Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Pediatr Surg Int ; 40(1): 35, 2024 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-38216767

RESUMO

PURPOSE: Necrotizing enterocolitis (NEC) is a severe intestinal disease primarily affecting premature infants, marked by impaired epithelial regeneration. Breastfed infants are less susceptible to NEC than formula-fed ones, and human milk oligosaccharides (HMO) found in breast milk have prebiotic properties that can protect against NEC. However, it is unclear how HMOs influence intestinal epithelium regeneration in relation to the gut microbiota. METHODS: Broad-spectrum antibiotics were administered to pregnant dams to reduce the microbiota in offspring. NEC was induced through administration of hyperosmolar formula, lipopolysaccharide, and hypoxia from postnatal days (p) 5-9. Intestinal epithelial organoids were derived from p9 mice. HMOs were isolated from human donor breast milk and then solubilized in the formula for each feed or culture media for organoids. RESULTS: HMOs did not alter the microbiota profile in the presence of a normal or reduced microbiota. In the reduced microbiota, HMO treatment decreased NEC intestinal injury, and increased proliferation and stem cell activity. Additionally, in the complete absence of the microbiota, HMOs stimulated intestinal organoid growth. CONCLUSION: This study demonstrates that HMOs promoted intestinal epithelial regeneration independent of the gut microbiota. These findings provide further insight into the various benefits HMOs may have in the protection against NEC.


Assuntos
Enterocolite Necrosante , Doenças do Recém-Nascido , Microbiota , Lactente , Feminino , Gravidez , Recém-Nascido , Animais , Humanos , Camundongos , Leite Humano , Enterocolite Necrosante/prevenção & controle , Mucosa Intestinal , Oligossacarídeos/farmacologia , Regeneração
2.
Front Immunol ; 14: 1043680, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36969170

RESUMO

Zetomipzomib (KZR-616) is a selective inhibitor of the immunoproteasome currently undergoing clinical investigation in autoimmune disorders. Here, we characterized KZR-616 in vitro and in vivo using multiplexed cytokine analysis, lymphocyte activation and differentiation, and differential gene expression analysis. KZR-616 blocked production of >30 pro-inflammatory cytokines in human peripheral blood mononuclear cells (PBMCs), polarization of T helper (Th) cells, and formation of plasmablasts. In the NZB/W F1 mouse model of lupus nephritis (LN), KZR-616 treatment resulted in complete resolution of proteinuria that was maintained at least 8 weeks after the cessation of dosing and was mediated in part by alterations in T and B cell activation, including reduced numbers of short and long-lived plasma cells. Gene expression analysis of human PBMCs and tissues from diseased mice revealed a consistent and broad response focused on inhibition of T, B, and plasma cell function and the Type I interferon pathway and promotion of hematopoietic cell lineages and tissue remodeling. In healthy volunteers, KZR-616 administration resulted in selective inhibition of the immunoproteasome and blockade of cytokine production following ex vivo stimulation. These data support the ongoing development of KZR-616 in autoimmune disorders such as systemic lupus erythematosus (SLE)/LN.


Assuntos
Leucócitos Mononucleares , Nefrite Lúpica , Humanos , Animais , Camundongos , Leucócitos Mononucleares/metabolismo , Citocinas/metabolismo , Imunidade
3.
Pediatr Res ; 91(1): 101-106, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34561550

RESUMO

BACKGROUND: Stem cell therapy has been proven to rescue intestinal injury and stimulate intestinal regeneration in necrotizing enterocolitis (NEC). Specifically, stem cells derived from amniotic fluid (AFSCs) and mesenchymal stem cells (MSCs) derived from bone marrow have shown promising results in the treatment of experimental NEC. This study aims to examine the effects of AFSCs and MSCs on the prevention of intestinal injury during experimental NEC. METHODS: Supernatants from AFSC and MSC cultures were collected to perform proteomic analysis. Prior to NEC induction, mice received intraperitoneal injections of phosphate-buffered saline (PBS), 2 × 106 AFSCs, or 2 × 106 MSCs. RESULTS: We found that AFSCs grew faster than MSCs. Proteomic analysis indicated that AFSCs are primarily involved in cell development and growth, while MSCs are involved in immune regulation. Administering AFSCs before NEC induction decreased NEC severity and mucosal inflammation. Intestinal proliferation and endogenous stem cell activation were increased after AFSC administration. However, administering MSCs before NEC induction had no beneficial effects. CONCLUSIONS: This study demonstrated that AFSCs and MSCs have different protein release profiles. AFSCs can potentially be used as a preventative strategy for neonates at risk of NEC, while MSCs cannot be used. IMPACT: AFSCs and MSCs have distinct protein secretory profiles, and AFSCs are primarily involved in cell development and growth, while MSCs are involved in immune regulation. AFSCs are unique in transiently enhancing healthy intestinal epithelial cell growth, which offers protection against the development of experimental NEC. The prevention of NEC via the administration of AFSCs should be evaluated in infants at great risk of developing NEC or in infants with early signs of NEC.


Assuntos
Líquido Amniótico/citologia , Transplante de Células-Tronco , Animais , Enterocolite Necrosante , Humanos , Recém-Nascido , Camundongos
4.
Drug Metab Dispos ; 49(9): 810-821, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34234005

RESUMO

KZR-616 is an irreversible tripeptide epoxyketone-based selective inhibitor of the human immunoproteasome. Inhibition of the immunoproteasome results in anti-inflammatory activity in vitro and based on promising therapeutic activity in animal models of rheumatoid arthritis and systemic lupus erythematosus KZR-616 is being developed for potential treatment of multiple autoimmune and inflammatory diseases. The presence of a ketoepoxide pharmacophore presents unique challenges in the study of drug metabolism during lead optimization and clinical candidate profiling. This study presents a thorough and systematic in vitro and cell-based enzymatic metabolism and kinetic investigation to identify the major enzymes involved in the metabolism and elimination of KZR-616. Upon exposure to liver microsomes in the absence of NADPH, KZR-616 and its analogs were converted to their inactive diol derivatives with varying degrees of stability. Diol formation was also shown to be the major metabolite in pharmacokinetic studies in monkeys and correlated with in vitro stability results for individual compounds. Further study in intact hepatocytes revealed that KZR-616 metabolism was sensitive to an inhibitor of microsomal epoxide hydrolase (mEH) but not inhibitors of cytochrome P450 (P450) or soluble epoxide hydrolase (sEH). Primary human hepatocytes were determined to be the most robust source of mEH activity for study in vitro. These findings also suggest that the exposure of KZR-616 in vivo is unlikely to be affected by coadministration of inhibitors or inducers of P450 and sEH. SIGNIFICANCE STATEMENT: This work presents a thorough and systematic investigation of metabolism and kinetics of KZR-616 and related analogs in in vitro and cell-based enzymatic systems. Information gained could be useful in assessing novel covalent proteasome inhibitors during lead compound optimization. These studies also demonstrate a robust source in vitro test system that correlated with in vivo pharmacokinetics for KZR-616 and two additional tripeptide epoxyketones.


Assuntos
Cisteína Endopeptidases/imunologia , Sistema Enzimático do Citocromo P-450/metabolismo , Epóxido Hidrolases/metabolismo , Morfolinas/farmacologia , Complexo de Endopeptidases do Proteassoma/imunologia , Proteínas/imunologia , Animais , Doenças Autoimunes/tratamento farmacológico , Células Cultivadas , Cisteína Endopeptidases/metabolismo , Epóxido Hidrolases/imunologia , Hepatócitos/metabolismo , Humanos , Inativação Metabólica , Inflamação/tratamento farmacológico , Macaca fascicularis , Inibidores de Proteassoma/farmacologia
5.
Cell Death Dis ; 10(10): 743, 2019 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-31582728

RESUMO

Necrotizing enterocolitis (NEC) is a devastating neonatal disease characterized by acute intestinal injury. Intestinal stem cell (ISC) renewal is required for gut regeneration in response to acute injury. The Wnt/ß-catenin pathway is essential for intestinal renewal and ISC maintenance. We found that ISC expression, Wnt activity and intestinal regeneration were all decreased in both mice with experimental NEC and in infants with acute active NEC. Moreover, intestinal organoids derived from NEC-injured intestine of both mice and humans failed to maintain proliferation and presented more differentiation. Administration of Wnt7b reversed these changes and promoted growth of intestinal organoids. Additionally, administration of exogenous Wnt7b rescued intestinal injury, restored ISC, and reestablished intestinal epithelial homeostasis in mice with NEC. Our findings demonstrate that during NEC, Wnt/ß-catenin signaling is decreased, ISC activity is impaired, and intestinal regeneration is defective. Administration of Wnt resulted in the maintenance of intestinal epithelial homeostasis and avoidance of NEC intestinal injury.


Assuntos
Enterocolite Necrosante/fisiopatologia , Intestinos/fisiopatologia , Regeneração/fisiologia , Via de Sinalização Wnt , Animais , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Enterocolite Necrosante/genética , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Intestinos/efeitos dos fármacos , Intestinos/patologia , Camundongos Endogâmicos C57BL , Modelos Biológicos , Organoides/efeitos dos fármacos , Organoides/metabolismo , Proteínas Proto-Oncogênicas/administração & dosagem , Proteínas Proto-Oncogênicas/farmacologia , Regeneração/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Análise de Sobrevida , Proteínas Wnt/administração & dosagem , Proteínas Wnt/farmacologia , Via de Sinalização Wnt/efeitos dos fármacos , Via de Sinalização Wnt/genética
6.
J Med Chem ; 61(24): 11127-11143, 2018 12 27.
Artigo em Inglês | MEDLINE | ID: mdl-30380863

RESUMO

Selective immunoproteasome inhibition is a promising approach for treating autoimmune disorders, but optimal proteolytic active site subunit inhibition profiles remain unknown. We reveal here our design of peptide epoxyketone-based selective low molecular mass polypeptide-7 (LMP7) and multicatalytic endopeptidase complex subunit-1 (MECL-1) subunit inhibitors. Utilizing these and our previously disclosed low molecular mass polypeptide-2 (LMP2) inhibitor, we demonstrate a requirement of dual LMP7/LMP2 or LMP7/MECL-1 subunit inhibition profiles for potent cytokine expression inhibition and in vivo efficacy in an inflammatory disease model. These and additional findings toward optimized solubility led the design and selection of KZR-616 disclosed here and presently in clinical trials for treatment of rheumatic disease.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Artrite Reumatoide/tratamento farmacológico , Morfolinas/farmacologia , Inibidores de Proteassoma/farmacologia , Administração Intravenosa , Animais , Anti-Inflamatórios não Esteroides/química , Anti-Inflamatórios não Esteroides/farmacocinética , Artrite Experimental/tratamento farmacológico , Linhagem Celular Tumoral , Cisteína Endopeptidases/metabolismo , Citocinas/metabolismo , Desenho de Fármacos , Feminino , Humanos , Camundongos Endogâmicos BALB C , Morfolinas/química , Morfolinas/farmacocinética , Oligopeptídeos/farmacologia , Complexo de Endopeptidases do Proteassoma/imunologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/química , Inibidores de Proteassoma/farmacocinética , Relação Estrutura-Atividade
7.
ACS Med Chem Lett ; 8(4): 413-417, 2017 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-28435528

RESUMO

Building upon the success of bortezomib (VELCADE) and carfilzomib (KYPROLIS), the design of a next generation of inhibitors targeting specific subunits within the immunoproteasome is of interest for the treatment of autoimmune disease. There are three catalytic subunits within the immunoproteasome (low molecular mass polypeptide-7, -2, and multicatalytic endopeptidase complex subunit-1; LMP7, LMP2, and MECL-1), and a campaign was undertaken to design a potent and selective LMP2 inhibitor with sufficient properties to allow for sustained inhibition in vivo. Screening a focused library of epoxyketones revealed a series of potent dipeptides that were optimized to provide the highly selective inhibitor KZR-504 (12).

8.
J Biomed Inform ; 64: 116-121, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27693764

RESUMO

Medical errors and patient safety issues remain a significant problem for the healthcare industry in the United States. The Institute of Medicine report To Err is Human reported that there were as many as 98,000 deaths per year due to medical error as of 1999. Many authors and government officials believe that the first step on the path to improvement in patient safety is more comprehensive collection and analysis of patient safety events. The belief is that this will enable safety improvements based on data showing the nature and frequency of events that occur, and the effectiveness of interventions. This systematization of healthcare practice can be a step in the right direction toward a value based, safety conscious and effective healthcare system. To help standardize this reporting and analysis, AHRQ created Common Formats for Patient Safety data collection and reporting. This manuscript describes the development of patient safety reporting and learning through the Patient Safety Organizations (PSO)s and the Common Formats and gives readers an overview of how the system is expected to function and the breadth of development of the Common Formats to date.


Assuntos
Coleta de Dados , Erros Médicos , Segurança do Paciente , Confiabilidade dos Dados , Humanos , Estados Unidos
9.
Diabetes ; 64(7): 2537-49, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25735732

RESUMO

Obesity and diabetes are characterized by increased inflammation reflecting disordered control of innate immunity. We reveal a local intestinal intraepithelial lymphocyte (IEL)-GLP-1 receptor (GLP-1R) signaling network that controls mucosal immune responses. Glp1r expression was enriched in intestinal IEL preparations and copurified with markers of Tαß and Tγδ IELs, the two main subsets of intestinal IELs. Exendin-4 increased cAMP accumulation in purified IELs and reduced the production of cytokines from activated IELs but not from splenocytes ex vivo. These actions were mimicked by forskolin, absent in IELs from Glp1r(-/-) mice, and attenuated by the GLP-1R agonist exendin (9-39) consistent with a GLP-1R-dependent mechanism of action. Furthermore, Glp1r(-/-) mice exhibited dysregulated intestinal gene expression, an abnormal representation of microbial species in feces, and enhanced sensitivity to intestinal injury following administration of dextran sodium sulfate. Bone marrow transplantation using wild-type C57BL/6 donors normalized expression of multiple genes regulating immune function and epithelial integrity in Glp1r(-/-) recipient mice, whereas acute exendin-4 administration robustly induced the expression of genes encoding cytokines and chemokines in normal and injured intestine. Taken together, these findings define a local enteroendocrine-IEL axis linking energy availability, host microbial responses, and mucosal integrity to the control of innate immunity.


Assuntos
Mucosa Intestinal/imunologia , Linfócitos/imunologia , Receptores de Glucagon/agonistas , Receptores de Glucagon/fisiologia , Animais , Citocinas/análise , Exenatida , Feminino , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Receptor do Peptídeo Semelhante ao Glucagon 1 , Imunidade Inata , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Peptídeos/farmacologia , Transdução de Sinais , Peçonhas/farmacologia
10.
Inflamm Bowel Dis ; 21(2): 297-306, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25590952

RESUMO

BACKGROUND: Adherent-invasive Escherichia coli (AIEC) colonization has been strongly implicated in the pathogenesis of Crohn's disease. Environmental triggers such as vitamin D deficiency have emerged as key factors in the pathogenesis of inflammatory bowel diseases. The aim of this study was to investigate the effects of 1,25(OH)2D3 on AIEC infection-induced changes in vivo and in vitro. METHODS: Barrier function was assessed in polarized epithelial Caco-2-bbe cells grown in medium with or without vitamin D and challenged with AIEC strain LF82. Weaned C57BL/6 mice were fed either a vitamin D-sufficient or -deficient diet for 5 weeks and then infected with AIEC, in the absence and presence of low-dose dextran sodium sulphate. Disease severity was assessed by histological analysis and in vivo intestinal permeability assay. Presence of invasive bacteria was assessed by transmission electron microscopy. RESULTS: Caco-2-bbe cells incubated with 1,25(OH)2D3 were protected against AIEC-induced disruption of transepithelial electrical resistance and tight-junction protein redistribution. Vitamin D-deficient C57BL/6 mice given a course of 2% dextran sodium sulphate exhibited pronounced epithelial barrier dysfunction, were more susceptible to AIEC colonization, and showed exacerbated colonic injury. Transmission electron microscopy of colonic tissue from infected mice demonstrated invasion of AIEC and fecal microbiome analysis revealed shifts in microbial communities. CONCLUSIONS: These data show that vitamin D is able to mitigate the deleterious effects of AIEC on the intestinal mucosa, by maintaining intestinal epithelial barrier homeostasis and preserving tight-junction architecture. This study highlights the association between vitamin D status, dysbiosis, and Crohn's disease.


Assuntos
Calcitriol/farmacologia , Colite/etiologia , Doença de Crohn/etiologia , Modelos Animais de Doenças , Infecções por Escherichia coli/etiologia , Escherichia coli/patogenicidade , Mucosa Intestinal/patologia , Deficiência de Vitamina D/fisiopatologia , Animais , Apoptose , Aderência Bacteriana/efeitos dos fármacos , Western Blotting , Células CACO-2 , Permeabilidade da Membrana Celular/efeitos dos fármacos , Proliferação de Células , Células Cultivadas , Colite/metabolismo , Colite/patologia , Doença de Crohn/metabolismo , Doença de Crohn/patologia , Sulfato de Dextrana/farmacologia , Condutividade Elétrica , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/patologia , Feminino , Imunofluorescência , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/patologia , Humanos , Técnicas Imunoenzimáticas , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/patologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Deficiência de Vitamina D/complicações
11.
Shock ; 43(5): 483-9, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25608140

RESUMO

Intestinal epithelia regulate barrier integrity when challenged by inflammation, oxidative stress, and microbes. Transforming growth factor-ß1 (TGF-ß1) is a cytokine with known beneficial effects on intestinal epithelia, including barrier enhancement, after exposure to proinflammatory cytokines and infectious agents. The aim of this study was to determine whether TGF-ß1 directly protects intestinal epithelia during hypoxia-reoxygenation (HR). Intestinal epithelial monolayers (T84, Caco-2) were exposed to either hypoxia (1% O2, 1 h) or oxidative stress (hydrogen peroxide, 1 mM), followed by normoxic atmosphere for different time points in the absence and presence of varying concentrations of TGF-ß1. Transepithelial electrical resistance (TER) assessed barrier function, with RNA extracted for reverse transcription polymerase chain reaction analysis of GPx-1, HIF-1, heme-oxygenase-1 (HO-1), and NOX-1. In some experiments, intestinal epithelia were exposed to enterohemorrhagic Escherichia coli (EHEC) O157:H7 during the reoxygenation period and TER recorded 7 h after the infectious challenge. Hypoxia-reoxygenation significantly decreased TER in intestinal epithelia compared with normoxic controls. Transforming growth factor-ß1 pretreatment ameliorated HR-induced epithelial barrier dysfunction in T84 (at 1 - 3 h) and Caco-2 (1 h) monolayers. Transforming growth factor-ß1 preserved barrier integrity for up to 16 h after challenge with hydrogen peroxide. In TGF-ß1-treated epithelial monolayers, only HO-1 mRNA significantly increased after HR (P < 0.05 vs. normoxic controls). The EHEC-induced epithelial barrier dysfunction was significantly worsened by intestinal HR (P < 0.05 vs. normoxia-EHEC-infected cells), but this was not protected by TGF-ß1 pretreatment. Transforming growth factor-ß1 preserves loss of epithelial barrier integrity caused by the stress of HR via a mechanism that may involve the upregulation of HO-1 transcription. Targeted treatment with TGF-ß could lead to novel therapies in enteric diseases characterized by HR injury.


Assuntos
Mucosa Intestinal/efeitos dos fármacos , Estresse Oxidativo , Fator de Crescimento Transformador beta1/farmacologia , Células CACO-2 , Hipóxia Celular , Citocinas/metabolismo , Impedância Elétrica , Escherichia coli Êntero-Hemorrágica/patogenicidade , Células Epiteliais/efeitos dos fármacos , Epitélio/metabolismo , Expressão Gênica , Heme Oxigenase-1/metabolismo , Humanos , Peróxido de Hidrogênio/química , Enteropatias/metabolismo , Mucosa Intestinal/patologia , Oxigênio/química , RNA Mensageiro/metabolismo , Proteínas Recombinantes/farmacologia , Fatores de Tempo
12.
J Nutr ; 144(11): 1725-33, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25143376

RESUMO

BACKGROUND: Few studies have focused on the ability of prebiotics to prevent pathogen-induced cellular changes or alter the composition of the intestinal microbiota in complimentary relevant cell and animal models of inflammatory bowel disease. OBJECTIVE: The objective of this study was to determine if pretreatment with inulin and a short-chain fructo-oligosaccharide (sc-FOS) prevents enterohemorrhagic Escherichia coli (EHEC) O157:H7 infection in Caco2-bbe epithelial cells and what effect 10% wt:v sc-FOS or inulin has on C57BL/6 mice under sham conditions or pretreatment with prebiotics before Citrobacter rodentium infection (10(8) colony-forming units). METHODS: Actin rearrangement and tight junction protein (zona occludin-1) were examined with immunofluorescence. Barrier function was assessed by a fluorescent probe and by measuring transepithelial electrical resistance (TER). Alterations in cytokine gene expression and microbiome were assessed with quantitative reverse transcriptase-polymerase chain reaction and fluorescence in situ hybridization. Short-chain fatty acids (SCFAs) were measured by GC. RESULTS: sc-FOS added to monolayers altered actin polymerization without affecting TER or permeability to a fluorescein isothiocyanate (FITC) probe, whereas inulin increased TER (P < 0.005) and altered actin arrangement without affecting FITC permeability. Neither prebiotic attenuated EHEC-induced decreases in barrier function. Prebiotics increased interleukin 10 (Il10) and transforming growth factor-ß (Tgfß) cytokine responses alone (P < 0.05) or with EHEC O157:H7 infection (P < 0.05) in vitro. Increases in tumor necrosis factor-α (Tnfα) (P < 0.05) and decreases in chemokine CXC motif ligand 8 (Cxcl8) (P < 0.05) expression were observed with prebiotic treatment prior to EHEC infection. No differences were noted in barrier function or cytokine responses in the absence or presence of C. rodentium in vivo. Alterations in microbiome were evident at 6 d and 10 d postinfection in treatment groups, but a change in C. rodentium load was not observed. Inulin and sc-FOS (P < 0.05) increased fecal SCFAs in the absence of infection. CONCLUSION: This study provides new insights as to how prebiotics act in complementary in vitro and in vivo models of intestinal injury.


Assuntos
Infecções por Enterobacteriaceae/complicações , Escherichia coli O157 , Inflamação/tratamento farmacológico , Inulina/farmacologia , Oligossacarídeos/farmacologia , Animais , Anti-Inflamatórios/química , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Células CACO-2 , Citrobacter rodentium , Colite/tratamento farmacológico , Colite/microbiologia , Fezes/microbiologia , Feminino , Humanos , Inulina/química , Camundongos , Camundongos Endogâmicos C57BL , Oligossacarídeos/química , Prebióticos
13.
Infect Immun ; 82(4): 1648-56, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24491575

RESUMO

Enterohemorrhagic Escherichia coli serotype O157:H7 causes outbreaks of diarrhea, hemorrhagic colitis, and the hemolytic-uremic syndrome. E. coli O157:H7 intimately attaches to epithelial cells, effaces microvilli, and recruits F-actin into pedestals to form attaching and effacing lesions. Lipid rafts serve as signal transduction platforms that mediate microbe-host interactions. The aims of this study were to determine if protein kinase C (PKC) is recruited to lipid rafts in response to E. coli O157:H7 infection and what role it plays in attaching and effacing lesion formation. HEp-2 and intestine 407 tissue culture epithelial cells were challenged with E. coli O157:H7, and cell protein extracts were then separated by buoyant density ultracentrifugation to isolate lipid rafts. Immunoblotting for PKC was performed, and localization in lipid rafts was confirmed with an anti-caveolin-1 antibody. Isoform-specific PKC small interfering RNA (siRNA) was used to determine the role of PKC in E. coli O157:H7-induced attaching and effacing lesions. In contrast to uninfected cells, PKC was recruited to lipid rafts in response to E. coli O157:H7. Metabolically active bacteria and cells with intact lipid rafts were necessary for the recruitment of PKC. PKC recruitment was independent of the intimin gene, type III secretion system, and the production of Shiga toxins. Inhibition studies, using myristoylated PKCζ pseudosubstrate, revealed that atypical PKC isoforms were activated in response to the pathogen. Pretreating cells with isoform-specific PKC siRNA showed that PKCζ plays a role in E. coli O157:H7-induced attaching and effacing lesions. We concluded that lipid rafts mediate atypical PKC signal transduction responses to E. coli O157:H7. These findings contribute further to the understanding of the complex array of microbe-eukaryotic cell interactions that occur in response to infection.


Assuntos
Aderência Bacteriana/fisiologia , Infecções por Escherichia coli/microbiologia , Escherichia coli O157/fisiologia , Proteína Quinase C/fisiologia , Células Cultivadas , Células Epiteliais/microbiologia , Escherichia coli O157/patogenicidade , Humanos , Immunoblotting , Microdomínios da Membrana/fisiologia , Transdução de Sinais/fisiologia , Fatores de Virulência/fisiologia
14.
J Med Chem ; 56(6): 2218-34, 2013 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-23394126

RESUMO

A series of novel, highly potent, selective, and ATP-competitive mammalian target of rapamycin (mTOR) inhibitors based on a benzoxazepine scaffold have been identified. Lead optimization resulted in the discovery of inhibitors with low nanomolar activity and greater than 1000-fold selectivity over the closely related PI3K kinases. Compound 28 (XL388) inhibited cellular phosphorylation of mTOR complex 1 (p-p70S6K, pS6, and p-4E-BP1) and mTOR complex 2 (pAKT (S473)) substrates. Furthermore, this compound displayed good pharmacokinetics and oral exposure in multiple species with moderate bioavailability. Oral administration of compound 28 to athymic nude mice implanted with human tumor xenografts afforded significant and dose-dependent antitumor activity.


Assuntos
Trifosfato de Adenosina/metabolismo , Ligação Competitiva , Descoberta de Drogas , Inibidores de Proteínas Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Administração Oral , Animais , Benzoxazinas/química , Benzoxazinas/metabolismo , Benzoxazinas/farmacocinética , Benzoxazinas/farmacologia , Disponibilidade Biológica , Linhagem Celular Tumoral , Cães , Feminino , Humanos , Masculino , Camundongos , Modelos Moleculares , Conformação Proteica , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacocinética , Ratos , Especificidade por Substrato , Serina-Treonina Quinases TOR/química
15.
Benef Microbes ; 4(2): 195-209, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23443951

RESUMO

Enterohaemorrhagic Escherichia coli O157:H7 and adherent-invasive Escherichia coli are two groups of enteric bacterial pathogens associated with haemorrhagic colitis and Crohn's Disease, respectively. Bacterial contact with host epithelial cells stimulates an immediate innate immune response designed to combat infection. In this study, immune responses of human epithelial cells to pathogens, either alone or in combination with probiotic bacteria were studied. Industrially prepared Lactobacillus helveticus strain R0052 was first examined by microarray analysis and then compared to broth-grown strains of R0052 and Lactobacillus rhamnosus strain GG using quantitative realt-time polymerase chain reaction. Results showed host immune activation responses increased following pathogen exposure, which were differentially ameliorated using probiotics depending on both the preparation of probiotics employed and conditions of exposure. These findings provide additional support for the concept that specific probiotic strains serve as a promising option for use in preventing the risk of enteric bacterial infections.


Assuntos
Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Escherichia coli/imunologia , Fatores Imunológicos/farmacologia , Lacticaseibacillus rhamnosus/imunologia , Lactobacillus helveticus/imunologia , Probióticos/farmacologia , Células CACO-2 , Perfilação da Expressão Gênica , Humanos , Análise em Microsséries , Reação em Cadeia da Polimerase em Tempo Real
16.
Bioorg Med Chem Lett ; 22(24): 7653-8, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23127890

RESUMO

We report the discovery of a series of 4-aryl-2-aminoalkylpyrimidine derivatives as potent and selective JAK2 inhibitors. High throughput screening of our in-house compound library led to the identification of hit 1, from which optimization resulted in the discovery of highly potent and selective JAK2 inhibitors. Advanced lead 10d demonstrated a significant dose-dependent pharmacodynamic and antitumor effect in a mouse xenograft model. Based upon the desirable profile of 10d (XL019) it was advanced into clinical trials.


Assuntos
Antineoplásicos/farmacologia , Janus Quinase 2/antagonistas & inibidores , Neoplasias Experimentais/tratamento farmacológico , Prolina/análogos & derivados , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Cristalografia por Raios X , Cães , Relação Dose-Resposta a Droga , Haplorrinos , Ensaios de Triagem em Larga Escala , Janus Quinase 2/metabolismo , Camundongos , Camundongos Nus , Modelos Moleculares , Estrutura Molecular , Neoplasias Experimentais/patologia , Prolina/administração & dosagem , Prolina/química , Prolina/farmacologia , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/química , Pirimidinas/administração & dosagem , Pirimidinas/química , Ratos , Relação Estrutura-Atividade , Ensaios Antitumorais Modelo de Xenoenxerto
17.
J Med Chem ; 55(11): 5467-82, 2012 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-22548342

RESUMO

The phosphoinositide 3-kinases (PI3Ks) have been linked to an extraordinarily diversified group of cellular functions making these enzymes compelling targets for the treatment of disease. A large body of evidence has linked PI3Kγ to the modulation of autoimmune and inflammatory processes making it an intriguing target for drug discovery. Our high-throughput screening (HTS) campaign revealed two hits that were nominated for further optimization studies. The in vitro activity of the first HTS hit, designated as the sulfonylpiperazine scaffold, was optimized utilizing structure-based design. However, nonoptimal pharmacokinetic properties precluded this series from further studies. An overlay of the X-ray structures of the sulfonylpiperazine scaffold and the second HTS hit within their complexes with PI3Kγ revealed a high degree of overlap. This feature was utilized to design a series of hybrid analogues including advanced leads such as 31 with desirable potency, selectivity, and oral bioavailability.


Assuntos
Inibidores de Fosfoinositídeo-3 Quinase , Piperazinas/síntese química , Sulfonamidas/síntese química , Sulfonas/síntese química , Administração Oral , Animais , Disponibilidade Biológica , Linhagem Celular , Cristalografia por Raios X , Feminino , Ensaios de Triagem em Larga Escala , Humanos , Isoenzimas/antagonistas & inibidores , Medições Luminescentes , Camundongos , Microssomos Hepáticos/metabolismo , Modelos Moleculares , Estrutura Molecular , Fosforilação , Piperazinas/farmacocinética , Piperazinas/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade , Sulfonamidas/farmacocinética , Sulfonamidas/farmacologia , Sulfonas/farmacocinética , Sulfonas/farmacologia
18.
Infect Immun ; 80(7): 2307-15, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22526675

RESUMO

Enterohemorrhagic Escherichia coli (EHEC) serotype O157:H7 is a food-borne pathogen that causes significant morbidity and mortality in developing and industrialized nations. EHEC infection of host epithelial cells is capable of inhibiting the gamma interferon (IFN-γ) proinflammatory pathway through the inhibition of Stat-1 phosphorylation, which is important for host defense against microbial pathogens. The aim of this study was to determine the bacterial factors involved in the inhibition of Stat-1 tyrosine phosphorylation. Human HEp-2 and Caco-2 epithelial cells were challenged directly with either EHEC or bacterial culture supernatants and stimulated with IFN-γ, and then the protein extracts were analyzed by immunoblotting. The data showed that IFN-γ-mediated Stat-1 tyrosine phosphorylation was inhibited by EHEC secreted proteins. Using two-dimensional difference gel electrophoresis, EHEC Shiga toxins were identified as candidate inhibitory factors. EHEC Shiga toxin mutants were then generated and complemented in trans, and mutant culture supernatant was supplemented with purified Stx to confirm their ability to subvert IFN-γ-mediated cell activation. We conclude that while other factors are likely involved in the suppression of IFN-γ-mediated Stat-1 tyrosine phosphorylation, E. coli-derived Shiga toxins represent a novel mechanism by which EHEC evades the host immune system.


Assuntos
Escherichia coli O157/imunologia , Escherichia coli O157/patogenicidade , Evasão da Resposta Imune , Interferon gama/imunologia , Fator de Transcrição STAT1/imunologia , Toxinas Shiga/imunologia , Toxinas Shiga/toxicidade , Western Blotting , Linhagem Celular , Eletroforese em Gel Bidimensional , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Humanos , Interferon gama/antagonistas & inibidores , Fosforilação , Proteoma/análise , Fator de Transcrição STAT1/metabolismo
19.
J Infect Dis ; 206(1): 99-109, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22430833

RESUMO

BACKGROUND: Probiotics prevent disease induced by Citrobacter rodentium, a murine-specific enteric pathogen. Whether probiotics can be used to interrupt the infectious process following initiation of infection was determined. METHODS: C57BL/6 adult and neonatal mice were challenged with C. rodentium, and a probiotic mixture containing Lactobacillus helveticus and Lactobacillus rhamnosus was provided 1 week before bacterial challenge, concurrently with infection, or 3 days and 6 days after infection. Mice were sacrificed 10 days after infection, and disease severity was assessed by histological analysis and in vivo intestinal permeability assay. Inflammatory pathways and the composition of the fecal microbiome were assessed in adult mice. RESULTS: Preadministration and coadministration of probiotics ameliorated C. rodentium-induced barrier dysfunction, epithelial hyperplasia, and binding of the pathogen to host colonocytes in adults, with similar findings in neonatal mice. Upregulated tumor necrosis factor α and interferon γ transcripts were suppressed in the pretreated probiotic group, whereas interleukin 17 transcription was suppressed with probiotics given up to 3 days after infection. Probiotics promoted transcription of interleukin 10 and FOXP3, and increased follicular T-regulatory cells in pretreatment mice. C. rodentium infection resulted in an altered fecal microbiome, which was normalized with probiotic intervention. CONCLUSIONS: This study provides evidence that probiotics can prevent illness and treat disease in an animal model of infectious colitis.


Assuntos
Citrobacter rodentium/crescimento & desenvolvimento , Colite/terapia , Infecções por Enterobacteriaceae/terapia , Lacticaseibacillus rhamnosus , Lactobacillus helveticus , Probióticos/farmacologia , Animais , Citrobacter rodentium/metabolismo , Colite/metabolismo , Colite/microbiologia , Colo/metabolismo , Colo/microbiologia , Infecções por Enterobacteriaceae/metabolismo , Infecções por Enterobacteriaceae/microbiologia , Fezes/microbiologia , Feminino , Fatores de Transcrição Forkhead/metabolismo , Hiperplasia/metabolismo , Hiperplasia/microbiologia , Hiperplasia/prevenção & controle , Hiperplasia/terapia , Interferon gama/metabolismo , Interleucina-10/metabolismo , Interleucina-17/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Metagenoma , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/microbiologia , Fator de Necrose Tumoral alfa/metabolismo
20.
J Med Chem ; 55(3): 1368-81, 2012 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-22214363

RESUMO

A series of subtype selective sphingosine 1-phosphate receptor 1 (S1P(1)) antagonists are disclosed. Our high-throughput screening campaign revealed hit 1 for which an increase in potency and mouse oral exposure was achieved with minor modifications to the chemical scaffold. In vivo efficacy revealed that at high doses compounds 12 and 15 inhibited tumor growth. Further optimization of our lead series led to the discovery of proline derivatives 37 (XL541) and 38 which had similar efficacy as our first generation analogues at significantly lower doses. Analogue 37 displayed excellent pharmacokinetics and oral exposure in multiple species.


Assuntos
Antineoplásicos/síntese química , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Administração Oral , Amidas/síntese química , Amidas/farmacocinética , Amidas/farmacologia , Inibidores da Angiogênese/síntese química , Inibidores da Angiogênese/farmacocinética , Inibidores da Angiogênese/farmacologia , Compostos de Anilina/síntese química , Compostos de Anilina/farmacocinética , Compostos de Anilina/farmacologia , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Disponibilidade Biológica , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Cães , Haplorrinos , Ensaios de Triagem em Larga Escala , Camundongos , Neovascularização Patológica , Prolina/análogos & derivados , Prolina/síntese química , Prolina/farmacocinética , Prolina/farmacologia , Ratos , Serina/análogos & derivados , Serina/síntese química , Serina/farmacocinética , Serina/farmacologia , Estereoisomerismo , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA