Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Biochem Soc Trans ; 2024 May 16.
Article in English | MEDLINE | ID: mdl-38752830

ABSTRACT

Despite having the highest risk of progressing to severe disease due to lack of acquired immunity, the youngest children living in areas of highly intense malaria transmission have long been observed to be infected at lower rates than older children. Whether this observation is due to reduced exposure to infectious mosquito bites from behavioral and biological factors, maternally transferred immunity, genetic factors, or enhanced innate immunity in the young child has intrigued malaria researchers for over half a century. Recent evidence suggests that maternally transferred immunity may be limited to early infancy and that the young child's own immune system may contribute to control of malarial symptoms early in life and prior to the development of more effective adaptive immunity. Prospective studies of active and passive detection of Plasmodium falciparum blood-stage infections have identified young children (<5 years old) who remain uninfected through a defined surveillance period despite living in settings of highly intense malaria transmission. Yet, little is known about the potential immunological basis for this 'aparasitemic' phenotype. In this review, we summarize the observational evidence for this phenotype in field studies and examine potential reasons why these children escape detection of parasitemia, covering factors that are either extrinsic or intrinsic to their developing immune system. We discuss the challenges of distinguishing malaria protection from lack of malaria exposure in field studies. We also identify gaps in our knowledge regarding cellular immunity in the youngest age group and propose directions that researchers may take to address these gaps.

2.
JCI Insight ; 9(11)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38687615

ABSTRACT

A systems analysis was conducted to determine the potential molecular mechanisms underlying differential immunogenicity and protective efficacy results of a clinical trial of the radiation-attenuated whole-sporozoite PfSPZ vaccine in African infants. Innate immune activation and myeloid signatures at prevaccination baseline correlated with protection from P. falciparum parasitemia in placebo controls. These same signatures were associated with susceptibility to parasitemia among infants who received the highest and most protective PfSPZ vaccine dose. Machine learning identified spliceosome, proteosome, and resting DC signatures as prevaccination features predictive of protection after highest-dose PfSPZ vaccination, whereas baseline circumsporozoite protein-specific (CSP-specific) IgG predicted nonprotection. Prevaccination innate inflammatory and myeloid signatures were associated with higher sporozoite-specific IgG Ab response but undetectable PfSPZ-specific CD8+ T cell responses after vaccination. Consistent with these human data, innate stimulation in vivo conferred protection against infection by sporozoite injection in malaria-naive mice while diminishing the CD8+ T cell response to radiation-attenuated sporozoites. These data suggest a dichotomous role of innate stimulation for malaria protection and induction of protective immunity by whole-sporozoite malaria vaccines. The uncoupling of vaccine-induced protective immunity achieved by Abs from more protective CD8+ T cell responses suggests that PfSPZ vaccine efficacy in malaria-endemic settings may be constrained by opposing antigen presentation pathways.


Subject(s)
Immunity, Innate , Malaria Vaccines , Malaria, Falciparum , Plasmodium falciparum , Sporozoites , Vaccines, Attenuated , Malaria Vaccines/immunology , Malaria Vaccines/administration & dosage , Immunity, Innate/immunology , Humans , Animals , Malaria, Falciparum/prevention & control , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Mice , Vaccines, Attenuated/immunology , Vaccines, Attenuated/administration & dosage , Sporozoites/immunology , Sporozoites/radiation effects , CD8-Positive T-Lymphocytes/immunology , Infant , Protozoan Proteins/immunology , Antibodies, Protozoan/immunology , Female , Parasitemia/immunology , Parasitemia/prevention & control , Immunoglobulin G/immunology , Immunoglobulin G/blood , Vaccine Efficacy
3.
Proc Natl Acad Sci U S A ; 121(13): e2313672121, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38502693

ABSTRACT

Memory B cells (MBCs) play a critical role in protection against homologous and variant pathogen challenge by either differentiating to plasma cells (PCs) or to germinal center (GC) B cells. The human MBC compartment contains both switched IgG+ and unswitched IgM+ MBCs; however, whether these MBC subpopulations are equivalent in their response to B cell receptor cross-linking and their resulting fates is incompletely understood. Here, we show that IgG+ and IgM+ MBCs can be distinguished based on their response to κ-specific monoclonal antibodies of differing affinities. IgG+ MBCs responded only to high-affinity anti-κ and differentiated almost exclusively toward PC fates. In contrast, IgM+ MBCs were eliminated by apoptosis by high-affinity anti-κ but responded to low-affinity anti-κ by differentiating toward GC B cell fates. These results suggest that IgG+ and IgM+ MBCs may play distinct yet complementary roles in response to pathogen challenge ensuring the immediate production of high-affinity antibodies to homologous and closely related challenges and the generation of variant-specific MBCs through GC reactions.


Subject(s)
Immunoglobulin Class Switching , Memory B Cells , Humans , B-Lymphocytes , Antigens , Immunoglobulin G , Immunoglobulin M , Immunologic Memory
4.
Curr Opin Immunol ; 77: 102227, 2022 08.
Article in English | MEDLINE | ID: mdl-35724448

ABSTRACT

The world's struggle to contain the SARS-CoV-2 epidemic, primarily through vaccination, has highlighted the importance of better understanding the biology of B cells that participate in defense against infectious diseases, both acute and chronic. Here, we focus on a population of human B cells, termed atypical B cells (ABCs), that comprise a distinct B-cell lineage that differentiates from naive B cells in an interferon-γ-driven process, and are infrequent in healthy individuals but significantly expanded in chronic infectious diseases, including malaria, as well as in systemic autoimmune diseases such as systemic lupus erythematosus (SLE). Recent comparisons of ABCs by single-cell RNAseq provided evidence that ABCs in diverse chronic infectious diseases and in systemic autoimmune diseases are highly related and share common drivers of differentiation and expansion. However, ABCs in different diseases are not identical and also show discrete disease-specific features. Here, we compare and contrast key features of two ABC populations, namely those that are expanded in individuals living in malaria-endemic areas of the world versus those in SLE patients. This comparison is of interest as it appears that unique features of these two diseases result in participation of autoreactive ABCs in parasite-specific responses in malaria but in pathogenic autoimmune responses in SLE. A better understanding of the commonality and differences in the ABC responses in these two diseases may provide critical insights into the development of vaccines that drive pathogen-specific antibody responses and avoid autoimmunity.


Subject(s)
COVID-19 , Communicable Diseases , Lupus Erythematosus, Systemic , Malaria , Autoimmunity , Humans , SARS-CoV-2
5.
Sci Adv ; 7(22)2021 05.
Article in English | MEDLINE | ID: mdl-34039612

ABSTRACT

Chronic infectious diseases have a substantial impact on the human B cell compartment including a notable expansion of B cells here termed atypical B cells (ABCs). Using unbiased single-cell RNA sequencing (scRNA-seq), we uncovered and characterized heterogeneities in naïve B cell, classical memory B cells, and ABC subsets. We showed remarkably similar transcriptional profiles for ABC clusters in malaria, HIV, and autoimmune diseases and demonstrated that interferon-γ drove the expansion of ABCs in malaria. These observations suggest that ABCs represent a separate B cell lineage with a common inducer that further diversifies and acquires disease-specific characteristics and functions. In malaria, we identified ABC subsets based on isotype expression that differed in expansion in African children and in B cell receptor repertoire characteristics. Of particular interest, IgD+IgMlo and IgD-IgG+ ABCs acquired a high antigen affinity threshold for activation, suggesting that ABCs may limit autoimmune responses to low-affinity self-antigens in chronic malaria.

6.
Nat Immunol ; 22(5): 537-539, 2021 05.
Article in English | MEDLINE | ID: mdl-33888897

Subject(s)
Malaria , Humans
7.
Immunol Rev ; 292(1): 139-148, 2019 11.
Article in English | MEDLINE | ID: mdl-31553065

ABSTRACT

T cells exposed to persistent antigen in the inflammatory environment of chronic infections often show progressive loss of effector functions, high expression of inhibitory receptors and distinct transcriptional programs. T cells in this functional state are termed "exhausted" and T cell exhaustion is associated with inefficient control of infections. A remarkably similar scenario has been described for B cells during chronic infections in humans, including malaria, in which case a subpopulation of atypical memory B cells (MBCs) greatly expands and these MBCs show attenuation of B cell receptor signaling, loss of the B cell effector functions of antibody and cytokine production, high expression of inhibitory receptors and distinct transcriptional profiles. The expansion of these MBCs is also associated with inefficient control of infections. Despite the similarities with exhausted T cells we speculate that at least in malaria, atypical MBCs may not be exhausted but rather may be functional, possibly even beneficial. Our recent results suggest that we simply may not have known how to ask an atypical MBC to function. Thus, exhaustion may not be in the human B cell's vocabulary, at least not in malaria.


Subject(s)
Antigens, Protozoan/immunology , B-Lymphocytes/immunology , Immunologic Memory/immunology , Malaria/immunology , T-Lymphocytes/immunology , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/parasitology , Humans , Malaria/parasitology , Receptors, Antigen, B-Cell/immunology , Receptors, Antigen, B-Cell/metabolism , Signal Transduction/immunology , T-Lymphocytes/parasitology
8.
Sci Immunol ; 3(29)2018 11 30.
Article in English | MEDLINE | ID: mdl-30504208

ABSTRACT

Protective antibody responses to vaccination or infection depend on affinity maturation, a process by which high-affinity germinal center (GC) B cells are selected on the basis of their ability to bind, gather, and present antigen to T follicular helper (Tfh) cells. Here, we show that human GC B cells have intrinsically higher-affinity thresholds for both B cell antigen receptor (BCR) signaling and antigen gathering as compared with naïve B cells and that these functions are mediated by distinct cellular structures and pathways that ultimately lead to antigen affinity- and Tfh cell-dependent differentiation to plasma cells. GC B cells bound antigen through highly dynamic, actin- and ezrin-rich pod-like structures that concentrated BCRs. The behavior of these structures was dictated by the intrinsic antigen affinity thresholds of GC B cells. Low-affinity antigens triggered continuous engagement and disengagement of membrane-associated antigens, whereas high-affinity antigens induced stable synapse formation. The pod-like structures also mediated affinity-dependent antigen internalization by unconventional pathways distinct from those of naïve B cells. Thus, intrinsic properties of human GC B cells set thresholds for affinity selection.


Subject(s)
Antibody Affinity/immunology , Antigen Presentation/immunology , B-Lymphocytes/immunology , Germinal Center/cytology , Germinal Center/immunology , Antigen-Antibody Reactions , Humans
9.
PLoS Pathog ; 13(9): e1006576, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28953967

ABSTRACT

Many chronic infections, including malaria and HIV, are associated with a large expansion of CD21-CD27- 'atypical' memory B cells (MBCs) that exhibit reduced B cell receptor (BCR) signaling and effector functions. Little is known about the conditions or transcriptional regulators driving atypical MBC differentiation. Here we show that atypical MBCs in malaria-exposed individuals highly express the transcription factor T-bet, and that T-bet expression correlates inversely with BCR signaling and skews toward IgG3 class switching. Moreover, a longitudinal analysis of a subset of children suggested a correlation between the incidence of febrile malaria and the expansion of T-bethi B cells. The Th1-cytokine containing supernatants of malaria-stimulated PBMCs plus BCR cross linking induced T-bet expression in naïve B cells that was abrogated by neutralizing IFN-γ or blocking the IFN-γ receptor on B cells. Accordingly, recombinant IFN-γ plus BCR cross-linking drove T-bet expression in peripheral and tonsillar B cells. Consistent with this, Th1-polarized Tfh (Tfh-1) cells more efficiently induced T-bet expression in naïve B cells. These data provide new insight into the mechanisms underlying atypical MBC differentiation.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Differentiation/immunology , Gene Expression Regulation/immunology , Immunologic Memory/immunology , Interferon-gamma/biosynthesis , Malaria/immunology , Adolescent , Adult , Child , Child, Preschool , Female , Fetal Proteins/metabolism , Humans , Infant , Malaria/metabolism , Male , Receptors, Antigen, B-Cell/metabolism , T-Box Domain Proteins/metabolism , Young Adult
10.
Nanomedicine (Lond) ; 11(4): 377-90, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26786134

ABSTRACT

AIM: To design a theranostic capsule using the virus-like nanoparticle of the hepatitis E virus modified to display breast cancer cell targeting functional group (LXY30). METHODS: Five surface-exposed residues were mutated to cysteine to allow conjugation to maleimide-linked chemical groups via thiol-selective linkages. Engineered virus-like nanoparticles were then covalently conjugated to a breast cancer recognized ligand, LXY30 and an amine-coupled near-infrared fluorescence dye. RESULTS: LXY30-HEV VLP was checked for its binding and entry to a breast cancer cell line and for tumor targeting in vivo to breast cancer tissue in mice. The engineered virus-like nanoparticle not only targeted cancer cells, but also appeared immune silent to native hepatitis E virus antibodies due to epitope disruption at the antibody-binding site. CONCLUSION: These results demonstrate the production of a theranostic capsule suitable for cancer diagnostics and therapeutics based on surface modification of a highly stable virus-like nanoparticle.


Subject(s)
Breast Neoplasms/therapy , Capsid/drug effects , Hepatitis E virus/genetics , Amino Acid Sequence , Animals , Capsid/chemistry , Cell Line, Tumor , Cryoelectron Microscopy , Female , Fluorescent Dyes/chemistry , Heterografts , Humans , Mice , Mice, Inbred BALB C , Molecular Sequence Data
11.
Sci Rep ; 5: 8639, 2015 Mar 02.
Article in English | MEDLINE | ID: mdl-25728138

ABSTRACT

A majority of the human genome is transcribed into noncoding RNAs, of which the functions of long noncoding RNAs (lncRNAs) are poorly understood. Many host proteins and RNAs have been characterized for their roles in HIV/AIDS pathogenesis, but there is only one lncRNA, NEAT1, which is shown to affect the HIV-1 life cycle. We profiled 90 disease-related lncRNAs and found NRON (noncoding repressor of Nuclear Factor of Activated T cells [NFAT]) to be one of several lncRNAs whose expression was significantly altered following HIV-1 infection. The regulation of NRON expression during the HIV-1 life cycle was complex; its levels were reduced by the early viral accessory protein Nef and increased by the late protein Vpu. Consequently, Nef and Vpu also modulated activity of the transcription factor NFAT. The knockdown of NRON enhanced HIV-1 replication through increased activity of NFAT and the viral LTR. Using siRNA-mediated NFAT knockdown, we show the effects of NRON on HIV-1 replication to be mediated by NFAT, and the viral Nef and Vpu proteins to modulate NFAT activity through their effects on NRON. These findings add the lncRNA, NRON to the vast repertoire of host factors utilized by HIV for infection and persistence.


Subject(s)
HIV-1/physiology , Human Immunodeficiency Virus Proteins/metabolism , NFATC Transcription Factors/metabolism , RNA, Long Noncoding/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Virus Replication , nef Gene Products, Human Immunodeficiency Virus/metabolism , HEK293 Cells , HIV Infections/genetics , HIV Infections/virology , HIV Long Terminal Repeat/genetics , Humans , Jurkat Cells , Models, Biological
12.
Traffic ; 16(4): 398-416, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25615268

ABSTRACT

The hepatitis E virus (HEV) causes large outbreaks and sporadic cases of acute viral hepatitis in developing countries. In the developed world, HEV occurrence has increased as a result of zoonotic transmission from swine. The cellular aspects of HEV infection, especially the determinants of entry, are poorly understood. In the absence of a robust in vitro culture system for HEV, it is not possible to produce high titre infectious virus that can be labeled for tracking its internalization. We have therefore used an Escherichia coli expressed HEV-like particle (HEV-LP) to study HEV entry. Following internalization, the HEV-LP initially trafficks to Rab5-positive compartments en route to acidic lysosomal compartments where it is degraded. Using pharmacological inhibitors, dominant negative and constitutively active mutants, and siRNA-mediated perturbations, we show that HEV entry requires dynamin-2, clathrin, membrane cholesterol and actin, but is independent of factors associated with macropinocytosis. The HEV-LP results were further validated through infection of liver cells with virus from the stool of an infected patient. The comparative analysis also showed involvement of the phosphatidylinositol-3-kinase/Akt pathway in an early post-entry step of viral replication. This report provides a detailed description of endocytic processes associated with HEV infection.


Subject(s)
Cholesterol/metabolism , Clathrin/metabolism , Dynamin II/metabolism , Hepatitis E virus/physiology , Hepatocytes/virology , Liver/virology , Virus Internalization , Actins/metabolism , Cell Line , Cell Line, Tumor , Escherichia coli/metabolism , HEK293 Cells , HeLa Cells , Hepatocytes/metabolism , Humans , Liver/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Pinocytosis/physiology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Virus Replication/physiology
13.
PLoS One ; 9(5): e95920, 2014.
Article in English | MEDLINE | ID: mdl-24828336

ABSTRACT

BACKGROUND: The surrogate markers of HIV/AIDS progression include CD4 T cell count and plasma viral load. But, their reliability has been questioned in patients on anti-retroviral therapy (ART). Five microRNAs (miRNAs) - miR-16, miR-146b-5p, miR-150, miR-191 and miR-223 in peripheral blood mononuclear cells (PBMCs) were earlier found to assign HIV/AIDS patients into groups with varying CD4 T cell counts and viral loads. In this pilot study, we profiled the expression of these five miRNAs in PBMCs, and two of these miRNAs (miR-146b-5p and miR-150) in the plasma of HIV/AIDS patients, including those on ART and those who developed ART resistance, to evaluate if these are biomarkers of disease progression and therapy. RESULTS: We quantified miRNA levels by quantitative reverse transcription polymerase chain reaction (qRT-PCR) using RNA isolated from PBMCs and plasma of healthy persons or HIV-infected patients who were (1) asymptomatic; (2) symptomatic and ART naïve; (3) on ART; and (4) failing ART. Our results show miR-150 (p<0.01) and to a lesser extent miR-146b-5p (p<0.05) levels in PBMCs to reliably distinguish between ART-naïve AIDS patients, those on ART, and those developing drug resistance and failing ART. The plasma levels of these two miRNAs also varied significantly between patients in these groups and between patients and healthy controls (p values <0.05). CONCLUSIONS: We report for the first time that PBMC and plasma levels of miR-150 and miR-146b-5p are predictive of HIV/AIDS disease progression and therapy.


Subject(s)
Acquired Immunodeficiency Syndrome/genetics , MicroRNAs/genetics , Acquired Immunodeficiency Syndrome/drug therapy , Acquired Immunodeficiency Syndrome/pathology , Acquired Immunodeficiency Syndrome/virology , Adult , Alkynes , Antiretroviral Therapy, Highly Active , Benzoxazines/therapeutic use , Biomarkers/metabolism , CD4 Lymphocyte Count , Cyclopropanes , Disease Progression , Drug Resistance, Viral , Female , Gene Expression Regulation , HIV/drug effects , HIV/physiology , Humans , Lamivudine/therapeutic use , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , Male , Nevirapine/therapeutic use , Stavudine/therapeutic use , Treatment Outcome , Viral Load/drug effects , Zidovudine/therapeutic use
14.
Biochim Biophys Acta ; 1830(3): 2763-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23238519

ABSTRACT

BACKGROUND: The p38alpha MAP kinase pathway is involved in inflammation, cell differentiation, growth, apoptosis and production of pro-inflammatory cytokines TNF-alpha and IL-1beta. The overproduction of these cytokines plays an important role in cancer. The aim of this work was to design a peptide inhibitor on the basis of structural information of the active site of p38alpha. METHODS: A tetrapeptide, VWCS as p38alpha inhibitor was designed on the basis of structural information of the ATP binding site by molecular modeling. The inhibition study of peptide with p38alpha was performed by ELISA, binding study by Surface Plasmon Resonance and anti-proliferative assays by MTT and flow cytometry. RESULTS: The percentage inhibition of designed VWCS against pure p38alpha protein and serum of HNSCC patients was 70.30 and 71.5%, respectively. The biochemical assay demonstrated the K(D) and IC50 of the selective peptide as 7.22 x 10(-9) M and 20.08 nM, respectively. The VWCS as inhibitor significantly reduced viability of oral cancer KB cell line with an IC50 value of 10 microM and induced apoptosis by activating Caspase 3 and 7. CONCLUSIONS: VWCS efficiently interacted at the ATP binding pocket of p38alpha with high potency and can be used as a potent inhibitor in case of HNSCC. GENERAL SIGNIFICANCE: VWCS can act as an anticancer agent as it potentially inhibits the cell growth and induces apoptosis in oral cancer cell-line in a dose as well as time dependent manner. Hence, p38alpha MAP kinase inhibitor can be a potential therapeutic agent for human oral cancer.


Subject(s)
Antineoplastic Agents/chemistry , Carcinoma, Squamous Cell/drug therapy , Head and Neck Neoplasms/drug therapy , Mitogen-Activated Protein Kinase 14/chemistry , Neoplasm Proteins/chemistry , Oligopeptides/chemistry , Adenosine Triphosphate/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Binding Sites , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Caspases/genetics , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Design , Enzyme-Linked Immunosorbent Assay , Gene Expression/drug effects , Head and Neck Neoplasms/enzymology , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , Inhibitory Concentration 50 , Kinetics , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Mitogen-Activated Protein Kinase 14/genetics , Molecular Docking Simulation , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Oligopeptides/chemical synthesis , Oligopeptides/pharmacology , Protein Binding , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Squamous Cell Carcinoma of Head and Neck
15.
PLoS One ; 6(7): e22412, 2011.
Article in English | MEDLINE | ID: mdl-21799848

ABSTRACT

The hepatitis E virus (HEV) is a small RNA virus and the cause of acute viral hepatitis E. The open reading frame 3 protein (pORF3) of HEV appears to be a pleiotropic regulatory protein that helps in the establishment, propagation and progression of viral infection. However, the global cellular effects of this protein remain to be explored. In the absence of traditional in vitro viral infection systems or efficient replicon systems, we made an adenovirus based ORF3 protein expression system to study its effects on host cell gene expression. We infected Huh7 hepatoma cells with recombinant adenoviruses expressing pORF3 and performed microarray-based gene expression analyses. Several genes down regulated in pORF3-expressing cells were found to be under regulation of the liver-enriched hepatocyte nuclear factor 4 (HNF4), which regulates hepatocyte-specific gene expression. While HNF4 localizes to the nucleus, its phosphorylation results in impaired nuclear localization of HNF4. Here we report that pORF3 increases HNF4 phosphorylation through the ERK and Akt kinases, which results in impaired nuclear translocation of HNF4 and subsequently the down modulation of HNF4-responsive genes in pORF3-expressing cells. We propose that modulation of several hepatocyte specific genes by pORF3 will create an environment favorable for viral replication and pathogenesis.


Subject(s)
Cell Nucleus/metabolism , Gene Expression Profiling , Hepatitis E virus , Hepatocyte Nuclear Factor 4/metabolism , Liver/cytology , Liver/metabolism , Viral Proteins/metabolism , Active Transport, Cell Nucleus/genetics , Adenoviridae/genetics , Cell Line, Tumor , DNA, Recombinant/genetics , Down-Regulation/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , HEK293 Cells , Hepatitis E virus/physiology , Humans , Liver/virology , Organ Specificity , Phosphorylation/genetics , Proto-Oncogene Proteins c-akt/metabolism , Transcription, Genetic/genetics , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...