomarkers such as creatinine. CD1pcy/pcy (pcy) mice have a slowly progressive disease phenotype that resembles human autosomal dominant polycystic kidney disease with renal cyst formation and inflammation. Previous reports suggest that dietary protein restriction may slow disease progression in mice and humans with polycystic kidney disease. Accordingly, we fed pcy mice either a standard chow (22.5% protein) or a protein-restricted (11.5% soy-based protein) diet from weaning until 34 wk of age. Every 6 wk we measured markers of kidney disease, including serum creatinine, BUN, and serum albumin as well as urinary monocyte chemoattractant protein 1 (MCP1), microalbumin, and specific gravity. Progression of kidney disease was equivalent for both diet groups despite dietary protein restriction. Urinary biomarkers proved useful for early detection of disease, in that urinary microalbumin was elevated as early as 22 wk of age and urinary MCP1 was increased by 28 wk of age, whereas increases in serum creatinine and BUN were detected later (at 34 wk of age) in both diet groups. Thus, urinary microalbumin and MCP1 analyses provided earlier, noninvasive indicators for detection of kidney disease and disease progression in pcy mice than did serum creatinine and BUN.Abbreviations: ADPKD, autosomal dominant polycystic kidney disease; MCP1, monocyte chemoattractant protein 1; PE diet, protein-restricted experimental dietAutosomal dominant polycystic kidney disease (ADPKD) is one of the most common heritable diseases in people and is the most frequently inherited nephropathy in North America.19 Mouse models of ADPKD have been described, in which mutant phenotypes result from spontaneous mutations or gene-specific targeting in mouse orthologs of human polycystic kidney disease genes.8 CD1pcy/pcy (pcy) mice, which have a mutated NPHP3 gene, develop similar renal pathology to human ADPKD including cyst development, interstitial nephritis, and fibrosis.8 The disease is transmitted as an autosomal recessive trait, and 100% affected offspring can be achieved by intercrossing homozygous pcy mice.24 The murine pcy phenotype recapitulates human ADPKD, with renal cyst location along the entire nephron and slow disease progression.8 Restricted protein diets have been reported to modulate the progression of polycystic kidney disease in humans and pcy mice.8,14 Compared with standard casein-based diets, soy-protein–based diets attenuated the disease course in one mouse study, in which feeding a low concentration of soy protein (6%) resulted in lower kidney weights, lower cyst scores (% cyst area times relative kidney weight), and reduced renal cyst growth in pcy mice at 23 wk of age.2 In addition, dietary fat type can influence kidney injury; for example, low or high amounts (7% or 20%) of flaxseed, a rich source of ω3 fatty acid and phytoestrogens, reportedly slowed early fibrosis progression in pcy mice, compared with diets containing either corn oil (rich in linoleic acid, an ω6 fatty acid, 18:2n-6) or an oil rich in docosahexaenoic acid, an ω3 fatty acid (22:6n-3).20Compared with traditional serum biomarkers such as creatinine and BUN, urinary microalbumin, creatinine, and monocyte chemoattractant protein (MCP1) are well-described renal biomarkers and early predictors of kidney disease progression in humans with polycystic kidney disease.26 Urinary biomarkers can provide an adjunct to traditional renal biomarkers to assess disease such as glomerular or tubular damage.12,16,28 Increased urinary albumin and MCP1 excretion are detected earlier than are altered glomerular filtration rate and azotemia in human ADPKD patients,28 and microalbuminuria is associated with disease progression.12,16 To assess the use of urinary biomarkers as a potentially more sensitive and less invasive means of monitoring and comparing kidney disease progression in different diet treatment groups, we fed pcy mice either a standard or protein-restricted diet and measured urinary microalbumin and MCP1 excretion from weaning until 34 wk of age, near end-stage kidney disease. These values were compared with concurrent serum creatinine, BUN, and albumin data. In addition, body weight and urine specific gravity were measured serially at the same time points, and CBC results and morphologic pathology were evaluated at the end of study.  相似文献   

15.

Introduction

Epigallocatechin 3-gallate (EGCG), a polyphenol present in green tea, was shown to exert chondroprotective effects in vitro. In this study, we used a posttraumatic osteoarthritis (OA) mouse model to test whether EGCG could slow the progression of OA and relieve OA-associated pain.

Methods

C57BL/6 mice were subjected to surgical destabilization of the medial meniscus (DMM) or sham surgery. EGCG (25 mg/kg) or vehicle control was administered daily for 4 or 8 weeks by intraperitoneal injection starting on the day of surgery. OA severity was evaluated using Safranin O staining and Osteoarthritis Research Society International (OARSI) scores, as well as by immunohistochemical analysis to detect cleaved aggrecan and type II collagen and expression of proteolytic enzymes matrix metalloproteinase 13 (MMP-13) and A disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTS5). Real-time PCR was performed to characterize the expression of genes critical for articular cartilage homeostasis. During the course of the experiments, tactile sensitivity testing (von Frey test) and open-field assays were used to evaluate pain behaviors associated with OA, and expression of pain expression markers and inflammatory cytokines in the dorsal root ganglion (DRG) was determined by real-time PCR.

Results

Four and eight weeks after DMM surgery, the cartilage in EGCG-treated mice exhibited less Safranin O loss and cartilage erosion, as well as lower OARSI scores compared to vehicle-treated controls, which was associated with reduced staining for aggrecan and type II collagen cleavage epitopes, and reduced staining for MMP-13 and ADAMTS5 in the articular cartilage. Articular cartilage in the EGCG-treated mice also exhibited reduced levels of Mmp1, Mmp3, Mmp8, Mmp13,Adamts5, interleukin 1 beta (Il1b) and tumor necrosis factor alpha (Tnfa) mRNA and elevated gene expression of the MMP regulator Cbp/p300 interacting transactivator 2 (Cited2). Compared to vehicle controls, mice treated with EGCG exhibited reduced OA-associated pain, as indicated by higher locomotor behavior (that is, distance traveled). Moreover, expression of the chemokine receptor Ccr2 and proinflammatory cytokines Il1b and Tnfa in the DRG were significantly reduced to levels similar to those of sham-operated animals.

Conclusions

This study provides the first evidence in an OA animal model that EGCG significantly slows OA disease progression and exerts a palliative effect.

Electronic supplementary material

The online version of this article (doi:10.1186/s13075-014-0508-y) contains supplementary material, which is available to authorized users.  相似文献   

16.
Previous studies have shown that tumor progression in the transgenic adenocarcinoma of mouse prostate (TRAMP) model is characterized by global DNA hypomethylation initiated during early-stage disease and locus-specific DNA hypermethylation occurring predominantly in late-stage disease. Here, we utilized Dnmt1 hypomorphic alleles to examine the role of Dnmt1 in normal prostate development and in prostate cancer in TRAMP. Prostate tissue morphology and differentiation status was normal in Dnmt1 hypomorphic mice, despite global DNA hypomethylation. TRAMP; Dnmt1 hypomorphic mice also displayed global DNA hypomethylation, but were characterized by altered tumor phenotype. Specifically, TRAMP; Dnmt1 hypomorphic mice exhibited slightly increased tumor incidence and significantly increased pathological progression at early ages and, conversely, displayed slightly decreased tumor incidence and significantly decreased pathological progression at advanced ages. Remarkably, hypomorphic Dnmt1 expression abrogated local and distant site macrometastases. Thus, Dnmt1 has tumor suppressor activity in early-stage prostate cancer, and oncogenic activity in late stage prostate cancer and metastasis. Consistent with the biological phenotype, epigenomic studies revealed that TRAMP; Dnmt1 hypomorphic mice show dramatically reduced CpG island and promoter DNA hypermethylation in late-stage primary tumors compared to control mice. Taken together, the data reveal a crucial role for Dnmt1 in prostate cancer and suggest that Dnmt1-targeted interventions may have utility specifically for advanced and/or metastatic prostate cancer.Changes in DNA methyltransferase (Dnmt) expression and DNA methylation are observed in human prostate cancer (3, 38, 41). Of particular interest, genes with tumor suppressive function become hypermethylated and silenced, which correlates with the development of specific disease phenotypes (2, 3, 38). Although an association between prostate cancer and alterations in DNA methylation has been established, in vivo models are required to determine whether these changes functionally contribute to the disease. In this context, studies in which pharmacological inhibitors of Dnmts were shown to inhibit prostate cancer in murine models have proven informative (34, 56). However, it remains unknown whether genetic disruption of epigenetic components, such as Dnmts, also impacts prostate cancer development. This is a critical question since the pharmacological inhibitors of Dnmts have pleiotropic effects, including those unrelated to activation of methylation-silenced genes (21, 23, 31). Moreover, no studies to date have examined whether Dnmts or DNA methylation play roles in normal prostate development; this information is vital to fully understanding the effects that inhibiting DNA methylation may have on prostate cancer.Dnmt1 is a maintenance DNA methyltransferase that propagates preexisting DNA methylation patterns in genomic DNA (44). Dnmt1 also is involved in de novo DNA methylation in cancer cells and interacts with other key epigenetic control molecules, including histone-modifying enzymes (11, 19). Murine models have been used to investigate the in vivo functions of Dnmt1. Complete genetic knockout of Dnmt1 is embryonic lethal in mice (29). However, hypomorphic expression of Dnmt1 allows murine development to proceed but causes global DNA hypomethylation and impacts cancer development and progression (7, 14, 28). Specifically, hypomorphic expression of Dnmt1 can lead to the development of lymphoma (14). Furthermore, crossing Dnmt1 hypomorphic mice with murine tumor models alters tumor progression, resulting in either increased or decreased tumor development, depending on the disease stage and tissue site (1, 7, 53). For example, reduced expression of Dnmt1 dramatically decreases intestinal polyp formation in ApcMin/+ mice, either alone or in combination with 5-aza-2′-deoxycytidine treatment (7, 27). However, it was later noted that reduced expression of Dnmt1 has a dual effect on intestinal cancer in ApcMin/+ mice, in which the development of early stage intestinal microadenomas is accelerated, whereas the formation of adenomatous polyps is significantly reduced (53). In addition, ApcMin/+ Dnmt1 hypomorphic mice develop liver cancer associated with the loss of heterozygosity of Apc (53). Similarly, in Dnmt1 hypomorphic mice crossed to Mlh1−/− mice, a dual effect was noted wherein mice developed fewer intestinal cancers but displayed increased T- and B-cell lymphomas (52). In addition, a recent study demonstrated that hypomorphic Dnmt1 expression is associated with reduced squamous cell carcinoma of the tongue and esophagus, resulting in decreased invasive cancer (1). Taken together, the data suggest that Dnmt1 has diverse effects on cancer development, which are dependent on tissue context and tumor stage.TRAMP is a well-established transgenic prostate cancer model driven by prostate-specific expression of the simian virus 40 (SV40) T/t oncogenes (16). TRAMP mice are characterized by Dnmt mRNA and protein overexpression, altered DNA methylation, and altered gene expression during prostate cancer development (2, 33, 35, 37). Of the three enzymatically active Dnmts, Dnmt1 shows the greatest level of overexpression in TRAMP, and this correlates with Rb inactivation, a key genetic event driving prostate cancer in the model (37). Most critically, global DNA hypomethylation occurs during early and late disease stages, while DNA hypermethylation occurs primarily at late disease stages in TRAMP (35).Here, we utilized Dnmt1 hypomorphic mice and the TRAMP model to assess the role of DNA methylation in both normal prostatic development and prostate cancer. The Dnmt1 hypomorphic mouse model used involves two different hypomorphic alleles (N and R), resulting in four genotypes with progressively reduced DNA methylation (Dnmt1+/+, Dnmt1R/+, Dnmt1N/+, and Dnmt1N/R) (7, 52). The N allele consists of a PGK-Neo insertion that deletes a portion of exon 4 of Dnmt1, resulting in severely reduced Dnmt1 expression, while the R allele involves a lacO insertion into intron 3 of Dnmt1, which partially reduces Dnmt1 expression (7, 52). Based on our previous work establishing the timing of DNA hypomethylation and DNA hypermethylation in TRAMP, we hypothesized that hypomorphic Dnmt1 expression in TRAMP may have tumor-promoting effects at early disease stages and tumor-inhibitory effects at later stages of prostate cancer progression. Our data are consistent with this hypothesis and, more importantly, reveal a critical and unanticipated role for Dnmt1 in prostate cancer metastasis.  相似文献   

17.

Background

Chagas disease is a neglected disease caused by the intracellular parasite Trypanosoma cruzi. Around 30% of the infected patients develop chronic cardiomyopathy or megasyndromes, which are high-cost morbid conditions. Immune response against myocardial self-antigens and exacerbated Th1 cytokine production has been associated with the pathogenesis of the disease. As IL-17 is involved in the pathogenesis of several autoimmune, inflammatory and infectious diseases, we investigated its role during the infection with T. cruzi.

Methodology/Principal Findings

First, we detected significant amounts of CD4, CD8 and NK cells producing IL-17 after incubating live parasites with spleen cells from normal BALB/c mice. IL-17 is also produced in vivo by CD4+, CD8+ and NK cells from BALB/c mice on the early acute phase of infection. Treatment of infected mice with anti-mouse IL-17 mAb resulted in increased myocarditis, premature mortality, and decreased parasite load in the heart. IL-17 neutralization resulted in increased production of IL-12, IFN-γ and TNF-α and enhanced specific type 1 chemokine and chemokine receptors expression. Moreover, the results showed that IL-17 regulates T-bet, RORγt and STAT-3 expression in the heart, showing that IL-17 controls the differentiation of Th1 cells in infected mice.

Conclusion/Significance

These results show that IL-17 controls the resistance to T. cruzi infection in mice regulating the Th1 cells differentiation, cytokine and chemokine production and control parasite-induced myocarditis, regulating the influx of inflammatory cells to the heart tissue. Correlations between the levels of IL-17, the extent of myocardial destruction, and the evolution of cardiac disease could identify a clinical marker of disease progression and may help in the design of alternative therapies for the control of chronic morbidity of chagasic patients.  相似文献   

18.
Bcl2-associated athanogene 3 (BAG3) mutations have been reported to cause the myofibrillar myopathy (MFM) which shows progressive limb muscle weakness, respiratory failure, and cardiomyopathy. Myopathy patients with BAG3 mutation are very rare. We described a patient showing atypical phenotypes. We aimed to find the genetic cause of Korean patients with sensory motor polyneuropathy, myopathy and rigid spine. We performed whole exome sequencing (WES) with 423 patients with sensory motor polyneuropathy. We found BAG3 mutation in one patient with neuropathy, myopathy and rigid spine syndrome, and performed electrophysiological study, whole body MRI and muscle biopsy on the patient. A de novo heterozygous p.Pro209Leu (c.626C>T) mutation in BAG3 was identified in a female myopathy. She first noticed a gait disturbance and spinal rigidity at the age of 11, and serum creatine kinase levels were elevated ninefolds than normal. She showed an axonal sensory-motor polyneuropathy like Charcot–Marie–Tooth disease (CMT), myopathy, rigid spine and respiratory dysfunction; however, she did not show any cardiomyopathy, which is a common symptom in BAG3 mutation. Lower limb MRI and whole spine MRI showed bilateral symmetric fatty atrophy of muscles at the lower limb and paraspinal muscles. When we track traceable MRI 1 year later, the muscle damage progressed slowly. As far as our knowledge, this is the first Korean patient with BAG3 mutation. We described a BAG3 mutation patient with atypical phenotype of CMT and myopathy, and those are expected to broaden the clinical spectrum of the disease and help to diagnose it.  相似文献   

19.
20.
First Person is a series of interviews with the first authors of a selection of papers published in Disease Models & Mechanisms, helping early-career researchers promote themselves alongside their papers. Chady Hakim is first author on ‘ Extensor carpi ulnaris muscle shows unexpected slow-to-fast fiber-type switch in Duchenne muscular dystrophy dogs’, published in DMM. Chady is a Research Assistant Professor in the lab of Dongsheng Duan at the University of Missouri, Colombia, MO, USA, investigating the preclinical development of gene therapy for Duchenne muscular dystrophy (DMD), with a particular interest in using the canine DMD model.

Chady Hakim How would you explain the main findings of your paper to non-scientific family and friends? DMD is a severe muscle disease caused by dystrophin deficiency. Loss of dystrophin leads to muscle degeneration and remodeling, and eventually to muscle death and replacement by fatty and fibrotic tissues. The canine DMD model shares clinical and pathophysiological similarities to that of human patients. Therefore, studies performed with the canine model provide critical insight into understanding muscle disease in DMD. In this study, we were first interested in developing a force assay platform to evaluate the contractile force and characterize the kinetic properties of a single muscle in the canine DMD model. We focused on the extensor carpi ulnaris (ECU) muscle from the forelimb muscle group. As expected, we saw a loss of muscle force in affected dogs. Surprisingly, we observed an unexpected contractile kinetic profile. It has been well established that the dystrophic muscle undergoes a fast-to-slow fiber-type switch. This led us to predict that the affected muscle would exhibit slow contraction and relaxation. Surprisingly, we saw just the opposite. There was a decrease in the time taken to reach peak tension and relax the affected ECU muscle, indicating a faster contraction and relaxation. Additional characterization of myofiber-type composition in the normal and affected ECU muscle confirmed the kinetic assay results.
“[…] studies performed with the canine model provide critical insight into understanding muscle disease in DMD.”
What are the potential implications of these results for your field of research? The unexpected slow-to-fast myofiber-type switch highlights the complexity of muscle remodeling in dystrophic large mammals and paves the way for better utilizing dystrophic canines as a preclinical model in the study of DMD pathogenesis. Additionally, the fiber-type switch phenomenon offers a unique entry point for (1) investigating the molecular mechanism(s) that lead to this phenomenon and how it directly correlates to the loss of dystrophin, and (2) evaluating the pathophysiological implications for muscle strength and in determining whether this is unique to canine muscle. Most importantly, these results have significant implications for therapeutic approaches to DMD, such as gene replacement and editing, and evaluating their efficacy in correcting the fiber-type switch. What are the main advantages and drawbacks of the model system you have used as it relates to the disease you are investigating? When initiating this study, our goal was to evaluate muscle strength in the affected dogs. To achieve this goal, we developed an all-in-one automated in situ force assay platform. This novel platform has several advantages. First, we designed all the components to be adjustable to meet the need for studying muscles at different anatomic locations or with different sizes. Our design was also made with the consideration to adopt the platform to accommodate other large animal models besides the canine. Second, we developed a detailed protocol to optimize the stimulation parameters, allowing the muscle to reach its optimal force during contraction. This allowed the comprehensive evaluation of the contractile and kinetic properties of a single muscle. Together, this novel platform offers a unique ability to correlate the physiological findings with the molecular, cellular, biochemical and histological changes in a single muscle. This ability is critical for evaluating preclinical intervention studies. Unfortunately, this is a terminal assay, limiting the investigators to follow disease progression and therapeutic response in the same animal over time. What has surprised you the most while conducting your research? It is well established that the dystrophic muscle undergoes a fast-to-slow, rather than a slow-to-fast, transition in fiber type. In this study, we observed the opposite in affected canine muscles as they were mainly composed of the fast fiber type. The underlying mechanism of this fiber-type switch needs to be further investigated so it can be determined whether it is unique to canine muscle. Furthermore, muscles that are mainly composed of the fast fiber type are characterized by a higher force, higher contraction and relaxation rate, and less time needed to achieve full contraction and relaxation. In affected dogs, we noticed a reduction in the time taken for contraction and relaxation. Surprisingly, the force, contraction rate and relaxation rate were significantly reduced in the affected muscle compared to the normal muscle. Open in a separate windowRepresentative myosin heavy chain isoform immunostaining photomicrographs of a normal (left) and an affected (right) ECU muscle. Blue, type I myofiber; red, type IIa myofiber; magenta, type I/IIa hybrid myofiber; green, laminin immunostaining Describe what you think is the most significant challenge impacting your research at this time and how will this be addressed over the next 10 years? Unlike other genetic diseases, DMD is very challenging to treat. First, it is caused by mutations in the second largest gene in the body. The large size of the dystrophin gene makes it impossible to replace it through the gene replacement approach unless a truncated gene with a similar function to the full-length gene is used. Second, DMD affects every muscle type in the body, making a whole-body treatment necessary to achieve a complete cure. Gene therapy using adeno-associated virus (AAV)-mediated CRISPR/Cas9 gene editing shows much promise for treating DMD. It allows for the restoration of a near full-length dystrophin protein without the need for using a highly truncated microgene that can only result in limited function rescue (Hakim et al. 2018). We recently showed that AAV CRISPR therapy resulted in efficient dystrophin restoration in affected dogs but resulted in a Cas9-specific immune response that eliminated the edited cell. This unfortunate response is a critical barrier to advancing CRISPR therapy into clinics. With further advances in gene therapy, combined with advances in the understanding of CRISPR genome editing and how to evade the Cas9-specific T-cell response, AAV CRISPR therapy will be suitable for treating DMD. What changes do you think could improve the professional lives of early-career scientists? I believe it is critical for early-career scientists to collaborate and interact with other related research fields. This empowers and extends their knowledge, and also has a positive influence on their research focus. Before I started my PhD, I had gained some knowledge about muscle physiology. I wanted to extend this knowledge in my PhD studies by building a bridge between muscle physiology and molecular biology, and the perfect application was the field of DMD gene therapy. Through my previous experience, I was able to develop tools, such as the platform presented in this study, to answer critical molecular questions in the field of gene therapy. As a matter of fact, the outcome observation of the fiber-type switch was a result of analyzing the kinetic properties of the affected muscle force. This observation will now become an important biomarker in the evaluation of the efficacy of novel therapy.
“I believe it is critical for early-career scientists to collaborate and interact with other related research fields.”
What''s next for you? With the novelty of the data presented in this study, I''m excited about finding out whether gene therapy approaches would correct the fiber-type switch and reverse the remodeling observed in the affected canine muscle. I''m currently working with my mentor Dr Dongsheng Duan to evaluate fiber-type composition-affected canine muscles treated with gene therapy.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号