Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
iScience ; 26(5): 106628, 2023 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-37192971

RESUMEN

This study was undertaken to investigate the role of CD4+FoxP3+ regulatory T cells (Tregs) in regulating neuroinflammation during viral Ag-challenge and re-challenge. CD8+ lymphocytes persisting within tissues are designated tissue-resident memory T cells (TRM), within brain: bTRM. Reactivation of bTRM with T cell epitope peptides generates rapid antiviral recall, but repeated stimulation leads to cumulative dysregulation of microglial activation, proliferation, and prolonged neurotoxic mediator production. Here, we show Tregs were recruited into murine brains following prime-CNS boost, but displayed altered phenotypes following repeated Ag-challenge. In response to repeated Ag, brain Tregs (bTregs) displayed inefficient immunosuppressive capacity, along with reduced expression of suppression of tumorigenicity 2 (ST2) and amphiregulin (Areg). Ex vivo Areg treatment revealed reduced production of neurotoxic mediators such as iNOS, IL-6, and IL-1ß, and decreased microglial activation and proliferation. Taken together, these data indicate bTregs display an unstable phenotype and fail to control reactive gliosis in response to repeated Ag-challenge.

2.
Cells ; 11(18)2022 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-36139401

RESUMEN

Regulatory T-cells (Tregs) play pivotal roles during infection, cancer, and autoimmunity. In our previous study, we demonstrated a role for the PD-1:PD-L1 pathway in controlling cytolytic responses of CD8+ T lymphocytes against microglial cells presenting viral peptides. In this study, we investigated the role of Tregs in suppressing CD8+ T-cell-mediated cytotoxicity against primary microglial cells. Using in vitro cytotoxicity assays and flow cytometry, we demonstrated a role for Tregs in suppressing antigen-specific cytotoxic T-lymphocyte (CTL) responses against microglia loaded with a model peptide (SIINFEKL). We went on to show a significant decrease in the frequency of IFN-γ- and TNF-producing CD8+ T-cells when cultured with Tregs. Interestingly, a significant increase in the frequency of granzyme B- and Ki67-producing CTLs was observed. We also observed a significant decrease in the production of interleukin (IL)-6 by microglia. On further investigation, we found that Tregs significantly reduced MHC class 1 (MHC-1) expression on IFN-γ-treated microglial cells. Taken together, these studies demonstrate an immunosuppressive role for Tregs on CTL responses generated against primary microglia. Hence, modulation of Treg cell activity in combination with negative immune checkpoint blockade may stimulate anti-viral T-cell responses to more efficiently clear viral infection from microglial cell reservoirs.


Asunto(s)
Linfocitos T Citotóxicos , Linfocitos T Reguladores , Antígeno B7-H1/metabolismo , Linfocitos T CD8-positivos , Granzimas/metabolismo , Inhibidores de Puntos de Control Inmunológico , Interferón gamma/metabolismo , Interleucinas/metabolismo , Antígeno Ki-67/metabolismo , Microglía/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo
3.
Brain Sci ; 11(11)2021 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-34827481

RESUMEN

The role of select pro- and anti-inflammatory mediators in driving microglial cell polarization into classically (M1), or alternatively, (M2) activated states, as well as the subsequent differential responses of these induced phenotypes, was examined. Expression of PD-L1, MHC-II, MHC-I, arginase 1 (Arg-1), and inducible nitric oxide synthase (iNOS) was assessed using multi-color flow cytometry. We observed that both pro- and anti-inflammatory mediators induced PD-L1 expression on non-polarized microglia. Moreover, IFN-γ stimulated significant MHC class I and II expression on these cells. Interestingly, we observed that only IL-4 treatment induced Arg-1 expression, indicating M2 polarization. These M2 cells were refractory to subsequent depolarization and maintained their alternatively activated state. Furthermore, PD-L1 expression was significantly induced on these M2-polarized microglia after treatment with pro-inflammatory mediators, but not anti-inflammatory cytokines. In addition, we observed that only LPS induced iNOS expression in microglial cells, indicating M1 polarization. Furthermore, IFN-γ significantly increased the percentage of M1-polarized microglia expressing iNOS. Surprisingly, when these M1-polarized microglia were treated with either IL-6 or other anti-inflammatory cytokines, they returned to their non-polarized state, as demonstrated by significantly reduced expression of iNOS. Taken together, these results demonstrate differential responses of microglial cells to mediators present in dissimilar microenvironments.

4.
Front Cell Neurosci ; 15: 686340, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34447297

RESUMEN

Upon reactivation of quiescent neurotropic viruses antigen (Ag)-specific brain resident-memory CD8+ T-cells (bTRM) may respond to de novo-produced viral Ag through the rapid release of IFN-γ, which drives subsequent interferon-stimulated gene expression in surrounding microglia. Through this mechanism, a small number of adaptive bTRM may amplify responses to viral reactivation leading to an organ-wide innate protective state. Over time, this brain-wide innate immune activation likely has cumulative neurotoxic and neurocognitive consequences. We have previously shown that HIV-1 p24 Ag-specific bTRM persist within the murine brain using a heterologous prime-CNS boost strategy. In response to Ag restimulation, these bTRM display rapid and robust recall responses, which subsequently activate glial cells. In this study, we hypothesized that repeated challenges to viral antigen (Ag) (modeling repeated episodes of viral reactivation) culminate in prolonged reactive gliosis and exacerbated neurotoxicity. To address this question, mice were first immunized with adenovirus vectors expressing the HIV p24 capsid protein, followed by a CNS-boost using Pr55Gag/Env virus-like particles (HIV-VLPs). Following the establishment of the bTRM population [>30 days (d)], prime-CNS boost animals were then subjected to in vivo challenge, as well as re-challenge (at 14 d post-challenge), using the immunodominant HIV-1 AI9 CD8+ T-cell epitope peptide. In these studies, Ag re-challenge resulted in prolonged expression of microglial activation markers and an increased proliferative response, longer than the challenge group. This continued expression of MHCII and PD-L1 (activation markers), as well as Ki67 (proliferative marker), was observed at 7, 14, and 30 days post-AI9 re-challenge. Additionally, in vivo re-challenge resulted in continued production of inducible nitric oxide synthase (iNOS) with elevated levels observed at 7, 14 and 30 days post re-challenge. Interestingly, iNOS expression was significantly lower among challenged animals when compared to re-challenged groups. Furthermore, in vivo specific Ag re-challenge produced lower levels of arginase (Arg)-1 when compared with the challenged group. Taken together, these results indicate that repeated Ag-specific stimulation of adaptive immune responses leads to cumulative dysregulated microglial cell activation.

5.
Glia ; 69(4): 858-871, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33128485

RESUMEN

Microglial cells are the main reservoir for HIV-1 within the brain and potential exists for negative immune checkpoint blockade therapies to purge this viral reservoir. Here, we investigated cytolytic responses of CD8+ T lymphocytes against microglia loaded with peptide epitopes. Initially, flow cytometric analysis demonstrated efficient killing of HIV-1 p24 AI9 or YI9 peptide-loaded splenocytes in MHC-matched recipients. Cytolytic killing of microglia was first demonstrated using ovalbumin (OVA) as a model antigen for in vitro cytotoxic T lymphocyte (CTL) assays. Peptide-loaded primary microglia obtained from programmed death ligand (PD-L) 1 knockout (KO) animals showed significantly more killing than cells from wild-type (WT) animals when co-cultured with activated CD8+ T-cells isolated from rAd5-OVA primed animals. Moreover, when peptide loaded-microglial cells from WT animals were treated with neutralizing α-PD-L1 Ab, significantly more killing was observed compared to either untreated or IgG isotype-treated cells. Most importantly, significantly increased in vivo killing of HIV-1 p24 YI9 peptide-loaded microglia from PD-L1 KO animals, as well as AI9 peptide-loaded BALB/c microglial cells treated with α-PD-L1, was observed within brains of rAd5-p24 primed-CNS boosted C57BL/6 or BALB/c mice, respectively. Finally, ex vivo responses of brain CD8+ T-cells in response to AI9 stimulation showed significantly increased IFN-γ and IL-2 production when treated with α-PD-1 Abs. Greater proliferation of CD8+ T-cells from the brain was also observed following blockade. Taken together, these studies demonstrate that PD-L1 induction on microglia restrains CTL responses and indicate that immune checkpoint blockade targeting this pathway may be beneficial in clearing viral brain reservoirs.


Asunto(s)
Antígeno B7-H1 , Linfocitos T Citotóxicos , Animales , Linfocitos T CD8-positivos , Inhibidores de Puntos de Control Inmunológico , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microglía , Péptidos/farmacología , Receptor de Muerte Celular Programada 1
6.
iScience ; 20: 512-526, 2019 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-31655062

RESUMEN

HIV-associated neurocognitive disorders (HAND) persist even during effective combination antiretroviral therapy (cART). Although the cause of HAND is unknown, studies link chronic immune activation, neuroinflammation, and cerebrospinal fluid viral escape to disease progression. In this study, we tested the hypothesis that specific, recall immune responses from brain-resident memory T cells (bTRM) could activate glia and induce neurotoxic mediators. To address this question, we developed a heterologous prime-central nervous system (CNS) boost strategy in mice. We observed that the murine brain became populated with long-lived CD8+ bTRM, some being specific for an immunodominant Gag epitope. Recall stimulation using HIV-1 AI9 peptide administered in vivo resulted in microglia displaying elevated levels of major histocompatibility complex class II and programmed death-ligand 1, and demonstrating tissue-wide reactive gliosis. Immunostaining further confirmed this glial activation. Taken together, these results indicate that specific, adaptive recall responses from bTRM can induce reactive gliosis and production of neurotoxic mediators.

7.
Pain Res Manag ; 2019: 1260353, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31354896

RESUMEN

The most common neurological complication in patients receiving successful combination antiretroviral therapy (cART) is peripheral neuropathic pain. Data show that distal symmetric polyneuropathy (DSP) also develops along with murine acquired immunodeficiency syndrome (MAIDS) after infection with the LP-BM5 murine retrovirus mixture. Links between cannabinoid receptor 2 (CB2R) and peripheral neuropathy have been established in animal models using nerve transection, chemotherapy-induced pain, and various other stimuli. Diverse types of neuropathic pain respond differently to standard drug intervention, and little is currently known regarding the effects of modulation through CB2Rs. In this study, we evaluated whether treatment with the exogenous synthetic CB2R agonists JWH015, JWH133, Gp1a, and HU308 controls neuropathic pain and neuroinflammation in animals with chronic retroviral infection. Hind-paw mechanical hypersensitivity in CB2R agonist-treated versus untreated animals was assessed using the MouseMet electronic von Frey system. Multicolor flow cytometry was used to determine the effects of CB2R agonists on macrophage activation and T-lymphocyte infiltration into dorsal root ganglia (DRG) and lumbar spinal cord (LSC). Results demonstrated that, following weekly intraperitoneal injections starting at 5 wk p.i., JWH015, JWH133, and Gp1a, but not HU308 (5 mg/kg), significantly ameliorated allodynia when assessed 2 h after ligand injection. However, these same agonists (2x/wk) did not display antiallodynic effects when mechanical sensitivity was assessed 24 h after ligand injection. Infection-induced macrophage activation and T-cell infiltration into the DRG and LSC were observed at 12 wk p.i., but this neuroinflammation was not affected by treatment with any CB2R agonist. Activation of JAK/STAT3 has been shown to contribute to development of neuropathic pain in the LSC and pretreatment of primary murine microglia (2 h) with JWH015-, JWH133-, or Gp1a-blocked IFN-gamma-induced phosphorylation of STAT1 and STAT3. Taken together, these data show that CB2R agonists demonstrate acute, but not long-term, antiallodynic effects on retrovirus infection-induced neuropathic pain.


Asunto(s)
Agonistas de Receptores de Cannabinoides/farmacología , Neuralgia/virología , Infecciones por Retroviridae/complicaciones , Animales , Cannabinoides/farmacología , Modelos Animales de Enfermedad , Humanos , Hiperalgesia/virología , Indenos/farmacología , Indoles/farmacología , Masculino , Ratones , Pirazoles/farmacología , Retroviridae
8.
Curr Opin Obstet Gynecol ; 30(2): 111-115, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29489501

RESUMEN

PURPOSE OF REVIEW: To review the current medical and stem-cell therapy for spinal muscular atrophy (SMA) and prenatal transplantation of amniotic fluid stem cells in the future. RECENT FINDINGS: SMA is an autosomal recessive inheritance of neurodegenerative disease, which is caused of the mutation in survival motor neuron. The severe-type SMA patients usually die from the respiratory failure within 2 years after birth. Recently, researchers had found that 3-methyladenine could inhibit the autophagy and had the capacity to reduce death of the neurons. The first food and drug administration-approved drug to treat SMA, Nusinersen, is a modified antisense oligonucleotide to target intronic splicing silencer N1 just recently launched. Not only medical therapy, but also stem cells including neural stem cells, embryonic stem cells, mesenchymal stem cells, and induced pluripotent stem cells could show the potential to repair the injured tissue and differentiate into neuron cells to rescue the SMA animal models. Human amniotic fluid stem cells (HAFSCs) share the potential of mesenchymal stem cells and could differentiate into tri-lineage-relative cells, which are also having the ability to restore the injured neuro-muscular function. In this review, we further demonstrate the therapeutic effect of using HAFSCs to treat type III SMA prenatally. HAFSCs, similar to other stem cells, could also help the improvement of SMA with even longer survival. SUMMARY: The concept of prenatal stem-cell therapy preserves the time window to treat disease in utero with much less cell number. Stem cell alone might not be enough to correct or cure the SMA but could be applied as the additional therapy combined with antisense oligonucleotide in the future.


Asunto(s)
Líquido Amniótico/citología , Atrofia Muscular Espinal/terapia , Trasplante de Células Madre , Animales , Femenino , Humanos , Atrofia Muscular Espinal/clasificación , Embarazo
9.
Immun Inflamm Dis ; 6(2): 332-344, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29602245

RESUMEN

INTRODUCTION: Previous work from our laboratory has demonstrated in vivo persistence of CD103+ CD69+ brain resident memory CD8+ T-cells (bTRM ) following viral infection, and that the PD-1: PD-L1 pathway promotes development of these TRM cells within the brain. Although glial cells express low basal levels of PD-L1, its expression is upregulated upon IFN-γ-treatment, and they have been shown to modulate antiviral T-cell effector responses through the PD-1: PD-L1 pathway. METHODS: We performed flow cytometric analysis of cells from co-cultures of mixed glia and CD8+ T-cells obtained from wild type mice to investigate the role of glial cells in the development of bTRM . RESULTS: In this study, we show that interactions between reactive glia and anti-CD3 Ab-stimulated CD8+ T-cells promote development of CD103+ CD69+ CD8+ T-cells through engagement of the PD-1: PD-L1 pathway. These studies used co-cultures of primary murine glial cells obtained from WT animals along with CD8+ T-cells obtained from either WT or PD-1 KO mice. We found that αCD3 Ab-stimulated CD8+ T-cells from WT animals increased expression of CD103 and CD69 when co-cultured with primary murine glial cells. In contrast, significantly reduced expression of CD103 and CD69 was observed using CD8+ T-cells from PD-1 KO mice. We also observed that reactive glia promoted high levels of CD127, a marker of memory precursor effector cells (MPEC), on CD69+ CD8+ T-cells, which promotes development of TRM cells. Interestingly, results obtained using T-cells from PD-1 KO animals showed significantly reduced expression of CD127 on CD69+ CD8+ cells. Additionally, blocking of glial PD-L1 resulted in decreased expression of CD103, along with reduced CD127 on CD69+ CD8+ T-cells. CONCLUSIONS: Taken together, these results demonstrate a role for activated glia in promoting development of bTRM through the PD-1: PD-L1 pathway.


