Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
JCI Insight ; 6(12)2021 06 22.
Article in English | MEDLINE | ID: mdl-34014838

ABSTRACT

Although antiretroviral therapy suppresses HIV replication, it does not eliminate viral reservoirs or restore damaged lymphoid tissue, posing obstacles to HIV eradication. Using the SIV model of AIDS, we investigated the effect of mesenchymal stem/stromal cell (MSC) infusions on gut mucosal recovery, antiviral immunity, and viral suppression and determined associated molecular/metabolic signatures. MSC administration to SIV-infected macaques resulted in viral reduction and heightened virus-specific responses. Marked clearance of SIV-positive cells from gut mucosal effector sites was correlated with robust regeneration of germinal centers, restoration of follicular B cells and T follicular helper (Tfh) cells, and enhanced antigen presentation by viral trapping within the follicular DC network. Gut transcriptomic analyses showed increased antiviral response mediated by pathways of type I/II IFN signaling, viral restriction factors, innate immunity, and B cell proliferation and provided the molecular signature underlying enhanced host immunity. Metabolic analysis revealed strong correlations between B and Tfh cell activation, anti-SIV antibodies, and IL-7 expression with enriched retinol metabolism, which facilitates gut homing of antigen-activated lymphocytes. We identified potentially new MSC functions in modulating antiviral immunity for enhanced viral clearance predominantly through type I/II IFN signaling and B cell signature, providing a road map for multipronged HIV eradication strategies.


Subject(s)
Germinal Center , Intestinal Mucosa/immunology , Mesenchymal Stem Cells , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Animals , Cytokines/metabolism , Germinal Center/cytology , Germinal Center/immunology , Germinal Center/metabolism , Immunity, Humoral/immunology , Macaca mulatta , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/immunology
2.
Proc Natl Acad Sci U S A ; 116(49): 24819-24829, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31740620

ABSTRACT

Chronic gut inflammatory diseases are associated with disruption of intestinal epithelial barriers and impaired mucosal immunity. HIV-1 (HIV) causes depletion of mucosal CD4+ T cells early in infection and disruption of gut epithelium, resulting in chronic inflammation and immunodeficiency. Although antiretroviral therapy (ART) is effective in suppressing viral replication, it is incapable of restoring the "leaky gut," which poses an impediment for HIV cure efforts. Strategies are needed for rapid repair of the epithelium to protect intestinal microenvironments and immunity in inflamed gut. Using an in vivo nonhuman primate intestinal loop model of HIV/AIDS, we identified the pathogenic mechanism underlying sustained disruption of gut epithelium and explored rapid repair of gut epithelium at the intersection of microbial metabolism. Molecular, immunological, and metabolomic analyses revealed marked loss of peroxisomal proliferator-activated receptor-α (PPARα) signaling, predominant impairment of mitochondrial function, and epithelial disruption both in vivo and in vitro. To elucidate pathways regulating intestinal epithelial integrity, we introduced probiotic Lactobacillus plantarum into Simian immunodeficiency virus (SIV)-inflamed intestinal lumen. Rapid recovery of the epithelium occurred within 5 h of L. plantarum administration, independent of mucosal CD4+ T cell recovery, and in the absence of ART. This intestinal barrier repair was driven by L. plantarum-induced PPARα activation and restoration of mitochondrial structure and fatty acid ß-oxidation. Our data highlight the critical role of PPARα at the intersection between microbial metabolism and epithelial repair in virally inflamed gut and as a potential mitochondrial target for restoring gut barriers in other infectious or gut inflammatory diseases.


Subject(s)
Energy Metabolism/physiology , Gastrointestinal Microbiome/physiology , Intestines/immunology , Intestines/microbiology , Mitochondria/metabolism , PPAR alpha/metabolism , Animals , Antiretroviral Therapy, Highly Active , CD4-Positive T-Lymphocytes/immunology , Disease Models, Animal , Energy Metabolism/drug effects , Epithelium/immunology , HIV Infections , Humans , Immunity, Mucosal , Interleukin-1beta/metabolism , Intestines/pathology , Lactobacillus plantarum/physiology , Macaca mulatta , Male , Metabolomics , Mitochondria/drug effects , Probiotics/administration & dosage , Probiotics/therapeutic use , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology
3.
J Virol ; 92(13)2018 07 01.
Article in English | MEDLINE | ID: mdl-29669841

ABSTRACT

Subclinical viral infections (SVI), including cytomegalovirus (CMV), are highly prevalent in humans, resulting in lifelong persistence. However, the impact of SVI on the interplay between the host immunity and gut microbiota in the context of environmental exposures is not well defined. We utilized the preclinical nonhuman primate (NHP) model consisting of SVI-free (specific-pathogen-free [SPF]) rhesus macaques and compared them to the animals with SVI (non-SPF) acquired through natural exposure and investigated the impact of SVI on immune cell distribution and function, as well as on gut microbiota. These changes were examined in animals housed in the outdoor environment compared to the controlled indoor environment. We report that SVI are associated with altered immune cell subsets and gut microbiota composition in animals housed in the outdoor environment. Non-SPF animals harbored a higher proportion of potential butyrate-producing Firmicutes and higher numbers of lymphocytes, effector T cells, and cytokine-producing T cells. Surprisingly, these differences diminished following their transfer to the controlled indoor environment, suggesting that non-SPFs had increased responsiveness to environmental exposures. An experimental infection of indoor SPF animals with CMV resulted in an increased abundance of butyrate-producing bacteria, validating that CMV enhanced colonization of butyrate-producing commensals. Finally, non-SPF animals displayed lower antibody responses to influenza vaccination compared to SPF animals. Our data show that subclinical CMV infection heightens host immunity and gut microbiota changes in response to environmental exposures. This may contribute to the heterogeneity in host immune response to vaccines and environmental stimuli at the population level.IMPORTANCE Humans harbor several latent viruses that modulate host immunity and commensal microbiota, thus introducing heterogeneity in their responses to pathogens, vaccines, and environmental exposures. Most of our understanding of the effect of CMV on the immune system is based on studies of children acquiring CMV or of immunocompromised humans with acute or reactivated CMV infection or in ageing individuals. The experimental mouse models are genetically inbred and are completely adapted to the indoor laboratory environment. In contrast, nonhuman primates are genetically outbred and are raised in the outdoor environment. Our study is the first to report the impact of long-term subclinical CMV infection on host immunity and gut microbiota, which is evident only in the outdoor environment but not in the indoor environment. The significance of this study is in highlighting the impact of SVI on enhancing host immune susceptibility to environmental exposures and immune heterogeneity.


Subject(s)
Bacteria/classification , Cytomegalovirus Infections/veterinary , Cytomegalovirus/pathogenicity , Monkey Diseases/immunology , Monkey Diseases/microbiology , Animals , Bacteria/isolation & purification , Cytokines/metabolism , Cytomegalovirus Infections/immunology , Disease Models, Animal , Gastrointestinal Microbiome , Housing, Animal , Lymphocytes/metabolism , Macaca mulatta , Phylogeny , Specific Pathogen-Free Organisms , T-Lymphocytes/immunology
4.
mBio ; 8(3)2017 05 02.
Article in English | MEDLINE | ID: mdl-28465428

ABSTRACT

Antiviral innate host defenses against acute viral infections include suppression of host protein synthesis to restrict viral protein production. Less is known about mechanisms by which viral pathogens subvert host antiviral innate responses for establishing their replication and dissemination. We investigated early innate defense against human immunodeficiency virus (HIV) infection and viral evasion by utilizing human CD4+ T cell cultures in vitro and a simian immunodeficiency virus (SIV) model of AIDS in vivo Our data showed that early host innate defense against the viral infection involves GCN2-ATF4 signaling-mediated suppression of global protein synthesis, which is exploited by the virus for supporting its own replication during early viral infection and dissemination in the gut mucosa. Suppression of protein synthesis and induction of protein kinase GCN2-ATF4 signaling were detected in the gut during acute SIV infection. These changes diminished during chronic viral infection. HIV replication induced by serum deprivation in CD4+ T cells was linked to the induction of ATF4 that was recruited to the HIV long terminal repeat (LTR) to promote viral transcription. Experimental inhibition of GCN2-ATF4 signaling either by a specific inhibitor or by amino acid supplementation suppressed the induction of HIV expression. Enhancing ATF4 expression through selenium administration resulted in reactivation of latent HIV in vitro as well as ex vivo in the primary CD4+ T cells isolated from patients receiving suppressive antiretroviral therapy (ART). In summary, HIV/SIV exploits the early host antiviral response through GCN2-ATF4 signaling by utilizing ATF4 for activating the viral LTR transcription to establish initial viral replication and is a potential target for HIV prevention and therapy.IMPORTANCE Understanding how HIV overcomes host antiviral innate defense response in order to establish infection and dissemination is critical for developing prevention and treatment strategies. Most investigations focused on the viral pathogenic mechanisms leading to immune dysfunction following robust viral infection and dissemination. Less is known about mechanisms that enable HIV to establish its presence despite rapid onset of host antiviral innate response. Our novel findings provide insights into the viral strategy that hijacks the host innate response of the suppression of protein biosynthesis to restrict the virus production. The virus leverages transcription factor ATF4 expression during the GCN2-ATF4 signaling response and utilizes it to activate viral transcription through the LTR to support viral transcription and production in both HIV and SIV infections. This unique viral strategy is exploiting the innate response and is distinct from the mechanisms of immune dysfunction after the critical mass of viral loads is generated.


Subject(s)
Activating Transcription Factor 4/metabolism , CD4-Positive T-Lymphocytes/virology , HIV-1/physiology , Host-Pathogen Interactions , Immunity, Innate , Protein Serine-Threonine Kinases/metabolism , Virus Replication , Activating Transcription Factor 4/genetics , Animals , CD4-Positive T-Lymphocytes/drug effects , Cells, Cultured , Disease Models, Animal , Gastrointestinal Tract/virology , HIV Infections/drug therapy , HIV Infections/immunology , HIV Infections/virology , HIV-1/immunology , HIV-1/pathogenicity , Humans , Immune Evasion , Macaca mulatta , Protein Serine-Threonine Kinases/genetics , Selenium/pharmacology , Signal Transduction , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus , Viral Load , Virus Latency
5.
Sci Rep ; 6: 24723, 2016 04 22.
Article in English | MEDLINE | ID: mdl-27102350

ABSTRACT

Chronic HIV infection results in impairment of gut-associated lymphoid tissue leading to systemic immune activation. We previously showed that in early SIV-infected rhesus macaques intestinal dysfunction is initiated with the induction of the IL-1ß pathway in the small intestine and reversed by treatment with an exogenous Lactobacillus plantarum strain. Here, we provide evidence that the transcriptomes of L. plantarum and ileal microbiota are not altered shortly after SIV infection. L. plantarum adapts to the small intestine by expressing genes required for tolerating oxidative stress, modifying cell surface composition, and consumption of host glycans. The ileal microbiota of L. plantarum-containing healthy and SIV+ rhesus macaques also transcribed genes for host glycan metabolism as well as for cobalamin biosynthesis. Expression of these pathways by bacteria were proposed but not previously demonstrated in the mammalian small intestine.


Subject(s)
Gastrointestinal Microbiome , Gene Expression Profiling , Ileum/pathology , Lactobacillus plantarum/genetics , Simian Acquired Immunodeficiency Syndrome/pathology , Animals , Ileum/microbiology , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/microbiology
6.
mBio ; 7(2): e02265, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26933058

ABSTRACT

UNLABELLED: Nontyphoidal Salmonella enterica serovar Typhimurium is a frequent cause of bloodstream infections in children and HIV-infected adults in sub-Saharan Africa. Most isolates from African patients with bacteremia belong to a single sequence type, ST313, which is genetically distinct from gastroenteritis-associated ST19 strains, such as 14028s and SL1344. Some studies suggest that the rapid spread of ST313 across sub-Saharan Africa has been facilitated by anthroponotic (person-to-person) transmission, eliminating the need for Salmonella survival outside the host. While these studies have not ruled out zoonotic or other means of transmission, the anthroponotic hypothesis is supported by evidence of extensive genomic decay, a hallmark of host adaptation, in the sequenced ST313 strain D23580. We have identified and demonstrated 2 loss-of-function mutations in D23580, not present in the ST19 strain 14028s, that impair multicellular stress resistance associated with survival outside the host. These mutations result in inactivation of the KatE stationary-phase catalase that protects high-density bacterial communities from oxidative stress and the BcsG cellulose biosynthetic enzyme required for the RDAR (red, dry, and rough) colonial phenotype. However, we found that like 14028s, D23580 is able to elicit an acute inflammatory response and cause enteritis in mice and rhesus macaque monkeys. Collectively, these observations suggest that African S. Typhimurium ST313 strain D23580 is becoming adapted to an anthroponotic mode of transmission while retaining the ability to infect and cause enteritis in multiple host species. IMPORTANCE: The last 3 decades have witnessed an epidemic of invasive nontyphoidal Salmonella infections in sub-Saharan Africa. Genomic analysis and clinical observations suggest that the Salmonella strains responsible for these infections are evolving to become more typhoid-like with regard to patterns of transmission and virulence. This study shows that a prototypical African nontyphoidal Salmonella strain has lost traits required for environmental stress resistance, consistent with an adaptation to a human-to-human mode of transmission. However, in contrast to predictions, the strain remains capable of causing acute inflammation in the mammalian intestine. This suggests that the systemic clinical presentation of invasive nontyphoidal Salmonella infections in Africa reflects the immune status of infected hosts rather than intrinsic differences in the virulence of African Salmonella strains. Our study provides important new insights into the evolution of host adaptation in bacterial pathogens.


Subject(s)
Adaptation, Biological , Salmonella Infections/microbiology , Salmonella typhimurium/enzymology , Salmonella typhimurium/physiology , Stress, Physiological , Africa South of the Sahara/epidemiology , Animals , Catalase/genetics , Catalase/metabolism , Disease Models, Animal , Epidemics , Glucosyltransferases/genetics , Glucosyltransferases/metabolism , Humans , Macaca mulatta , Mice , Mutant Proteins/genetics , Mutant Proteins/metabolism , Salmonella Infections/epidemiology , Salmonella typhimurium/genetics , Salmonella typhimurium/isolation & purification
7.
J Med Primatol ; 44(5): 241-52, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26275157

ABSTRACT

BACKGROUND: The impact of HIV infection on pattern recognition receptor (PRR) expression in gut-associated lymphoid tissue and its association with dysbiosis is not well understood. METHODS: PRR and cytokine gene expression were examined in mesenteric lymph nodes (mLN) of rhesus macaques during acute and chronic (untreated and early antiretroviral (ART) treated) infections. Gene expression was correlated with microbial abundance in the gut and immune activation. RESULTS: PRR expression rapidly increases during acute infection and is significantly decreased in chronic infection. Early ART maintains elevated PRR expression. Correlation analysis revealed three distinct groups of bacterial taxa that were associated with gene expression changes in infection. CONCLUSIONS: PRR and cytokine gene expression in the gut-draining mLN are rapidly modulated in response to viral infection and are correlated with gut dysbiosis. These data suggest that the dysregulation of PRR and related cytokine expression may contribute to chronic immune activation in SIV infection.


Subject(s)
Anti-Retroviral Agents/pharmacology , Cytokines/genetics , Gastrointestinal Microbiome , Gene Expression Regulation , Receptors, Pattern Recognition/genetics , Simian Acquired Immunodeficiency Syndrome/genetics , Acute Disease , Animals , Chronic Disease , Cytokines/metabolism , Lymph Nodes/immunology , Lymph Nodes/virology , Receptors, Pattern Recognition/metabolism , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/physiology
8.
PLoS Pathog ; 10(8): e1004311, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25166758

ABSTRACT

HIV causes rapid CD4+ T cell depletion in the gut mucosa, resulting in immune deficiency and defects in the intestinal epithelial barrier. Breakdown in gut barrier integrity is linked to chronic inflammation and disease progression. However, the early effects of HIV on the gut epithelium, prior to the CD4+ T cell depletion, are not known. Further, the impact of early viral infection on mucosal responses to pathogenic and commensal microbes has not been investigated. We utilized the SIV model of AIDS to assess the earliest host-virus interactions and mechanisms of inflammation and dysfunction in the gut, prior to CD4+ T cell depletion. An intestinal loop model was used to interrogate the effects of SIV infection on gut mucosal immune sensing and response to pathogens and commensal bacteria in vivo. At 2.5 days post-SIV infection, low viral loads were detected in peripheral blood and gut mucosa without CD4+ T cell loss. However, immunohistological analysis revealed the disruption of the gut epithelium manifested by decreased expression and mislocalization of tight junction proteins. Correlating with epithelial disruption was a significant induction of IL-1ß expression by Paneth cells, which were in close proximity to SIV-infected cells in the intestinal crypts. The IL-1ß response preceded the induction of the antiviral interferon response. Despite the disruption of the gut epithelium, no aberrant responses to pathogenic or commensal bacteria were observed. In fact, inoculation of commensal Lactobacillus plantarum in intestinal loops led to rapid anti-inflammatory response and epithelial tight junction repair in SIV infected macaques. Thus, intestinal Paneth cells are the earliest responders to viral infection and induce gut inflammation through IL-1ß signaling. Reversal of the IL-1ß induced gut epithelial damage by Lactobacillus plantarum suggests synergistic host-commensal interactions during early viral infection and identify these mechanisms as potential targets for therapeutic intervention.


Subject(s)
Interleukin-1beta/biosynthesis , Paneth Cells/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Animals , Fluorescent Antibody Technique , Host-Parasite Interactions/immunology , Immunohistochemistry , Interleukin-1beta/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/ultrastructure , Intestinal Mucosa/virology , Macaca mulatta , Male , Microscopy, Electron, Transmission , Oligonucleotide Array Sequence Analysis , Paneth Cells/metabolism , Paneth Cells/virology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tight Junctions/ultrastructure , Viral Load
9.
PLoS One ; 8(8): e72789, 2013.
Article in English | MEDLINE | ID: mdl-24023646

ABSTRACT

Chronic immune activation despite long-term therapy poses an obstacle to immune recovery in HIV infection. The role of antigen presenting cells (APCs) in chronic immune activation during HIV infection remains to be fully determined. APCs, the frontline of immune defense against pathogens, are capable of distinguishing between pathogens and non-pathogenic, commensal bacteria. We hypothesized that HIV infection induces dysfunction in APC immune recognition and response to some commensal bacteria and that this may promote chronic immune activation. Therefore we examined APC inflammatory cytokine responses to commensal lactobacilli. We found that APCs from HIV-infected patients produced an enhanced inflammatory response to Lactobacillus plantarum WCFS1 as compared to APCs from healthy, HIV-negative controls. Increased APC expression of TLR2 and CD36, signaling through p38-MAPK, and decreased expression of MAP kinase phosphatase-1 (MKP-1) in HIV infection was associated with this heightened immune response. Our findings suggest that chronic HIV infection enhances the responsiveness of APCs to commensal lactobacilli, a mechanism that may partly contribute to chronic immune activation.


Subject(s)
Antigen-Presenting Cells/immunology , HIV Infections/blood , HIV Infections/immunology , Lactobacillus/immunology , Adult , Aged , CD36 Antigens/metabolism , Chronic Disease , Cohort Studies , Dendritic Cells/metabolism , Female , HIV Infections/enzymology , HIV Infections/microbiology , Humans , Immunity/immunology , Inflammation/blood , Inflammation/immunology , Inflammation/pathology , MAP Kinase Signaling System , Male , Middle Aged , Monocytes/metabolism , Phosphorylation , Receptors, Immunologic/metabolism , Toll-Like Receptor 2/metabolism , Young Adult , p38 Mitogen-Activated Protein Kinases/metabolism
10.
PLoS One ; 6(9): e24250, 2011.
Article in English | MEDLINE | ID: mdl-21935390

ABSTRACT

Costimulatory molecules play a central role in the development of cellular immunity. Understanding how costimulatory pathways can be directed to positively influence the immune response may be critical for the generation of an effective HIV vaccine. Here, we evaluated the ability of intravenous administration of a blocking monoclonal antibody (mAb) directed against the negative costimulatory molecule CTLA-4, and an agonist mAb directed against the positive costimulatory molecule 4-1BB, either alone or in combination, to augment intramuscular SIV DNA immunizations. We then tested the ability these of these responses to impact a high-dose SIVmac251 challenge. Following immunization, the groups infused with the anti-4-1BB mAb exhibited enhanced IFN-γ responses compared to the DNA vaccine only group. Interestingly, although CTLA-4 blockade alone did not enhance IFN-γ responses it did increase the proliferative capacity of the CD4(+) and CD8(+) T cells. The combination of both mAbs enhanced the magnitude of the polyfunctional CD8(+) T cell response. Following challenge, the group that received both mAbs exhibited a significant, ∼2.0 log, decrease in plasma viral load compared to the naïve group the included complete suppression of viral load in some animals. Furthermore, the use of the CTLA-4 blocking antibody resulted in significantly higher viral loads during chronic infection compared to animals that received the 4-1BB mAb, likely due to the higher CD4(+) T cell proliferative responses which were driven by this adjuvant following immunization. These novel studies show that these adjuvants induce differential modulation of immune responses, which have dramatically different consequences for control of SIV replication, suggesting important implications for HIV vaccine development.


Subject(s)
Macaca fascicularis/microbiology , Macaca fascicularis/virology , Simian Immunodeficiency Virus/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Vaccines, DNA/immunology , Adjuvants, Immunologic , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/metabolism , Female , Flow Cytometry , Male , Simian Immunodeficiency Virus/pathogenicity , Tumor Necrosis Factor Receptor Superfamily, Member 9/antagonists & inhibitors
11.
Curr Opin HIV AIDS ; 6(3): 221-7, 2011 May.
Article in English | MEDLINE | ID: mdl-21399494

ABSTRACT

PURPOSE OF REVIEW: We present current findings about two subsets of CD4+ T cells that play an important part in the initial host response to infection with the HIV type 1: those producing IL-17 (Th17 cells) and those with immunosuppressive function (CD25+FoxP3+ regulatory T cells or T-reg). The role of these cells in the control of viral infection and immune activation as well as in the prevention of immune deficiency in HIV-infected elite controllers will be examined. We will also discuss the use of the simian immunodeficiency virus (SIV)-infected macaque model of AIDS to study the interplay between these cells and lentiviral infection in vivo. RECENT FINDINGS: Study of Th17 cells in humans and nonhuman primates (NHPs) has shown that depletion of these cells is associated with the dissemination of microbial products from the infected gut, increased systemic immune activation, and disease progression. Most impressively, having a smaller Th17-cell compartment has been found to predict these outcomes. T-reg have been associated with the reduced antiviral T-cell responses but not with the suppression of generalized T cell activation. Both cell subsets influence innate immune responses and, in doing so, may shape the inflammatory milieu of the host at infection. SUMMARY: Interactions between Th17 cells, T-reg, and cells of the innate immune system influence the course of HIV and SIV infection from its earliest stages, even before the appearance of adaptive immunity. Such interactions may be pivotal for elite control over disease progression.


Subject(s)
HIV Infections/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , CD4-Positive T-Lymphocytes , HIV Long-Term Survivors , Humans , Immunity, Innate , Macaca
12.
J Infect Dis ; 203(1): 95-102, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21148501

ABSTRACT

The threat of a smallpox-based bioterrorist event or a human monkeypox outbreak has heightened the importance of new, safe vaccine approaches for these pathogens to complement older poxviral vaccine platforms. As poxviruses are large, complex viruses, they present technological challenges for simple recombinant vaccine development where a multicomponent mixtures of vaccine antigens are likely important in protection. We report that a synthetic, multivalent, highly concentrated, DNA vaccine delivered by a minimally invasive, novel skin electroporation microarray can drive polyvalent immunity in macaques, and offers protection from a highly pathogenic monkeypox challenge. Such a diverse, high-titer antibody response produced against 8 different DNA-encoded antigens delivered simultaneously in microvolumes has not been previously described. These studies represent a significant improvement in the efficiency of the DNA vaccine platform, resulting in immune responses that mimic live viral infections, and would likely have relevance for vaccine design against complex human and animal pathogens.


Subject(s)
Mpox (monkeypox)/prevention & control , Smallpox Vaccine/immunology , Vaccines, DNA/immunology , Animals , Antibodies, Viral/blood , Electroporation/methods , Humans , Macaca mulatta , Smallpox Vaccine/administration & dosage , Survival Analysis , Vaccines, DNA/administration & dosage
13.
Mol Ther ; 18(8): 1568-76, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20551910

ABSTRACT

DNA vaccines have undergone important enhancements in their design, formulation, and delivery process. Past literature supports that DNA vaccines are not as immunogenic in nonhuman primates as live vector systems. The most potent recombinant vector system for induction of cellular immune responses in macaques and humans is adenovirus serotype 5 (Ad5), an important benchmark for new vaccine development. Here, we performed a head-to-head evaluation of the Merck Ad5 SIV vaccine and an optimized electroporation (EP) delivered SIV DNA vaccine in macaques. Animals receiving the Ad5 vaccine were immunized three times, whereas the DNA-vaccinated animals were immunized up to four times based on optimized protocols. We observed significant differences in the quantity of IFNgamma responses by enzyme-linked immunosorbent spot (ELISpot), greater proliferative capacity of CD8(+) T cells, and increased polyfunctionality of both CD4(+) and CD8(+) T cells in the DNA-vaccinated group. Importantly, Ad5 immunizations failed to boost following the first immunization, whereas DNA responses were continually boosted with all four immunizations demonstrating a major advantage of these improved DNA vaccines. These optimized DNA vaccines induce very different immune phenotypes than traditional Ad5 vaccines, suggesting that they could play an important role in vaccine research and development.


Subject(s)
Adenoviridae/genetics , Genetic Vectors/genetics , SAIDS Vaccines/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Macaca mulatta , Plasmids/genetics , Vaccines, DNA/immunology
14.
Vaccine ; 26(25): 3112-20, 2008 Jun 13.
Article in English | MEDLINE | ID: mdl-18430495

ABSTRACT

DNA vaccines are a promising technology. Historically, however, the ability of DNA vaccines to induce high response rates and strong immune responses, especially antibody responses, in non-human primates and human clinical trials has proven suboptimal. Here, we performed a pilot study in rhesus macaques to evaluate whether we could improve the immunogenicity of DNA vaccines through the use of adjuvant technology and improved delivery systems. The study consisted of four groups of animals that received: DNA by intramuscular (IM) injection, DNA with plasmid-encoded IL-12 by IM injection, DNA by IM injection with in vivo electroporation (EP), and DNA with IL-12 by IM EP. Each group was immunized three times with optimized HIV gag and env constructs. Vaccine immunogenicity was assessed by IFNgamma ELISpot, CFSE proliferation, polyfunctional flow cytometry, and antibody ELISA. Similar to previous studies, use of IL-12 as an adjuvant increased the gag and env-specific cellular responses. The use of EP to enhance plasmid delivery resulted in dramatically higher cellular as well as humoral responses. Interestingly, the use of EP to administer the DNA and IL-12 adjuvant combination resulted in the induction of higher, more efficient responses such that a 10-fold increase in antigen-specific IFNgamma(+) cells compared to IM DNA immunization was observed after a single immunization. In addition to increases in the magnitude of IFNgamma production in the initial and memory responses, the combined approach resulted in enhancements in the proliferative capacity of antigen-specific CD8(+) T cells and the amount of polyfunctional cells capable of producing IL-2 and TNFalpha in addition to IFNgamma. These data suggest that adjuvant and improved delivery methods may be able to overcome previous immunogenicity limitations in DNA vaccine technology.


Subject(s)
Drug Delivery Systems , Electroporation , HIV Antibodies/blood , HIV Infections/immunology , HIV-1/immunology , Interleukin-12/pharmacology , Vaccines, DNA/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/genetics , AIDS Vaccines/immunology , Adjuvants, Immunologic , Animals , Drug Synergism , HIV Infections/prevention & control , HIV-1/genetics , Interleukin-12/immunology , Macaca , Pilot Projects , Vaccination , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
15.
Vaccine ; 26(3): 440-8, 2008 Jan 17.
Article in English | MEDLINE | ID: mdl-18082294

ABSTRACT

In an effort to improve DNA vaccine immune potency electroporation has emerged as a method of delivery of plasmids to target tissues. However, few studies have examined the use of this technology to deliver plasmid vaccines to the skin. Here we studied the effect of electroporation on DNA vaccine potency and gene delivery using skin as a target tissue in larger animal species. Using a pig model, we determined that high plasmid concentrations resulted in improved gene expression for plasmid GFP delivered by the intradermal/subcutaneous (ID/SQ) route. In a macaque model, we observed higher cellular and humoral responses to an HIV DNA vaccine, which included plasmid-encoded IL-12, with electroporation compared to ID/SQ injection alone. The induced responses were TH1 mediated. These results support that skin electroporation may have importance as an immunization approach in larger animal models.


Subject(s)
AIDS Vaccines/administration & dosage , Electroporation/methods , HIV Antibodies/blood , HIV Infections/prevention & control , Plasmids/administration & dosage , Th1 Cells/immunology , Vaccines, DNA/administration & dosage , AIDS Vaccines/genetics , AIDS Vaccines/immunology , Animals , Enzyme-Linked Immunosorbent Assay/methods , Female , HIV Infections/immunology , HIV Infections/virology , HIV-1/immunology , Injections, Intradermal , Injections, Subcutaneous , Interferon-gamma/biosynthesis , Macaca mulatta , Male , Plasmids/genetics , Plasmids/immunology , Swine , Vaccines, DNA/genetics , Vaccines, DNA/immunology
16.
J Med Primatol ; 37 Suppl 2: 62-8, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19187432

ABSTRACT

BACKGROUND: Previously, we examined the effects of in vivo CTLA-4 blockade using a fully human monoclonal antibody as a part of a DNA vaccination regimen in cynomolgus macaques (Macaca fascicularis). We observed that while the antibody had little effect on the IFN-gamma ELISpot response, CTLA-4 blockade enhanced antigen-specific cellular proliferation in both CD4(+) and CD8(+)T-cell compartments. METHODS: We examine the specific effects of CTLA-4 blockade on memory T-cell compartments following the third immunization and 10 months following a fourth immunization, during the memory phase of the immune response. RESULTS: CLTA-4 blockade enhanced CD4(+) and CD8(+) central memory (CD28(hi), CD95(hi)) T-cell responses as well as a short-lived CD8(+) effector (CD28(lo), CD95(hi)) T-cell response. CONCLUSIONS: These data suggest differing effects of CTLA-4 blockade on CD4(+) and CD8(+) T cells with implications on the clinical use of anti-CTLA-4 antibodies for enhancement of vaccine strategies or treatment of human disease.


Subject(s)
Antigens, CD/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Animals , Antibodies, Monoclonal , Antigens, CD/metabolism , CTLA-4 Antigen , Female , Flow Cytometry , Macaca fascicularis , Male , Models, Immunological , Vaccines, DNA/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...