Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Redox Biol ; 46: 102092, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34418598

RESUMO

Environmental inhalation exposures are inherently mixed (gases and particles), yet regulations are still based on single toxicant exposures. While the impacts of individual components of environmental pollution have received substantial attention, the impact of inhalation co-exposures is poorly understood. Here, we mechanistically investigated pulmonary inflammation and lung function decline after inhalation co-exposure and individual exposures to ozone (O3) and ultrafine carbon black (CB). Environmentally/occupationally relevant lung deposition levels in mice were achieved after inhalation of stable aerosols with similar aerodynamic and mass median distributions. X-ray photoemission spectroscopy detected increased surface oxygen contents on particles in co-exposure aerosols. Compared with individual exposures, co-exposure aerosols produced greater acellular and cellular oxidants detected by electron paramagnetic resonance (EPR) spectroscopy, and in vivo immune-spin trapping (IST), as well as synergistically increased lavage neutrophils, lavage proteins and inflammation related gene/protein expression. Co-exposure induced a significantly greater respiratory function decline compared to individual exposure. A synthetic catalase-superoxide dismutase mimetic (EUK-134) significantly blunted lung inflammation and respiratory function decline confirming the role of oxidant imbalance. We identified a significant induction of epithelial alarmin (thymic stromal lymphopoietin-TSLP)-dependent interleukin-13 pathway after co-exposure, associated with increased mucin and interferon gene expression. We provided evidence of interactive outcomes after air pollution constituent co-exposure and identified a key mechanistic pathway that can potentially explain epidemiological observation of lung function decline after an acute peak of air pollution. Developing and studying the co-exposure scenario in a standardized and controlled fashion will enable a better mechanistic understanding of how environmental exposures result in adverse outcomes.


Assuntos
Poluentes Atmosféricos , Ozônio , Pneumonia , Poluentes Atmosféricos/toxicidade , Alarminas/farmacologia , Animais , Carbono/farmacologia , Exposição por Inalação , Pulmão , Camundongos , Oxidantes/farmacologia , Ozônio/toxicidade , Tamanho da Partícula , Pneumonia/induzido quimicamente
3.
mBio ; 12(2)2021 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-33849978

RESUMO

Mammalian cells detect microbial molecules known as pathogen-associated molecular patterns (PAMPs) as indicators of potential infection. Upon PAMP detection, diverse defensive responses are induced by the host, including those that promote inflammation and cell-intrinsic antimicrobial activities. Host-encoded molecules released from dying or damaged cells, known as damage-associated molecular patterns (DAMPs), also induce defensive responses. Both DAMPs and PAMPs are recognized for their inflammatory potential, but only the latter are well established to stimulate cell-intrinsic host defense. Here, we report a class of DAMPs that engender an antiviral state in human epithelial cells. These DAMPs include oxPAPC (oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine), PGPC (1-palmitoyl-2-glutaryl phosphatidylcholine), and POVPC [1-palmitoyl-2-(5-oxovaleroyl)-sn-glycero-3-phosphatidylcholine], oxidized lipids that are naturally released from dead or dying cells. Exposing cells to these DAMPs prior to vesicular stomatitis virus (VSV) infection limits viral replication. Mechanistically, these DAMPs prevent viral entry, thereby limiting the percentage of cells that are productively infected and consequently restricting viral load. We found that the antiviral actions of oxidized lipids are distinct from those mediated by the PAMP Poly I:C, in that the former induces a more rapid antiviral response without the induction of the interferon response. These data support a model whereby interferon-independent defensive activities can be induced by DAMPs, which may limit viral replication before PAMP-mediated interferon responses are induced. This antiviral activity may impact viruses that disrupt interferon responses in the oxygenated environment of the lung, such as influenza virus and SARS-CoV-2.IMPORTANCE In this work, we explored how a class of oxidized lipids, spontaneously created during tissue damage and unprogrammed cell lysis, block the earliest events in RNA virus infection in the human epithelium. This gives us novel insight into the ways that we view infection models, unveiling a built-in mechanism to slow viral growth that neither engages the interferon response nor is subject to known viral antagonism. These oxidized phospholipids act prior to infection, allowing time for other, better-known innate immune mechanisms to take effect. This discovery broadens our understanding of host defenses, introducing a soluble factor that alters the cellular environment to protect from RNA virus infection.


Assuntos
Alarminas/farmacologia , Antivirais/farmacologia , Vírus de RNA/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Células A549 , Morte Celular/efeitos dos fármacos , Humanos , Imunidade Inata , Interferons/genética , Interferons/metabolismo , Cinética , Moléculas com Motivos Associados a Patógenos/farmacologia , Fosfatidilcolinas/farmacologia , Vírus de RNA/fisiologia , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/fisiologia , Vesiculovirus/efeitos dos fármacos , Vesiculovirus/fisiologia , Carga Viral
4.
Cell Host Microbe ; 29(2): 222-235.e4, 2021 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-33388094

RESUMO

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic poses an unprecedented public health crisis. Evidence suggests that SARS-CoV-2 infection causes dysregulation of the immune system. However, the unique signature of early immune responses remains elusive. We characterized the transcriptome of rhesus macaques and mice infected with SARS-CoV-2. Alarmin S100A8 was robustly induced in SARS-CoV-2-infected animal models as well as in COVID-19 patients. Paquinimod, a specific inhibitor of S100A8/A9, could rescue the pneumonia with substantial reduction of viral loads in SARS-CoV-2-infected mice. Remarkably, Paquinimod treatment resulted in almost 100% survival in a lethal model of mouse coronavirus infection using the mouse hepatitis virus (MHV). A group of neutrophils that contributes to the uncontrolled pathological damage and onset of COVID-19 was dramatically induced by coronavirus infection. Paquinimod treatment could reduce these neutrophils and regain anti-viral responses, unveiling key roles of S100A8/A9 and aberrant neutrophils in the pathogenesis of COVID-19, highlighting new opportunities for therapeutic intervention.


Assuntos
Alarminas/farmacologia , Antivirais/farmacologia , Tratamento Farmacológico da COVID-19 , Neutrófilos/efeitos dos fármacos , SARS-CoV-2/efeitos dos fármacos , Animais , COVID-19/metabolismo , COVID-19/virologia , Modelos Animais de Doenças , Feminino , Humanos , Macaca mulatta , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/metabolismo , Transcriptoma , Carga Viral
5.
Brain Behav Immun ; 82: 319-337, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31505255

RESUMO

High mobility group box 1 (HMGB1) is an endogenous alarmin that drives the pathogenesis of neurodegenerative disorders including cognitive decline. Therefore, HMGB1 is thought to be a common biomarker as well as promising therapeutic target for neuroinflammation associated with neurocognitive disorders. Here, for the first time, we have unmasked the potential inhibitory effect of a novel receptor of HMGB1-CXCL12 complex; atypical chemokine receptor 3 (ACKR3/CXCR7) on HMGB1 induced glial phenotype switching, neuroinflammation, and subsequent memory loss. Upregulation of CXCR7 inhibits HMGB1-CXCL12 complex induced peripheral immune cells infiltration to CNS by regulating blood-brain barrier (BBB) integrity in HMGB1 induced dementia model of mice. Whereas, gene knockdown study by RNA interference (non-invasive intranasal delivery to animal model) shows CXCR7 ablation aggravates inflammatory responses in hippocampus region and immune cell infiltration to CNS tissue by breached BBB. This study also indicates the important role of CXCR7 molecule in maintaining CNS homeostasis by balancing M1/M2 microglia, A1/A2 astrocytes, long term potentiation/long term depression markers which ultimately ameliorates HMGB1 induced neurodegeneration, synaptic depression and memory loss (assessed by both radial arm maze and Morris water maze) in male mice model of dementia. Overall, the study summarizes several significant protective functions afforded by CXCR7 against HMGB1 induced disbalance in neuroimmunological axis, neurodegeneration and memory loss and thereby provides a new paradigm for strategic development of novel therapeutics against neurodegenerative diseases with dementia as a common symptom.


Assuntos
Alarminas/farmacologia , Transtornos da Memória/metabolismo , Receptores CXCR/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Quimiocina CXCL12/metabolismo , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/patologia , Feminino , Masculino , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/patologia , Camundongos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Microglia/patologia , Neuroglia/metabolismo , Neuroimunomodulação/fisiologia , Neuroproteção , Receptores CXCR/agonistas , Receptores CXCR4/metabolismo , Proteínas Recombinantes/farmacologia , Transdução de Sinais
6.
Jpn J Ophthalmol ; 62(1): 92-100, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29094325

RESUMO

PURPOSE: Sterile ulceration is frequently observed in the cornea following persistent corneal epithelial damage. We examined the effect of alarmins released by necrotic corneal epithelial cells (HCE) on the production of matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) by corneal fibroblasts. METHODS: IL-1α and high-mobility group box 1 protein (HMGB1) released into the supernatant derived from necrotic HCE cells were measured with enzyme-linked immunosorbent assay (ELISA). MMPs and TIMPs produced by corneal fibroblasts, stimulated with the supernatant from necrotic HCE cells, were analyzed and measured with protein array and ELISA. To investigate dynamic expression of alarmins in the corneal epithelium, we used immunohistochemistry to observe the expression of human IL-1α in the corneal epithelium of human IL-1α Tg mice with or without cryopexy. We also investigated the expression of MMPs in corneal stroma of the mice treated with cryopexy, using RT-PCR. RESULTS: We detected IL-1α and HMGB-1 in the supernatant of necrotic HCE cells. These supernatants increased the expression of MMP-3 and MMP-1, and decreased that of TIMP-1 and TIMP-2 in human corneal fibroblasts. Almost always these were inhibited by IL-1 receptor antagonist. Recombinant IL-1α increased the production MMP-3 and MMP-1 in corneal fibroblasts. After cryopexy of the epithelium of human IL-1α Tg mice, the expression of human IL-1α was recognized in the cytoplasm but not nucleus of epithelial cells. The level of MMP-3 and MMP-1 mRNAs was elevated in the corneal stroma in mice treated with cryopexy. CONCLUSION: Alarmins, especially IL-1α, released from necrotic HCE cells may play an important role in the expression of MMPs and TIMPs by corneal fibroblast, resulting in sterile ulceration.


Assuntos
Alarminas/farmacologia , Ceratócitos da Córnea/efeitos dos fármacos , Epitélio Corneano/patologia , Metaloproteinases da Matriz/metabolismo , Inibidores Teciduais de Metaloproteinases/metabolismo , Alarminas/metabolismo , Animais , Células Cultivadas , Ceratócitos da Córnea/metabolismo , Meios de Cultura , Ensaio de Imunoadsorção Enzimática , Epitélio Corneano/metabolismo , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Proteína HMGB1/metabolismo , Proteína HMGB1/farmacologia , Humanos , Interleucina-1alfa/metabolismo , Interleucina-1alfa/farmacologia , Metaloproteinases da Matriz/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Necrose , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Inibidores Teciduais de Metaloproteinases/genética
7.
Mediators Inflamm ; 2017: 2642549, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28804219

RESUMO

OBJECTIVE AND DESIGN: To investigate whether endogenous damage-associated molecular patterns (DAMPs) or alarmins originated from mitochondria or nucleus stimulates inflammatory response in articular chondrocytes to cause chondrolysis which leads to cartilage degradation featured in posttraumatic osteoarthritis (PTOA). MATERIALS: Primary cultures of bovine or human chondrocytes isolated from cartilage of weight-bearing joints. TREATMENT: Chondrocytes were subjected to mitochondrial DAMPs (MTDs) or HMGB1, a nuclear DAMP (NuD), with or without the presence of an N-terminal 29 kDa fibronectin fragment (Fn-f) or proinflammatory cytokines (IL-1ß and TNF-α). Injured cartilage-conditioned culturing medium containing a mixture of DAMPs was employed as a control. After 24 hrs, the protein expression of cartilage degrading metalloproteinases and iNOS in culture medium or cell lysates was examined with Western blotting, respectively. RESULTS: HMGB1 was synergized with IL-1ß in upregulating expression of MMP-3, MMP-13, ADAMTS-5, ADAM-8, and iNOS. Moreover, a moderate synergistic effect was detected between HMGB1 and Fn-f or between MTDs and TNF-α on MMP-3 expression. However, when acting alone, MTDs or HMGB1 did not upregulate cartilage degrading enzymes or iNOS. CONCLUSION: MTDs or HMGB1 could only stimulate inflammatory response in chondrocytes with the presence of cytokines or Fn-f.


Assuntos
Alarminas/farmacologia , Condrócitos/citologia , Condrócitos/efeitos dos fármacos , Citocinas/farmacologia , Fibronectinas/farmacologia , Proteínas ADAM/metabolismo , Proteína ADAMTS5/metabolismo , Animais , Bovinos , Células Cultivadas , Proteína HMGB1/metabolismo , Humanos , Interleucina-1beta/metabolismo , Metaloproteinase 13 da Matriz/metabolismo , Metaloproteinase 3 da Matriz/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
8.
Acta Derm Venereol ; 97(3): 305-312, 2017 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-27868148

RESUMO

The epidermal-derived "alarmins" high-mobility group box 1 (HMGB1) protein and interleukin-33 (IL-33) are upregulated in patients with atopic dermatitis. How-ever, their capacity as pro-inflammatory cytokines and their derived effects on skin barrier regulation are poorly elucidated. We investigated the impact of HMGB1 and IL-33 on gene transcription, protein expression and epidermal differentiation across 3 distinct keratinocyte in vitro models. Primary keratinocytes from healthy donors were used in submerged monolayer cultures, 3D human epidermis equivalents and 3D human skin equivalents. All keratinocyte models underwent 96-h stimulation with HMGB1 (100 µM) or IL-33 (100 ng/ml) using IL-4 (50 ng/ml) as positive control of regulation and vehicle as negative control. We found that HMGB1 and IL-33 downregulated transcription of several genes from members of the epidermal differentiation complex, including filaggrin. Furthermore, HMGB1 downregulated the expression of the encoded proteins in the upper epidermis. Finally, IL-33 and HMGB1 ultimately led to impaired epidermal growth and maturation. In conclusion, HMGB1 and IL-33 could play a significant role in the atopic dermatitis pathophysiology due to negative regulation of structural proteins, stratum corneum formation and epidermal growth.


Assuntos
Alarminas/farmacologia , Proliferação de Células/efeitos dos fármacos , Epiderme/efeitos dos fármacos , Proteína HMGB1/farmacologia , Interleucina-33/farmacologia , Queratinócitos/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Alarminas/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Dermatite Atópica/genética , Dermatite Atópica/metabolismo , Dermatite Atópica/patologia , Relação Dose-Resposta a Droga , Regulação para Baixo , Epiderme/crescimento & desenvolvimento , Epiderme/metabolismo , Epiderme/patologia , Proteínas Filagrinas , Proteína HMGB1/metabolismo , Voluntários Saudáveis , Humanos , Interleucina-33/metabolismo , Queratinócitos/metabolismo , Queratinócitos/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos
9.
PLoS One ; 11(6): e0156486, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27310139

RESUMO

BACKGROUND: Prothymosin α (ProTα) (isoform 2: iso2) is a widely distributed, small acidic protein with intracellular and extracellular-associated functions. Recently, we identified two new ProTα variants with potent anti-HIV activity from CD8+ T cells and cervicovaginal lavage. The first is a splice variant of the ProTα gene known as isoB and the second is the product of ProTα pseudogene 7 (p7). Similarly to iso2, the anti-HIV activity of both variants is mediated by type I IFN. Here we tested whether the immunomodulatory activity of isoB and p7 are also TLR4 dependent and determined their kinetic of release in response to HIV-1 infection. METHODS: Type I, type III, TNF-α and IL-6 mRNA inducing activity was determined in macrophages from wild type and TLR4 knockout mice treated with recombinant ProTα variants. Supernatants from mock and HIV infected cells were analyzed by mass spectrometry in positive and negative modes for the presence of ProTα variants. In silico structural and functional analysis of ProTα variants were performed. RESULTS: We show that both isoB and p7 upregulate IFN-ß, IFN-λ1, IL-6, TNF-α and RANTES mRNAs in primary human macrophages. The potent stimulation of IFN-ß by the recombinant ProTα variants in human macrophages is dependent on the TLR4 pathway, whereas the induction of TNF-α and IL-6 may also occur independently of TLR4, suggesting the interaction of ProTα variants with other signaling molecules/receptors. In silico analyses confirmed that the novel isoB and p7 variants are intrinsically disordered proteins, which lack the NLS and mass spectrometry showed release of ProTα variants within minutes post HIV-1 infection. These features are consistent with the function of ProTα variants as damage associate molecular patterns (DAMPs). CONCLUSIONS: Our findings indicate that ProTα variants strongly inhibit viral replication mainly, but not exclusively, through TLR4 signaling and that they are released within minutes of viral infection suggesting that they may function as DAMPs.


Assuntos
Alarminas/farmacologia , Infecções por HIV/tratamento farmacológico , HIV-1/efeitos dos fármacos , Proteínas Intrinsicamente Desordenadas/farmacologia , Precursores de Proteínas/farmacologia , Timosina/análogos & derivados , Receptor 4 Toll-Like/imunologia , Alarminas/genética , Alarminas/imunologia , Sequência de Aminoácidos , Animais , Quimiocina CCL5/genética , Quimiocina CCL5/imunologia , Regulação da Expressão Gênica , Infecções por HIV/genética , Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV-1/crescimento & desenvolvimento , HIV-1/imunologia , Humanos , Interferon beta/genética , Interferon beta/imunologia , Interferons , Interleucina-6/genética , Interleucina-6/imunologia , Interleucinas/genética , Interleucinas/imunologia , Proteínas Intrinsicamente Desordenadas/genética , Proteínas Intrinsicamente Desordenadas/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/virologia , Camundongos , Camundongos Knockout , Cultura Primária de Células , Ligação Proteica , Mapeamento de Interação de Proteínas , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Isoformas de Proteínas/farmacologia , Precursores de Proteínas/genética , Precursores de Proteínas/imunologia , Alinhamento de Sequência , Transdução de Sinais , Timosina/genética , Timosina/imunologia , Timosina/farmacologia , Receptor 4 Toll-Like/deficiência , Receptor 4 Toll-Like/genética , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
10.
Arthritis Rheumatol ; 68(1): 152-63, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26360647

RESUMO

OBJECTIVE: Both alarmins S100A8/A9 and canonical Wnt signaling have been found to play active roles in the development of experimental osteoarthritis (OA). However, what activates canonical Wnt signaling remains unknown. This study was undertaken to investigate whether S100A8 induces canonical Wnt signaling and whether S100 proteins exert their effects via activation of Wnt signaling. METHODS: Expression of the genes for S100A8/A9 and Wnt signaling pathway members was measured in an experimental OA model. Selected Wnt signaling pathway members were overexpressed, and levels of S100A8/A9 were measured. Activation of canonical Wnt signaling was determined after injection of S100A8 into naive joints and induction of collagenase-induced OA in S100A9-deficient mice. Expression of Wnt signaling pathway members was tested in macrophages and fibroblasts after S100A8 stimulation. Canonical Wnt signaling was inhibited in vivo to determine if the effects of S100A8 injections were dependent on Wnt signaling. RESULTS: The alarmins S100A8/A9 and members of the Wnt signaling pathway showed coinciding expression in synovial tissue in an experimental OA model. Synovial overexpression of selected Wnt signaling pathway members did not result in increased expression of S100 proteins. In contrast, intraarticular injection of S100A8 increased canonical Wnt signaling, whereas canonical Wnt signaling was decreased after induction of experimental OA in S100A9-deficient mice. S100A8 stimulation of macrophages, but not fibroblasts, resulted in increased expression of canonical Wnt signaling members. Overexpression of Dkk-1 to inhibit canonical Wnt signaling decreased the induction of matrix metalloproteinase 3, interleukin-6, and macrophage inflammatory protein 1α after injection of S100A8. CONCLUSION: Our findings indicate that the alarmin S100A8 induces canonical Wnt signaling in macrophages and murine knee joints. The effects of S100A8 are partially dependent on activation of canonical Wnt signaling.


Assuntos
Artrite Experimental/genética , Calgranulina A/genética , Calgranulina B/genética , Macrófagos/metabolismo , Osteoartrite do Joelho/genética , Joelho de Quadrúpedes/metabolismo , Membrana Sinovial/metabolismo , Via de Sinalização Wnt/genética , Alarminas/farmacologia , Animais , Artrite Experimental/induzido quimicamente , Artrite Experimental/metabolismo , Quimiocina CCL3/efeitos dos fármacos , Quimiocina CCL3/metabolismo , Colagenases/toxicidade , Modelos Animais de Doenças , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intercelular/genética , Interleucina-6/metabolismo , Macrófagos/efeitos dos fármacos , Metaloproteinase 3 da Matriz/efeitos dos fármacos , Metaloproteinase 3 da Matriz/metabolismo , Camundongos , Camundongos Knockout , Osteoartrite do Joelho/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...