Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Vaccine ; 42(7): 1831-1840, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-37479613

ABSTRACT

mRNA technology has emerged as a successful vaccine platform that offered a swift response to the COVID-19 pandemic. Accumulating evidence shows that vaccine efficacy, thermostability, and other important properties, are largely impacted by intrinsic properties of the mRNA molecule, such as RNA sequence and structure, both of which can be optimized. Designing mRNA sequence for vaccines presents a combinatorial problem due to an extremely large selection space. For instance, due to the degeneracy of the genetic code, there are over 10632 possible mRNA sequences that could encode the spike protein, the COVID-19 vaccines' target. Moreover, designing different elements of the mRNA sequence simultaneously against multiple objectives such as translational efficiency, reduced reactogenicity, and improved stability requires an efficient and sophisticated optimization strategy. Recently, there has been a growing interest in utilizing computational tools to redesign mRNA sequences to improve vaccine characteristics and expedite discovery timelines. In this review, we explore important biophysical features of mRNA to be considered for vaccine design and discuss how computational approaches can be applied to rapidly design mRNA sequences with desirable characteristics.


Subject(s)
COVID-19 , mRNA Vaccines , Humans , COVID-19 Vaccines , Pandemics , COVID-19/prevention & control , RNA, Messenger/genetics
2.
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
3.
RNA Biol ; 11(2): 106-10, 2014.
Article in English | MEDLINE | ID: mdl-24525859

ABSTRACT

Following reports by ENCyclopedia Of DNA Elements (ENCODE; GENCODE) Consortium and others, it is now fairly evident that the majority (70-80%) of the mammalian genome has the potential to be transcribed into non-protein-coding RNAs (ncRNAs). Critical to our understanding of genetic processes is the mechanism by which ncRNAs exert their roles. Accordingly, ncRNAs are shown to regulate the expression of protein-coding loci (i.e., genes) at the transcriptional as well as post-transcriptional stages. We recently reported on a widespread transcription at the DNA enhancer elements in myogenic cells. In our study, we found certain enhancer RNAs (eRNAs) regulate chromatin accessibility of the transcriptional machinery at loci encoding master regulators of myogenesis (i.e., MyoD/MyoG), thus suggesting their significance and site-specific impact in cellular programming. Here, we examine recent discoveries pertinent to the proposed role(s) of eRNAs in regulating gene expression. We will highlight consistencies, discuss confounding observations, and consider a lack of critical information in a way to prioritize future objectives.


Subject(s)
Chromatin/metabolism , Enhancer Elements, Genetic , Gene Regulatory Networks , RNA/metabolism , Transcription, Genetic , Genome, Human , Genomics , Humans , Models, Genetic , Muscle Development , Promoter Regions, Genetic
4.
Mol Cell ; 51(5): 606-17, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-23993744

ABSTRACT

Transcription factors and DNA regulatory binding motifs are fundamental components of the gene regulatory network. Here, by using genome-wide binding profiling, we show extensive occupancy of transcription factors of myogenesis (MyoD and Myogenin) at extragenic enhancer regions coinciding with RNA synthesis (i.e., eRNA). In particular, multiple regions were transcribed to eRNA within the regulatory region of MYOD1, including previously characterized distal regulatory regions (DRR) and core enhancer (CE). While (CE)RNA enhanced RNA polymerase II (Pol II) occupancy and transcription at MYOD1, (DRR)RNA acted to activate the downstream myogenic genes. The deployment of transcriptional machinery to appropriate loci is contingent on chromatin accessibility, a rate-limiting step preceding Pol II assembly. By nuclease sensitivity assay, we found that eRNAs regulate genomic access of the transcriptional complex to defined regulatory regions. In conclusion, our data suggest that eRNAs contribute to establishing a cell-type-specific transcriptional circuitry by directing chromatin-remodeling events.


