Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Ravi Kambadur is active.

Publication


Featured researches published by Ravi Kambadur.


Journal of Cellular Physiology | 1999

Myostatin, a transforming growth factor-β superfamily member, is expressed in heart muscle and is upregulated in cardiomyocytes after infarct

Mridula Sharma; Ravi Kambadur; Kenneth G. Matthews; Wayne G. Somers; G. Devlin; John V. Conaglen; Peter J. Fowke; John J. Bass

Myostatin is a secreted growth and differentiating factor (GDF‐8) that belongs to the transforming growth factor‐beta (TGF‐β) superfamily. Targeted disruption of the myostatin gene in mice and a mutation in the third exon of the myostatin gene in double‐muscled Belgian Blue cattle breed result in skeletal muscle hyperplasia. Hence, myostatin has been shown to be involved in the regulation of skeletal muscle mass in both mice and cattle. Previous published reports utilizing Northern hybridization had shown that myostatin expression was seen exclusively in skeletal muscle. A significantly lower level of myostatin mRNA was also reported in adipose tissue. Using a sensitive reverse transcription‐polymerase chain reaction (RT‐PCR) technique and Western blotting with anti‐myostatin antibodies, we show that myostatin mRNA and protein are not restricted to skeletal muscle. We also show that myostatin expression is detected in the muscle of both fetal and adult hearts. Sequence analysis reveals that the Belgian Blue heart myostatin cDNA sequence contains an 11 nucleotide deletion in the third exon that causes a frameshift that eliminates virtually all of the mature, active region of the protein. Anti‐myostatin immunostaining on heart sections also demonstrates that myostatin protein is localized in Purkinje fibers and cardiomyocytes in heart tissue. Furthermore, following myocardial infarction, myostatin expression is upregulated in the cardiomyocytes surrounding the infarct area. Given that myostatin is expressed in fetal and adult hearts and that myostatin expression is upregulated in cardiomyocytes after the infarction, myostatin could play an important role in cardiac development and physiology. J. Cell. Physiol. 180:1–9, 1999.


Journal of Cellular Physiology | 2006

Myostatin induces cachexia by activating the ubiquitin proteolytic system through an NF-κB-independent, FoxO1-dependent mechanism†

Craig McFarlane McFarlane; Erin Plummer; Mark Thomas; Alex Hennebry; Murray Ashby; Nicholas Ling; Heather K. Smith; Mridula Sharma; Ravi Kambadur

Myostatin, a transforming growth factor‐beta (TGF‐β) super‐family member, has been well characterized as a negative regulator of muscle growth and development. Myostatin has been implicated in several forms of muscle wasting including the severe cachexia observed as a result of conditions such as AIDS and liver cirrhosis. Here we show that Myostatin induces cachexia by a mechanism independent of NF‐κB. Myostatin treatment resulted in a reduction in both myotube number and size in vitro, as well as a loss in body mass in vivo. Furthermore, the expression of the myogenic genes myoD and pax3 was reduced, while NF‐κB (the p65 subunit) localization and expression remained unchanged. In addition, promoter analysis has confirmed Myostatin inhibition of myoD and pax3. An increase in the expression of genes involved in ubiquitin‐mediated proteolysis is observed during many forms of muscle wasting. Hence we analyzed the effect of Myostatin treatment on proteolytic gene expression. The ubiquitin associated genes atrogin‐1, MuRF‐1, and E214k were upregulated following Myostatin treatment. We analyzed how Myostatin may be signaling to induce cachexia. Myostatin signaling reversed the IGF‐1/PI3K/AKT hypertrophy pathway by inhibiting AKT phosphorylation thereby increasing the levels of active FoxO1, allowing for increased expression of atrophy‐related genes. Therefore, our results suggest that Myostatin induces cachexia through an NF‐κB‐independent mechanism. Furthermore, increased Myostatin levels appear to antagonize hypertrophy signaling through regulation of the AKT‐FoxO1 pathway. J. Cell. Physiol. 209: 501–514, 2006.


Journal of Cell Science | 2005

Improved muscle healing through enhanced regeneration and reduced fibrosis in myostatin-null mice

Seumas McCroskery; Mark G. Thomas; Leanne Platt; Alex Hennebry; Takanori Nishimura; Lance McLeay; Mridula Sharma; Ravi Kambadur

Numerous stimulatory growth factors that can influence muscle regeneration are known. Recently, it has been demonstrated that neutralization of muscle growth inhibitory factors, such as myostatin (Mstn; also known as growth differentiation factor 8, Gdf8), also leads to increased muscle regeneration in mdx mice that are known to have cycles of degeneration. However, the precise mechanism by which Mstn regulates muscle regeneration has not yet been fully determined. To investigate the role of Mstn in adult skeletal muscle regeneration, wild-type and myostatin-null (Mstn-/-) mice were injured with notexin. Forty-eight hours after injury, accelerated migration and enhanced accretion of myogenic cells (MyoD1+) and macrophages (Mac-1+) was observed at the site of regeneration in Mstn-/- muscle as compared with wild-type muscle. Inflammatory cell numbers decreased more rapidly in the Mstn-/- muscle, indicating that the whole process of inflammatory cell response is accelerated in Mstn-/- mice. Consistent with this result, the addition of recombinant Mstn reduced the activation of satellite cells (SCs) and chemotactic movements of both myoblasts and macrophages ex vivo. Examination of regenerated muscle (28 days after injury) also revealed that Mstn-/- mice showed increased expression of decorin mRNA, reduced fibrosis and improved healing as compared with wild-type mice. On the basis of these results, we propose that Mstn negatively regulates muscle regeneration not only by controlling SC activation but also by regulating the migration of myoblasts and macrophages to the site of injury. Thus, antagonists of Mstn could potentially be useful as pharmacological agents for the treatment of disorders of overt degeneration and regeneration.


Molecular and Cellular Biology | 2002

The Myostatin Gene Is a Downstream Target Gene of Basic Helix-Loop-Helix Transcription Factor MyoD

Michael P. Spiller; Ravi Kambadur; Ferenc Jeanplong; Mark G. Thomas; Julie K. Martyn; John J. Bass; Mridula Sharma

ABSTRACT Myostatin is a negative regulator of myogenesis, and inactivation of myostatin leads to heavy muscle growth. Here we have cloned and characterized the bovine myostatin gene promoter. Alignment of the upstream sequences shows that the myostatin promoter is highly conserved during evolution. Sequence analysis of 1.6 kb of the bovine myostatin gene upstream region revealed that it contains 10 E-box motifs (E1 to E10), arranged in three clusters, and a single MEF2 site. Deletion and mutation analysis of the myostatin gene promoter showed that out of three important E boxes (E3, E4, and E6) of the proximal cluster, E6 plays a significant role in the regulation of a reporter gene in C2C12 cells. We also demonstrate by band shift and chromatin immunoprecipitation assay that the E6 E-box motif binds to MyoD in vitro and in vivo. Furthermore, cotransfection experiments indicate that among the myogenic regulatory factors, MyoD preferentially up-regulates myostatin promoter activity. Since MyoD expression varies during the myoblast cell cycle, we analyzed the myostatin promoter activity in synchronized myoblasts and quiescent “reserve” cells. Our results suggest that myostatin promoter activity is relatively higher during the G1 phase of the cell cycle, when MyoD expression levels are maximal. However, in the reserve cells, which lack MyoD expression, a significant reduction in the myostatin promoter activity is observed. Taken together, these results suggest that the myostatin gene is a downstream target gene of MyoD. Since the myostatin gene is implicated in controlling G1-to-S progression of myoblasts, MyoD could be triggering myoblast withdrawal from the cell cycle by regulating myostatin gene expression.


Cell Metabolism | 2012

RETRACTED: The Ubiquitin Ligase Mul1 Induces Mitophagy in Skeletal Muscle in Response to Muscle-Wasting Stimuli

Sudarsanareddy Lokireddy; Isuru Wijerupage Wijesoma; Serena Teng; Sabeera Bonala; Peter D. Gluckman; Craig McFarlane; Mridula Sharma; Ravi Kambadur

Recent research reveals that dysfunction and subsequent loss of mitochondria (mitophagy) is a potent inducer of skeletal muscle wasting. However, the molecular mechanisms that govern the deregulation of mitochondrial function during muscle wasting are unclear. In this report, we show that different muscle-wasting stimuli upregulated mitochondrial E3 ubiquitin protein ligase 1 (Mul1), through a mechanism involving FoxO1/3 transcription factors. Overexpression of Mul1 in skeletal muscles and myoblast cultures was sufficient for the induction of mitophagy. Consistently, Mul1 suppression not only protected against mitophagy but also partially rescued the muscle wasting observed in response to muscle-wasting stimuli. In addition, upregulation of Mul1, while increasing mitochondrial fission, resulted in ubiquitination and degradation of the mitochondrial fusion protein Mfn2. Collectively, these data explain the molecular basis for the loss of mitochondrial number during muscle wasting.


Journal of Cellular Physiology | 2000

Myostatin regulation during skeletal muscle regeneration

Sonnie P. Kirk; Jenny M. Oldham; Ravi Kambadur; Mridula Sharma; Pete Dobbie; John J. Bass

Myostatin, a member of the TGF‐β superfamily, is a key negative regulator of skeletal muscle growth. The role of myostatin during skeletal muscle regeneration has not previously been reported. In the present studies, normal Sprague‐Dawley and growth hormone (GH)‐deficient (dw/dw) rats were administered the myotoxin, notexin, in the right M. biceps femoris on day 0. The dw/dw rats then received either saline or human‐N‐methionyl GH (200μg/100g body weight/day) during the ensuing regeneration. Normal and dw/dw M. biceps femoris were dissected on days 1, 2, 3, 5, 9 and 13, formalin‐fixed, then immunostained for myostatin protein. Immunostaining for myostatin revealed high levels of protein within necrotic fibres and connective tissue of normal and dw/dw damaged muscles. Regenerating myotubes contained no myostatin at the time of fusion (peak fusion on day 5), and only low levels of myostatin were observed during subsequent myotube enlargement. Fibres which survived assault by notexin (survivor fibres) contained moderate to high myostatin immunostaining initially. The levels in both normal and dw/dw rat survivor fibres decreased on days 2–3, then increased on days 9–13. In dw/dw rats, there was no observed effect of GH administration on the levels of myostatin protein in damaged muscle. The low level of myostatin observed in regenerating myotubes in these studies suggests a negative regulatory role for myostatin in muscle regeneration. J. Cell. Physiol. 184:356–363, 2000.


American Journal of Physiology-cell Physiology | 2009

Myostatin regulates fiber-type composition of skeletal muscle by regulating MEF2 and MyoD gene expression

Alex Hennebry; Carole Berry; Victoria Siriett; Paul O'Callaghan; Linda Chau; Trevor Watson; Mridula Sharma; Ravi Kambadur

Myostatin (Mstn) is a secreted growth factor belonging to the tranforming growth factor (TGF)-beta superfamily. Inactivation of murine Mstn by gene targeting, or natural mutation of bovine or human Mstn, induces the double muscling (DM) phenotype. In DM cattle, Mstn deficiency increases fast glycolytic (type IIB) fiber formation in the biceps femoris (BF) muscle. Using Mstn null ((-/-)) mice, we suggest a possible mechanism behind Mstn-mediated fiber-type diversity. Histological analysis revealed increased type IIB fibers with a concomitant decrease in type IIA and type I fibers in the Mstn(-/-) tibialis anterior and BF muscle. Functional electrical stimulation of Mstn(-/-) BF revealed increased fatigue susceptibility, supporting increased type IIB fiber content. Given the role of myocyte enhancer factor 2 (MEF2) in oxidative type I fiber formation, MEF2 levels in Mstn(-/-) tissue were quantified. Results revealed reduced MEF2C protein in Mstn(-/-) muscle and myoblast nuclear extracts. Reduced MEF2-DNA complex was also observed in electrophoretic mobility-shift assay using Mstn(-/-) nuclear extracts. Furthermore, reduced expression of MEF2 downstream target genes MLC1F and calcineurin were found in Mstn(-/-) muscle. Conversely, Mstn addition was sufficient to directly upregulate MLC promoter-enhancer activity in cultured myoblasts. Since high MyoD levels are seen in fast fibers, we analyzed MyoD levels in the muscle. In contrast to MEF2C, MyoD levels were increased in Mstn(-/-) muscle. Together, these results suggest that while Mstn positively regulates MEF2C levels, it negatively regulates MyoD expression in muscle. We propose that Mstn could regulate fiber-type composition by regulating the expression of MEF2C and MyoD during myogenesis.


Journal of Cellular Physiology | 2006

Prolonged absence of myostatin reduces sarcopenia

Victoria Siriett; Leanne Platt; Mônica Senna Salerno; Nicholas Ling; Ravi Kambadur; Mridula Sharma

Sarcopenia is a progressive age‐related loss of skeletal muscle mass and strength. Parabiotic experiments show that circulating factors positively influence the proliferation and regenerative capacity of satellite cells in aged mice. In addition, we believe that negative regulators of muscle mass also serve to balance the signals that influence satellite cell activation and regeneration capacity with ageing. Myostatin, a negative regulator of pre‐ and postnatal myogenesis, inhibits satellite cell activation and muscle regeneration postnatally. To investigate the role of myostatin during age‐related sarcopenia, we examined muscle mass and regeneration in young and old myostatin‐null mice. Young myostatin‐null muscle fibers were characterized by massive hypertrophy and hyperplasia and an increase in type IIB fibers, resulting in a more glycolytic muscle. With ageing, wild‐type muscle became increasingly oxidative and fiber atrophy was prominent. In contrast no fiber type switching was observed and atrophy was minimal in aged myostatin‐null muscle. The effect of ageing on satellite cell numbers appeared minimal, however, satellite cell activation declined significantly in both wild‐type and myostatin‐null muscles. In young mice, lack of myostatin resulted in increased satellite cell number and activation compared to wild‐type, suggesting a greater propensity to undergo myogenesis, a difference maintained in the aged mice. In addition, muscle regeneration of myostatin‐null muscle following notexin injury was accelerated and fiber hypertrophy and type were recovered with regeneration, unlike in wild‐type muscle. In conclusion, a lack of myostatin appears to reduce age‐related sarcopenia and loss of muscle regenerative capacity. J. Cell. Physiol. 209: 866–873, 2006.


Medicine and Science in Sports and Exercise | 2004

Resistance training alters plasma myostatin but not IGF-1 in healthy men.

Kylie S. Walker; Ravi Kambadur; Mridula Sharma; Heather K. Smith

PURPOSE We determined and compared the magnitude of changes in resting plasma myostatin and IGF-1, muscle strength, and size in response to whole body or local muscle resistance training in healthy men. METHODS Volunteers performed high-intensity resistance exercise of major muscle groups of the whole body (N = 11), or of the elbow flexors only (N = 6), twice per week for 10 wk. Strength was assessed by elbow flexor one-repetition maximum (1-RM) and repetitions at 80% of 1-RM, muscle cross-sectional area by MRI, and plasma IGF-1 by RIA and myostatin by Western analyses, before and after the training program. RESULTS In subjects of both groups, elbow flexor 1-RM and cross-sectional area increased (P = 0.05) by 30 +/- 8% (mean +/- SD) and 12 +/- 4%, respectively. Individual changes in myostatin ranged from 5.9 to -56.9%, with a mean decrease of 20 +/- 16%, whereas IGF-1 did not change from pre- to posttraining. There were no significant differences in any of the responses of the subjects between the two training programs. CONCLUSION Myostatin may play a role in exercise-induced increases in muscle size, its circulating levels decreasing with resistance training in healthy men. Exercise of the whole body versus the elbow flexors alone did not provide a supplementary stimulus in altering resting plasma IGF-1 or myostatin, or in increasing muscle strength or size. Thus, by default, growth factor responses local to the muscle may be more important than circulating factors in contributing to muscle hypertrophy with resistance training.


Journal of Cellular Physiology | 2002

Titin-cap associates with, and regulates secretion of, Myostatin

Gina Nicholas; Mark Thomas; Brett Langley; Wayne G. Somers; Ketan Patel; C. Fred Kemp; Mridula Sharma; Ravi Kambadur

Myostatin, a secreted growth factor, is a key negative regulator of skeletal muscle growth. To identify modifiers of Myostatin function, we screened for Myostatin interacting proteins. Using a yeast two‐hybrid screen, we identified Titin‐cap (T‐cap) protein as interacting with Myostatin. T‐cap is a sarcomeric protein that binds to the N‐terminal domain of Titin and is a substrate of the titin kinase. Mammalian two‐hybrid studies, in vitro binding assays and protein truncations in the yeast two‐hybrid system verified the specific interaction between processed mature Myostatin and full‐length T‐cap. Analysis of protein–protein interaction using surface plasmon resonance (Biacore, Uppsala, Sweden) kinetics revealed a high affinity between Myostatin and T‐cap with a KD of 40 nM. When T‐cap was stably overexpressed in C2C12 myoblasts, the rate of cell proliferation was significantly increased. Western analyses showed that production and processing of Myostatin were not altered in cells overexpressing T‐cap, but an increase in the retention of mature Myostatin indicated that T‐cap may block Myostatin secretion. Bioassay for Myostatin confirmed that conditioned media from myoblasts overexpressing T‐cap contained lower levels of Myostatin. Given that Myostatin negatively regulates myoblast proliferation, the increase in proliferation observed in myoblasts overexpressing T‐cap could thus be due to reduced Myostatin secretion. These results suggest that T‐cap, by interacting with Myostatin, controls Myostatin secretion in myogenic precursor cells without affecting the processing step of precursor Myostatin. J. Cell. Physiol. 193: 120–131, 2002.

Collaboration


Dive into the Ravi Kambadur's collaboration.

Top Co-Authors

Avatar

Mridula Sharma

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sudarsanareddy Lokireddy

Nanyang Technological University

View shared research outputs
Top Co-Authors

Avatar

Mridula Sharma

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sabeera Bonala

Nanyang Technological University

View shared research outputs
Top Co-Authors

Avatar

Xiaojia Ge

Nanyang Technological University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge