Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Cell Transplant ; 31: 9636897221136149, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36367048

RESUMO

CD8 T cells play a key role in cancer immunotherapy and allograft rejection. However, it is not clear how they kill cells and tissues that do not have the agonist peptide-major histocompatibility complex (MHC) on their surface, as in the settings of MHC class I deficient tumors and indirect rejection of MHC-mismatched transplants. CD8 T cells might respond to agonist antigen cross-presented on hematopoietic cells, leading to a "bystander" rejection. Alternatively, they may recognize agonist antigen cross-presented on recipient endothelial cells and kill the tissue's vital blood supply. The latter mechanism predicts that all non-vascularized grafts, grafts dependent on in-growth of recipient blood vessels, will be susceptible to CD8 T cell mediated indirect rejection. In contrast, we show here that non-vascularized transplants, bearing the same agonist antigen, are not universally susceptible to this rejection pathway. Non-vascularized skin, but not islet or heart tissue transplants were indirectly rejected by CD8 T cells. Furthermore, CD8 T cells were able to indirectly reject skin grafts when recipient MHC class I expression was restricted to bone marrow derived cells but not when it was restricted to radioresistant cells (e.g. endothelial cells). These findings argue against a major role for endothelial cell cross-presentation in killing of tissue that does not present the agonist peptide-MHC class I. Instead, the data suggests that cross-presentation by recipient hematopoietic cells underlies the CD8 T cell mediated killing of tissue that is unable to directly present the target peptide-MHC class I.


Assuntos
Medula Óssea , Células Endoteliais , Transplante Homólogo , Rejeição de Enxerto , Linfócitos T CD8-Positivos , Complexo Principal de Histocompatibilidade , Antígenos de Histocompatibilidade Classe I , Peptídeos , Linfócitos T CD4-Positivos
2.
J Control Release ; 338: 557-570, 2021 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-34474072

RESUMO

Nanoparticles (NPs) coated with autoimmune disease-relevant peptide-major histocompatibility complexes (pMHCs) can blunt autoimmune diseases by re-programming cognate effector T-lymphocytes into disease-suppressing regulatory T-cells, followed by massive expansion. Here, a method to quantify the absolute amounts of the active drug product is developed, to understand the relationship between bioavailability and pharmacodynamics. Incubation with plasma results in the formation of a protein corona that stabilizes the directional pMHC coat, shielding it from proteolysis or anti-drug antibody recognition, without any appreciable loss in biological potency. A quantitative method that harnesses these features indicates that the half-life of these compounds in the circulation and organs is an order of magnitude shorter (minutes vs. hours) than that measured using commonly-used semi-quantitative methods. Extensive transmission electron microscopy-based organ scanning and flow cytometry-based enumeration of pMHCII-NP capturing cells confirmed that these compounds are rapidly captured (within 1 min) by liver sinusoidal endothelial cells, Kupffer cells, splenic phagocytes and cognate T-cells, leading to a fast decline in the circulation. Therefore, the powerful pharmacodynamic effects of these compounds are dissociated from long bioavailability, implying a hit-and-run event. Collectively, these data provide a detailed view of the life-cycle of a nanoimmunomedicine, and suggest that the real half-lives of intact nanomedicines may be much shorter than those estimated using indirect approaches.


Assuntos
Doenças Autoimunes , Nanomedicina , Autoantígenos , Disponibilidade Biológica , Células Endoteliais , Humanos
3.
J Clin Invest ; 130(4): 1823-1829, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32125290

RESUMO

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


Assuntos
Autoimunidade , Encefalomielite Autoimune Experimental/imunologia , Hepatite Autoimune/imunologia , Fígado/imunologia , Linfócitos T Reguladores/imunologia , Animais , Autoantígenos/imunologia , Encefalomielite Autoimune Experimental/patologia , Hepatite Autoimune/patologia , Antígenos de Histocompatibilidade Classe II/imunologia , Fígado/patologia , Camundongos , Camundongos Endogâmicos NOD , Linfócitos T Reguladores/patologia
4.
Nat Commun ; 10(1): 4917, 2019 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-31664029

RESUMO

Assembly of soluble peptide-major histocompatibility complex class II (pMHCII) monomers into multimeric structures enables the detection of antigen-specific CD4+ T cells in biological samples and, in some configurations, their reprogramming in vivo. Unfortunately, current MHCII-αß chain heterodimerization strategies are typically associated with low production yields and require the use of foreign affinity tags for purification, precluding therapeutic applications in humans. Here, we show that fusion of peptide-tethered or empty MHCII-αß chains to the IgG1-Fc mutated to form knob-into-hole structures results in the assembly of highly stable pMHCII monomers. This design enables the expression and rapid purification of challenging pMHCII types at high yields without the need for leucine zippers and purification affinity tags. Importantly, this design increases the antigen-receptor signaling potency of multimerized derivatives useful for therapeutic applications and facilitates the detection and amplification of low-avidity T cell specificities in biological samples using flow cytometry.


Assuntos
Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/metabolismo , Animais , Dimerização , Antígenos de Histocompatibilidade Classe II/química , Camundongos , Camundongos Endogâmicos NOD , Peptídeos/genética , Peptídeos/metabolismo , Engenharia de Proteínas , Solubilidade , Linfócitos T/metabolismo
5.
ACS Nano ; 12(11): 10621-10635, 2018 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-30481968

RESUMO

Vaccination using nanocarrier-based delivery systems has recently emerged as a promising approach for meeting the continued challenge posed by infectious diseases and cancer. A diverse portfolio of nanocarriers of various sizes, compositions, and physical parameters have now been developed, and this diversity provides an opportunity for the rational design of vaccines that can mediate targeted delivery of various antigens and adjuvants or immune regulatory agents in ways unachievable with classical vaccination approaches. This flexibility allows control over the characteristics of vaccine-elicited immune responses such that they can be tailored to be effective in circumstances where classical vaccines have failed. Furthermore, the utility of nanocarrier-based immune modulation extends to the treatment of autoimmune disease where precisely targeted inhibition of immune responses is desirable. Clearly, the selection of appropriate nanocarriers, antigens, adjuvants, and other components underpins the efficacy of these nanoimmune interventions. Herein, we provide an overview of currently available nanocarriers of various types and their physical and pharmacological properties with the goal of providing a resource for researchers exploring nanomaterial-based approaches for immune modulation and identify some information gaps and unexplored questions to help guide future investigation.


Assuntos
Doenças Autoimunes/imunologia , Autoimunidade/imunologia , Fatores Imunológicos/imunologia , Infecções/imunologia , Nanopartículas/química , Neoplasias/imunologia , Animais , Doenças Autoimunes/terapia , Portadores de Fármacos/química , Humanos , Fatores Imunológicos/química , Infecções/terapia , Neoplasias/terapia
6.
Front Immunol ; 9: 12, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29416537

RESUMO

Lymphopenia can result from various factors, including viral infections, clinical interventions, or as a normal property of the fetal/neonatal period. T cells in a lymphopenic environment undergo lymphopenia-induced proliferation (LIP) to fill the available "niche" as defined by peptide-MHC and homeostatic cytokine resources. We recently reported systemic autoimmunity following reconstitution of the lymphoid compartment of Rag1-/- mice with PD-1-/- hematopoietic stem cells or by transfer of thymocytes, but not splenocytes, suggesting that programmed death-1 (PD-1) plays a crucial role in controlling recent thymic emigrants (RTE) and preventing autoimmunity upon their LIP. However, it is unclear whether RTE residing within the periphery of a lymphoreplete host maintain enhanced autoimmune generating potential or if this property only manifests if RTE experience a lymphopenic periphery immediately after export from the thymus. Furthermore, it is unclear which of a variety of T cell effector mechanisms generate pathology when control of RTE by PD-1 is lacking. Herein, we determined that PD-1 is upregulated on CD4 T cells undergoing the natural LIP characteristic of the neonatal period. Newly generated T cells lacking PD-1 maintained an enhanced autoimmune potential even after residence in a lymphoreplete periphery, emphasizing the importance of PD-1 in the establishment of peripheral tolerance. Neither Fas nor perforin-dependent killing mechanisms were required for autoimmunity, while host MHC-II expression was critical, suggesting that LIP-driven autoimmunity in the absence of PD-1 may primarily result from a CD4 T cell-mediated systemic cytokinemia, a feature potentially shared by other autoimmune or inflammatory syndromes associated with immune reconstitution and LIP.


Assuntos
Autoimunidade , Linfócitos T CD4-Positivos/imunologia , Perforina/imunologia , Tolerância Periférica , Receptor de Morte Celular Programada 1/imunologia , Receptor fas/imunologia , Animais , Diabetes Mellitus Experimental/cirurgia , Feminino , Transplante das Ilhotas Pancreáticas , Masculino , Camundongos
7.
Front Immunol ; 8: 1289, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29075267

RESUMO

Recovery of the T lymphocyte compartment within a lymphopenic host by lymphopenia-induced proliferation (LIP) is regulated by inter- and intraclonal competition for limited resources, including homeostatic cytokines and peptide:MHC (pMHC) complexes with which the TCR can interact at least weakly to yield a tonic signal. Importantly, the process of LIP can synergize with other factors that promote T cell activation to drive inflammatory disease. While reconstitution of the lymphoid compartment of immune deficient Rag-/- mice by transfer of wild-type hematopoietic stem cells (HSC) does not generally result in an overt disease phenotype, transfer of HSC deficient in expression of the co-inhibitory molecule PD-1 results in severe systemic autoimmunity driven by newly generated T cells that emerge from the thymus into the periphery and undergo LIP. Importantly, autoimmunity does not appear to depend on a response to exogenous (i.e., gut flora-derived) antigens. PD-1 is well known to be upregulated during T cell activation in response to cognate antigens, but it is unclear whether PD-1 has a role in controlling LIP of T cells in the absence of cognate antigen, i.e., in response to tonic pMHC. We examined whether PD-1 controls LIP of newly generated T cells by controlling the response to tonic pMHC or the homeostatic cytokine IL-7. We found that PD-1-deficient T cells have a proliferative advantage over WT T cells during LIP and this effect is MHC-II dependent and independent of IL-7Rα signaling. Furthermore, our data suggest that signals through IL-7Rα can be dispensable for LIP and may instead be of increased importance for T cell survival in conditions of high competition for limited pMHC (e.g., post-LIP, in a lymphoreplete host). We hypothesize that autoimmunity post-PD-1-/- HSC transplant is the result of an overzealous T cell response to normally tonic self-pMHC precipitated by the synergy of LIP and PD-1 deficiency. Furthermore, potentiation of TCR signals in response to normally tonic self-pMHC may contribute to the success of PD-1 blockade in cancer immunotherapy.

8.
J Immunol ; 198(7): 2534-2541, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28320914

RESUMO

Lymphopenia-induced proliferation (LIP) occurs when resources for T cell survival in a host are in excess. LIP has been associated with the development of inflammatory disease in situations where an additional disease-predisposing cofactor is present during LIP. This has led to the view of LIP-driven autoimmunity as a two hit model; however, not all cofactors have equal ability to precipitate autoimmunity and we have recently shown that in some circumstances, such as the absence of the coinhibitory molecule PD-1, additional hits are required. Herein we review factors controlling LIP, including coinhibitory molecules and other attenuators of TCR signaling, with a focus on their contribution to LIP-driven autoimmunity. Rather than viewing LIP-associated autoimmunity as an n-hit model, we suggest a more quantitative view of lymphopenia with respect to the factors that promote LIP as a tool to predict autoimmune potential and to inform tumor immunotherapy approaches.


Assuntos
Autoimunidade/imunologia , Proliferação de Células/fisiologia , Linfócitos T/imunologia , Animais , Humanos , Ativação Linfocitária/imunologia , Linfopenia/imunologia
9.
Eur J Immunol ; 44(12): 3560-72, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25236923

RESUMO

The expression of the coinhibitor PD-1 on T cells is important for the establishment of immune homeostasis. We previously found that PD-1 is particularly critical for the control of self-tolerance during lymphopenia-induced proliferation of recent thymic emigrants (RTEs). Previous studies suggested that PD-1 modulates the generation of Treg cells, particularly peripherally induced Treg (pTreg) cells, and controls Th17 cells. However, these conclusions were derived indirectly from studies on the ligand PD-L1, and not PD-1 itself. Herein we directly tested whether T-cell PD-1 expression was needed for Treg cell generation and examined if a paucity of Treg cells or enhanced Th17 cells could explain the severe lymphopenia-potentiated autoimmunity caused by PD-1 KO RTEs. Employing the murine FoxP3(EGFP) reporter system to simultaneously monitor conversion of WT and PD-1 KO T cells to pTreg cells in the same animal, we found that PD-1 deficiency did not inhibit pTreg cell generation or lead to Th17-cell-mediated autoimmunity. Surprisingly, pTreg cell numbers were increased in PD-1 KO versus WT cell populations. Furthermore, we noted an increased conversion to pTreg cells by RTEs. Our data suggest that the primary role for PD-1 is to restrain T-cell activation/proliferation to self-Ags rather than promote generation of Treg cells.


Assuntos
Proliferação de Células/fisiologia , Ativação Linfocitária/fisiologia , Receptor de Morte Celular Programada 1/imunologia , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Animais , Autoantígenos/genética , Autoantígenos/imunologia , Antígeno B7-H1/genética , Antígeno B7-H1/imunologia , Contagem de Linfócitos , Linfopenia/imunologia , Camundongos , Camundongos Knockout , Receptor de Morte Celular Programada 1/genética , Linfócitos T Reguladores/citologia , Células Th17/citologia
10.
PLoS One ; 9(7): e97984, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24988390

RESUMO

Human endogenous retroviruses (HERVs) are differentially expressed depending on the cell type and physiological circumstances. HERV-K has been implicated in the pathogenesis of several diseases although the functional consequences of its expression remain unknown. Human immunodeficiency virus (HIV) infection causes neuroinflammation with neuronal damage and death. Herein, we investigated HERV-K(II)/(HML-2) envelope (Env) expression and its actions in the brain during HIV/AIDS. HERV-K(II) Env expression was assessed in healthy brain tissues, autopsied HIV HIV- infected (HIV+) and uninfected (HIV-) brains and in neural cell cultures by real time RT-PCR, massively parallel (deep) sequencing, immunoblotting and immunohistochemistry. Neuronal and neural stem cells expressing HERV-K(II) Env were analyzed in assays of host responses including cellular viability, immune responses and neurobehavioral outcomes. Deep sequencing of human brain transcriptomes disclosed that RNA sequences encoded by HERV-K were among the most abundant HERV sequences detected in human brain. Comparison of different cell types revealed that HERV-K(II) env RNA abundance was highest in cultured human neurons but was suppressed by epidermal growth factor exposure. HERV-K(II) Env immunoreactivity was increased in the cerebral cortex from persons with HIV/AIDS, principally localized in neurons. Human neuronal cells transfected with HERV-K(II) Env exhibited increased NGF and BDNF expression. Expression of HERV-K(II) Env in neuronal cells increased cellular viability and prevented neurotoxicity mediated by HIV-1 Vpr. Intracerebral delivery of HERV-K(II) Env expressed by neural stem cells suppressed TNF-α expression and microglial activation while also improving neurobehavioral deficits in vpr/RAG1-/- mice. HERV-K(II) Env was highly expressed in human neurons, especially during HIV/AIDS, but in addition exerted neuroprotective effects. These findings imply that HERV gene products might exert adaptive effects in circumstances of pathophysiological stress, perhaps underlying the conservation of HERVs within the human genome.


Assuntos
Retrovirus Endógenos/metabolismo , HIV/metabolismo , Neurônios/metabolismo , Proteínas do Envelope Viral/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Células Cultivadas , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Córtex Cerebral/virologia , Fator de Crescimento Epidérmico/farmacologia , HIV/patogenicidade , Sequenciamento de Nucleotídeos em Larga Escala , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Fator de Crescimento Neural/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/virologia , Neurônios/citologia , Neurônios/virologia , Análise de Sequência de DNA , Regulação para Cima/efeitos dos fármacos , Proteínas do Envelope Viral/genética , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/genética , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/metabolismo
11.
J Immunol ; 191(4): 1686-91, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23851694

RESUMO

An ongoing dilemma faced during an immune response is generating an effective, often proinflammatory response to eliminate pathogens and/or infected cells while also minimizing collateral damage to adjacent noninfected tissues. The factors limiting bystander cell injury during an Ag-specific immune response in vivo are largely unknown. In this study, using an in vivo model of islet transplants in TCR transgenic mice, we show that both CD4 and CD8 T cells do have the capacity to inflict adjacent tissue damage and that this injury is greatly enhanced in sensitized hosts. CD4 T cell-mediated killing of specific and bystander cells occurred via different mechanisms. Unlike specific target cell killing, CD4-mediated bystander injury required tissue Fas expression and was inhibited with anti-IFN-γ Ab treatment in vivo. Moreover, bystander cell injury was not entirely nonspecific but rather required, in naive recipients, that the MHC allele expressed by the bystanders was self. Importantly, the coinhibitor programmed death-1 plays an important role in restraining bystander cell injury mediated either by defined TCR transgenic T cells or by polyclonal T cell populations. Thus, the differential requirements for specific versus bystander cell injury suggest that there are opportunities for inhibiting immune pathology without compromising Ag-specific immunity in vivo.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Imunidade Celular , Transplante das Ilhotas Pancreáticas/imunologia , Subpopulações de Linfócitos T/imunologia , Animais , Efeito Espectador/imunologia , Citotoxicidade Imunológica , Diabetes Mellitus Experimental/cirurgia , Feminino , Antígenos H-2/imunologia , Imunização , Inflamação , Interferon gama/antagonistas & inibidores , Interferon gama/fisiologia , Transplante das Ilhotas Pancreáticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Modelos Imunológicos , Ovalbumina/imunologia , Receptor de Morte Celular Programada 1 , Receptor fas/imunologia
12.
PLoS One ; 8(1): e54673, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23355888

RESUMO

The brain is assumed to be a sterile organ in the absence of disease although the impact of immune disruption is uncertain in terms of brain microbial diversity or quantity. To investigate microbial diversity and quantity in the brain, the profile of infectious agents was examined in pathologically normal and abnormal brains from persons with HIV/AIDS [HIV] (n = 12), other disease controls [ODC] (n = 14) and in cerebral surgical resections for epilepsy [SURG] (n = 6). Deep sequencing of cerebral white matter-derived RNA from the HIV (n = 4) and ODC (n = 4) patients and SURG (n = 2) groups revealed bacterially-encoded 16 s RNA sequences in all brain specimens with α-proteobacteria representing over 70% of bacterial sequences while the other 30% of bacterial classes varied widely. Bacterial rRNA was detected in white matter glial cells by in situ hybridization and peptidoglycan immunoreactivity was also localized principally in glia in human brains. Analyses of amplified bacterial 16 s rRNA sequences disclosed that Proteobacteria was the principal bacterial phylum in all human brain samples with similar bacterial rRNA quantities in HIV and ODC groups despite increased host neuroimmune responses in the HIV group. Exogenous viruses including bacteriophage and human herpes viruses-4, -5 and -6 were detected variably in autopsied brains from both clinical groups. Brains from SIV- and SHIV-infected macaques displayed a profile of bacterial phyla also dominated by Proteobacteria but bacterial sequences were not detected in experimentally FIV-infected cat or RAG1⁻/⁻ mouse brains. Intracerebral implantation of human brain homogenates into RAG1⁻/⁻ mice revealed a preponderance of α-proteobacteria 16 s RNA sequences in the brains of recipient mice at 7 weeks post-implantation, which was abrogated by prior heat-treatment of the brain homogenate. Thus, α-proteobacteria represented the major bacterial component of the primate brain's microbiome regardless of underlying immune status, which could be transferred into naïve hosts leading to microbial persistence in the brain.


Assuntos
Síndrome da Imunodeficiência Adquirida , Alphaproteobacteria , Infecções Bacterianas do Sistema Nervoso Central , Cérebro , RNA Bacteriano , RNA Ribossômico , Síndrome da Imunodeficiência Adquirida/complicações , Síndrome da Imunodeficiência Adquirida/genética , Síndrome da Imunodeficiência Adquirida/metabolismo , Síndrome da Imunodeficiência Adquirida/patologia , Alphaproteobacteria/genética , Alphaproteobacteria/metabolismo , Animais , Autopsia , Gatos , Infecções Bacterianas do Sistema Nervoso Central/etiologia , Infecções Bacterianas do Sistema Nervoso Central/genética , Infecções Bacterianas do Sistema Nervoso Central/metabolismo , Infecções Bacterianas do Sistema Nervoso Central/microbiologia , Infecções Bacterianas do Sistema Nervoso Central/patologia , Cérebro/metabolismo , Cérebro/microbiologia , Cérebro/patologia , Síndrome de Imunodeficiência Adquirida Felina/genética , Síndrome de Imunodeficiência Adquirida Felina/metabolismo , Síndrome de Imunodeficiência Adquirida Felina/microbiologia , Síndrome de Imunodeficiência Adquirida Felina/patologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Neuroglia/metabolismo , Neuroglia/microbiologia , Neuroglia/patologia , RNA Bacteriano/genética , RNA Bacteriano/metabolismo , RNA Ribossômico/genética , RNA Ribossômico/metabolismo
13.
J Immunol ; 187(9): 4788-99, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21964030

RESUMO

Endoplasmic reticulum (ER) stress is a homeostatic mechanism, which is used by cells to adapt to intercellular and intracellular changes. Moreover, ER stress is closely linked to inflammatory pathways. We hypothesized that ER stress is an integral component of neuroinflammation and contributes to the development of neurological diseases. In autopsied brain specimens from multiple sclerosis (MS) and non-MS patients, XBP-1 spliced variant (XBP-1/s) was increased in MS brains (p < 0.05) and was correlated with the expression of the human endogenous retrovirus-W envelope transcript, which encodes the glycoprotein, Syncytin-1 (p < 0.05). In primary human fetal astrocytes transfected with a Syncytin-1-expressing plasmid, XBP-1/s, BiP, and NOS2 were induced, which was suppressed by crocin treatment (p < 0.05). Crocin also protected oligodendrocytes exposed to cytotoxic supernatants derived from Syncytin-1-expressing astrocytes (p < 0.05) and NO-mediated oligodendrocytotoxicity (p < 0.05). During experimental autoimmune encephalomyelitis (EAE), the transcript levels of the ER stress genes XBP-1/s, BiP, PERK, and CHOP were increased in diseased spinal cords compared with healthy littermates (p < 0.05), although CHOP expression was not involved in the EAE disease phenotype. Daily treatment with crocin starting on day 7 post-EAE induction suppressed ER stress and inflammatory gene expression in spinal cords (p < 0.05), which was accompanied by preserved myelination and axonal density, together with reduced T cell infiltration and macrophage activation. EAE-associated neurobehavioral deficits were also ameliorated by crocin treatment (p < 0.05). These findings underscored the convergent roles of pathogenic ER stress and immune pathways in neuroinflammatory disease and point to potential therapeutic applications for crocin.


Assuntos
Carotenoides/uso terapêutico , Doenças Desmielinizantes/prevenção & controle , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/patologia , Mediadores da Inflamação/uso terapêutico , Doenças Neurodegenerativas/prevenção & controle , Animais , Carotenoides/administração & dosagem , Células Cultivadas , Doenças Desmielinizantes/patologia , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/patologia , Retículo Endoplasmático/efeitos dos fármacos , Feminino , Sequestradores de Radicais Livres/uso terapêutico , Lobo Frontal/efeitos dos fármacos , Lobo Frontal/metabolismo , Lobo Frontal/patologia , Humanos , Mediadores da Inflamação/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Doenças Neurodegenerativas/patologia , Ratos , Ratos Sprague-Dawley
14.
Brain ; 134(Pt 9): 2703-21, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21908875

RESUMO

High-throughput technologies have led to advances in the recognition of disease pathways and their underlying mechanisms. To investigate the impact of micro-RNAs on the disease process in multiple sclerosis, a prototypic inflammatory neurological disorder, we examined cerebral white matter from patients with or without the disease by micro-RNA profiling, together with confirmatory reverse transcription-polymerase chain reaction analysis, immunoblotting and gas chromatography-mass spectrometry. These observations were verified using the in vivo multiple sclerosis model, experimental autoimmune encephalomyelitis. Brains of patients with or without multiple sclerosis demonstrated differential expression of multiple micro-RNAs, but expression of three neurosteroid synthesis enzyme-specific micro-RNAs (miR-338, miR-155 and miR-491) showed a bias towards induction in patients with multiple sclerosis (P < 0.05). Analysis of the neurosteroidogenic pathways targeted by micro-RNAs revealed suppression of enzyme transcript and protein levels in the white matter of patients with multiple sclerosis (P < 0.05). This was confirmed by firefly/Renilla luciferase micro-RNA target knockdown experiments (P < 0.05) and detection of specific micro-RNAs by in situ hybridization in the brains of patients with or without multiple sclerosis. Levels of important neurosteroids, including allopregnanolone, were suppressed in the white matter of patients with multiple sclerosis (P < 0.05). Induction of the murine micro-RNAs, miR-338 and miR-155, accompanied by diminished expression of neurosteroidogenic enzymes and allopregnanolone, was also observed in the brains of mice with experimental autoimmune encephalomyelitis (P < 0.05). Allopregnanolone treatment of the experimental autoimmune encephalomyelitis mouse model limited the associated neuropathology, including neuroinflammation, myelin and axonal injury and reduced neurobehavioral deficits (P < 0.05). These multi-platform studies point to impaired neurosteroidogenesis in both multiple sclerosis and experimental autoimmune encephalomyelitis. The findings also indicate that allopregnanolone and perhaps other neurosteroid-like compounds might represent potential biomarkers or therapies for multiple sclerosis.


Assuntos
MicroRNAs/metabolismo , Esclerose Múltipla/metabolismo , Neurotransmissores/biossíntese , 20-Hidroxiesteroide Desidrogenases/genética , 20-Hidroxiesteroide Desidrogenases/metabolismo , Anestésicos/farmacologia , Anestésicos/uso terapêutico , Animais , Células Cultivadas , Biologia Computacional , Encefalite/tratamento farmacológico , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Esclerose Múltipla/genética , Esclerose Múltipla/patologia , Neurotransmissores/genética , Oligodendroglia/citologia , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/fisiologia , Pregnanolona/farmacologia , Pregnanolona/uso terapêutico , Ratos , Ratos Sprague-Dawley
15.
PLoS One ; 6(4): e19176, 2011 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-21559469

RESUMO

BACKGROUND: The glycoprotein, Syncytin-1, is encoded by a human endogenous retrovirus (HERV)-W env gene and is capable of inducing neuroinflammation. The specific allele(s) responsible for Syncytin-1 expression in the brain is uncertain. Herein, HERV-W env diversity together with Syncytin-1 abundance and host immune gene profiles were examined in the nervous system using a multiplatform approach. RESULTS: HERV-W env sequences were encoded by multiple chromosomal encoding loci in primary human neurons compared with less chromosomal diversity in astrocytes and microglia (p<0.05). HERV-W env RNA sequences cloned from brains of patients with systemic or neurologic diseases were principally derived from chromosomal locus 7q21.2. Within the same specimens, HERV-W env transcript levels were correlated with the expression of multiple proinflammatory genes (p<0.05). Deep sequencing of brain transcriptomes disclosed the env transcripts to be the most abundant HERV-W transcripts, showing greater expression in fetal compared with healthy adult brain specimens. Syncytin-1's expression in healthy brain specimens was derived from multiple encoding loci and linked to distinct immune and developmental gene profiles. CONCLUSIONS: Syncytin-1 expression in the brain during disease was associated with neuroinflammation and was principally encoded by a full length provirus. The present studies also highlighted the diversity in HERV gene expression within the brain and reinforce the potential contributions of HERV expression to neuroinflammatory diseases.


Assuntos
Encéfalo/virologia , Retrovirus Endógenos/genética , Regulação da Expressão Gênica , Genes env , Adolescente , Adulto , Fatores Etários , Idoso , Alelos , Encéfalo/embriologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Produtos do Gene env/biossíntese , Variação Genética , Genoma Humano , Humanos , Sistema Imunitário , Inflamação , Masculino , Pessoa de Meia-Idade , Neurônios/patologia , Proteínas da Gravidez/biossíntese , Análise de Sequência de RNA
16.
Virology ; 404(2): 246-60, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20570310

RESUMO

Computational analysis of feline immunodeficiency virus (FIV) RNA sequences indicated that common FIV strains contain a rev response element (RRE) defined by a long unbranched hairpin with 6 stem-loop sub-domains, termed stem-loop A (SLA). To examine the role of the RNA secondary structure of the RRE, mutational analyses were performed in both an infectious FIV molecular clone and a FIV CAT-RRE reporter system. These studies disclosed that the stems within SLA (SA1, 2, 3, 4, and 5) of the RRE were critical but SA6 was not essential for FIV replication and CAT expression. These studies also revealed that the secondary structure rather than an antisense protein (ASP) mediates virus expression and replication in vitro. In addition, a single synonymous mutation within the FIV-RRE, SA3/45, reduced viral reverse transcriptase activity and p24 expression after transfection but in addition also showed a marked reduction in viral expression and production following infection.


Assuntos
Genes env/fisiologia , Vírus da Imunodeficiência Felina/metabolismo , Animais , Sequência de Bases , Gatos , Linhagem Celular , Regulação Viral da Expressão Gênica/fisiologia , Vírus da Imunodeficiência Felina/genética , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Estrutura Terciária de Proteína , RNA Viral/química , Replicação Viral/fisiologia
17.
FASEB J ; 24(6): 1799-812, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20097875

RESUMO

MicroRNAs (miRNAs) are small noncoding RNA molecules, which are known to regulate gene expression in physiological and pathological conditions. miRNA profiling was performed using brain tissue from patients with HIV encephalitis (HIVE), a neuroinflammatory/degenerative disorder caused by HIV infection of the brain. Microarray analysis showed differential expression of multiple miRNAs in HIVE compared to control brains. Target prediction and gene ontology enrichment analysis disclosed targeting of several gene families/biological processes by differentially expressed miRNAs (DEMs), with cell death-related genes, including caspase-6, showing a bias toward down-regulated DEMs. Consistent with the miRNA data, HIVE brains exhibited higher levels of caspase-6 transcripts compared with control patients. Immunohistochemical analysis showed localization of the cleaved form of caspase-6 in astrocytes in HIVE brain sections. Exposure of cultured human primary astrocytes to HIV viral protein R (Vpr) induced p53 up-regulation, loss of mitochondrial membrane potential, and caspase-6 activation followed by cell injury. Transgenic mice, expressing Vpr in microglial cells, demonstrated astrocyte apoptosis in brain, which was associated with caspase-6 activation and neurobehavioral abnormalities. Overall, these data point to previously unrecognized alterations in miRNA profile in the brain during HIV infection, which contribute to cell death through dysregulation of cell death machinery.


Assuntos
Complexo AIDS Demência/etiologia , Astrócitos/citologia , Encéfalo/metabolismo , Caspase 6/metabolismo , Infecções por HIV/complicações , MicroRNAs/fisiologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/fisiologia , Adulto , Animais , Astrócitos/metabolismo , Biomarcadores/metabolismo , Western Blotting , Encéfalo/citologia , Sinalização do Cálcio , Caspase 6/genética , Sobrevivência Celular , Feminino , Feto/citologia , Feto/metabolismo , Imunofluorescência , Perfilação da Expressão Gênica , Infecções por HIV/genética , HIV-1 , Humanos , Técnicas Imunoenzimáticas , Masculino , Potencial da Membrana Mitocondrial , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
J Immunol ; 184(3): 1566-74, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20042580

RESUMO

Lentivirus infections including HIV and feline immunodeficiency virus (FIV) cause neurovirulence, which is largely mediated by innate immunity. To investigate the interactions between neurovirulence and repeated conditioning by innate immune activation, models of lentivirus infection were exposed to LPS. Gene expression in HIV-infected (HIV+) and control (HIV-) patient brains was compared by real time RT-PCR and immunocytochemistry. Supernatants from mock and HIV-infected monocyte-derived macrophages exposed to LPS were applied to human neurons. FIV-infected (FIV+) and control (FIV-) animals were exposed repeatedly to LPS postinfection together with concurrent neurobehavioral testing, viral load, and host gene analyses. Brains from HIV+ individuals exhibited induction of CD3epsilon, CXCL10, and granzyme A expression (p < 0.05). Supernatants from HIV+ monocyte-derived macrophages induced CXCL10 expression in neurons, which was diminished by IL-10 treatment (p < 0.05). LPS-exposed FIV+ animals demonstrated lower plasma and brain viral loads (p < 0.05). Neuronal CXCL10 expression was increased in FIV+ animals but was suppressed by LPS exposure, together with reduced brain CD3epsilon and granzyme A expression (p < 0.05). In conjunction with preserved NeuN-positive neuronal counts in parietal cortex (p < 0.05), FIV+ animals exposed to LPS also showed less severe neurobehavioral deficits (p < 0.05). Repeated LPS exposures suppressed CXCL10 in the brain and ensuing T cell infiltration with a concomitant reduction in neurovirulence. Thus, innate immune chronic conditioning exerted beneficial effects on neurovirulence through suppression of a specific chemotactic factor, CXCL10, mediated by IL-10, leading to reduced leukocyte infiltration and release of neurotoxic factors.


Assuntos
Encéfalo/imunologia , Quimiocina CXCL10/antagonistas & inibidores , Infecções por HIV/imunologia , Vírus da Imunodeficiência Felina/imunologia , Vírus da Imunodeficiência Felina/patogenicidade , Interleucina-10/fisiologia , Lipopolissacarídeos/farmacologia , Fármacos Neuroprotetores/farmacologia , Animais , Encéfalo/patologia , Encéfalo/virologia , Gatos , Linhagem Celular Tumoral , Movimento Celular/imunologia , Células Cultivadas , Quimiocina CXCL10/fisiologia , Infecções por HIV/patologia , HIV-1/imunologia , Humanos , Vírus da Imunodeficiência Felina/crescimento & desenvolvimento , Infecções por Lentivirus/imunologia , Infecções por Lentivirus/patologia , Infecções por Lentivirus/prevenção & controle , Neurônios/imunologia , Neurônios/patologia , Neurônios/virologia , Carga Viral/imunologia , Virulência/imunologia
19.
Can J Neurol Sci ; 37 Suppl 2: S24-33, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21246932

RESUMO

The human microbiome is comprised of commensal and pathogenic microorganisms, which exert diverse effects in close proximity to the site of intection as well as in remote tissues through immune-mediated mechanisms. Multiple infectious agents have been implicated in the pathogenesis of multiple sclerosis (MS) with variable findings depending on the agent, techniques, and disease phenotype. Herein, the contributions of individual infectious agents to MS and their effects on the immune and nervous systems are reviewed, focusing on herpes viruses, coronaviruses, retroviruses, and synchronic infections. While infectious agents are often assumed to be pathogenic, their effects might also be beneficial to the host in the long-term, depending on age and the type of immunogen/pathogen exposure, as proposed by the hygiene hypothesis. The human microbiome has potential impact on future diagnostic and therapeutic issues in MS.


Assuntos
Viroses do Sistema Nervoso Central/complicações , Sistema Nervoso Central , Metagenoma , Esclerose Múltipla/microbiologia , Animais , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/microbiologia , Sistema Nervoso Central/patologia , Humanos , Metagenoma/genética , Modelos Biológicos , Esclerose Múltipla/genética , Esclerose Múltipla/patologia
20.
J Immunol ; 183(1): 298-309, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19542441

RESUMO

The rising incidence of autoimmune diseases such as multiple sclerosis (MS) in developed countries might be due to a more hygienic environment, particularly during early life. To investigate this concept, we developed a model of neonatal exposure to a common pathogen-associated molecular pattern, LPS, and determined its impact on experimental autoimmune encephalomyelitis (EAE). Mice exposed to LPS at 2 wk of age showed a delayed onset and diminished severity of myelin oligodendrocyte glycoprotein (MOG)-induced EAE, induced at 12 wk, compared with vehicle-exposed animals. Spinal cord transcript levels of CD3epsilon and F4/80 were lower in LPS- compared with PBS-exposed EAE animals with increased IL-10 levels in the LPS-exposed group. Splenic CD11c(+) cells from LPS-exposed animals exhibited reduced MHC class II and CD83 expression but increased levels of CD80 and CD86 both before and during EAE. MOG-treated APC from LPS-exposed animals stimulated less T lymphocyte proliferation but increased expansion of CD4(+)FoxP3(+) T cells compared with APC from PBS-exposed animals. Neuropathological studies disclosed reduced myelin and axonal loss in spinal cords from LPS-exposed compared with PBS-exposed animals with EAE, and this neuroprotective effect was associated with an increased number of CD3(+)FoxP3(+) immunoreactive cells. Analyses of human brain tissue revealed that FoxP3 expression was detected in lymphocytes, albeit reduced in MS compared with non-MS patients' brains. These findings support the concept of early-life microbial exposure influencing the generation of neuroprotective regulatory T cells and may provide insights into new immunotherapeutic strategies for MS.


Assuntos
Diferenciação Celular/imunologia , Células Dendríticas/imunologia , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/prevenção & controle , Tolerância Imunológica , Lipopolissacarídeos/administração & dosagem , Fármacos Neuroprotetores/administração & dosagem , Linfócitos T Reguladores/imunologia , Animais , Animais Recém-Nascidos , Encéfalo/imunologia , Encéfalo/metabolismo , Encéfalo/patologia , Diferenciação Celular/genética , Movimento Celular/genética , Movimento Celular/imunologia , Células Cultivadas , Técnicas de Cocultura , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Encefalomielite Autoimune Experimental/patologia , Feminino , Fatores de Transcrição Forkhead/antagonistas & inibidores , Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/genética , Tolerância Imunológica/genética , Interleucina-10/biossíntese , Interleucina-10/genética , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/imunologia , Esclerose Múltipla/metabolismo , Índice de Gravidade de Doença , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA