Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters

Publication year range
1.
Nature ; 574(7777): 259-263, 2019 10.
Article in English | MEDLINE | ID: mdl-31554973

ABSTRACT

Chikungunya virus (CHIKV) is a re-emerging alphavirus that is transmitted to humans by mosquito bites and causes musculoskeletal and joint pain1,2. Despite intensive investigations, the human cellular factors that are critical for CHIKV infection remain unknown, hampering the understanding of viral pathogenesis and the development of anti-CHIKV therapies. Here we identified the four-and-a-half LIM domain protein 1 (FHL1)3 as a host factor that is required for CHIKV permissiveness and pathogenesis in humans and mice. Ablation of FHL1 expression results in the inhibition of infection by several CHIKV strains and o'nyong-nyong virus, but not by other alphaviruses and flaviviruses. Conversely, expression of FHL1 promotes CHIKV infection in cells that do not normally express it. FHL1 interacts directly with the hypervariable domain of the nsP3 protein of CHIKV and is essential for the replication of viral RNA. FHL1 is highly expressed in CHIKV-target cells and is particularly abundant in muscles3,4. Dermal fibroblasts and muscle cells derived from patients with Emery-Dreifuss muscular dystrophy that lack functional FHL15 are resistant to CHIKV infection. Furthermore,  CHIKV infection  is undetectable in Fhl1-knockout mice. Overall, this study shows that FHL1 is a key factor expressed by the host that enables CHIKV infection and identifies the interaction between nsP3 and FHL1 as a promising target for the development of anti-CHIKV therapies.


Subject(s)
Chikungunya Fever/virology , Chikungunya virus/pathogenicity , Host-Derived Cellular Factors/metabolism , Host-Pathogen Interactions , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/metabolism , Muscle Proteins/metabolism , Animals , Cells, Cultured , Chikungunya Fever/drug therapy , Chikungunya virus/drug effects , Chikungunya virus/genetics , Chikungunya virus/growth & development , Female , Fibroblasts/virology , HEK293 Cells , Host-Derived Cellular Factors/genetics , Humans , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , LIM Domain Proteins/deficiency , LIM Domain Proteins/genetics , Male , Mice , Muscle Proteins/deficiency , Muscle Proteins/genetics , Myoblasts/virology , O'nyong-nyong Virus/growth & development , O'nyong-nyong Virus/pathogenicity , Protein Binding , RNA, Viral/biosynthesis , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication
2.
BMC Genomics ; 21(1): 45, 2020 Jan 14.
Article in English | MEDLINE | ID: mdl-31937237

ABSTRACT

BACKGROUND: Clostridium perfringens is a Gram-positive anaerobic pathogen that causes multiple diseases in humans and animals. C. perfringens lack flagella but have type IV pili (TFP) and can glide on agar surfaces. When C. perfringens bacteria are placed on surfaces, they become elongated, flexible and have TFP on their surface, traits not seen in liquid-grown cells. In addition, the main pilin in C. perfringens TFP, PilA2, undergoes differential post-translational modification when grown in liquid or on plates. To understand the mechanisms underlying these phenotypes, bacteria were grown in three types of liquid media and on agar plates with the same medium to compare gene expression using RNA-Seq. RESULTS: Hundreds of genes were differentially expressed, including transcriptional regulatory protein-encoding genes and genes associated with TFP functions, which were higher on plates than in liquid. Transcript levels of TFP genes reflected the proportion of each protein predicted to reside in a TFP assembly complex. To measure differences in rates of translation, the Escherichia coli reporter gene gusA gene (encoding ß-glucuronidase) was inserted into the chromosome downstream of TFP promoters and in-frame with the first gene of the operon. ß-glucuronidase expression was then measured in cells grown in liquid or on plates. ß-glucuronidase activity was proportional to mRNA levels in liquid-grown cells, but not plate-grown cells, suggesting significant levels of post-transcriptional regulation of these TFP-associated genes occurs when cells are grown on surfaces. CONCLUSIONS: This study reveals insights into how a non-flagellated pathogenic rod-shaped bacterium senses and responds to growth on surfaces, including inducing transcriptional regulators and activating multiple post-transcriptional regulatory mechanisms associated with TFP functions.


Subject(s)
Clostridium perfringens/physiology , Fimbriae Proteins/genetics , Gene Expression Regulation, Bacterial , Animals , Bacterial Adhesion , Bacterial Toxins/genetics , Base Sequence , Gene Expression Profiling , Gene Order , High-Throughput Nucleotide Sequencing , Humans , Mice , Myoblasts/virology , Operon , Promoter Regions, Genetic , Temperature , Transcriptome
3.
J Fish Dis ; 41(9): 1331-1338, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30003544

ABSTRACT

A continuous cell line consisting mostly of epithelioid cells was established from the caudal fin of marbled eels (Anguilla marmorata) and designated as marbled eel caudal fin (MECF)-1. The cells multiplied well in Leibovitz's L-15 medium containing 2% to 15% foetal bovine serum at temperatures of 20°C to 35°C and were subcultured for >90 passages during a 5-year period from 2012 to 2017. Transcripts of ictacalcin, keratin 13, cd146, nestin, ncam1 and myod1 were demonstrated in the cells using reverse transcription polymerase chain reaction. The results indicated that MECF-1 was composed of epidermal and mesenchyme stem and progenitor cells including myoblasts. MECF-1 was susceptible to Japanese eel herpesvirus HVA980811, marbled eel polyoma-like virus (MEPyV), aquabirnavirus MEIPNV1310 and aquareovirus CSV. By contrast, MECF-1 was noted refractory to megalocytiviruses RSIV-Ku and GSIV-K1 infection. Moreover, the cells were resistant to betanodavirus infection. In conclusion, MECF-1 derived from marbled eel is suitable for studies on anguillid viruses and interaction with host cells.


Subject(s)
Anguilla/anatomy & histology , Anguilla/virology , Animal Fins/cytology , Animal Fins/virology , Cell Line/virology , Tissue Culture Techniques , Animals , Cell Culture Techniques/veterinary , Cell Line/cytology , Culture Media/chemistry , Disease Susceptibility , Epidermal Cells , Epidermis/virology , Fish Diseases/virology , Herpesviridae/physiology , Myoblasts/virology , Polyomavirus/physiology , Reoviridae/physiology
4.
J Gen Virol ; 98(12): 3008-3025, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29120300

ABSTRACT

Enterovirus 71 (EV71) has caused major outbreaks of hand, foot and mouth disease. EV71 infections increase the production of many host cytokines and pro-inflammatory factors, including interleukin (IL)-6, IL-10 and COX-2. Some of these molecules could stimulate the signal transducer and activator of transcription 3 (STAT3), which plays a key role in regulating host immune responses and several viral diseases. However, the role of STAT3 in EV71 infection remains unknown. This study found that the phosphorylation levels of STAT3 (pY705-STAT3) are closely related to EV71 infection. Further experiments revealed that STAT3 exerts an anti-EV71 activity. However, the antiviral activity of STAT3 is partially antagonized by EV71-induced miR-124, which directly targets STAT3 mRNA. Similarly, IL-6R, the α-subunit of the IL-6 receptor complex, exhibits anti-EV71 activity and is directly targeted by the virus-induced miR-124. These results indicate that EV71 can evade host IL-6R- and STAT3-mediated antiviral activities by EV71-induced miR-124. This suggests that controlling miR-124 and the downstream targets, IL-6R and STAT3, might benefit the antiviral treatment of EV71 infection.


Subject(s)
Enterovirus A, Human/genetics , Immune Evasion , MicroRNAs/genetics , RNA, Messenger/genetics , Receptors, Interleukin-6/genetics , STAT3 Transcription Factor/genetics , Base Sequence , Binding Sites , Cell Line, Tumor , Cyclooxygenase 2/genetics , Cyclooxygenase 2/immunology , Enterovirus A, Human/growth & development , Enterovirus A, Human/immunology , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-6/genetics , Interleukin-6/immunology , MicroRNAs/immunology , Myoblasts/immunology , Myoblasts/virology , Neurons/immunology , Neurons/virology , Phosphorylation , RNA, Messenger/immunology , Receptors, Interleukin-6/immunology , STAT3 Transcription Factor/immunology , Signal Transduction , Virus Replication
5.
Am J Physiol Regul Integr Comp Physiol ; 313(3): R240-R250, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28637658

ABSTRACT

Skeletal muscle stem cells play a critical role in regeneration of myofibers. We previously demonstrated that chronic binge alcohol (CBA) markedly attenuates myoblast differentiation potential and myogenic gene expression. Muscle-specific microRNAs (miRs) are implicated in regulation of myogenic genes. The aim of this study was to determine whether myoblasts isolated from asymptomatic CBA-administered simian immunodeficiency virus (SIV)-infected macaques treated with antiretroviral therapy (ART) showed similar impairments and, if so, to elucidate potential underlying mechanisms. Myoblasts were isolated from muscle at 11 mo after SIV infection from CBA/SIV macaques and from time-matched sucrose (SUC)-treated SIV-infected (SUC/SIV) animals and age-matched controls. Myoblast differentiation and myogenic gene expression were significantly decreased in myoblasts from SUC/SIV and CBA/SIV animals compared with controls. SIV and CBA decreased muscle-specific miR-206 in plasma and muscle and SIV decreased miR-206 expression in myoblasts, with no statistically significant changes in other muscle-specific miRs. These findings were associated with a significant increase in histone deacetylase 4 (HDAC4) and decrease in myogenic enhancer factor 2C (MEF2C) expression in CBA/SIV muscle. Transfection with miR-206 inhibitor decreased myotube differentiation, increased expression of HDAC4, and decreased MEF2C, suggesting a critical role of miR-206 in myogenesis. Moreover, HDAC4 was confirmed to be a direct miR-206 target. These results support a mechanistic role for decreased miR-206 in suppression of myoblast differentiation resulting from chronic alcohol and SIV infection. The parallel changes in skeletal muscle and circulating levels of miR-206 warrant studies to establish the possible use of plasma miR-206 as an indicator of impaired muscle function.


Subject(s)
Binge Drinking/physiopathology , Cell Differentiation , MicroRNAs/metabolism , Muscle Development , Myoblasts/cytology , Myoblasts/pathology , Simian Acquired Immunodeficiency Syndrome/physiopathology , Animals , Binge Drinking/pathology , Binge Drinking/virology , Down-Regulation , Macaca mulatta , Male , MicroRNAs/genetics , Myoblasts/virology , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Acquired Immunodeficiency Syndrome/virology
6.
Antimicrob Agents Chemother ; 60(9): 5357-67, 2016 09.
Article in English | MEDLINE | ID: mdl-27353263

ABSTRACT

Enterovirus 71 (EV-A71) is a major causative pathogen of hand, foot, and mouth disease (HFMD) epidemics. No antiviral therapies are currently available for treating EV-A71 infections. Here, we selected five reported enterovirus inhibitors (suramin, itraconazole [ITZ], GW5074, rupintrivir, and favipiravir) with different mechanisms of action to test their abilities to inhibit EV-A71 replication alone and in combination. All selected compounds have anti-EV-A71 activities in cell culture. The combination of rupintrivir and ITZ or favipiravir was synergistic, while the combination of rupintrivir and suramin was additive. The combination of suramin and favipiravir exerted a strong synergistic antiviral effect. The observed synergy was not due to cytotoxicity, as there was no significant increase in cytotoxicity when compounds were used in combinations at the tested doses. To investigate the potential inhibitory mechanism of favipiravir against enterovirus, two favipiravir-resistant EV-A71 variants were independently selected, and both of them carried an S121N mutation in the finger subdomain of the 3D polymerase. Reverse engineering of this 3D S121N mutation into an infectious clone of EV-A71 confirmed the resistant phenotype. Moreover, viruses resistant to ITZ or favipiravir remained susceptible to other inhibitors. Most notably, combined with ITZ, rupintrivir prevented the development of ITZ-resistant variants. Taken together, these results provide a rational basis for the design of combination regimens for use in the treatment of EV-A71 infections.


Subject(s)
Antiviral Agents/pharmacology , Enterovirus A, Human/drug effects , Isoxazoles/pharmacology , Itraconazole/pharmacology , Pyrrolidinones/pharmacology , Suramin/pharmacology , Viral Nonstructural Proteins/genetics , Amides/pharmacology , Amino Acid Sequence , Animals , Binding Sites , Cell Line, Tumor , Chlorocebus aethiops , Drug Combinations , Drug Resistance, Viral/genetics , Drug Synergism , Enterovirus A, Human/genetics , Enterovirus A, Human/growth & development , Humans , Indoles/pharmacology , Molecular Docking Simulation , Mutation , Myoblasts/drug effects , Myoblasts/virology , Phenols/pharmacology , Phenylalanine/analogs & derivatives , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Pyrazines/pharmacology , Sequence Alignment , Sequence Homology, Amino Acid , Valine/analogs & derivatives , Vero Cells , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/metabolism , Virus Replication/drug effects
7.
J Med Virol ; 87(5): 733-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25656089

ABSTRACT

In addition to fever, rash, and arthralgia/arthritis, myalgia is another dominant symptom in Chikungunya virus (CHIKV) infection. How CHIKV induces myalgia is unclear. To better understand the viral factors involved in CHIKV-induced myalgia, CHIKVs were isolated from patients with and without myalgia designated myalgia-CHIKV and mild-CHIKV, respectively. The response of myoblasts to infection by the two groups of clinical isolates of CHIKV was investigated. Both groups of CHIKV replicated well in primary human myoblasts. However, the myalgia-CHIKVs replicated to a higher titer and caused the death of infected myoblast more rapidly than the mild-CHIKVs. CHIKV-infected myoblasts increased production of four out of five inflammatory cytokines examined (MCP-1, IP-10, MIP-1α, and IL-8) in comparison to mock-infected cells. Comparison between the myoblast inflammatory cytokine responses showed that myalgia-CHIKVs were stronger activators of cytokines than mild-CHIKVs. This means that recent epidemic strains of CHIKV exhibited different degrees of myoblast permissiveness as evidenced by differences in the ability to replicate and to stimulate inflammatory responses in myoblasts. This data suggest that the myopathic syndrome in recent epidemics is dependent upon the strain of CHIKV.


Subject(s)
Chikungunya Fever/virology , Chikungunya virus/physiology , Myalgia/virology , Myoblasts/immunology , Myoblasts/virology , Virus Replication , Adult , Cells, Cultured , Chikungunya Fever/epidemiology , Chikungunya Fever/pathology , Chikungunya virus/growth & development , Chikungunya virus/isolation & purification , Cytokines/metabolism , Humans , Viral Load
8.
J Virol ; 87(22): 12327-38, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24027304

ABSTRACT

Rabies virus (RABV), which is transmitted via a bite wound caused by a rabid animal, infects peripheral nerves and then spreads to the central nervous system (CNS) before causing severe neurological symptoms and death in the infected individual. Despite the importance of this ability of the virus to spread from a peripheral site to the CNS (neuroinvasiveness) in the pathogenesis of rabies, little is known about the mechanism underlying the neuroinvasiveness of RABV. In this study, to obtain insights into the mechanism, we conducted comparative analysis of two fixed RABV strains, Nishigahara and the derivative strain Ni-CE, which cause lethal and asymptomatic infections, respectively, in mice after intramuscular inoculation. Examination of a series of chimeric viruses harboring the respective genes from Nishigahara in the genetic background of Ni-CE revealed that the Nishigahara phosphoprotein (P) gene plays a major role in the neuroinvasiveness by mediating infection of peripheral nerves. The results obtained from both in vivo and in vitro experiments strongly suggested that the Nishigahara P gene, but not the Ni-CE P gene, is important for stable viral replication in muscle cells. Further investigation based on the previous finding that RABV phosphoprotein counteracts the host interferon (IFN) system demonstrated that the Nishigahara P gene, but not the Ni-CE P gene, functions to suppress expression of the beta interferon (IFN-ß) gene (Ifn-ß) and IFN-stimulated genes in muscle cells. In conclusion, we provide the first data strongly suggesting that RABV phosphoprotein assists viral replication in muscle cells by counteracting the host IFN system and, consequently, enhances infection of peripheral nerves.


Subject(s)
Muscle Cells/virology , Myoblasts/virology , Peripheral Nerves/virology , Phosphoproteins/metabolism , Rabies virus/pathogenicity , Rabies/virology , Viral Structural Proteins/metabolism , 2',5'-Oligoadenylate Synthetase/genetics , 2',5'-Oligoadenylate Synthetase/metabolism , Animals , Blotting, Western , Cells, Cultured , Female , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Interferons/pharmacology , Mice , Molecular Chaperones , Muscle Cells/metabolism , Muscle Cells/pathology , Myoblasts/metabolism , Myoblasts/pathology , Myxovirus Resistance Proteins/genetics , Myxovirus Resistance Proteins/metabolism , Neuroblastoma/genetics , Neuroblastoma/pathology , Neuroblastoma/virology , Peripheral Nerves/metabolism , Peripheral Nerves/pathology , Phosphoproteins/genetics , RNA, Messenger/genetics , Rabies/genetics , Rabies/pathology , Rabies virus/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/pathology , Rhabdomyosarcoma/virology , Viral Structural Proteins/genetics , Virulence , Virus Replication
9.
Avian Pathol ; 43(5): 414-9, 2014.
Article in English | MEDLINE | ID: mdl-25204414

ABSTRACT

Marek's disease virus (MDV; also known as Gallid herpesvirus 2, MDV-1) causes oncogenic disease in chickens producing clinical signs that include lymphomas, visceral tumours, nerve lesions, and immunosuppression. MDV vaccines are widely used and mostly produced using primary cells: chicken embryo fibroblast cells, duck embryo fibroblast cells, chicken embryo kidney cells or chicken kidney cells. An immortalized cell line that can be used to manufacture the virus has long been desired. In this report, we demonstrate that QM7 cells were susceptible to infection with either MDV or herpesvirus of turkey (HVT; also known as Meleagrid herpesvirus 1, MDV-3). Polymerase chain reaction analysis with primers amplifying selected MDV genes revealed that QM7 cells did not contain these sequences. However, MDV genes were detected in QT35 cells, which have been reported to harbour latent MDV virus. Transfection of naked MDV DNA initiated efficient infection of QM7 cells. In addition, QM7 cell lysate, clarified supernatant, and QM7 cell pellet infected with MDV were negative for reverse transcriptase activity, indicating absence of endogenous retrovirus. QM7 cells were also found to be free of other avian pathogens in a chick embryo inoculation test. In vivo studies of MDV growing in QM7 cells showed the virus retained its pathogenicity and virulence. In ovo experiments demonstrated that both HVT and MDV propagated in QM7 cells did not interfere with hatchability of injected eggs, and viruses could be re-isolated from hatched chicks. The results suggest that QM7 could be a good host cell line for growing both MDV and HVT.


Subject(s)
Herpesvirus 1, Meleagrid/physiology , Mardivirus/physiology , Myoblasts/virology , Virus Cultivation , Animals , Cell Line , Chickens , Genome, Viral , Mardivirus/pathogenicity , Marek Disease/virology , Quail , Specific Pathogen-Free Organisms , Virus Latency
10.
J Infect Dis ; 206(3): 407-14, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22615321

ABSTRACT

BACKGROUND: Sindbis virus (SINV) is a mosquito-borne alphavirus found in Eurasia, Africa, and Oceania. Clinical SINV infection is characterized by febrile rash and arthritis and sometimes prolonged arthralgia and myalgia. The pathophysiological mechanisms of musculoskeletal and rheumatic disease caused by SINV are inadequately understood. METHODS: We studied the muscle pathology of SINV infection ex vivo by examining a unique muscle biopsy obtained from a patient with chronic myalgia and arthralgia 6 months after acute SINV infection and assessed potential genetic predisposing factors by determining the human leukocyte antigen (HLA) and complement factor C4 genes and proteins. In addition, we performed in vitro SINV infections of primary human myoblasts and myotubes. RESULTS: In the muscle biopsy we found evidence of muscle regeneration due to previous necrotic lesions likely caused by earlier SINV infection. We showed that human myoblasts and myotubes were susceptible in vitro for SINV infection as the cells became immunoreactive for viral antigens and cytopathic effect was observed. The patient was homozygous for HLA-B*35 alleles and heterozygous for HLA-DRB1*01 and HLA-DRB1*03 alleles and had total deficiency of C4B protein. CONCLUSIONS: This study provides new insights concerning pathological processes leading to chronic symptoms in SINV infection and demonstrates for the first time the susceptibility of human myogenic cells to SINV infection.


Subject(s)
Alphavirus Infections/complications , Muscle Fibers, Skeletal/virology , Muscular Diseases/virology , Myoblasts/virology , Pain/complications , Sindbis Virus , Humans , Male , Middle Aged , Pain/virology , RNA, Viral/isolation & purification , Real-Time Polymerase Chain Reaction/methods , Reverse Transcriptase Polymerase Chain Reaction/methods
11.
Med Sci Monit ; 17(12): BR364-71, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22129894

ABSTRACT

BACKGROUND: The expression of microRNA-206 (miR-206) is high in skeletal muscle but low in most other tissues. The expression of miR-206 is increased in muscular dystrophy, suggesting its involvement in the pathogenesis of muscle diseases. To determine the role of miR-206 in muscle cell differentiation and explore a possible gene therapy vector, we constructed a miR-206 adenoviral expression vector (AdvmiR-206) and tested for transfection into C2C12 stem cells. MATERIAL/METHODS: A 355-bp PCR amplicon from C57B6 mouse skeletal muscle genomic DNA was inserted into the adenoviral shuttle vector pAdTrack-CMV, which was then co-transformed with the adenoviral backbone plasmid pAdEasy-1 into competent E. coli BJ5183 bacteria. The specificity and function of this recombinant adenoviral MiR-206 were studied in C2C12 cells by Northern blot, immunofluorescence, Western blot, and flow cytometry. RESULTS: Increased expression of miR-206 in AdvmiR-206 transfected C2C12 cells (P < 0.001) and resulted in morphological and biochemical changes over time that were similar to serum deprivation, including elongated cells and increased myosin heavy chain proteins. Even in the absence of serum deprivation, miR-206 overexpression accounted for a 50% reduction of S-phase cells (P < 0.01). Moreover, in untransfected C2C12 cells, the introduction of miR-206-specific antisense oligoribonucleotides inhibited the normal response to serum deprivation. Twenty-four hours after lipofection of antisense oligoribonucleotides, the number of elongated cells was reduced by half (P < 0.01). CONCLUSIONS: Collectively, these data support a role for miR-206 in myoblast differentiation. We foresee potential applications for the AdvmiR-206 vector in research and therapy.


Subject(s)
Adenoviridae/genetics , MicroRNAs/genetics , Muscle Development/genetics , Myoblasts/metabolism , Myoblasts/virology , Recombination, Genetic/genetics , Animals , Cell Cycle , Cell Differentiation/genetics , Cell Line , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal , Organ Specificity/genetics , Transfection
12.
PLoS Negl Trop Dis ; 14(8): e0008282, 2020 08.
Article in English | MEDLINE | ID: mdl-32817655

ABSTRACT

Muscle cells are potential targets of many arboviruses, such as Ross River, Dengue, Sindbis, and chikungunya viruses, that may be involved in the physiopathological course of the infection. During the recent outbreak of Zika virus (ZIKV), myalgia was one of the most frequently reported symptoms. We investigated the susceptibility of human muscle cells to ZIKV infection. Using an in vitro model of human primary myoblasts that can be differentiated into myotubes, we found that myoblasts can be productively infected by ZIKV. In contrast, myotubes were shown to be resistant to ZIKV infection, suggesting a differentiation-dependent susceptibility. Infection was accompanied by a caspase-independent cytopathic effect, associated with paraptosis-like cytoplasmic vacuolization. Proteomic profiling was performed 24h and 48h post-infection in cells infected with two different isolates. Proteome changes indicate that ZIKV infection induces an upregulation of proteins involved in the activation of the Interferon type I pathway, and a downregulation of protein synthesis. This work constitutes the first observation of primary human muscle cells susceptibility to ZIKV infection, and differentiation-dependent restriction of infection from myoblasts to myotubes. Since myoblasts constitute the reservoir of stem cells involved in reparation/regeneration in muscle tissue, the infection of muscle cells and the viral-induced alterations observed here could have consequences in ZIKV infection pathogenesis.


Subject(s)
Cell Differentiation , Muscle Cells/metabolism , Muscle Cells/virology , Proteomics , Zika Virus Infection , Cell Death , Cell Line , Cytopathogenic Effect, Viral , Disease Susceptibility , Host-Pathogen Interactions , Humans , Interferon Type I/metabolism , Muscle Cells/pathology , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/virology , Myoblasts/metabolism , Myoblasts/virology , Proteins/metabolism , Stem Cells , Virus Replication , Zika Virus/pathogenicity , Zika Virus Infection/pathology , Zika Virus Infection/virology
13.
Wei Sheng Wu Xue Bao ; 49(9): 1259-64, 2009 Sep.
Article in Zh | MEDLINE | ID: mdl-20030068

ABSTRACT

OBJECTIVE: To construct the recombinant baculovirus expressing Infectious bursal disease (IBDV) VP2 gene in the chicken primary myoblast cells. METHODS: A proteinase K digestion and phenol-chloroform extraction method was used to extract dsRNA genome from IBDV. VP2 gene was amplified by Reverse Transcription Polymerase Chain Reaction (RT-PCR) with the genome RNA as template. The pFastBac-pCMV-VP2 baculovirus transfer vector was constructed by inserting VP2 gene under the immediate-early promoter of cytomegalovirus. The VP2 recombinant bacmid was obtained by Bac-to-Bac system and transfected sf9 insect cell to acquire VP2 recombinant baculovirus. After amplification of recombinant baculovirus on cell passages, the recombinant virus was seeded on chicken primary myoblast cells with 50 multiplicity of infection (MOI), and the cells were harvested at 72 hours after infection. RESULTS: Sodium Dodecyl Sulphate Poly-Acrylamide Gel Electrophoresis (SDS-PAGE) and Western blot results showed that the VP2 gene was successfully expressed in chicken primary myoblast cells. The product was a 48kDa protein and could be recognized by anti-IBDV serum. CONCLUSION: The recombinant baculovirus could efficiently delivery IBDV VP2 gene into chicken primary cells and that CMV, a mammalian-cell-active promoter, was functional in chicken primary cells and could direct the expression of VP2 antigen protein. The research can be a potential basis for the development of baculovirus vector vaccines for IBDV and other avian infectious disease.


Subject(s)
Baculoviridae/genetics , Gene Expression , Genetic Vectors/genetics , Myoblasts/metabolism , Viral Structural Proteins/genetics , Animals , Baculoviridae/physiology , Cell Line , Cells, Cultured , Chickens , Cloning, Molecular , Genetic Vectors/physiology , Infectious bursal disease virus/genetics , Infectious bursal disease virus/metabolism , Myoblasts/virology , Spodoptera , Viral Structural Proteins/metabolism
14.
FASEB J ; 21(9): 2074-85, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17325231

ABSTRACT

The major vascular endothelial growth factor (VEGF) isoforms are splice variants from a single gene that differ in their extent of heparin affinity due to the absence of the heparin binding domain in the smallest isoform (mouse VEGF120, human VEGF121). A long-held assumption that has guided the use of VEGF isoforms clinically has been that their differences in heparin binding dictate their ability to diffuse through tissue, with VEGF121 moving most freely and that the distribution of recombinant VEGF would have therapeutically relevant consequences. To test this assumption, we delivered the genes encoding these isoforms by myoblast-mediated gene transfer, a means of delivering genes to highly localized sites within muscle. Surprisingly, all isoforms induced comparable extremely localized physiological effects. Significantly, irrespective of the isoform delivered, the vessels passing within several micrometers of muscle fibers expressing VEGF displayed sharply delineated changes in morphology. The induction of capillary wrapping around VEGF-producing fibers, and of vascular malformations in the muscle at high levels, did not differ among isoforms. These results indicate that heparin binding is not essential for the localization of VEGF in adult tissue and suggest that the preferential delivery of VEGF121 cDNA for clinical applications may not have a physiological basis.


Subject(s)
Heparin/metabolism , Neovascularization, Physiologic/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Alternative Splicing , Animals , Arterioles/drug effects , Arterioles/growth & development , Biological Availability , Capillaries/drug effects , Capillaries/growth & development , Cells, Cultured/metabolism , Cells, Cultured/transplantation , Cells, Cultured/virology , Diffusion , Ear, External/blood supply , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Hemangioma/etiology , Hemangioma/pathology , Male , Mice , Mice, SCID , Molecular Weight , Muscle Neoplasms/etiology , Muscle Neoplasms/pathology , Muscle, Skeletal/blood supply , Myoblasts/metabolism , Myoblasts/transplantation , Myoblasts/virology , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/pharmacology , RNA, Messenger/biosynthesis , Retroviridae/genetics , Transduction, Genetic , Transgenes , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
15.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 30(4): 498-505, 2008 Aug.
Article in Zh | MEDLINE | ID: mdl-18795628

ABSTRACT

OBJECTIVE: To construct the recombinant adenovirus containing herpes simplex virus-1 virion protein (VP) 22 and human microdystrophin gene, then the adenovirus was transfected into C2C12 myoblast and studied on the property of protein transduction with VP22-mediated microdystrophin in C2C12 myoblast. METHODS: The full-length VP22 cDNA was obtained from recombinant plasmid pSINrep5-VP22 with PCR, and the product was directionally inserted into pShuttle-CMV to acquire the plasmid pCMV-VP22. Microdystrophin cDNA was obtained from recombinant plasmid pBSK-micro digested with restrictive endonuclease NotI, and the product was directionally inserted into pCMV-VP22 to acquire the plasmid pCMV-VP22-MICDYS. The plasmid of pCMV-VP22-MICDYS was lined with Pme I, and the fragment containing VP22-microdystrophin was reclaimed and transfected into E1 coli BJ5183 with plasmid pAdeasy-1. After having been screened by selected media, the extracted plasmid of positive bacteria was transfected into HEK293 cells with liposome and was identified by observing the cytopathic effect of cells and by PCR method to acquire the recombinant adenovirus Ad-VP22-MICDYS. Finally, the C2C12 myoblast were transfected with the recombinant adenovirus Ad-VP22-MICDYS and Ad-MICDYS, and the expression of microdystrophin was detected by RT-PCR, Western blot and immunocytochemistry. RESULTS: The recombinant adenovirus including VP22 and microdystrophin gene was successfully constructed. VP22 transferred VP22-microdystrophin fused protein from infected C2C12 myoblast into uninfected cells and enhance the expression of microdystrophin in myoblast. CONCLUSIONS: Recombinant adenovirus containing VP22 and microdystrophin gene was constructed successfully. VP22 can enhance the expression with microdystrophin in myoblast. It lays the foundation for further studying on VP22-mediated recombinant including microdystrophin gene to cure Duchenne muscular dystrophy.


Subject(s)
Adenoviridae/genetics , Dystrophin/genetics , Genetic Vectors/genetics , Transduction, Genetic , Viral Structural Proteins/genetics , Adenoviridae/physiology , Animals , Cell Line , Dystrophin/metabolism , Genetic Vectors/metabolism , Humans , Mice , Myoblasts/metabolism , Myoblasts/virology , Simplexvirus/genetics , Simplexvirus/metabolism , Viral Structural Proteins/metabolism , Virion/genetics , Virion/metabolism
16.
Skelet Muscle ; 7(1): 7, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28506310

ABSTRACT

BACKGROUND: shRNA lentiviral vectors are extensively used for gene knockdowns in mammalian cells, and non-target shRNAs typically are considered the proper experimental control for general changes caused by RNAi. However, the effects of non-target lentivirus controls on the modulation of cell signaling pathways remain largely unknown. In this study, we evaluated the effect of control lentiviral transduction on oxytocin receptor (OXTR) expression through the ERK/MAPK pathway in mouse and human skeletal muscle cells, on myogenic activity, and in vivo on mouse muscle regeneration. Furthermore, we mined published data for the influence of viral infections on OXTR levels in human populations and found that unrelated viral pathologies have a common consequence: diminished levels of OXTR. METHODS: We examined the change in OXTR mRNA expression upon transduction with control and Smad3-targeting viral vectors through real time RT-PCR and Western blotting, and confirmed with immunofluorescence. Changes in Smad3 and OXTR expression were examined both in vitro with mouse and human myoblasts and in vivo in mouse satellite cells. The general effects of viral infections on OXTR downregulation in humans were also examined by analyzing published Gene Expression Omnibus (GEO) datasets. The change in myoblast myogenic activity caused by the viral transduction (the percent of Pax7 + Ki67+ cells) was examined by immunofluorescence. RESULTS: Results shown in this work establish that lentiviral control vectors significantly downregulate OXTR expression at mRNA and protein levels and diminish key downstream effectors of OXTR, ERK signaling, reducing the myogenic proliferation of infected cells. This effect is evolutionarily conserved between mouse and human myogenic cells, and it manifests in satellite cells after control lentiviral transduction of mice in vivo. Furthermore, an examination of published datasets uncovered similar OXTR downregulation in humans that are afflicted with different viral infections. Additionally, cells transduced with Smad3-targeting shRNA downregulate OXTR even more than cells transduced with control viruses. CONCLUSIONS: Our work suggests that experimental cohorts transduced with control viruses may not behave the same as un-transduced cells and animals, specifically that control viral vectors significantly change the intensity of key cell-signaling pathways, such as OXTR/ERK. Our results further demonstrate that lentiviral transduction significantly decreases myogenic proliferation and suggest that viral infections in general may play a role in decreasing muscle health and regeneration, a decline in metabolic health, and a lower sense of well-being, as these rely on effective OXTR signaling. Additionally, our data suggest pathway crosstalk between TGF-ß/pSmad3 and OXTR, implying that sustained attenuation of the TGF-ß/pSmad3 pathway will reduce pro-regenerative OXTR/pERK signaling.


Subject(s)
Receptors, Oxytocin/genetics , Signal Transduction , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Virus Diseases/metabolism , Animals , Cells, Cultured , Down-Regulation , Evolution, Molecular , Humans , Lentivirus/pathogenicity , Mice , Mice, Inbred C57BL , Myoblasts/metabolism , Myoblasts/virology , Receptors, Oxytocin/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/virology , Virus Diseases/genetics
17.
PLoS One ; 12(1): e0170080, 2017.
Article in English | MEDLINE | ID: mdl-28095450

ABSTRACT

Infectious bursal disease virus (IBDV), a member of the Birnaviridae family, is responsible for a devastating immunosuppressive disease affecting juvenile domestic chickens. IBDV particles are naked icosahedrons enclosing a bipartite double-stranded RNA genome harboring three open reading frames (ORF). One of these ORFs codes for VP5, a non-structural polypeptide dispensable for virus replication in tissue culture but essential for IBDV pathogenesis. Using two previously described recombinant viruses, whose genomes differ in a single nucleotide, expressing or not the VP5 polypeptide, we have analyzed the role of this polypeptide during the IBDV replication process. Here, we show that VP5 is not involved in house-keeping steps of the virus replication cycle; i.e. genome transcription/replication, protein translation and virus assembly. Although infection with the VP5 expressing and non-expressing viruses rendered similar intracellular infective progeny yields, striking differences were detected on the ability of their progenies to exiting infected cells. Experimental data shows that the bulk of the VP5-expressing virus progeny efficiently egresses infected cells during the early phase of the infection, when viral metabolism is peaking and virus-induced cell death rates are as yet minimal, as determined by qPCR, radioactive protein labeling and quantitative real-time cell death analyses. In contrast, the release of the VP5-deficient virus progeny is significantly abridged and associated to cell death. Taken together, data presented in this report show that IBDV uses a previously undescribed VP5-dependent non-lytic egress mechanism significantly enhancing the virus dissemination speed. Ultrastructural analyses revealed that newly assembled IBDV virions associate to a vesicular network apparently facilitating their trafficking from virus assembly factories to the extracellular milieu, and that this association requires the expression of the VP5 polypeptide.


Subject(s)
Birnaviridae Infections/virology , Infectious bursal disease virus/pathogenicity , Viral Nonstructural Proteins/metabolism , Virion/metabolism , Virus Release/physiology , Virus Replication , Animals , Birnaviridae Infections/metabolism , Cells, Cultured , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Muscle, Skeletal/virology , Myoblasts/cytology , Myoblasts/metabolism , Myoblasts/virology , Quail
18.
Hum Gene Ther ; 17(2): 193-205, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16454653

ABSTRACT

Until recently, adenovirus-based gene therapy has been almost exclusively based on human adenovirus serotype 5 (Ad5). The aim of this study was to systematically compare the efficiency of transduction of primary muscle cells from various species by two adenoviral vectors from subgroups C and D. Transduction of a panel of myoblasts demonstrated a striking specificity of an Ad19a-based replication-defective E1-deleted vector (Ad19aEGFP) for human cells, whereas the Ad5-based vector had high affinity for nonhuman primate myoblasts. Transgene expression correlated well with cell-associated vector genomes. Up to 6.59% of the initially applied Ad19aEGFP vector particles were taken up by human myoblasts, as compared with 0.1% of the corresponding Ad5 vector. Remarkably, Ad19aEGFP but not Ad5EGFP efficiently transduced differentiated human myotubes, an in vitro model for skeletal muscle transduction. Uptake of Ad19aEGFP vector particles in human myotubes was 12-fold more efficient than that of Ad5EGFP. Moreover, both vectors demonstrated an early block at the level of vector uptake in mouse myoblasts and rat L6 cells. Investigation of the underlying mechanism for binding and uptake of the two vectors by human myoblasts showed high susceptibility for Ad19a to neuraminidase and wheat germ agglutinin (WGA) lectin, whereas Ad5-mediated transduction was dependent on binding to the coxsackie-adenovirus receptor (CAR) and sensitive to soluble RGD peptide and heparin. Our study offers insights into species-dependent factors that determine Ad tropism and, moreover, provides a basis for application of the novel Ad19a-based vector for gene transfer into human skeletal muscle.


Subject(s)
Adenoviruses, Human/genetics , Genetic Therapy/methods , Genetic Vectors/pharmacology , Muscle, Skeletal/virology , Animals , Cells, Cultured , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Epitopes/chemistry , Epitopes/metabolism , Flow Cytometry/methods , Genetic Vectors/genetics , Heparitin Sulfate/metabolism , Humans , Mice , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/virology , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Myoblasts/cytology , Myoblasts/virology , N-Acetylneuraminic Acid/chemistry , N-Acetylneuraminic Acid/metabolism , Rats , Receptors, Virus/chemistry , Receptors, Virus/metabolism , Species Specificity , Transduction, Genetic , Tropism , Virus Replication
19.
PLoS One ; 10(7): e0132099, 2015.
Article in English | MEDLINE | ID: mdl-26154387

ABSTRACT

Myogenesis is defined as growth, differentiation and repair of muscles where cell fusion of myoblasts to multinucleated myofibers is one major characteristic. Other cell fusion events in humans are found with bone resorbing osteoclasts and placental syncytiotrophoblasts. No unifying gene regulation for natural cell fusions has been found. We analyzed skeletal muscle biopsies of competitive cyclists for muscle-specific attributes and expression of human endogenous retrovirus (ERV) envelope genes due to their involvement in cell fusion of osteoclasts and syncytiotrophoblasts. Comparing muscle biopsies from post- with the pre-competitive seasons a significant 2.25-fold increase of myonuclei/mm fiber, a 2.38-fold decrease of fiber area/nucleus and a 3.1-fold decrease of satellite cells (SCs) occurred. We propose that during the pre-competitive season SC proliferation occurred following with increased cell fusion during the competitive season. Expression of twenty-two envelope genes of muscle biopsies demonstrated a significant increase of putative muscle-cell fusogenic genes Syncytin-1 and Syncytin-3, but also for the non-fusogenic erv3. Immunohistochemistry analyses showed that Syncytin-1 mainly localized to the sarcolemma of myofibers positive for myosin heavy-chain isotypes. Cellular receptors SLC1A4 and SLC1A5 of Syncytin-1 showed significant decrease of expression in post-competitive muscles compared with the pre-competitive season, but only SLC1A4 protein expression localized throughout the myofiber. Erv3 protein was strongly expressed throughout the myofiber, whereas envK1-7 localized to SC nuclei and myonuclei. Syncytin-1 transcription factors, PPARγ and RXRα, showed no protein expression in the myofiber, whereas the pCREB-Ser133 activator of Syncytin-1 was enriched to SC nuclei and myonuclei. Syncytin-1, Syncytin-3, SLC1A4 and PAX7 gene regulations along with MyoD1 and myogenin were verified during proliferating or actively-fusing human primary myoblast cell cultures, resembling muscle biopsies of cyclists. Myoblast treatment with anti-Synycytin-1 abrogated cell fusion in vitro. Our findings support functional roles for ERV envelope proteins, especially Syncytin-1, contributing to cell fusion of myotubes.


Subject(s)
Endogenous Retroviruses/genetics , Exercise , Genes, Viral , Myoblasts/cytology , Myoblasts/virology , Physical Endurance , Adolescent , Bicycling , Cell Fusion , Cells, Cultured , Cryoultramicrotomy , Fluorescent Antibody Technique , Gene Expression Profiling , Gene Expression Regulation , Humans , Male , Muscle Fibers, Skeletal/metabolism , Real-Time Polymerase Chain Reaction , Satellite Cells, Skeletal Muscle/metabolism , Time Factors
20.
PLoS One ; 8(11): e79628, 2013.
Article in English | MEDLINE | ID: mdl-24223983

ABSTRACT

Besides the classical respiratory and systemic symptoms, unusual complications of influenza A infection in humans involve the skeletal muscles. Numerous cases of acute myopathy and/or rhabdomyolysis have been reported, particularly following the outbreak of pandemic influenza A(H1N1) in 2009. The pathogenesis of these influenza-associated myopathies (IAM) remains unkown, although the direct infection of muscle cells is suspected. Here, we studied the susceptibility of cultured human primary muscle cells to a 2009 pandemic and a 2008 seasonal influenza A(H1N1) isolate. Using cells from different donors, we found that differentiated muscle cells (i. e. myotubes) were highly susceptible to infection by both influenza A(H1N1) isolates, whereas undifferentiated cells (i. e. myoblasts) were partially resistant. The receptors for influenza viruses, α2-6 and α2-3 linked sialic acids, were detected on the surface of myotubes and myoblasts. Time line of viral nucleoprotein (NP) expression and nuclear export showed that the first steps of the viral replication cycle could take place in muscle cells. Infected myotubes and myoblasts exhibited budding virions and nuclear inclusions as observed by transmission electron microscopy and correlative light and electron microscopy. Myotubes, but not myoblasts, yielded infectious virus progeny that could further infect naive muscle cells after proteolytic treatment. Infection led to a cytopathic effect with the lysis of muscle cells, as characterized by the release of lactate dehydrogenase. The secretion of proinflammatory cytokines by muscle cells was not affected following infection. Our results are compatible with the hypothesis of a direct muscle infection causing rhabdomyolysis in IAM patients.


Subject(s)
Influenza A Virus, H1N1 Subtype/physiology , Muscle, Skeletal/cytology , Muscle, Skeletal/virology , Pandemics , Seasons , Cell Death , Cell Differentiation , Cell Proliferation , Humans , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/virology , Myoblasts/cytology , Myoblasts/virology , Receptors, Cell Surface/metabolism , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL