Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Nicotine Tob Res ; 26(3): 289-297, 2024 Feb 22.
Article in English | MEDLINE | ID: mdl-37462724

ABSTRACT

INTRODUCTION: Roll-your-own (RYO) tobacco is a popular choice in Australia, with some people who smoke finding these products more attractive than factory-made cigarettes (FMC). Differences in visual and tactile properties and in the feel and taste of the smoke may contribute to this attractiveness. These differences may be driven by variation in tobacco constituents and wrapping paper permeability. However, to date, there has been no comparison of RYO and FMC products on the Australian market. AIMS AND METHODS: Chemical constituents, pH, flavorants, and paper permeability were compared in unburned RYO tobacco and tobacco from FMC. RYO and FMC products from matched brands were compared, as were products from the most popular FMC and RYO brands on the Australian market in 2018. RESULTS: RYO tobacco had higher moisture and humectant content (glycerol and propylene glycol) than FMC tobacco. RYO tobacco also had higher amounts of total and reducing sugars and lower nicotine when comparing the most popular brands. RYO papers were less permeable than FMC papers. Both RYO and FMC tobacco contained many chemicals identified as flavorants, including fourteen with known potential health risks. For most measured constituents and flavorants, RYO tobaccos had more in common with other RYO than FMC, with the commonalities remaining even when matched brands were compared. CONCLUSIONS: Higher levels of moisture, humectants, and sugars in Australian RYO tobacco compared to FMC may be increasing attractiveness of RYO by reducing the harsh taste of the smoke and increasing the moist feel of the tobacco. IMPLICATIONS: While price is the main factor driving the use of RYO tobacco, some people who smoke find these products more attractive. This study has shown that Australian RYO tobacco contains higher amounts of glycerol, propylene glycol, and sugars than FMC. These chemicals may be improving the taste of the tobacco, as well as creating a moist feel that is falsely perceived as indicating that the tobacco is "fresh" and "less chemically." Ironically, it may be that higher amounts of some added chemicals in RYO contribute to false perceptions of a more natural and less harmful product.


Subject(s)
Glycerol , Tobacco Products , Humans , Australia , Sugars , Propylene Glycols
2.
Hum Vaccin Immunother ; 13(11): 2726-2737, 2017 11 02.
Article in English | MEDLINE | ID: mdl-28949787

ABSTRACT

While broadly neutralizing antibodies (bnAbs) are a promising preventative and therapeutic tool for HIV infection, production is difficult and expensive. Production of antibody-like fragments in bacterial cytoplasm provides a cheaper alternative. This work explored the transplantation of the complementarity determining regions of the anti-HIV bnAbs PGT121 and 10E8 onto a single-chain variable fragment (scFv) scaffold, previously discovered through a novel screening platform. The scaffolded 10E8 scFv, but not the scaffolded PGT121 scFv, was soluble in bacterial cytoplasm, enabling efficient production in bacteria. Three additional multimeric constructs employing the scaffolded 10E8 scFv were also generated and soluble versions produced in bacteria. However, the constructs were found to have substantially lost anti-HIV binding function and had completely abrogated neutralizing activity. Overall, while this study provides a proof-of-concept for anti-HIV bnAb construct production in bacterial cytoplasm, future refinement of these technologies will be required to realize the goal of producing inexpensive and effective bnAb-like tools for the control of HIV.


Subject(s)
Antibodies, Neutralizing/therapeutic use , Bacteria/immunology , HIV Antibodies/therapeutic use , HIV Infections/therapy , Single-Chain Antibodies/immunology , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/biosynthesis , Escherichia coli/genetics , Escherichia coli/immunology , HIV Antibodies/administration & dosage , HIV Antibodies/biosynthesis , HIV Antibodies/chemistry , HIV Infections/immunology , HIV Infections/prevention & control , HIV Infections/virology , HIV-1/immunology , Humans , Neutralization Tests , Proof of Concept Study , Single-Chain Antibodies/biosynthesis
3.
Virology ; 492: 1-10, 2016 May.
Article in English | MEDLINE | ID: mdl-26896929

ABSTRACT

The low fidelity of HIV replication facilitates immune and drug escape. Some reverse transcriptase (RT) inhibitor drug-resistance mutations increase RT fidelity in biochemical assays but their effect during viral replication is unclear. We investigated the effect of RT mutations K65R, Q151N and V148I on SIV replication and fidelity in vitro, along with SIV replication in pigtailed macaques. SIVmac239-K65R and SIVmac239-V148I viruses had reduced replication capacity compared to wild-type SIVmac239. Direct virus competition assays demonstrated a rank order of wild-type>K65R>V148I mutants in terms of viral fitness. In single round in vitro-replication assays, SIVmac239-K65R demonstrated significantly higher fidelity than wild-type, and rapidly reverted to wild-type following infection of macaques. In contrast, SIVmac239-Q151N was replication incompetent in vitro and in pigtailed macaques. Thus, we showed that RT mutants, and specifically the common K65R drug-resistance mutation, had impaired replication capacity and higher fidelity. These results have implications for the pathogenesis of drug-resistant HIV.


Subject(s)
Drug Resistance, Viral/genetics , RNA-Directed DNA Polymerase/genetics , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Viral Proteins/genetics , Virus Replication/genetics , Animals , Base Sequence , Cell Line , HEK293 Cells , Humans , Macaca nemestrina , Male , Molecular Sequence Data , Mutation , Plasmids/chemistry , Plasmids/immunology , RNA-Directed DNA Polymerase/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , T-Lymphocytes/immunology , T-Lymphocytes/virology , Viral Load , Viral Proteins/immunology
4.
Biotechnol J ; 10(5): 783-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25712138

ABSTRACT

Conventional antibody surface display requires fusion protein export through at least one cellular membrane, constraining the yield and occasioning difficulties in achieving scaled production. To circumvent this limitation, we developed a novel cytoplasmic display platform, Retained Display (ReD), and used it to screen for human scFv frameworks that are highly soluble and stable in the bacterial cytoplasm. ReD, based on the retention of high-molecular weight complexes within detergent-permeabilized Escherichia coli, enabled presentation of exogenous targets to antibodies that were expressed and folded in the cytoplasm. All human λ and κ light chain family genes were expressed as IGHV3-23 fusions. Members of the λ subfamilies 1, 3 and 6 were soluble cytoplasmic partners of IGHV3-23. Contrary to previous in vivo screens for soluble reduced scFvs, the pairings identified by ReD were identical to the human germline sequences for the framework, CDR1 and CDR2 regions. Using the most soluble scFv scaffold identified, we demonstrated tolerance to CDR3 diversification and isolated a binding scFv to an exogenous protein target. This screening system has the potential to rapidly produce antibodies to target threats such as emerging infectious diseases and bioterror agents.


Subject(s)
Cytoplasm/metabolism , Escherichia coli/genetics , Single-Chain Antibodies/biosynthesis , Autoantigens/genetics , Cloning, Molecular , Escherichia coli/metabolism , Humans , Nerve Tissue Proteins/genetics , Single-Chain Antibodies/genetics
5.
J Leukoc Biol ; 97(3): 599-609, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25605872

ABSTRACT

The testis is a site of immune privilege in rodents, and there is evidence that T cell responses are also suppressed in the primate testis. Local immunosuppression is a potential mechanism for HIV persistence in tissue reservoirs that few studies have examined. The response of the pig-tailed macaque testis to SIVmac239 infection was characterized to test this possibility. Testes were surgically removed during early-chronic (10 wk) and late-chronic (24-30 wk) SIV infection in 4 animals and compared with those from 7 uninfected animals. SIV infection caused only minor disruption to the seminiferous epithelium without marked evidence of inflammation or consistent changes in total intratesticular leukocyte numbers. Infection also led to an increase in the relative proportion of testicular effector memory CD8(+) T cell numbers and a corresponding reduction in central memory CD4(+) T cells. A decrease in the relative proportion of resident-type CD163(+) macrophages and DCs was also observed. SIV-specific CD8(+) T cells were detectable in the testis, 10-11 wk after infection by staining with SIV Gag-specific or Tat-specific MHC-I tetramers. However, testicular CD8(+) T cells from the infected animals had suppressed cytokine responses to mitogen activation. These results support the possibility that local immunosuppression in the testis may be restricting the ability of T cells to respond to SIV or HIV infection. Local immunosuppression in the testis may be an underexplored mechanism allowing HIV persistence.


Subject(s)
Immunity , Macaca nemestrina/immunology , Macaca nemestrina/virology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/immunology , Testis/immunology , Animals , CD4 Lymphocyte Count , CD8-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Dendritic Cells/pathology , Granulocytes/pathology , HEK293 Cells , Humans , Killer Cells, Natural/pathology , Leukocyte Common Antigens/metabolism , Macrophages/pathology , Male , Phenotype , Seminiferous Tubules/immunology , Seminiferous Tubules/pathology , Seminiferous Tubules/virology , Simian Acquired Immunodeficiency Syndrome/blood , Testis/pathology , Testis/virology
6.
J Infect Dis ; 211(4): 529-38, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25170105

ABSTRACT

BACKGROUND: Combination antiretroviral therapy (cART) effectively controls human immunodeficiency virus (HIV) infection but does not eliminate HIV, and lifelong treatment is therefore required. HIV-specific cytotoxic T lymphocyte (CTL) responses decline following cART initiation. Alterations in other HIV-specific immune responses that may assist in eliminating latent HIV infection, specifically antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent phagocytosis (ADP), are unclear. METHODS: A cohort of 49 cART-naive HIV-infected subjects from Thailand (mean baseline CD4 count, 188 cells/µL; mean viral load, 5.4 log10 copies/mL) was followed for 96 weeks after initiating cART. ADCC and ADP assays were performed using serum samples obtained at baseline and after 96 weeks of cART. RESULTS: A 35% reduction in HIV type 1 envelope (Env)-specific ADCC-mediated killing of target cells (P<.001) was observed after 96 weeks of cART. This was corroborated by a significant reduction in the ability of Env-specific ADCC antibodies to activate natural killer cells (P<.001). Significantly reduced ADP was also observed after 96 weeks of cART (P=.018). CONCLUSIONS: This longitudinal study showed that cART resulted in significant reductions of HIV-specific effector antibody responses, including ADCC and ADP. Therapeutic vaccines or other immunomodulatory approaches may be required to improve antibody-mediated control of HIV during cART.


Subject(s)
Anti-Retroviral Agents/therapeutic use , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/immunology , HIV Infections/drug therapy , HIV Infections/immunology , Anti-Retroviral Agents/administration & dosage , Anti-Retroviral Agents/adverse effects , Cohort Studies , HIV Infections/epidemiology , HIV-1/immunology , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Killer Cells, Natural/immunology , Longitudinal Studies , Monocytes/immunology , Viral Load , env Gene Products, Human Immunodeficiency Virus/immunology
7.
AIDS ; 28(13): 1859-70, 2014 Aug 24.
Article in English | MEDLINE | ID: mdl-24937308

ABSTRACT

OBJECTIVE: The objective of this study is to determine the breadth of HIV-1 Env-specific antibody-dependent cellular cytotoxicity (ADCC) in HIV controllers and HIV progressors with a view to design globally relevant HIV vaccines. DESIGN: The breadth of ADCC towards four major HIV-1 Env subtypes was measured in vitro for 11 HIV controllers and 11 HIV progressors. METHODS: Plasma from 11 HIV controllers (including long-term slow progressors, viremic controllers, elite controller and posttreatment controller) and 11 HIV progressors, mostly infected with HIV-1 subtype B, was analysed for ADCC responses. ADCC assays were performed against 10 HIV-1 gp120 and 8 gp140 proteins from four major HIV-1 subtypes (A, B, C and E) and 3 glycosylation-mutant gp140 proteins. RESULTS: ADCC-mediated natural killer cell activation was significantly broader (P = 0.02) and of higher magnitude (P < 0.001) in HIV controllers than in HIV progressors. HIV controllers also showed significantly higher magnitude of ADCC-mediated killing of Env-coated target cells than HIV progressors to both HIV-1 subtype B and the heterologous subtype E gp140 (P = 0.001). We found good ADCC reactivity to subtype B and E Envs, less cross-reactivity to subtype A and minimal cross-reactivity to subtype C Envs. Glycosylation-dependent ADCC epitopes comprise a significant proportion of the total Env-specific ADCC response, as evident from the reduction in ADCC to nonglycosylated form of HIV-1 gp140 (P = 0.004). CONCLUSION: HIV controllers have robust ADCC responses that recognize a broad range of HIV-1 Env. Glycosylation of Env was found to be important for recognition of ADCC epitopes. Identifying conserved ADCC epitopes will assist in designing globally relevant ADCC-based HIV vaccines.


Subject(s)
AIDS Vaccines/immunology , HIV Antibodies/immunology , HIV Infections/immunology , HIV-1/immunology , T-Lymphocytes, Cytotoxic/immunology , env Gene Products, Human Immunodeficiency Virus/immunology , Adult , Epitopes/immunology , Female , Genotype , HIV-1/classification , HIV-1/genetics , Humans , Male , Middle Aged , Young Adult , env Gene Products, Human Immunodeficiency Virus/genetics
8.
J Infect Dis ; 210(11): 1811-22, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-24916185

ABSTRACT

BACKGROUND: Intravenous immunoglobulin (IVIG) is a purified pool of human antibodies from thousands of donors that is used to prevent or treat primary immune deficiency, several infectious diseases, and autoimmune diseases. The antibodies that mediate antibody-dependent cellular cytotoxicity (ADCC) against heterologous influenza strains may be present in IVIG preparations. METHODS: We tested 8 IVIG preparations prior to the 2009 H1N1 swine-origin influenza pandemic and 10 IVIG preparations made after 2010 for their ability to mediate influenza-specific ADCC. RESULTS: ADCC mediating antibodies to A(H1N1)pdm09 hemagglutinin (HA) and neuraminidase (NA) were detected in IVIG preparations prior to the 2009-H1N1 pandemic. The HA-specific ADCC targeted both the HA1 and HA2 regions of A(H1N1)pdm09 HA and was capable of recognizing a broad range of HA proteins including those from recent avian influenza strains A(H5N1) and A(H7N9). The low but detectable ADCC recognition of A(H7N9) was likely due to rare individuals in the population contributing cross-reactive antibodies to IVIG. CONCLUSIONS: IVIG preparations contain broadly cross-reactive ADCC mediating antibodies. IVIG may provide at least some level of protection for individuals at high risk of severe influenza disease, especially during influenza pandemics prior to the development of effective vaccines.


Subject(s)
Antibodies, Viral/immunology , Antibody-Dependent Cell Cytotoxicity/immunology , Cross Reactions/immunology , Immunoglobulins, Intravenous/therapeutic use , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/immunology , Influenza, Human/therapy , Adult , Aged , Aged, 80 and over , Antibodies, Viral/blood , Female , Hemagglutination Tests , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Influenza A Virus, H7N9 Subtype/immunology , Killer Cells, Natural/immunology , Male , Middle Aged , Young Adult
9.
Immunol Cell Biol ; 92(7): 570-7, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24797582

ABSTRACT

Antibodies are one of our most useful biological tools. Indeed, improvements in antibody-based technologies have ushered in a new era of antibody-based therapeutics, research and diagnostic tools. Although improved technologies have led to the development of therapeutic antibodies for treatment of malignancies and inflammatory conditions, the use of advanced antibody technology in the therapy of viral infections is in its infancy. Non-human primate studies have demonstrated that antibodies against the HIV envelope can both prevent viral infection and control viremia. Despite the obvious potential of antibody therapies against HIV, there remain limitations in production and purification capacity that require further research. Recent advances in recombinant antibody technology have led to the development of a range of novel antibody fragments, such as single-domain nanobodies and bispecific antibodies, that are capable of targeting cancer cells to cytotoxic T cells. Novel antibody production techniques have also been designed, allowing antibodies to be obtained from non-mammalian cells, bovine colostrum and the periplasm and cytoplasm of bacteria. These advances may allow large-scale production of HIV antibodies that are capable of protecting against HIV infection or serving as therapeutics that reduce the need for life-long antiretroviral treatment. This review summarises recent advances in antibody-based technologies and discusses the possibilities and challenges of using these advances to design prophylactics and therapeutics against HIV.


Subject(s)
Biotechnology , HIV Antibodies/immunology , HIV Infections/immunology , HIV-1/immunology , Animals , Antibodies, Bispecific/immunology , Antibodies, Bispecific/metabolism , Biotechnology/trends , HIV Infections/epidemiology , HIV Infections/prevention & control , HIV Infections/therapy , HIV Infections/transmission , Humans , Immunoglobulin Fc Fragments/immunology , Immunoglobulin Fc Fragments/metabolism , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Single-Domain Antibodies/immunology , Single-Domain Antibodies/metabolism
10.
AIDS Res Hum Retroviruses ; 30(1): 8-16, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24180375

ABSTRACT

The notoriously low fidelity of HIV-1 replication is largely responsible for the virus's rapid mutation rate, facilitating escape from immune or drug control. The error-prone activity of the viral reverse transcriptase (RT) is predicted to be the most influential mechanism for generating mutations. The low fidelity of RT has been successfully exploited by nucleoside and nucleotide analogue reverse transcriptase inhibitors (NRTIs) that halt viral replication upon incorporation. Consequently, drug-resistant strains have arisen in which the viral RT has an increased fidelity of replication, thus reducing analogue incorporation. Higher fidelity, however, impacts on viral fitness. The appearance of compensatory mutations in combination with higher fidelity NRTI resistance mutations and the subsequent reversion of NRTI-resistant mutations upon cessation of antiretroviral treatment lend support to the notion that higher fidelity exacts a fitness cost. Potential mechanisms for reduced viral fitness are a smaller pool of mutant strains available to respond to immune or drug pressure, slower rates of replication, and a limitation to the dNTP tropism of the virus. Unraveling the relationship between replication fidelity and fitness should lead to a greater understanding of the evolution and control of HIV.


Subject(s)
HIV Reverse Transcriptase/genetics , HIV-1/genetics , Virus Replication/genetics , Anti-HIV Agents/pharmacology , Genetic Variation , HIV Infections/drug therapy , HIV Infections/genetics , HIV Infections/virology , HIV-1/growth & development , Humans , Mutation Rate , Reverse Transcriptase Inhibitors/pharmacology
11.
J Reprod Immunol ; 100(2): 146-56, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24139314

ABSTRACT

The rodent testis is well established as a site of immune privilege where both innate and acquired immune responses are suppressed. Immune cells and responses within human or non-human primate testes, by contrast, are poorly characterised. This study used multi-colour flow cytometry to characterise the leukocytes in testicular cells isolated from 12 young adult pigtail macaques (Macaca nemestrina) by collagenase dispersal, and to measure the cytokine responses of macaque testicular T-lymphocytes to mitogens. B-lymphocytes and granulocytes were present in very low numbers (0.24% and 3.3% of leukocytes respectively), indicating minimal blood contamination. A median of 30.8% of the recovered testicular leukocytes were CD3+ lymphocytes, with CD4 and CD8 T-lymphocyte proportions similar to those in the blood. The proportion of naïve T-lymphocytes in the testis was low, with significantly higher frequencies of central memory cells, compared with the blood. A median of 42.7% of the testicular leukocytes were CD163+ macrophages, while 4.5% were CD14+CD163- monocyte-like macrophages. Small populations of myeloid and plasmacytoid dendritic cells, NK cells and NKT cells were also detected. Following mitogen stimulation, 19.7% of blood T-lymphocytes produced IFNγ and/or TNF, whereas significantly fewer (4.4%) of the testicular T-lymphocytes responded to stimulation. Our results characterise the immune cells within the adult macaque testis and identify a suppression of T-lymphocyte responses. This study provides a baseline to examine the immunology of the primate testis and suggests that testicular immune privilege could also be present in primates.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Lymphocyte Subsets/immunology , Macaca/immunology , Macrophages/immunology , Testis/immunology , Animals , Antigens, CD/metabolism , Cell Separation , Cells, Cultured , Flow Cytometry , Humans , Immunity, Cellular , Immunologic Memory , Interferon-gamma/metabolism , Lymphocyte Activation , Male , Mice , Phytohemagglutinins/immunology , Rats , Testis/pathology , Tetradecanoylphorbol Acetate/immunology , Tumor Necrosis Factor-alpha/metabolism
12.
J Infect Dis ; 208(7): 1051-61, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23812238

ABSTRACT

BACKGROUND: During the 2009 pandemic of influenza A virus subtype H1N1 (A[H1N1]pdm09) infection, older individuals were partially protected from severe disease. It is not known whether preexisting antibodies with effector functions such as antibody-dependent cellular cytotoxicity (ADCC) contributed to the immunity observed. METHODS: We tested serum specimens obtained from 182 individuals aged 1-72 years that were collected either immediately before or after the A(H1N1)pdm09 pandemic for ADCC antibodies to the A(H1N1)pdm09 hemagglutinin (HA) protein. RESULTS: A(H1N1)pdm09 HA-specific ADCC antibodies were detected in almost all individuals aged >45 years (28/31 subjects) before the 2009 A(H1N1) pandemic. Conversely, only approximately half of the individuals aged 1-14 years (11/31) and 15-45 years (17/31) had cross-reactive ADCC antibodies before the 2009 A(H1N1) pandemic. The A(H1N1)pdm09-specific ADCC antibodies were able to efficiently mediate the killing of influenza virus-infected respiratory epithelial cells. Further, subjects >45 years of age had higher ADCC titers to a range of seasonal H1N1 HA proteins, including from the 1918 virus, compared with younger individuals. CONCLUSIONS: ADCC antibodies may have contributed to the protection exhibited in older individuals during the 2009 A(H1N1) pandemic. This work has significant implications for improved vaccination strategies for future influenza pandemics.


Subject(s)
Antibodies, Viral/immunology , Antibody-Dependent Cell Cytotoxicity/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/immunology , Adolescent , Adult , Age Factors , Child , Child, Preschool , Female , Humans , Infant , Influenza, Human/virology , Male , Middle Aged , Young Adult
13.
J Reprod Immunol ; 97(2): 147-58, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23415010

ABSTRACT

Testicular macrophages (TMs) are important contributors to the response of the testis to infection, as well as the regulation of spermatogenesis, steroidogenesis and other homeostatic functions of the testis. The TMs are the largest population of immune cells in a region of tight immunoregulation, where both innate and acquired immune responses are effectively suppressed, and these cells are predicted to be responsible for regulating this immunosuppression. In the rat, TMs have been broadly classified into two main populations, designated "newly arrived" and "resident", the latter being characterised by expression of the scavenger receptor, CD163. Systemic inflammation in response to lipopolysaccharide leads to an influx of CD163(-) monocyte-like ("infiltrating") macrophages into the rodent testis, which have a pro-inflammatory phenotype and express interleukin-1ß, tumour necrosis factor-α, inducible nitric oxide synthase and other inflammatory factors. The resident CD163(+) TMs, on the other hand, constitutively produce IL-10 and are poor stimulators of T-cell proliferation in vitro, indicating that they contribute to testicular immunosuppression. However, our recent studies have demonstrated that the "newly-arrived" CD163(-) TMs present in the rat testis under normal homeostatic conditions show very little response to LPS stimulation in vitro. We here propose a modified model of TM heterogeneity whereby the CD163(-) TMs of the normal rat testis are derived from a monocyte subset that continuously repopulates the testis, and is distinct from the monocyte-like "infiltrating" subset from which pro-inflammatory CD163(-) TMs may be derived during systemic inflammation.


Subject(s)
Antigens, CD/analysis , Antigens, Differentiation, Myelomonocytic/analysis , Inflammation/immunology , Macrophage Activation , Macrophages/immunology , Receptors, Cell Surface/analysis , Testis/cytology , Animals , Cell Proliferation , Inflammation/chemically induced , Interleukin-10/biosynthesis , Interleukin-1beta/biosynthesis , Lipopolysaccharides , Macrophages/metabolism , Male , Monocytes/immunology , Monocytes/metabolism , Nitric Oxide Synthase Type II/biosynthesis , Rats , Testis/immunology , Tumor Necrosis Factor-alpha/biosynthesis
14.
J Immunol ; 190(4): 1837-48, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23319732

ABSTRACT

A better understanding of immunity to influenza virus is needed to generate cross-protective vaccines. Engagement of Ab-dependent cellular cytotoxicity (ADCC) Abs by NK cells leads to killing of virus-infected cells and secretion of antiviral cytokines and chemokines. ADCC Abs may target more conserved influenza virus Ags compared with neutralizing Abs. There has been minimal interest in influenza-specific ADCC in recent decades. In this study, we developed novel assays to assess the specificity and function of influenza-specific ADCC Abs. We found that healthy influenza-seropositive young adults without detectable neutralizing Abs to the hemagglutinin of the 1968 H3N2 influenza strain (A/Aichi/2/1968) almost always had ADCC Abs that triggered NK cell activation and in vitro elimination of influenza-infected human blood and respiratory epithelial cells. Furthermore, we detected ADCC in the absence of neutralization to both the recent H1N1 pandemic strain (A/California/04/2009) as well as the avian H5N1 influenza hemagglutinin (A/Anhui/01/2005). We conclude that there is a remarkable degree of cross-reactivity of influenza-specific ADCC Abs in seropositive humans. Targeting cross-reactive influenza-specific ADCC epitopes by vaccination could lead to improved influenza vaccines.


Subject(s)
Antibodies, Viral/metabolism , Antibody Specificity/immunology , Antibody-Dependent Cell Cytotoxicity/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/immunology , Neutralization Tests/methods , Adult , Animals , Child, Preschool , Cross Reactions/immunology , Hemagglutination Inhibition Tests/methods , Hemagglutinins, Viral/metabolism , Humans , Influenza A Virus, H1N1 Subtype/metabolism , Influenza Vaccines/metabolism , Influenza Vaccines/therapeutic use , Influenza, Human/immunology , Influenza, Human/prevention & control , Influenza, Human/virology , Macaca nemestrina , Middle Aged , Protein Binding/immunology , Young Adult
15.
Cytokine ; 61(1): 199-204, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23116663

ABSTRACT

Activin A, a transforming growth factor-ß family cytokine, plays a crucial role in regulating the onset and severity of many inflammatory conditions, such as acute lipopolysaccharide (LPS)-induced inflammation. Activin A is also implicated in type 2 diabetes (T2D), a disease characterised by insulin resistance, hyperglycaemia and chronic elevation of pro-inflammatory cytokines, including tumour necrosis factor (TNF-α). In the human, neutrophils contain activin A that can be released in response to TNF-α. Studies of inflammatory disease in vivo, however, generally use the mouse, so it is essential to know if murine neutrophils have similar properties. Regulation of activin A was investigated in bone marrow-derived neutrophil precursors (BMNPs) from 8 to 10 weeks old C57BL6/J male mice. The BMNPs contained 7-fold higher concentrations of activin A than bone marrow mononuclear cells. Release of activin A from isolated BMNPs was stimulated by TNF-α, but this was not due to increased activin A production. In contrast to TNF-α, LPS had no effect on isolated BMNPs, but stimulated activin A release and production in total bone marrow cell cultures. Moreover, activin A release in response to LPS, was not prevented in TNF-α null mice. Increased glucose and insulin had no effect on base-line activin A secretion by BMNPs in culture, but pre-treatment with insulin blocked the TNF-α induced release of activin A. These results indicate that murine neutrophils are a source of stored activin A, the release of which can be directly stimulated by TNF-α, although TNF-α is not the only stimulator of activin A release during inflammation. Furthermore, regulation of neutrophil activin A release by insulin may also play a role in the inflammation associated with T2D.


Subject(s)
Activins/metabolism , Bone Marrow Cells/metabolism , Insulin/pharmacology , Neutrophils/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cells, Cultured , Glucose/pharmacology , Inflammation , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Tumor Necrosis Factor-alpha/genetics
16.
Reprod Fertil Dev ; 25(3): 570-80, 2013.
Article in English | MEDLINE | ID: mdl-23164397

ABSTRACT

Activin A and its inhibitors follistatin and inhibin play key roles in development and function of the male reproductive tract. Quantitative (q) polymerase chain reaction (PCR) was used to evaluate the expression of Inhba (the gene encoding activin A subunits), Inha and Inhbb (genes encoding the inhibin B subunits), as well as the genes for follistatin (Fst) and follistatin-like 3 (Fstl3) and the activin receptor subunits, in the male mouse reproductive tract. A qPCR assay that discriminated between the two follistatin variants of Fst288 (tissue-bound form) and Fst315 (circulating) was established. Activin A protein was measured by ELISA, whereas the inhibin α-subunit and total follistatin proteins were measured by radioimmunoassay (RIA). A screen of 22 tissues demonstrated tissue-specific regulation of the follistatin variants, with Fst288 highly expressed in the vas deferens and Fst315 most highly expressed in the skin. The expression of Fst288 and Fst315 and follistatin protein levels increased progressively from the testis through to the distal vas deferens. Inhba and the activin receptors were highly expressed in the epididymis, but activin A protein was elevated in both the epididymis and vas deferens. Inhibin α-subunit mRNA and protein and Inhbb expression were highest in the testis. These results indicate a role for activin A within the epididymis, but also that activin A bioactivity may be increasingly inhibited by follistatin distally along the male reproductive tract.


Subject(s)
Activins/metabolism , Epididymis/metabolism , Follistatin/metabolism , Gene Expression Regulation , Inhibins/metabolism , Vas Deferens/metabolism , 3' Untranslated Regions , Activin Receptors/genetics , Activin Receptors/metabolism , Activins/genetics , Animals , Base Sequence , Exons , Follistatin/chemistry , Follistatin/genetics , Genitalia, Male/metabolism , Inhibins/genetics , Male , Mice , Mice, Inbred Strains , Molecular Sequence Data , Organ Specificity , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism
17.
Am J Physiol Regul Integr Comp Physiol ; 303(6): R665-75, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22855279

ABSTRACT

Activin A, a member of the transforming growth factor-ß family, increases in the circulation within 1 h after administration of bacterial LPS. To clarify the origins of this rapid increase, the distribution of activin A and its binding protein, follistatin, and their production following LPS treatment, were assessed in adult male mice. In untreated mice, activin A was detectable in all 23 tissues examined, with highest mRNA expression (as measured by quantitative RT-PCR) was found in the liver, and the largest concentration of activin A protein (by ELISA) was found in the bone marrow. Likewise, follistatin mRNA and protein were present in all tissues, with highest expression in the vas deferens. Activin A and follistatin mRNA did not increase significantly in any tissue within the first hour after LPS, but activin A protein decreased by 35% in the bone marrow and increased 5-fold in the lung. No significant changes were observed in any other tissue. Activin A reached a peak in the circulation 1 h following LPS, and then declined. Cycloheximide, an inhibitor of protein translation, reduced this increase of activin A by more than 50%. Actinomycin D, an inhibitor of mRNA transcription, had no effect. Circulating follistatin did not increase until 4 h after LPS and was not affected by either inhibitor. These data indicate that the rapid increase in circulating activin A during LPS-induced inflammation is regulated at the posttranscriptional level, apparently from newly translated and stored protein, and implicate bone marrow-derived cells, and, in particular, neutrophils, as a significant source of this preformed activin A.


Subject(s)
Activins/metabolism , Follistatin/metabolism , Lipopolysaccharides/toxicity , Activins/blood , Activins/genetics , Animals , Dactinomycin/pharmacology , Follistatin/blood , Follistatin/genetics , Gene Expression Regulation/physiology , Male , Mice , Protein Subunits , RNA, Messenger/genetics , RNA, Messenger/metabolism
18.
Virology ; 428(1): 11-20, 2012 Jun 20.
Article in English | MEDLINE | ID: mdl-22507218

ABSTRACT

Defining which cells become infected with simian immunodeficiency virus (SIV) in vivo should assist in unravelling the pathogenesis of human immunodeficiency virus (HIV)/SIV infection. HIV/SIV infection of CD4(+) T cells resulted in down-regulation of CD3 and CD4 surface molecules in vitro, however this phenomenon is poorly characterised in vivo. Intracellular SIV p27 was studied by flow cytometry in serial blood samples and lymph node samples during acute infection of 17 SIVmac-infected pigtail macaques. Two weeks after infection, a mean of 56±6.8% the p27(+) cells were lymphocytes negative for surface CD4 and CD3, and indeed the highest proportion of SIV infected cells were found in the small subset of CD3(Lo)CD4(-)CD8(-) lymphocytes, indicating that infection has lead to down-regulation of these markers in vivo. Furthermore, the relative amount of SIV p27 within lymphocytes (based of mean fluorescence intensity) was higher in CD3(Lo)CD4(-) and CD3(-) infected cells than in CD3(+) or CD4(+) p27(+) populations, consistent with greater viral production in CD4(+) T cells down-regulating CD3 and CD4 molecules. The CD3(-)CD4(-) infected cells expressed T cell markers CD2 and CD5 and were negative for monocyte, NK and B cell markers. The majority of infected cells were CD28(+)CD95(+) central memory T cells. Surprisingly, p27(+) blood lymphocytes were mostly negative for activation markers CD25 and CD69, but most of the infected lymph nodes cells were activated. Our results characterise productively-infected macaque lymphocytes in vivo. The high proportion of SIV-infected lymphocytes that are CD3(-)CD4(-) has important implications for the in vivo study of pathogenesis of SIV/HIV infections.


Subject(s)
Disease Models, Animal , HIV Infections/immunology , Macaca nemestrina , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/physiology , Animals , CD3 Complex/genetics , CD3 Complex/immunology , CD4 Antigens/genetics , CD4 Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Down-Regulation , HIV Infections/genetics , HIV Infections/virology , Humans , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Viral Proteins/genetics , Viral Proteins/metabolism
19.
Mol Cell Endocrinol ; 359(1-2): 30-42, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-21964464

ABSTRACT

Activin A provides a unique link between reproduction and immunity, which is especially significant in the adult testis. This cytokine, together with inhibin B and follistatin acting as regulators of activin A activity, is fundamentally involved in the regulation of spermatogenesis and testicular steroidogenesis. However, activin A also has a much broader role in control of inflammation, fibrosis and immunity. In the Sertoli cell, activin A is regulated by signalling pathways that normally regulate stress and inflammation, signalling pathways that intersect with the classical hormonal regulatory pathways mediated by FSH. Modulation of activin A production and activity during spermatogenesis is implicated in the fine control of the cycle of the seminiferous epithelium. The immunoregulatory properties of activin A also suggest that it may be involved in maintaining testicular immune privilege. Consequently, elevated activin A production within the testis during inflammation and infection may contribute to spermatogenic failure, fibrosis and testicular damage.


Subject(s)
Activins/metabolism , Immunomodulation , Inhibins/metabolism , Spermatogenesis , Testis/metabolism , Activins/physiology , Animals , Humans , Inflammation/metabolism , Inhibins/physiology , Male , Signal Transduction , Testis/immunology , Testis/pathology
20.
J Leukoc Biol ; 90(1): 133-43, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21498587

ABSTRACT

The ability of the rodent testis to tolerate graft alloantigens and spermatogenic cell autoantigens is well known. The mechanisms underlying this "immune privilege" are poorly understood, but the numerous resident TMs have been implicated. Although it has been assumed that TMs display a phenotype consistent with immune privilege, this has not been formally established. Consequently, TMs were isolated from adult rats and cultured under basal conditions and following stimulation with LPS and IFN-γ (classical activation) or IL-4 (alternative activation). BMMs matured in vitro were used as control. Expression of the classical (proinflammatory) activation markers TNF-α, IL-1ß, iNOS, IL-6, RANTES, IL-12p40, and SOCS3 and alternative (immunoregulatory) activation markers IL-10, TGF-ß1, CXCL2, and SOCS1 was measured by QPCR or ELISA. In culture, TMs were characterized by poor expression of classical activation genes and TGF-ß1 but constitutively high IL-10 production and reduced costimulatory activity in a polyclonal T cell activation assay. This pattern of gene expression was associated with TMs expressing the scavenger receptor CD163, which is characteristic of tissue resident macrophages and alternative activation. By contrast, CD163-negative TMs displayed reduced inflammatory gene expression but did not constitutively produce IL-10. These data indicate that under the influence of the testicular environment, macrophages adopt an alternatively activated phenotype, involving reduced capacity for proinflammatory gene expression, constitutive IL-10 production, and impaired ability to support T cell activation, consistent with a role in maintaining testicular immune privilege.


Subject(s)
Gene Expression Regulation/immunology , Immune Tolerance/immunology , Interleukin-10/biosynthesis , Macrophage Activation/immunology , Macrophages/immunology , Testis/immunology , Animals , Cell Separation , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression , Interleukin-10/immunology , Lymphocyte Activation/immunology , Macrophages/cytology , Macrophages/metabolism , Male , Phenotype , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , Testis/cytology , Testis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...