Asunto(s)
Antígeno B7-H1/metabolismo , Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica , Neuroglía/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Antígeno B7-H1/inmunología , Encéfalo/citología , Encéfalo/inmunología , Linfocitos T CD8-positivos/metabolismo , Células Cultivadas , Enfermedades Virales del Sistema Nervioso Central/inmunología , Enfermedades Virales del Sistema Nervioso Central/virología , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Humanos , Cadenas alfa de Integrinas/metabolismo , Lectinas Tipo C/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Muromegalovirus/inmunología , Neuroglía/metabolismo , Cultivo Primario de Células , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología , Transducción de Señal/inmunología
10.
J Neuroinflammation ; 15(1): 66, 2018 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-29506535

RESUMEN

BACKGROUND: Peripheral neuropathy is currently the most common neurological complication in HIV-infected individuals, occurring in 35-50% of patients undergoing combination anti-retroviral therapy. Data have shown that distal symmetric polyneuropathy develops in mice by 6 weeks following infection with the LP-BM5 retrovirus mixture. Previous work from our laboratory has demonstrated that glial cells modulate antiviral T-cell effector responses through the programmed death (PD)-1: PD-L1 pathway, thereby limiting the deleterious consequences of unrestrained neuroinflammation. METHODS: Using the MouseMet electronic von Frey system, we assessed hind-paw mechanical hypersensitivity in LP-BM5-infected wild-type (WT) and PD-1 KO animals. Using multi-color flow cytometry, we quantitatively assessed cellular infiltration and microglial activation. Using real-time RT-PCR, we assessed viral load, expression of IFN-γ, iNOS, and MHC class II. Using western blotting, we measured protein nitrosylation within the lumbar spinal cord (LSC) and dorsal root ganglion (DRG). Histochemical staining was performed to analyze the presence of CD3, ionized calcium binding adaptor molecule (Iba)-1, MHCII, nitrotyrosine, isolectin B4 (IB4) binding, and neurofilament 200 (NF200). Statistical analyses were carried out using graphpad prism. RESULTS: Hind-paw mechanical hypersensitivity observed in LP-BM5-infected animals was associated with significantly increased lymphocyte infiltration into the spinal cord and DRG. We also observed elevated expression of IFN-γ (in LSC and DRG) and MHC II (on resident microglia in LSC). We detected elevated levels of 3-nitrotyrosine within the LSC and DRG of LP-BM5-infected animals, an indicator of nitric oxide (NO)-induced protein damage. Moreover, we observed 3-nitrotyrosine in both small (IB4+) and large (NF200+) DRG sensory neurons. Additionally, infected PD-1 KO animals displayed significantly greater mechanical hypersensitivity than WT or uninfected mice at 4 weeks post-infection (p.i.). Accelerated onset of hind-paw hypersensitivity in PD-1 KO animals was associated with significantly increased infiltration of CD4+ and CD8+ T lymphocytes, macrophages, and microglial activation at early time points. Importantly, we also observed elevated levels of 3-nitrotyrosine and iNOS in infected PD-1 KO animals when compared with WT animals. CONCLUSIONS: Results reported here connect peripheral immune cell infiltration and reactive gliosis with nitrosative damage. These data may help elucidate how retroviral infection-induced neuroinflammatory networks contribute to nerve damage and neuropathic pain.


Asunto(s)
Neuralgia/etiología , Infecciones por Retroviridae/complicaciones , Animales , Antígenos CD/metabolismo , Modelos Animales de Enfermedad , Ganglios Espinales , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/metabolismo , Interferón gamma/metabolismo , Leucocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuralgia/patología , Neuralgia/virología , Óxido Nítrico Sintasa de Tipo II , Receptor de Muerte Celular Programada 1/deficiencia , Receptor de Muerte Celular Programada 1/genética , ARN Mensajero , Retroviridae/patogenicidad , Médula Espinal/patología
11.
J Neuroinflammation ; 14(1): 82, 2017 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-28407741

RESUMEN

BACKGROUND: Previous work from our laboratory has demonstrated that during acute viral brain infection, glial cells modulate antiviral T cell effector responses through the PD-1: PD-L1 pathway, thereby limiting the deleterious consequences of unrestrained neuroinflammation. Here, we evaluated the PD-1: PD-L1 pathway in development of brain-resident memory T cells (bTRM) following murine cytomegalovirus (MCMV) infection. METHODS: Flow cytometric analysis of immune cells was performed at 7, 14, and 30 days post-infection (dpi) to assess the shift of brain-infiltrating CD8+ T cell populations from short-lived effector cells (SLEC) to memory precursor effector cells (MPEC), as well as generation of bTRMs. RESULTS: In wild-type (WT) animals, we observed a switch in the phenotype of brain-infiltrating CD8+ T cell populations from KLRG1+ CD127- (SLEC) to KLRG1- CD127+ (MPEC) during transition from acute through chronic phases of infection. At 14 and 30 dpi, the majority of CD8+ T cells expressed CD127, a marker of memory cells. In contrast, fewer CD8+ T cells expressed CD127 within brains of infected, PD-L1 knockout (KO) animals. Notably, in WT mice, a large population of CD8+ T cells was phenotyped as CD103+ CD69+, markers of bTRM, and differences were observed in the numbers of these cells when compared to PD-L1 KOs. Immunohistochemical studies revealed that brain-resident CD103+ bTRM cells were localized to the parenchyma. Higher frequencies of CXCR3 were also observed among WT animals in contrast to PD-L1 KOs. CONCLUSIONS: Taken together, our results indicate that bTRMs are present within the CNS following viral infection and the PD-1: PD-L1 pathway plays a role in the generation of this brain-resident population.


Asunto(s)
Antígeno B7-H1/deficiencia , Encéfalo/metabolismo , Linfocitos T CD8-positivos/metabolismo , Encefalitis Viral/metabolismo , Receptor de Muerte Celular Programada 1/deficiencia , Animales , Encéfalo/inmunología , Linfocitos T CD8-positivos/inmunología , Encefalitis Viral/inmunología , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Células 3T3 NIH , Transducción de Señal/fisiología
12.
Sci Rep ; 7: 41889, 2017 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-28165503

RESUMEN

Fcγ receptors (FcγRs) for IgG couple innate and adaptive immunity through activation of effector cells by antigen-antibody complexes. We investigated relative levels of activating and inhibitory FcγRs on brain-resident microglia following murine cytomegalovirus (MCMV) infection. Flow cytometric analysis of microglial cells obtained from infected brain tissue demonstrated that activating FcγRs were expressed maximally at 5 d post-infection (dpi), while the inhibitory receptor (FcγRIIB) remained highly elevated during both acute and chronic phases of infection. The highly induced expression of activating FcγRIV during the acute phase of infection was also noteworthy. Furthermore, in vitro analysis using cultured primary microglia demonstrated the role of interferon (IFN)γ and interleukin (IL)-4 in polarizing these cells towards a M1 or M2 phenotype, respectively. Microglial cell-polarization correlated with maximal expression of either FcγRIV or FcγRIIB following stimulation with IFNγ or IL-4, respectively. Finally, we observed a significant delay in polarization of microglia towards an M2 phenotype in the absence of FcγRs in MCMV-infected Fcer1g and FcgR2b knockout mice. These studies demonstrate that neuro-inflammation following viral infection increases expression of activating FcγRs on M1-polarized microglia. In contrast, expression of the inhibitory FcγRIIB receptor promotes M2-polarization in order to shut-down deleterious immune responses and limit bystander brain damage.


Asunto(s)
Encéfalo/metabolismo , Infecciones por Herpesviridae/metabolismo , Microglía/metabolismo , Receptores de IgG/metabolismo , Células 3T3 , Animales , Encéfalo/citología , Diferenciación Celular , Células Cultivadas , Femenino , Interferón gamma/farmacología , Interleucina-4/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microglía/citología , Microglía/efectos de los fármacos , Muromegalovirus , Receptores de IgG/genética
13.
Glia ; 63(11): 1982-1996, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26041050

RESUMEN

Long-term, persistent central nervous system inflammation is commonly seen following brain infection. Using a murine model of viral encephalitis (murine cytomegalovirus, MCMV) we have previously shown that post-encephalitic brains are maintained in an inflammatory state consisting of glial cell reactivity, retention of brain-infiltrating tissue-resident memory CD8+ T-cells, and long-term persistence of antibody-producing cells of the B-lineage. Here, we report that this neuroinflammation occurs concomitantly with accumulation and retention of immunosuppressive regulatory T-cells (Tregs), and is exacerbated following their ablation. However, the extent to which these Tregs function to control neuroimmune activation following MCMV encephalitis is unknown. In this study, we used Foxp3-diphtheria toxin receptor-GFP (Foxp3-DTR-GFP) transgenic mice, which upon administration of low-dose diphtheria toxin (DTx) results in the specific depletion of Tregs, to investigate their function. We found treatment with DTx during the acute phase of viral brain infection (0-4 dpi) resulted in depletion of Tregs from the brain, exacerbation of encephalitis (i.e., increased presence of CD4+ and CD8+ T-cells), and chronic reactive phenotypes of resident glial cells (i.e., elevated MHC Class II as well as PD-L1 levels, sustained microgliosis, and increased glial fibrillary acidic protein (GFAP) expression on astrocytes) versus untreated, infected animals. This chronic proinflammatory environment was associated with reduced cognitive performance in spatial learning and memory tasks (Barnes Maze) by convalescent animals. These data demonstrate that chronic glial cell activation, unremitting post-encephalitic neuroinflammation, and its associated long-term neurological sequelae in response to viral brain infection are modulated by the immunoregulatory properties of Tregs. GLIA 2015;63:1982-1996.

14.
PLoS One ; 10(12): e0145457, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26720146

RESUMEN

Accumulation and retention of regulatory T-cells (Tregs) has been reported within post viral-encephalitic brains, however, the full extent to which these cells modulate neuroinflammation is yet to be elucidated. Here, we used Foxp3-DTR (diphtheria toxin receptor) knock-in transgenic mice, which upon administration of low dose diphtheria toxin (DTx) results in specific deletion of Tregs. We investigated the proliferation status of various immune cell subtypes within inflamed central nervous system (CNS) tissue. Depletion of Tregs resulted in increased proliferation of both CD8+ and CD4+ T-cell subsets within the brain at 14 d post infection (dpi) when compared to Treg-sufficient animals. At 30 dpi, while proliferation of CD8+ T-cells was controlled within brains of both Treg-depleted and undepleted mice, proliferation of CD4+ T-cells remained significantly enhanced with DTx-treatment. Previous studies have demonstrated that Treg numbers within the brain rebound following DTx treatment to even higher numbers than in untreated animals. Despite this rebound, CD8+ and CD4+ T-cells proliferated at a higher rate when compared to that of Treg-sufficient mice, thus maintaining sustained neuroinflammation. Furthermore, at 30 dpi we found the majority of CD8+ T-cells were CD127hi KLRG1- indicating that the cells were long lived memory precursor cells. These cells showed marked elevation of CD103 expression, a marker of tissue resident-memory T-cells (TRM) in the CNS, in untreated animals when compared to DTx-treated animals suggesting that generation of TRM is impaired upon Treg depletion. Moreover, the effector function of TRM as indicated by granzyme B production in response to peptide re-stimulation was found to be more potent in Treg-sufficient animals. Taken together, our findings demonstrate that Tregs limit neuroinflammatory responses to viral infection by controlling cell proliferation and may direct a larger proportion of lymphocytes within the brain to be maintained as TRM cells.


Asunto(s)
Encéfalo/inmunología , Encéfalo/virología , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Memoria Inmunológica , Activación de Linfocitos/inmunología , Muromegalovirus/fisiología , Linfocitos T Reguladores/inmunología , Antígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Citocinas/biosíntesis , Granzimas/biosíntesis , Humanos , Depleción Linfocítica , Linfocitos T Reguladores/citología
15.
Glia ; 62(10): 1582-94, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24890099

RESUMEN

Engagement of the programmed death (PD)-1 receptor on activated cells by its ligand (PD-L1) is a mechanism for suppression of activated T-lymphocytes. Microglia, the resident inflammatory cells of the brain, are important for pathogen detection and initiation of innate immunity, however, a novel role for these cells as immune regulators has also emerged. PD-L1 on microglia has been shown to negatively regulate T-cell activation in models of multiple sclerosis and acute viral encephalitis. In this study, we investigated the role of glial cell PD-L1 in controlling encephalitogenic CD8(+) T-lymphocytes, which infiltrate the brain to manage viral infection, but remain to produce chronic neuroinflammation. Using a model of chronic neuroinflammation following murine cytomegalovirus (MCMV)-induced encephalitis, we found that CD8(+) T-cells persisting within the brain expressed PD-1. Conversely, activated microglia expressed PD-L1. In vitro, primary murine microglia, which express low basal levels of PD-L1, upregulated the co-inhibitory ligand on IFN-γ-treatment. Blockade of the PD-1: PD-L1 pathway in microglial: CD8(+) T-cell co-cultures increased T-cell IFN-γ and interleukin (IL)-2 production. We observed a similar phenomenon following blockade of this co-inhibitory pathway in astrocyte: CD8(+) T-cell co-cultures. Using ex vivo cultures of brain leukocytes, including microglia and CD8(+) T-cells, obtained from mice with MCMV-induced chronic neuroinflammation, we found that neutralization of either PD-1 or PD-L1 increased IFN-γ production from virus-specific CD8(+) T-cells stimulated with MCMV IE1168-176 peptide. These data demonstrate that microglia and astrocytes control antiviral T-cell responses and suggest a therapeutic potential of PD1: PD-L1 modulation to manage the deleterious consequences of uncontrolled neuroinflammation.


Asunto(s)
Astrocitos/fisiología , Antígeno B7-H1/metabolismo , Linfocitos T CD8-positivos/fisiología , Encefalitis/fisiopatología , Microglía/fisiología , Receptor de Muerte Celular Programada 1/metabolismo , Animales , Encéfalo/fisiopatología , Enfermedad Crónica , Técnicas de Cocultivo , Citomegalovirus , Infecciones por Citomegalovirus , Modelos Animales de Enfermedad , Femenino , Interferón gamma/metabolismo , Interleucina-2/metabolismo , Leucocitos/fisiología , Ratones Endogámicos BALB C , Neuroinmunomodulación/fisiología , Transducción de Señal
16.
PLoS One ; 8(10): e77577, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24147028

RESUMEN

Despite the therapeutic impact of anti-retroviral therapy, HIV-1-associated neurocognitive disorder (HAND) remains a serious threat to AIDS patients, and there currently remains no specific therapy for the neurological manifestations of HIV-1. Recent work suggests that the nigrostriatal dopaminergic area is a critical brain region for the neuronal dysfunction and death seen in HAND and that human dopaminergic neurons have a particular sensitivity to gp120-induced damage, manifested as reduced function (decreased dopamine uptake), morphological changes, and reduced viability. Synthetic cannabinoids inhibit HIV-1 expression in human microglia, suppress production of inflammatory mediators in human astrocytes, and there is substantial literature demonstrating the neuroprotective properties of cannabinoids in other neuropathogenic processes. Based on these data, experiments were designed to test the hypothesis that synthetic cannabinoids will protect dopaminergic neurons against the toxic effects of the HIV-1 protein gp120. Using a human mesencephalic neuronal/glial culture model, which contains dopaminergic neurons, microglia, and astrocytes, we were able to show that the CB1/CB2 agonist WIN55,212-2 blunts gp120-induced neuronal damage as measured by dopamine transporter function, apoptosis and lipid peroxidation; these actions were mediated principally by the CB2 receptor. Adding supplementary human microglia to our cultures enhances gp120-induced damage; WIN55,212-2 is able to alleviate this enhanced damage. Additionally, WIN55,212-2 inhibits gp120-induced superoxide production by purified human microglial cells, inhibits migration of human microglia towards supernatants generated from gp120-stimulated human mesencephalic neuronal/glial cultures and reduces chemokine and cytokine production from the human mesencephalic neuronal/glial cultures. These data suggest that synthetic cannabinoids are capable of protecting human dopaminergic neurons from gp120 in a variety of ways, acting principally through the CB2 receptors and microglia.


Asunto(s)
Benzoxazinas/farmacología , Neuronas Dopaminérgicas/efectos de los fármacos , Proteína gp120 de Envoltorio del VIH/farmacología , Morfolinas/farmacología , Naftalenos/farmacología , Fármacos Neuroprotectores/farmacología , Receptor Cannabinoide CB2/agonistas , Proteínas Recombinantes/farmacología , Apoptosis/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Citocinas/biosíntesis , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Humanos , Peroxidación de Lípido , Mesencéfalo/citología , Microglía/efectos de los fármacos , Microglía/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Receptor Cannabinoide CB2/metabolismo , Superóxidos/metabolismo
17.
Neurochem Res ; 38(10): 2148-59, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23918204

RESUMEN

Reactive oxygen species (ROS) have been shown to be a contributor to aging and disease. ROS also serve as a trigger switch for signaling cascades leading to corresponding cellular and molecular events. In the central nervous system (CNS), microglial cells are likely the main source of ROS production. However, activated astrocytes also appear to be capable of generating ROS. In this study we investigated ROS production in human astrocytes stimulated with interleukin (IL)-1ß and interferon (IFN)-γ and its potential harmful effects. Although IFN-γ alone had no effect, it potentiated IL-1ß-induced ROS production in a time-dependent manner. One of the sources of ROS in IL-1ß-activated astrocytes was from increased superoxide production in mitochondria accompanied by enhanced manganese superoxide dismutase and inhibited catalase expression. NADPH oxidase (NOX) may also contribute to ROS production as astrocytes express NOX isoforms. Glutamate uptake, which represents one of the most important methods of astrocytes to prevent excitotoxicity, was down-regulated in IL-1ß-activated astrocytes, and was further suppressed in the presence of IFN-γ; IFN-γ itself exerted minimal effect. Elevated levels of 8-isoprostane in IL-1ß ± IFN-γ-activated human astrocytes indicate downstream lipid peroxidation. Pretreatment with diphenyleneiodonium abolished the IL-1ß ± IFN-γ-induced ROS production, restored glutamate uptake function and reduced 8-isoprostane to near control levels suggesting that ROS contributes to the dysfunction of activated astrocytes. These results support the notion that dampening activated human astrocytes to maintain the redox homeostasis is vital to preserve their neuroprotective potential in the CNS.


Asunto(s)
Astrocitos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Astrocitos/efectos de los fármacos , Células Cultivadas , Dinoprost/análogos & derivados , Dinoprost/metabolismo , Sinergismo Farmacológico , Ácido Glutámico/metabolismo , Humanos , Interferón gamma/farmacología , Interleucina-1beta/farmacología , Peroxidación de Lípido/efectos de los fármacos , Mitocondrias/metabolismo , NADPH Oxidasas/metabolismo , Compuestos Onio/farmacología , Oxidación-Reducción
18.
J Neuroinflammation ; 8: 123, 2011 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-21943001

RESUMEN

BACKGROUND: Production of reactive oxygen species (ROS) and proinflammatory cytokines by microglial cells in response to viral brain infection contributes to both pathogen clearance and neuronal damage. In the present study, we examined the effect of herpes simplex virus (HSV)-1-induced, NADPH oxidase-derived ROS in activating mitogen-activated protein kinases (MAPKs) as well as driving cytokine and chemokine expression in primary murine microglia. METHODS: Oxidation of 2', 7'-dichlorodihydrofluorescin diacetate (H2DCFDA) was used to measure production of intracellular ROS in microglial cell cultures following viral infection. Virus-induced cytokine and chemokine mRNA and protein levels were assessed using real-time RT-PCR and ELISA, respectively. Virus-induced phosphorylation of microglial p38 and p44/42 (ERK1/2) MAPKs was visualized using Western Blot, and levels of phospho-p38 were quantified using Fast Activated Cell-based ELISA (FACE assay). Diphenyleneiodonium (DPI) and apocynin (APO), inhibitors of NADPH oxidases, were used to investigate the role of virus-induced ROS in MAPK activation and cytokine, as well as chemokine, production. RESULTS: Levels of intracellular ROS were found to be highly elevated in primary murine microglial cells following infection with HSV and the majority of this virus-induced ROS was blocked following DPI and APO treatment. Correspondingly, inhibition of NADPH oxidase also decreased virus-induced proinflammatory cytokine and chemokine production. In addition, microglial p38 and p44/42 MAPKs were found to be phosphorylated in response to viral infection and this activation was also blocked by inhibitors of NADPH oxidase. Finally, inhibition of either of these ROS-induced signaling pathways suppressed cytokine (TNF-α and IL-1ß) production, while chemokine (CCL2 and CXCL10) induction pathways were sensitive to inhibition of p38, but not ERK1/2 MAPK. CONCLUSIONS: Data presented herein demonstrate that HSV infection induces proinflammatory responses in microglia through NADPH oxidase-dependent ROS and the activation of MAPKs.


Asunto(s)
Citocinas/inmunología , Herpesvirus Humano 1/inmunología , Microglía/inmunología , Microglía/virología , NADPH Oxidasas/inmunología , Especies Reactivas de Oxígeno/metabolismo , Animales , Células Cultivadas , Quimiocinas/genética , Quimiocinas/inmunología , Citocinas/genética , Activación Enzimática , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C , Microglía/citología , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/inmunología , NADPH Oxidasas/antagonistas & inhibidores , Oxidación-Reducción , Transducción de Señal/inmunología
19.
J Neuroimmune Pharmacol ; 6(4): 528-39, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21850403

RESUMEN

Anti-retroviral therapy (ART) has had a tremendous impact on the clinical outcomes of HIV-1 infected individuals. While ART has produced many tangible benefits, chronic, long-term consequences of HIV infection have grown in importance. HIV-1-associated neurocognitive disorder (HAND) represents a collection of neurological syndromes that have a wide range of functional cognitive impairments. HAND remains a serious threat to AIDS patients, and there currently remains no specific therapy for the neurological manifestations of HIV-1. Based upon work in other models of neuroinflammation, kappa opioid receptors (KOR) and synthetic cannabinoids have emerged as having neuroprotective properties and the ability to dampen pro-inflammatory responses of glial cells; properties that may have a positive influence in HIV-1 neuropathogenesis. The ability of KOR ligands to inhibit HIV-1 production in human microglial cells and CD4 T lymphocytes, demonstrate neuroprotection, and dampen chemokine production in astrocytes provides encouraging data to suggest that KOR ligands may emerge as potential therapeutic agents in HIV neuropathogenesis. Based upon findings that synthetic cannabinoids inhibit HIV-1 expression in human microglia and suppress production of inflammatory mediators such as nitric oxide (NO) in human astrocytes, as well as a substantial literature demonstrating neuroprotective properties of cannabinoids in other systems, synthetic cannabinoids have also emerged as potential therapeutic agents in HIV neuropathogenesis. This review focuses on these two classes of compounds and describes the immunomodulatory and neuroprotective properties attributed to each in the context of HIV neuropathogenesis.


Asunto(s)
Complejo SIDA Demencia/inmunología , Analgésicos Opioides/farmacología , Cannabinoides/farmacología , VIH-1/efectos de los fármacos , Neuroinmunomodulación/inmunología , Fármacos Neuroprotectores/farmacología , Complejo SIDA Demencia/prevención & control , Analgésicos Opioides/inmunología , Animales , Cannabinoides/inmunología , Humanos , Fármacos Neuroprotectores/inmunología , Receptores Opioides kappa/inmunología
20.
Curr Opin Obstet Gynecol ; 23(2): 109-16, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21386681

RESUMEN

PURPOSE OF REVIEW: To review the potential of stem cells derived from amniotic fluid and applications in prenatal and postnatal therapy. RECENT FINDINGS: We have recently described that pluripotent stem cells can be isolated from amniotic fluid defined as amniotic fluid stem (AFS) cells by selection for expression of the membrane stem cell factor receptor c-Kit. AFS cells maintained for over 250 population doublings retained long telomeres and normal karyotype. Clonal human lines verified by retroviral marking were induced to differentiate into cell types representing each embryonic germ layer, including adipogenic, osteogenic, myogenic, endothelial, neuronal, and hepatic lineages. Rat AFS cells have been able to improve the repair of damaged smooth muscle in cryoinjury bladders. Furthermore, AFS cells could be differentiated toward cardiomyogenic lineages, when co-cultured with neonatal cardiomyocytes and have potential to generate hematopoietic lineages both in vitro and in vivo. These cells have been applied into fetal therapy, and widely used for tissue repair in animal models. Finally, we demonstrated a feasible way to do in-utero autologous AFS transplantation in sheep. SUMMARY: Stem cells derived from amniotic fluid are a relatively new source of cells that could have a therapeutic value in various diseases prenatally and/or postnatally.


Asunto(s)
Líquido Amniótico/citología , Líquido Amniótico/metabolismo , Células Madre/citología , Animales , Linaje de la Célula , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Femenino , Células Madre Hematopoyéticas/citología , Humanos , Cariotipificación , Células Madre Mesenquimatosas/citología , Ratones , Embarazo , Ratas , Retroviridae/metabolismo , Ovinos , Telómero/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA