Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Am J Physiol Renal Physiol ; 326(5): F862-F875, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38511222

RESUMO

IgA nephropathy (IgAN) is characterized by glomerular deposition of immune complexes (ICs) consisting of IgA1 with O-glycans deficient in galactose (Gd-IgA1) and Gd-IgA1-specific IgG autoantibodies. These ICs induce kidney injury, and in the absence of disease-specific therapy, up to 40% of patients with IgAN progress to kidney failure. IgA1 with its clustered O-glycans is unique to humans, which hampered development of small-animal models of IgAN. Here, we used a model wherein engineered ICs (EICs) formed from human Gd-IgA1 and recombinant human IgG autoantibody are injected into nude mice to induce glomerular injury mimicking human IgAN. In this model, we assessed the protective effects of sparsentan, a single-molecule dual endothelin angiotensin receptor antagonist (DEARA) versus vehicle on EIC-induced glomerular proliferation and dysregulation of gene expression in the kidney. Oral administration of sparsentan (60 or 120 mg/kg daily) to mice intravenously injected with EIC attenuated the EIC-induced glomerular hypercellularity. Furthermore, analysis of changes in the whole kidney transcriptome revealed that key inflammatory and proliferative biological genes and pathways that are upregulated in this EIC model of IgAN were markedly reduced by sparsentan, including complement genes, integrin components, members of the mitogen-activated protein kinase family, and Fc receptor elements. Partial overlap between mouse and human differentially expressed genes in IgAN further supported the translational aspect of the immune and inflammatory components from our transcriptional findings. In conclusion, our data indicate that in the mouse model of IgAN, sparsentan targets immune and inflammatory processes leading to protection from mesangial hypercellularity.NEW & NOTEWORTHY The mechanisms by which deposited IgA1 immune complexes cause kidney injury during early phases of IgA nephropathy are poorly understood. We used an animal model we recently developed that involves IgA1-IgG immune complex injections and determined pathways related to the induced mesangioproliferative changes. Treatment with sparsentan, a dual inhibitor of endothelin type A and angiotensin II type 1 receptors, ameliorated the induced mesangioproliferative changes and the associated alterations in the expression of inflammatory genes and networks.


Assuntos
Complexo Antígeno-Anticorpo , Modelos Animais de Doenças , Glomerulonefrite por IGA , Imunoglobulina A , Imunoglobulina G , Glomérulos Renais , Animais , Glomerulonefrite por IGA/imunologia , Glomerulonefrite por IGA/tratamento farmacológico , Glomerulonefrite por IGA/genética , Glomerulonefrite por IGA/patologia , Glomerulonefrite por IGA/metabolismo , Imunoglobulina A/metabolismo , Imunoglobulina A/imunologia , Glomérulos Renais/patologia , Glomérulos Renais/metabolismo , Glomérulos Renais/efeitos dos fármacos , Glomérulos Renais/imunologia , Complexo Antígeno-Anticorpo/metabolismo , Redes Reguladoras de Genes , Camundongos Nus , Humanos , Camundongos , Proliferação de Células/efeitos dos fármacos
2.
Infect Immun ; 91(12): e0041623, 2023 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-37909751

RESUMO

Cystic fibrosis (CF) is a genetic disease affecting epithelial ion transport, resulting in thickened mucus and impaired mucociliary clearance. Persons with CF (pwCF) experience life-long infections of the respiratory mucosa caused by a diverse array of opportunists, which are leading causes of morbidity and mortality. In recent years, there has been increased appreciation for the range and diversity of microbes causing CF-related respiratory infections. The introduction of new therapeutics and improved detection methodology has revealed CF-related opportunists such as Achromobacter xylosoxidans (Ax). Ax is a Gram-negative bacterial species which is widely distributed in environmental sources and has been increasingly observed in sputa and other samples from pwCF, typically in patients in later stages of CF disease. In this study, we characterized CF clinical isolates of Ax and tested colonization and persistence of Ax in respiratory infection using immortalized human CF respiratory epithelial cells and BALB/c mice. Genomic analyses of clinical Ax isolates showed homologs for factors including flagellar synthesis, antibiotic resistance, and toxin secretion systems. Ax isolates adhered to polarized cultures of CFBE41o- human immortalized CF bronchial epithelial cells and caused significant cytotoxicity and depolarization of cell layers. Ax colonized and persisted in mouse lungs for up to 72 h post infection, with inflammatory consequences that include increased neutrophil influx in the lung, lung damage, cytokine production, and mortality. We also identified genes that are differentially expressed in synthetic CF sputum media. Based on these results, we conclude that Ax is an opportunistic pathogen of significance in CF.


Assuntos
Achromobacter denitrificans , Fibrose Cística , Infecções por Bactérias Gram-Negativas , Infecções Respiratórias , Animais , Camundongos , Humanos , Achromobacter denitrificans/genética , Fibrose Cística/complicações , Fibrose Cística/microbiologia , Escarro/microbiologia , Infecções por Bactérias Gram-Negativas/microbiologia , Perfilação da Expressão Gênica
3.
Microbiology (Reading) ; 169(1)2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36748431

RESUMO

Cystic fibrosis (CF) is a genetic disorder affecting epithelial ion transport, which among other impacts results in defective mucociliary clearance and innate defenses in the respiratory tract. Consequently, people with CF experience lifelong infections of the respiratory mucosa that are chronic and polymicrobial in nature. Young children with CF are initially colonized by opportunists like nontypeable Haemophilus influenzae (NTHi), which normally resides within the microbiome of the nasopharynx and upper airways and can also cause infections of the respiratory mucosa that include bronchitis and otitis media. NTHi is typically supplanted by other microbes as patients age; for example, people with CF are often chronically infected with mucoid strains of Pseudomonas aeruginosa, which prior work in our laboratory has shown to promote colonization and persistence by other opportunists that include Stenotrophomonas maltophilia. Our previous work has shown that polymicrobial infection impacts host colonization and persistence of incoming microbes via diverse mechanisms that include priming of host immunity that can promote microbial clearance, and cooperativity within polymicrobial biofilms, which can promote persistence. In infection studies with BALB/c Cftrtm1UNC mice, results showed, as previously observed for WT BALB/c mice, preceding infection with NTHi decreased colonization and persistence by P. aeruginosa. Likewise, polymicrobial infection of BALB/c Cftrtm1UNC and C57BL/6 Cftrtm1UncTg(FABPhCFTR)1Jaw/J mice showed correlation between S. maltophilia and P. aeruginosa, with increased bacterial colonization and lung pathology. Based on these results, we conclude that our previous observations regarding polymicrobial infections with CF opportunists in WT mice are also validated using CF mice.


Assuntos
Coinfecção , Fibrose Cística , Infecções por Pseudomonas , Camundongos , Animais , Fibrose Cística/microbiologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Coinfecção/microbiologia , Camundongos Endogâmicos C57BL , Sistema Respiratório , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/genética
4.
Infect Immun ; 90(2): e0056821, 2022 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-34780275

RESUMO

Patients with cystic fibrosis (CF) experience lifelong respiratory infections, which are a significant cause of morbidity and death. These infections are polymicrobial in nature, and the predominant bacterial species undergo a predictable series of changes as patients age. Young patients have populations dominated by opportunists that are typically found within the microbiome of the human nasopharynx, such as nontypeable Haemophilus influenzae (NTHi); these are eventually supplanted, and the population within the CF lung is later dominated by pathogens such as Pseudomonas aeruginosa. In this study, we investigated how initial colonization with NTHi impacts colonization and persistence of P. aeruginosa in the respiratory tract. Analysis of polymicrobial biofilms in vitro by confocal microscopy revealed that NTHi promoted greater P. aeruginosa biofilm volume and diffusion. However, sequential respiratory infection of mice with NTHi followed by P. aeruginosa resulted in significantly lower levels of P. aeruginosa, compared to infection with P. aeruginosa alone. Coinfected mice also had reduced airway tissue damage and lower levels of inflammatory cytokines, compared with P. aeruginosa-infected mice. Similar results were observed after instillation of heat-inactivated NTHi bacteria or purified NTHi lipooligosaccharide endotoxin prior to P. aeruginosa introduction. Based on these results, we conclude that NTHi significantly reduces susceptibility to subsequent P. aeruginosa infection, most likely due to priming of host innate immunity rather than a direct competitive interaction between species. These findings have potential significance with regard to therapeutic management of early-life infections in patients with CF.


Assuntos
Fibrose Cística , Infecções por Haemophilus , Infecções Respiratórias , Animais , Biofilmes , Infecções por Haemophilus/microbiologia , Haemophilus influenzae , Humanos , Camundongos , Pseudomonas aeruginosa , Sistema Respiratório
5.
Gynecol Oncol ; 164(1): 170-180, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34844776

RESUMO

BACKGROUND: Progress in immunotherapy use for gynecologic malignancies is hampered by poor tumor antigenicity and weak T cell infiltration of the tumor microenvironment (TME). Wnt/ß-catenin pathway modulation demonstrated patient benefit in clinical trials as well as enhanced immune cell recruitment in preclinical studies. The purpose of this study was to characterize the pathways by which Wnt/ß-catenin modulation facilitates a more immunotherapy-favorable TME. METHODS: Human tumor samples and in vivo patient-derived xenograft and syngeneic murine models were administered Wnt/ß-catenin modulating agents DKN-01 and CGX-1321 individually or in sequence. Analytical methods included immunohistochemistry, flow cytometry, multiplex cytokine/chemokine array, and RNA sequencing. RESULTS: DKK1 blockade via DKN-01 increased HLA/MHC expression in human and murine tissues, correlating with heightened expression of known MHC I regulators: NFkB, IL-1, LPS, and IFNy. PORCN inhibition via CGX-1321 increased production of T cell chemoattractant CXCL10, providing a mechanism for observed increases in intra-tumoral T cells. Diverse leukocyte recruitment was noted with elevations in B cells and macrophages, with increased tumor expression of population-specific chemokines. Sequential DKK1 blockade and PORCN inhibition decreased tumor burden as evidenced by reduced omental weights. CONCLUSIONS: Wnt/ß-catenin pathway modulation increases MHC I expression and promotes tumor leukocytic infiltration, facilitating a pro-immune TME associated with decreased tumor burden. This intervention overcomes common tumor immune-evasion mechanisms and may render ovarian tumors susceptible to immunotherapy.


Assuntos
Antineoplásicos/farmacologia , Neoplasias dos Genitais Femininos/genética , Via de Sinalização Wnt/efeitos dos fármacos , beta Catenina/efeitos dos fármacos , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/uso terapêutico , Modelos Animais de Doenças , Sinergismo Farmacológico , Feminino , Genes MHC Classe I/genética , Neoplasias dos Genitais Femininos/patologia , Neoplasias dos Genitais Femininos/terapia , Humanos , Imunoterapia , Camundongos , Camundongos Endogâmicos C57BL , Microambiente Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
6.
J Autoimmun ; 118: 102593, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33508637

RESUMO

BACKGROUND: IgA nephropathy is thought to be an autoimmune disease wherein galactose-deficient IgA1 (Gd-IgA1) is recognized by IgG autoantibodies, resulting in formation and renal accumulation of nephritogenic immune complexes. Although this hypothesis is supported by recent findings that, in renal immunodeposits of IgA nephropathy patients, IgG is enriched for Gd-IgA1-specific autoantibodies, experimental proof is still lacking. METHODS: IgG isolated from sera of IgA nephropathy patients or produced as a recombinant IgG (rIgG) was mixed with human Gd-IgA1 to form immune complexes. IgG from healthy individuals served as a control. Nude and SCID mice were injected with human IgG and Gd-IgA1, in immune complexes or individually, and their presence in kidneys was ascertained by immunofluorescence. Pathologic changes in the glomeruli were evaluated by quantitative morphometry and exploratory transcriptomic profiling was performed by RNA-Seq. RESULTS: Immunodeficient mice injected with Gd-IgA1 mixed with IgG autoantibodies from patients with IgA nephropathy, but not Gd-IgA1 mixed with IgG from healthy individuals, displayed IgA, IgG, and mouse complement C3 glomerular deposits and mesangioproliferative glomerular injury with hematuria and proteinuria. Un-complexed Gd-IgA1 or IgG did not induce pathological changes. Moreover, Gd-IgA1-rIgG immune complexes injected into immunodeficient mice induced histopathological changes characteristic of human disease. Exploratory transcriptome profiling of mouse kidney tissues indicated that these immune complexes altered gene expression of multiple pathways, in concordance with the changes observed in kidney biopsies of patients with IgA nephropathy. CONCLUSIONS: This study provides the first in vivo evidence for a pathogenic role of IgG autoantibodies specific for Gd-IgA1 in the pathogenesis of IgA nephropathy.


Assuntos
Autoanticorpos/imunologia , Glomerulonefrite por IGA/imunologia , Imunoglobulina G/imunologia , Animais , Complexo Antígeno-Anticorpo/administração & dosagem , Complexo Antígeno-Anticorpo/imunologia , Autoanticorpos/sangue , Modelos Animais de Doenças , Glomerulonefrite por IGA/sangue , Glomerulonefrite por IGA/patologia , Humanos , Imunoglobulina A/imunologia , Glomérulos Renais/imunologia , Glomérulos Renais/patologia , Camundongos
7.
J Am Soc Nephrol ; 30(10): 2017-2026, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31444275

RESUMO

BACKGROUND: IgA nephropathy (IgAN) is the leading primary GN worldwide. The disease is thought to result from glomerular deposition of circulating immune complexes of IgG bound to galactose-deficient IgA1 (Gd-IgA1). However, routine immunofluorescence microscopy fails to detect IgG in many kidney biopsies from patients with IgAN and the specificity of IgG in immunodeposits has not been tested. METHODS: We used remnant frozen kidney-biopsy specimens from 34 patients with IgAN; 14 were IgG-positive and 20 were IgG-negative by routine immunofluorescence microscopy. Six patients with primary membranous nephropathy (MN) and eight with lupus nephritis (LN) served as controls. IgG in the kidney tissue was extracted and its amount determined by ELISA. IgG molecular integrity was assessed by SDS-PAGE immunoblotting. Antigenic specificity of extracted IgG was determined by ELISA using phospholipase A2 receptor (PLA2R) or Gd-IgA1 as antigen. In addition, ten other IgAN cases, six IgG-positive and four IgG-negative by routine immunofluorescence, were used for colocalization studies by confocal microscopy. RESULTS: IgG extracted from MN but not IgAN immunodeposits reacted with PLA2R. Conversely, IgG extracted from IgAN but not MN or LN immunodeposits reacted with Gd-IgA1. Even IgAN kidney-biopsy specimens without IgG by routine immunofluorescence microscopy had IgG specific for Gd-IgA1. Confocal microscopy confirmed the presence of IgG in the IgAN biopsies with colocalization of glomerular IgA and IgG. CONCLUSIONS: These results reveal for the first time that IgAN kidney biopsies, with or without IgG by routine immunofluorescence, contain Gd-IgA1-specific IgG autoantibodies. These findings support the importance of these autoantibodies in the pathogenesis of IgAN.


Assuntos
Autoanticorpos/imunologia , Glomerulonefrite por IGA/imunologia , Imunoglobulina A/imunologia , Glomérulos Renais/imunologia , Adulto , Idoso , Especificidade de Anticorpos , Feminino , Galactose/deficiência , Humanos , Imunoglobulina A/metabolismo , Masculino , Pessoa de Meia-Idade , Adulto Jovem
8.
Gynecol Oncol ; 151(1): 6-9, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29887484

RESUMO

A 32 year-old nulligravid woman with a uterine mass underwent exploratory laparotomy with myomectomy. Final pathology revealed a low-grade endometrial stromal sarcoma (ESS) with positive margins. She subsequently underwent definitive robotic hysterectomy and bilateral salpingectomy with ovarian preservation. She was diagnosed with a stage IB low-grade ESS. She is currently undergoing observation. Discussion of classification, surgical options, and adjuvant therapy is presented.


Assuntos
Neoplasias do Endométrio/terapia , Tumores do Estroma Endometrial/terapia , Tratamentos com Preservação do Órgão/métodos , Adulto , Biópsia , Quimioterapia Adjuvante/métodos , Neoplasias do Endométrio/diagnóstico por imagem , Neoplasias do Endométrio/patologia , Tumores do Estroma Endometrial/diagnóstico por imagem , Tumores do Estroma Endometrial/patologia , Tubas Uterinas/diagnóstico por imagem , Tubas Uterinas/cirurgia , Feminino , Preservação da Fertilidade/métodos , Humanos , Histerectomia , Laparoscopia/métodos , Margens de Excisão , Estadiamento de Neoplasias , Ovário/diagnóstico por imagem , Procedimentos Cirúrgicos Robóticos/métodos , Salpingectomia , Miomectomia Uterina , Útero/diagnóstico por imagem , Útero/patologia , Útero/cirurgia
9.
J Immunol ; 196(5): 2239-48, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26829988

RESUMO

We used two different infection models to investigate the kinetics of the PcpA-dependent pneumococcal disease in mice. In a bacteremic pneumonia model, we observed a PcpA-dependent increase in bacterial burden in the lungs, blood, liver, bronchoalveolar lavage, and spleens of mice at 24 h postinfection. This PcpA-dependent effect on bacterial burden appeared earlier (within 12 h) in the focal pneumonia model, which lacks bacteremia or sepsis. Histological changes show that the ability of pneumococci to make PcpA was associated with unresolved inflammation in both models of infection. Using our bacteremic pneumonia model we further investigated the effects of PcpA on recruitment of innate immune regulatory cells. The presence of PcpA was associated with increased IL-6 levels, suppressed production of TRAIL, and reduced infiltration of polymorphonuclear cells. The ability of pneumococci to make PcpA negatively modulated both the infiltration and apoptosis of macrophages and the recruitment of myeloid-derived suppressor-like cells. The latter have been shown to facilitate the clearance and control of bacterial pneumonia. Taken together, the ability to make PcpA was strongly associated with increased bacterial burden, inflammation, and negative regulation of innate immune cell recruitment to the lung tissue during bacteremic pneumonia.


Assuntos
Proteínas de Bactérias/imunologia , Proteínas de Transporte/imunologia , Imunomodulação , Células Mieloides/imunologia , Pneumonia Pneumocócica/imunologia , Pneumonia Pneumocócica/microbiologia , Streptococcus pneumoniae/imunologia , Animais , Bacteriemia , Carga Bacteriana , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Citocinas/biossíntese , Modelos Animais de Doenças , Feminino , Regulação Bacteriana da Expressão Gênica , Mediadores da Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Transgênicos , Mutação , Células Mieloides/metabolismo , Pneumonia Pneumocócica/mortalidade , Pneumonia Pneumocócica/patologia , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Fatores de Tempo
10.
J Immunol ; 193(11): 5604-12, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25355920

RESUMO

Circulating monocytes carrying human CMV (HCMV) migrate into tissues, where they differentiate into HCMV-infected resident macrophages that upon interaction with bacterial products may potentiate tissue inflammation. In this study, we investigated the mechanism by which HCMV promotes macrophage-orchestrated inflammation using a clinical isolate of HCMV (TR) and macrophages derived from primary human monocytes. HCMV infection of the macrophages, which was associated with viral DNA replication, significantly enhanced TNF-α, IL-6, and IL-8 gene expression and protein production in response to TLR4 ligand (LPS) stimulation compared with mock-infected LPS-stimulated macrophages during a 6-d in vitro infection. HCMV infection also potentiated TLR5 ligand-stimulated cytokine production. To elucidate the mechanism by which HCMV infection potentiated inducible macrophage responses, we show that infection by HCMV promoted the maintenance of surface CD14 and TLR4 and TLR5, which declined over time in mock-infected macrophages, and enhanced both the intracellular expression of adaptor protein MyD88 and the inducible phosphorylation of IκBα and NF-κB. These findings provide additional information toward elucidating the mechanism by which HCMV potentiates bacteria-induced NF-κB-mediated macrophage inflammatory responses, thereby enhancing organ inflammation in HCMV-infected tissues.


Assuntos
Infecções por Citomegalovirus/imunologia , Citomegalovirus/fisiologia , Macrófagos/imunologia , Células Cultivadas , Citocinas/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Receptores de Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/imunologia , Macrófagos/virologia , Fator 88 de Diferenciação Mieloide/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Receptor 4 Toll-Like/imunologia , Receptor 5 Toll-Like/imunologia , Replicação Viral
11.
J Virol ; 88(13): 7683-8, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24741097

RESUMO

We report that primary human vaginal dendritic cells (DCs) display a myeloid phenotype and express CD4, CCR5, and CXCR4. Vaginal CD13(+) CD11c(+) DCs rapidly and efficiently bound transmitted/founder (T/F) CCR5-tropic (R5) viruses, transported them through explanted vaginal mucosa, and transmitted them in trans to vaginal and blood lymphocytes. Vaginal myeloid DCs may play a key role in capturing and disseminating T/F R5 HIV-1 in vivo and are candidate "gatekeeper" cells in HIV-1 transmission.


Assuntos
Células Dendríticas/virologia , Infecções por HIV/transmissão , HIV-1/patogenicidade , Células Mieloides/virologia , Linfócitos T/virologia , Vagina/virologia , Células Cultivadas , Células Dendríticas/metabolismo , Feminino , Infecções por HIV/metabolismo , Humanos , Células Mieloides/metabolismo , Receptores CCR5/metabolismo , Linfócitos T/metabolismo , Vagina/metabolismo
12.
PLoS Pathog ; 7(5): e1002060, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21637819

RESUMO

Tissue macrophages are derived exclusively from blood monocytes, which as monocyte-derived macrophages support HIV-1 replication. However, among human tissue macrophages only intestinal macrophages are non-permissive to HIV-1, suggesting that the unique microenvironment in human intestinal mucosa renders lamina propria macrophages non-permissive to HIV-1. We investigated this hypothesis using blood monocytes and intestinal extracellular matrix (stroma)-conditioned media (S-CM) to model the exposure of newly recruited monocytes and resident macrophages to lamina propria stroma, where the cells take up residence in the intestinal mucosa. Exposure of monocytes to S-CM blocked up-regulation of CD4 and CCR5 expression during monocyte differentiation into macrophages and inhibited productive HIV-1 infection in differentiated macrophages. Importantly, exposure of monocyte-derived macrophages simultaneously to S-CM and HIV-1 also inhibited viral replication, and sorted CD4+ intestinal macrophages, a proportion of which expressed CCR5+, did not support HIV-1 replication, indicating that the non-permissiveness to HIV-1 was not due to reduced receptor expression alone. Consistent with this conclusion, S-CM also potently inhibited replication of HIV-1 pseudotyped with vesicular stomatitis virus glycoprotein, which provides CD4/CCR5-independent entry. Neutralization of TGF-ß in S-CM and recombinant TGF-ß studies showed that stromal TGF-ß inhibited macrophage nuclear translocation of NF-κB and HIV-1 replication. Thus, the profound inability of intestinal macrophages to support productive HIV-1 infection is likely the consequence of microenvironmental down-regulation of macrophage HIV-1 receptor/coreceptor expression and NF-κB activation.


Assuntos
Antígenos CD4/metabolismo , Regulação para Baixo/fisiologia , HIV-1/fisiologia , Mucosa Intestinal/virologia , Macrófagos/metabolismo , NF-kappa B/metabolismo , Receptores CCR5/metabolismo , Células Cultivadas , Meios de Cultivo Condicionados/farmacologia , HIV-1/isolamento & purificação , HIV-1/patogenicidade , Humanos , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/farmacologia , Replicação Viral/efeitos dos fármacos , Replicação Viral/fisiologia
13.
Front Cell Infect Microbiol ; 13: 1144157, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37305417

RESUMO

Pseudomonas aeruginosa dominates the complex polymicrobial cystic fibrosis (CF) airway and is a leading cause of death in persons with CF. Interestingly, oral streptococcal colonization has been associated with stable CF lung function. The most abundant streptococcal species found in stable patients, Streptococcus salivarius, has been shown to downregulate pro-inflammatory cytokines in multiple colonization models. However, no studies have demonstrated how S. salivarius potentially improves lung function. Our lab previously demonstrated that the P. aeruginosa exopolysaccharide Psl promotes S. salivarius biofilm formation in vitro, suggesting a possible mechanism by which S. salivarius is incorporated into the CF airway microbial community. In this study, we demonstrate that co-infection of rats leads to enhanced S. salivarius colonization and reduced P. aeruginosa colonization. Histological scores for tissue inflammation and damage are lower in dual-infected rats compared to P. aeruginosa infected rats. Additionally, pro-inflammatory cytokines IL-1ß, IL-6, CXCL2, and TNF-α are downregulated during co-infection compared to P. aeruginosa single-infection. Lastly, RNA sequencing of cultures grown in synthetic CF sputum revealed that P. aeruginosa glucose metabolism genes are downregulated in the presence of S. salivarius, suggesting a potential alteration in P. aeruginosa fitness during co-culture. Overall, our data support a model in which S. salivarius colonization is promoted during co-infection with P. aeruginosa, whereas P. aeruginosa airway bacterial burden is reduced, leading to an attenuated host inflammatory response.


Assuntos
Coinfecção , Fibrose Cística , Animais , Ratos , Pseudomonas aeruginosa , Fibrose Cística/complicações , Escarro , Citocinas
14.
J Cyst Fibros ; 22(6): 1104-1112, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37714777

RESUMO

BACKGROUND: Mucus stasis, a hallmark of muco-obstructive disease, results from impaired mucociliary transport and leads to lung function decline and chronic infection. Although therapeutics that target mucus stasis in the airway, such as hypertonic saline or rhDNAse, show some therapeutic benefit, they do not address the underlying electrostatic defect apparent in mucins in CF and related conditions. We have previously shown poly (acetyl, arginyl) glucosamine (PAAG, developed as SNSP113), a soluble, cationic polymer, significantly improves mucociliary transport in a rat model of CF by normalizing the charge defects of CF mucin. Here, we report efficacy in the CFTR-sufficient, ENaC hyperactive, Scnn1b-Tg mouse model that develops airway muco-obstruction due to sodium hyperabsorption and airway dehydration. METHODS: Scnn1b-Tg mice were treated with either 250 µg/mL SNSP113 or vehicle control (1.38% glycerol in PBS) via nebulization once daily for 7 days and then euthanized for analysis. Micro-Optical Coherence Tomography-based evaluation of excised mouse trachea was used to determine the effect on the functional microanatomy. Tissue analysis was performed by routine histopathology. RESULTS: Nebulized treatment of SNSP113 significantly improved mucociliary transport in the airways of Scnn1b-Tg mice, without altering the airway surface or periciliary liquid layer. In addition, SNSP113 significantly reversed epithelial hypertrophy and goblet cell metaplasia. Finally, SNSP113 significantly ameliorated eosinophilic crystalline pneumonia and lung consolidation in addition to inflammatory macrophage influx in this model. CONCLUSION: Overall, this study extends the efficacy of SNSP113 as a potential therapeutic to alleviate mucus stasis in muco-obstructive diseases in CF and potentially in related conditions.


Assuntos
Obstrução das Vias Respiratórias , Fibrose Cística , alfa 2-Macroglobulinas Associadas à Gravidez , Feminino , Gravidez , Camundongos , Animais , Ratos , Depuração Mucociliar , Camundongos Transgênicos , Modelos Animais de Doenças , Camundongos Endogâmicos CFTR , Pulmão , Canais Epiteliais de Sódio/genética
15.
J Clin Med ; 12(23)2023 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-38068413

RESUMO

Routine immunofluorescence microscopy of glomerular immunodeposits in IgA nephropathy shows IgA, C3, and lambda light chains, and sometimes IgG, IgM, and kappa light chains. However, a previous study using high-resolution confocal microscopy showed IgG in all IgA nephropathy cases, likely representing autoantibodies specific for galactose-deficient IgA1. Here, we used high-resolution confocal microscopy to examine the composition of glomerular immunodeposits and colocalization of kappa and lambda light chains with IgA or IgG heavy chains in kidney-biopsy samples from twenty patients with IgA nephropathy, seventeen without IgG, and nine with no or trace kappa light chains by routine immunofluorescence microscopy. IgG was detected in all biopsies by high-resolution confocal microscopy. Single-optical-plane images showed similar colocalization of IgG heavy chains with kappa and lambda light chains. Colocalization of IgA heavy chains was greater with lambda light chains than with kappa light chains. Colocalization of IgG heavy chain with kappa light chains was higher than with lambda light chains in biopsies with endocapillary hypercellularity and crescents, i.e., biopsies with active lesions. We confirmed the utility of high-resolution confocal microscopy to detect components of glomerular immunodeposits not apparent on routine immunofluorescence microscopy and for colocalization of different components, potentially clarifying the pathogenesis of IgA nephropathy.

16.
Microbiol Spectr ; 11(6): e0219823, 2023 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-37800950

RESUMO

IMPORTANCE: Respiratory infections are a leading cause of morbidity and mortality in people with cystic fibrosis (CF). These infections are polymicrobial in nature with overt pathogens and other colonizing microbes present. Microbiome data have indicated that the presence of oral commensal bacteria in the lungs is correlated with improved outcomes. We hypothesize that one oral commensal, Streptococcus parasanguinis, inhibits CF pathogens and modulates the host immune response. One major CF pathogen is Pseudomonas aeruginosa, a Gram-negative, opportunistic bacterium with intrinsic drug resistance and an arsenal of virulence factors. We have previously shown that S. parasanguinis inhibits P. aeruginosa in vitro in a nitrite-dependent manner through the production of reactive nitrogen intermediates. In this study, we demonstrate that while this mechanism is evident in a cell culture model of the CF airway, an alternative mechanism by which S. parasanguinis may improve outcomes for people with CF is through immunomodulation.


Assuntos
Fibrose Cística , Infecções por Pseudomonas , Humanos , Nitritos , Pseudomonas aeruginosa/fisiologia , Biofilmes , Infecções por Pseudomonas/microbiologia , Fibrose Cística/microbiologia , Pulmão , Mucosa Respiratória
18.
J Biol Chem ; 285(25): 19593-604, 2010 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-20388715

RESUMO

Human intestinal macrophages contribute to tissue homeostasis in noninflamed mucosa through profound down-regulation of pro-inflammatory cytokine release. Here, we show that this down-regulation extends to Toll-like receptor (TLR)-induced cytokine release, as intestinal macrophages expressed TLR3-TLR9 but did not release cytokines in response to TLR-specific ligands. Likely contributing to this unique functional profile, intestinal macrophages expressed markedly down-regulated adapter proteins MyD88 and Toll interleukin receptor 1 domain-containing adapter-inducing interferon beta, which together mediate all TLR MyD88-dependent and -independent NF-kappaB signaling, did not phosphorylate NF-kappaB p65 or Smad-induced IkappaBalpha, and did not translocate NF-kappaB into the nucleus. Importantly, transforming growth factor-beta released from intestinal extracellular matrix (stroma) induced identical down-regulation in the NF-kappaB signaling and function of blood monocytes, the exclusive source of intestinal macrophages. Our findings implicate stromal transforming growth factor-beta-induced dysregulation of NF-kappaB proteins and Smad signaling in the differentiation of pro-inflammatory blood monocytes into noninflammatory intestinal macrophages.


Assuntos
Regulação Enzimológica da Expressão Gênica , Proteínas I-kappa B/metabolismo , Inflamação , Mucosa Intestinal/metabolismo , Macrófagos/metabolismo , NF-kappa B/metabolismo , Proteínas Smad/metabolismo , Citocinas/metabolismo , Inibidores Enzimáticos/química , Humanos , Monócitos/metabolismo , Inibidor de NF-kappaB alfa , Análise de Sequência com Séries de Oligonucleotídeos , Fosforilação , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo
19.
J Clin Invest ; 118(2): 629-39, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18172551

RESUMO

Aberrant glycosylation of IgA1 plays an essential role in the pathogenesis of IgA nephropathy. This abnormality is manifested by a deficiency of galactose in the hinge-region O-linked glycans of IgA1. Biosynthesis of these glycans occurs in a stepwise fashion beginning with the addition of N-acetylgalactosamine by the enzyme N-acetylgalactosaminyltransferase 2 and continuing with the addition of either galactose by beta1,3-galactosyltransferase or a terminal sialic acid by a N-acetylgalactosamine-specific alpha2,6-sialyltransferase. To identify the molecular basis for the aberrant IgA glycosylation, we established EBV-immortalized IgA1-producing cells from peripheral blood cells of patients with IgA nephropathy. The secreted IgA1 was mostly polymeric and had galactose-deficient O-linked glycans, characterized by a terminal or sialylated N-acetylgalactosamine. As controls, we showed that EBV-immortalized cells from patients with lupus nephritis and healthy individuals did not produce IgA with the defective galactosylation pattern. Analysis of the biosynthetic pathways in cloned EBV-immortalized cells from patients with IgA nephropathy indicated a decrease in beta1,3-galactosyltransferase activity and an increase in N-acetylgalactosamine-specific alpha2,6-sialyltransferase activity. Also, expression of beta1,3-galactosyltransferase was significantly lower, and that of N-acetylgalactosamine-specific alpha2,6-sialyltransferase was significantly higher than the expression of these genes in the control cells. Thus, our data suggest that premature sialylation likely contributes to the aberrant IgA1 glycosylation in IgA nephropathy and may represent a new therapeutic target.


Assuntos
Glomerulonefrite por IGA/imunologia , Imunoglobulina A/metabolismo , Leucócitos Mononucleares/imunologia , Adulto , Linhagem Celular Transformada , Feminino , Galactosiltransferases/antagonistas & inibidores , Galactosiltransferases/metabolismo , Glucosiltransferases , Glicosilação , Complexo de Golgi/imunologia , Herpesvirus Humano 4 , Humanos , Imunoglobulina A/análise , Nefrite Lúpica/imunologia , Masculino , Pessoa de Meia-Idade , Ácido N-Acetilneuramínico/metabolismo , Sialiltransferases/metabolismo
20.
Nephrol Dial Transplant ; 26(11): 3451-7, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21828345

RESUMO

BACKGROUND: Circulating immune complexes (CIC) containing galactose (Gal)-deficient IgA1 from adults with IgA nephropathy (IgAN) induce proliferation of cultured mesangial cells, but activities of CIC from pediatric patients with the disease have not been studied. METHODS: CIC of different sizes were isolated from sera of pediatric and adult IgAN patients and their effects on cultured human mesangial cells (MC) were assessed by measuring cellular proliferation, expression of IL-6 and IL-8 and laminin and phosphotyrosine signaling. RESULTS: Large CIC from pediatric IgAN patients (>800 kDa) containing Gal-deficient IgA1 stimulated cellular proliferation, whereas in some patients, smaller CIC were inhibitory. Addition of stimulatory and inhibitory CIC to MC differentially altered phosphorylation patterns of three major tyrosine-phosphorylated proteins of molecular mass 37, 60 and 115 kDa. The stimulatory CIC transiently increased tyrosine-phosphorylation of the 37-kDa protein and decreased phosphorylation of the other two proteins, whereas the inhibitory CIC increased phosphorylation of all three proteins. Furthermore, we investigated the influence of IgA1-containing CIC from sera of children with IgAN with clinically active disease (i.e., abnormal urinalysis and/or serum creatinine concentration) or inactive disease (i.e., normal urinalysis and serum creatinine concentration) on the expression of IL-6 and IL-8 genes by mesangial cells. Real-time reverse transcription-polymerase chain reaction results showed that the CIC from a patient with active disease stimulated MC to express the two cytokine genes at higher levels than did the CIC from a patient with inactive disease. Moreover, stimulatory CIC increased production of the extracellular matrix protein laminin. CONCLUSION: These data indicate that sera of pediatric IgAN patients contain biologically active CIC with Gal-deficient IgA1.


Assuntos
Complexo Antígeno-Anticorpo/farmacologia , Proliferação de Células , Mesângio Glomerular/citologia , Glomerulonefrite por IGA/fisiopatologia , Imunoglobulina A/metabolismo , Células Mesangiais/efeitos dos fármacos , Adolescente , Adulto , Complexo Antígeno-Anticorpo/sangue , Western Blotting , Células Cultivadas , Criança , Pré-Escolar , Ensaio de Imunoadsorção Enzimática , Feminino , Galactose/deficiência , Taxa de Filtração Glomerular , Mesângio Glomerular/imunologia , Mesângio Glomerular/metabolismo , Glomerulonefrite por IGA/etiologia , Glicosilação , Humanos , Técnicas Imunoenzimáticas , Imunoglobulina A/imunologia , Interleucina-6/genética , Interleucina-6/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Masculino , Células Mesangiais/citologia , Células Mesangiais/imunologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA