Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Rheumatology (Oxford) ; 62(8): 2864-2871, 2023 08 01.
Article in English | MEDLINE | ID: mdl-36478205

ABSTRACT

OBJECTIVES: The B-cell depleting biologic, rituximab, is used to treat refractory autoimmune myositis. However, the beneficial effects of rituximab appear to outweigh the known contribution of B cells in myositis. We aimed to elucidate how myositis patients respond differently to rituximab and possible alternative mechanisms of action. METHODS: Here we have: (i) comprehensively investigated concurrent mRNA and microRNA expression in muscle biopsies taken at baseline and 16 weeks post treatment in 10 patients who were part of the rituximab in myositis (RIM) trial; and (ii) investigated the beneficial effect of rituximab on myositis muscle cells. RESULTS: Our analyses identified an increased number of changes in gene expression in biopsies from patients who had a clinical response to rituximab (n = 5) compared with non-responders (n = 5). The two groups had completely different changes in microRNA and mRNA expression following rituximab therapy, with the exception of one mRNA, BHMT2. Networks of mRNA and microRNA with opposite direction of expression changes highlighted ESR1 as upregulated in responders. We confirmed ESR1 upregulation upon rituximab treatment of immortalized myotubes and primary human dermatomyositis muscle cells in vitro, demonstrating a direct effect of rituximab on muscle cells. Notably, despite showing a response to rituximab, human dermatomyositis primary muscle cells did not express the rituximab target, CD20. However, these cells expressed a possible alternative target of rituximab, sphingomyelinase-like phosphodiesterase 3 b (SMPDL3B). CONCLUSION: In addition to B-cell depletion, rituximab may be beneficial in myositis due to increased ESR1 signalling mediated by rituximab binding to SMPDL3B on skeletal muscle cells.


Subject(s)
Dermatomyositis , MicroRNAs , Myositis , Humans , Rituximab/pharmacology , Rituximab/therapeutic use , Sphingomyelin Phosphodiesterase/therapeutic use , Dermatomyositis/drug therapy , Estrogen Receptor alpha , Myositis/drug therapy , Phosphoric Diester Hydrolases
2.
Article in English | MEDLINE | ID: mdl-36483401

ABSTRACT

Changes in antimicrobial use during the pandemic in relation to long-term trends in utilization among different antimicrobial stewardship program models have not been fully characterized. We analyzed data from an integrated health system using joinpoint regression and found temporal fluctuations in prescribing as well as continuation of existing trends.

4.
Skelet Muscle ; 10(1): 37, 2020 12 11.
Article in English | MEDLINE | ID: mdl-33308300

ABSTRACT

BACKGROUND: Nonsense or loss-of-function mutations in the non-lysosomal cysteine protease calpain-3 result in limb-girdle muscular dystrophy type 2A (LGMD2A). While calpain-3 is implicated in muscle cell differentiation, sarcomere formation, and muscle cytoskeletal remodeling, the physiological basis for LGMD2A has remained elusive. METHODS: Cell growth, gene expression profiling, and mitochondrial content and function were analyzed using muscle and muscle cell cultures established from healthy and calpain-3-deficient mice. Calpain-3-deficient mice were also treated with PPAR-delta agonist (GW501516) to assess mitochondrial function and membrane repair. The unpaired t test was used to assess the significance of the differences observed between the two groups or treatments. ANOVAs were used to assess significance over time. RESULTS: We find that calpain-3 deficiency causes mitochondrial dysfunction in the muscles and myoblasts. Calpain-3-deficient myoblasts showed increased proliferation, and their gene expression profile showed aberrant mitochondrial biogenesis. Myotube gene expression analysis further revealed altered lipid metabolism in calpain-3-deficient muscle. Mitochondrial defects were validated in vitro and in vivo. We used GW501516 to improve mitochondrial biogenesis in vivo in 7-month-old calpain-3-deficient mice. This treatment improved satellite cell activity as indicated by increased MyoD and Pax7 mRNA expression. It also decreased muscle fatigability and reduced serum creatine kinase levels. The decreased mitochondrial function also impaired sarcolemmal repair in the calpain-3-deficient skeletal muscle. Improving mitochondrial activity by acute pyruvate treatment improved sarcolemmal repair. CONCLUSION: Our results provide evidence that calpain-3 deficiency in the skeletal muscle is associated with poor mitochondrial biogenesis and function resulting in poor sarcolemmal repair. Addressing this deficit by drugs that improve mitochondrial activity offers new therapeutic avenues for LGMD2A.


Subject(s)
Calpain/metabolism , Mitochondria, Muscle/metabolism , Muscle Proteins/metabolism , Animals , Calpain/genetics , Cell Line , Cells, Cultured , Loss of Function Mutation , Mice , Mice, Inbred C57BL , Mitochondria, Muscle/pathology , Muscle Proteins/genetics , MyoD Protein/genetics , MyoD Protein/metabolism , Myoblasts/drug effects , Myoblasts/metabolism , Myoblasts/pathology , Organelle Biogenesis , PAX7 Transcription Factor/genetics , PAX7 Transcription Factor/metabolism , PPAR delta/agonists , Thiazoles/pharmacology
5.
J Genet Couns ; 29(4): 587-593, 2020 08.
Article in English | MEDLINE | ID: mdl-32196827

ABSTRACT

For poorly understood reasons, Black non-Hispanic (BNH) women meeting National Comprehensive Cancer Network (NCCN) criteria for genetic testing for breast cancer risk are less likely than White non-Hispanic (WNH) women to undergo testing (Armstrong, Micco, Carney, Stopfer, & Putt, JAMA, 293, 1729 and 2005). We compared physician referral rates and uptake for genetic testing of BNH and WNH women meeting select NCCN criteria (breast cancer under age 50, two primary breast cancers, triple-negative disease under age 60) in the Cancer Center at George Washington University (GWCC) between 2015 and 2018. Of the 723 BNH and WNH patients treated for breast cancer at GWCC, 28% met study criteria for genetic counseling referral (n = 252; BNH n = 115, WNH n = 137). Physician referral rates to genetic counseling differed significantly by race (BNH 75.7%, n = 87 and WNH 92.7%; n = 127; χ2  = 14.19, p-value < .01). Once referred, though, there was no significant difference in uptake of genetic counseling by race (BNH 95.4%, n = 83; WNH 97.6%, n = 124, χ2  = 1.33, p-value = .25) for patients appropriately referred.


Subject(s)
Black or African American , Breast Neoplasms/genetics , White People , Adult , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/ethnology , Ethnicity , Female , Genetic Counseling , Genetic Testing , Humans , Incidence , Middle Aged , Referral and Consultation , Risk Assessment
6.
Nat Commun ; 9(1): 3431, 2018 08 24.
Article in English | MEDLINE | ID: mdl-30143619

ABSTRACT

Duchenne muscular dystrophy (DMD) is a neuromuscular disorder causing progressive muscle degeneration. Although cardiomyopathy is a leading mortality cause in DMD patients, the mechanisms underlying heart failure are not well understood. Previously, we showed that NF-κB exacerbates DMD skeletal muscle pathology by promoting inflammation and impairing new muscle growth. Here, we show that NF-κB is activated in murine dystrophic (mdx) hearts, and that cardiomyocyte ablation of NF-κB rescues cardiac function. This physiological improvement is associated with a signature of upregulated calcium genes, coinciding with global enrichment of permissive H3K27 acetylation chromatin marks and depletion of the transcriptional repressors CCCTC-binding factor, SIN3 transcription regulator family member A, and histone deacetylase 1. In this respect, in DMD hearts, NF-κB acts differently from its established role as a transcriptional activator, instead promoting global changes in the chromatin landscape to regulate calcium genes and cardiac function.


Subject(s)
Muscular Dystrophy, Duchenne/metabolism , Myocytes, Cardiac/metabolism , NF-kappa B/metabolism , Animals , CCCTC-Binding Factor/metabolism , Calcium/metabolism , Cells, Cultured , Chromatin Assembly and Disassembly/genetics , Chromatin Assembly and Disassembly/physiology , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Male , Mice , Mice, Inbred mdx , Muscular Dystrophy, Duchenne/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction/physiology , Sin3 Histone Deacetylase and Corepressor Complex , Sodium-Calcium Exchanger/genetics , Sodium-Calcium Exchanger/metabolism
7.
J Clin Invest ; 127(10): 3796-3809, 2017 Oct 02.
Article in English | MEDLINE | ID: mdl-28891811

ABSTRACT

Macrophages are attracted to developing tumors and can participate in immune surveillance to eliminate neoplastic cells. In response, neoplastic cells utilize NF-κB to suppress this killing activity, but the mechanisms underlying their self-protection remain unclear. Here, we report that this dynamic interaction between tumor cells and macrophages is integrally linked by a soluble factor identified as growth and differentiation factor 15 (GDF-15). In vitro, tumor-derived GDF-15 signals in macrophages to suppress their proapoptotic activity by inhibiting TNF and nitric oxide (NO) production. In vivo, depletion of GDF-15 in Ras-driven tumor xenografts and in an orthotopic model of pancreatic cancer delayed tumor development. This delay correlated with increased infiltrating antitumor macrophages. Further, production of GDF-15 is directly regulated by NF-κB, and the colocalization of activated NF-κB and GDF-15 in epithelial ducts of human pancreatic adenocarcinoma supports the importance of this observation. Mechanistically, we found that GDF-15 suppresses macrophage activity by inhibiting TGF-ß-activated kinase (TAK1) signaling to NF-κB, thereby blocking synthesis of TNF and NO. Based on these results, we propose that the NF-κB/GDF-15 regulatory axis is important for tumor cells in evading macrophage immune surveillance during the early stages of tumorigenesis.


Subject(s)
Adenocarcinoma/immunology , Growth Differentiation Factor 15/immunology , Immunologic Surveillance , Macrophages/immunology , NF-kappa B/immunology , Neoplasm Proteins/immunology , Neoplasms, Experimental/immunology , Pancreatic Neoplasms/immunology , Signal Transduction/immunology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Female , Growth Differentiation Factor 15/genetics , Heterografts , MAP Kinase Kinase Kinases , Macrophages/pathology , Male , Mice , Mice, Knockout , NF-kappa B/genetics , Neoplasm Proteins/genetics , Neoplasm Transplantation , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Nitric Oxide/genetics , Nitric Oxide/immunology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Signal Transduction/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
8.
Cell Rep ; 17(2): 514-526, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27705798

ABSTRACT

MyoD is a key regulator of skeletal myogenesis that directs contractile protein synthesis, but whether this transcription factor also regulates skeletal muscle metabolism has not been explored. In a genome-wide ChIP-seq analysis of skeletal muscle cells, we unexpectedly observed that MyoD directly binds to numerous metabolic genes, including those associated with mitochondrial biogenesis, fatty acid oxidation, and the electron transport chain. Results in cultured cells and adult skeletal muscle confirmed that MyoD regulates oxidative metabolism through multiple transcriptional targets, including PGC-1ß, a master regulator of mitochondrial biogenesis. We find that PGC-1ß expression is cooperatively regulated by MyoD and the alternative NF-κB signaling pathway. Bioinformatics evidence suggests that this cooperativity between MyoD and NF-κB extends to other metabolic genes as well. Together, these data identify MyoD as a regulator of the metabolic capacity of mature skeletal muscle to ensure that sufficient energy is available to support muscle contraction.


Subject(s)
Mitochondria/metabolism , Muscle, Skeletal/metabolism , MyoD Protein/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Animals , Mice , Mitochondria/genetics , Muscle Contraction/genetics , Muscle Development/genetics , MyoD Protein/metabolism , Myoblasts/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Oxidation-Reduction , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Protein Binding , Signal Transduction , Transcription Factor RelB/genetics , Transcription Factor RelB/metabolism
9.
Dev Cell ; 36(2): 215-24, 2016 Jan 25.
Article in English | MEDLINE | ID: mdl-26777211

ABSTRACT

Skeletal muscle growth immediately following birth is critical for proper body posture and locomotion. However, compared with embryogenesis and adulthood, the processes regulating the maturation of neonatal muscles is considerably less clear. Studies in the 1960s predicted that neonatal muscle growth results from nuclear accretion of myoblasts preferentially at the tips of myofibers. Remarkably, little information has been added since then to resolve how myoblasts migrate to the ends of fibers. Here, we provide insight into this process by revealing a unique NF-κB-dependent communication between NG2(+) interstitial cells and myoblasts. NF-κB in NG2(+) cells promotes myoblast migration to the tips of myofibers through cell-cell contact. This occurs through expression of ephrinA5 from NG2(+) cells, which we further deduce is an NF-κB target gene. Together, these results suggest that NF-κB plays an important role in the development of newborn muscles to ensure proper myoblast migration for fiber growth.


Subject(s)
Antigens/metabolism , Cell Differentiation/physiology , Ephrin-A5/metabolism , Muscle Development/physiology , Myoblasts/metabolism , NF-kappa B/metabolism , Proteoglycans/metabolism , Animals , Animals, Newborn , Cell Movement/physiology , Male , Mice, Knockout , Mice, Transgenic , Muscle, Skeletal/metabolism
10.
PLoS One ; 10(3): e0119128, 2015.
Article in English | MEDLINE | ID: mdl-25760867

ABSTRACT

Few studies have empirically examined the suite of mechanisms that underlie the distributional shifts displayed by organisms in response to changing climatic condition. Mangrove forests are expected to move inland as sea-level rises, encroaching on saltmarsh plants inhabiting higher elevations. Mangrove propagules are transported by tidal waters and propagule dispersal is likely modified upon encountering the mangrove-saltmarsh ecotone, the implications of which are poorly known. Here, using an experimental approach, we record landward and seaward dispersal and subsequent establishment of mangrove propagules that encounter biotic boundaries composed of two types of saltmarsh taxa: succulents and grasses. Our findings revealed that propagules emplaced within saltmarsh vegetation immediately landward of the extant mangrove fringe boundary frequently dispersed in the seaward direction. However, propagules moved seaward less frequently and over shorter distances upon encountering boundaries composed of saltmarsh grasses versus succulents. We uniquely confirmed that the small subset of propagules dispersing landward displayed proportionately higher establishment success than those transported seaward. Although impacts of ecotones on plant dispersal have rarely been investigated in situ, our experimental results indicate that the interplay between tidal transport and physical attributes of saltmarsh vegetation influence boundary permeability to propagules, thereby directing the initial phase of shifting mangrove distributions. The incorporation of tidal inundation information and detailed data on landscape features, such as the structure of saltmarsh vegetation at mangrove boundaries, should improve the accuracy of models that are being developed to forecast mangrove distributional shifts in response to sea-level rise.


Subject(s)
Avicennia/physiology , Poaceae/physiology , Wetlands , Population Dynamics , Seawater
11.
Muscle Nerve ; 52(5): 795-802, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25728314

ABSTRACT

INTRODUCTION: We investigated the extent to which intercellular adhesion molecule-1 (ICAM-1), a critical protein of the inflammatory response, is expressed in skeletal muscles of mdx mice (a murine model of Duchenne muscular dystrophy). METHODS: Muscles were collected from control and mdx mice at 2-24 weeks of age and analyzed for ICAM-1 expression by means of Western blot and immunofluorescence. RESULTS: Western blot revealed higher expression of ICAM-1 in mdx compared with control muscles through 24 weeks of age. In contrast to control muscles, ICAM-1 was expressed on the membrane of damaged, regenerating, and normal myofibers of mdx mice. CD11b+ myeloid cells also expressed ICAM-1 in mdx muscles, and CD11b+ cells were closely associated with the membrane of myofibers expressing ICAM-1. CONCLUSIONS: These findings support a paradigm in which ICAM-1 and its localization to myofibers in muscles of mdx mice contributes to the dystrophic pathology.


Subject(s)
Intercellular Adhesion Molecule-1/biosynthesis , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Animals , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/diagnosis
12.
Skelet Muscle ; 4: 18, 2014.
Article in English | MEDLINE | ID: mdl-25789154

ABSTRACT

BACKGROUND: Duchenne muscular dystrophy (DMD) is caused by mutations in the dystrophin gene and afflicts skeletal and cardiac muscles. Previous studies showed that DMD is associated with constitutive activation of NF-κB, and in dystrophin-deficient mdx and utrophin/dystrophin (utrn (-/-) ;mdx) double knock out (dko) mouse models, inhibition of NF-κB with the Nemo Binding Domain (NBD) peptide led to significant improvements in both diaphragm and cardiac muscle function. METHODS: A trial in golden retriever muscular dystrophy (GRMD) canine model of DMD was initiated with four primary outcomes: skeletal muscle function, MRI of pelvic limb muscles, histopathologic features of skeletal muscles, and safety. GRMD and wild type dogs at 2 months of age were treated for 4 months with NBD by intravenous infusions. Results were compared with those collected from untreated GRMD and wild type dogs through a separate, natural history study. RESULTS: Results showed that intravenous delivery of NBD in GRMD dogs led to a recovery of pelvic limb muscle force and improvement of histopathologic lesions. In addition, NBD-treated GRMD dogs had normalized postural changes and a trend towards lower tissue injury on magnetic resonance imaging. Despite this phenotypic improvement, NBD administration over time led to infusion reactions and an immune response in both treated GRMD and wild type dogs. CONCLUSIONS: This GRMD trial was beneficial both in providing evidence that NBD is efficacious in a large animal DMD model and in identifying potential safety concerns that will be informative moving forward with human trials.

13.
Ecology ; 93(7): 1648-58, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22919911

ABSTRACT

Field experiments were conducted at a black mangrove-salt-marsh ecotone in southwest Florida (U.S.A.) to investigate retention of propagules of the black mangrove, Avicennia germinans, by salt-marsh plants as a mechanism of facilitation operating on recruitment success at landward boundaries. Buoyant A. germinans propagules are dispersed by tides, and stranding is required for establishment; therefore, processes that enable stranding should facilitate mangrove recruitment. We expected the physical structure of salt-marsh vegetation to define propagule retention capacity, and we predicted that salt-marsh plants with distinct growth forms would differentially retain propagules. Experimental monoculture plots (1 m2) of salt-marsh plants with different growth forms (Sporobolus virginicus [grass], Sesuvium portulacastrum [succulent forb], and Batis maritima [succulent scrub]) were created, and A. germinans propagules were emplaced into these plots and monitored over time. For comparison, propagules were also placed into natural polyculture plots (1 m2). Polyculture plots contained at least two of the salt-marsh plant taxa selected for monoculture treatments, and S. virginicus was always present within these polyculture plots. Natural polyculture plots retained 59.3% +/- 11.0% (mean +/- SE) of emplaced propagules. Monocultures varied in their propagule retention capacities with plots of S. virginicus retaining on average 65.7% +/- 11.5% of transplanted propagules compared to 7.2% +/- 1.8% by B. maritima and 5.0% +/- 1.9% by S. portulacastrum. Plots containing S. virginicus retained a significantly greater percentage of emplaced propagules relative to the two succulent salt-marsh taxa. Furthermore, propagule entrapment, across all treatments, was strongly correlated with salt-marsh structure (r2 = 0.6253, P = 0.00001), which was estimated using an indirect quantitative metric (lateral obstruction) calculated from digital images of plots. Overall, our findings imply that entrapment of propagules by salt-marsh plants may be facilitative if propagules are dispersed beyond the established tree line by spring or storm tides, and that facilitation may be sustained over time. We conclude that salt-marsh ecotone permeability may modulate landward encroachment by A. germinans, and that interactions among the early life history stages of black mangroves and neighboring plants may direct community responses to climate change.


Subject(s)
Avicennia/physiology , Tidal Waves , Wetlands , Florida , Population Dynamics
14.
Curr Top Dev Biol ; 96: 85-119, 2011.
Article in English | MEDLINE | ID: mdl-21621068

ABSTRACT

Muscle development, growth, and maintenance require an intricate and timely series of events initiated through a multitude of signaling pathways. The very nature of skeletal muscle requires tremendous plasticity to accommodate the need for anabolism or catabolism, and deregulation of these processes may be a tipping point in the development or progression of various skeletal muscle disorders. Among the relevant signaling pathways, NF-κB has emerged as a critical factor involved in various facets of muscle homeostasis. In this review, we summarize the NF-κB signaling pathway and provide a fresh perspective into the regulation and function of this transcription factor, underlying both the physiological and pathophysiological states of skeletal muscle.


Subject(s)
Muscle, Skeletal/metabolism , NF-kappa B/metabolism , Signal Transduction , Adaptation, Physiological , Animals , Humans , Muscle Development , Muscle, Skeletal/cytology
15.
J Transl Med ; 9: 68, 2011 May 17.
Article in English | MEDLINE | ID: mdl-21586145

ABSTRACT

BACKGROUND: Duchenne muscular dystrophy (DMD) is an inherited and progressive disease causing striated muscle deterioration. Patients in their twenties generally die from either respiratory or cardiac failure. In order to improve the lifespan and quality of life of DMD patients, it is important to prevent or reverse the progressive loss of contractile function of the heart. Recent studies by our labs have shown that the peptide NBD (Nemo Binding Domain), targeted at blunting Nuclear Factor κB (NF-κB) signaling, reduces inflammation, enhances myofiber regeneration, and improves contractile deficits in the diaphragm in dystrophin-deficient mdx mice. METHODS: To assess whether cardiac function in addition to diaphragm function can be improved, we investigated physiological and histological parameters of cardiac muscle in mice deficient for both dystrophin and its homolog utrophin (double knockout = dko) mice treated with NBD peptide. These dko mice show classic pathophysiological hallmarks of heart failure, including myocyte degeneration, an impaired force-frequency response and a severely blunted ß-adrenergic response. Cardiac contractile function at baseline and frequencies and pre-loads throughout the in vivo range as well as ß-adrenergic reserve was measured in isolated cardiac muscle preparations. In addition, we studied histopathological and inflammatory markers in these mice. RESULTS: At baseline conditions, active force development in cardiac muscles from NBD treated dko mice was more than double that of vehicle-treated dko mice. NBD treatment also significantly improved frequency-dependent behavior of the muscles. The increase in force in NBD-treated dko muscles to ß-adrenergic stimulation was robustly restored compared to vehicle-treated mice. However, histological features, including collagen content and inflammatory markers were not significantly different between NBD-treated and vehicle-treated dko mice. CONCLUSIONS: We conclude that NBD can significantly improve cardiac contractile dysfunction in the dko mouse model of DMD and may thus provide a novel therapeutic treatment for heart failure.


Subject(s)
Dystrophin/deficiency , Muscular Dystrophy, Animal/physiopathology , Myocardial Contraction/drug effects , NF-kappa B/antagonists & inhibitors , Peptides/pharmacology , Utrophin/deficiency , Animals , Disease Models, Animal , Dystrophin/metabolism , Mice , Mice, Knockout , Muscular Dystrophy, Animal/drug therapy , Myocardium/metabolism , Myocardium/pathology , NF-kappa B/metabolism , Peptides/therapeutic use , Receptors, Adrenergic, beta/metabolism , Utrophin/metabolism
16.
Mol Med ; 17(5-6): 508-15, 2011.
Article in English | MEDLINE | ID: mdl-21267511

ABSTRACT

Deterioration of diaphragm function is one of the prominent factors that contributes to the susceptibility of serious respiratory infections and development of respiratory failure in patients with Duchenne Muscular Dystrophy (DMD). The NF-κB signaling pathway has been implicated as a contributing factor of dystrophic pathology, making it a potential therapeutic target. Previously, we demonstrated that pharmacological inhibition of NF-κB via a small NEMO Binding Domain (NBD) peptide was beneficial for reducing pathological features of mdx mice. Now, we stringently test the effectiveness and clinical potential of NBD by treating mdx mice with various formulations of NBD and use diaphragm function as our primary outcome criteria. We found that administering DMSO-soluble NBD rescued 78% of the contractile deficit between mdx and wild-type (WT) diaphragm. Interestingly, synthesis of a GLP NBD peptide as an acetate salt permitted its solubility in water, but as a negative consequence, also greatly attenuated functional efficacy. However, replacing the acetic acid counterion of the NBD peptide with trifluoroacetic acid retained the peptide's water solubility and significantly restored mdx diaphragm contractile function and improved histopathological indices of disease in both diaphragm and limb muscle. Together, these results support the feasibility of using a mass-produced, water-soluble NBD peptide for clinical use.


Subject(s)
Diaphragm/drug effects , Muscle Contraction/drug effects , Muscular Dystrophy, Duchenne/drug therapy , NF-kappa B/metabolism , Peptides/therapeutic use , Animals , Electrophoretic Mobility Shift Assay , Female , Male , Mice , Mice, Inbred mdx , Muscular Dystrophy, Duchenne/metabolism , Peptides/chemical synthesis , Peptides/chemistry
17.
J Appl Physiol (1985) ; 106(1): 130-7, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18974369

ABSTRACT

We tested the hypothesis that cytokines derived from differentiated skeletal muscle cells in culture induce neutrophil chemotaxis after mechanical strain. Flexible-bottom plates with cultured human muscle cells attached were exposed to mechanical strain regimens (ST) of 0, 10, 30, 50, or 70 kPa of negative pressure. Conditioned media were tested for the ability to induce chemotaxis of human blood neutrophils in vitro and for a marker of muscle cell injury (lactate dehydrogenase). Conditioned media promoted neutrophil chemotaxis in a manner that was related both to the degree of strain and to the magnitude of muscle cell injury (ST 70 > ST 50 > ST 30). Protein profiling using a multiplex cytokine assay revealed that mechanical strain increased the presence of IL-8, granulocyte-macrophage colony-stimulating factor (GM-CSF), granulocyte colony-stimulating factor, monocyte chemotactic protein (MCP)-1, and IL-6 in conditioned media. We also detected 14 other cytokines in conditioned media from control cultures that did not respond to mechanical strain. Neutralization of IL-8 and GM-CSF completely inhibited the chemotactic response for ST 30 and ST 50 and reduced the chemotactic response for ST 70 by 40% and 47%, respectively. Neutralization of MCP-1 or IL-6 did not reduce chemotaxis after ST 70. This study enhances our understanding of the immunobiology of skeletal muscle by revealing that skeletal muscle cell-derived IL-8 and GM-CSF promote neutrophil chemotaxis after injurious mechanical strain.


Subject(s)
Chemotaxis, Leukocyte , Cytokines/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Interleukin-8/metabolism , Myoblasts, Skeletal/immunology , Neutrophils/immunology , Paracrine Communication , Cell Survival , Cells, Cultured , Chemokine CCL2/metabolism , Culture Media, Conditioned/metabolism , Female , Humans , Interleukin-6/metabolism , L-Lactate Dehydrogenase/metabolism , Myoblasts, Skeletal/enzymology , Myoblasts, Skeletal/pathology , Protein Array Analysis , Stress, Mechanical
18.
Int Rev Immunol ; 27(5): 375-87, 2008.
Article in English | MEDLINE | ID: mdl-18853344

ABSTRACT

Signaling through nuclear factor-kappa B (NF-kappaB) is emerging as an important regulator of muscle development, maintenance, and regeneration. Classic signaling modulates early muscle development by enhancing proliferation and inhibiting differentiation, and alternative signaling promotes myofiber maintenance and metabolism. Likewise, NF-kappaB signaling is critical for the development of immunity. Although these processes occur normally, dysregulation of NF-kappaB signaling has prohibitive effects on muscle growth and regeneration and can perpetuate inflammation in muscle diseases. Aberrant NF-kappaB signaling from immune and muscle cells has been detected and implicated in the pathologic progression of numerous dystrophies and myopathies, indicating that targeted NF-kappaB inhibitors may prove clinically beneficial.


Subject(s)
Dystrophin/metabolism , Muscular Diseases/immunology , Muscular Dystrophies/immunology , NF-kappa B/metabolism , Animals , Curcumin/pharmacology , Dystrophin/genetics , Dystrophin/immunology , Glucocorticoids/pharmacology , Humans , I-kappa B Kinase/metabolism , Inflammation , Mice , Mice, Inbred mdx , Muscle Development/immunology , Muscular Diseases/metabolism , Muscular Diseases/therapy , Muscular Dystrophies/metabolism , Muscular Dystrophies/therapy , NF-kappa B/immunology , Rats , Signal Transduction , Transcriptional Activation
19.
J Appl Physiol (1985) ; 101(5): 1394-9, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16840574

ABSTRACT

Tumor necrosis factor-alpha (TNF-alpha) has been associated with cachexia and is known to regulate multiple inflammatory cell (neutrophil and macrophage) responses. We tested the hypothesis that neutrophils and macrophages accumulate in the extensor digitorum longus (EDL) and soleus muscles of mice after chronic TNF-alpha administration. Murine recombinant TNF-alpha (approximately 100 microg x kg(-1) x day(-1)) in vehicle solution or vehicle solution alone (sham) was administered to C57BL/6 mice for 7 days via osmotic minipumps. In EDL muscles from TNF-alpha-treated mice, neutrophil and macrophage concentrations were elevated seven- and threefold, respectively, compared with sham mice. Neutrophil and macrophage concentrations were also elevated five- and twofold, respectively, in solei of TNF-alpha- relative to sham-treated mice. Treatment with TNF-alpha elevated ubiquitin content by approximately 25% relative to sham values for both the EDL and soleus muscles; however, these elevations were not statistically significant. No differences were observed between TNF-alpha- and sham-treated mice in body weight, food consumption, muscle mass, myofiber cross-sectional area, carbonyl groups, total protein content, or relative abundance of myosin heavy chain protein. Furthermore, no overt signs of muscle injury or regeneration were observed in muscles from TNF-alpha-treated mice in either the EDL or soleus muscles. These observations suggest that 7 days of TNF-alpha administration promote muscle inflammation as indicated by the accumulation of neutrophils and macrophages without overt signs of atrophy, injury, or regeneration.


Subject(s)
Macrophages/drug effects , Muscle, Skeletal/drug effects , Myositis/chemically induced , Neutrophils/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Animals , Infusion Pumps, Implantable , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/pathology , Myositis/pathology , Neutrophils/pathology , Recombinant Proteins/pharmacology , Ubiquitin/metabolism
20.
J Physiol ; 562(Pt 3): 899-913, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15550464

ABSTRACT

We tested the hypotheses that: (1) neutrophil accumulation after contraction-induced muscle injury is dependent on the beta(2) integrin CD18, (2) neutrophils contribute to muscle injury and oxidative damage after contraction-induced muscle injury, and (3) neutrophils aid the resolution of contraction-induced muscle injury. These hypotheses were tested by exposing extensor digitorum longus (EDL) muscles of mice deficient in CD18 (CD18(-/-); Itgb2(tm1Bay)) and of wild type mice (C57BL/6) to in situ lengthening contractions and by quantifying markers of muscle inflammation, injury, oxidative damage and regeneration/repair. Neutrophil concentrations were significantly elevated in wild type mice at 6 h and 3 days post-lengthening contractions; however, neutrophils remained at control levels at these time points in CD18-/- mice. These data indicate that CD18 is required for neutrophil accumulation after contraction-induced muscle injury. Histological and functional (isometric force deficit) signs of muscle injury and total carbonyl content, a marker of oxidative damage, were significantly higher in wild type relative to CD18-/- mice 3 days after lengthening contractions. These data show that neutrophils exacerbate contraction-induced muscle injury. After statistically controlling for differences in the force deficit at 3 days, wild type mice also demonstrated a higher force deficit at 7 days, a lower percentage of myofibres expressing embryonic myosin heavy chain at 3 and 7 days, and a smaller cross sectional area of central nucleated myofibres at 14 days relative to CD18-/- mice. These observations suggest that neutrophils impair the restoration of muscle structure and function after injury. In conclusion, neutrophil accumulation after contraction-induced muscle injury is dependent on CD18. Furthermore, neutrophils appear to contribute to muscle injury and impair some of the events associated with the resolution of contraction-induced muscle injury.


Subject(s)
CD18 Antigens/immunology , Muscle Contraction/immunology , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Atrophy/physiopathology , Neutrophils/immunology , Neutrophils/pathology , Animals , Male , Mice , Mice, Inbred C57BL , Muscle Proteins/metabolism , Muscle, Skeletal/injuries , Muscular Atrophy/pathology , Recovery of Function/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...