Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Clin Infect Dis ; 73(1): e184-e190, 2021 07 01.
Article in English | MEDLINE | ID: mdl-32915202

ABSTRACT

BACKGROUND: Several chronic diseases accelerate biological aging. We investigated age acceleration and the association between peripheral blood DNA methylation (DNAm) and immune cell markers in patients chronically infected with the hepatitis B virus (HBV) or the hepatitis C virus (HCV) with and without human immunodeficiency virus (HIV) co-infection. METHODS: Age acceleration was measured as the difference between epigenetic age (Horvath clock) and chronological age. The immune marker model of age acceleration was developed using Elastic Net regression to select both the immune markers and their associated weights in the final linear model. RESULTS: Patients with chronic HBV (n = 51) had a significantly higher median epigenetic age compared to chronological age (age accelerated) (P < .001). In patients with chronic HCV infection (n = 63), age acceleration was associated with liver fibrosis as assessed by histology (P < .05), or presence of HIV co-infection (P < .05), but not HCV mono-infection. Age acceleration defined by immune markers was concordant with age acceleration by DNA methylation (correlation coefficient = .59 in HBV; P = .0025). One-year treatment of HBV patients with nucleoside therapy was associated with a modest reduction in age acceleration, as measured using the immune marker model (-.65 years, P = .018). CONCLUSION: Our findings suggest that patients with chronic viral hepatitis have accelerated epigenetic aging, that immune markers define biological age, and have the potential to assess the effects of therapeutic intervention on age acceleration.


Subject(s)
Coinfection , HIV Infections , Hepatitis B, Chronic , Hepatitis B , Hepatitis C , Aging , Biomarkers , DNA Methylation , HIV Infections/complications , Hepacivirus , Hepatitis B virus/genetics , Hepatitis B, Chronic/complications , Humans
2.
J Med Virol ; 93(6): 3752-3760, 2021 06.
Article in English | MEDLINE | ID: mdl-33150966

ABSTRACT

Combination regimens of direct-acting antiviral agents (DAAs) for chronic genotype 1 hepatitis C virus (HCV) infection given for 8 or 12 weeks have high cure rates. Shortened treatment durations that maintain high cure rates may lessen treatment barriers related to affordability and drug adherence. We enrolled 12 treatment-naïve adults with chronic genotype 1 HCV infection without cirrhosis in a single-center, open-label trial to receive 2 weeks of the highly potent and selective non-nucleoside inhibitor (NNI) CDI-31244 concurrent with 6 weeks of sofosbuvir/velpatasvir. The main efficacy endpoints were sustained virologic response at 12 (SVR12) and 24 (SVR24) weeks after treatment completion. In all patients, plasma HCV RNA levels rapidly decreased during the first 2 days of treatment and were below the lower limit of quantification by the end of the 6-week treatment period. Eight of 12 (67%) patients achieved both SVR12 and SVR24. Four patients had virological relapse at Week 10, 4 weeks after end of treatment. The most common adverse event was headache, occurring in five (42%) patients. Pharmacokinetic analysis showed no relevant drug interactions between CDI-31244, sofosbuvir, and velpatasvir. In this pilot study of short-duration combination therapy involving a novel NNI with a fixed-combination DAA, 8 of 12 treatment-naïve patients with chronic genotype 1 HCV infection without cirrhosis achieved virologic cure. Future trials might evaluate whether extending the NNI duration beyond 2 weeks with combination DAAs results in higher cure rates comparable with currently approved longer duration therapy.


Subject(s)
Antiviral Agents/therapeutic use , Carbamates/therapeutic use , Hepacivirus/drug effects , Hepatitis C, Chronic/drug therapy , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Sofosbuvir/therapeutic use , Adult , Antiviral Agents/adverse effects , Carbamates/adverse effects , Drug Therapy, Combination , Female , Genotype , Hepacivirus/genetics , Heterocyclic Compounds, 4 or More Rings/adverse effects , Humans , Male , Middle Aged , Pilot Projects , RNA, Viral/blood , Sofosbuvir/adverse effects , Sustained Virologic Response , Time Factors
3.
Int J Mol Sci ; 21(20)2020 Oct 10.
Article in English | MEDLINE | ID: mdl-33050486

ABSTRACT

Chronic HCV (CHC) infection is the only chronic viral infection for which curative treatments have been discovered. These direct acting antiviral (DAA) agents target specific steps in the viral replication cycle with remarkable efficacy and result in sustained virologic response (SVR) or cure in high (>95%) proportions of patients. These treatments became available 6-7 years ago and it is estimated that their real impact on HCV related morbidity, including outcomes such as cirrhosis and hepatocellular carcinoma (HCC), will not be known for the next decade or so. The immune system of a chronically infected patient is severely dysregulated and questions remain regarding the immune system's capacity in limiting liver pathology in a cured individual. Another important consequence of impaired immunity in patients cleared of HCV with DAA will be the inability to generate protective immunity against possible re-infection, necessitating retreatments or developing a prophylactic vaccine. Thus, the impact of viral clearance on restoring immune homeostasis is being investigated by many groups. Among the important questions that need to be answered are how much the immune system normalizes with cure, how long after viral clearance this recalibration occurs, what are the consequences of persisting immune defects for protection from re-infection in vulnerable populations, and does viral clearance reduce liver pathology and the risk of developing hepatocellular carcinoma in individuals cured with these agents. Here, we review the recent literature that describes the defects present in various lymphocyte populations in a CHC patient and their status after viral clearance using DAA treatments.


Subject(s)
Hepacivirus/immunology , Hepatitis C, Chronic/etiology , Hepatitis C, Chronic/metabolism , Host-Pathogen Interactions/immunology , Lymphocytes/immunology , Lymphocytes/metabolism , Adaptive Immunity , Animals , Antiviral Agents/therapeutic use , Disease Management , Disease Susceptibility , Hepatitis C, Chronic/drug therapy , Humans , Immunity, Innate , Liver/innervation , Liver/metabolism , Liver/pathology , Liver/virology , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Viral Load
4.
J Viral Hepat ; 26(9): 1105-1116, 2019 09.
Article in English | MEDLINE | ID: mdl-31074195

ABSTRACT

Immune dysfunction is a hallmark of chronic HCV infection and viral clearance with direct antivirals recover some of these immune defects. TCRVγ9Vδ2 T-cell dysfunction in treated HCV patients however is not well studied and was the subject of this investigation. Peripheral blood cells from patients who had achieved sustained virologic response (SVR) or those who had relapsed after interferon-free therapy were phenotyped using flow cytometry. Functional potential of Vγ9Vδ2 T cells was tested by measuring proliferation in response to aminobisphosphonate zoledronic acid, and cytotoxicity against HepG2 hepatoma cell line. TCR sequencing was performed to analyse impact of HCV infection on Vδ2 T-cell repertoire. Vγ9Vδ2 cells from patients were activated and therapy resulted in reduction of CD38 expression on these cells in SVR group. Relapsed patients had Vδ2 cells with persistently activated and terminally differentiated cytotoxic phenotype (CD38+ CD45RA+ CD27- CD107a+ ). Irrespective of outcome with therapy, majority of patients had persistently poor Vδ2 T-cell proliferative response to zoledronate along with lower expression of CD56, which identifies anti-tumour cytotoxic subset, relative to healthy controls. There was no association between the number of antigen reactive Vγ2-Jγ1.2 TCR rearrangements at baseline and levels of proliferation indicating nonresponse to zoledronate is not due to depletion of phosphoantigen responding chains. Thus, HCV infection results in circulating Vγ9Vδ2 T cells with a phenotype equipped for immediate effector function but poor cytokine response and expansion in response to antigen, a functional defect that may have implications for susceptibility for carcinogenesis despite HCV cure.


Subject(s)
Antiviral Agents/therapeutic use , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/immunology , Sustained Virologic Response , T-Lymphocytes/pathology , Adult , Aged , Cell Proliferation/drug effects , Clinical Trials as Topic , Cohort Studies , Cytokines/immunology , Cytotoxicity Tests, Immunologic , Female , Hep G2 Cells , Humans , Lymphocyte Activation , Male , Middle Aged , Phenotype , Receptors, Antigen, T-Cell, gamma-delta , T-Lymphocytes/immunology , Zoledronic Acid/pharmacology
5.
Immunogenetics ; 70(3): 179-183, 2018 03.
Article in English | MEDLINE | ID: mdl-28785825

ABSTRACT

Rhesus macaque is an important animal model for studies testing interventions like antibody therapeutics; as such knowledge of inter-individual variations in function of genes affecting antibody recycling is important for optimal experimental design. Neonatal Fc receptor (FcRn), a heterodimer composed of FCGRT and ß2-m chains, plays critical role in extending catabolic half-life of IgG. We studied genomic polymorphisms in rhesus macaque FcRn and asked if they are functional by assessing correlations with serum IgG or ß2-m levels. We tested 75 animals and report the presence of a VNTR polymorphism in promoter of FcRn as well as a single nucleotide polymorphism in the signal peptide of ß2-m. A VNTR minor allele was associated with lower levels of serum IgG. This polymorphism may account for inter-animal variation in antibody levels and has relevance for effective design of rhesus macaque studies investigating vaccine-induced antibody responses and passive immunizations.


Subject(s)
Antibodies/genetics , Histocompatibility Antigens Class I/genetics , Immunoglobulin G/immunology , Macaca mulatta/genetics , Receptors, Fc/genetics , Alleles , Animals , Antibodies/immunology , Histocompatibility Antigens Class I/immunology , Humans , Immunoglobulin G/blood , Immunoglobulin G/genetics , Macaca mulatta/immunology , Minisatellite Repeats/genetics , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Receptors, Fc/immunology
6.
J Med Virol ; 90(5): 936-941, 2018 05.
Article in English | MEDLINE | ID: mdl-29236302

ABSTRACT

Mixed cryoglobulinemic vasculitis is associated with monoclonal B cell expansion in patients with chronic hepatitis C (HCV) infection. B cell depletion therapy using rituximab, a CD20 monoclonal antibody, has been successful in achieving remission from symptomatic disease. This study investigated whether B cell depletion therapy has an impact on activation of HCV-specific T cell phenotype and function. Nineteen patients with Hepatitis C mixed cryoglobulinemic vasculitis were treated with 4 cycles of rituximab (375 mg/m2 ) and variables were measured 6 months after therapy. Using flow cytometry and Enzyme-Linked Immunospot assay, the number of activated and tissue-like B cells and number of T cells expressing Programmed cell death protein 1 (PD-1), T-cell immunoglobulin and mucin-domain containing-3 (TIM-3), and multiple cytokines were measured before and after rituximab therapy. B cell depletion therapy is associated with a significant (P < 0.0001) decline in peripheral T cells with exhaustive phenotype, from pre-therapy to post-therapy-of rituximab (mean ± standard error): CD4+ (16.9 ± 0.9% to 8.9 ± 1.0%) and CD8+ (6.8 ± 0.6% to 3.0 ± 0.5%) T cells expressing PD-1 and CD4+ (11.0 ± 1.0% to 6.1 ± 0.8%) and CD8+ (12.7 ± 0.7% to 6.4 ± 0.4%) T cells expressing TIM-3. In addition, there was a significantly higher percentage of peripheral CD8+ T cells responding to HCV peptide stimulation in vitro secreting IFN-γ (4.55 ± 0.3 to 9.6 ± 1.0 IFN-γ/106 PBMCs, P < 0.0001), and more than one cytokine (1.3 ± 0.1% to 3.8 ± 0.2%, P < 0.0001) after therapy compared to pre-therapy. B cell depletion therapy results in recovery of T cell exhaustion and function in patients with HCV cryoglobulinemic vasculitis.


Subject(s)
Cryoglobulinemia/complications , Hepacivirus/immunology , Hepatitis C, Chronic/immunology , Immunologic Factors/therapeutic use , Rituximab/therapeutic use , T-Lymphocytes/immunology , Vasculitis/drug therapy , B-Lymphocytes/immunology , Cytokines/analysis , Enzyme-Linked Immunospot Assay , Flow Cytometry , Hepatitis A Virus Cellular Receptor 2/analysis , Hepatitis C, Chronic/complications , Humans , Lymphocyte Subsets/immunology , Programmed Cell Death 1 Receptor/analysis , Treatment Outcome , Vasculitis/complications
7.
Virol J ; 10: 52, 2013 Feb 12.
Article in English | MEDLINE | ID: mdl-23402317

ABSTRACT

BACKGROUND: Lassa hemorrhagic fever (LHF) is a rodent-borne viral disease that can be fatal for human beings. In this study, an attenuated Lassa vaccine candidate, ML29, was tested in SIV-infected rhesus macaques for its ability to elicit immune responses without instigating signs pathognomonic for arenavirus disease. ML29 is a reassortant between Lassa and Mopeia viruses that causes a transient infection in non-human primates and confers sterilizing protection from lethal Lassa viral challenge. However, since the LHF endemic area of West Africa also has high HIV seroprevalence, it is important to determine whether vaccination could be safe in the context of HIV infection. RESULTS: SIV-infected and uninfected rhesus macaques were vaccinated with the ML29 virus and monitored for specific humoral and cellular immune responses, as well as for classical and non-classical signs of arenavirus disease. Classical disease signs included viremia, rash, respiratory distress, malaise, high liver enzyme levels, and virus invasion of the central nervous system. Non-classical signs, derived from profiling the blood transcriptome of virulent and non-virulent arenavirus infections, included increased expression of interferon-stimulated genes (ISG) and decreased expression of COX2, IL-1ß, coagulation intermediates and nuclear receptors needed for stress signaling. All vaccinated monkeys showed ML29-specific antibody responses and ML29-specific cell-mediated immunity. CONCLUSION: SIV-infected and uninfected rhesus macaques responded similarly to ML29 vaccination, and none developed chronic arenavirus infection. Importantly, none of the macaques developed signs, classical or non-classical, of arenavirus disease.


Subject(s)
Coinfection/immunology , HIV Infections/immunology , Lassa Fever/prevention & control , Lassa virus/immunology , Viral Vaccines/immunology , Animals , Antibodies, Viral/immunology , Coinfection/prevention & control , Coinfection/virology , HIV Infections/complications , HIV Infections/virology , Humans , Lassa Fever/complications , Lassa Fever/immunology , Lassa Fever/virology , Macaca mulatta , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Viral Vaccines/administration & dosage
8.
Front Immunol ; 14: 1146791, 2023.
Article in English | MEDLINE | ID: mdl-37180151

ABSTRACT

CD38, a nicotinamide adenine dinucleotide (NAD)+ glycohydrolase, is considered an activation marker of T lymphocytes in humans that is highly expressed during certain chronic viral infections. T cells constitute a heterogeneous population; however, the expression and function of CD38 has been poorly defined in distinct T cell compartments. We investigated the expression and function of CD38 in naïve and effector T cell subsets in the peripheral blood mononuclear cells (PBMCs) from healthy donors and people with HIV (PWH) using flow cytometry. Further, we examined the impact of CD38 expression on intracellular NAD+ levels, mitochondrial function, and intracellular cytokine production in response to virus-specific peptide stimulation (HIV Group specific antigen; Gag). Naïve T cells from healthy donors showed remarkably higher levels of CD38 expression than those of effector cells with concomitant reduced intracellular NAD+ levels, decreased mitochondrial membrane potential and lower metabolic activity. Blockade of CD38 by a small molecule inhibitor, 78c, increased metabolic function, mitochondrial mass and mitochondrial membrane potential in the naïve T lymphocytes. PWH exhibited similar frequencies of CD38+ cells in the T cell subsets. However, CD38 expression increased on Gag-specific IFN-γ and TNF-α producing cell compartments among effector T cells. 78c treatment resulted in reduced cytokine production, indicating its distinct expression and functional profile in different T cell subsets. In summary, in naïve cells high CD38 expression reflects lower metabolic activity, while in effector cells it preferentially contributes to immunopathogenesis by increasing inflammatory cytokine production. Thus, CD38 may be considered as a therapeutic target in chronic viral infections to reduce ongoing immune activation.


Subject(s)
HIV Infections , Virus Diseases , Humans , ADP-ribosyl Cyclase 1/metabolism , NAD/metabolism , Leukocytes, Mononuclear/metabolism , Cytokines
9.
Cytotherapy ; 14(2): 173-81, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22029653

ABSTRACT

BACKGROUND AIMS: Immunotherapy using γδ T cells capable of mediating antibody-dependent cellular cytotoxicity (ADCC) is a promising anti-human immunodeficiency virus (HIV) strategy. Approved aminobispohsphonate drugs, for example zoledronate (Zometa), stimulate γδ T cells in cancer patients, where they may promote direct tumor killing. Knowing that γδ T cells are modulated during HIV disease, documenting their responses and potential for controlling HIV is important. We investigated whether zoledronate/interleukin (IL)-2 could expand cytotoxic Vδ2 cells from HIV+ donors and whether these cells functioned in ADCC. METHODS: Peripheral blood mononuclear cells from uninfected controls and HIV+ individuals receiving anti-retroviral therapy were treated with isopentenyl pyrophosphate (IPP) or zoledronate plus IL-2 to expand the Vδ2+ subset. Immunophenotyping and functional analyzes (cytotoxicity or cytokine expression) allowed us to compare cell properties from individual donors and to compare the responses to each stimulating agent. RESULTS: Zoledronate stimulated a greater expansion of Vδ2 cells in HIV+ individuals compared with phosphoantigen IPP, and these cells expressed CD16. CD56 expression (a marker for cytotoxic cells) was lower on zoledronate-expanded cells, consistent with significantly lower cytotoxicity against the Daudi tumor cell line. Cells expanded with either zoledronate or IPP were active in ADCC, were similar in terms of interferon (IFN)-γ and tumor necrosis factor (TNF)-α expression, and degranulated in response to Fc receptor cross-linking. CONCLUSIONS: Zoledronate causes ex vivo expansion of Vδ2 cells from HIV+ individuals. Despite lower expression of CD56 and decreased direct cytotoxicity, these effectors were potent in ADCC. Zoledronate/IL-2- expanded cells have potential for immunotherapy to activate Vδ2 cells in HIV patients and enhance ADCC.


Subject(s)
Anti-Retroviral Agents/pharmacology , Antibody-Dependent Cell Cytotoxicity/immunology , Diphosphonates/pharmacology , HIV Infections/immunology , Imidazoles/pharmacology , Interleukin-2/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , CD56 Antigen/immunology , CD56 Antigen/metabolism , Cell Line, Tumor , GPI-Linked Proteins/immunology , GPI-Linked Proteins/metabolism , HIV/immunology , HIV Infections/therapy , Hemiterpenes/pharmacology , Humans , Immunophenotyping , Interleukin-2/metabolism , Lymphocyte Activation/drug effects , Organophosphorus Compounds/pharmacology , Receptors, IgG/immunology , Receptors, IgG/metabolism , Zoledronic Acid
10.
Blood ; 114(6): 1196-204, 2009 Aug 06.
Article in English | MEDLINE | ID: mdl-19498020

ABSTRACT

Immune deficiency viruses such as SIV in macaques or HIV-1 in human beings have evolved mechanisms to defeat host immunity that also impact the efficacy of vaccines. A key factor for vaccine protection is whether immune responses elicited by prior immunization remain at levels sufficient to limit disease progression once a host is exposed to the pathogen. One potential mechanism for escaping pre-existing immunity is to trigger death among antigen-activated cells. We tested whether FasL/CD178 is involved in destroying preexisting immunity. Rhesus macaques were immunized with recombinant vesicular stomatitis virus vaccine expressing SIV Gag to elicit cellular immune responses, then treated with antibody that neutralizes FasL and challenged with intravenous SIVmac251. Compared with animals injected with control antibody, anti-FasL-treated macaques had superior preservation of central memory CD4(+) and CD8(+) cells and decreased regulatory T cells in the blood. The CD4(+) and CD8(+) lymphocytes from treated animals responded better to SIV Gag compared with controls, evidenced by higher cell-mediated immune responses to viral antigens for at least 17 weeks after SIV challenge. Anti-FasL treatment during the initial stages of acute SIV infection preserved the T-cell compartment and sustained cell-mediated immunity to SIV.


Subject(s)
Antibodies/pharmacology , Fas Ligand Protein/immunology , Gene Products, gag/immunology , Immunologic Memory , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Animals , Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Fas Ligand Protein/antagonists & inhibitors , HIV-1/immunology , Humans , Immunity, Cellular , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/therapy , T-Lymphocytes, Regulatory/immunology
11.
Viruses ; 13(12)2021 11 30.
Article in English | MEDLINE | ID: mdl-34960669

ABSTRACT

TLR8 agonists have the potential for use as immunomodulatory components in therapeutic modalities for viral infections such as chronic HBV (CHB) and HIV. In this study, using peripheral blood samples from a phase 1a clinical trial, we examined the acute effects of a single oral administration of a selective TLR8 agonist on immune cell phenotypes. Administration of the TLR8 agonist selgantolimod (SLGN) in healthy individuals resulted in alteration in frequencies of peripheral blood monocytes, pDCs, mDCs and MAIT cells. Frequencies of mDCs and lymphoid cells significantly reduced after 8 h of SLGN administration, whereas pDC frequencies significantly increased, with changes possibly reflecting migration of different cell types between peripheral and tissue compartments in response to the agonist. Myeloid cell activation was evident by an upregulated expression of co-stimulatory molecules CD40 and CD86 accompanied by the production of IL-6 and IL-18 from these cells. Concomitantly, there was induction of the early activation marker CD69 on innate and adaptive lymphoid cells, including MAIT and NK cell subsets. Further, these activated lymphoid cells had enhanced expression of the effector molecules granzyme B and perforin. Microarray analysis of isolated lymphocytes and monocytes from baseline and post-SLGN treatment revealed changes in expression of genes involved in cellular response to cytokine stimulus, innate immune response, myeloid cell differentiation and antigen receptor-mediated signaling pathway. In a preliminary analysis of samples from CHB patients treated with selgantolimod, activation of innate and adaptive lymphocytes was evident. In conclusion, this first in-human study shows that selgantolimod administration in humans results in activation of multiple immune cell responses with antiviral potential.


Subject(s)
Hexanols/administration & dosage , Lymphocytes/drug effects , Pyrimidines/administration & dosage , Toll-Like Receptor 8/agonists , Adaptive Immunity/drug effects , Administration, Oral , Granzymes/genetics , Granzymes/immunology , Humans , Immunity, Innate/drug effects , Interleukin-18/genetics , Interleukin-18/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lymphocytes/immunology , Mucosal-Associated Invariant T Cells/drug effects , Mucosal-Associated Invariant T Cells/immunology , Toll-Like Receptor 8/genetics , Toll-Like Receptor 8/immunology
12.
Front Immunol ; 12: 735913, 2021.
Article in English | MEDLINE | ID: mdl-34512670

ABSTRACT

Identifying signaling pathways that induce B cell response can aid functional cure strategies for chronic hepatitis B infection (CHB). TLR8 activation with ssRNA was shown to enhance follicular helper T cell (TFH) function leading to improved B cell responses in vitro. We investigated whether this mechanism can rescue an exhausted immune response in CHB infection. Effect of TLR8 agonism on supporting cytokines and TFH and B cells were evaluated using ex vivo and in vitro assays. The ability of an oral TLR8 agonist to promote TFH and B cell response was tested in samples from phase 1b clinical trial. TLR8 agonism induced TFH polarizing cytokine IL-12 in monocytes. Treatment of peripheral blood mononuclear cells (PBMCs) from CHB patients with TLR8 agonists induced cytokine IL-21 by TFH cells with enhanced IL-21+BCL-6+ and ICOS+BCL-6+ co-expression. Mechanistically, incubation of isolated naïve CD4+ T cells with TLR8 triggered monocytes resulted in their differentiation into IL-21+ICOS+BCL-6+ TFH in an IL-12 dependent manner. Furthermore, co-culture of these IL-21 producing TFH with autologous naïve B cells led to enhanced memory (CD19+CD27+) and plasma B cell generation (CD19+CD27++CD38+) and IgG production. Importantly, in TFH from CHB patients treated with an oral TLR8 agonist, HBsAg-specific BCL-6, ICOS, IL-21 and CD40L expression and rescue of defective activation induced marker (AIM) response along with partial restoration of HBsAg-specific B cell ELISPOT response was evident. TLR8 agonism can thus enhance HBV-specific B cell responses in CHB patients by improving monocyte-mediated TFH function and may play a role in achieving HBV functional cure.


Subject(s)
Antiviral Agents/therapeutic use , B-Lymphocytes/drug effects , Hepatitis B Surface Antigens/immunology , Hepatitis B virus/immunology , Hepatitis B, Chronic/drug therapy , Hexanols/therapeutic use , Pyrimidines/therapeutic use , T Follicular Helper Cells/drug effects , Toll-Like Receptor 8/agonists , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/virology , CD40 Ligand/metabolism , Case-Control Studies , Cells, Cultured , Enzyme-Linked Immunospot Assay , Hepatitis B virus/pathogenicity , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/metabolism , Hepatitis B, Chronic/virology , Host-Pathogen Interactions , Humans , Inducible T-Cell Co-Stimulator Protein/metabolism , Interleukins/metabolism , Proto-Oncogene Proteins c-bcl-6/metabolism , Signal Transduction , T Follicular Helper Cells/immunology , T Follicular Helper Cells/metabolism , T Follicular Helper Cells/virology , Toll-Like Receptor 8/metabolism , Treatment Outcome
13.
Front Immunol ; 11: 569611, 2020.
Article in English | MEDLINE | ID: mdl-33133083

ABSTRACT

COVID-19 caused by SARS CoV2 emerged in China at the end of 2019 and soon become a pandemic. Since the virus is novel, pre-existing CoV2-specific immunity is not expected to exist in humans, although studies have shown presence of CoV2 cross-reactive T cells in unexposed individuals. Lack of effective immunity in most individuals along with high infectiousness of the virus has resulted in massive global public health emergency. Intense efforts are on to study viral pathogenesis and immune response to help guide prophylactic and therapeutic interventions as well as epidemiological assessments like transmission modeling. To develop an effective vaccine or biologic therapeutic, it is critical to understand the immune correlates of COVID-19 control. At the same time, whether immunity in recovered individuals is effective for preventing re-infection will be important for informing interventions like social distancing. Key questions that are being investigated regarding immune response in COVID-19 which will help these efforts include, investigations of immune response that distinguishes patients with severe versus mild infection or those that recover relative to those that succumb, durability of immunity in recovered patients and relevance of developed immunity in a cured patient for protection against re-infection as well as value of convalescent plasma from recovered patients as a potential therapeutic modality. This is a broad and rapidly evolving area and multiple reports on status of innate and adaptive immunity against SARS-CoV2 are emerging on a daily basis. While many questions remain unanswered for now, the purpose of this focused review is to summarize the current understanding regarding immune correlates of COVID-19 severity and resolution in order to assist researchers in the field to pursue new directions in prevention and control.


Subject(s)
Coronavirus Infections/immunology , Coronavirus Infections/prevention & control , Pandemics/prevention & control , Pneumonia, Viral/immunology , Pneumonia, Viral/prevention & control , Antibodies, Viral/immunology , B-Lymphocytes/immunology , COVID-19 , COVID-19 Vaccines , Coronavirus Infections/drug therapy , Cytokines/immunology , Humans , Inflammation Mediators/immunology , Patient Acuity , Recurrence , T-Lymphocytes/immunology , Viral Vaccines/immunology , COVID-19 Drug Treatment
14.
Front Immunol ; 11: 599648, 2020.
Article in English | MEDLINE | ID: mdl-33584666

ABSTRACT

Chronic Hepatitis B (CHB) affects over 350 million people worldwide. Current treatment does result in reduced complications; however, a cure (development of antibodies to the S antigen) is not achieved, requiring life-long therapy. Humoral responses contribute to viral elimination by secreting neutralizing antibodies; though, effective induction of humoral immunity require CD4T cell differentiation into T follicular helper (TFH) cells that support B cell response through interleukin-21 (IL-21). In CHB, mechanism of TFH-B interactions is seldom described. During CHB, TFH cells are defective in producing IL-21 in response to hepatitis B surface antigen (HBsAg). However, regardless of low IL-21, TFH cells efficiently support B cell responses by producing interleukin-27 (IL-27), which directs the formation of plasmablasts and plasma cells from memory and naïve B cells by enhancing B lymphocyte-induced maturation protein-1. IL-27 not only improved total antibody production but HBsAg-specific IgG and IgM secretion that are essential for viral clearance. Importantly, IL-27+TFH cells were significantly associated with HBV DNA reduction. Therefore, these findings imply a novel mechanism of TFH mediated B cell help in CHB and suggest that IL-27 effectively compensate the function of IL-21 by supporting TFH-B cell function, required for protective antibody response and may contribute to viral clearance by providing potential target for achieving a functional cure.


Subject(s)
B-Lymphocytes/immunology , Hepatitis B virus/immunology , Hepatitis B, Chronic/immunology , Interleukins/deficiency , Interleukins/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adult , B-Lymphocytes/pathology , Female , Hepatitis B Surface Antigens/immunology , Hepatitis B, Chronic/pathology , Humans , Immunologic Memory , Male , Middle Aged , T-Lymphocytes, Helper-Inducer/pathology
15.
Clin Cardiol ; 43(3): 222-234, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31785111

ABSTRACT

Hepatitis C virus (HCV) infects 180 million people worldwide and over 4 million people in the United States. HCV infection is a major cause of chronic liver disease and is recognized as a risk factor for clinical cardiovascular disease (CVD). Many studies have shown increased prevalence of cardiac and inflammatory biomarkers in patients with chronic HCV infection (CHC), and though these markers may be used to risk stratify people for cardiac disease in the general population their role in the HCV population is unknown. Patients with CHC have elevated cardiac and inflammatory biomarkers compared to noninfected controls which may play a role in CVD risk stratification. We undertook a systematic review of inflammatory and cardiac biomarkers in people with HCV infection with a focus on the effect of CHC on serum levels of these markers and their utility as predictors of CVD in this population. Medline, EMBASE, and Cochrane databases were searched for relevant articles until June 2019. A total of 2430 results were reviewed with 115 studies included. Our review revealed that HCV infection significantly alters serum levels of markers of inflammation, endothelial function, and cardiac dysfunction prior to HCV treatment, and some of which may change in response to HCV therapy. Current risk stratification tools for development of CVD in the general population may not account for the increased inflammatory markers that appear to be elevated among HCV-infected patients contributing to increased CVD risk.


Subject(s)
Cardiovascular Diseases/blood , Hepatitis C, Chronic/blood , Inflammation Mediators/blood , Biomarkers/blood , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/epidemiology , Heart Disease Risk Factors , Hepatitis C, Chronic/diagnosis , Hepatitis C, Chronic/epidemiology , Humans , Prognosis , Risk Assessment
16.
17.
Sci Rep ; 10(1): 1835, 2020 02 04.
Article in English | MEDLINE | ID: mdl-32020034

ABSTRACT

Chronic hepatitis B (CHB) infection functional cure is defined as sustained loss of HBsAg and several therapeutic strategies are in clinical development designed to pharmacologically reduce serum HBsAg, break immune tolerance, and increase functional cure rates. However, little is known about pre-treatment HBsAg levels as an indicator of HBV immune potential. Here, we compared the phenotypes and HBV-specific response of lymphocytes in CHB patients stratified by serum HBsAg levels <500 (HBslo) or >50,000 IU/ml (HBshi) using immunological assays (flow cytometry, ICS, ELISPOT). HBshi patients had significantly higher expression of inhibitory PD-1 on CD4+ T cells, particularly among TEMRA subset, and higher FcRL5 expression on B cells. Upon HBcAg(core) or HBsAg(env)-stimulation, 85% and 60% of HBslo patients had IFNγ+TNFα+ and IFNγ+ IL2+ CD4+ T cell responses respectively, in comparison to 33% and 13% of HBshi patients. Checkpoint blockade with αPD-1 improved HBV-specific CD4+ T cell function only in HBslo patients. HBsAg-specific antibody-secreting cells (ASCs) response was not different between these groups, yet αPD-1 treatment resulted in significantly higher fold change in ASCs among patients with HBsAg <100 IU/ml compared to patients with HBsAg >5,000 IU/ml. Thus, serum HBsAg correlates with inhibitory receptor expression, HBV-specific CD4+ T cell responses, and augmentation by checkpoint blockade.


Subject(s)
B-Lymphocytes/immunology , Hepatitis B Surface Antigens/blood , Hepatitis B virus/immunology , Hepatitis B, Chronic/immunology , T-Lymphocytes/immunology , Biomarkers/blood , Flow Cytometry , Hepatitis B, Chronic/blood , Humans , Programmed Cell Death 1 Receptor/metabolism
18.
Antivir Ther ; 25(3): 171-180, 2020.
Article in English | MEDLINE | ID: mdl-32667286

ABSTRACT

BACKGROUND: Selgantolimod is a novel oral, selective Toll-like receptor 8 (TLR8) agonist in development for the treatment of chronic hepatitis B (CHB). TLR8 is an endosomal innate immune receptor and a target for treatment of viral infections. This first-in-human study investigated the safety, tolerability, pharmacokinetics (PK) and pharmacodynamics (PD) of selgantolimod in healthy volunteers. METHODS: Of 71 subjects enrolled, 59 received a single dose of selgantolimod (0.5, 1.5, 3 or 5 mg) or placebo, and 12 were evaluated for food effect. Safety, PK and PD activity by induction of cytokines, chemokines and acute phase proteins were assessed. PK/PD analyses were conducted. RESULTS: Single doses of 0.5-5 mg were generally safe. No serious adverse events (AEs) or AEs leading to discontinuation were reported, and most were Grade 1 in severity. Selgantolimod displayed rapid absorption and dose-proportional PK and PD activity. Food had minimal effect on PK but resulted in diminished PD activity. In PK/PD analyses, near-saturation of induction for most evaluated biomarkers occurred at the 5-mg dose. CONCLUSIONS: Single doses of up to 5 mg selgantolimod were safe and induced dose-dependent PD responses. These data support evaluation of selgantolimod in combination with other agents in future clinical studies of CHB. Australian New Zealand Clinical Trials Registration: ACTRN12616001646437.


Subject(s)
Antiviral Agents/pharmacology , Hexanols/pharmacology , Pyrimidines/pharmacology , Toll-Like Receptor 8/agonists , Administration, Oral , Adult , Antiviral Agents/administration & dosage , Antiviral Agents/adverse effects , Antiviral Agents/pharmacokinetics , Chemokines/blood , Dose-Response Relationship, Drug , Female , Hepatitis B, Chronic/drug therapy , Hexanols/administration & dosage , Hexanols/adverse effects , Hexanols/pharmacokinetics , Humans , Interleukin 1 Receptor Antagonist Protein/blood , Interleukin-12/blood , Male , Pyrimidines/administration & dosage , Pyrimidines/adverse effects , Pyrimidines/pharmacokinetics , Young Adult
19.
Retrovirology ; 6: 91, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19832988

ABSTRACT

Human immunodeficiency virus disease involves progressive destruction of host immunity leading to opportunistic infections and increased rates for malignancies. Both depletion in immune cell numbers as well as defects in their effector functions are responsible for this immunodeficiency The broad impact of HIV reflects a similarly broad pattern of cell depletion including subsets that do not express viral receptors or support viral replication. Indirect cell killing, the destruction of uninfected cells, is due partly to activation of the Fas/FasL system for cell death. This death-signaling pathway is induced during HIV disease and contributes significantly to viral pathogenesis and disease.


Subject(s)
Cell Death , Fas Ligand Protein/physiology , HIV Infections/virology , HIV/pathogenicity , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/pathogenicity , fas Receptor/physiology , Animals , Haplorhini , Humans , Models, Biological
20.
Front Immunol ; 10: 1470, 2019.
Article in English | MEDLINE | ID: mdl-31316516

ABSTRACT

Direct acting antiviral (DAA) regimens of 12 weeks result in HCV clearance in vast majority of patients across genotypes. We previously demonstrated an ultra-short regimen of 4 weeks DAA cleared HCV in a subset of patients. Here, we hypothesized that individual level of antiviral immunity differentially influenced viral clearance and investigated biomarkers of a successful response. Cohorts of HCV patients treated for 4 weeks with DAA therapy who either achieved sustained virologic response (SVR) or relapsed were compared at baseline and at end of therapy (EOT) for immune cell phenotypes and HCV specific immunity. Higher levels of PD-1+ CD8+ and CD4+ T lymphocytes co-expressing inhibitory receptors (IR) were present at baseline and at EOT in HCV patients who eventually achieved SVR compared with those who relpased. HCV specific CD8+ T cells were predominantly contained within these IR expressing PD-1+ subsets. Patients in the SVR group had significantly higher CD8+ T cell degranulation in response to HCV peptides at baseline and higher levels of cytokine producing T cells at EOT time-point, relative to those who relapsed. In ex vivo cultures, PD-1+CD160+ CD8+ T cells had higher HCV specific degranulation and PD-1+2B4+ CD8+ T cells had higher cytokine expression (IFNγ+TNFα+ or IFNγ+CD107a+) compared with single or no IR expressing subsets, indicating higher virus specific functional capacity of these subsets. Receiver operating characteristics curve (ROC) for baseline circulating frequencies of PD-1+CD160+, PD-1+Tim-3+ CD8+ T cells and PD-1+CD160+, PD-1+Blimp-1+, PD-1-CTLA4+ CD4+ T cells respectively, had associated C-statistics of 0.8214 and 0.9451 for discriminatin of patients who successfully cleared HCV with 4 weeks treatment. Thus, PD-1+ virus-specific CD8+ T cell subsets with cytotoxic capacity are present in a subset of chronic HCV infected individuals that associate with ability to achieve SVR, indicating role of immunity in DAA mediated viral clearance with short duration therapy.


Subject(s)
Antiviral Agents/therapeutic use , Hepacivirus/drug effects , Hepatitis C, Chronic/drug therapy , Programmed Cell Death 1 Receptor/metabolism , Receptors, Immunologic/metabolism , T-Lymphocytes/drug effects , Adult , Benzimidazoles/therapeutic use , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cytokines/metabolism , Drug Therapy, Combination , Female , Fluorenes/therapeutic use , Furans/therapeutic use , Hepacivirus/physiology , Hepatitis C, Chronic/metabolism , Hepatitis C, Chronic/virology , Humans , Male , Middle Aged , Pilot Projects , Quinolines/therapeutic use , Sofosbuvir/therapeutic use , Sustained Virologic Response , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Thiophenes/therapeutic use , Viral Load/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL