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











Publication year range
1.
Sci Signal ; 5(237): ra58, 2012 Aug 14.
Article in English | MEDLINE | ID: mdl-22894835

ABSTRACT

PTEN (phosphatase and tensin homolog deleted on chromosome 10) and MAST2 (microtubule-associated serine and threonine kinase 2) interact with each other through the PDZ domain of MAST2 (MAST2-PDZ) and the carboxyl-terminal (C-terminal) PDZ domain-binding site (PDZ-BS) of PTEN. These two proteins function as negative regulators of cell survival pathways, and silencing of either one promotes neuronal survival. In human neuroblastoma cells infected with rabies virus (RABV), the C-terminal PDZ domain of the viral glycoprotein (G protein) can target MAST2-PDZ, and RABV infection triggers neuronal survival in a PDZ-BS-dependent fashion. These findings suggest that the PTEN-MAST2 complex inhibits neuronal survival and that viral G protein disrupts this complex through competition with PTEN for binding to MAST2-PDZ. We showed that the C-terminal sequences of PTEN and the viral G protein bound to MAST2-PDZ with similar affinities. Nuclear magnetic resonance structures of these complexes exhibited similar large interaction surfaces, providing a structural basis for their binding specificities. Additionally, the viral G protein promoted the nuclear exclusion of PTEN in infected neuroblastoma cells in a PDZ-BS-dependent manner without altering total PTEN abundance. These findings suggest that formation of the PTEN-MAST2 complex is specifically affected by the viral G protein and emphasize how disruption of a critical protein-protein interaction regulates intracellular PTEN trafficking. In turn, the data show how the viral protein might be used to decipher the underlying molecular mechanisms and to clarify how the subcellular localization of PTEN regulates neuronal survival.


Subject(s)
Glycoproteins/metabolism , Microtubule-Associated Proteins/metabolism , Models, Molecular , Neurons/physiology , PTEN Phosphohydrolase/metabolism , Protein Serine-Threonine Kinases/metabolism , Rabies virus/metabolism , Viral Proteins/metabolism , Binding, Competitive , Blotting, Western , Calorimetry , Cell Line, Tumor , Cell Survival/physiology , Glycoproteins/chemistry , Humans , Immunohistochemistry , Isotope Labeling , Microtubule-Associated Proteins/chemistry , Neurons/metabolism , Nuclear Magnetic Resonance, Biomolecular , PDZ Domains/physiology , PTEN Phosphohydrolase/chemistry , Protein Serine-Threonine Kinases/chemistry , Spectrometry, Fluorescence , Viral Proteins/chemistry
2.
Structure ; 19(10): 1518-24, 2011 Oct 12.
Article in English | MEDLINE | ID: mdl-22000519

ABSTRACT

PTPN4, a human tyrosine phosphatase, protects cells against apoptosis. This protection could be abrogated by targeting the PDZ domain of this phosphatase with a peptide mimicking the C-terminal sequence of the G protein of an attenuated rabies virus strain. Here, we demonstrate that glioblastoma death is triggered upon intracellular delivery of peptides, either from viral origin or from known endogenous ligands of PTPN4-PDZ, such as the C terminus sequence of the glutamate receptor subunit GluN2A. The killing efficiency of peptides closely reflects their affinities for the PTPN4-PDZ. The crystal structures of two PTPN4-PDZ/peptide complexes allow us to pinpoint the main structural determinants of binding and to synthesize a peptide of high affinity for PTPN4-PDZ enhancing markedly its cell death capacity. These results allow us to propose a potential mechanism for the efficiency of peptides and provide a target and a robust framework for the design of new pro-death compounds.


Subject(s)
Cell Death , Glioblastoma/pathology , PDZ Domains , Peptides/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 4/metabolism , Amino Acid Sequence , Cell Line, Tumor , Flow Cytometry , Glioblastoma/metabolism , Humans , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Multiprotein Complexes/metabolism , Point Mutation , Protein Binding , Protein Structure, Secondary , Rabies virus/chemistry , Receptors, N-Methyl-D-Aspartate/metabolism , Sequence Alignment , Viral Proteins/chemical synthesis , Viral Proteins/pharmacology
3.
J Neurovirol ; 17(4): 353-67, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21805057

ABSTRACT

Rabies virus (RABV) is a neurotropic virus transmitted by the bite of an infected animal that triggers a fatal encephalomyelitis. During its migration in the nervous system (NS), RABV triggers an innate immune response, including a type I IFN response well known to limit viral infections. We showed that although the neuroinvasive RABV strain CVS-NIV dampens type I IFN signaling by inhibiting IRF3 phosphorylation and STAT2 translocation, an early and transient type I IFN response is still triggered in the infected neuronal cells and NS. This urged us to investigate the role of type I IFN on RABV infection. We showed that primary mouse neurons (DRGs) of type I IFN(α/ß) receptor deficient mice (IFNAR(-/-) mice) were more susceptible to RABV than DRGs of WT mice. In addition, exogenous type I IFN is partially efficient in preventing and slowing down infection in human neuroblastoma cells. Intra-muscular inoculation of type I IFNAR deficient mice [IFNAR(-/-) mice and NesCre ((+/-)) IFNAR ((flox/flox)) mice lacking IFNAR in neural cells of neuroectodermal origin only] with RABV reveals that the type I IFN response limits RABV dissemination in the inoculated muscle, slows down invasion of the spinal cord, and delays mortality. Thus, the type I IFN which is still produced in the NS during RABV infection is efficient enough to reduce neuroinvasiveness and pathogenicity and partially protect the host from fatal infection.


Subject(s)
Interferon Type I , Neurons/immunology , Rabies virus/physiology , Rabies/immunology , Receptor, Interferon alpha-beta/deficiency , Signal Transduction/immunology , Spinal Cord/immunology , Animals , Cell Line, Tumor , Female , Flow Cytometry , Humans , Immunohistochemistry , Injections, Intramuscular , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon Type I/biosynthesis , Interferon Type I/immunology , Interferon Type I/pharmacology , Mice , Mice, Knockout , Neuroblastoma/immunology , Neuroblastoma/pathology , Neuroblastoma/virology , Neurons/virology , Primary Cell Culture , Rabies/mortality , Rabies/pathology , Rabies/virology , Real-Time Polymerase Chain Reaction , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/immunology , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/metabolism , Spinal Cord/virology , Survival Rate , Viral Load/immunology
4.
J Virol ; 85(13): 6657-68, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21525357

ABSTRACT

The neurotropic rabies virus (RABV) has developed several evasive strategies, including immunoevasion, to successfully infect the nervous system (NS) and trigger a fatal encephalomyelitis. Here we show that expression of LGP2, a protein known as either a positive or negative regulator of the RIG-I-mediated innate immune response, is restricted in the NS. We used a new transgenic mouse model (LGP2 TG) overexpressing LGP2 to impair the innate immune response to RABV and thus revealed the role of the RIG-I-mediated innate immune response in RABV pathogenesis. After infection, LGP2 TG mice exhibited reduced expression of inflammatory/chemoattractive molecules, beta interferon (IFN-ß), and IFN-stimulated genes in their NS compared to wild-type (WT) mice, demonstrating the inhibitory function of LGP2 in the innate immune response to RABV. Surprisingly, LGP2 TG mice showed more viral clearance in the brain and lower morbidity than WT mice, indicating that the host innate immune response, paradoxically, favors RABV neuroinvasiveness and morbidity. LGP2 TG mice exhibited similar neutralizing antibodies and microglia activation to those of WT mice but showed a reduction of infiltrating CD4(+) T cells and less disappearance of infiltrating CD8(+) T cells. This occurred concomitantly with reduced neural expression of the IFN-inducible protein B7-H1, an immunoevasive protein involved in the elimination of infiltrated CD8(+) T cells. Our study shows that the host innate immune response favors the infiltration of T cells and, at the same time, promotes CD8(+) T cell elimination. Thus, to a certain extent, RABV exploits the innate immune response to develop its immunoevasive strategy.


