Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Antiviral Res ; 216: 105656, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37327877

RESUMO

The ongoing COVID-19 pandemic caused by SARS-CoV-2 infection has threatened global health. Since the first case of infection was reported in December 2019, SARS-CoV-2 has rapidly spread worldwide and caused millions of deaths. As vaccination is the best way to protect the host from invading pathogens, several vaccines have been developed to prevent the infection of SARS-CoV-2, saving numerous lives thus far. However, SARS-CoV-2 constantly changes its antigens, resulting in escape from vaccine-induced protection, and the longevity of immunity induced by vaccines remains an issue. Additionally, traditional intramuscular COVID-19 vaccines are insufficient at evoking mucosal-specific immune responses. Because the respiratory tract is the primary route of SARS-CoV-2 entry, the need for mucosal vaccines is strong. Using an adenoviral (Ad) vector platform, we generated Ad5-S.Mod, a recombinant COVID-19 vaccine that encodes modified-spike (S) antigen and the genetic adjuvant human CXCL9. Intranasal delivery of Ad5-S.Mod elicited superior airway humoral and T-cell responses over traditional intramuscular vaccines and protected mice from lethal SARS-CoV-2 infection. cDC1 cells were required for the generation of antigen-specific CD8+ T-cell responses and CD8+ tissue-resident memory T-cell development in intranasal Ad5-S.Mod vaccinated mice. Furthermore, we confirmed the efficacy of the intranasal Ad5-S.Mod vaccine in terms of transcriptional changes and identified lung macrophages as a key supporter of maintenance of lung-resident memory T and B cells. Our study demonstrates Ad5-S.Mod has the potential to confer protective immunity against SARS-CoV-2 and that lung macrophages support the maintenance of vaccine-induced tissue-resident memory lymphocytes.


Assuntos
Infecções por Adenoviridae , Vacinas contra Adenovirus , COVID-19 , Camundongos , Humanos , Animais , Adenoviridae/genética , Vacinas contra COVID-19 , SARS-CoV-2/genética , COVID-19/prevenção & controle , Imunidade nas Mucosas , Glicoproteína da Espícula de Coronavírus/genética , Pandemias , Adjuvantes Imunológicos , Anticorpos Antivirais , Anticorpos Neutralizantes
2.
J Virol ; 97(6): e0055623, 2023 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-37191498

RESUMO

During the 2015-2016 Zika virus (ZIKV) epidemic, ZIKV-associated neurological diseases were reported in adults, including microcephaly, Guillain-Barre syndrome, myelitis, meningoencephalitis, and fatal encephalitis. However, the mechanisms underlying the neuropathogenesis of ZIKV infection are not yet fully understood. In this study, we used an adult ZIKV infection mouse model (Ifnar1-/-) to investigate the mechanisms underlying neuroinflammation and neuropathogenesis. ZIKV infection induced the expression of proinflammatory cytokines, including interleukin-1ß (IL-1ß), IL-6, gamma interferon, and tumor necrosis factor alpha, in the brains of Ifnar1-/- mice. RNA-seq analysis of the infected mouse brain also revealed that genes involved in innate immune responses and cytokine-mediated signaling pathways were significantly upregulated at 6 days postinfection. Furthermore, ZIKV infection induced macrophage infiltration and activation and augmented IL-1ß expression, whereas microgliosis was not observed in the brain. Using human monocyte THP-1 cells, we confirmed that ZIKV infection promotes inflammatory cell death and increases IL-1ß secretion. In addition, expression of the complement component C3, which is associated with neurodegenerative diseases and known to be upregulated by proinflammatory cytokines, was induced by ZIKV infection through the IL-1ß-mediated pathway. An increase in C5a produced by complement activation in the brains of ZIKV-infected mice was also verified. Taken together, our results suggest that ZIKV infection in the brain of this animal model augments IL-1ß expression in infiltrating macrophages and elicits IL-1ß-mediated inflammation, which can lead to the destructive consequences of neuroinflammation. IMPORTANCE Zika virus (ZIKV) associated neurological impairments are an important global health problem. Our results suggest that ZIKV infection in the mouse brain can induce IL-1ß-mediated inflammation and complement activation, thereby contributing to the development of neurological disorders. Thus, our findings reveal a mechanism by which ZIKV induces neuroinflammation in the mouse brain. Although we used adult type I interferon receptor IFNAR knockout (Ifnar1-/-) mice owing to the limited mouse models of ZIKV pathogenesis, our conclusions contributed to the understanding ZIKV-associated neurological diseases to develop treatment strategies for patients with ZIKV infection based on these findings.


Assuntos
Encéfalo , Interleucina-1beta , Macrófagos , Infecção por Zika virus , Animais , Humanos , Camundongos , Encéfalo/imunologia , Citocinas/imunologia , Inflamação/imunologia , Interleucina-1beta/imunologia , Macrófagos/imunologia , Doenças Neuroinflamatórias/imunologia , Doenças Neuroinflamatórias/virologia , Zika virus , Infecção por Zika virus/imunologia , Transcriptoma/imunologia , Modelos Animais de Doenças , Neurônios/imunologia , Neurônios/virologia
3.
Nat Immunol ; 24(6): 1007-1019, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37069398

RESUMO

Adoptive transfer of genetically engineered chimeric antigen receptor (CAR) T cells is becoming a promising treatment option for hematological malignancies. However, T cell immunotherapies have mostly failed in individuals with solid tumors. Here, with a CRISPR-Cas9 pooled library, we performed an in vivo targeted loss-of-function screen and identified ST3 ß-galactoside α-2,3-sialyltransferase 1 (ST3GAL1) as a negative regulator of the cancer-specific migration of CAR T cells. Analysis of glycosylated proteins revealed that CD18 is a major effector of ST3GAL1 in activated CD8+ T cells. ST3GAL1-mediated glycosylation induces the spontaneous nonspecific tissue sequestration of T cells by altering lymphocyte function-associated antigen-1 (LFA-1) endocytic recycling. Engineered CAR T cells with enhanced expression of ßII-spectrin, a central LFA-1-associated cytoskeleton molecule, reversed ST3GAL1-mediated nonspecific T cell migration and reduced tumor growth in mice by improving tumor-specific homing of CAR T cells. These findings identify the ST3GAL1-ßII-spectrin axis as a major cell-intrinsic program for cancer-targeting CAR T cell migration and as a promising strategy for effective T cell immunotherapy.


Assuntos
Receptores de Antígenos Quiméricos , Animais , Camundongos , Linfócitos T CD8-Positivos , Linhagem Celular Tumoral , Movimento Celular , Imunoterapia Adotiva , Antígeno-1 Associado à Função Linfocitária , Espectrina , Humanos , Feminino
4.
Microbiol Spectr ; 10(3): e0109122, 2022 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-35510852

RESUMO

Accumulating evidence suggests that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection causes various neurological symptoms in patients with coronavirus disease 2019 (COVID-19). The most dominant immune cells in the brain are microglia. Yet, the relationship between neurological manifestations, neuroinflammation, and host immune response of microglia to SARS-CoV-2 has not been well characterized. Here, we reported that SARS-CoV-2 can directly infect human microglia, eliciting M1-like proinflammatory responses, followed by cytopathic effects. Specifically, SARS-CoV-2 infected human microglial clone 3 (HMC3), leading to inflammatory activation and cell death. RNA sequencing (RNA-seq) analysis also revealed that endoplasmic reticulum (ER) stress and immune responses were induced in the early, and apoptotic processes in the late phases of viral infection. SARS-CoV-2-infected HMC3 showed the M1 phenotype and produced proinflammatory cytokines, such as interleukin (IL)-1ß, IL-6, and tumor necrosis factor α (TNF-α), but not the anti-inflammatory cytokine IL-10. After this proinflammatory activation, SARS-CoV-2 infection promoted both intrinsic and extrinsic death receptor-mediated apoptosis in HMC3. Using K18-hACE2 transgenic mice, murine microglia were also infected by intranasal inoculation of SARS-CoV-2. This infection induced the acute production of proinflammatory microglial IL-6 and TNF-α and provoked a chronic loss of microglia. Our findings suggest that microglia are potential mediators of SARS-CoV-2-induced neurological problems and, consequently, can be targets of therapeutic strategies against neurological diseases in patients with COVID-19. IMPORTANCE Recent studies reported neurological and cognitive sequelae in patients with COVID-19 months after the viral infection with several symptoms, including ageusia, anosmia, asthenia, headache, and brain fog. Our conclusions raise awareness of COVID-19-related microglia-mediated neurological disorders to develop treatment strategies for the affected patients. We also indicated that HMC3 was a novel human cell line susceptible to SARS-CoV-2 infection that exhibited cytopathic effects, which could be further used to investigate cellular and molecular mechanisms of neurological manifestations of patients with COVID-19.


Assuntos
Apoptose , COVID-19 , Microglia , Animais , Linhagem Celular , Citocinas/metabolismo , Humanos , Interleucina-6 , Camundongos , Camundongos Transgênicos , Microglia/virologia , SARS-CoV-2 , Fator de Necrose Tumoral alfa
5.
J Med Chem ; 64(20): 14955-14967, 2021 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-34624194

RESUMO

Blocking the association between the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein receptor-binding domain (RBD) and the human angiotensin-converting enzyme 2 (ACE2) is an attractive therapeutic approach to prevent the virus from entering human cells. While antibodies and other modalities have been developed to this end, d-amino acid peptides offer unique advantages, including serum stability, low immunogenicity, and low cost of production. Here, we designed potent novel D-peptide inhibitors that mimic the ACE2 α1-binding helix by searching a mirror-image version of the PDB. The two best designs bound the RBD with affinities of 29 and 31 nM and blocked the infection of Vero cells by SARS-CoV-2 with IC50 values of 5.76 and 6.56 µM, respectively. Notably, both D-peptides neutralized with a similar potency the infection of two variants of concern: B.1.1.7 and B.1.351 in vitro. These potent D-peptide inhibitors are promising lead candidates for developing SARS-CoV-2 prophylactic or therapeutic treatments.


Assuntos
Peptídeos , SARS-CoV-2 , Animais , Chlorocebus aethiops , Simulação de Acoplamento Molecular , Células Vero
6.
Front Immunol ; 12: 666231, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34149701

RESUMO

Although cancer immunotherapy is effective against hematological malignancies, it is less effective against solid tumors due in part to significant metabolic challenges present in the tumor microenvironment (TME), where infiltrated CD8+ T cells face fierce competition with cancer cells for limited nutrients. Strong metabolic suppression in the TME is often associated with impaired T cell recruitment to the tumor site and hyporesponsive effector function via T cell exhaustion. Increasing evidence suggests that mitochondria play a key role in CD8+ T cell activation, effector function, and persistence in tumors. In this study, we showed that there was an increase in overall mitochondrial function, including mitochondrial mass and membrane potential, during both mouse and human CD8+ T cell activation. CD8+ T cell mitochondrial membrane potential was closely correlated with granzyme B and IFN-γ production, demonstrating the significance of mitochondria in effector T cell function. Additionally, activated CD8+ T cells that migrate on ICAM-1 and CXCL12 consumed significantly more oxygen than stationary CD8+ T cells. Inhibition of mitochondrial respiration decreased the velocity of CD8+ T cell migration, indicating the importance of mitochondrial metabolism in CD8+ T cell migration. Remote optical stimulation of CD8+ T cells that express our newly developed "OptoMito-On" successfully enhanced mitochondrial ATP production and improved overall CD8+ T cell migration and effector function. Our study provides new insight into the effect of the mitochondrial membrane potential on CD8+ T cell effector function and demonstrates the development of a novel optogenetic technique to remotely control T cell metabolism and effector function at the target tumor site with outstanding specificity and temporospatial resolution.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/efeitos da radiação , Optogenética/métodos , Trifosfato de Adenosina/metabolismo , Animais , Linfócitos T CD8-Positivos/metabolismo , Movimento Celular/efeitos da radiação , Citocinas/metabolismo , Citoesqueleto/metabolismo , Citoesqueleto/efeitos da radiação , Humanos , Imunoterapia , Ativação Linfocitária/efeitos da radiação , Potencial da Membrana Mitocondrial/genética , Potencial da Membrana Mitocondrial/efeitos da radiação , Camundongos , Mitocôndrias/metabolismo , Mitocôndrias/efeitos da radiação , Neoplasias/imunologia , Neoplasias/terapia
7.
Microorganisms ; 9(3)2021 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-33800763

RESUMO

Zika virus (ZIKV), which is associated with severe diseases in humans, has spread rapidly and globally since its emergence. ZIKV and dengue virus (DENV) are closely related, and antibody-dependent enhancement (ADE) of infection between cocirculating ZIKV and DENV may exacerbate disease. Despite these serious threats, there are currently no approved antiviral drugs against ZIKV and DENV. The NS2B-NS3 viral protease is an attractive antiviral target because it plays a pivotal role in polyprotein cleavage, which is required for viral replication. Thus, we sought to identify novel inhibitors of the NS2B-NS3 protease. To that aim, we performed structure-based virtual screening using 467,000 structurally diverse chemical compounds. Then, a fluorescence-based protease inhibition assay was used to test whether the selected candidates inhibited ZIKV protease activity. Among the 123 candidate inhibitors selected from virtual screening, compound 1 significantly inhibited ZIKV NS2B-NS3 protease activity in vitro. In addition, compound 1 effectively inhibited ZIKV and DENV infection of human cells. Molecular docking analysis suggested that compound 1 binds to the NS2B-NS3 protease of ZIKV and DENV. Thus, compound 1 could be used as a new therapeutic option for the development of more potent antiviral drugs against both ZIKV and DENV, reducing the risks of ADE.

8.
BMB Rep ; 54(1): 31-43, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33298246

RESUMO

Dendritic cells (DC), which consist of several different subsets, specialize in antigen presentation and are critical for mediating the innate and adaptive immune responses. DC subsets can be classified into conventional, plasmacytoid, and monocyte-derived DC in the tumor microenvironment, and each subset plays a different role. Because of the role of intratumoral DCs in initiating antitumor immune responses with tumor-derived antigen presentation to T cells, DCs have been targeted in the treatment of cancer. By regulating the functionality of DCs, several DCbased immunotherapies have been developed, including administration of tumor-derived antigens and DC vaccines. In addition, DCs participate in the mechanisms of classical cancer therapies, such as radiation therapy and chemotherapy. Thus, regulating DCs is also important in improving current cancer therapies. Here, we will discuss the role of each DC subset in antitumor immune responses, and the current status of DC-related cancer therapies. [BMB Reports 2021; 54(1): 31-43].


Assuntos
Neoplasias/terapia , Apresentação de Antígeno/imunologia , Células Dendríticas/imunologia , Humanos , Neoplasias/imunologia , Microambiente Tumoral/imunologia
9.
Nat Commun ; 8: 15365, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28504276

RESUMO

Adoptive cell transfer utilizing tumour-targeting cytotoxic T lymphocytes (CTLs) is one of the most effective immunotherapies against haematological malignancies, but significant clinical success has not yet been achieved in solid tumours due in part to the strong immunosuppressive tumour microenvironment. Here, we show that suppression of CTL killing by CD4+CD25+Foxp3+ regulatory T cell (Treg) is in part mediated by TGFß-induced inhibition of inositol trisphosphate (IP3) production, leading to a decrease in T cell receptor (TCR)-dependent intracellular Ca2+ response. Highly selective optical control of Ca2+ signalling in adoptively transferred CTLs enhances T cell activation and IFN-γ production in vitro, leading to a significant reduction in tumour growth in mice. Altogether, our findings indicate that the targeted optogenetic stimulation of intracellular Ca2+ signal allows for the remote control of cytotoxic effector functions of adoptively transferred T cells with outstanding spatial resolution by boosting T cell immune responses at the tumour sites.


Assuntos
Cálcio/imunologia , Neoplasias Experimentais/imunologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Reguladores/imunologia , Microambiente Tumoral/imunologia , Animais , Cálcio/metabolismo , Linhagem Celular Tumoral , Células HEK293 , Humanos , Imunoterapia Adotiva/métodos , Interferon gama/genética , Interferon gama/imunologia , Interferon gama/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Neoplasias Experimentais/genética , Neoplasias Experimentais/terapia , Linfócitos T Citotóxicos/metabolismo , Linfócitos T Citotóxicos/transplante , Linfócitos T Reguladores/metabolismo , Carga Tumoral/genética , Carga Tumoral/imunologia , Microambiente Tumoral/genética
10.
Methods Enzymol ; 543: 1-20, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24924125

RESUMO

Ca(2+) is a ubiquitous second messenger that is involved in regulation of various signaling pathways. Cytoplasmic Ca(2+) is maintained at low concentrations (~100 nM) by many active mechanisms. Increases in intracellular Ca(2+) concentration ([Ca(2+)]i) indeed can initiate multiple signaling pathways, depending both on their pattern and subcellular localization. In T cells, the stimulation of T-cell receptor leads to an increase in [Ca(2+)]i upon the opening of Ca(2+) release-activated calcium (CRAC) channels. T cells can actually sustain high [Ca(2+)]i for several hours, resulting in the activation of transcriptional programs orchestrated by members of the nuclear factor of activated T-cell (NFAT) protein family. Here, we describe an imaging method widely employed to measure cytoplasmic [Ca(2+)] in naïve and effector T cells based on the ratiometric dye Fura-2. Furthermore, we discuss a pharmacological method relying on an inhibitor of CRAC channels, 2-aminoethyldiphenyl borate, to validate the role of CRAC channels in cytoplasmic Ca(2+) elevation. Finally, we describe an approach to measure mitochondrial [Ca(2+)] based on another fluorescent dye, Rhod-2. With appropriate variations, our methodological approach can be employed to assess the effect and regulation of cytosolic and mitochondrial Ca(2+) waves in multiple experimental settings, including cultured cancer cells.


Assuntos
Cálcio/metabolismo , Corantes/farmacologia , Citoplasma/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Equorina/metabolismo , Animais , Compartimento Celular , Linhagem Celular Tumoral , Citoplasma/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Mitocôndrias/metabolismo
11.
Proc Natl Acad Sci U S A ; 109(22): 8682-7, 2012 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-22586105

RESUMO

Orai1 and stromal interaction molecule (STIM)1 are critical components of Ca(2+) release-activated Ca(2+) (CRAC) channels. Orai1 is a pore subunit of CRAC channels, and STIM1 acts as an endoplasmic reticulum (ER) Ca(2+) sensor that detects store depletion. Upon store depletion after T-cell receptor stimulation, STIM1 translocates and coclusters with Orai1 at sites of close apposition of the plasma membrane (PM) and the ER membrane. However, the molecular components of these ER-PM junctions remain poorly understood. Using affinity protein purification, we uncovered junctate as an interacting partner of Orai1-STIM1 complex. Furthermore, we identified a Ca(2+)-binding EF-hand motif in the ER-luminal region of junctate. Mutation of this EF-hand domain of junctate impaired its Ca(2+) binding and resulted in partial activation of CRAC channels and clustering of STIM1 independently of store depletion. In addition to the known mechanisms of STIM1 clustering (i.e., phosphoinositide and Orai1 binding), our study identifies an alternate mechanism to recruit STIM1 into the ER-PM junctions via binding to junctate. We propose that junctate, a Ca(2+)-sensing ER protein, is a structural component of the ER-PM junctions where Orai1 and STIM1 cluster and interact in T cells.


Assuntos
Canais de Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Membrana/metabolismo , Oxigenases de Função Mista/metabolismo , Proteínas Musculares/metabolismo , Proteínas de Neoplasias/metabolismo , Cálcio/metabolismo , Canais de Cálcio/genética , Proteínas de Ligação ao Cálcio/genética , Membrana Celular/metabolismo , Motivos EF Hand/genética , Retículo Endoplasmático/metabolismo , Células HEK293 , Células HeLa , Humanos , Immunoblotting , Células Jurkat , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Proteínas de Membrana/genética , Microscopia de Fluorescência , Oxigenases de Função Mista/genética , Proteínas Musculares/genética , Mutação , Proteínas de Neoplasias/genética , Proteína ORAI1 , Ligação Proteica , Transporte Proteico , Molécula 1 de Interação Estromal
12.
Analyst ; 136(13): 2831-6, 2011 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-21623432

RESUMO

Leukocyte adhesion to the endothelium through surface molecules such as E-selectin and intercellular adhesion molecule-1 (ICAM-1) is a critical cellular event reflecting the physiological status of both cell types. Here we present a microfluidic system that can not only easily monitor the interaction between leukocytes and endothelial cells under physiological conditions, but also screen drug candidates for potential modulation of this interaction. Shear stress, which is an important factor for the binding of activated T cells to tumor necrosis factor-alpha (TNF-α)-treated human umbilical vein endothelial cells (HUVECs), was easily controlled by adjusting the flow rate in the microfluidic system. Whole blood of patients with systemic lupus erythematosus (SLE) who have auto-reactive T cells were infused into the activated HUVECs which subsequently showed a higher level of binding compared to a control blood sample from a person without SLE. When these autoreactive T cells were treated with immunosuppressors tacrolimus and cyclosporin A, the binding of the T cells to HUVECs was dramatically decreased. Therefore, this microfluidic system is capable of differentiating the physiological status of T cells or endothelial cells representing different disease conditions, as well as being useful for the identification of novel reagents that modulate the functions of leukocytes or endothelial cells.


Assuntos
Técnicas Analíticas Microfluídicas/métodos , Linfócitos T/citologia , Moléculas de Adesão Celular/metabolismo , Ciclosporina/imunologia , Avaliação Pré-Clínica de Medicamentos , Células Endoteliais/citologia , Humanos , Imunossupressores/imunologia , Células Jurkat , Leucócitos/citologia , Leucócitos/imunologia , Leucócitos/metabolismo , Técnicas Analíticas Microfluídicas/instrumentação , Linfócitos T/imunologia , Linfócitos T/metabolismo , Tacrolimo/imunologia
13.
Nat Cell Biol ; 12(5): 436-46, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20418871

RESUMO

Orai1 and STIM1 are critical components of Ca(2+) release-activated Ca(2+) (CRAC) channels that mediate store-operated Ca(2+) entry (SOCE) in immune cells. Although it is known that Orai1 and STIM1 co-cluster and physically interact to mediate SOCE, the cytoplasmic machinery modulating these functions remains poorly understood. We sought to find modulators of Orai1 and STIM1 using affinity protein purification and identified a novel EF-hand protein, CRACR2A (also called CRAC regulator 2A, EFCAB4B or FLJ33805). We show that CRACR2A interacts directly with Orai1 and STIM1, forming a ternary complex that dissociates at elevated Ca(2+) concentrations. Studies using knockdown mediated by small interfering RNA (siRNA) and mutagenesis show that CRACR2A is important for clustering of Orai1 and STIM1 upon store depletion. Expression of an EF-hand mutant of CRACR2A enhanced STIM1 clustering, elevated cytoplasmic Ca(2+) and induced cell death, suggesting its active interaction with CRAC channels. These observations implicate CRACR2A, a novel Ca(2+) binding protein that is highly expressed in T cells and conserved in vertebrates, as a key regulator of CRAC channel-mediated SOCE.


Assuntos
Canais de Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Cálcio/metabolismo , Linfócitos T/metabolismo , Sinalização do Cálcio , Proteínas de Ligação ao Cálcio/fisiologia , Citosol , Motivos EF Hand , Humanos , Proteínas de Membrana/metabolismo , Dados de Sequência Molecular , Proteínas de Neoplasias/metabolismo , Proteína ORAI1 , Estabilidade Proteica , Molécula 1 de Interação Estromal
14.
Nat Med ; 12(5): 574-9, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16604087

RESUMO

CTLA-4 is a negative regulator of T-cell activation, and its inhibitory effects can be accomplished either by competition with CD28 or by transmitting negative signals through its intracellular domain. To utilize the cytoplasmic domain of CTLA-4 to suppress allergic inflammation, we fused it to a novel protein-transduction domain in the human transcriptional factor Hph-1. Transduction efficiency was verified in vitro and in vivo after ocular, intranasal and intradermal administration. After transduction into T cells, the Hph-1-ctCTLA-4 fusion protein inhibited the production of interleukin (IL)-2, and downregulated CD69 and CD25. Intranasal administration of Hph-1-ctCTLA-4 resulted in markedly reduced infiltration of inflammatory cells, secretion of T helper type 2 (T(H)2) cytokines, serum IgE levels and airway hyper-responsiveness in a mouse model of allergic airway inflammation. These results indicated that Hph-1-ctCTLA-4 constitutes an effective immunosuppressive protein drug for potential use in the treatment of allergic asthma, via nasal administration.


Assuntos
Administração Intranasal , Antígenos de Diferenciação/administração & dosagem , Antígenos de Diferenciação/imunologia , Asma , Proteínas de Transporte/metabolismo , Imunossupressores , Inflamação , Animais , Antígenos CD , Antígenos de Diferenciação/genética , Asma/imunologia , Asma/prevenção & controle , Hiper-Reatividade Brônquica , Antígeno CTLA-4 , Proteínas de Transporte/genética , Feminino , Humanos , Imunoconjugados/administração & dosagem , Imunoconjugados/genética , Imunoconjugados/imunologia , Imunossupressores/administração & dosagem , Imunossupressores/imunologia , Inflamação/imunologia , Inflamação/prevenção & controle , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina/imunologia , Complexo Repressor Polycomb 1 , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Transdução Genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA