Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Med Virol ; 96(3): e29504, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38445794

RESUMO

While most NOD-like receptors (NLRs) are predominately expressed by innate immune cells, NLRC3, an inhibitory NLR of immune signaling, exhibits the highest expression in lymphocytes. The role of NLRC3 or any NLRs in B lymphocytes is completely unknown. Gammaherpesviruses, including human Epstein-Barr virus (EBV) and murine gammaherpesvirus 68 (MHV-68), establish latent infection in B lymphocytes, which requires elevated NF-κB. This study shows that during latent EBV infection of human B cells, viral-encoded latent membrane protein 1 (LMP1) decreases NLRC3 transcript. LMP1-induced-NF-κB activation suppresses the promoter activity of NLRC3 via p65 binding to the promoter. Conversely, NLRC3 inhibits NF-κB activation by promoting the degradation of LMP1 in a proteasome-dependent manner. In vivo, MHV-68 infection reduces Nlrc3 transcripts in splenocytes, and Nlrc3-deficient mice show greater viral latency than controls. These results reveal a bidirectional regulatory circuit in B lymphocytes, where viral latent protein LMP1 reduces NLRC3 expression, while NLRC3 disrupts gammaherpesvirus latency, which is an important step for tumorigenesis.


Assuntos
Infecções por Vírus Epstein-Barr , Latência Viral , Animais , Humanos , Camundongos , Herpesvirus Humano 4/genética , NF-kappa B , Linfócitos B , Peptídeos e Proteínas de Sinalização Intercelular
3.
Brain ; 146(7): 2989-3002, 2023 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-36795624

RESUMO

Spreading depolarization (SD), the underlying mechanism of migraine aura, may trigger the opening of the pannexin 1 (PANX1) pore to sustain the cortical neuroinflammatory cascades involved in the genesis of headache. Yet, the mechanism underlying SD-evoked neuroinflammation and trigeminovascular activation remains incompletely understood. We characterized the identity of inflammasome activated following SD-evoked PANX1 opening. Pharmacological inhibitors targeting PANX1 or NLRP3 as well as genetic ablation of Nlrp3 and Il1b were applied to investigate the molecular mechanism of the downstream neuroinflammatory cascades. In addition, we examined whether SD-triggered microglial activation facilitates neuronal NLRP3-mediated inflammatory cascades. Pharmacological inhibition of toll-like receptors TLR2/4, the potential receptors of the damage-associated molecular pattern HMGB1, was further employed to interrogate the neuron-microglia interplay in SD-induced neuroinflammation. We found that NLRP3 but not NLRP1 or NLRP2 inflammasome was activated following PANX1 opening after single or multiple SDs evoked by either KCl topical application or non-invasively with optogenetics. The SD-evoked NLRP3 inflammasome activation was observed exclusively in neurons but not microglia or astrocytes. Proximity ligation assay demonstrated that the assembly of the NLRP3 inflammasome occurred as early as 15 min after SD. Genetic ablation of Nlrp3 or Il1b or pharmacological inhibition of PANX1 or NLRP3 ameliorated SD-induced neuronal inflammation, middle meningeal artery dilatation, calcitonin gene-related peptide expression in trigeminal ganglion and c-Fos expression in trigeminal nucleus caudalis. Moreover, multiple SDs induced microglial activation subsequent to neuronal NLRP3 inflammasome activation, which in turn orchestrated with neurons to mediate cortical neuroinflammation, as demonstrated by decreased neuronal inflammation after pharmacological inhibition of microglia activation or blockade of the TLR2/4 receptors. To conclude, single or multiple SDs evoked activation of neuronal NLRP3 inflammasomes and its downstream inflammatory cascades to mediate cortical neuroinflammation and trigeminovascular activation. In the context of multiple SDs, the cortical inflammatory processes could be facilitated by SD-evoked microglia activation. These findings may implicate the potential role of innate immunity in migraine pathogenesis.


Assuntos
Inflamassomos , Transtornos de Enxaqueca , Humanos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Doenças Neuroinflamatórias , Receptor 2 Toll-Like , Inflamação , Neurônios/metabolismo , Proteínas do Tecido Nervoso , Conexinas
4.
J Clin Invest ; 133(3)2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36719379

RESUMO

Signaling driven by nucleic acid sensors participates in interferonopathy-mediated autoimmune diseases. NLRP12, a pyrin-containing NLR protein, is a negative regulator of innate immune activation and type I interferon (IFN-I) production. Peripheral blood mononuclear cells (PBMCs) derived from systemic lupus erythematosus (SLE) patients expressed lower levels of NLRP12, with an inverse correlation with IFNA expression and high disease activity. NLRP12 expression was transcriptionally suppressed by runt-related transcription factor 1-dependent (RUNX1-dependent) epigenetic regulation under IFN-I treatment, which enhanced a negative feedback loop between low NLRP12 expression and IFN-I production. Reduced NLRP12 protein levels in SLE monocytes was linked to spontaneous activation of innate immune signaling and hyperresponsiveness to nucleic acid stimulations. Pristane-treated Nlrp12-/- mice exhibited augmented inflammation and immune responses; and substantial lymphoid hypertrophy was characterized in NLRP12-deficient lupus-prone mice. NLRP12 deficiency mediated the increase of autoantibody production, intensive glomerular IgG deposition, monocyte recruitment, and the deterioration of kidney function. These were bound in an IFN-I signature-dependent manner in the mouse models. Collectively, we reveal a remarkable link between low NLRP12 expression and lupus progression, which suggests the impact of NLRP12 on homeostasis and immune resilience.


Assuntos
Lúpus Eritematoso Sistêmico , Nefrite Lúpica , Ácidos Nucleicos , Animais , Camundongos , Epigênese Genética , Imunidade Inata , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Leucócitos Mononucleares , Lúpus Eritematoso Sistêmico/genética , Interferons/metabolismo
5.
Front Microbiol ; 13: 883597, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35663899

RESUMO

COVID-19 is currently global pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Accompanying the rapid spread of the error-prone RNA-based genome, several dominant SARS-CoV-2 variants have been genetically identified. The mutations in the spike protein, which are essential for receptor binding and fusion, have been intensively investigated for their contributions to viral transmission. Nevertheless, the importance of other viral proteins and their mutations in SARS-CoV-2 lifecycle and transmission remains fairly understood. Here, we report the strong potency of an accessory protein ORF8 in modulating the level and processing of the spike protein. The expression of ORF8 protein does not affect propagation but expression of spike protein, which may lead to pseudovirions with less spike protein on the surface, therefore less infection potential. At the protein level, ORF8 expression led to downregulation and insufficient S1/S2 cleavage of the spike protein in a dose-dependent manner. ORF8 exhibits a strong interaction with the spike protein mainly at S1 domains and mediates its degradation through multiple pathways. The dominant clinical isolated ORF8 variants with the reduced protein stability exhibited the increased capacity of viral transmission without compromising their inhibitory effects on HLA-A2. Although the increase in spike protein level and Spike pseudovirus production observed by using highly transmissible clinical spike variants, there was no significant compromise in ORF8-mediated downregulation. Because ORF8 is important for immune surveillance and might be required for viral fitness in vivo, the alteration of the spike protein might be an optional strategy used by SARS-CoV-2 to promote viral transmission by escaping the inhibitory effects of ORF8. Therefore, our report emphasized the importance of ORF8 in SARS-CoV-2 spike protein production, maturation, and possible evolution.

6.
Int J Mol Sci ; 22(21)2021 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-34768828

RESUMO

Cell death is an essential immunological apparatus of host defense, but dysregulation of mutually inclusive cell deaths poses severe threats during microbial and parasitic infections leading to deleterious consequences in the pathological progression of infectious diseases. Nucleotide-binding oligomerization domain (NOD)-Leucine-rich repeats (LRR)-containing receptors (NLRs), also called nucleotide-binding oligomerization (NOD)-like receptors (NLRs), are major cytosolic pattern recognition receptors (PRRs), their involvement in the orchestration of innate immunity and host defense against bacteria, viruses, fungi and parasites, often results in the cleavage of gasdermin and the release of IL-1ß and IL-18, should be tightly regulated. NLRs are functionally diverse and tissue-specific PRRs expressed by both immune and non-immune cells. Beyond the inflammasome activation, NLRs are also involved in NF-κB and MAPK activation signaling, the regulation of type I IFN (IFN-I) production and the inflammatory cell death during microbial infections. Recent advancements of NLRs biology revealed its possible interplay with pyroptotic cell death and inflammatory mediators, such as caspase 1, caspase 11, IFN-I and GSDMD. This review provides the most updated information that caspase 8 skews the NLRP3 inflammasome activation in PANoptosis during pathogen infection. We also update multidimensional roles of NLRP12 in regulating innate immunity in a content-dependent manner: novel interference of NLRP12 on TLRs and NOD derived-signaling cascade, and the recently unveiled regulatory property of NLRP12 in production of type I IFN. Future prospects of exploring NLRs in controlling cell death during parasitic and microbial infection were highlighted.


Assuntos
Infecções/imunologia , Proteínas NLR/fisiologia , Doenças Parasitárias/imunologia , Animais , Morte Celular/imunologia , Interações entre Hospedeiro e Microrganismos , Interações Hospedeiro-Parasita , Humanos , Mediadores da Inflamação/metabolismo , Proteínas NLR/genética , Proteínas NLR/imunologia , Proteínas NLR/metabolismo , Receptores de Reconhecimento de Padrão/genética , Receptores de Reconhecimento de Padrão/imunologia , Transdução de Sinais , Vírus/imunologia
7.
Plast Reconstr Surg Glob Open ; 8(5): e2829, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-33154871

RESUMO

Injectable poly-d,l-lactic acid (PDLLA) is a new collagen-stimulating filler containing PDLLA microspheres and carboxymethyl cellulose. It is available as a lyophilized powder that must be reconstituted with a diluent before administration. The aims of this study were to investigate the efficacy of different diluents and a new accelerating "back-and-forth" method. METHODS: Six different diluents, sodium bicarbonate, sterile water for injection (SWFI), normal saline, lidocaine, lidocaine with epinephrine (lidocaine + E), and mannitol, were tested. The recommended "vortex" method for preparation of thin suspensions and a new back-and-forth method suitable for both thin and thick suspensions were compared. Gross and microscopic views of the prepared suspensions were examined. RESULTS: Using the vortex method, only mannitol and SWFI are found to be effective reconstitution diluents for injectable PDLLA. Using the back-and-forth method, all six diluents can be used for reconstitution of injectable PDLLA. Moreover, the time needed for reconstitution of injectable PDLLA by this back-and-forth method is very short, regardless of the thickness of the suspension. CONCLUSIONS: Clinically, only SWFI can be used for reconstitution of injectable PDLLA by "hand-shaking" or vortex method. To accelerate the reconstitution time especially when using small amount of SWFI, back-and-forth is the method of choice. Besides, when SWFI is not available, other diluents such as normal saline, lidocaine, or lidocaine + E can be used by this novel back-and-forth reconstitution method.

8.
Sci Rep ; 10(1): 8422, 2020 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-32439945

RESUMO

Toll-like receptors (TLRs) play crucial roles in host immune defenses. Recently, TLR-mediated autophagy is reported to promote immune responses via increasing antigen processing and presentation in antigen presenting cells. The present study examined whether the synthetic TLR4 activator (CCL-34) could induce autophagy to promote innate and adaptive immunity. In addition, the potential of CCL-34 as an immune adjuvant in vivo was also investigated. Our data using RAW264.7 cells and bone marrow-derived macrophages showed that CCL-34 induced autophagy through a TLR4-NF-κB pathway. The autophagy-related molecules (Nrf2, p62 and Beclin 1) were activated in RAW264.7 cells and bone marrow-derived macrophages under CCL-34 treatment. CCL-34-stimulated macrophages exhibited significant antigen-processing activity and induced the proliferation of antigen-specific CD4+T cells as well as the production of activated T cell-related cytokines, IL-2 and IFN-γ. Furthermore, CCL-34 immunization in mice induced infiltration of monocytes in the peritoneal cavity and elevation of antigen-specific IgG in the serum. CCL-34 treatment in vivo did not cause toxicity based on serum biochemical profiles. Notably, the antigen-specific responses induced by CCL-34 were attenuated by the autophagy inhibitor, 3-methyladenine. In summary, we demonstrated CCL-34 can induce autophagy to promote antigen-specific immune responses and act as an efficient adjuvant.


Assuntos
Adjuvantes Imunológicos/farmacologia , Autofagia/imunologia , Glicolipídeos/farmacologia , Imunogenicidade da Vacina/imunologia , Serina/análogos & derivados , Receptor 4 Toll-Like/metabolismo , Adenina/análogos & derivados , Adenina/farmacologia , Animais , Proteína Beclina-1/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Imunoglobulina G/sangue , Interferon gama/metabolismo , Interleucina-2/metabolismo , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Monócitos/imunologia , Fator 2 Relacionado a NF-E2/metabolismo , Células RAW 264.7 , Serina/farmacologia , Vacinas/imunologia
9.
J Virol ; 94(3)2020 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-31723026

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) is the causative agent of Kaposi's sarcoma (KS), an AIDS-defining cancer with abnormal angiogenesis. The high incidence of KS in human immunodeficiency virus (HIV)-infected AIDS patients has been ascribed to an interaction between HIV type 1 (HIV-1) and KSHV, focusing on secretory proteins. The HIV-1 secreted protein HIV Tat has been found to synergize with KSHV lytic proteins to induce angiogenesis. However, the impact and underlying mechanisms of HIV Tat in KSHV-infected endothelial cells undergoing viral lytic reactivation remain unclear. Here, we identified LINC00313 as a novel KSHV reactivation-activated long noncoding RNA (lncRNA) that interacts with HIV Tat. We found that LINC00313 overexpression inhibits cell migration, invasion, and tube formation, and this suppressive effect was relieved by HIV Tat. In addition, LINC00313 bound to polycomb repressive complex 2 (PRC2) complex components, and this interaction was disrupted by HIV Tat, suggesting that LINC00313 may mediate transcription repression through recruitment of PRC2 and that HIV Tat alleviates repression through disruption of this association. This notion was further supported by bioinformatics analysis of transcriptome profiles in LINC00313 overexpression combined with HIV Tat treatment. Ingenuity Pathway Analysis (IPA) showed that LINC00313 overexpression negatively regulates cell movement and migration pathways, and enrichment of these pathways was absent in the presence of HIV Tat. Collectively, our results illustrate that an angiogenic repressive lncRNA, LINC00313, which is upregulated during KSHV reactivation, interacts with HIV Tat to promote endothelial cell motility. These results demonstrate that an lncRNA serves as a novel connector in HIV-KSHV interactions.IMPORTANCE KS is a prevalent tumor associated with infections with two distinct viruses, KSHV and HIV. Since KSHV and HIV infect distinct cell types, the virus-virus interaction associated with KS formation has focused on secretory factors. HIV Tat is a well-known RNA binding protein secreted by HIV. Here, we revealed LINC00313, an lncRNA upregulated during KSHV lytic reactivation, as a novel HIV Tat-interacting lncRNA that potentially mediates HIV-KSHV interactions. We found that LINC00313 can repress endothelial cell angiogenesis-related properties potentially by interacting with chromatin remodeling complex PRC2 and downregulation of cell migration-regulating genes. An interaction between HIV Tat and LINC00313 contributed to the dissociation of PRC2 from LINC00313 and the disinhibition of LINC00313-induced repression of cell motility. Given that lncRNAs are emerging as key players in tissue physiology and disease progression, including cancer, the mechanism identified in this study may help decipher the mechanisms underlying KS pathogenesis induced by HIV and KSHV coinfection.


Assuntos
HIV-1/fisiologia , Herpesvirus Humano 8/fisiologia , RNA Longo não Codificante/metabolismo , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo , Coinfecção , Células Endoteliais/metabolismo , Perfilação da Expressão Gênica , Infecções por HIV/virologia , Humanos , Complexo Repressor Polycomb 2 , Sarcoma de Kaposi/virologia , Ativação Transcricional , Regulação para Cima , Ativação Viral/genética , Replicação Viral
10.
Cell Host Microbe ; 25(4): 602-616.e7, 2019 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-30902577

RESUMO

Establishing the balance between positive and negative innate immune mechanisms is crucial for maintaining homeostasis. Here we uncover the regulatory crosstalk between two previously unlinked innate immune receptor families: RIG-I, an anti-viral cytosolic receptor activated type I interferon production, and NLR (nucleotide-binding domain, leucine repeat domain-containing protein). We show that NLRP12 dampens RIG-I-mediated immune signaling against RNA viruses by controlling RIG-I's association with its adaptor MAVS. The nucleotide-binding domain of NLRP12 interacts with the ubiquitin ligase TRIM25 to prevent TRIM25-mediated, Lys63-linked ubiquitination and activation of RIG-I. NLRP12 also enhances RNF125-mediated, Lys48-linked degradative ubiquitination of RIG-I. Vesicular stomatitis virus (VSV) infection downregulates NLRP12 expression to allow RIG-I activation. Myeloid-cell-specific Nlrp12-deficient mice display a heightened interferon and TNF response and are more resistant to VSV infection. These results indicate that NLRP12 functions as a checkpoint for anti-viral RIG-I activation.


Assuntos
Proteína DEAD-box 58/imunologia , Proteínas de Ligação a DNA/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Infecções por Vírus de RNA/imunologia , Vírus de RNA/fisiologia , Fatores de Transcrição/imunologia , Animais , Proteína DEAD-box 58/genética , Proteínas de Ligação a DNA/genética , Feminino , Humanos , Interferons/genética , Interferons/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Infecções por Vírus de RNA/genética , Infecções por Vírus de RNA/virologia , Vírus de RNA/genética , Fatores de Transcrição/genética , Ubiquitinação
11.
Sports Biomech ; 17(2): 143-156, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28632060

RESUMO

The purpose of this study was to compare the lower extremity inter-joint coordination of different collision forces runners during running braking phase. A dynamical system approach was used to analyse the inter-joint coordination parameters. Data were collected with six infra-red cameras and two force plates. According to the impact peak of the vertical ground reaction force, twenty habitually rearfoot-strike runners were categorised into three groups: high collision forces runners (HF group, n = 8), medium collision forces runners (MF group, n = 5), and low collision forces runners (LF group, n = 7). There were no significant differences among the three groups in the ankle and knee joint angle upon landing and in the running velocity (p > 0.05). The HF group produced significantly smaller deviation phase (DP) of the hip flexion/extension-knee flexion/extension during the braking phase compared with the MF and LF groups (p < 0.05). The DP of the hip flexion/extension-knee flexion/extension during the braking phase correlated negatively with the collision force (p < 0.05). The disparities regarding the flexibility of lower extremity inter-joint coordination were found in high collision forces runners. The efforts of the inter-joint coordination and the risk of running injuries need to be clarified further.


Assuntos
Articulação do Tornozelo/fisiologia , Articulação do Quadril/fisiologia , Articulação do Joelho/fisiologia , Corrida/fisiologia , Fenômenos Biomecânicos/fisiologia , Marcha/fisiologia , Humanos , Fatores de Risco , Corrida/lesões , Estudos de Tempo e Movimento , Adulto Jovem
12.
Nat Commun ; 8(1): 299, 2017 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-28824166

RESUMO

The C-type lectin member 5A (CLEC5A) is a pattern recognition receptor for members of the Flavivirus family and has critical functions in response to dengue virus and Japanese encephalitis virus. Here we show that CLEC5A is involved in neutrophil extracellular trap formation and the production of reactive oxygen species and proinflammatory cytokines in response to Listeria monocytogenes. Inoculation of Clec5a -/- mice with L. monocytogenes causes rapid bacterial spreading, increased bacterial loads in the blood and liver, and severe liver necrosis. In these mice, IL-1ß, IL-17A, and TNF expression is inhibited, CCL2 is induced, and large numbers of CD11b+Ly6ChiCCR2hiCX3CR1low inflammatory monocytes infiltrate the liver. By day 5 of infection, these mice also have fewer IL-17A+ γδ T cells, severe liver necrosis and a higher chance of fatality. Thus, CLEC5A has a pivotal function in the activation of multiple aspects of innate immunity against bacterial invasion.The lectin receptor CLEC5A is a pattern recognition receptor that has been shown to detect dengue and Japanese encephalitis virus. Here the authors show that CLEC5A is needed for optimal ROS production, NET formation and other immune responses to Listeria monocytogenes in mice.


Assuntos
Imunidade Inata/imunologia , Lectinas Tipo C/imunologia , Listeria monocytogenes/imunologia , Listeriose/imunologia , Receptores de Superfície Celular/imunologia , Animais , Citocinas/imunologia , Citocinas/metabolismo , Armadilhas Extracelulares/genética , Armadilhas Extracelulares/imunologia , Armadilhas Extracelulares/microbiologia , Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata/genética , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Listeria monocytogenes/fisiologia , Listeriose/genética , Listeriose/microbiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Espécies Reativas de Oxigênio/imunologia , Espécies Reativas de Oxigênio/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo
13.
J Virol ; 91(1)2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27795434

RESUMO

Human infections with influenza viruses exhibit mild to severe clinical outcomes as a result of complex virus-host interactions. Induction of inflammatory mediators via pattern recognition receptors may dictate subsequent host responses for pathogen clearance and tissue damage. We identified that human C-type lectin domain family 5 member A (CLEC5A) interacts with the hemagglutinin protein of influenza viruses expressed on lentiviral pseudoparticles through lectin screening. Silencing CLEC5A gene expression, blocking influenza-CLEC5A interactions with anti-CLEC5A antibodies, or dampening CLEC5A-mediated signaling using a spleen tyrosine kinase inhibitor consistently reduced the levels of proinflammatory cytokines produced by human macrophages without affecting the replication of influenza A viruses of different subtypes. Infection of bone marrow-derived macrophages from CLEC5A-deficient mice showed reduced levels of tumor necrosis factor alpha (TNF-α) and IP-10 but elevated alpha interferon (IFN-α) compared to those of wild-type mice. The heightened type I IFN response in the macrophages of CLEC5A-deficient mice was associated with upregulated TLR3 mRNA after treatment with double-stranded RNA. Upon lethal challenges with a recombinant H5N1 virus, CLEC5A-deficient mice showed reduced levels of proinflammatory cytokines, decreased immune cell infiltration in the lungs, and improved survival compared to the wild-type mice, despite comparable viral loads noted throughout the course of infection. The survival difference was more prominent at a lower dose of inoculum. Our results suggest that CLEC5A-mediated enhancement of the inflammatory response in myeloid cells contributes to influenza pathogenicity in vivo and may be considered a therapeutic target in combination with effective antivirals. Well-orchestrated host responses together with effective viral clearance are critical for optimal clinical outcome after influenza infections. IMPORTANCE: Multiple pattern recognition receptors work in synergy to sense viral RNA or proteins synthesized during influenza replication and mediate host responses for viral control. Well-orchestrated host responses may help to maintain the inflammatory response to minimize tissue damage while inducing an effective adaptive immune response for viral clearance. We identified that CLEC5A, a C-type lectin receptor which has previously been reported to mediate flavivirus-induced inflammatory responses, enhanced induction of proinflammatory cytokines and chemokines in myeloid cells after influenza infections. CLEC5A-deficient mice infected with influenza virus showed reduced inflammation in the lungs and improved survival compared to that of the wild-type mice despite comparable viral loads. The survival difference was more prominent at a lower dose of inoculum. Collectively, our results suggest that dampening CLEC5A-mediated inflammatory responses in myeloid cells reduces immunopathogenesis after influenza infections.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Vírus da Influenza A Subtipo H1N1/patogenicidade , Virus da Influenza A Subtipo H5N1/patogenicidade , Lectinas Tipo C/imunologia , Infecções por Orthomyxoviridae/imunologia , Receptores de Superfície Celular/imunologia , Animais , Anticorpos/farmacologia , Quimiocina CXCL10/genética , Quimiocina CXCL10/imunologia , Regulação da Expressão Gênica , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Interações Hospedeiro-Patógeno , Humanos , Vírus da Influenza A Subtipo H1N1/crescimento & desenvolvimento , Vírus da Influenza A Subtipo H1N1/imunologia , Virus da Influenza A Subtipo H5N1/crescimento & desenvolvimento , Virus da Influenza A Subtipo H5N1/imunologia , Interferon-alfa/genética , Interferon-alfa/imunologia , Lectinas Tipo C/antagonistas & inibidores , Lectinas Tipo C/genética , Lentivirus/genética , Lentivirus/imunologia , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/virologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/virologia , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/mortalidade , Infecções por Orthomyxoviridae/virologia , Cultura Primária de Células , Ligação Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores de Superfície Celular/antagonistas & inibidores , Receptores de Superfície Celular/genética , Análise de Sobrevida , Receptor 3 Toll-Like/genética , Receptor 3 Toll-Like/imunologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
14.
J Mol Med (Berl) ; 94(9): 1025-37, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27033255

RESUMO

UNLABELLED: Osteoclasts are bone tissue macrophages critical to maintain bone homeostasis. However, whether osteoclasts are susceptible to flaviviral infections and involved in dengue virus (DV)-induced disease pathogenesis is still unknown. In this study, we found that osteoclasts were preferentially susceptible to DV infection and produced similar amounts of cytokines and infectious virions as macrophages. Interestingly, DV-induced cytokine secretion and nuclear translocation of the transcription factor NFATc1 in osteoclast via the Syk-coupled myeloid C-type lectin member 5A (CLEC5A). Moreover, DV caused transient inflammatory reaction in bone tissue and upregulated osteolytic activity to release C-telopeptide of type I collagen (CTX-1) from bone tissue. Furthermore, DV-induced osteolytic activity was attenuated in CLEC5A-deficient mice, and administration of antagonistic anti-CLEC5A mAb inhibited DV-activated osteolytic activity and reduced CTX-1 serum level in vivo. This observation suggests that osteoclasts serve as a novel target for DV, and transient upregulation of osteolytic activity may contribute to the clinical symptoms in dengue patients. KEY MESSAGES: Cultured osteoclasts were susceptible to DV infection. Osteoclasts produced similar amounts of cytokines and infectious virions as macrophages. DV induced nuclear translocation of NFATc1 in osteoclast via CLEC5A. DV caused transient inflammatory reaction in bone tissue and upregulated osteolytic activity. Antagonistic anti-CLEC5A mAb inhibited DV-activated osteolytic activity in vivo.


Assuntos
Osso e Ossos/metabolismo , Vírus da Dengue/fisiologia , Homeostase , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Osteoclastos/metabolismo , Osteoclastos/virologia , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Animais , Biomarcadores , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/patologia , Células Cultivadas , Citocinas/metabolismo , Dengue/genética , Dengue/metabolismo , Dengue/patologia , Dengue/virologia , Humanos , Imuno-Histoquímica , Mediadores da Inflamação/metabolismo , Camundongos , Camundongos Knockout , Modelos Animais , Fatores de Transcrição NFATC/metabolismo , Osteólise/genética , Osteólise/metabolismo , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Tomografia Computadorizada com Tomografia Computadorizada de Emissão de Fóton Único
15.
World Neurosurg ; 89: 732.e19-23, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26882971

RESUMO

BACKGROUND: Central nervous system (CNS) melioidosis is notorious because of the difficulty in bacteria eradication and the destruction of brain structures. Early manifestation of CNS melioidosis mimics malignancy or stroke. We present a case of CNS melioidosis that initially manifested as malignancy. CASE DESCRIPTION: A 30-year-old man presented with sudden onset of left limb weakness and seizure. Computed tomography of the brain showed a low-density lesion over the right parietal lobe, and magnetic resonance imaging showed a well-enhanced lobulated lesion. Neuronavigation-guided open surgery was performed but failed to find a malignancy. The patient presented 3 days later with sudden loss of consciousness, pupil dilation, and high fever. Emergent craniectomy was performed for severe right hemisphere swelling with midline shift. After craniectomy, pus was found in the previous operative field. Burkholderia pseudomallei was cultured from pus and blood samples 1 week after collection. The brain lesion developed into an organized abscess and led to mass effect and ventriculitis. Extraventricular drainage and débridement was performed repeatedly accompanied by systemic and intraventricular antibiotic administration. After 4 months of treatment, the patient achieved a complete consciousness recover while left hemiparesis. CONCLUSIONS: CNS melioidosis requires accurate pathogen identification and appropriate long-term antibiotic treatment for eradication of bacteria and prevention of relapse. Débridement and adequate drainage provide better infection control and outcome.


Assuntos
Infecções Bacterianas do Sistema Nervoso Central/diagnóstico por imagem , Neoplasias do Sistema Nervoso Central/fisiopatologia , Melioidose/diagnóstico por imagem , Adulto , Antibacterianos/uso terapêutico , Infecções Bacterianas do Sistema Nervoso Central/tratamento farmacológico , Humanos , Imageamento por Ressonância Magnética , Masculino , Melioidose/tratamento farmacológico
16.
J Mol Med (Berl) ; 93(10): 1131-43, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25940317

RESUMO

UNLABELLED: Influenza A virus (IAV) infects macrophages and stimulates innate immunity receptors and sensors to produce proinflammatory cytokines and chemokines, which are responsible for IAV-induced pulmonary inflammation and injury. Decoy receptor 3 (DcR3) is a soluble protein belonging to the tumor necrosis factor receptor superfamily (TNFRSF), and is able to skew macrophage differentiation into an M2 phenotype. We demonstrated that DcR3 attenuated IAV-induced secretion of proinflammatory cytokines and chemokine from macrophages, and mitigated pulmonary infiltration and reduce lethality. Proteome-wide phosphoproteomic mapping revealed that DcR3 not only activated STK10, a negative regulator of cell migration, but also inactivated PKC-α, which are crucial for the activation of ERK and JNK in human macrophages. Furthermore, less pulmonary infiltration with lower levels of proinflammatory cytokines and chemokine in bronchoalveolar lavage fluid (BALF) were observed in DcR3-transgenic mice. Moreover, recombinant DcR3.Fc and heparan sulfate proteoglycan binding domain of DcR3.Fc (HBD.Fc) fusion proteins attenuated weight loss and protected mice from IAV-induced lethality. Thus, DcR3-mediated protection is not only via suppression of proinflammatory cytokine and chemokine release, but also via activation of STK10 to inhibit cell infiltration. DcR3 fusion proteins may become therapeutic agents to protect host from IAV-induced lethality in the future. KEY MESSAGE: • DcR3 suppresses IAV-induced cytokine secretion.• DcR3 inhibits IAV-induced JNK and ERK activation in human macrophages.• DcR3 downregulates TLR3 and 7 expressions in human macrophages.• DcR3 protects mice from IAV-induced lethality.


Assuntos
Vírus da Influenza A , Ativação de Macrófagos/imunologia , Proteínas Quinases Ativadas por Mitógeno/imunologia , Infecções por Orthomyxoviridae/imunologia , Pneumonia/imunologia , Membro 6b de Receptores do Fator de Necrose Tumoral/imunologia , Animais , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Citocinas/imunologia , Proteoglicanas de Heparan Sulfato/genética , Humanos , Imunoglobulina G/farmacologia , Pulmão/virologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Estrutura Terciária de Proteína , Membro 6b de Receptores do Fator de Necrose Tumoral/genética , Proteínas Recombinantes de Fusão/farmacologia , Receptores Toll-Like/imunologia
17.
PLoS One ; 9(1): e86105, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24465901

RESUMO

The pathogenic roles of myeloid DAP12-associating lectin-1(MDL-1) and DAP12 in human rheumatoid arthritis (RA) remain unknown. Frequencies of MDL-1-expressing monocytes in 22 active RA patients, 16 inactive RA patients, 12 osteoarthritis (OA) patients and 10 healthy controls (HC) were determined by flow-cytometry analysis. The mRNA expression levels of MDL-1 and DAP12 on PBMCs were evaluated by quantitative PCR, and their protein expression levels in the synovium were examined by immunohistochemistry. Significantly higher median percentages of circulating MDL-1-expressing monocytes were observed in active RA patients (53.6%) compared to inactive RA patients (34.1%), OA patients (27.9%), and HC (21.2%). Levels of MDL-1 and DAP12 gene expression in PBMCs and their protein expression in the synovium were significantly higher in active RA patients than in inactive RA or OA patients. MDL-1 levels were positively correlated with parameters of disease activity, articular damage, and levels of proinflammatory cytokines. MDL-1 activator (Dengue virus type 2 antigen) stimulation on PBMCs resulted in significantly enhanced levels of proinflammatory cytokines in RA patients compared to those in OA patients or HC, indicating that MDL-1 activation is functional. Frequencies of MDL-1-expressing monocytes and levels of MDL-1 and DAP12 gene expression significantly decreased after effective therapy. Concordant overexpression of MDL-1 and DAP12 were correlated with increased production of proinflammatory cytokines in RA patients, suggesting their roles in regulating articular inflammation.


Assuntos
Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Inflamação/metabolismo , Lectinas Tipo C/metabolismo , Receptores de Superfície Celular/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Artrite Reumatoide/sangue , Artrite Reumatoide/genética , Proteína C-Reativa/metabolismo , Citocinas/sangue , Feminino , Citometria de Fluxo , Regulação da Expressão Gênica , Humanos , Imuno-Histoquímica , Inflamação/genética , Inflamação/patologia , Mediadores da Inflamação/sangue , Articulações/patologia , Lectinas Tipo C/genética , Leucócitos Mononucleares/metabolismo , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Pessoa de Meia-Idade , Monócitos/metabolismo , Monócitos/patologia , Osteoartrite/genética , Osteoartrite/metabolismo , Osteoartrite/patologia , Reação em Cadeia da Polimerase , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Superfície Celular/genética , Membrana Sinovial/metabolismo , Membrana Sinovial/patologia
18.
J Biomed Sci ; 20: 36, 2013 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-23742038

RESUMO

Macrophages (Mϕ) are the major source of inflammatory cytokines and are target cells for dengue virus (DV) replication. However, Mϕ are heterogeneous and their phenotypic and functional diversities are influenced by cytokines that regulate their differentiation, tissue distribution, and defense against invading pathogens. In vitro, human primary macrophages are derived from peripheral blood CD14+ monocytes in the presence of macrophage colony-stimulating factor (M-CSF) or granulocyte macrophage colony-stimulating factor (GM-CSF). These are essential for developing tissue/resting macrophages (M-Mϕ) and inflammatory macrophages (GM-Mϕ), respectively. While IFN production is similar between M-Mϕ and GM-Mϕ, M-Mϕ cannot produce IL-1ß after DV infection. In contrast, GM-Mϕ is more susceptible to DV infection and DV triggers CLEC5A in GM-Mϕ to activate NLRP3 inflammasomes, which in turn release IL-18 and IL-1ß that are critical for Th17 activation and contribute to disease severity. Thus, GM-Mϕ is more representative than M-Mϕ for investigating inflammasome activation in dengue infection, and is invaluable for revealing the molecular mechanism of pathogen-induced inflammatory reaction. Distinct phenotypes of macrophage subsets under the influence of M-CSF and GM-CSF raise the question of optimal conditions for culturing primary macrophages to study host-pathogen interaction.


Assuntos
Vírus da Dengue/imunologia , Inflamassomos/metabolismo , Macrófagos/metabolismo , Diferenciação Celular , Vírus da Dengue/patogenicidade , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Ativação de Macrófagos , Macrófagos/citologia , Macrófagos/imunologia
19.
Blood ; 121(1): 95-106, 2013 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-23152543

RESUMO

Persistent high fever is one of the most typical clinical symptoms in dengue virus (DV)-infected patients. However, the source of endogenous pyrogen (eg, IL-1ß) and the signaling cascade leading to the activation of inflammasome and caspase-1, which are essential for IL-1ß and IL-18 secretion, during dengue infection have not been elucidated yet. Macrophages can be polarized into distinct phenotypes under the influence of GM-CSF or M-CSF, denoted as GM-Mϕ and M-Mϕ, respectively. We found that DV induced high levels of IL-1ß and IL-18 from GM-Mϕ (inflammatory macrophage) and caused cell death (pyroptosis), whereas M-Mϕ (resting macrophage) did not produce IL-1ß and IL-18 on DV infection even with lipopolysaccharide priming. This observation demonstrates the distinct responses of GM-Mϕ and M-Mϕ to DV infection. Moreover, up-regulation of pro-IL-1ß, pro-IL-18, and NLRP3 associated with caspase-1 activation was observed in DV-infected GM-Mϕ, whereas blockade of CLEC5A/MDL-1, a C-type lectin critical for dengue hemorrhagic fever and Japanese encephalitis virus infection, inhibits NLRP3 inflammasome activation and pyrotopsis in GM-Mϕ. Thus, DV can activate NLRP3 inflammasome via CLEC5A, and GM-Mϕ plays a more important role than M-Mϕ in the pathogenesis of DV infection.


Assuntos
Vírus da Dengue/fisiologia , Dengue/imunologia , Inflamassomos/imunologia , Lectinas Tipo C/fisiologia , Ativação de Macrófagos , Macrófagos/imunologia , Receptores de Superfície Celular/fisiologia , Apoptose , Permeabilidade Capilar , Proteínas de Transporte/imunologia , Inibidores de Caspase/farmacologia , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Dengue/genética , Endotélio Vascular/fisiologia , Febre/etiologia , Febre/fisiopatologia , Regulação da Expressão Gênica/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Humanos , Interleucina-18/biossíntese , Interleucina-18/genética , Interleucina-1beta/biossíntese , Interleucina-1beta/genética , Lectinas Tipo C/antagonistas & inibidores , Lipopolissacarídeos/farmacologia , Fator Estimulador de Colônias de Macrófagos/farmacologia , Macrófagos/classificação , Macrófagos/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR , RNA Interferente Pequeno/farmacologia , Receptores de Superfície Celular/antagonistas & inibidores , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética , Replicação Viral
20.
Proteomics ; 12(21): 3251-62, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22945439

RESUMO

Prostate cancer has been known to be the second highest cause of death in cancer among men. Pomegranate is rich in polyphenols with the potent antioxidant activity and inhibits cell proliferation, invasion, and promotes apoptosis in various cancer cells. This study demonstrated that pomegranate fruit juice could effectively hinder the proliferation of human prostate cancer DU145 cell. The results of apoptotic analyses implicated that fruit juice might trigger the apoptosis in DU145 cells via death receptor signaling and mitochondrial damage pathway. In this study, we exploited 2DE-based proteomics to compare nine pairs of the proteome maps collected from untreated and treated DU145 cells to identify the differentially expressed proteins. Comparative proteomics indicated that 11 proteins were deregulated in affected DU145 cells with three upregulated and eight downregulated proteins. These dys-regulated proteins participated in cytoskeletal functions, antiapoptosis, proteasome activity, NF-κB signaling, cancer cell proliferation, invasion, and angiogenesis. Western immunoblotting were implemented to confirm the deregulated proteins and the downstream signaling proteins. The analytical results of this study help to provide insight into the molecular mechanism of inducing prostate cancer cell apoptosis by pomegranate fruit juice and to develop a novel mechanism-based chemopreventive strategy for prostate cancer.


Assuntos
Frutas , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Lythraceae , Extratos Vegetais/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/genética , Proteoma/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Bebidas , Western Blotting , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Eletroforese em Gel Bidimensional , Humanos , Masculino , Neoplasias da Próstata/metabolismo , Proteoma/análise , Proteoma/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...