Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Nat Med ; 28(12): 2633-2645, 2022 12.
Article in English | MEDLINE | ID: mdl-36456835

ABSTRACT

Osteoarthritis (OA) is a common, debilitating, chronic disease with no disease-modifying drug approved to date. We discovered LNA043-a derivative of angiopoietin-like 3 (ANGPTL3)-as a potent chondrogenesis inducer using a phenotypic screen with human mesenchymal stem cells. We show that LNA043 promotes chondrogenesis and cartilage matrix synthesis in vitro and regenerates hyaline articular cartilage in preclinical OA and cartilage injury models in vivo. LNA043 exerts at least part of these effects through binding to the fibronectin receptor, integrin α5ß1 on mesenchymal stem cells and chondrocytes. In a first-in-human (phase 1), randomized, double-blinded, placebo-controlled, single ascending dose, single-center trial ( NCT02491281 ; sponsored by Novartis Pharmaceuticals), 28 patients with knee OA were injected intra-articularly with LNA043 or placebo (3:1 ratio) either 2 h, 7 d or 21 d before total knee replacement. LNA043 met its primary safety endpoint and showed short serum pharmacokinetics, cartilage penetration and a lack of immunogenicity (secondary endpoints). Post-hoc transcriptomics profiling of cartilage revealed that a single LNA043 injection reverses the OA transcriptome signature over at least 21 d, inducing the expression of hyaline cartilage matrix components and anabolic signaling pathways, while suppressing mediators of OA progression. LNA043 is a novel disease-modifying OA drug candidate that is currently in a phase 2b trial ( NCT04864392 ) in patients with knee OA.


Subject(s)
Cartilage, Articular , Osteoarthritis, Knee , Humans , Osteoarthritis, Knee/drug therapy , Chondrocytes , Signal Transduction , Angiopoietins/metabolism , Angiopoietins/pharmacology , Angiopoietins/therapeutic use , Angiopoietin-Like Protein 3
2.
iScience ; 24(12): 103434, 2021 Dec 17.
Article in English | MEDLINE | ID: mdl-34877494

ABSTRACT

Inflammatory responses are crucial for regeneration following peripheral nerve injury (PNI). PNI triggers inflammatory responses at the site of injury. The DNA-sensing receptor cyclic GMP-AMP synthase (cGAS) and its downstream effector stimulator of interferon genes (STING) sense foreign and self-DNA and trigger type I interferon (IFN) immune responses. We demonstrate here that following PNI, the cGAS/STING pathway is upregulated in the sciatic nerve of naive rats and dysregulated in old rats. In a nerve crush mouse model where STING is knocked out, myelin content in sciatic nerve is increased resulting in accelerated functional axon recovery. STING KO mice have lower macrophage number in sciatic nerve and decreased microglia activation in spinal cord 1 week post injury. STING activation regulated processing of colony stimulating factor 1 receptor (CSF1R) and microglia survival in vitro. Taking together, these data highlight a previously unrecognized role of STING in the regulation of nerve regeneration.

4.
Cell Rep ; 29(6): 1539-1554.e7, 2019 Nov 05.
Article in English | MEDLINE | ID: mdl-31693894

ABSTRACT

Age-related loss of skeletal muscle innervation by motor neurons leads to impaired neuromuscular function and is a well-established clinical phenomenon. However, the underlying pathogenesis remains unclear. Studying mice, we find that the number of motor units (MUs) can be maintained by counteracting neurotoxic microglia in the aged spinal cord. We observe that marked innervation changes, detected by motor unit number estimation (MUNE), occur prior to loss of muscle function in aged mice. This coincides with gene expression changes indicative of neuronal remodeling and microglial activation in aged spinal cord. Voluntary exercise prevents loss of MUs and reverses microglia activation. Depleting microglia by CSF1R inhibition also prevents the age-related decline in MUNE and neuromuscular junction disruption, implying a causal link. Our results suggest that age-related changes in spinal cord microglia contribute to neuromuscular decline in aged mice and demonstrate that removal of aged neurotoxic microglia can prevent or reverse MU loss.


Subject(s)
Aging/metabolism , Microglia/metabolism , Motor Neurons/metabolism , Physical Conditioning, Animal/physiology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Aging/pathology , Animals , Cell Line , Databases, Genetic , Humans , Induced Pluripotent Stem Cells , Macrophages , Male , Mice , Mice, Inbred C57BL , Microglia/enzymology , Microglia/physiology , Motor Neurons/cytology , Motor Neurons/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Neuromuscular Junction/metabolism , Neuronal Plasticity/genetics , RNA-Seq , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Spinal Cord/enzymology , Spinal Cord/metabolism , Spinal Cord/physiopathology
5.
JMIR Mhealth Uhealth ; 7(11): e15191, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31774406

ABSTRACT

BACKGROUND: Digital technologies and advanced analytics have drastically improved our ability to capture and interpret health-relevant data from patients. However, only limited data and results have been published that demonstrate accuracy in target indications, real-world feasibility, or the validity and value of these novel approaches. OBJECTIVE: This study aimed to establish accuracy, feasibility, and validity of continuous digital monitoring of walking speed in frail, elderly patients with sarcopenia and to create an open source repository of raw, derived, and reference data as a resource for the community. METHODS: Data described here were collected as a part of 2 clinical studies: an independent, noninterventional validation study and a phase 2b interventional clinical trial in older adults with sarcopenia. In both studies, participants were monitored by using a waist-worn inertial sensor. The cross-sectional, independent validation study collected data at a single site from 26 naturally slow-walking elderly subjects during a parcours course through the clinic, designed to simulate a real-world environment. In the phase 2b interventional clinical trial, 217 patients with sarcopenia were recruited across 32 sites globally, where patients were monitored over 25 weeks, both during and between visits. RESULTS: We have demonstrated that our approach can capture in-clinic gait speed in frail slow-walking adults with a residual standard error of 0.08 m per second in the independent validation study and 0.08, 0.09, and 0.07 m per second for the 4 m walk test (4mWT), 6-min walk test (6MWT), and 400 m walk test (400mWT) standard gait speed assessments, respectively, in the interventional clinical trial. We demonstrated the feasibility of our approach by capturing 9668 patient-days of real-world data from 192 patients and 32 sites, as part of the interventional clinical trial. We derived inferred contextual information describing the length of a given walking bout and uncovered positive associations between the short 4mWT gait speed assessment and gait speed in bouts between 5 and 20 steps (correlation of 0.23) and longer 6MWT and 400mWT assessments with bouts of 80 to 640 steps (correlations of 0.48 and 0.59, respectively). CONCLUSIONS: This study showed, for the first time, accurate capture of real-world gait speed in slow-walking older adults with sarcopenia. We demonstrated the feasibility of long-term digital monitoring of mobility in geriatric populations, establishing that sufficient data can be collected to allow robust monitoring of gait behaviors outside the clinic, even in the absence of feedback or incentives. Using inferred context, we demonstrated the ecological validity of in-clinic gait assessments, describing positive associations between in-clinic performance and real-world walking behavior. We make all data available as an open source resource for the community, providing a basis for further study of the relationship between standardized physical performance assessment and real-world behavior and independence.


Subject(s)
Frailty/complications , Monitoring, Physiologic/instrumentation , Walking Speed/physiology , Aged , Aged, 80 and over , Cross-Sectional Studies , Female , Fitness Trackers/statistics & numerical data , Frailty/physiopathology , Humans , Longitudinal Studies , Male , Middle Aged , Monitoring, Physiologic/methods , Monitoring, Physiologic/statistics & numerical data , Validation Studies as Topic
6.
Sci Rep ; 9(1): 9397, 2019 06 28.
Article in English | MEDLINE | ID: mdl-31253821

ABSTRACT

Microgravity exposure is associated with loss of muscle mass and strength. The E3 ubiquitin ligase MuRF1 plays an integral role in degrading the contractile apparatus of skeletal muscle; MuRF1 null (KO) mice have shown protection in ground-based models of muscle atrophy. In contrast, MuRF1 KO mice subjected to 21 days of microgravity on the International Space Station (ISS) were not protected from muscle atrophy. In a time course experiment microgravity-induced muscle loss on the ISS showed MuRF1 gene expression was not upregulated. A comparison of the soleus transcriptome profiles between spaceflight and a publicly available data set for hindlimb suspension, a claimed surrogate model of microgravity, showed only marginal commonalities between the models. These findings demonstrate spaceflight induced atrophy is unique, and that understanding of effects of space requires study situated beyond the Earth's mesosphere.


Subject(s)
Hypogravity , Muscle Proteins/deficiency , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/etiology , Muscular Atrophy/metabolism , Space Flight , Tripartite Motif Proteins/deficiency , Ubiquitin-Protein Ligases/deficiency , Animals , Disease Susceptibility , Gene Expression Profiling , Gene Expression Regulation , Hindlimb Suspension , Mice , Mice, Knockout , Muscular Atrophy/pathology , Organ Size
7.
Dev Cell ; 34(2): 181-91, 2015 Jul 27.
Article in English | MEDLINE | ID: mdl-26143994

ABSTRACT

Increasing evidence suggests that long non-coding RNAs (LncRNAs) represent a new class of regulators of stem cells. However, the roles of LncRNAs in stem cell maintenance and myogenesis remain largely unexamined. For this study, hundreds of intergenic LncRNAs were identified that are expressed in myoblasts and regulated during differentiation. One of these LncRNAs, termed LncMyoD, is encoded next to the Myod gene and is directly activated by MyoD during myoblast differentiation. Knockdown of LncMyoD strongly inhibits terminal muscle differentiation, largely due to a failure to exit the cell cycle. LncMyoD directly binds to IGF2-mRNA-binding protein 2 (IMP2) and negatively regulates IMP2-mediated translation of proliferation genes such as N-Ras and c-Myc. While the RNA sequence of LncMyoD is not well conserved between human and mouse, its locus, gene structure, and function are preserved. The MyoD-LncMyoD-IMP2 pathway elucidates a mechanism as to how MyoD blocks proliferation to create a permissive state for differentiation.


Subject(s)
Muscle Development/genetics , MyoD Protein/genetics , Myoblasts/cytology , RNA, Long Noncoding/genetics , RNA-Binding Proteins/genetics , Animals , Cell Differentiation/genetics , Cell Line , Cell Proliferation/genetics , HMGA2 Protein/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Monomeric GTP-Binding Proteins/biosynthesis , Muscle, Skeletal/embryology , Muscle, Skeletal/growth & development , Proto-Oncogene Proteins c-myc/biosynthesis , RNA Interference , RNA, Small Interfering
8.
Cell Metab ; 22(1): 164-74, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26001423

ABSTRACT

Age-related frailty may be due to decreased skeletal muscle regeneration. The role of TGF-ß molecules myostatin and GDF11 in regeneration is unclear. Recent studies showed an age-related decrease in GDF11 and that GDF11 treatment improves muscle regeneration, which were contrary to prior studies. We now show that these recent claims are not reproducible and the reagents previously used to detect GDF11 are not GDF11 specific. We develop a GDF11-specific immunoassay and show a trend toward increased GDF11 levels in sera of aged rats and humans. GDF11 mRNA increases in rat muscle with age. Mechanistically, GDF11 and myostatin both induce SMAD2/3 phosphorylation, inhibit myoblast differentiation, and regulate identical downstream signaling. GDF11 significantly inhibited muscle regeneration and decreased satellite cell expansion in mice. Given early data in humans showing a trend for an age-related increase, GDF11 could be a target for pharmacologic blockade to treat age-related sarcopenia.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Growth Differentiation Factors/metabolism , Muscle, Skeletal/physiology , Regeneration , Aging , Animals , Bone Morphogenetic Proteins/blood , Bone Morphogenetic Proteins/genetics , Cell Differentiation , Cell Line , Growth Differentiation Factors/blood , Growth Differentiation Factors/genetics , Humans , Mice , Myoblasts/cytology , Myoblasts/metabolism , Myostatin/metabolism , Rats , Signal Transduction , Up-Regulation
9.
PLoS One ; 9(7): e102359, 2014.
Article in English | MEDLINE | ID: mdl-25020062

ABSTRACT

Mesenchymal stromal/stem cell (MSC) expansion in conventional monolayer culture on plastic dishes (2D) leads to progressive loss of functionality and thus challenges fundamental studies on the physiology of skeletal progenitors, as well as translational applications for cellular therapy and molecular medicine. Here we demonstrate that 2D MSC expansion can be entirely bypassed by culturing freshly isolated bone marrow nucleated cells within 3D porous scaffolds in a perfusion-based bioreactor system. The 3D-perfusion system generated a stromal tissue that could be enzymatically treated to yield CD45- MSC. As compared to 2D-expanded MSC (control), those derived from 3D-perfusion culture after the same time (3 weeks) or a similar extent of proliferation (7-8 doublings) better maintained their progenitor properties, as assessed by a 4.3-fold higher clonogenicity and the superior differentiation capacity towards all typical mesenchymal lineages. Transcriptomic analysis of MSC from 5 donors validated the robustness of the process and indicated a reduced inter-donor variability and a significant upregulation of multipotency-related gene clusters following 3D-perfusion--as compared to 2D-expansion. Interestingly, the differences in functionality and transcriptomics between MSC expanded in 2D or under 3D-perfusion were only partially captured by cytofluorimetric analysis using conventional surface markers. The described system offers a multidisciplinary approach to study how factors of a 3D engineered niche regulate MSC function and, by streamlining conventional labor-intensive processes, is prone to automation and scalability within closed bioreactor systems.


Subject(s)
Cell Culture Techniques/methods , Mesenchymal Stem Cells/cytology , Perfusion/methods , Bioreactors , Cell Culture Techniques/instrumentation , Cell Proliferation , Cell Separation , Humans , Oligonucleotide Array Sequence Analysis , Perfusion/instrumentation , Phenotype
10.
Mol Cell Biol ; 34(4): 606-18, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24298022

ABSTRACT

The myostatin/activin type II receptor (ActRII) pathway has been identified to be critical in regulating skeletal muscle size. Several other ligands, including GDF11 and the activins, signal through this pathway, suggesting that the ActRII receptors are major regulatory nodes in the regulation of muscle mass. We have developed a novel, human anti-ActRII antibody (bimagrumab, or BYM338) to prevent binding of ligands to the receptors and thus inhibit downstream signaling. BYM338 enhances differentiation of primary human skeletal myoblasts and counteracts the inhibition of differentiation induced by myostatin or activin A. BYM338 prevents myostatin- or activin A-induced atrophy through inhibition of Smad2/3 phosphorylation, thus sparing the myosin heavy chain from degradation. BYM338 dramatically increases skeletal muscle mass in mice, beyond sole inhibition of myostatin, detected by comparing the antibody with a myostatin inhibitor. A mouse version of the antibody induces enhanced muscle hypertrophy in myostatin mutant mice, further confirming a beneficial effect on muscle growth beyond myostatin inhibition alone through blockade of ActRII ligands. BYM338 protects muscles from glucocorticoid-induced atrophy and weakness via prevention of muscle and tetanic force losses. These data highlight the compelling therapeutic potential of BYM338 for the treatment of skeletal muscle atrophy and weakness in multiple settings.


Subject(s)
Activin Receptors, Type II/immunology , Activins/metabolism , Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , Hypertrophy/metabolism , Myoblasts, Skeletal/metabolism , Activin Receptors, Type II/metabolism , Animals , Antibodies, Blocking/metabolism , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Humanized , Atrophy/immunology , Atrophy/metabolism , Cell Differentiation/physiology , Humans , Hypertrophy/pathology , Mice , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myoblasts, Skeletal/immunology , Signal Transduction/physiology , Smad2 Protein/metabolism , Smad3 Protein/metabolism
11.
Microbiol Res ; 169(2-3): 107-20, 2014.
Article in English | MEDLINE | ID: mdl-24360837

ABSTRACT

Due to evolutionary conservation of biology, experimental knowledge captured from genetic studies in eukaryotic model organisms provides insight into human cellular pathways and ultimately physiology. Yeast chemogenomic profiling is a powerful approach for annotating cellular responses to small molecules. Using an optimized platform, we provide the relative sensitivities of the heterozygous and homozygous deletion collections for nearly 1800 biologically active compounds. The data quality enables unique insights into pathways that are sensitive and resistant to a given perturbation, as demonstrated with both known and novel compounds. We present examples of novel compounds that inhibit the therapeutically relevant fatty acid synthase and desaturase (Fas1p and Ole1p), and demonstrate how the individual profiles facilitate hypothesis-driven experiments to delineate compound mechanism of action. Importantly, the scale and diversity of tested compounds yields a dataset where the number of modulated pathways approaches saturation. This resource can be used to map novel biological connections, and also identify functions for unannotated genes. We validated hypotheses generated by global two-way hierarchical clustering of profiles for (i) novel compounds with a similar mechanism of action acting upon microtubules or vacuolar ATPases, and (ii) an un-annotated ORF, YIL060w, that plays a role in respiration in the mitochondria. Finally, we identify and characterize background mutations in the widely used yeast deletion collection which should improve the interpretation of past and future screens throughout the community. This comprehensive resource of cellular responses enables the expansion of our understanding of eukaryotic pathway biology.


Subject(s)
Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/genetics , Antifungal Agents/pharmacology , Biosynthetic Pathways , Drug Resistance, Fungal , Gene Expression Regulation, Fungal , High-Throughput Screening Assays , Molecular Sequence Data , Phylogeny , Saccharomyces cerevisiae/classification , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae Proteins/metabolism
12.
PLoS One ; 8(8): e71084, 2013.
Article in English | MEDLINE | ID: mdl-23976982

ABSTRACT

The marbling of skeletal muscle by ectopic adipose tissue is a hallmark of many muscle diseases, including sarcopenia and muscular dystrophies, and generally associates with impaired muscle regeneration. Although the etiology and the molecular mechanisms of ectopic adipogenesis are poorly understood, fatty regeneration can be modeled in mice using glycerol-induced muscle damage. Using comprehensive molecular and histological profiling, we compared glycerol-induced fatty regeneration to the classical cardiotoxin (CTX)-induced regeneration model previously believed to lack an adipogenic response in muscle. Surprisingly, ectopic adipogenesis was detected in both models, but was stronger and more persistent in response to glycerol. Importantly, extensive differential transcriptomic profiling demonstrated that glycerol induces a stronger inflammatory response and promotes adipogenic regulatory networks while reducing fatty acid ß-oxidation. Altogether, these results provide a comprehensive mapping of gene expression changes during the time course of two muscle regeneration models, and strongly suggest that adipogenic commitment is a hallmark of muscle regeneration, which can lead to ectopic adipocyte accumulation in response to specific physio-pathological challenges.


Subject(s)
Adipocytes/metabolism , Adipogenesis/genetics , Adipose Tissue/metabolism , Muscle Proteins/genetics , Muscle Weakness/genetics , Muscle, Skeletal/metabolism , Regeneration/genetics , Adipocytes/pathology , Adipose Tissue/pathology , Animals , Cardiotoxins , Disease Models, Animal , Fatty Acids/metabolism , Gene Expression Profiling , Glycerol , Lipid Metabolism/genetics , Male , Mice , Mice, Inbred C57BL , Muscle Proteins/metabolism , Muscle Weakness/chemically induced , Muscle Weakness/pathology , Muscle, Skeletal/pathology
13.
Proc Natl Acad Sci U S A ; 110(26): E2371-80, 2013 Jun 25.
Article in English | MEDLINE | ID: mdl-23757500

ABSTRACT

The clinical severity of the neurodegenerative disorder spinal muscular atrophy (SMA) is dependent on the levels of functional Survival Motor Neuron (SMN) protein. Consequently, current strategies for developing treatments for SMA generally focus on augmenting SMN levels. To identify additional potential therapeutic avenues and achieve a greater understanding of SMN, we applied in vivo, in vitro, and in silico approaches to identify genetic and biochemical interactors of the Drosophila SMN homolog. We identified more than 300 candidate genes that alter an Smn-dependent phenotype in vivo. Integrating the results from our genetic screens, large-scale protein interaction studies, and bioinformatic analysis, we define a unique interactome for SMN that provides a knowledge base for a better understanding of SMA.


Subject(s)
Drosophila Proteins/genetics , Genes, Insect , RNA-Binding Proteins/genetics , Animals , Animals, Genetically Modified , Gene Regulatory Networks , Humans , Knowledge Bases , Neuromuscular Junction/genetics , Phenotype , RNA Interference , Species Specificity , Spinal Muscular Atrophies of Childhood/genetics
14.
Mol Cell Biol ; 33(2): 194-212, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23109432

ABSTRACT

Molecular mechanisms underlying sarcopenia, the age-related loss of skeletal muscle mass and function, remain unclear. To identify molecular changes that correlated best with sarcopenia and might contribute to its pathogenesis, we determined global gene expression profiles in muscles of rats aged 6, 12, 18, 21, 24, and 27 months. These rats exhibit sarcopenia beginning at 21 months. Correlation of the gene expression versus muscle mass or age changes, and functional annotation analysis identified gene signatures of sarcopenia distinct from gene signatures of aging. Specifically, mitochondrial energy metabolism (e.g., tricarboxylic acid cycle and oxidative phosphorylation) pathway genes were the most downregulated and most significantly correlated with sarcopenia. Also, perturbed were genes/pathways associated with neuromuscular junction patency (providing molecular evidence of sarcopenia-related functional denervation and neuromuscular junction remodeling), protein degradation, and inflammation. Proteomic analysis of samples at 6, 18, and 27 months confirmed the depletion of mitochondrial energy metabolism proteins and neuromuscular junction proteins. Together, these findings suggest that therapeutic approaches that simultaneously stimulate mitochondrogenesis and reduce muscle proteolysis and inflammation have potential for treating sarcopenia.


Subject(s)
Aging/genetics , Mitochondria/metabolism , Neuromuscular Junction/pathology , Proteome/analysis , Sarcopenia/pathology , Transcriptome , Aging/metabolism , Animals , DNA, Mitochondrial/genetics , Energy Metabolism , Gene Expression Profiling , Immunohistochemistry , Linear Models , Male , Microarray Analysis , Mitochondria/genetics , Mitochondria/pathology , Muscle Strength/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Neuromuscular Junction/genetics , Neuromuscular Junction/metabolism , Postmortem Changes , Proteomics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Up-Regulation
15.
Mol Cell Biol ; 32(14): 2871-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22586266

ABSTRACT

Brown adipose tissue (BAT) is a key tissue for energy expenditure via fat and glucose oxidation for thermogenesis. In this study, we demonstrate that the myostatin/activin receptor IIB (ActRIIB) pathway, which serves as an important negative regulator of muscle growth, is also a negative regulator of brown adipocyte differentiation. In parallel to the anticipated hypertrophy of skeletal muscle, the pharmacological inhibition of ActRIIB in mice, using a neutralizing antibody, increases the amount of BAT without directly affecting white adipose tissue. Mechanistically, inhibition of ActRIIB inhibits Smad3 signaling and activates the expression of myoglobin and PGC-1 coregulators in brown adipocytes. Consequently, ActRIIB blockade in brown adipose tissue enhances mitochondrial function and uncoupled respiration, translating into beneficial functional consequences, including enhanced cold tolerance and increased energy expenditure. Importantly, ActRIIB inhibition enhanced energy expenditure only at ambient temperature or in the cold and not at thermoneutrality, where nonshivering thermogenesis is minimal, strongly suggesting that brown fat activation plays a prominent role in the metabolic actions of ActRIIB inhibition.


Subject(s)
Activin Receptors, Type II/antagonists & inhibitors , Adipogenesis/physiology , Adipose Tissue, Brown/metabolism , Thermogenesis/physiology , Activin Receptors, Type II/immunology , Activin Receptors, Type II/metabolism , Adipocytes, Brown/cytology , Adipocytes, Brown/metabolism , Adipose Tissue, Brown/ultrastructure , Animals , Antibodies, Neutralizing , Cell Differentiation , Energy Metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, SCID , Mice, Transgenic , Microscopy, Electron, Transmission , Mitochondria/metabolism , Muscle, Skeletal/metabolism , Myostatin/metabolism , Signal Transduction , Smad3 Protein/metabolism , Transcription Factors/metabolism
16.
Res Microbiol ; 156(7): 822-9, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16040234

ABSTRACT

Phospholipases play an important role as virulence factors in human pathogens. Candida albicans, the major fungal pathogen of humans, encodes phospholipases of type A, B, C and D. Type B Plb2 and type D Pld1 phospholipases have been shown to contribute to virulence in this organism. We analyzed, in C. albicans, PLC2 and PLC3, two highly conserved genes coding for phosphatidylinositol-dependent phospholipases C with homology to the known virulence factor PlcA in the human pathogen Listeria monocytogenes. We show that expression of PLC2 and PLC3 is upregulated under different filament-inducing conditions and in the constitutive filamentous mutant tup1Delta. In order to analyze PLC2 and PLC3 function in C. albicans, we constructed strains that carry PLC2 or PLC3 under a constitutive promoter and strains that lack all four PLC2/3 alleles. These strains were not affected in their ability to produce filaments under non-inducing conditions, nor was filamentation modified under inducing conditions, suggesting that PLC2/3 are not critical determinants of the yeast-to-hypha switch. In a cell culture model for macrophage interaction, phagocytosis of C. albicans and subsequent killing were not influenced by PLC2/3. These results demonstrate that C. albicans PLC2 and PLC3 are dispensable for virulence; moreover, they underline the sharp contrast with the function of plcA in L. monocytogenes.


Subject(s)
Candida albicans/enzymology , Candida albicans/growth & development , Type C Phospholipases/genetics , Type C Phospholipases/metabolism , Amino Acid Sequence , Animals , Candida albicans/cytology , Candida albicans/genetics , Cell Line , Fungal Proteins/genetics , Fungal Proteins/physiology , Gene Deletion , Gene Expression Regulation, Fungal , Genes, Fungal , Hyphae/cytology , Hyphae/genetics , Hyphae/growth & development , Macrophages/microbiology , Mice , Molecular Sequence Data , Morphogenesis , Phagocytosis , Sequence Homology, Amino Acid , Type C Phospholipases/chemistry , Virulence
17.
Eukaryot Cell ; 4(6): 1102-15, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15947202

ABSTRACT

The genomes of the hemiascomycetes Saccharomyces cerevisiae and Ashbya gossypii have been completely sequenced, allowing a comparative analysis of these two genomes, which reveals that a small number of genes appear to have entered these genomes as a result of horizontal gene transfer from bacterial sources. One potential case of horizontal gene transfer in A. gossypii and 10 potential cases in S. cerevisiae were identified, of which two were investigated further. One gene, encoding the enzyme dihydroorotate dehydrogenase (DHOD), is potentially a case of horizontal gene transfer, as shown by sequencing of this gene from additional bacterial and fungal species to generate sufficient data to construct a well-supported phylogeny. The DHOD-encoding gene found in S. cerevisiae, URA1 (YKL216W), appears to have entered the Saccharomycetaceae after the divergence of the S. cerevisiae lineage from the Candida albicans lineage and possibly since the divergence from the A. gossypii lineage. This gene appears to have come from the Lactobacillales, and following its acquisition the endogenous eukaryotic DHOD gene was lost. It was also shown that the bacterially derived horizontally transferred DHOD is required for anaerobic synthesis of uracil in S. cerevisiae. The other gene discussed in detail is BDS1, an aryl- and alkyl-sulfatase gene of bacterial origin that we have shown allows utilization of sulfate from several organic sources. Among the eukaryotes, this gene is found in S. cerevisiae and Saccharomyces bayanus and appears to derive from the alpha-proteobacteria.


Subject(s)
Evolution, Molecular , Gene Transfer, Horizontal , Genes, Fungal , Saccharomyces cerevisiae/genetics , Amino Acid Sequence , Bayes Theorem , Crosses, Genetic , Dihydroorotate Dehydrogenase , Fungal Proteins/chemistry , Genetic Complementation Test , Genome, Fungal , Molecular Sequence Data , Oxidoreductases Acting on CH-CH Group Donors/genetics , Phylogeny , Saccharomyces cerevisiae/growth & development , Saccharomycetales/genetics , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Sulfatases/genetics
18.
Nucleic Acids Res ; 33(Database issue): D348-52, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15608214

ABSTRACT

The Ashbya Genome Database (AGD) is a comprehensive online source of information covering genes from the filamentous fungus Ashbya gossypii. The database content is based upon comparative genome annotation between A.gossypii and the closely related budding yeast Saccharomyces cerevisiae taking both sequence similarity and synteny (conserved order and orientation) into account. Release 2 of AGD contains 4718 protein-encoding loci located across seven chromosomes. Information can be retrieved using systematic or standard locus names from A.gossypii as well as budding and fission yeast. Approximately 90% of the genes in the genome of A.gossypii are homologous and syntenic to loci of budding yeast. Therefore, AGD is a useful tool not only for the various yeast communities in general but also for biologists who are interested in evolutionary aspects of genome research and comparative genome annotation. The database provides scientists with a convenient graphical user interface that includes various locus search and genome browsing options, data download and export functionalities and numerous reciprocal links to external databases including SGD, MIPS, GeneDB, KEGG, GermOnline and Swiss-Prot/TrEMBL. AGD is accessible at http://agd.unibas.ch/.


Subject(s)
Databases, Genetic , Genome, Fungal , Genomics , Saccharomycetales/genetics , Computer Graphics , Software , User-Computer Interface
19.
Science ; 304(5668): 304-7, 2004 Apr 09.
Article in English | MEDLINE | ID: mdl-15001715

ABSTRACT

We have sequenced and annotated the genome of the filamentous ascomycete Ashbya gossypii. With a size of only 9.2 megabases, encoding 4718 protein-coding genes, it is the smallest genome of a free-living eukaryote yet characterized. More than 90% of A. gossypii genes show both homology and a particular pattern of synteny with Saccharomyces cerevisiae. Analysis of this pattern revealed 300 inversions and translocations that have occurred since divergence of these two species. It also provided compelling evidence that the evolution of S. cerevisiae included a whole genome duplication or fusion of two related species and showed, through inferred ancient gene orders, which of the duplicated genes lost one copy and which retained both copies.


Subject(s)
Chromosome Mapping , Genome, Fungal , Saccharomyces cerevisiae/genetics , Saccharomycetales/genetics , Sequence Analysis, DNA , Base Composition , Biological Evolution , Centromere/genetics , Chromosome Inversion , Computational Biology , Fungal Proteins/genetics , Gene Duplication , Gene Order , Genes, Fungal , Molecular Sequence Data , Open Reading Frames , Sequence Alignment , Sequence Homology, Nucleic Acid , Synteny , Translocation, Genetic
20.
Genome Biol ; 4(7): R45, 2003.
Article in English | MEDLINE | ID: mdl-12844361

ABSTRACT

BACKGROUND: The recently sequenced genome of the filamentous fungus Ashbya gossypii revealed remarkable similarities to that of the budding yeast Saccharomyces cerevisiae both at the level of homology and synteny (conservation of gene order). Thus, it became possible to reinvestigate the S. cerevisiae genome in the syntenic regions leading to an improved annotation. RESULTS: We have identified 23 novel S. cerevisiae open reading frames (ORFs) as syntenic homologs of A. gossypii genes; for all but one, homologs are present in other eukaryotes including humans. Other comparisons identified 13 overlooked introns and suggested 69 potential sequence corrections resulting in ORF extensions or ORF fusions with improved homology to the syntenic A. gossypii homologs. Of the proposed corrections, 25 were tested and confirmed by resequencing. In addition, homologs of nearly 1,000 S. cerevisiae ORFs, presently annotated as hypothetical, were found in A. gossypii at syntenic positions and can therefore be considered as authentic genes. Finally, we suggest that over 400 S. cerevisiae ORFs that overlap other ORFs in S. cerevisiae and for which no homolog can be detected in A. gossypii should be regarded as spurious. CONCLUSIONS: Although, the S. cerevisiae genome is rightly considered as one of the most accurately sequenced and annotated eukaryotic genomes, we have shown that it still benefits substantially from comparison to the completed sequence and syntenic gene map of A. gossypii, an evolutionarily related fungus. This type of approach will strongly support the annotation of more complex genomes such as the human and murine genomes.


Subject(s)
Genome, Fungal , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Amino Acid Sequence , Artificial Gene Fusion , Exons , Genomics , Introns , Molecular Sequence Data , Open Reading Frames , Protein Structure, Tertiary , Saccharomyces cerevisiae Proteins/chemistry , Saccharomycetales/genetics , Sequence Homology, Amino Acid , Synteny
SELECTION OF CITATIONS
SEARCH DETAIL
...