Subject(s)
Chromatin/metabolism , Enhancer Elements, Genetic/genetics , MyoD Protein/metabolism , Myogenin/metabolism , RNA/metabolism , Animals , Binding Sites , Cell Line , Chromatin/genetics , Chromatin Assembly and Disassembly , Gene Expression Regulation , Gene Regulatory Networks , Mice , MyoD Protein/genetics , Myogenin/genetics , Promoter Regions, Genetic , RNA/biosynthesis , RNA/genetics , RNA Polymerase II/genetics , RNA Polymerase II/metabolism
5.
Nature ; 498(7455): 506-10, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23728300

ABSTRACT

Through their functional diversification, distinct lineages of CD4(+) T cells can act to either drive or constrain immune-mediated pathology. Transcription factors are critical in the generation of cellular diversity, and negative regulators antagonistic to alternate fates often act in conjunction with positive regulators to stabilize lineage commitment. Genetic polymorphisms within a single locus encoding the transcription factor BACH2 are associated with numerous autoimmune and allergic diseases including asthma, Crohn's disease, coeliac disease, vitiligo, multiple sclerosis and type 1 diabetes. Although these associations point to a shared mechanism underlying susceptibility to diverse immune-mediated diseases, a function for BACH2 in the maintenance of immune homeostasis has not been established. Here, by studying mice in which the Bach2 gene is disrupted, we define BACH2 as a broad regulator of immune activation that stabilizes immunoregulatory capacity while repressing the differentiation programs of multiple effector lineages in CD4(+) T cells. BACH2 was required for efficient formation of regulatory (Treg) cells and consequently for suppression of lethal inflammation in a manner that was Treg-cell-dependent. Assessment of the genome-wide function of BACH2, however, revealed that it represses genes associated with effector cell differentiation. Consequently, its absence during Treg polarization resulted in inappropriate diversion to effector lineages. In addition, BACH2 constrained full effector differentiation within TH1, TH2 and TH17 cell lineages. These findings identify BACH2 as a key regulator of CD4(+) T-cell differentiation that prevents inflammatory disease by controlling the balance between tolerance and immunity.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Homeostasis/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmunity/immunology , Basic-Leucine Zipper Transcription Factors/deficiency , Basic-Leucine Zipper Transcription Factors/genetics , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Homeostasis/genetics , Humans , Immune Tolerance/genetics , Immune Tolerance/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/mortality , Inflammation/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/pharmacology
6.
EMBO J ; 32(8): 1075-86, 2013 Apr 17.
Article in English | MEDLINE | ID: mdl-23503590

ABSTRACT

Histone chaperones affect chromatin structure and gene expression through interaction with histones and RNA polymerase II (PolII). Here, we report that the histone chaperone Spt6 counteracts H3K27me3, an epigenetic mark deposited by the Polycomb Repressive Complex 2 (PRC2) and associated with transcriptional repression. By regulating proper engagement and function of the H3K27 demethylase KDM6A (UTX), Spt6 effectively promotes H3K27 demethylation, muscle gene expression, and cell differentiation. ChIP-Seq experiments reveal an extensive genome-wide overlap of Spt6, PolII, and KDM6A at transcribed regions that are devoid of H3K27me3. Mammalian cells and zebrafish embryos with reduced Spt6 display increased H3K27me3 and diminished expression of the master regulator MyoD, resulting in myogenic differentiation defects. As a confirmation for an antagonistic relationship between Spt6 and H3K27me3, inhibition of PRC2 permits MyoD re-expression in myogenic cells with reduced Spt6. Our data indicate that, through cooperation with PolII and KDM6A, Spt6 orchestrates removal of H3K27me3, thus controlling developmental gene expression and cell differentiation.


Subject(s)
Histone Demethylases/metabolism , Histones/metabolism , Muscle Development , RNA Polymerase II/metabolism , Animals , Cell Differentiation , Cell Line , Chromatin Immunoprecipitation , Methylation , Mice , Transcription Factors , Zebrafish
7.
Mol Cell ; 45(2): 255-62, 2012 Jan 27.
Article in English | MEDLINE | ID: mdl-22196887

ABSTRACT

Polycomb group (PcG) proteins initiate the formation of repressed chromatin domains and regulate developmental gene expression. A mammalian PcG protein, enhancer of zeste homolog 2 (Ezh2), triggers transcriptional repression by catalyzing the addition of methyl groups onto lysine 27 of histone H3 (H3K27me2/3). This action facilitates the binding of other PcG proteins to chromatin for purposes of transcriptional silencing. Interestingly, there exists a paralog of Ezh2, termed Ezh1, whose primary function remains unclear. Here, we provide evidence for genome-wide association of Ezh1 complex with active epigenetic mark (H3K4me3), RNA polymerase II (Pol II), and mRNA production. Ezh1 depletion reduced global Pol II occupancy within gene bodies and resulted in delayed transcriptional activation during differentiation of skeletal muscle cells. Conversely, overexpression of wild-type Ezh1 led to premature gene activation and rescued Pol II occupancy defects in Ezh1-depleted cells. Collectively, these findings reveal a role for a PcG complex in promoting mRNA transcription.


Subject(s)
DNA-Binding Proteins/physiology , Histone-Lysine N-Methyltransferase/physiology , Peptide Chain Elongation, Translational/physiology , RNA Polymerase II/physiology , Transcription Factors/physiology , Animals , Cell Differentiation , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Methylation , Mice , Muscle Development , Polycomb Repressive Complex 2 , RNA Interference , RNA Polymerase II/metabolism , Repressor Proteins/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism
8.
PLoS One ; 6(12): e29057, 2011.
Article in English | MEDLINE | ID: mdl-22216166

ABSTRACT

A wave of structural reorganization involving centrosomes, microtubules, Golgi complex and ER exit sites takes place early during skeletal muscle differentiation and completely remodels the secretory pathway. The mechanism of these changes and their functional implications are still poorly understood, in large part because all changes occur seemingly simultaneously. In an effort to uncouple the reorganizations, we have used taxol, nocodazole, and the specific GSK3-ß inhibitor DW12, to disrupt the dynamic microtubule network of differentiating cultures of the mouse skeletal muscle cell line C2. Despite strong effects on microtubules, cell shape and cell fusion, none of the treatments prevented early differentiation. Redistribution of centrosomal proteins, conditional on differentiation, was in fact increased by taxol and nocodazole and normal in DW12. Redistributions of Golgi complex and ER exit sites were incomplete but remained tightly linked under all circumstances, and conditional on centrosomal reorganization. We were therefore able to uncouple microtubule reorganization from the other events and to determine that centrosomal proteins lead the reorganization hierarchy. In addition, we have gained new insight into structural and functional aspects of the reorganization of microtubule nucleation during myogenesis.


Subject(s)
Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Microtubules/physiology , Animals , Biological Transport , Cells, Cultured , Enzyme Inhibitors/pharmacology , Mice , Microtubules/drug effects , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Nocodazole/pharmacology , Paclitaxel/pharmacology
9.
Sci Signal ; 3(152): pe49, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21156935

ABSTRACT

In the field of molecular oncology, the Myc basic helix-loop-helix family of transcription factors has been extensively studied. The Myc proto-oncogene c-Myc binds DNA, activates or represses gene transcription, and consequently affects cellular proliferation. However, emerging evidence presents the existence of c-Myc variants that lack transcriptional activity. A cytoplasmic variant of c-Myc called "Myc-nick," which arises from calpain-mediated cleavage of c-Myc, assists in stable microtubule assembly. Furthermore, Myc-nick promotes MyoD-mediated myogenic differentiation, thus antagonizing its precursor. These results provide exciting new opportunities in formulating molecular approaches for treatment of cancer and in our understanding of cell differentiation.


Subject(s)
Calpain/metabolism , Proto-Oncogene Proteins c-myc/physiology , Humans , Hydrolysis , Microtubules , Muscle Development , Neoplasms , Proto-Oncogene Mas , Proto-Oncogene Proteins c-myc/genetics
10.
J Neurosci ; 27(3): 665-75, 2007 Jan 17.
Article in English | MEDLINE | ID: mdl-17234598

ABSTRACT

After axotomy, expression of acetylcholinesterase (AChE) is greatly reduced in the superior cervical ganglion (SCG); however, the molecular events involved in this response remain unknown. Here, we first examined AChE mRNA levels in the brain of transgenic mice that overexpress human HuD. Both in situ hybridization and reverse transcription-PCR demonstrated that AChE transcript levels were increased by more than twofold in the hippocampus of HuD transgenic mice. Additionally, direct interaction between the HuD transgene product and AChE mRNA was observed. Next, we examined the role of HuD in regulating AChE expression in intact and axotomized rat SCG neurons. After axotomy of the adult rat SCG neurons, AChE transcript levels decreased by 50 and 85% by the first and fourth day, respectively. In vitro mRNA decay assays indicated that the decrease in AChE mRNA levels resulted from changes in the stability of presynthesized transcripts. A combination of approaches performed using the region that directly encompasses an adenylate and uridylate (AU)-rich element within the AChE 3'-untranslated region demonstrated a decrease in RNA-protein complexes in response to axotomy of the SCG and, specifically, a decrease in HuD binding. After axotomy, HuD transcript and protein levels also decreased. Using a herpes simplex virus construct containing the human HuD sequence to infect SCG neurons in vivo, we found that AChE and GAP-43 mRNA levels were maintained in the SCG after axotomy. Together, the results of this study demonstrate that AChE expression in neurons of the rat SCG is regulated via post-transcriptional mechanisms that involve the AU-rich element and HuD.


Subject(s)
Acetylcholinesterase/metabolism , ELAV Proteins/metabolism , Gene Expression Regulation, Enzymologic/physiology , Neurons/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Acetylcholinesterase/genetics , Animals , Axotomy , ELAV Proteins/genetics , ELAV-Like Protein 4 , Female , Humans , Mice , Mice, Transgenic , Protein Binding/physiology , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Rats , Rats, Sprague-Dawley , Superior Cervical Ganglion/metabolism
11.
J Neurosci ; 26(21): 5739-49, 2006 May 24.
Article in English | MEDLINE | ID: mdl-16723531

ABSTRACT

In skeletal muscle, brain-derived neurotrophic factor (BDNF) has long been thought to serve as a retrograde trophic factor for innervating motor neurons throughout their lifespan. However, its localization in mature muscle fibers has remained elusive. Given the postulated roles of BDNF in skeletal muscle, we performed a series of complementary experiments aimed at defining the localization of BDNF and its transcripts in adult muscle. By reverse transcription-PCR, in situ hybridization, and immunofluorescence, we show that BDNF, along with the receptor p75NTR, is not expressed at significant levels within mature myofibers and that it does not accumulate preferentially within subsynaptic regions of neuromuscular junctions. Interestingly, expression of BDNF correlated with that of Pax3, a marker of muscle progenitor cells, in several different adult skeletal muscles. Additionally, BDNF was expressed in Pax7+ satellite cells where it colocalized with p75NTR. In complementary cell culture experiments, we detected high levels of BDNF and p75NTR in myoblasts. During myogenic differentiation, expression of BDNF became drastically reduced. Using small interfering RNA (siRNA) technology to knock down BDNF expression, we demonstrate enhanced myogenic differentiation of myoblasts. This accelerated rate of myogenic differentiation seen in myoblasts expressing BDNF siRNA was normalized by administration of recombinant BDNF. Collectively, these findings show that BDNF plays an important regulatory function during myogenic differentiation. In addition, the expression of BDNF in satellite cells is coherent with the notion that BDNF serves a key role in maintaining the population of muscle progenitors in adult muscle.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Receptor, Nerve Growth Factor/metabolism , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Animals , Cell Differentiation , Cells, Cultured , Female , Rats , Rats, Sprague-Dawley , Tissue Distribution
12.
J Biol Chem ; 280(2): 1132-41, 2005 Jan 14.
Article in English | MEDLINE | ID: mdl-15528206

ABSTRACT

Both the transcription factor c-Jun and the c-Jun N-terminal kinases (JNKs) have been associated with neuronal loss in several death paradigms. JNK are key regulators of c-Jun and a common accepted model has been that JNKs mediate neuronal death through modulation of c-Jun activation. In the present study, we examined whether JNK2 and -3 (JNK members most associated with neuronal loss) deficiency can rescue neuronal loss caused by facial and sciatic nerve axotomy in the neonate in vivo. JNK2, JNK3, and JNK2/3 double-deficient neurons displayed significantly less death in the facial nerves of the CNS when compared with controls. JNK2 and JNK2/3 double-deficient animals also showed reduced c-Jun phosphorylation and induction following axotomy, consistent with the model that JNK acts to regulate death by activating c-Jun. Of significance, however, protection of facial nerves in JNK3-deficient animals was not accompanied by reduction in c-Jun activation. These results suggest that JNKs can mediate death independently of c-Jun. Importantly, the lack of correlation between JNK3 deficiency and c-Jun induction was not universal. In a sciatic axotomy model of neuronal injury in the neonate, death of DRG neurons was also reduced by JNK3 deficiency. However, in this case, c-Jun activation was also eliminated.


Subject(s)
Mitogen-Activated Protein Kinase 10/deficiency , Motor Neurons/cytology , Motor Neurons/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Animals , Axotomy , Cell Death , Face/innervation , Ganglia, Spinal/cytology , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 10/genetics , Mitogen-Activated Protein Kinase 10/metabolism , Mitogen-Activated Protein Kinase 9/deficiency , Mitogen-Activated Protein Kinase 9/metabolism , Motor Neurons/enzymology , Motor Neurons/physiology , Phosphorylation , Phosphoserine/metabolism , Sciatic Nerve/physiology , Sciatic Nerve/surgery
13.
Am J Physiol Cell Physiol ; 287(1): C22-9, 2004 Jul.
Article in English | MEDLINE | ID: mdl-14973145

ABSTRACT

Neonatal sciatic nerve injury is known to result in an extensive loss of lumbar motor neurons as well as the disappearance of their respective muscle fibers in the hindlimb musculature. The loss of motor neurons and muscle fibers can be prevented by immediate administration of target-derived neurotrophic factors to the site of injury. In the present study, we investigated the role of ciliary neurotrophic factor (CNTF) and brain-derived neurotrophic factor (BDNF) in the survival and maturation of a subset of motor neurons innervating the extensor digitorum longus (EDL) and tibialis anterior (TA) muscles. We have shown that combined administration of CNTF and BDNF prevented the loss of motor units after neonatal nerve injury and contributed to the maintenance of muscle mass. Importantly, this combined neurotrophin regimen also prevented the disappearance of muscle fibers that express myosin heavy chain IIB (MyHC IIB) in both EDL and TA muscles 3 mo after neonatal sciatic nerve crush. In parallel studies, we observed a higher level of BDNF in EDL muscle during the critical period of development when motor neurons are highly susceptible to target removal. Given our previous findings that combined administration of CNTF with neurotrophin-3 (NT-3) or neurotrophin-4/5 (NT-4/5) did not result in the rescue of MyHC IIB fibers in EDL, the present results show the importance of muscle-derived BDNF in the survival and maturation of a subpopulation of motor neurons and of MyHC IIB muscle fibers during neonatal development of the neuromuscular system.


Subject(s)
Animals, Newborn , Brain-Derived Neurotrophic Factor/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Myosin Heavy Chains/metabolism , Nonmuscle Myosin Type IIB/metabolism , Sciatic Nerve/injuries , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Ciliary Neurotrophic Factor/pharmacology , Drug Combinations , Hindlimb , Muscle Fibers, Skeletal/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Nerve Crush , Rats , Wounds and Injuries/physiopathology
14.
Am J Physiol Cell Physiol ; 283(3): C950-9, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12176751

ABSTRACT

Neonatal sciatic nerve crush results in a sustained reduction of the mass of both extensor digitorum longus (EDL) and soleus (SOL) muscles in the rat. Type IIB fibers are selectively lost from EDL. We have investigated the effects of ciliary neurotrophic factor (CNTF) combined with neurotrophin (NT)-3 or NT-4 on muscle mass, as well as the number, cross-sectional area, and distribution of muscle fiber types and the number of motor neurons innervating EDL and SOL 3 mo after transient axotomy 5 days after birth. Both NT treatments prevented the axotomy-induced loss of muscle mass in both EDL and SOL and of total number of muscle fibers in EDL but not in SOL. Although IIB fiber loss was not prevented, both NT treatments resulted in altered fiber type distribution. Both NT combinations also reduced the loss of EDL motor neurons. These data suggest that a differential distribution of NT receptors on either motor neurons or muscle fibers may lead to different levels of susceptibility to neonatal axotomy.


Subject(s)
Muscle Fibers, Skeletal/drug effects , Muscle, Skeletal/drug effects , Nerve Growth Factors/pharmacology , Sciatic Neuropathy/drug therapy , Animals , Animals, Newborn , Axotomy , Cell Survival/drug effects , Ciliary Neurotrophic Factor/pharmacology , Disease Models, Animal , Drug Therapy, Combination , Immunohistochemistry , Motor Neurons/drug effects , Motor Neurons/pathology , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle Fibers, Skeletal/classification , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/innervation , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Myosin Heavy Chains/biosynthesis , Nerve Crush , Neurotrophin 3/pharmacology , Rats , Rats, Sprague-Dawley , Sciatic Neuropathy/pathology , Sciatic Neuropathy/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...