Subject(s)
B7-1 Antigen/metabolism , Immunity, Innate , Membrane Glycoproteins/metabolism , Peptides/metabolism , RNA Helicases/metabolism , Rabies virus/immunology , Rabies virus/pathogenicity , Rabies/immunology , Animals , B7-1 Antigen/genetics , B7-H1 Antigen , Brain/immunology , Brain/virology , Cell Line , Cell Line, Tumor , Humans , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , NIH 3T3 Cells , Neurons/immunology , Neurons/virology , Peptides/genetics , RNA Helicases/genetics , Rabies/virology , T-Lymphocytes/immunology
5.
Sci Signal ; 3(105): ra5, 2010 Jan 19.
Article in English | MEDLINE | ID: mdl-20086240

ABSTRACT

The capacity of a rabies virus to promote neuronal survival (a signature of virulence) or death (a marker of attenuation) depends on the cellular partners recruited by the PDZ-binding site (PDZ-BS) of its envelope glycoprotein (G). Neuronal survival requires the selective association of the PDZ-BS of G with the PDZ domains of two closely related serine-threonine kinases, MAST1 and MAST2. Here, we found that a single amino acid change in the PDZ-BS triggered the apoptotic death of infected neurons and enabled G to interact with additional PDZ partners, in particular the tyrosine phosphatase PTPN4. Knockdown of PTPN4 abrogated virus-mediated apoptosis. Thus, we propose that attenuation of rabies virus requires expansion of the set of host PDZ proteins with which G interacts, which interferes with the finely tuned homeostasis required for survival of the infected neuron.


Subject(s)
Rabies virus/pathogenicity , Viral Envelope Proteins/physiology , Amino Acid Substitution , Animals , Apoptosis , Cytoplasm , Mice , Neurons/virology , PDZ Domains , Protein Serine-Threonine Kinases/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 4 , Rabies , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Virulence
6.
PLoS Pathog ; 5(2): e1000315, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19247444

ABSTRACT

Human neurons express the innate immune response receptor, Toll-like receptor 3 (TLR3). TLR3 levels are increased in pathological conditions such as brain virus infection. Here, we further investigated the production, cellular localisation, and function of neuronal TLR3 during neuronotropic rabies virus (RABV) infection in human neuronal cells. Following RABV infection, TLR3 is not only present in endosomes, as observed in the absence of infection, but also in detergent-resistant perinuclear inclusion bodies. As well as TLR3, these inclusion bodies contain the viral genome and viral proteins (N and P, but not G). The size and composition of inclusion bodies and the absence of a surrounding membrane, as shown by electron microscopy, suggest they correspond to the previously described Negri Bodies (NBs). NBs are not formed in the absence of TLR3, and TLR3(-/-) mice -- in which brain tissue was less severely infected -- had a better survival rate than WT mice. These observations demonstrate that TLR3 is a major molecule involved in the spatial arrangement of RABV-induced NBs and viral replication. This study shows how viruses can exploit cellular proteins and compartmentalisation for their own benefit.


Subject(s)
Inclusion Bodies, Viral , Neurons/virology , Rabies virus/physiology , Rabies/pathology , Rabies/virology , Toll-Like Receptor 3/metabolism , Animals , Cell Compartmentation , Cells, Cultured , Data Interpretation, Statistical , Endosomes/metabolism , Endosomes/virology , Humans , Inclusion Bodies, Viral/immunology , Inclusion Bodies, Viral/metabolism , Inclusion Bodies, Viral/virology , Kaplan-Meier Estimate , Mice , Mice, Knockout , Microscopy, Electron , Neurons/metabolism , Nucleocapsid/metabolism , Rabies/immunology , Rabies/metabolism , Toll-Like Receptor 3/genetics , Virus Replication
7.
J Immunol ; 180(11): 7506-15, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18490751

ABSTRACT

Rabies virus is the etiological agent of an acute encephalitis, which in absence of post exposure treatment is fatal in almost all cases. Virus lethality rests on its ability to evade the immune response. In this study, we analyzed the role of the immuno-inhibitory molecule B7-H1 in this virus strategy. We showed that in the brain and spinal cord of mice, rabies virus infection resulted in significant up-regulation of B7-H1 expression, which is specifically expressed in infected neurons. Correlatively, clinical rabies in B7-H1(-/-) mice is markedly less severe than in wild-type mice. B7-H1(-/-) mice display resistance to rabies. Virus invasion is reduced and the level of migratory CD8 T cells increases into the nervous system, while CD4/CD8 ratio remains unchanged in the periphery. In vivo, neuronal B7-H1 expression is critically depending on TLR3 signaling and IFN-beta, because TLR3(-/-) mice--in which IFN-beta production is reduced--showed only a limited increase of B7-H1 transcripts after infection. These data provide evidence that neurons can express the B7-H1 molecule after viral stress or exposure to a particular cytokine environment. They show that the B7-H1/PD-1 pathway can be exploited locally and in an organ specific manner--here the nervous system--by a neurotropic virus to promote successful host invasion.


Subject(s)
Antigens, CD/metabolism , B7-1 Antigen/metabolism , Encephalitis, Viral/immunology , Membrane Glycoproteins/metabolism , Neurons/metabolism , Peptides/metabolism , Rabies virus/immunology , Rabies/immunology , Animals , Antigens, CD/immunology , B7-1 Antigen/immunology , B7-H1 Antigen , Brain/immunology , Brain/metabolism , Cell Line, Tumor , Cells, Cultured , Encephalitis, Viral/virology , Humans , Interferon-beta/immunology , Interferon-beta/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Membrane Glycoproteins/immunology , Mice , Mice, Mutant Strains , Neurons/immunology , Neurons/virology , Peptides/immunology , Rabies/metabolism , Rabies/virology , Spinal Cord/immunology , Spinal Cord/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
8.
Hum Immunol ; 68(4): 294-302, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17400066

ABSTRACT

Human Leukocyte Antigen (HLA)-G and E are nonclassical human MHC class I molecules. They may promote tolerance leading to virus and tumor immune escape. We recently described that the herpes simplex virus type 1 (HSV-1), a neurotropic virus inducing chronic infection and neuron latency, and rabies virus (RABV), a neuronotropic virus triggering acute neuron infection, up-regulate HLA-G expression in human neurons (NT2-N). Surface expression was only detected after RABV infection. We investigated here whether RABV and HSV-1 up-regulate HLA-E expression in human neuronal precursors (Ntera-2D/1). We found that RABV, not HSV-1, up-regulates HLA-E expression, nevertheless HLA-E could not be detected on the surface of RABV-infected Ntera-2D/1. Altogether these data suggest that HLA-G and not HLA-E could contribute to the immune escape of RABV. In contrast, there was no evidence that these molecules are used by latent HSV-1 infection. Thus, neurotropic viruses that escape the host immune response totally (RABV) or partially (HSV-1) regulate HLA-G expression on human neuronal cells differentially.


Subject(s)
HLA Antigens/genetics , Herpesvirus 1, Human/immunology , Histocompatibility Antigens Class I/genetics , Neurons/immunology , Rabies virus/immunology , Cell Line , Cell Membrane/immunology , HLA Antigens/biosynthesis , HLA-G Antigens , Histocompatibility Antigens Class I/biosynthesis , Humans , Neurons/metabolism , Neurons/virology , Up-Regulation/immunology , HLA-E Antigens
10.
J Mol Neurosci ; 29(3): 185-94, 2006.
Article in English | MEDLINE | ID: mdl-17085778

ABSTRACT

Inflammation is an important factor in the pathogenesis of neurodegenerative diseases, such as Alzheimer's disease or multiple sclerosis, and during microbial infections of the nervous system. Glial cells were thought to be the main contributor for cytokine and chemokine production and Toll-like receptor (TLR) expression in the brain. Here, we report that human neurons express TLR-3, a major receptor in virus-mediated innate immune response. We established that these cells can mount a strong inflammatory response characterized by the expression of inflammatory cytokines (TNF-alpha, IL-6), chemokines (CCL-5 and CXCL-10), and antiviral molecules (2'5'OAS and IFN-beta) after treatment with dsRNA - a by-product of viral infection and ligand of TLR-3. This work firmly establishes that human neurons, in absence of glia, have the intrinsic machinery to trigger robust inflammatory, chemoattractive, and antiviral responses.


Subject(s)
Brain/immunology , Nerve Tissue Proteins/biosynthesis , Neurons/metabolism , Toll-Like Receptor 3/biosynthesis , Cell Differentiation/drug effects , Cell Line/drug effects , Cell Line/metabolism , Chemokines/biosynthesis , Chemokines/genetics , Cytokines/biosynthesis , Cytokines/genetics , Gene Expression Regulation/drug effects , Humans , Inflammation/immunology , Interferon Type I/pharmacology , Interferon-beta/biosynthesis , Interferon-beta/genetics , Lipopolysaccharides/pharmacology , Nerve Tissue Proteins/genetics , Neurons/immunology , Organ Specificity , Poly I-C/pharmacology , RNA, Double-Stranded/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Recombinant Proteins , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 1/genetics , Toll-Like Receptor 2/genetics , Toll-Like Receptor 3/genetics , Toll-Like Receptor 4/genetics , Virus Diseases/immunology
11.
J Virol ; 79(24): 15226-37, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16306594

ABSTRACT

HLA-G is a nonclassical human major histocompatibility complex class I molecule. It may promote tolerance, leading to acceptance of the semiallogeneic fetus and tumor immune escape. We show here that two viruses-herpes simplex virus type 1 (HSV-1), a neuronotropic virus inducing acute infection and neuron latency; and rabies virus (RABV), a neuronotropic virus triggering acute neuron infection-upregulate the neuronal expression of several HLA-G isoforms, including HLA-G1 and HLA-G5, the two main biologically active isoforms. RABV induces mostly HLA-G1, and HSV-1 induces mostly HLA-G3 and HLA-G5. HLA-G expression is upregulated in infected cells and neighboring uninfected cells. Soluble mediators, such as beta interferon (IFN-beta) and IFN-gamma, upregulate HLA-G expression in uninfected cells. The membrane-bound HLA-G1 isoform was detected on the surface of cultured RABV-infected neurons but not on the surface of HSV-1-infected cells. Thus, neuronotropic viruses that escape the host immune response totally (RABV) or partially (HSV-1) regulate HLA-G expression on human neuronal cells differentially. HLA-G may therefore be involved in the escape of certain viruses from the immune response in the nervous system.


Subject(s)
HLA Antigens/metabolism , Herpesvirus 1, Human/physiology , Histocompatibility Antigens Class I/metabolism , Nervous System/virology , Neurons/metabolism , HLA Antigens/genetics , HLA-G Antigens , Histocompatibility Antigens Class I/genetics , Humans , Nervous System/pathology , Neurons/virology , RNA, Messenger/biosynthesis , Rabies virus/physiology , Transcription, Genetic , Tumor Cells, Cultured , Up-Regulation/immunology
12.
J Virol ; 79(20): 12893-904, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16188991

ABSTRACT

To study the capacity of human neurons to mount innate immunity responses to viral infections, we infected cells of a human postmitotic neuron-derivative cell line, NT2-N, with rabies virus (RABV) and herpes simplex type 1 (HSV-1). Changes in neuronal gene expression were analyzed by use of Affymetrix microarrays. Applying a twofold cutoff, RABV increased the transcription of 228 genes, and HSV-1 increased the transcription of 263 genes. The most striking difference between the two infections concerns genes involved in immunity. These genes represent 24% of the RABV-upregulated genes and only 4.9% of the HSV-1-upregulated genes. Following RABV infection, the most upregulated genes belong to the immunity cluster and included almost exclusively genes for beta interferon (IFN-beta) primary and secondary responses as well as genes for chemokines (CCL-5, CXCL-10) and inflammatory cytokines (interleukin 6 [IL-6], tumor necrosis factor alpha, interleukin 1 alpha). In contrast, HSV-1 infection did not increase IFN-beta gene transcripts and triggered the production of only IL-6 and interferon regulatory factor 1 mRNAs. The microarray results were confirmed by real-time PCR, immunocytochemistry, and enzyme-linked immunosorbent assay. Human neurons were found to express Toll-like receptor 3. They produced IFN-beta after treatment with poly(I:C) but not with lipopolysaccharide. Thus, human neurons can mount an innate immunity response to double-stranded RNA. These observations firmly establish that human neurons, in absence of glia, have the intrinsic machinery to sense virus infection.


Subject(s)
Herpes Simplex/virology , Herpesvirus 1, Human/immunology , Interferon-beta/biosynthesis , Membrane Glycoproteins/metabolism , Neurons/immunology , Neurons/virology , Rabies virus/immunology , Rabies/virology , Receptors, Cell Surface/metabolism , Cell Line , Enzyme-Linked Immunosorbent Assay , Herpes Simplex/immunology , Humans , Immunity, Innate , Immunohistochemistry , Interferon-beta/genetics , Neurons/metabolism , Polymerase Chain Reaction , RNA, Messenger/genetics , Rabies/immunology , Toll-Like Receptor 3 , Toll-Like Receptors
13.
Virology ; 314(2): 549-61, 2003 Sep 30.
Article in English | MEDLINE | ID: mdl-14554083

ABSTRACT

We report here that rabies virus strains, currently used to immunize wildlife against rabies, induce not only caspase-dependent apoptosis in the human lymphoblastoid Jurkat T cell line (Jurkat-vect), but also a caspase-independent pathway involving the apoptosis-inducing factor (AIF). In contrast, a strain of neurotropic RV that does not induce apoptosis did not activate caspases or induce AIF translocation. Bcl-2 overproduction in Jurkat T cells (Jurkat-Bcl-2) abolished both pathways. ERA infection and production were similar in Jurkat-vect and Jurkat-Bcl-2 cells, indicating Bcl-2 has no direct antiviral effects. Bcl-2 production is naturally upregulated by day 3 in ERA-infected Jurkat-vect cultures. The increase in Bcl-2 levels seems to be controlled by the virus infection itself and results in the establishment of long-term, persistently infected cultures that continue to produce virus. Thus, in infections with live RV vaccine strains, infected cells may be productive reservoirs of virus in the long term. This may account for the high efficacy of live rabies vaccines.


Subject(s)
Apoptosis , Jurkat Cells/virology , Proto-Oncogene Proteins c-bcl-2/metabolism , Rabies Vaccines , Rabies virus/physiology , Apoptosis Inducing Factor , Caspases/metabolism , Cell Line , Flavoproteins/genetics , Flavoproteins/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Rabies/physiopathology , Rabies/virology , Rabies virus/pathogenicity , Vaccines, Attenuated
14.
Ann N Y Acad Sci ; 1010: 598-603, 2003 Dec.
Article in English | MEDLINE | ID: mdl-15033799

ABSTRACT

We report that non-neurotropic rabies virus (RV) strains, currently used to immunize wildlife against rabies, induces not only a caspase-dependent apoptosis in the human lymphoblastoid Jurkat T cell line (Jurkat-vect), but also a caspase-independent pathway. Cell redistribution of the apoptosis-inducing factor (AIF) was observed in Jurkat-vect infected with RV vaccine strain. Bcl-2 overproduction in Jurkat T cells (Jurkat-Bcl-2) abolished both caspase activation and AIF distribution. In contrast, strain of neurotropic RV did not induce apoptosis. The inverse correlation of the induction of apoptosis and the capacity of a virus strain to invade the brain suggests that blockage of apoptosis could be a strategy selected by neurotropic virus to favor its progression through the nervous system.


Subject(s)
Apoptosis/physiology , Brain/virology , Rabies virus/pathogenicity , Apoptosis Inducing Factor , Caspases/metabolism , Flavoproteins/metabolism , Humans , Jurkat Cells , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL