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








Base de dados
Intervalo de ano de publicação
1.
mBio ; 15(2): e0292823, 2024 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-38193729

RESUMO

Serum titers of SARS-CoV-2-neutralizing antibodies (nAbs) correlate well with protection from symptomatic COVID-19 but decay rapidly in the months following vaccination or infection. In contrast, measles-protective nAb titers are lifelong after measles vaccination, possibly due to persistence of the live-attenuated virus in lymphoid tissues. We, therefore, sought to generate a live recombinant measles vaccine capable of driving high SARS-CoV-2 nAb responses. Since previous clinical testing of a live measles vaccine encoding a SARS-CoV-2 spike glycoprotein resulted in suboptimal anti-spike antibody titers, our new vectors were designed to encode prefusion-stabilized SARS-CoV-2 spike glycoproteins, trimerized via an inserted peptide domain, and displayed on a dodecahedral miniferritin scaffold. Additionally, to circumvent the blunting of vaccine efficacy by preformed anti-measles antibodies, we extensively modified the measles surface glycoproteins. Comprehensive in vivo mouse testing demonstrated the potent induction of high titer nAbs in measles-immune mice and confirmed the significant contributions to overall potency afforded by prefusion stabilization, trimerization, and miniferritin display of the SARS-CoV-2 spike glycoprotein. In animals primed and boosted with a measles virus (MeV) vaccine encoding the ancestral SARS-CoV-2 spike, high-titer nAb responses against ancestral virus strains were only weakly cross-reactive with the Omicron variant. However, in primed animals that were boosted with a MeV vaccine encoding the Omicron BA.1 spike, antibody titers to both ancestral and Omicron strains were robustly elevated, and the passive transfer of serum from these animals protected K18-ACE2 mice from infection and morbidity after exposure to BA.1 and WA1/2020 strains. Our results demonstrate that by engineering the antigen, we can develop potent measles-based vaccine candidates against SARS-CoV-2.IMPORTANCEAlthough the live-attenuated measles virus (MeV) is one of the safest and most efficacious human vaccines, a measles-vectored COVID-19 vaccine candidate expressing the SARS-CoV-2 spike failed to elicit neutralizing antibody (nAb) responses in a phase-1 clinical trial, especially in measles-immune individuals. Here, we constructed a comprehensive panel of MeV-based COVID-19 vaccine candidates using a MeV with extensive modifications on the envelope glycoproteins (MeV-MR). We show that artificial trimerization of the spike is critical for the induction of nAbs and that their magnitude can be significantly augmented when the spike protein is synchronously fused to a dodecahedral scaffold. Furthermore, preexisting measles immunity did not abolish heterologous immunity elicited by our vector. Our results highlight the importance of antigen optimization in the development of spike-based COVID-19 vaccines and therapies.


Assuntos
COVID-19 , Sarampo , Humanos , Animais , Camundongos , Vacinas contra COVID-19 , Anticorpos Neutralizantes , SARS-CoV-2/genética , Glicoproteína da Espícula de Coronavírus/genética , COVID-19/prevenção & controle , Vacina contra Sarampo/genética , Vírus do Sarampo/genética , Anticorpos Antivirais , Glicoproteínas de Membrana
2.
Mol Ther ; 32(1): 241-256, 2024 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-37927036

RESUMO

Oncolytic virotherapy aims to activate host antitumor immunity. In responsive tumors, intratumorally injected herpes simplex viruses (HSVs) have been shown to lyse tumor cells, resulting in local inflammation, enhanced tumor antigen presentation, and boosting of antitumor cytotoxic lymphocytes. In contrast to HSV, cytomegalovirus (CMV) is nonlytic and reprograms infected myeloid cells, limiting their antigen-presenting functions and protecting them from recognition by natural killer (NK) cells. Here, we show that when co-injected into mouse tumors with an oncolytic HSV, mouse CMV (mCMV) preferentially targeted tumor-associated myeloid cells, promoted the local release of proinflammatory cytokines, and enhanced systemic antitumor immune responses, leading to superior control of both injected and distant contralateral tumors. Deletion of mCMV genes m06, which degrades major histocompatibility complex class I (MHC class I), or m144, a viral MHC class I homolog that inhibits NK activation, was shown to diminish the antitumor activity of the HSV/mCMV combination. However, an mCMV recombinant lacking the m04 gene, which escorts MHC class I to the cell surface, showed superior HSV adjuvanticity. CMV is a potentially promising agent with which to reshape and enhance antitumor immune responses following oncolytic HSV therapy.


Assuntos
Infecções por Citomegalovirus , Herpesvirus Humano 1 , Neoplasias , Terapia Viral Oncolítica , Vírus Oncolíticos , Animais , Camundongos , Herpesvirus Humano 1/genética , Citomegalovirus , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Apresentação de Antígeno , Vírus Oncolíticos/genética , Vírus Oncolíticos/metabolismo
3.
Front Immunol ; 14: 1279387, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38022659

RESUMO

Introduction: Metastatic uveal melanoma (MUM) has a poor prognosis and treatment options are limited. These patients do not typically experience durable responses to immune checkpoint inhibitors (ICIs). Oncolytic viruses (OV) represent a novel approach to immunotherapy for patients with MUM. Methods: We developed an OV with a Vesicular Stomatitis Virus (VSV) vector modified to express interferon-beta (IFN-ß) and Tyrosinase Related Protein 1 (TYRP1) (VSV-IFNß-TYRP1), and conducted a Phase 1 clinical trial with a 3 + 3 design in patients with MUM. VSV-IFNß-TYRP1 was injected into a liver metastasis, then administered on the same day as a single intravenous (IV) infusion. The primary objective was safety. Efficacy was a secondary objective. Results: 12 patients with previously treated MUM were enrolled. Median follow up was 19.1 months. 4 dose levels (DLs) were evaluated. One patient at DL4 experienced dose limiting toxicities (DLTs), including decreased platelet count (grade 3), increased aspartate aminotransferase (AST), and cytokine release syndrome (CRS). 4 patients had stable disease (SD) and 8 patients had progressive disease (PD). Interferon gamma (IFNγ) ELIspot data showed that more patients developed a T cell response to virus encoded TYRP1 at higher DLs, and a subset of patients also had a response to other melanoma antigens, including gp100, suggesting epitope spreading. 3 of the patients who responded to additional melanoma antigens were next treated with ICIs, and 2 of these patients experienced durable responses. Discussion: Our study found that VSV-IFNß -TYRP1 can be safely administered via intratumoral (IT) and IV routes in a previously treated population of patients with MUM. Although there were no clear objective radiographic responses to VSV-IFNß-TYRP1, dose-dependent immunogenicity to TYRP1 and other melanoma antigens was seen.


Assuntos
Terapia Viral Oncolítica , Vírus Oncolíticos , Estomatite Vesicular , Animais , Humanos , Interferon beta/metabolismo , Antígenos Específicos de Melanoma , Monofenol Mono-Oxigenase/metabolismo , Terapia Viral Oncolítica/efeitos adversos , Vírus Oncolíticos/genética , Linfócitos T/metabolismo , Vírus da Estomatite Vesicular Indiana
4.
bioRxiv ; 2022 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-36561187

RESUMO

Serum titers of SARS-CoV-2 neutralizing antibodies (nAb) correlate well with protection from symptomatic COVID-19, but decay rapidly in the months following vaccination or infection. In contrast, measles-protective nAb titers are life-long after measles vaccination, possibly due to persistence of the live-attenuated virus in lymphoid tissues. We therefore sought to generate a live recombinant measles vaccine capable of driving high SARS-CoV-2 nAb responses. Since previous clinical testing of a live measles vaccine encoding a SARS-CoV-2 spike glycoprotein resulted in suboptimal anti-spike antibody titers, our new vectors were designed to encode prefusion-stabilized SARS-CoV-2 spike glycoproteins, trimerized via an inserted peptide domain and displayed on a dodecahedral miniferritin scaffold. Additionally, to circumvent the blunting of vaccine efficacy by preformed anti-measles antibodies, we extensively modified the measles surface glycoproteins. Comprehensive in vivo mouse testing demonstrated potent induction of high titer nAb in measles-immune mice and confirmed the significant incremental contributions to overall potency afforded by prefusion stabilization, trimerization, and miniferritin-display of the SARS-CoV-2 spike glycoprotein, and vaccine resurfacing. In animals primed and boosted with a MeV vaccine encoding the ancestral SARS-CoV-2 spike, high titer nAb responses against ancestral virus strains were only weakly cross-reactive with the omicron variant. However, in primed animals that were boosted with a MeV vaccine encoding the omicron BA.1 spike, antibody titers to both ancestral and omicron strains were robustly elevated and the passive transfer of serum from these animals protected K18-ACE2 mice from infection and morbidity after exposure to BA.1 and WA1/2020 strains. Our results demonstrate that antigen engineering can enable the development of potent measles-based SARS-CoV-2 vaccine candidates.

5.
Vaccine ; 40(15): 2342-2351, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35282925

RESUMO

An orally active vaccine capable of boosting SARS-CoV-2 immune responses in previously infected or vaccinated individuals would help efforts to achieve and sustain herd immunity. Unlike mRNA-loaded lipid nanoparticles and recombinant replication-defective adenoviruses, replicating vesicular stomatitis viruses with SARS-CoV-2 spike glycoproteins (VSV-SARS2) were poorly immunogenic after intramuscular administration in clinical trials. Here, by G protein trans-complementation, we generated VSV-SARS2(+G) virions with expanded target cell tropism. Compared to parental VSV-SARS2, G-supplemented viruses were orally active in virus-naive and vaccine-primed cynomolgus macaques, powerfully boosting SARS-CoV-2 neutralizing antibody titers. Clinical testing of this oral VSV-SARS2(+G) vaccine is planned.


Assuntos
COVID-19 , Rhabdoviridae , Vacinas Virais , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais , COVID-19/prevenção & controle , Lipossomos , Nanopartículas , Primatas , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus/genética
6.
Blood Adv ; 6(11): 3268-3279, 2022 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-35175355

RESUMO

Clinical success with intravenous (IV) oncolytic virotherapy (OV) has to-date been anecdotal. We conducted a phase 1 clinical trial of systemic OV and investigated the mechanisms of action in responding patients. A single IV dose of vesicular stomatitis virus (VSV) interferon-ß (IFN-ß) with sodium iodide symporter (NIS) was administered to patients with relapsed/refractory hematologic malignancies to determine safety and efficacy across 4 dose levels (DLs). Correlative studies were undertaken to evaluate viremia, virus shedding, virus replication, and immune responses. Fifteen patients received VSV-IFNß-NIS. Three patients were treated at DL1 through DL3 (0.05, 0.17, and 0.5 × 1011 TCID50), and 6 were treated at DL4 (1.7 × 1011 TCID50) with no dose-limiting toxicities. Three of 7 patients with T-cell lymphoma (TCL) had responses: a 3-month partial response (PR) at DL2, a 6-month PR, and a complete response (CR) ongoing at 20 months at DL4. Viremia peaked at the end of infusion, g was detected. Plasma IFN-ß, a biomarker of VSV-IFNß-NIS replication, peaked between 4 hours and 48 hours after infusion. The patient with CR had robust viral replication with increased plasma cell-free DNA, high peak IFN-ß of 18 213 pg/mL, a strong anti-VSV neutralizing antibody response, and increased numbers of tumor reactive T-cells. VSV-IFNß-NIS as a single agent was effective in patients with TCL, resulting in durable disease remissions in heavily pretreated patients. Correlative analyses suggest that responses may be due to a combination of direct oncolytic tumor destruction and immune-mediated tumor control. This trial is registered at www.clinicaltrials.gov as #NCT03017820.


Assuntos
Linfoma de Células T , Terapia Viral Oncolítica , Humanos , Interferon beta/genética , Recidiva Local de Neoplasia , Terapia Viral Oncolítica/métodos , Vírus da Estomatite Vesicular Indiana/genética , Viremia/etiologia
7.
Leukemia ; 34(12): 3310-3322, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32327728

RESUMO

Oncolytic virus therapy leads to immunogenic death of virus-infected tumor cells and this has been shown in preclinical models to enhance the cytotoxic T-lymphocyte response against tumor-associated antigens (TAAs), leading to killing of uninfected tumor cells. To investigate whether oncolytic virotherapy can increase immune responses to tumor antigens in human subjects, we studied T-cell responses against a panel of known myeloma TAAs using PBMC samples obtained from ten myeloma patients before and after systemic administration of an oncolytic measles virus encoding sodium iodide symporter (MV-NIS). Despite their prior exposures to multiple immunosuppressive antimyeloma treatment regimens, T-cell responses to some of the TAAs were detectable even before measles virotherapy. Measurable baseline T-cell responses against MAGE-C1 and hTERT were present. Furthermore, MV-NIS treatment significantly (P < 0.05) increased T-cell responses against MAGE-C1 and MAGE-A3. Interestingly, one patient who achieved complete remission after MV-NIS therapy had strong baseline T-cell responses both to measles virus proteins and to eight of the ten tested TAAs. Our data demonstrate that oncolytic virotherapy can function as an antigen agnostic vaccine, increasing cytotoxic T-lymphocyte responses against TAAs in patients with multiple myeloma, providing a basis for continued exploration of this modality in combination with immune checkpoint blockade.


Assuntos
Vírus do Sarampo/imunologia , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/terapia , Vírus Oncolíticos/imunologia , Antígenos de Neoplasias/imunologia , Células Cultivadas , Humanos , Leucócitos Mononucleares , Terapia Viral Oncolítica/métodos , Simportadores/imunologia , Linfócitos T/imunologia , Linfócitos T Citotóxicos/imunologia
8.
Mayo Clin Proc ; 94(9): 1834-1839, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31235278

RESUMO

Recent measles epidemics in US and European cities where vaccination coverage has declined are providing a harsh reminder for the need to maintain protective levels of immunity across the entire population. Vaccine uptake rates have been declining in large part because of public misinformation regarding a possible association between measles vaccination and autism for which there is no scientific basis. The purpose of this article is to address a new misinformed antivaccination argument-that measles immunity is undesirable because measles virus is protective against cancer. Having worked for many years to develop engineered measles viruses as anticancer therapies, we have concluded (1) that measles is not protective against cancer and (2) that its potential utility as a cancer therapy will be enhanced, not diminished, by prior vaccination.


Assuntos
Comunicação , Vírus do Sarampo/imunologia , Sarampo/epidemiologia , Sarampo/prevenção & controle , Terapia Viral Oncolítica/métodos , Vacinação/efeitos adversos , Criança , Pré-Escolar , Controle de Doenças Transmissíveis/organização & administração , Europa (Continente) , Feminino , Humanos , Masculino , Prevalência , Medição de Risco , Estados Unidos , Vacinação/métodos
9.
Inflammation ; 41(1): 328-336, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29127663

RESUMO

Sepsis continues to be a major healthcare issue with one of the highest mortality rates in intensive care units. Toll-like receptors are pattern recognition receptors that are intricately involved in the pathogenesis of sepsis. TLR3 is a major receptor for double-stranded RNA and is largely associated with immunity to viral infection. In this study, we examined the role of TLR3 priming in the immunopathology of sepsis using cecal-ligation and puncture (CLP) model of sepsis in mice. Mice injected with vehicle or poly(I:C) were subjected to sham or CLP surgery and various parameters of sepsis, including mortality, inflammation, and bacterial clearance were assessed. Poly(I:C) pre-treatment significantly enhanced mortality in mice subjected to CLP. Consistent with this, inflammatory cytokines including TNFα, IL-12p40, IFNγ, and MCP-1 were enhanced both systemically and locally in the poly(I:C)-treated group compared to the vehicle control. In addition, bacterial load was significantly higher in the poly(I:C)-treated septic mice. These changes were associated with reduced macrophage activation (but not neutrophils) in the peritoneal cavity of poly(I:C) pre-treated mice compared to vehicle pre-treatment. Together our results demonstrate that poly(I:C) priming in sepsis is likely to be detrimental to the host due to effects on systemic inflammatory cytokines and bacterial clearance.


Assuntos
Ceco/microbiologia , Poli I-C/toxicidade , Sepse/induzido quimicamente , Animais , Carga Bacteriana , Ceco/cirurgia , Citocinas/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Ligadura , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos Endogâmicos C57BL , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Neutrófilos/metabolismo , Neutrófilos/microbiologia , Punções , Sepse/imunologia , Sepse/metabolismo , Sepse/microbiologia , Fatores de Tempo
10.
Physiol Genomics ; 49(11): 682-689, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-28939643

RESUMO

G protein-coupled receptor kinase-6 (GRK6) is a serine/threonine kinase that is important in inflammatory processes. In this study, we examined the role of GRK6 in Escherichia coli-induced lung infection and inflammation using GRK6 knockout (KO) and wild-type (WT) mice. Intratracheal instillation of E. coli significantly enhanced bacterial load in the bronchoalveolar lavage (BAL) of KO compared with WT mice. Reduced bacterial clearance in the KO mice was not due to an intrinsic defect in neutrophil phagocytosis or killing but as a result of reduced neutrophil numbers in the KO BAL. Interestingly, neutrophil numbers in the lung were increased in the KO compared with WT mice, suggesting a potential dysfunction in transepithelial migration of neutrophils from the lungs to the bronchoalveolar space. This effect was selective for lung tissue because peritoneal neutrophil numbers were similar between the two genotypes following peritoneal infection. Although neutrophil expression of CXCR2/CXCR3 was similar between WT and KO, IL-17A expression was higher in the KO compared with WT mice. These results suggest that enhanced neutrophil count in the KO lungs but reduced numbers in BAL are likely due to transepithelial migration defect and/or altered chemokines/cytokines. Together, our studies suggest a previously unrecognized and novel role for GRK6 in neutrophil migration specific to pulmonary tissue during bacterial infection.


Assuntos
Infecções por Escherichia coli/enzimologia , Infecções por Escherichia coli/microbiologia , Quinases de Receptores Acoplados a Proteína G/metabolismo , Pneumopatias/enzimologia , Pneumopatias/microbiologia , Animais , Apoptose/genética , Carga Bacteriana , Líquido da Lavagem Broncoalveolar/citologia , Modelos Animais de Doenças , Infecções por Escherichia coli/genética , Infecções por Escherichia coli/patologia , Regulação da Expressão Gênica , Inflamação/genética , Inflamação/patologia , Pneumopatias/genética , Pneumopatias/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Viabilidade Microbiana , Neutrófilos/metabolismo , Fagocitose , Receptores de Quimiocinas/metabolismo
11.
World J Gastroenterol ; 23(19): 3427-3439, 2017 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-28596679

RESUMO

AIM: To determine how a normal human colon cell line reacts to microbial challenge as a way to study oxidative stress-induced responses associated with inflammatory bowel disease. METHODS: Normal human colon epithelial cells (ATCC® CRL.1790™) were stimulated with either heat killed E. coli or heat killed murine cecal contents (HKC) and examined for several relevant biomarkers associated with inflammation and oxidative stress including cytokine production, mitochondrial autophagy and oxidant status. TNFα, IL-1ß and IL-8 protein concentrations were measured within the supernatants. Fluorescent microscopy was performed to quantify the production of reactive oxygen species (ROS) using an oxidation responsive fluorogenic probe. Mitochondrial morphology and mitochondrial membrane potential was assessed by dual staining using COXIV antibody and a dye concentrating in active mitochondria. Mitochondrial ROS scavenger was used to determine the source of ROS in stimulated cells. Autophagy was detected by staining for the presence of autophagic vesicles. Positive controls for autophagy and ROS/RNS experiments were treated with rapamycin and chloroquine. Mitochondrial morphology, ROS production and autophagy microscopy experiments were analyzed using a custom acquisition and analysis microscopy software (ImageJ). RESULTS: Exposing CRL.1790 cells to microbial challenge stimulated cells to produce several relevant biomarkers associated with inflammation and oxidative stress. Heat killed cecal contents treatment induced a 10-12 fold increase in IL-8 production by CRL.1790 cells compared to unstimulated controls at 6 and 12 h (P < 0.001). Heat killed E. coli stimulation resulted in a 4-5 fold increase in IL-8 compared to the unstimulated control cells at each time point (P < 0.001). Both heat killed E. coli and HKC stimulated robust ROS production at 6 (P < 0.001), and 12 h (P < 0.01). Mitochondrial morphologic abnormalities were detected at 6 and 12 h based on reduced mitochondrial circularity and decreased mitochondrial membrane potential, P < 0.01. Microbial stimulation also induced significant autophagy at 6 and 12 h, P < 0.01. Lastly, blocking mitochondrial ROS generation using mitochondrial specific ROS scavenger reversed microbial challenge induced mitochondrial morphologic abnormalities and autophagy. CONCLUSION: The findings from this study suggest that CRL.1790 cells may be a useful alternative to other colon cancer cell lines in studying the mechanisms of oxidative stress events associated with intestinal inflammatory disorders.


Assuntos
Colo/citologia , Células Epiteliais/citologia , Doenças Inflamatórias Intestinais/microbiologia , Mitocôndrias/patologia , Estresse Oxidativo , Animais , Autofagia , Biomarcadores/metabolismo , Ceco/microbiologia , Linhagem Celular , Linhagem Celular Tumoral , Quimiocinas/metabolismo , Citocinas/metabolismo , Escherichia coli/metabolismo , Temperatura Alta , Humanos , Inflamação , Doenças Inflamatórias Intestinais/fisiopatologia , Potencial da Membrana Mitocondrial , Camundongos , Microscopia de Fluorescência , Espécies Reativas de Oxigênio/metabolismo
12.
Mediators Inflamm ; 2017: 4207928, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29445256

RESUMO

Urinary tract infection (UTI) is an extremely common infectious disease. Uropathogenic Escherichia coli (UPEC) is the predominant etiological agent of UTI. Asymptomatic bacteriuric E. coli (ABEC) strains successfully colonize the urinary tract resulting in asymptomatic bacteriuria (ABU) and do not induce symptoms associated with UTI. Oxylipids are key signaling molecules involved in inflammation. Based on the distinct clinical outcomes of E. coli colonization, we hypothesized that UPEC triggers the production of predominantly proinflammatory oxylipids and ABEC leads to production of primarily anti-inflammatory or proresolving oxylipids in the urinary tract. We performed quantitative detection of 39 oxylipid mediators with proinflammatory, anti-inflammatory, and proresolving properties, during UTI and ABU caused by genetically distinct E. coli strains in the murine urinary bladder. Our results reveal that infection with UPEC causes an increased accumulation of proinflammatory oxylipids as early as 6 h postinoculation, compared to controls. To the contrary, ABEC colonization leads to decreased accumulation of proinflammatory oxylipids at the early time point compared to UPEC infection but does not affect the level of proresolving oxylipids. This report represents the first comprehensive investigation on the oxylipidome during benign ABEC colonization observed in ABU and acute inflammation triggered by UPEC leading to UTI.


Assuntos
Infecções por Escherichia coli/etiologia , Mediadores da Inflamação/fisiologia , Lipídeos/fisiologia , Bexiga Urinária/microbiologia , Infecções Urinárias/etiologia , Animais , Ácidos Graxos/análise , Feminino , Lipídeos/análise , Lipoxigenase/fisiologia , Camundongos , Camundongos Endogâmicos CBA , Prostaglandina-Endoperóxido Sintases/fisiologia , Bexiga Urinária/química
13.
Mediators Inflamm ; 2016: 9867138, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27818578

RESUMO

Inflammation is an essential host response during bacterial infections such as bovine mastitis. Endothelial cells are critical for an appropriate inflammatory response and loss of vascular barrier integrity is implicated in the pathogenesis of Streptococcus uberis-induced mastitis. Previous studies suggested that accumulation of linoleic acid (LA) oxygenation products derived from 15-lipoxygenase-1 (15-LOX-1) metabolism could regulate vascular functions. The initial LA derivative from the 15-LOX-1 pathway, 13-hydroperoxyoctadecadienoic acid (HPODE), can induce endothelial death, whereas the reduced hydroxyl product, 13-hydroxyoctadecadienoic acid (HODE), is abundantly produced during vascular activation. However, the relative contribution of specific LA-derived metabolites on impairment of mammary endothelial integrity is unknown. Our hypothesis was that S. uberis-induced LA-derived 15-LOX-1 oxygenation products impair mammary endothelial barrier integrity by apoptosis. Exposure of bovine mammary endothelial cells (BMEC) to S. uberis did not increase 15-LOX-1 LA metabolism. However, S. uberis challenge of bovine monocytes demonstrated that monocytes may be a significant source of both 13-HPODE and 13-HODE during mastitis. Exposure of BMEC to 13-HPODE, but not 13-HODE, significantly reduced endothelial barrier integrity and increased apoptosis. Changing oxidant status by coexposure to an antioxidant during 13-HPODE treatment prevented adverse effects of 13-HPODE, including amelioration of apoptosis. A better understanding of how the oxidant status of the vascular microenvironment impacts endothelial barrier properties could lead to more efficacious treatments for S. uberis mastitis.


Assuntos
Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Ácidos Linoleicos/farmacologia , Peróxidos Lipídicos/farmacologia , Acetilcisteína/farmacologia , Animais , Apoptose/efeitos dos fármacos , Bovinos , Células Cultivadas , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Glândulas Mamárias Humanas/citologia , Oxilipinas/metabolismo , Streptococcus/patogenicidade
14.
Infect Immun ; 84(5): 1633-1641, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26975990

RESUMO

G protein-coupled receptor kinase 5 (GRK5) is a serine/threonine kinase previously shown to mediate polymicrobial sepsis-induced inflammation. The goal of the present study was to examine the role of GRK5 in monomicrobial pulmonary infection by using an intratracheal Escherichia coli infection model of pneumonia. We used sublethal and lethal doses of E. coli to examine the mechanistic differences between low-grade and high-grade inflammation induced by E. coli infection. With a sublethal dose of E. coli, GRK5 knockout (KO) mice exhibited higher plasma CXCL1/KC levels and enhanced lung neutrophil recruitment early after infection, and lower bacterial loads, than wild-type (WT) mice. The inflammatory response was also diminished, and resolution of inflammation advanced, in the lungs of GRK5 KO mice. In contrast to the reduced bacterial loads in GRK5 KO mice following a sublethal dose, at a lethal dose of E. coli, the bacterial burdens remained high in GRK5 KO mice relative to those in WT mice. This occurred in spite of enhanced plasma CXCL1 levels as well as neutrophil recruitment in the KO mice. But the recruited neutrophils (following high-dose infection) exhibited decreased CD11b expression and reduced reactive oxygen species production, suggesting decreased neutrophil activation or increased neutrophil exhaustion in the GRK5 KO mice. In agreement with the increased bacterial burden, KO mice showed poorer survival than WT mice following E. coli infection at a lethal dose. Overall, our data suggest that GRK5 negatively regulates CXCL1/KC levels during bacterial pneumonia but that the role of GRK5 in the clinical outcome in this model is dependent on the bacterial dose.


Assuntos
Carga Bacteriana , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/patologia , Escherichia coli/isolamento & purificação , Quinase 5 de Receptor Acoplado a Proteína G/metabolismo , Pneumonia Bacteriana/microbiologia , Pneumonia Bacteriana/patologia , Animais , Quinase 5 de Receptor Acoplado a Proteína G/deficiência , Pulmão/microbiologia , Pulmão/patologia , Masculino , Camundongos Knockout , Análise de Sobrevida
15.
Anim Health Res Rev ; 16(2): 135-49, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26303748

RESUMO

The bovine mammary gland is a dynamic and complex organ composed of various cell types that work together for the purpose of milk synthesis and secretion. A layer of endothelial cells establishes the blood-milk barrier, which exists to facilitate the exchange of solutes and macromolecules necessary for optimal milk production. During bacterial challenge, however, endothelial cells divert some of their lactation function to protect the underlying tissue from damage by initiating inflammation. At the onset of inflammation, endothelial cells tightly regulate the movement of plasma components and leukocytes into affected tissue. Unfortunately, endothelial dysfunction as a result of exacerbated or sustained inflammation can negatively affect both barrier integrity and the health of surrounding extravascular tissue. The objective of this review is to highlight the role of endothelial cells in supporting milk production and regulating optimal inflammatory responses. The consequences of endothelial dysfunction and sustained inflammation on milk synthesis and secretion are discussed. Given the important role of endothelial cells in orchestrating the inflammatory response, a better understanding of endothelial function during mastitis may support development of targeted therapies to protect bovine mammary tissue and mammary endothelium.


Assuntos
Células Endoteliais/fisiologia , Glândulas Mamárias Animais/citologia , Mastite Bovina/fisiopatologia , Animais , Permeabilidade Capilar , Bovinos , Feminino , Humanos , Inflamação , Lactação , Leucócitos/citologia , Glândulas Mamárias Animais/patologia , Glândulas Mamárias Humanas/fisiologia , Mastite Bovina/microbiologia , Leite , Estresse Oxidativo , Fenótipo
16.
Am J Pathol ; 184(8): 2297-309, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24946011

RESUMO

ß-Arrestin-1 (ßArr1), a scaffolding protein critical in G-protein coupled receptor desensitization has more recently been found to be important in the pathogenesis of various inflammatory diseases. We sought to understand the role of ßArr1 in sepsis pathogenesis using a mouse model of polymicrobial sepsis. Although in previous studies we established that ßArr1 deficiency protects mice from endotoxemia, here we demonstrate that the absence of ßArr1 remarkably renders mice more susceptible to mortality in polymicrobial sepsis. In accordance with the mortality pattern, early production of inflammatory mediators was markedly enhanced in ßArr1 knockout mice systemically and locally in various organs. In addition, enhanced inflammation in the heart was associated with increased NFκB activation. Compared to these effects, immune cell infiltration, thymic apoptosis, and immune suppression during polymicrobial sepsis were unaffected by a deficiency of ßArr1. Additionally, enhanced inflammation and consequent higher mortality were not observed in heterozygous mice, suggesting that one allele of ßArr1 was sufficient for this protective negative regulatory role. We further demonstrate that, unexpectedly, ßArr1 in nonhematopoietic cells is critical and sufficient for inhibiting sepsis-induced inflammation, whereas hematopoietic ßArr1 is likely redundant. Taken together, our results reveal a novel and previously unrecognized negative regulatory role of the nonhematopoietic ßArr1 in sepsis-induced inflammation.


Assuntos
Arrestinas/metabolismo , Sepse/metabolismo , Animais , Western Blotting , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Sepse/fisiopatologia , beta-Arrestina 1 , beta-Arrestinas
17.
J Innate Immun ; 5(4): 401-13, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23485819

RESUMO

NFκB-dependent signaling is an important modulator of inflammation in several diseases including sepsis. G-protein-coupled receptor kinase-5 (GRK5) is an evolutionarily conserved regulator of the NFκB pathway. We hypothesized that GRK5 via NFκB regulation plays an important role in the pathogenesis of sepsis. To test this we utilized a clinically relevant polymicrobial sepsis model in mice that were deficient in GRK5. We subjected wild-type (WT) and GRK5 knockout (KO) mice to cecal ligation and puncture (CLP)-induced polymicrobial sepsis and assessed the various events in sepsis pathogenesis. CLP induced a significant inflammatory response in the WT and this was markedly attenuated in the KO mice. To determine the signaling mechanisms and the role of NFκB activation in sepsis-induced inflammation, we assessed the levels of IκBα phosphorylation and expression of NFκB-dependent genes in the liver in the two genotypes. Both IκBα phosphorylation and gene expression were significantly inhibited in the GRK5 KO compared to the WT mice. Interestingly, however, GRK5 did not modulate either immune cell infiltration (to the primary site of infection) or local/systemic bacterial load subsequent to sepsis induction. In contrast GRK5 deficiency significantly inhibited sepsis-induced plasma corticosterone levels and the consequent thymocyte apoptosis in vivo. Associated with these outcomes, CLP-induced mortality was significantly prevented in the GRK5 KO mice in the presence of antibiotics. Together, our studies demonstrate that GRK5 is an important regulator of inflammation and thymic apoptosis in polymicrobial sepsis and implicate GRK5 as a potential molecular target in sepsis.


Assuntos
Quinase 5 de Receptor Acoplado a Proteína G/metabolismo , Fígado/metabolismo , Sepse/imunologia , Animais , Apoptose/genética , Apoptose/imunologia , Carga Bacteriana/genética , Ceco/lesões , Ceco/cirurgia , Movimento Celular/genética , Células Cultivadas , Corticosterona/sangue , Modelos Animais de Doenças , Quinase 5 de Receptor Acoplado a Proteína G/genética , Quinase 5 de Receptor Acoplado a Proteína G/imunologia , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Humanos , Inflamação/genética , Inflamação/imunologia , Fígado/imunologia , Masculino , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Transdução de Sinais/genética , Timócitos/patologia
18.
Cell Immunol ; 272(2): 107-11, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22078319

RESUMO

G-protein coupled receptor kinase-5 (GRK5) is a recently described NFκB regulator in TLR4 signaling pathway. To determine whether the role of GRK5 is MyD88- or TRIF-dependent, we injected wild type and GRK5 knockout mice with Pam3CSK4 (MyD88-dependent TLR1/2 ligand) and Poly(I:C) (TRIF-dependent TLR3 ligand) and examined the in vivo systemic inflammatory response. Our results demonstrate that GRK5 regulates IL-12p40 and G-CSF via a mechanism that is common to both MyD88 and TRIF. However, GRK5 regulates IL-5 and MCP-1 in a MyD88-dependent but TNFα in a TRIF-dependent manner. Together, our results demonstrate multiple roles of GRK5 in TLR signaling.


Assuntos
Quinase 5 de Receptor Acoplado a Proteína G/metabolismo , Síndrome de Resposta Inflamatória Sistêmica/metabolismo , Receptor 2 Toll-Like/metabolismo , Receptor 3 Toll-Like/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Quimiocina CCL2/metabolismo , Quinase 5 de Receptor Acoplado a Proteína G/deficiência , Quinase 5 de Receptor Acoplado a Proteína G/genética , Fator Estimulador de Colônias de Granulócitos/metabolismo , Subunidade p40 da Interleucina-12/metabolismo , Interleucina-5/metabolismo , Ligantes , Lipopeptídeos/farmacologia , Masculino , Camundongos , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/metabolismo , Poli I-C/farmacologia , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
19.
Int J Clin Exp Med ; 4(4): 320-30, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22140603

RESUMO

Previous studies have implicated a critical role for G-protein coupled receptor kinase-2 (GRK2) in sepsis owing to its ability to regulate inflammatory response and chemotaxis of immune cells. We therefore, hypothesized that deletion of GRK2 in myeloid cells would significantly modulate the pathogenesis of polymicrobial sepsis. To test this hypothesis, we induced cecal ligation and puncture (CLP), in mice with myeloid-specific deletion of GRK2 and the corresponding GRK2 wild type littermates and determined the inflammatory response (IL-6 and IL-10), immune cell infiltration, bacterial load and survival. Six hours after surgery, plasma IL-6 and IL-6:IL-10 ratios were significantly enhanced in the GRK2 knockouts compared to the GRK2 wild type mice. Compared to these effects, IL-6was significantly elevated in the bronchoalveolar lavage but not in the peritoneal fluid of the GRK2 knockout mice. On the other hand, peritoneal IL-10 was significantly elevated in the GRK2 knockout mice compared to the GRK2 wild type. Even though GRK2 knockout mice exhibited an exaggerated cytokine response, there was no difference in immune cell infiltration into the primary site of infection or in bacterial clearance when compared between the GRK2 wild type and GRK2 knockout mice after surgery. Furthermore, in spite of the enhanced pro-inflammatory profile early after surgery, there was only a modest increase in mortality in the GRK2 knockout compared to the GRK2 wild type mice after CLP. Together, our studies demonstrate that myeloid-specific knockout of GRK2 renders the mice more susceptible to an early pro-inflammatory state. However, myeloid-specific GRK2 is not involved in immune cell infiltration to the primary site of infection or in bacterial clearance and does not significantly modulate mortality in the cecal ligation puncture model of polymicrobial sepsis.

20.
J Cell Physiol ; 226(5): 1323-33, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-20945396

RESUMO

G-protein coupled receptor kinase-5 (GRK5) is a serine/threonine kinase discovered for its role in the regulation of G-protein coupled receptor signaling. Recent studies have shown that GRK5 is also an important regulator of signaling pathways stimulated by non-GPCRs. This study was undertaken to determine the physiological role of GRK5 in Toll-like receptor-4-induced inflammatory signaling pathways in vivo and in vitro. Using mice genetically deficient in GRK5 (GRK5(-/-) ) we demonstrate here that GRK5 is an important positive regulator of lipopolysaccharide (LPS, a TLR4 agonist)-induced inflammatory cytokine and chemokine production in vivo. Consistent with this role, LPS-induced neutrophil infiltration in the lungs (assessed by myeloperoxidase activity) was markedly attenuated in the GRK5(-/-) mice compared to the GRK5(+/+) mice. Similar to the in vivo studies, primary macrophages from GRK5(-/-) mice showed attenuated cytokine production in response to LPS. Our results also identify TLR4-induced NFκB pathway in macrophages to be selectively regulated by GRK5. LPS-induced IκBα phosphorylation, NFκB p65 nuclear translocation, and NFκB binding were markedly attenuated in GRK5(-/-) macrophages. Together, our findings demonstrate that GRK5 is a positive regulator of TLR4-induced IκBα-NFκB pathway as well as a key modulator of LPS-induced inflammatory response.


Assuntos
Quinase 5 de Receptor Acoplado a Proteína G/efeitos dos fármacos , Inflamação/enzimologia , Lipopolissacarídeos/farmacologia , Pulmão/efeitos dos fármacos , Macrófagos Peritoneais/efeitos dos fármacos , NF-kappa B/metabolismo , Animais , Células Cultivadas , Citocinas/metabolismo , Feminino , Quinase 5 de Receptor Acoplado a Proteína G/deficiência , Quinase 5 de Receptor Acoplado a Proteína G/genética , Quinase 5 de Receptor Acoplado a Proteína G/metabolismo , Proteínas I-kappa B/metabolismo , Inflamação/induzido quimicamente , Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Pulmão/enzimologia , Pulmão/imunologia , Macrófagos Peritoneais/enzimologia , Macrófagos Peritoneais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Inibidor de NF-kappaB alfa , Infiltração de Neutrófilos/efeitos dos fármacos , Fosforilação , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Receptor 4 Toll-Like/agonistas , Receptor 4 Toll-Like/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA