Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Virol ; 96(8): e0025022, 2022 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-35352999

RESUMO

In late 2019, a novel coronavirus began circulating within humans in central China. It was designated SARS-CoV-2 because of its genetic similarities to the 2003 SARS coronavirus (SARS-CoV). Now that SARS-CoV-2 has spread worldwide, there is a risk of it establishing new animal reservoirs and recombination with native circulating coronaviruses. To screen local animal populations in the United States for exposure to SARS-like coronaviruses, we developed a serological assay using the receptor binding domain (RBD) from SARS-CoV-2. SARS-CoV-2's RBD is antigenically distinct from common human and animal coronaviruses, allowing us to identify animals previously infected with SARS-CoV or SARS-CoV-2. Using an indirect enzyme-linked immunosorbent assay (ELISA) for SARS-CoV-2's RBD, we screened serum from wild and domestic animals for the presence of antibodies against SARS-CoV-2's RBD. Surprisingly prepandemic feline serum samples submitted to the University of Tennessee Veterinary Hospital were ∼50% positive for anti-SARS RBD antibodies. Some of these samples were serologically negative for feline coronavirus (FCoV), raising the question of the etiological agent generating anti-SARS-CoV-2 RBD cross-reactivity. We also identified several white-tailed deer from South Carolina with anti-SARS-CoV-2 antibodies. These results are intriguing, as cross-reactive antibodies toward SARS-CoV-2 RBD have not been reported to date. The etiological agent responsible for seropositivity was not readily apparent, but finding seropositive cats prior to the current SARS-CoV-2 pandemic highlights our lack of information about circulating coronaviruses in other species. IMPORTANCE We report cross-reactive antibodies from prepandemic cats and postpandemic South Carolina white-tailed deer that are specific for that SARS-CoV RBD. There are several potential explanations for this cross-reactivity, each with important implications to coronavirus disease surveillance. Perhaps the most intriguing possibility is the existence and transmission of an etiological agent (such as another coronavirus) with similarity to SARS-CoV-2's RBD region. However, we lack conclusive evidence of prepandemic transmission of a SARS-like virus. Our findings provide impetus for the adoption of a One Health Initiative focusing on infectious disease surveillance of multiple animal species to predict the next zoonotic transmission to humans and future pandemics.


Assuntos
Anticorpos Antivirais , Gatos , Cervos , Glicoproteína da Espícula de Coronavírus , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , COVID-19/diagnóstico , COVID-19/veterinária , Gatos/virologia , Reações Cruzadas/imunologia , Cervos/virologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/imunologia , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus/metabolismo , Zoonoses Virais/diagnóstico , Zoonoses Virais/virologia
2.
Pathogens ; 10(12)2021 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-34959531

RESUMO

Human cytomegalovirus (HCMV) is a ubiquitous pathogen infecting a majority of people worldwide, with diseases ranging from mild to life-threatening. Its clinical relevance in immunocompromised people and congenital infections have made treatment and vaccine development a top priority. Because of cytomegaloviruses' species specificity, murine cytomegalovirus (MCMV) models have historically informed and advanced translational CMV therapies. Using the phenomenon of centrifugal enhancement, we explored differences between MCMVs derived in vitro and in vivo. We found centrifugal enhancement on tissue culture-derived virus (TCV) was ~3× greater compared with salivary gland derived virus (SGV). Using novel "flow virometry", we found that TCV contained a distinct submicron particle composition compared to SGV. Using an inhibitor of exosome production, we show these submicron particles are not extracellular vesicles that contribute to centrifugal enhancement. We examined how these differences in submicron particles potentially contribute to differing centrifugal enhancement phenotypes, as well as broader in vivo vs. in vitro MCMV differences.

3.
Mol Metab ; 44: 101140, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33285301

RESUMO

OBJECTIVE: The expression of the interleukin-1 receptor type I (IL-1R) is enriched in pancreatic islet ß-cells, signifying that ligands activating this pathway are important for the health and function of the insulin-secreting cell. Using isolated mouse, rat, and human islets, we identified the cytokine IL-1α as a highly inducible gene in response to IL-1R activation. In addition, IL-1α is elevated in mouse and rat models of obesity and Type 2 diabetes. Since less is known about the biology of IL-1α relative to IL-1ß in pancreatic tissue, our objective was to investigate the contribution of IL-1α to pancreatic ß-cell function and overall glucose homeostasis in vivo. METHODS: We generated a novel mouse line with conditional IL-1α alleles and subsequently produced mice with either pancreatic- or myeloid lineage-specific deletion of IL-1α. RESULTS: Using this in vivo approach, we discovered that pancreatic (IL-1αPdx1-/-), but not myeloid-cell, expression of IL-1α (IL-1αLysM-/-) was required for the maintenance of whole body glucose homeostasis in both male and female mice. Moreover, pancreatic deletion of IL-1α led to impaired glucose tolerance with no change in insulin sensitivity. This observation was consistent with our finding that glucose-stimulated insulin secretion was reduced in islets isolated from IL-1αPdx1-/- mice. Alternatively, IL-1αLysM-/- mice (male and female) did not have any detectable changes in glucose tolerance, respiratory quotient, physical activity, or food intake when compared with littermate controls. CONCLUSIONS: Taken together, we conclude that there is an important physiological role for pancreatic IL-1α to promote glucose homeostasis by supporting glucose-stimulated insulin secretion and islet ß-cell mass in vivo.


Assuntos
Glucose/metabolismo , Homeostase , Secreção de Insulina/fisiologia , Interleucina-1alfa/metabolismo , Células Mieloides/metabolismo , Pâncreas/metabolismo , Animais , Linhagem Celular , Citocinas/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Feminino , Intolerância à Glucose/metabolismo , Proteínas de Homeodomínio , Inflamação , Insulina/sangue , Insulina/metabolismo , Resistência à Insulina , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Ratos , Receptores de Citocinas , Receptores Tipo I de Interleucina-1/metabolismo , Transativadores
4.
J Steroid Biochem Mol Biol ; 195: 105468, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31536768

RESUMO

Clinical glucocorticoid use, and diseases that produce elevated circulating glucocorticoids, promote drastic changes in body composition and reduction in whole body insulin sensitivity. Because steroid-induced diabetes is the most common form of drug-induced hyperglycemia, we investigated mechanisms underlying the recognized phenotypes associated with glucocorticoid excess. Male C57BL/6 J mice were exposed to either 100ug/mL corticosterone (cort) or vehicle in their drinking water. Body composition measurements revealed an increase in fat mass with drastically reduced lean mass during the first week (i.e., seven days) of cort exposure. Relative to the vehicle control group, mice receiving cort had a significant reduction in insulin sensitivity (measured by insulin tolerance test) five days after drug intervention. The increase in insulin resistance significantly correlated with an increase in the number of Ki-67 positive ß-cells. Moreover, the ability to switch between fuel sources in liver tissue homogenate substrate oxidation assays revealed reduced metabolic flexibility. Furthermore, metabolomics analyses revealed a decrease in liver glycolytic metabolites, suggesting reduced glucose utilization, a finding consistent with onset of systemic insulin resistance. Physical activity was reduced, while respiratory quotient was increased, in mice receiving corticosterone. The majority of metabolic changes were reversed upon cessation of the drug regimen. Collectively, we conclude that changes in body composition and tissue level substrate metabolism are key components influencing the reductions in whole body insulin sensitivity observed during glucocorticoid administration.


Assuntos
Corticosterona/farmacologia , Glucocorticoides/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Fígado/efeitos dos fármacos , Locomoção/efeitos dos fármacos , Animais , Composição Corporal/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Dieta Hiperlipídica , Glucose/metabolismo , Glicólise/efeitos dos fármacos , Resistência à Insulina , Fígado/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Peritonite/induzido quimicamente , Peritonite/metabolismo , Tioglicolatos
5.
mBio ; 10(3)2019 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-31239384

RESUMO

Human cytomegalovirus (HCMV) is a betaherpesvirus that is a significant pathogen within newborn and immunocompromised populations. Morbidity associated with HCMV infection is the consequence of viral dissemination. HCMV has evolved to manipulate the host immune system to enhance viral dissemination and ensure long-term survival within the host. The immunomodulatory protein vCXCL-1, a viral chemokine functioning primarily through the CXCR2 chemokine receptor, is hypothesized to attract CXCR2+ neutrophils to infection sites, aiding viral dissemination. Neutrophils harbor HCMV in vivo; however, the interaction between vCXCL-1 and the neutrophil has not been evaluated in vivo Using the mouse model and mouse cytomegalovirus (MCMV) infection, we show that murine neutrophils harbor and transfer infectious MCMV and that virus replication initiates within this cell type. Utilizing recombinant MCMVs expressing vCXCL-1 from the HCMV strain (Toledo), we demonstrated that vCXCL-1 significantly enhances MCMV dissemination kinetics. Through cellular depletion experiments, we observe that neutrophils impact dissemination but that overall dissemination is largely neutrophil independent. This work adds neutrophils to the list of innate cells (i.e., dendritic and macrophages/monocytes) that contribute to MCMV dissemination but refutes the hypothesis that neutrophils are the primary cell responding to vCXCL-1.IMPORTANCE An adequate in vivo analysis of HCMV's viral chemokine vCXCL-1 has been lacking. Here we generate recombinant MCMVs expressing vCXCL-1 to study vCXCL-1 function in vivo using MCMV as a surrogate. We demonstrate that vCXCL-1 increases MCMV dissemination kinetics for both primary and secondary dissemination. Additionally, we provide evidence, that the murine neutrophil is largely a bystander in the mouse's response to vCXCL-1. We confirm the hypothesis that vCXCL-1 is a HCMV virulence factor. Infection of severely immunocompromised mice with MCMVs expressing vCXCL-1 was lethal in more than 50% of infected animals, while all animals infected with parental virus survived during a 12-day period. This work provides needed insights into vCXCL-1 function in vivo.


Assuntos
Quimiocina CXCL1/imunologia , Infecções por Citomegalovirus/imunologia , Citomegalovirus/imunologia , Muromegalovirus/imunologia , Neutrófilos/virologia , Animais , Quimiocina CXCL1/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Cinética , Camundongos , Camundongos Endogâmicos BALB C , Muromegalovirus/patogenicidade , Neutrófilos/imunologia , Receptores de Interleucina-8B/genética , Receptores de Interleucina-8B/imunologia , Fatores de Virulência/imunologia , Replicação Viral
6.
mSphere ; 4(1)2019 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-30760613

RESUMO

Human cytomegalovirus (HCMV) is a ubiquitous betaherpesvirus that can cause severe disease following in utero exposure, during primary infection, or latent virus reactivation in immunocompromised populations. These complications lead to a 1- to 2-billion-dollar economic burden, making vaccine development and/or alternative treatments a high priority. Current treatments for HCMV include nucleoside analogues such as ganciclovir (GCV), foscarnet, and cidofovir. Recently, letermovir, a terminase complex inhibitor, was approved for prophylaxis after stem cell transplantation. These treatments have unwanted side effects, and HCMV is becoming resistant to them. Therefore, we sought to develop an alternative treatment that targets a different stage in viral infection. Currently, small antiviral peptides are being investigated as anti-influenza and anti-HIV treatments. We have developed heparan sulfate-binding peptides as tools for preventing CMV infections. These peptides are highly effective at stopping infection of fibroblasts with in vitro-derived HCMV and murine cytomegalovirus (MCMV). However, they do not prevent MCMV infection in vivo Interestingly, these peptides inhibit infectivity of in vivo-derived CMVs, albeit not as well as tissue culture-grown CMVs. We further demonstrate that this class of heparan sulfate-binding peptides is incapable of inhibiting MCMV cell-to-cell spread, which is independent of heparan sulfate usage. These data indicate that inhibition of CMV infection can be achieved using synthetic polybasic peptides, but cell-to-cell spread and in vivo-grown CMVs require further investigation to design appropriate anti-CMV peptides.IMPORTANCE In the absence of an effective vaccine to prevent HCMV infections, alternative interventions must be developed. Prevention of viral entry into susceptible cells is an attractive alternative strategy. Here we report that heparan sulfate-binding peptides effectively inhibit entry into fibroblasts of in vitro-derived CMVs and partially inhibit in vivo-derived CMVs. This includes the inhibition of urine-derived HCMV (uCMV), which is highly resistant to antibody neutralization. While these antiviral peptides are highly effective at inhibiting cell-free virus, they do not inhibit MCMV cell-to-cell spread. This underscores the need to understand the mechanism of cell-to-cell spread and differences between in vivo-derived versus in vitro-derived CMV entry to effectively prevent CMV's spread.


Assuntos
Antivirais/farmacologia , Infecções por Citomegalovirus/prevenção & controle , Citomegalovirus/efeitos dos fármacos , Citomegalovirus/fisiologia , Peptídeos/farmacologia , Animais , Células Cultivadas , Infecções por Citomegalovirus/tratamento farmacológico , Modelos Animais de Doenças , Fibroblastos/virologia , Heparitina Sulfato/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Muromegalovirus/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
7.
J Mol Endocrinol ; 59(1): R33-R46, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28420714

RESUMO

Both type 1 and type 2 diabetes exhibit features of inflammation associated with alterations in pancreatic islet function and mass. These immunological disruptions, if unresolved, contribute to the overall pathogenesis of disease onset. This review presents the emerging role of pancreatic islet chemokine production as a critical factor regulating immune cell entry into pancreatic tissue as well as an important facilitator of changes in tissue resident leukocyte activity. Signaling through two specific chemokine receptors (i.e., CXCR2 and CXCR3) is presented to illustrate key points regarding ligand-mediated regulation of innate and adaptive immune cell responses. The prospective roles of chemokine ligands and their corresponding chemokine receptors to influence the onset and progression of autoimmune- and obesity-associated forms of diabetes are discussed.


Assuntos
Quimiocinas/imunologia , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 2/imunologia , Ilhotas Pancreáticas/imunologia , Receptores CXCR3/imunologia , Receptores de Interleucina-8B/imunologia , Imunidade Adaptativa , Animais , Quimiocinas/genética , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Imunidade Inata , Inflamação , Ilhotas Pancreáticas/patologia , Leucócitos/imunologia , Leucócitos/patologia , Obesidade/genética , Obesidade/imunologia , Obesidade/patologia , Receptores CXCR3/genética , Receptores de Interleucina-8B/genética , Transdução de Sinais
8.
Antiviral Res ; 135: 15-23, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27678155

RESUMO

Human cytomegalovirus (HCMV) infection in utero can lead to congenital sensory neural hearing loss and mental retardation. Reactivation or primary infection can increase the morbidity and mortality in immune suppressed transplant recipients and AIDS patients. The current standard of care for HCMV disease is nucleoside analogs, which can be nephrotoxic. In addition resistance to current treatments is becoming increasingly common. In an effort to develop novel CMV treatments, we tested the effectiveness of the D-form of a novel heparan sulfate binding peptide, p5RD, at reducing infection of ganciclovir (GCV) resistant HCMVs in vitro and MCMV in vivo. HCMV infection was reduced by greater than 90% when cells were pretreated with p5RD. Because p5RD acts by a mechanism unrelated to those used by current antivirals, it was effective at reducing GCV resistant HCMVs by 85%. We show that p5RD is resistant to common proteases and serum inactivation, which likely contributed to its ability to significantly reduced infection of peritoneal exudate cells and viral loads in the spleen and the lungs in vivo. The ability of p5RD to reduce HCMV infectivity in vitro including GCV resistant HCMVs and MCMV infection in vivo suggests that this peptide could be a novel anti-CMV therapeutic.


Assuntos
Infecções por Citomegalovirus/tratamento farmacológico , Citomegalovirus/efeitos dos fármacos , Heparitina Sulfato/metabolismo , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Citomegalovirus/fisiologia , Infecções por Citomegalovirus/virologia , Farmacorresistência Viral , Fibroblastos/efeitos dos fármacos , Fibroblastos/virologia , Ganciclovir/farmacologia , Humanos , Pulmão/efeitos dos fármacos , Pulmão/virologia , Peptídeos/química , Peptídeos/metabolismo , Baço/efeitos dos fármacos , Baço/virologia , Carga Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
9.
J Forensic Sci ; 60(5): 1234-40, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26096156

RESUMO

Postmortem succession of human-associated microbial communities ("human microbiome") has been suggested as a possible method for estimating postmortem interval (PMI) for forensic analyses. Here we evaluate human gut bacterial populations to determine quantifiable, time-dependent changes postmortem. Gut microflora were repeatedly sampled from the proximal large intestine of 12 deceased human individuals as they decayed under environmental conditions. Three intestinal bacterial genera were quantified by quantitative PCR (qPCR) using group-specific primers targeting 16S rRNA genes. Bacteroides and Lactobacillus relative abundances declined exponentially with increasing PMI at rates of Nt=0.977e(-0.0144t) (r2=0.537, p<0.001) and Nt=0.019e(-0.0087t) (r2=0.396, p<0.001), respectively, where Nt is relative abundance at time (t) in cumulative degree hours. Bifidobacterium relative abundances did not change significantly: Nt=0.003e(-0.002t) (r2=0.033, p=0.284). Therefore, Bacteroides and Lactobacillus abundances could be used as quantitative indicators of PMI.


Assuntos
Intestinos/microbiologia , Mudanças Depois da Morte , Idoso , Idoso de 80 Anos ou mais , Bacteroides/genética , Bacteroides/fisiologia , Bifidobacterium/genética , Bifidobacterium/fisiologia , Feminino , Microbioma Gastrointestinal , Humanos , Lactobacillus/genética , Lactobacillus/fisiologia , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , RNA Ribossômico 16S/metabolismo
10.
PLoS One ; 10(5): e0126239, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25992785

RESUMO

Human cytomegalovirus (HCMV) infection can lead to congenital hearing loss and mental retardation. Upon immune suppression, reactivation of latent HCMV or primary infection increases morbidity in cancer, transplantation, and late stage AIDS patients. Current treatments include nucleoside analogues, which have significant toxicities limiting their usefulness. In this study we screened a panel of synthetic heparin-binding peptides for their ability to prevent CMV infection in vitro. A peptide designated, p5+14 exhibited ~ 90% reduction in murine CMV (MCMV) infection. Because negatively charged, cell-surface heparan sulfate proteoglycans (HSPGs), serve as the attachment receptor during the adsorption phase of the CMV infection cycle, we hypothesized that p5+14 effectively competes for CMV adsorption to the cell surface resulting in the reduction in infection. Positively charged Lys residues were required for peptide binding to cell-surface HSPGs and reducing viral infection. We show that this inhibition was not due to a direct neutralizing effect on the virus itself and that the peptide blocked adsorption of the virus. The peptide also inhibited infection of other herpesviruses: HCMV and herpes simplex virus 1 and 2 in vitro, demonstrating it has broad-spectrum antiviral activity. Therefore, this peptide may offer an adjunct therapy for the treatment of herpes viral infections and other viruses that use HSPGs for entry.


Assuntos
Proteoglicanas de Heparan Sulfato/metabolismo , Proteoglicanas de Heparan Sulfato/farmacologia , Herpesviridae/efeitos dos fármacos , Herpesviridae/fisiologia , Internalização do Vírus/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Antivirais/química , Antivirais/farmacologia , Células Cultivadas , Citomegalovirus/efeitos dos fármacos , Citomegalovirus/patogenicidade , Citomegalovirus/fisiologia , Avaliação Pré-Clínica de Medicamentos , Proteoglicanas de Heparan Sulfato/química , Herpesviridae/patogenicidade , Infecções por Herpesviridae/tratamento farmacológico , Infecções por Herpesviridae/prevenção & controle , Infecções por Herpesviridae/virologia , Humanos , Técnicas In Vitro , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Muromegalovirus/efeitos dos fármacos , Muromegalovirus/patogenicidade , Muromegalovirus/fisiologia , Estrutura Secundária de Proteína , Ligação Viral/efeitos dos fármacos
11.
J Immunol ; 195(1): 227-36, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25987741

RESUMO

Human CMV (HCMV) uses members of the hematopoietic system including neutrophils for dissemination throughout the body. HCMV encodes a viral chemokine, vCXCL-1, that is postulated to attract neutrophils for dissemination within the host. The gene encoding vCXCL-1, UL146, is one of the most variable genes in the HCMV genome. Why HCMV has evolved this hypervariability and how this affects the virus' dissemination and pathogenesis is unknown. Because the vCXCL-1 hypervariability maps to important binding and activation domains, we hypothesized that vCXCL-1s differentially activate neutrophils, which could contribute to HCMV dissemination, pathogenesis, or both. To test whether these viral chemokines affect neutrophil function, we generated vCXCL-1 proteins from 11 different clades from clinical isolates from infants infected congenitally with HCMV. All vCXCL-1s were able to induce calcium flux at a concentration of 100 nM and integrin expression on human peripheral blood neutrophils, despite differences in affinity for the CXCR1 and CXCR2 receptors. In fact, their affinity for CXCR1 or CXCR2 did not correlate directly with chemotaxis, G protein-dependent and independent (ß-arrestin-2) activation, or secondary chemokine (CCL22) expression. Our data suggest that vCXCL-1 polymorphisms affect the binding affinity, receptor usage, and differential peripheral blood neutrophil activation that could contribute to HCMV dissemination and pathogenesis.


Assuntos
Quimiocinas CXC/imunologia , Infecções por Citomegalovirus/imunologia , Citomegalovirus/imunologia , Neutrófilos/imunologia , Receptores de Interleucina-8A/imunologia , Receptores de Interleucina-8B/imunologia , Proteínas Virais/imunologia , Animais , Arrestinas/genética , Arrestinas/imunologia , Cálcio/metabolismo , Quimiocina CCL22/genética , Quimiocina CCL22/imunologia , Quimiocinas CXC/genética , Citomegalovirus/genética , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/patologia , Infecções por Citomegalovirus/virologia , Regulação da Expressão Gênica , Variação Genética , Células HEK293 , Células HL-60 , Interações Hospedeiro-Patógeno , Humanos , Lactente , Neutrófilos/patologia , Neutrófilos/virologia , Cultura Primária de Células , Receptores de Interleucina-8A/genética , Receptores de Interleucina-8B/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Células Sf9 , Transdução de Sinais , Spodoptera , Proteínas Virais/genética , beta-Arrestina 2 , beta-Arrestinas
12.
Biochim Biophys Acta ; 1849(6): 637-52, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25882704

RESUMO

Enhanced leukocytic infiltration into pancreatic islets contributes to inflammation-based diminutions in functional ß-cell mass. Insulitis (aka islet inflammation), which can be present in both T1DM and T2DM, is one factor influencing pancreatic ß-cell death and dysfunction. IL-1ß, an inflammatory mediator in both T1DM and T2DM, acutely (within 1h) induced expression of the CCL20 gene in rat and human islets and clonal ß-cell lines. Transcriptional induction of CCL20 required the p65 subunit of NF-κB to replace the p50 subunit at two functional κB sites within the CCL20 proximal gene promoter. The NF-κB p50 subunit prevents CCL20 gene expression during unstimulated conditions and overexpression of p50 reduces CCL20, but enhances cyclooxygenase-2 (COX-2), transcript accumulation after exposure to IL-1ß. We also identified differential recruitment of specific co-activator molecules to the CCL20 gene promoter, when compared with the CCL2 and COX2 genes, revealing distinct transcriptional requirements for individual NF-κB responsive genes. Moreover, IL-1ß, TNF-α and IFN-γ individually increased the expression of CCR6, the receptor for CCL20, on the surface of human neutrophils. We further found that the chemokine CCL20 is elevated in serum from both genetically obese db/db mice and in C57BL6/J mice fed a high-fat diet. Taken together, these results are consistent with a possible activation of the CCL20-CCR6 axis in diseases with inflammatory components. Thus, interfering with this signaling pathway, either at the level of NF-κB-mediated chemokine production, or downstream receptor activation, could be a potential therapeutic target to offset inflammation-associated tissue dysfunction in obesity and diabetes.


Assuntos
Quimiocina CCL20/genética , Diabetes Mellitus/genética , Inflamação/genética , Obesidade/genética , Fator de Transcrição RelA/genética , Animais , Quimiocina CCL20/biossíntese , Quimiocina CCL20/metabolismo , Diabetes Mellitus/patologia , Humanos , Imunidade Inata/genética , Inflamação/patologia , Resistência à Insulina/genética , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Camundongos , Camundongos Obesos , NF-kappa B/genética , Obesidade/metabolismo , Obesidade/fisiopatologia , Ratos , Receptores CCR6/genética , Transdução de Sinais/genética , Fator de Transcrição RelA/biossíntese , Fator de Transcrição RelA/metabolismo
13.
Mol Immunol ; 62(1): 54-62, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24972324

RESUMO

Synthesis and secretion of immunomodulatory proteins, such as cytokines and chemokines, controls the inflammatory response within pancreatic islets. When this inflammation does not resolve, destruction of pancreatic islet ß-cells leads to diabetes mellitus. Production of the soluble mediators of inflammation, such as TNF-α and IL-1ß, from resident and invading immune cells, as well as directly from islet ß-cells, is also associated with suboptimal islet transplantation outcomes. In this study, we found that IL-1ß induces rapid increases in TNF-α mRNA in rat and human islets and the 832/13 clonal ß-cell line. The surge in transcription of the TNF-α gene required the inhibitor of kappa B kinase beta (IκKß), the p65 subunit of the NF-κB and a signal-specific recruitment of RNA polymerase II to the gene promoter. Of note was the increased intracellular production of TNF-α protein in a manner consistent with mRNA accumulation in response to IL-1ß, but no detectable secretion of TNF-α into the media. Additionally, TNF-α specifically induces expression of CD11b, but not CD11c, on neutrophils, which could contribute to the inflammatory milieu and diabetes progression. We conclude that activation of the NF-κB pathway in pancreatic ß-cells leads to rapid intracellular production of the pro-inflammatory TNF-α protein through a combination of specific histone covalent modifications and NF-κB signaling pathways.


Assuntos
Células Secretoras de Insulina/imunologia , Interleucina-1beta/farmacologia , Transcrição Gênica , Fator de Necrose Tumoral alfa/genética , Animais , Linhagem Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Quinase I-kappa B/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo
14.
Methods Mol Biol ; 1119: 267-88, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24639228

RESUMO

Although human cytomegalovirus (HCMV) primary infection is generally asymptomatic, in immune-compromised patients HCMV increases morbidity and mortality. As a member of the betaherpesvirus family, in vivo studies of HCMV are limited due to its species specificity. CMVs from other species are often used as surrogates to express HCMV genes/proteins or used as models for inferring HCMV protein function in humans. Using innovative experiments, these animal models have answered important questions about CMV's life cycle, dissemination, pathogenesis, immune evasion, and host immune response. This chapter provides CMV biologists with an overview of the insights gained using these animal models. Subsequent chapters will provide details of the specifics of the experimental methods developed for each of the animal models discussed here.


Assuntos
Citomegalovirus/genética , Evasão da Resposta Imune , Biologia Molecular/métodos , Animais , Citomegalovirus/imunologia , Citomegalovirus/patogenicidade , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/virologia , Modelos Animais de Doenças , Humanos
15.
Am J Physiol Endocrinol Metab ; 306(2): E131-49, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24280128

RESUMO

Diabetes mellitus results from immune cell invasion into pancreatic islets of Langerhans, eventually leading to selective destruction of the insulin-producing ß-cells. How this process is initiated is not well understood. In this study, we investigated the regulation of the CXCL1 and CXCL2 genes, which encode proteins that promote migration of CXCR2(+) cells, such as neutrophils, toward secreting tissue. Herein, we found that IL-1ß markedly enhanced the expression of the CXCL1 and CXCL2 genes in rat islets and ß-cell lines, which resulted in increased secretion of each of these proteins. CXCL1 and CXCL2 also stimulated the expression of specific integrin proteins on the surface of human neutrophils. Mutation of a consensus NF-κB genomic sequence present in both gene promoters reduced the ability of IL-1ß to promote transcription. In addition, IL-1ß induced binding of the p65 and p50 subunits of NF-κB to these consensus κB regulatory elements as well as to additional κB sites located near the core promoter regions of each gene. Additionally, serine-phosphorylated STAT1 bound to the promoters of the CXCL1 and CXCL2 genes. We further found that IL-1ß induced specific posttranslational modifications to histone H3 in a time frame congruent with transcription factor binding and transcript accumulation. We conclude that IL-1ß-mediated regulation of the CXCL1 and CXCL2 genes in pancreatic ß-cells requires stimulus-induced changes in histone chemical modifications, recruitment of the NF-κB and STAT1 transcription factors to genomic regulatory sequences within the proximal gene promoters, and increases in phosphorylated forms of RNA polymerase II.


Assuntos
Quimiocina CXCL1/genética , Quimiocina CXCL2/genética , Regulação da Expressão Gênica , Células Secretoras de Insulina/metabolismo , NF-kappa B/metabolismo , Fator de Transcrição STAT1/metabolismo , Animais , Células Cultivadas , Humanos , Células Secretoras de Insulina/efeitos dos fármacos , Interleucina-1beta/farmacologia , Ratos , Ratos Wistar , Fator de Transcrição STAT1/genética , Transcrição Gênica/efeitos dos fármacos
16.
J Am Assoc Lab Anim Sci ; 52(2): 176-9, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23562101

RESUMO

We evaluated the effect of repeated intraperitoneal administration of tribromoethanol on various parameters in C57BL/6NHsd mice. Mice (n = 68) were randomly assigned to 1 of 7 groups to receive tribromoethanol (500 mg/kg IP) on day 0 or days 0 and 8; vehicle (tert-amyl alcohol in sterile water) only on day 0 or days 0 and 8; sterile water injection on day 0 or days 0 and 8; or no treatment. A single dose of tribromoethanol failed to produce loss of pedal reflex and had no effect on median food and water consumption but altered median body weight on days 1 through 4 when compared with that in mice that received vehicle only or no treatment. Median body weight did not differ between mice that received a single dose of tribromoethanol and those that received an injection of water. Among mice given 2 doses of tribromoethanol, induction time, anesthetic duration, and recovery time varied widely. Repeated administration of tribromoethanol had no effect on median food and water consumption or body weight compared with those in controls. Median liver weight was significantly greater in mice that received 2 doses compared with a single dose of tribromoethanol. Median liver weight did not differ between untreated mice and those that received tribromoethanol. No significant organ or tissue pathology was observed in any study animal. Although tribromoethanol did not produce morbidity, mortality, or pathologic changes in treated animals, we urge caution in use of tribromoethanol in C57BL/6NHsd mice due to its variable anesthetic effectiveness.


Assuntos
Anestésicos/efeitos adversos , Etanol/análogos & derivados , Camundongos Endogâmicos C57BL , Anestésicos/administração & dosagem , Animais , Peso Corporal/efeitos dos fármacos , Etanol/administração & dosagem , Etanol/efeitos adversos , Feminino , Camundongos , Pentanóis/administração & dosagem , Distribuição Aleatória
17.
Methods Enzymol ; 485: 481-97, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21050933

RESUMO

Chemokines play an important role in inflammatory, developmental, and homeostatic processes. Deregulation of this system results in various diseases including tumorigenesis and cancer metastasis. Deregulation can occur when constitutively active mutant (CAM) chemokine receptors are locked in the "on" position. This can lead to cellular transformation/tumorigenesis. The CXC chemokine receptor 2 (CXCR2) is a G-protein-coupled receptor (GPCR) expressed on neutrophils, some monocytes, endothelial cells, and some epithelial cells. CXCR2 activation with CXC chemokines induces leukocyte migration, trafficking, leukocyte degranulation, cellular differentiation, and angiogenesis. Activation of CXCR2 can lead to cellular transformation. We hypothesized that CAM CXCR2s may play a role in cancer development. In order to identify CXCR2 CAMs, potential mutant CXCR2 receptors were screened using a modified Saccharomyces cerevisiae high-throughput system. S. cerevisiae has been used successfully to identify GPCR/G-protein interactions and autocrine selection for peptide agonists. The CXCR2 CAMs identified from this screen were characterized in mammalian cells. Their ability to transform cells in vitro was shown using foci formation, soft-agar growth, impedance measurement assays, and in vivo tumor growth following hind flank inoculation into mice. Signaling pathways contributing to cellular transformation were identified using luciferase reporter assays. Studying constitutively active GPCRs is an approach to "capturing" pluridimensional GPCRs in a "locked" activation state. In order to address the residues necessary for CXCR2 activation, we used S. cerevisiae for screening novel CAMs and characterized them using mammalian reporter assays.


Assuntos
Mutação , Receptores de Interleucina-8B/genética , Receptores de Interleucina-8B/metabolismo , Animais , Linhagem Celular , Proliferação de Células , Impedância Elétrica , Expressão Gênica , Genes Reporter , Humanos , Luciferases/genética , Camundongos , Mutagênese , Células NIH 3T3 , Neoplasias/patologia , Saccharomyces cerevisiae/genética , Transfecção/métodos
18.
Cell Biol Int ; 33(3): 429-33, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19356706

RESUMO

Cellular transformation is the first step in cancer development. Two features of cellular transformation are proliferation in reduced serum and loss of contact inhibition. Electronic Cell-Substrate Impedance Sensing (ECIS) measurements have been used to measure cellular proliferation, cytotoxicity, apoptosis, and attachment. We have used impedance measurements to distinguish normal cells from cells transformed with a constitutively active chemokine receptor, CXCR2. CXCR2, a member of the G-protein coupled receptor (GPCR) family, is normally involved in cellular activation and migration, but a single amino acid substitution leads to constitutive activity. NIH3T3 cells were transformed with a constitutively active CXCR2 (D143V_CXCR2) and growth in reduced serum and foci formation were measured using established biological assays and compared to data from ECIS. The results of this study show that impedance measurements provide a quick and reliable way of measuring cellular transformation and provide real time assessment of transformed cellular parameters. Use of the ECIS system could allow a rapid screening of anti-cancer drugs that alter cellular transformation.


Assuntos
Transformação Celular Neoplásica/metabolismo , Técnicas Eletroquímicas , Substituição de Aminoácidos , Animais , Técnicas Biossensoriais , Linhagem Celular , Impedância Elétrica , Camundongos , Células NIH 3T3 , Valor Preditivo dos Testes , Receptores de Interleucina-8B/biossíntese , Receptores de Interleucina-8B/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/metabolismo , Transfecção
19.
Virol J ; 6: 4, 2009 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-19134204

RESUMO

No one likes to feel like they have been manipulated, but in the case of cytomegalovirus (CMV) immune manipulation, we do not really have much choice. Whether you call it CMV immune modulation, manipulation, or evasion, the bottom line is that CMV alters the immune response in such a way to allow the establishment of latency with lifelong shedding. With millions of years of coevolution within their hosts, CMVs, like other herpesviruses, encode numerous proteins that can broadly influence the magnitude and quality of both innate and adaptive immune responses. These viral proteins include both homologues of host proteins, such as MHC class I or chemokine homologues, and proteins with little similarity to any other known proteins, such as the chemokine binding protein. Although a strong immune response is launched against CMV, these virally encoded proteins can interfere with the host's ability to efficiently recognize and clear virus, while others induce or alter specific immune responses to benefit viral replication or spread within the host. Modulation of host immunity allows survival of both the virus and the host. One way of describing it would be a kind of "mutually assured survival" (as opposed to MAD, Mutually Assured Destruction). Evaluation of this relationship provides important insights into the life cycle of CMV as well as a greater understanding of the complexity of the immune response to pathogens in general.


Assuntos
Infecções por Citomegalovirus/imunologia , Citomegalovirus/imunologia , Latência Viral , Animais , Citocinas/genética , Citocinas/imunologia , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/virologia , Humanos , Imunidade Inata , Células Matadoras Naturais/imunologia , Receptores de Citocinas/genética , Receptores de Citocinas/imunologia , Proteínas Virais/imunologia , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...