Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Ecotoxicol Environ Saf ; 264: 115447, 2023 Oct 01.
Article in English | MEDLINE | ID: mdl-37690176

ABSTRACT

As emerging pollutants in the environment, nanoplastics (NPs) can cross biological barriers and be enriched in organisms, posing a greatest threat to the health of livestock and humans. However, the size-dependent toxic effects of NPs in higher mammals remain largely unknown. To determine the size-dependent potential toxicities of NPs, we exposed mouse (AML-12) and human (L02) liver cell lines in vitro, and 6-week-old C57BL/6 mice (well-known preclinical model) in vivo to five different sizes of polystyrene NPs (PS-NPs) (20, 50, 100, 200 and 500 nm). We found that ultra-small NPs (20 nm) induced the highest cytotoxicity in mouse and human liver cell lines, causing oxidative stress and mitochondrial membrane potential loss on AML-12 cells. Unexpectedly in vivo, after long-term oral exposure to PS-NPs (75 mg/kg), medium NPs (200 nm) and large NPs (500 nm) induced significant hepatotoxicity, evidenced by increased oxidative stress, liver dysfunction, and lipid metabolism disorders. Most importantly, medium or large NPs generated local immunotoxic effects via recruiting and activating more numbers of neutrophils and monocytes in the liver or intestine, which potentially resulted in increased proinflammatory cytokine secretion and the tissue damage. The discrepancy in in vitro-in vivo toxic results might be attributed to the different properties of biodistribution and tissue accumulation of different sized NPs in vivo. Our study provides new insights regarding the hepatotoxicity and immunotoxicity of NPs on human and livestock health, warranting us to take immense measures to prevent these NPs-associated health damage.


Subject(s)
Antineoplastic Agents , Chemical and Drug Induced Liver Injury , Leukemia, Myeloid, Acute , Nanoparticles , Water Pollutants, Chemical , Humans , Animals , Mice , Mice, Inbred C57BL , Microplastics/toxicity , Polystyrenes/toxicity , Tissue Distribution , Livestock , Mammals
2.
J Clin Invest ; 130(4): 1823-1829, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32125290

ABSTRACT

Peptide MHC class II-based (pMHCII-based) nanomedicines trigger the formation of multicellular regulatory networks by reprogramming autoantigen-experienced CD4+ T cells into autoimmune disease-suppressing T regulatory type 1 (TR1) cells. We have shown that pMHCII-based nanomedicines displaying liver autoimmune disease-relevant yet ubiquitously expressed antigens can blunt various liver autoimmune disorders in a non-disease-specific manner without suppressing local or systemic immunity against infectious agents or cancer. Here, we show that such ubiquitous autoantigen-specific T cells are also awakened by extrahepatic tissue damage and that the corresponding TR1 progeny can suppress experimental autoimmune encephalomyelitis (EAE) and pancreatic ß cell autoreactivity. In mice having EAE, nanomedicines displaying either ubiquitous or CNS-specific epitopes triggered the formation and expansion of cognate TR1 cells and their recruitment to the CNS-draining lymph nodes, sparing their liver-draining counterparts. Surprisingly, in mice having both liver autoimmunity and EAE, liver inflammation sequestered these ubiquitous or even CNS-specific TR1 cells away from the CNS, abrogating their antiencephalitogenic activity. In these mice, only the ubiquitous antigen-specific TR1 cells suppressed liver autoimmunity. Thus, the scope of antigen spreading in autoimmune disorders is larger than previously anticipated, involving specificities expected to be silenced by mechanisms of tolerance; the regulatory activity, but not the retention of autoreactive TR1 cells, requires local autoantigen expression.


Subject(s)
Autoimmunity , Encephalomyelitis, Autoimmune, Experimental/immunology , Hepatitis, Autoimmune/immunology , Liver/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Autoantigens/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Hepatitis, Autoimmune/pathology , Histocompatibility Antigens Class II/immunology , Liver/pathology , Mice , Mice, Inbred NOD , T-Lymphocytes, Regulatory/pathology
3.
Nat Commun ; 10(1): 2150, 2019 05 14.
Article in English | MEDLINE | ID: mdl-31089130

ABSTRACT

Peptide-major histocompatibility complex class II (pMHCII)-based nanomedicines displaying tissue-specific autoantigenic epitopes can blunt specific autoimmune conditions by re-programming cognate antigen-experienced CD4+ T-cells into disease-suppressing T-regulatory type 1 (TR1) cells. Here, we show that single pMHCII-based nanomedicines displaying epitopes from mitochondrial, endoplasmic reticulum or cytoplasmic antigens associated with primary biliary cholangitis (PBC) or autoimmune hepatitis (AIH) can broadly blunt PBC, AIH and Primary Sclerosing Cholangitis in various murine models in an organ- rather than disease-specific manner, without suppressing general or local immunity against infection or metastatic tumors. Therapeutic activity is associated with cognate TR1 cell formation and expansion, TR1 cell recruitment to the liver and draining lymph nodes, local B-regulatory cell formation and profound suppression of the pro-inflammatory capacity of liver and liver-proximal myeloid dendritic cells and Kupffer cells. Thus, autoreactivity against liver-enriched autoantigens in liver autoimmunity is not disease-specific and can be harnessed to treat various liver autoimmune diseases broadly.


Subject(s)
Autoimmune Diseases/drug therapy , Histocompatibility Antigens Class II/immunology , Liver Diseases/drug therapy , Nanoparticles/administration & dosage , Peptides/administration & dosage , Aged , Animals , Autoantigens/immunology , Autoimmune Diseases/immunology , Cell Line , Disease Models, Animal , Epitopes/immunology , Female , Histocompatibility Antigens Class II/chemistry , Humans , Liver/drug effects , Liver/immunology , Liver Diseases/immunology , Male , Mice , Middle Aged , Nanomedicine/methods , Nanoparticles/chemistry , Peptides/chemistry , Peptides/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology
4.
Nat Nanotechnol ; 12(7): 701-710, 2017 07.
Article in English | MEDLINE | ID: mdl-28436959

ABSTRACT

We have shown that nanoparticles (NPs) can be used as ligand-multimerization platforms to activate specific cellular receptors in vivo. Nanoparticles coated with autoimmune disease-relevant peptide-major histocompatibility complexes (pMHC) blunted autoimmune responses by triggering the differentiation and expansion of antigen-specific regulatory T cells in vivo. Here, we define the engineering principles impacting biological activity, detail a synthesis process yielding safe and stable compounds, and visualize how these nanomedicines interact with cognate T cells. We find that the triggering properties of pMHC-NPs are a function of pMHC intermolecular distance and involve the sustained assembly of large antigen receptor microclusters on murine and human cognate T cells. These compounds show no off-target toxicity in zebrafish embryos, do not cause haematological, biochemical or histological abnormalities, and are rapidly captured by phagocytes or processed by the hepatobiliary system. This work lays the groundwork for the design of ligand-based NP formulations to re-program in vivo cellular responses using nanotechnology.


Subject(s)
Autoimmunity , Histocompatibility Antigens , Nanomedicine/methods , Nanoparticles/chemistry , Peptides , T-Lymphocytes, Regulatory/immunology , Animals , Histocompatibility Antigens/chemistry , Histocompatibility Antigens/immunology , Humans , Mice , Mice, Inbred NOD , Peptides/chemistry , Peptides/immunology , T-Lymphocytes, Regulatory/pathology
5.
J Immunol ; 193(7): 3296-307, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25165150

ABSTRACT

We investigated whether a prevalent epitope of the ß-cell-specific autoantigen islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP206-214) reaches regional Ag-presentation pathways via unprocessed polypeptide chains, as free IGRP206-214 peptide or via preformed IGRP206-214/K(d) complexes. This was accomplished by expressing bacterial artificial chromosome transgenes encoding wild-type (stable) or ubiquitinated (unstable) forms of IGRP in IGRP-deficient NOD mice carrying MHC class I-deficient ß-cells, dendritic cells, or B cells. We investigated the ability of the pancreatic lymph nodes of these mice to prime naive IGRP206-214-reactive CD8(+) T cells in vivo, either in response to spontaneous Ag shedding, or to synchronized forms of ß-cell necrosis or apoptosis. When IGRP was made unstable by targeting it for proteasomal degradation within ß-cells, the cross-priming, autoimmune-initiating potential of this autoantigen (designated autoantigenicity) was impaired. Yet at the same time, the direct presentation, CTL-targeting potential of IGRP (designated pathogenicity) was enhanced. The appearance of IGRP206-214 in regional Ag-presentation pathways was dissociated from transfer of IGRP206-214 or IGRP206-214/K(d) from ß cells to dendritic cells. These results indicate that autoantigenicity and pathogenicity are separable and inversely related properties and suggest that pathogenic autoantigens, capable of efficiently priming CTLs while marking target cells for CTL-induced killing, may have a critical balance of these two properties.


Subject(s)
Antigen Presentation , Autoantigens/immunology , Cross-Priming , Dendritic Cells/immunology , Glucose-6-Phosphatase/immunology , Insulin-Secreting Cells/immunology , Animals , Apoptosis/immunology , Autoantigens/genetics , B-Lymphocytes/immunology , B-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Dendritic Cells/pathology , Glucose-6-Phosphatase/genetics , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Insulin-Secreting Cells/pathology , Mice , Mice, Inbred NOD , Mice, Knockout , Necrosis
6.
PLoS One ; 8(6): e64787, 2013.
Article in English | MEDLINE | ID: mdl-23785406

ABSTRACT

Involvement of CD4(+) helper T (Th) cells is crucial for CD8(+) cytotoxic T lymphocyte (CTL)-mediated immunity. However, CD4(+) Th's signals that govern CTL survival and functional memory are still not completely understood. In this study, we assessed the role of CD4(+) Th cells with acquired antigen-presenting machineries in determining CTL fates. We utilized an adoptive co-transfer into CD4(+) T cell-sufficient or -deficient mice of OTI CTLs and OTII Th cells or Th cells with various gene deficiencies pre-stimulated in vitro by ovalbumin (OVA)-pulsed dendritic cell (DCova). CTL survival was kinetically assessed in these mice using FITC-anti-CD8 and PE-H-2K(b)/OVA257-264 tetramer staining by flow cytometry. We show that by acting via endogenous CD40L and IL-2, and acquired peptide-MHC-I (pMHC-I) complex signaling, CD4(+) Th cells enhance survival of transferred effector CTLs and their differentiation into the functional memory CTLs capable of protecting against highly-metastasizing tumor challenge. Moreover, RT-PCR, flow cytometry and Western blot analysis demonstrate that increased survival of CD4(+) Th cell-helped CTLs is matched with enhanced Akt1/NF-κB activation, down-regulation of TRAIL, and altered expression profiles with up-regulation of prosurvival (Bcl-2) and down-regulation of proapoptotic (Bcl-10, Casp-3, Casp-4, Casp-7) molecules. Taken together, our results reveal a previously unexplored mechanistic role for CD4(+) Th cells in programming CTL survival and memory recall responses. This knowledge could also aid in the development of efficient adoptive CTL cancer therapy.


Subject(s)
CD40 Ligand/metabolism , Histocompatibility Antigens Class I/immunology , Immunologic Memory , Interleukin-2/metabolism , Signal Transduction , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Apoptosis/genetics , Apoptosis/immunology , Cell Line, Tumor , Cell Survival/immunology , Cluster Analysis , Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression Profiling , Immunologic Memory/genetics , Mice , Mice, Knockout , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Helper-Inducer/metabolism
7.
Vet Res Commun ; 36(4): 245-50, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23007876

ABSTRACT

Mononuclear leukocytes of peripheral blood mononuclear cells (PBMCs) and regional lymphoid organs (RLOs) play a critical role in primary BTV replication and subsequent viral dissemination to distant systemic organs. The lesions in animals develop primarily as a result of vascular insult, presumably induced by the activity of viral and/or proinflammatory vasoactive mediators. Hence, the current study was designed in sheep to investigate the responses of potent vasoactivators, inducible nitric oxide synthase (iNOS) and/or nitric oxide (NO) in mononuclear leukocytes of PBMCs and RLOs. The results show that BTV infection of sheep led to enhanced transcription of iNOS in PBMCs and in particular RLOs. The BTV RNAs and/or antigens were readily demonstrable in these mononuclear leukocytes, suggesting the possible role of BTV in iNOS induction. Moreover, upon in vitro infection of PBMCs with BTV-23, iNOS was up-regulated in time-dependent fashion and correlated with increased NO production. The results from these in vivo and in vitro studies thus suggest iNOS and NO produced by mononuclear leukocytes may potentially contribute to vascular-related pathology of BT.


Subject(s)
Bluetongue virus/physiology , Bluetongue/virology , Leukocytes, Mononuclear/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/metabolism , Animals , Antigens, Viral/blood , Bluetongue/immunology , Bluetongue/pathology , Bluetongue virus/classification , Bluetongue virus/genetics , Female , Lymph Nodes/immunology , Lymph Nodes/metabolism , Male , RNA, Viral/blood , Real-Time Polymerase Chain Reaction/veterinary , Sheep , Spleen/immunology , Spleen/metabolism
8.
Vaccine ; 30(24): 3519-25, 2012 May 21.
Article in English | MEDLINE | ID: mdl-22484292

ABSTRACT

The limitations of highly active anti-retroviral therapy have necessitated the development of alternative therapeutics for human immunodeficiency virus type-1 (HIV-1)-infected patients with dysfunctional dendritic cells (DCs) and CD4(+) T cell deficiency. We previously demonstrated that HIV-1 Gp120-specific T cell-based Gp120-Texo vaccine by using ConA-stimulated C57BL/6 (B6) mouse CD8(+) T (ConA-T) cells with uptake of pcDNA(Gp120)-transfected B6 mouse DC line DC2.4 (DC2.4(Gp120))-released exosomes (EXO(Gp120)) was capable of stimulating DC and CD4(+) T cell-independent CD8(+) cytotoxic T lymphocyte (CTL) responses detected in wild-type B6 mice using non-specific PE-anti-CD44 and anti-IFN-γ antibody staining by flow cytometry. To assess effectiveness of Gp120-Texo vaccine in transgenic (Tg) HLA-A2 mice mimicking the human situation, we constructed adenoviral vector AdV(Gp120) expressing HIV-1 GP120 by recombinant DNA technology, and generated Gp120-Texo vaccine by using Tg HLA-A2 mouse CD8(+) ConA-T cells with uptake of AdV(Gp120)-transfected HLA-A2 mouse bone marrow DC (DC(Gp120))-released EXO(Gp120). We then performed animal studies to assess Gp120-Texo-induced stimulation of Gp120-specific CTL responses and antitumor immunity in Tg HLA-A2 mice. We demonstrate that Gp120-Texo vaccine stimulates Gp120-specific CTL responses detected in Tg HLA-A2 mice using Gp120-specific PE-HLA-A2/Gp120 peptide (KLTPLCVTL) tetramer staining by flow cytometry. These Gp120-specific CTLs are capable of further differentiating into functional effectors with killing activity to Gp120 peptide-pulsed splenocytes in vivo. In addition, Gp120-Texo vaccine also induces Gp120-specific preventive, therapeutic (for 6 day tumor lung metastasis) and CD4(+) T cell-independent long-term immunity against B16 melanoma BL6-10(Gp120/A2Kb) expressing both Gp120 and A2Kb (α1 and α2 domains of HLA-A2 and α3 domain of H-2K(b)) in Tg HLA-A2 mice. Taken together, the novel CD8(+) Gp120-Texo vaccine capable of stimulating efficient CD4(+) T cell-independent Gp120-specific CD8(+) CTL responses leading to therapeutic and long-term immunity in Tg HLA-A2 mice may represent a new immunotherapeutic vaccine for treatment of HIV-1 patients with CD4(+) T cell deficiency.


Subject(s)
AIDS Vaccines/immunology , CD8-Positive T-Lymphocytes/immunology , HIV Envelope Protein gp120/immunology , HIV Infections/therapy , Immunotherapy/methods , T-Lymphocytes, Cytotoxic/immunology , AIDS Vaccines/genetics , Adenoviridae/genetics , Animals , Disease Models, Animal , Genetic Vectors , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic
9.
Vet Immunol Immunopathol ; 141(3-4): 230-8, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21511346

ABSTRACT

The pathogenesis of bluetongue (BT) could vary with route of inoculation. Using laboratory-passaged moderately virulent bluetongue virus serotype 23 (BTV-23), one of the most prevalent Indian serotype, we investigated the pathogenesis of BT in intradermally (ID) and intravenously (IV) inoculated native sheep. The ID inoculation resulted in relatively increased clinical signs and lesions in many organs as compared to IV inoculation. BTV-23 detection by real-time RT-PCR and isolation studies revealed that ID inoculation can be more efficient than IV ones in disseminating and spreading virus to systemic organs, including pre-scapular draining lymph node, spleen, lungs and pulmonary artery. Furthermore, the ID inoculation resulted in early onset and increased humoral response with significant increase (P<0.01) in antibody titre at various intervals. Taken together, these data suggest that ID inoculation can be more potent in reproducing many aspects of natural infection, including clinical disease, viral and immune responses, and may be useful route in setting up experimental infections for challenge or pathogenesis studies using laboratory passaged BTVs.


Subject(s)
Bluetongue virus/pathogenicity , Bluetongue/virology , Animals , Bluetongue/immunology , Bluetongue/pathology , Bluetongue virus/classification , Bluetongue virus/genetics , Female , Lung/pathology , Male , Muscle, Skeletal/pathology , Muscle, Smooth, Vascular/pathology , Myocardium/pathology , RNA, Viral/genetics , RNA, Viral/metabolism , Sheep , Tongue/pathology
10.
Vaccine ; 29(19): 3538-47, 2011 Apr 27.
Article in English | MEDLINE | ID: mdl-21406265

ABSTRACT

The limitations of highly active anti-retroviral therapy (HAART) have necessitated the development of alternative therapeutics. In this study, we generated ovalbumin (OVA)-pulsed and pcDNAgp120-transfected dendritic cell (DC)-released exosomes (EXOova and EXOgp120) and ConA-stimulated C57BL/6 CD8(+) T cells. OVA- and Gp120-Texo vaccines were generated from CD8(+) T cells with uptake of EXOova and EXOgp120, respectively. We demonstrate that OVA-Texo stimulates in vitro and in vivo OVA-specific CD4(+) and CD8(+) cytotoxic T lymphocyte (CTL) responses leading to long-term immunity against OVA-expressing BL6-10(OVA) melanoma. Interestingly, CD8(+) T cell responses are DC and CD4(+) T cell independent. Importantly, Gp120-Texo also stimulates Gp120-specific CTL responses and long-term immunity against Gp120-expressing B16 melanoma. Therefore, this novel HIV-1-specific EXO-targeted Gp120-Texo vaccine may be useful in induction of efficient CTL responses in AIDS patients with DC dysfunction and CD4(+) T cell deficiency.


Subject(s)
AIDS Vaccines/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Dendritic Cells/immunology , HIV Envelope Protein gp120/immunology , Acquired Immunodeficiency Syndrome/prevention & control , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Line , Exosomes/immunology , Female , HIV-1/immunology , Immunity, Cellular , Immunologic Memory/immunology , Intercellular Adhesion Molecule-1/immunology , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/immunology
11.
Res Vet Sci ; 90(2): 212-4, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21349379

ABSTRACT

Bluetongue (BT), a serious disease of sheep and some wild ruminants, is caused by bluetongue virus (BTV), a member of the family, Reoviridae. The current research thrust for controlling BT is on development of efficient vaccines, necessitating clear understanding of ovine immunology. At present, comparative studies on cytokine gene expression profiles of naïve and BTV-sensitized peripheral blood mononuclear cells (PBMC) in sheep have not been clearly understood. In the present study, PBMC from naïve and BEI-inactivated-saponin-adjuvanted BTV-1 vaccinated sheep were stimulated in vitro with heterologous BTV-23. At various intervals, RT-qPCR was carried out to estimate cytokine (interferon-gamma, interleukin-12 and interleukin-2) mRNA expressions that are linked to cell-mediated immunity. The results showed that PBMC cytokine profiles were relatively increased both temporally and quantitatively in immunized sheep PBMC compared to naïve ones, suggesting that BTV-1 vaccination may prime immune system that can cross-react with BTV-23 antigens.


Subject(s)
Bluetongue virus , Cytokines/metabolism , Gene Expression Regulation/immunology , Leukocytes, Mononuclear/metabolism , Animals , Cytokines/genetics , Immunity, Cellular , Leukocytes, Mononuclear/immunology , Sheep
12.
Vet Microbiol ; 144(3-4): 310-8, 2010 Aug 26.
Article in English | MEDLINE | ID: mdl-20347236

ABSTRACT

The role of apoptosis in pathogenesis of bluetongue (BT) has been suggested from various in vitro studies. However, to date, no clear data are available regarding BTV-induced apoptosis and its consequences in natural host, sheep. In the present study, bluetongue virus (BTV)-induced apoptosis was studied in sheep blood and splenic mononuclear cells by analyzing annexin(+)-propidium iodide(-) early apoptotic cells, DNA ladder pattern, and caspase-3 gene expression. The onset of apoptosis and lymphocyte depletion in viraemic phase and IFN-alpha response indicated the involvement of BTV and IFN-alpha in the pathogenesis of BT. The development of Pasteurella pneumonia in 4 of 7 infected sheep during the experiment pointed to possible BTV-induced immuno-suppression and predisposition to secondary microbial infections. These results have significant implications not only in understanding immuno-pathological consequences but also in studying interactions of BTV with host cells.


Subject(s)
Bluetongue virus/classification , Bluetongue/virology , Immunosuppression Therapy/veterinary , Animals , Apoptosis , Caspase 3/genetics , Caspase 3/metabolism , Gene Expression Regulation , Leukocytes, Mononuclear/physiology , Leukocytes, Mononuclear/virology , Lung/cytology , Lymphoid Tissue/cytology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Serotyping , Sheep , Spleen/cytology , Time Factors , Viremia
13.
Vaccine ; 28(13): 2522-31, 2010 Mar 16.
Article in English | MEDLINE | ID: mdl-20117268

ABSTRACT

Bluetongue is a serious hemorrhagic disease of sheep, cattle and other ruminants causing economic losses worldwide. Recent invasion of multiple bluetongue virus serotypes (BTV) in various countries warrants immediate development of efficacious vaccine that targets more than one serotype. In the present study, the cross-protective efficacy of binary ethylenimine (BEI)-inactivated BTV-1 vaccine was evaluated in Indian native sheep against virulent heterologous BTV-23 serotype challenge. BTV-1 vaccination induced significant cell-mediated immunity (CMI) as determined by lymphoproliferative responses, and increased CD8 T cell, IL-2 and IFN-gamma responses. Both naïve and immunized sheep also showed increased CD4 T cell, IL-12 and IFN-alpha responses. Collectively, these data suggested that inactivated BTV-1 vaccine induced appreciable CMI and greatly reduced the severity of heterologous BTV-23 infection.


Subject(s)
Aziridines/pharmacology , Bluetongue virus/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cross Protection , Disinfectants/pharmacology , Viral Vaccines/immunology , Animals , Bluetongue/pathology , Bluetongue/prevention & control , Cell Proliferation , Cytotoxicity, Immunologic , India , Interferon-alpha/metabolism , Interferon-gamma/metabolism , Interleukin-12/metabolism , Interleukin-2/metabolism , Severity of Illness Index , Sheep , Vaccines, Inactivated/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...