Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros

Bases de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Immunol ; 201(2): 772-781, 2018 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-29884698

RESUMO

Leukocyte-associated Ig-like receptor 1 (LAIR1) is an ITIM-bearing collagen receptor expressed by leukocytes and is implicated in immune suppression. However, using a divalent soluble LAIR1/Fc recombinant protein to block interaction of cell surface LAIR1 with matrix collagen, we found that whereas Th1 responses were enhanced as predicted, Th17 responses were strongly inhibited. Indeed, LAIR1 on both T cells and monocytes was required for optimal Th17 responses to collagen type (Col)V. For pre-existing "natural" Th17 response to ColV, the LAIR1 requirement was absolute, whereas adaptive Th17 and Th1/17 immune responses in both mice and humans were profoundly reduced in the absence of LAIR1. Furthermore, the addition of C1q, a natural LAIR1 ligand, decreased Th1 responses in a dose-dependent manner, but it had no effect on Th17 responses. In IL-17-dependent murine organ transplant models of chronic rejection, LAIR1+/+ but not LAIR1-/- littermates mounted strong fibroproliferative responses. Surface LAIR1 expression was higher on human Th17 cells as compared with Th1 cells, ruling out a receptor deficiency that could account for the differences. We conclude that LAIR1 ligation by its natural ligands favors Th17 cell development, allowing for preferential activity of these cells in collagen-rich environments. The emergence of cryptic self-antigens such as the LAIR1 ligand ColV during ischemia/reperfusion injury and early acute rejection, as well as the tendency of macrophages/monocytes to accumulate in the allograft during chronic rejection, favors Th17 over Th1 development, posing a risk to long-term graft survival.


Assuntos
Rejeição de Enxerto/imunologia , Receptores Imunológicos/metabolismo , Células Th1/fisiologia , Células Th17/imunologia , Animais , Autoantígenos/imunologia , Células Cultivadas , Colágeno/metabolismo , Humanos , Imunidade Celular , Imunomodulação , Interleucina-17/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/imunologia , Transplante de Órgãos , Ligação Proteica , Receptores Imunológicos/genética
2.
Liver Transpl ; 22(1): 91-102, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26285140

RESUMO

Hepatic ischemia/reperfusion injury (IRI) is a critical component of hepatic surgery. Oxidative stress has long been implicated as a key player in IRI. In this study, we examine the cell-specific role of the nuclear factor (erythroid-derived 2)-like 2 (Nrf2)-antioxidant response element pathway in warm hepatic IRI. Nrf2 knockout (KO) and wild-type (WT) animals and novel transgenic mice expressing a constitutively active nuclear factor (erythroid-derived 2)-like 2 (caNrf2) mutant in hepatocytes (AlbCre+/caNrf2+) and their littermate controls underwent partial hepatic ischemia or sham surgery. The animals were killed 6 hours after reperfusion, and their serum and tissue were collected for analysis. As compared to WT animals after ischemia/reperfusion (IR), Nrf2 KO mice had increased hepatocellular injury with increased serum alanine aminotransferase and aspartate aminotransferase, Suzuki score, apoptosis, an increased inflammatory infiltrate, and enhanced inflammatory cytokine expression. On the other hand, AlbCre+/caNrf2+ that underwent IR had significantly reduced serum transaminases, less necrosis on histology, and a less pronounced inflammatory infiltrate and inflammatory cytokine expression as compared to the littermate controls. However, there were no differences in apoptosis. Taken together, Nrf2 plays a critical role in our murine model of warm hepatic IRI, with Nrf2 deficiency exacerbating hepatic IRI and hepatocyte-specific Nrf2 overactivation providing protection against warm hepatic IRI.


Assuntos
Hepatócitos/metabolismo , Fígado/irrigação sanguínea , Fator 2 Relacionado a NF-E2/metabolismo , Traumatismo por Reperfusão/metabolismo , Animais , Apoptose , Modelos Animais de Doenças , Expressão Gênica , Camundongos Transgênicos , Fator 2 Relacionado a NF-E2/genética , Transdução de Sinais
3.
Liver Transpl ; 22(8): 1115-28, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27113842

RESUMO

Hepatic ischemia/reperfusion injury (IRI) is an unavoidable consequence of liver transplantation that can lead to postoperative hepatic dysfunction. Myeloid cells that include Kupffer cells, monocytes, and neutrophils contribute to the inflammatory response and cellular injury observed during hepatic IRI. We hypothesize that overactivation of the nuclear erythroid 2 p45-related factor 2 (Nrf2)-antioxidant response element (ARE) pathway in myeloid cells leads to decreased cellular damage after hepatic IRI. We constructed transgenic mice with constitutively active nuclear erythroid 2 p45-related factor 2 (caNrf2) that over activates the Nrf2-ARE pathway in myeloid cells (lysozyme M cre recombinase [LysMcre]+/caNrf2+, n = 9), and their littermate controls lacking transgene expression (LysMcre+/caNrf2-, n = 11). The mice underwent either sham or partial hepatic ischemia surgery, with 60 minutes of ischemia followed by 6 hours of reperfusion. After IRI, LysMcre+/caNrf2+ mice demonstrated significantly decreased serum alanine aminotransferase and decreased areas of necrosis. Immunohistochemistry and immunoblot of caspase 3 showed a significantly decreased cleaved to full-length caspase 3 ratio in LysMcre+/caNrf2+ animals. Lymphocyte antigen 6 complex locus G and CD68 staining demonstrated reduced inflammatory cell infiltration. LysMcre+/caNrf2+ animals also had significantly decreased gene expression of proinflammatory cytokines, including interleukin (IL) 1ß, IL6, tumor necrosis factor α, chemokine (C-C motif) ligand 2, and chemokine (C-X-C motif) ligand 10, and significantly decreased levels of 8-isoprostanes. In our model, Nrf2 overactivation in myeloid cells leads to decreased hepatocellular damage, necrosis, apoptosis, inflammation, and oxidative stress. Pharmacologic targeting of the Nrf2-ARE pathway in myeloid cells may be a novel strategy to mitigate hepatic IRI. Liver Transplantation 22 1115-1128 2016 AASLD.


Assuntos
Doença Hepática Terminal/cirurgia , Transplante de Fígado/efeitos adversos , Fígado/patologia , Fator 2 Relacionado a NF-E2/metabolismo , Traumatismo por Reperfusão/metabolismo , Alanina Transaminase/sangue , Animais , Elementos de Resposta Antioxidante , Apoptose , Caspase 3/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Imuno-Histoquímica , Células de Kupffer/metabolismo , Fígado/citologia , Camundongos , Camundongos Transgênicos , Monócitos/metabolismo , Fator 2 Relacionado a NF-E2/genética , Necrose , Neutrófilos/metabolismo , Estresse Oxidativo , Traumatismo por Reperfusão/sangue , Traumatismo por Reperfusão/etiologia , Transdução de Sinais , Regulação para Cima
4.
Am J Pathol ; 185(7): 2000-11, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25987251

RESUMO

Null alleles for the COL5A1 gene and missense mutations for COL5A1 or the COL5A2 gene underlie cases of classic Ehlers-Danlos syndrome, characterized by fragile, hyperextensible skin and hypermobile joints. However, no classic Ehlers-Danlos syndrome case has yet been associated with COL5A2 null alleles, and phenotypes that might result from such alleles are unknown. We describe mice with null alleles for the Col5a2. Col5a2(-/-) homozygosity is embryonic lethal at approximately 12 days post conception. Unlike previously described mice null for Col5a1, which die at 10.5 days post conception and virtually lack collagen fibrils, Col5a2(-/-) embryos have readily detectable collagen fibrils, thicker than in wild-type controls. Differences in Col5a2(-/-) and Col5a1(-/-) fibril formation and embryonic survival suggest that α1(V)3 homotrimers, a rare collagen V isoform that occurs in the absence of sufficient levels of α2(V) chains, serve functional roles that partially compensate for loss of the most common collagen V isoform. Col5a2(+/-) adults have skin with marked hyperextensibility and reduced tensile strength at high strain but not at low strain. Col5a2(+/-) adults also have aortas with increased compliance and reduced tensile strength. Results thus suggest that COL5A2(+/-) humans, although unlikely to present with frank classic Ehlers-Danlos syndrome, are likely to have fragile connective tissues with increased susceptibility to trauma and certain chronic pathologic conditions.


Assuntos
Colágeno Tipo V/genética , Colágeno/genética , Síndrome de Ehlers-Danlos/genética , Adulto , Alelos , Animais , Colágeno/metabolismo , Colágeno Tipo V/metabolismo , Tecido Conjuntivo/anormalidades , Tecido Conjuntivo/patologia , Síndrome de Ehlers-Danlos/metabolismo , Síndrome de Ehlers-Danlos/patologia , Feminino , Heterozigoto , Homozigoto , Humanos , Masculino , Camundongos , Camundongos Knockout , Mutação , Fenótipo , Pele/patologia
5.
Biomacromolecules ; 14(5): 1696-704, 2013 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-23597075

RESUMO

We report an approach to deliver DNA to vascular tissue in vivo using intravascular stents coated with degradable, DNA-containing polyelectrolyte multilayers (PEMs). Ionically cross-linked multilayers ∼120 nm thick were fabricated layer-by-layer on the surfaces of balloon-mounted stainless steel stents using plasmid DNA and a hydrolytically degradable poly(ß-amino ester) (polymer 1). Characterization of stents coated using a fluorescently end-labeled analog of polymer 1 revealed film erosion to be uniform across the surfaces of the stents; differential stresses experienced upon balloon expansion did not lead to faster film erosion or dose dumping of DNA in areas near stent joints when stents were incubated in physiologically relevant media. The ability of film-coated stents to transfer DNA and transfect arterial tissue in vivo was then investigated in pigs and rabbits. Stents coated with films fabricated using fluorescently labeled DNA resulted in uniform transfer of DNA to sub-endothelial tissue in the arteries of pigs in patterns corresponding to the locations and geometries of stent struts. Stents coated with films fabricated using polymer 1 and plasmid DNA encoding EGFP resulted in expression of EGFP in the medial layers of stented tissue in both pigs and rabbits two days after implantation. The results of this study, combined with the modular and versatile nature of layer-by-layer assembly, provide a polymer-based platform that is well suited for fundamental studies of stent-mediated gene transfer. With further development, this approach could also prove useful for the design of nonviral, gene-based approaches for prevention of complications that arise from the implantation of stents and other implantable interventional devices.


Assuntos
Artérias Carótidas/metabolismo , DNA/genética , Endotélio Vascular/metabolismo , Técnicas de Transferência de Genes , Polímeros/química , Stents , Animais , Artérias Carótidas/citologia , Materiais Revestidos Biocompatíveis , DNA/metabolismo , Eletrólitos , Endotélio Vascular/citologia , Genes Reporter , Proteínas de Fluorescência Verde/genética , Plasmídeos , Coelhos , Suínos
6.
J Vasc Surg ; 56(2): 446-54, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22521802

RESUMO

INTRODUCTION: We have previously demonstrated that transforming growth factor-ß (TGF-ß) in the presence of elevated levels of Smad3, its primary signaling protein, stimulates rat vascular smooth muscle cell (VSMC) proliferation and intimal hyperplasia. The mechanism is partly through the nuclear exportation of phosphorylated cyclin-dependent kinase inhibitor p27. The objective of this study is to clarify the downstream pathways through which Smad3 produces its proliferative effect. Specifically, we evaluated the role of extracellular signal-regulated kinase (ERK) mitogen-activated protein kinase (MAPK) in TGF-ß-induced VSMC proliferation. METHODS: Cultured rat aortic VSMCs were incubated with TGF-ß at varying concentrations and times, and phosphorylated ERK was measured by Western blotting. Smad3 was enhanced in VSMCs using an adenovirus expressing Smad3 or inhibited with small interfering RNA (siRNA). For in vivo experiments, male Sprague-Dawley rats underwent carotid balloon injury, followed by intraluminal infection with an adenovirus expressing Smad3. Arteries were harvested at 3 days and subjected to immunohistochemistry for Smad3, phospho-ERK MAPK, and proliferating cell nuclear antigen. RESULTS: In cultured VSMCs, TGF-ß induced activation and phosphorylation of ERK MAPK in a time-dependent and concentration-dependent manner. Overexpression of the signaling protein Smad3 enhanced TGF-ß-induced activation of ERK MAPK, whereas inhibition of Smad3 with a siRNA blocked ERK MAPK phosphorylation in response to TGF-ß. These data suggest that Smad3 acts as a signaling intermediate between TGF-ß and ERK MAPK. Inhibition of ERK MAPK activation with PD98059 completely blocked the ability of TGF-ß/Smad3 to stimulate VSMC proliferation, demonstrating the importance of ERK MAPK in this pathway. Immunoprecipitation of phospho-ERK MAPK and blotting with Smad3 revealed a physical association, suggesting that activation of ERK MAPK by Smad3 requires a direct interaction. In an in vivo rat carotid injury model, overexpression of Smad3 resulted in an increase in phosphorylated ERK MAPK as well as increased VSMC proliferation as measured by proliferating cell nuclear antigen. CONCLUSIONS: Our findings demonstrate a mechanism through which TGF-ß stimulates VSMC proliferation. Although TGF-ß has been traditionally identified as an inhibitor of proliferation, our data suggest that TGF-ß enhances VSMC proliferation through a Smad3/ERK MAPK signaling pathway. These findings at least partly explain the mechanism by which TGF-ß enhances intimal hyperplasia. Knowledge of this pathway provides potential novel targets that may be used to prevent restenosis.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Músculo Liso Vascular/citologia , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Animais , Aorta/citologia , Proliferação de Células , Sobrevivência Celular , Imuno-Histoquímica , Masculino , Fosforilação , Ratos , Ratos Sprague-Dawley , Regulação para Cima/fisiologia
7.
Circ Res ; 107(9): 1106-16, 2010 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-20814021

RESUMO

RATIONALE: Considerable evidence shows atherosclerosis to be a chronic inflammatory disease in which immunity to self-antigens contributes to disease progression. We recently identified the collagen type V [col(V)] α1(V) chain as a key autoantigen driving the Th17-dependent cellular immunity underlying another chronic inflammatory disease, obliterative bronchiolitis. Because specific induction of α1(V) chains has previously been reported in human atheromas, we postulated involvement of col(V) autoimmunity in atherosclerosis. OBJECTIVE: To determine whether col(V) autoimmunity may be involved in the pathogenesis of atherosclerosis. METHODS AND RESULTS: Here, we demonstrate Th17-dependent anti-col(V) immunity to be characteristic of atherosclerosis in human coronary artery disease (CAD) patients and in apolipoprotein E-null (ApoE(-/-)) atherosclerotic mice. Responses were α1(V)-specific in CAD with variable Th1 pathway involvement. In early atherosclerosis in ApoE(-/-) mice, anti-col(V) immunity was tempered by an interleukin (IL)-10-dependent mechanism. In support of a causal role for col(V) autoimmunity in the pathogenesis of atherosclerosis, col(V) sensitization of ApoE(-/-) mice on a regular chow diet overcame IL-10-mediated inhibition of col(V) autoimmunity, leading to increased atherosclerotic burden in these mice and local accumulation of IL-17-producing cells, particularly in the col(V)-rich adventitia subjacent to the atheromas. CONCLUSIONS: These findings establish col(V) as an autoantigen in human CAD and show col(V) autoimmunity to be a consistent feature in atherosclerosis in humans and mice. Furthermore, data are consistent with a causative role for col(V) in the pathogenesis of atherosclerosis.


Assuntos
Aterosclerose/imunologia , Doenças Autoimunes/imunologia , Colágeno Tipo V/fisiologia , Interleucina-17/fisiologia , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/patologia , Doenças Autoimunes/genética , Doenças Autoimunes/patologia , Bovinos , Colágeno Tipo V/efeitos adversos , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Células Th1/imunologia , Células Th1/metabolismo , Células Th1/patologia
8.
J Surg Res ; 176(2): 639-48, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22440934

RESUMO

BACKGROUND: Donor brain death (BD) triggers a systemic inflammatory response that reduces organ quality and increases immunogenicity of the graft. We characterized the early innate immune response induced by BD in the liver and peripheral blood of hemodinamically stable non-human primates (NHP). METHODS: Rhesus macaques were assigned to either brain death or control group. BD was induced by inflation of a subdurally placed catheter and confirmed clinically and by cerebral angiography. Animals were monitored for 6 h after BD and managed to maintain hemodynamic stability. RESULTS: Cortisol, epinephrine, nor-epinephrine, and IL-6 levels were elevated immediately after BD induction. Neutrophils and monocytes significantly increased in circulation following BD induction, while dendritic cells were decreased at 6 h post-induction. Flow cytometry revealed increased expression of chemokine receptors CxCR1, CxCR2, CCR2, and CCR5 in peripheral blood leukocytes from NHP subjected to BD. Microarray analysis demonstrated a significant up-regulation of genes related to innate inflammatory responses, toll-like receptor signaling, stress pathways, and apoptosis/cell death in BD subjects. Conversely, pathways related to glucose, lipid, and protein metabolism were down-regulated. In addition, increased expression of SOCS3, S100A8/A9, ICAM-1, MHC class II, neutrophil accumulation, and oxidative stress markers (carboxy-methyl-lysine and hydroxynonenal) were detected by immunoblot and immunohistochemistry. CONCLUSIONS: Activation of the innate immune response after BD in association with a down-regulation of genes associated with cell metabolism pathways in the liver. These findings may provide a potential explanation for the reduced post-transplant function of organs from brain dead donors. In addition, this work suggests potential novel targets to improve donor management strategies.


Assuntos
Morte Encefálica/imunologia , Hepatite/imunologia , Imunidade Inata/imunologia , Transplante de Fígado , Fígado/imunologia , Doadores de Tecidos , Animais , Citocinas/sangue , Epinefrina/sangue , Perfilação da Expressão Gênica , Hepatite/genética , Hidrocortisona/sangue , Imunidade Inata/genética , Células de Kupffer/imunologia , Linfócitos/imunologia , Macaca mulatta , Neutrófilos/imunologia , Norepinefrina/sangue , Receptores Toll-Like/metabolismo
9.
Blood ; 114(17): 3578-87, 2009 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-19700665

RESUMO

In mice and humans, the immunologic effects of developmental exposure to noninherited maternal antigens (NIMAs) are quite variable. This heterogeneity likely reflects differences in the relative levels of NIMA-specific T regulatory (T(R)) versus T effector (T(E)) cells. We hypothesized that maintenance of NIMA-specific T(R) cells in the adult requires continuous exposure to maternal cells and antigens (eg, maternal microchimerism [MMc]). To test this idea, we used 2 sensitive quantitative polymerase chain reaction (qPCR) tests to detect MMc in different organs of NIMA(d)-exposed H2(b) mice. MMc was detected in 100% of neonates and a majority (61%) of adults; nursing by a NIMA+ mother was essential for preserving MMc into adulthood. MMc was most prevalent in heart, lungs, liver, and blood, but was rarely detected in unfractionated lymphoid tissues. However, MMc was detectable in isolated CD4+, CD11b+, and CD11c+ cell subsets of spleen, and in lineage-positive cells in heart. Suppression of delayed type hypersensitivity (DTH) and in vivo lymphoproliferation correlated with MMc levels, suggesting a link between T(R) and maternal cell engraftment. In the absence of neonatal exposure to NIMA via breastfeeding, MMc was lost, which was accompanied by sensitization to NIMA in some offspring, indicating a role of oral exposure in maintaining a favorable T(R) > T(E) balance.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Quimerismo , Antígenos H-2/fisiologia , Hipersensibilidade Tardia/imunologia , Tolerância Imunológica/imunologia , Linfócitos T Reguladores/imunologia , Animais , Linfócitos T CD4-Positivos/metabolismo , Cruzamentos Genéticos , Feminino , Citometria de Fluxo , Hipersensibilidade Tardia/metabolismo , Técnicas Imunoenzimáticas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Knockout , Mães , Reação em Cadeia da Polimerase , Linfócitos T Reguladores/metabolismo
10.
Cytotherapy ; 13(4): 400-6, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21090918

RESUMO

BACKGROUND AIMS: For many years the human heart has been considered a terminally differentiated organ with no regenerative potential after injury. Recent studies, however, have cast doubt on this long-standing dogma. The objective of this study was to investigate the presence of and characterize mesenchymal stromal cells (MSC) in the adult mouse heart. The impact of MSC on growth and differentiation of adult cardiac stem cells (CSC) was also analyzed. METHODS: A combination of lineage-negative/c-kit-negative (Lin(-)/c-kit(-)) immunoselection with a plastic-adhesion technique was used to isolate cardiac-derived MSC. The differentiation capacity and expression of surface markers were analyzed. To investigate the impact of MSC on growth and differentiation of adult CSC, Green Fluorescent Protein (GFP(+)) adult CSC were co-cultured with GFP(-) cardiac-derived MSC. RESULTS: MSC were present in the adult mouse heart and they met the criteria established to define mouse MSC. They expressed surface markers and were able to differentiate, in a controlled manner, into multiple lineages. In addition, cardiac-derived MSC promoted the survival and expansion of adult CSC in vitro. CONCLUSIONS: MSC can be isolated from the mouse heart and they promote growth and differentiation of adult CSC. The findings from this study could have a significant beneficial impact on future heart failure treatment. Co-culture and co-implantation of cardiac-derived MSC with adult CSC could provide extensive cardiac regeneration and maintenance of the CSC population after implanted into the heart.


Assuntos
Células-Tronco Adultas/citologia , Células-Tronco Mesenquimais/citologia , Miocárdio/citologia , Células Estromais/citologia , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Técnicas de Cocultura , Masculino , Camundongos , Camundongos Endogâmicos C57BL
11.
Sci Rep ; 7: 41916, 2017 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-28165488

RESUMO

To date, there is no periadventitial drug delivery method available in the clinic to prevent restenotic failure of open vascular reconstructions. Resveratrol is a promising anti-restenotic natural drug but subject to low bioavailability when systemically administered. In order to reconcile these two prominent issues, we tested effects of periadventitial delivery of resveratrol on all three major pro-restenotic pathologies including intimal hyperplasia (IH), endothelium impairment, and vessel shrinkage. In a rat carotid injury model, periadventitial delivery of resveratrol either via Pluronic gel (2-week), or polymer sheath (3-month), effectively reduced IH without causing endothelium impairment and vessel shrinkage. In an in vitro model, primary smooth muscle cells (SMCs) were stimulated with elevated transforming growth factor (TGFß) and its signaling protein Smad3, known contributors to IH. TGFß/Smad3 up-regulated Kruppel-like factor (KLF5) protein, and SMC de-differentiation which was reversed by KLF5 siRNA. Furthermore, TGFß/Smad3-stimulated KLF5 production and SMC de-differentiation were blocked by resveratrol via its inhibition of the Akt-mTOR pathway. Concordantly, resveratrol attenuated Akt phosphorylation in injured arteries. Taken together, periadventitial delivery of resveratrol produces durable inhibition of all three pro-restenotic pathologies - a rare feat among existing anti-restenotic methods. Our study suggests a potential anti-restenotic modality of resveratrol application suitable for open surgery.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Reestenose Coronária/prevenção & controle , Regulação da Expressão Gênica/efeitos dos fármacos , Músculo Liso Vascular/citologia , Proteína Smad3/metabolismo , Estilbenos/farmacologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Antioxidantes/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Reestenose Coronária/metabolismo , Reestenose Coronária/patologia , Masculino , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Fosforilação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Resveratrol , Transdução de Sinais/efeitos dos fármacos
12.
EBioMedicine ; 2(11): 1650-61, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26870791

RESUMO

BACKGROUND: Intimal hyperplasia is a common cause of many vasculopathies. There has been a recent surge of interest in the bromo and extra-terminal (BET) epigenetic "readers" including BRD4 since the serendipitous discovery of JQ1(+), an inhibitor specific to the seemingly undruggable BET bromodomains. The role of the BET family in the development of intimal hyperplasia is not known. METHODS: We investigated the effect of BET inhibition on intimal hyperplasia using a rat balloon angioplasty model. RESULTS: While BRD4 was dramatically up-regulated in the rat and human hyperplastic neointima, blocking BET bromodomains with JQ1(+) diminished neointima in rats. Knocking down BRD4 with siRNA, or treatment with JQ1(+) but not the inactive enantiomer JQ1(-), abrogated platelet-derived growth factor (PDGF-BB)-stimulated proliferation and migration of primary rat aortic smooth muscle cells. This inhibitory effect of JQ1(+) was reproducible in primary human aortic smooth muscle cells. In human aortic endothelial cells, JQ1(+) prevented cytokine-induced apoptosis and impairment of cell migration. Furthermore, either BRD4 siRNA or JQ1(+) but not JQ1(-), substantially down-regulated PDGF receptor-α which, in JQ1(+)-treated arteries versus vehicle control, was also reduced. CONCLUSIONS: Blocking BET bromodomains mitigates neointima formation, suggesting an epigenetic approach for effective prevention of intimal hyperplasia and associated vascular diseases.


Assuntos
Artérias Carótidas/metabolismo , Artérias Carótidas/patologia , Proteínas Nucleares/antagonistas & inibidores , Fatores de Transcrição/antagonistas & inibidores , Túnica Íntima/metabolismo , Túnica Íntima/patologia , Animais , Azepinas/farmacologia , Artérias Carótidas/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células , Sobrevivência Celular , Citocinas/metabolismo , Células Endoteliais/efeitos dos fármacos , Humanos , Hiperplasia , Imuno-Histoquímica , Mediadores da Inflamação/metabolismo , Masculino , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Domínios e Motivos de Interação entre Proteínas , Ratos , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Triazóis/farmacologia , Túnica Íntima/efeitos dos fármacos , Veias/metabolismo , Veias/patologia
13.
JPEN J Parenter Enteral Nutr ; 39(2): 190-9, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24121183

RESUMO

BACKGROUND: The parotid and submandibular salivary glands are gut-associated lymphoid tissues (GALTs) that secrete immune compounds into the oral cavity. Parenteral nutrition (PN) without enteral stimulation decreases GALT function, including intestinal lymphocyte counts and secretory immunoglobulin A (sIgA) levels. Since the neuropeptide bombesin (BBS), a gastrin-releasing peptide analogue, stimulates intestinal function and restores GALT parameters, we hypothesized that PN + BBS would stimulate parotid and salivary gland IgA levels, T lymphocytes, and IgA plasma cell counts compared with PN alone. METHODS: Male (Institute of Cancer Research) ICR mice received intravenous catheters and were randomized to chow with saline, PN, or PN + BBS (15 µg/tid/mouse) for 5 days (8/group), 2 days after cannulation. Salivary glands were weighed and either frozen for IgA and amylase analysis or fixed for histological analysis of acinar cells, IgA+ plasma cells, and T lymphocytes. Small intestinal wash fluid was collected for IgA regression analysis with salivary glands. RESULTS: PN reduced organ weight, acinar cell size, and amylase activity compared with chow; BBS had no significant effects on these parameters. Compared with chow, PN significantly reduced salivary gland IgA levels, IgA+ plasma cells, and T lymphocytes. PN + BBS significantly elevated IgA and restored cellularity compared with PN. Salivary gland tissue homogenate IgA levels significantly correlated with intestinal fluid IgA levels. CONCLUSIONS: Compared with chow, PN results in atrophy of the salivary glands characterized by reduced amylase, IgA, and immune cellularity. BBS has no effect on acinar cells or amylase activity compared with PN but maintains tissue IgA and plasma cells and T-lymphocyte numbers compared with chow.


Assuntos
Imunidade Adaptativa/efeitos dos fármacos , Bombesina/farmacologia , Neurotransmissores/farmacologia , Nutrição Parenteral/métodos , Glândulas Salivares/efeitos dos fármacos , Células Acinares/efeitos dos fármacos , Amilases/análise , Animais , Bombesina/administração & dosagem , Imunoglobulina A/análise , Masculino , Camundongos , Camundongos Endogâmicos ICR , Plasmócitos/efeitos dos fármacos , Glândulas Salivares/metabolismo , Linfócitos T/efeitos dos fármacos , Resultado do Tratamento
14.
Toxicol Sci ; 142(2): 361-74, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25294219

RESUMO

Oxidative stress is implicated in the development of non-alcoholic steatohepatitis (NASH). The Nrf2-antioxidant response element pathway protects cells from oxidative stress. Studies have shown that global Nrf2 deficiency hastens the progression of NASH. The purpose of this study was to determine whether long-term hepatocyte-specific activation of Nrf2 mitigates NASH progression. Transgenic mice expressing a constitutively active Nrf2 construct in hepatocytes (AlbCre+/caNrf2+) and littermate controls were generated. These mice were fed standard or methionine-choline-deficient (MCD) diet, a diet used to induce NASH development in rodents. After 28 days of MCD dietary feeding, mice developed significant increases in steatosis, inflammation, oxidative stress, and HSC activation compared with those mice on standard diet. AlbCre+/caNrf2+ animals had significantly decreased serum transaminases and reduced steatosis when compared with the AlbCre+/caNrf2- animals. This significant reduction in steatosis was associated with increased expression of genes involved in triglyceride export (MTTP) and ß-oxidation (CPT2). However, there were no differences in the increased oxidative stress, inflammation, and HSC activation from MCD diet administration between the AlbCre+/caNrf2- and AlbCre+/caNrf2+ animals. We conclude that hepatocyte-specific activation of Nrf2-mediated gene expression decreased hepatocellular damage and steatosis in a dietary model of NASH. However, hepatocyte-specific induction of Nrf2-mediated gene expression alone is insufficient to mitigate inflammation, oxidative stress, and HSC activation in this nutritional NASH model.


Assuntos
Elementos de Resposta Antioxidante/genética , Hepatócitos/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , Estresse Oxidativo , Animais , Western Blotting , Dieta , Expressão Gênica/fisiologia , Hepatócitos/patologia , Imuno-Histoquímica , Metabolismo dos Lipídeos/genética , Testes de Função Hepática , Camundongos Transgênicos , Fator 2 Relacionado a NF-E2/genética , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/patologia , Hepatopatia Gordurosa não Alcoólica/prevenção & controle , Regiões Promotoras Genéticas , Transdução de Sinais , Triglicerídeos/metabolismo
15.
PLoS One ; 9(2): e89349, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24586708

RESUMO

Intimal hyperplasia is the cause of the recurrent occlusive vascular disease (restenosis). Drugs currently used to treat restenosis effectively inhibit smooth muscle cell (SMC) proliferation, but also inhibit the growth of the protective luminal endothelial cell (EC) lining, leading to thrombosis. To identify compounds that selectively inhibit SMC versus EC proliferation, we have developed a high-throughput screening (HTS) format using human cells and have employed this to screen a multiple compound collection (NIH Clinical Collection). We developed an automated, accurate proliferation assay in 96-well plates using human aortic SMCs and ECs. Using this HTS format we screened a 447-drug NIH Clinical Library. We identified 11 compounds that inhibited SMC proliferation greater than 50%, among which idarubicin exhibited a unique feature of preferentially inhibiting SMC versus EC proliferation. Concentration-response analysis revealed this differential effect most evident over an ∼10 nM-5 µM window. In vivo testing of idarubicin in a rat carotid injury model at 14 days revealed an 80% reduction of intimal hyperplasia and a 45% increase of lumen size with no significant effect on re-endothelialization. Taken together, we have established a HTS assay of human vascular cell proliferation, and identified idarubicin as a selective inhibitor of SMC versus EC proliferation both in vitro and in vivo. Screening of larger and more diverse compound libraries may lead to the discovery of next-generation therapeutics that can inhibit intima hyperplasia without impairing re-endothelialization.


Assuntos
Proliferação de Células/efeitos dos fármacos , Idarubicina/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Túnica Íntima/efeitos dos fármacos , Animais , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Ensaios de Triagem em Larga Escala/métodos , Humanos , Hiperplasia/tratamento farmacológico , Masculino , Miócitos de Músculo Liso/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
16.
Circ Cardiovasc Interv ; 7(4): 594-601, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25074254

RESUMO

BACKGROUND: Three major processes, constrictive vessel remodeling, intimal hyperplasia (IH), and retarded re-endothelialization, contribute to restenosis after vascular reconstructions. Clinically used drugs inhibit IH but delay re-endothelialization and also cause constrictive remodeling. Here we have examined halofuginone, an herbal derivative, for its beneficial effects on vessel remodeling and differential inhibition of IH versus re-endothelialization. METHODS AND RESULTS: Two weeks after perivascular application to balloon-injured rat common carotid arteries, halofuginone versus vehicle (n=6 animals) enlarged luminal area 2.14-fold by increasing vessel size (adaptive remodeling; 123%), reducing IH (74.3%) without inhibiting re-endothelialization. Consistent with its positive effect on vessel expansion, halofuginone reduced collagen type 1 (but not type 3) production in injured arteries as well as that from adventitial fibroblasts in vitro. In support of its differential effects on IH versus re-endothelialization, halofuginone produced greater inhibition of vascular smooth muscle cell versus endothelial cell proliferation at concentrations ≈50 nmol/L. Furthermore, halofuginone at 50 nmol/L effectively blocked Smad3 phosphorylation in smooth muscle cells, which is known to promote smooth muscle cell proliferation, migration, and IH, but halofuginone had no effect on phospho-Smad3 in endothelial cells. CONCLUSIONS: Periadventitial delivery of halofuginone dramatically increased lumen patency via adaptive remodeling and selective inhibition of IH without affecting endothelium recovery. Halofuginone is the first reported small molecule that has favorable effects on all 3 major processes involved in restenosis.


Assuntos
Inibidores da Angiogênese/administração & dosagem , Angioplastia com Balão , Artérias Carótidas/efeitos dos fármacos , Lesões das Artérias Carótidas/tratamento farmacológico , Endotélio Vascular/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Hiperplasia/prevenção & controle , Miócitos de Músculo Liso/efeitos dos fármacos , Piperidinas/administração & dosagem , Complicações Pós-Operatórias/prevenção & controle , Quinazolinonas/administração & dosagem , Adaptação Biológica/efeitos dos fármacos , Inibidores da Angiogênese/efeitos adversos , Animais , Artérias Carótidas/patologia , Artérias Carótidas/cirurgia , Lesões das Artérias Carótidas/cirurgia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Colágeno Tipo I/metabolismo , Endotélio Vascular/patologia , Fibroblastos/patologia , Humanos , Hiperplasia/etiologia , Masculino , Modelos Animais , Miócitos de Músculo Liso/patologia , Especificidade de Órgãos , Piperidinas/efeitos adversos , Quinazolinonas/efeitos adversos , Ratos , Ratos Sprague-Dawley , Proteína Smad3/metabolismo , Remodelação Vascular/efeitos dos fármacos
17.
JPEN J Parenter Enteral Nutr ; 37(3): 401-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23064255

RESUMO

BACKGROUND: Lamina propria Th2 cytokines, interleukin (IL)-4 and IL-13, stimulate goblet cell (GC) proliferation and MUC2 production, which protect the intestinal mucosa. Elemental enteral nutrition (EEN) reduces tissue IL-4 and impairs barrier function. Proanthocyanidins (PACs) stimulate oral mucin levels. We hypothesized that adding PAC to EEN would maintain Th2-without stimulating Th1-cytokines and preserve luminal MUC2 vs EEN alone. MATERIALS AND METHODS: Seventy mice were randomized to 5 diet groups-standard chow, intragastric EEN, or EEN with lowPAC, midPAC (50 mg), or highPAC (100 mg PAC/kg BW)-for 5 days, starting 2 days after gastric cannulation. Ileal tissue was analyzed for histomorphology and the cytokines IL-4, IL-13, IL-1ß, IL-6, and TNF-α by enzyme-linked immunosorbent assay. MUC2 was measured in intestinal washes. RESULTS: EEN lowered IL-13 (P < .05) compared with standard chow, whereas IL-4 was not significant (P < .07). LowPAC and midPAC increased IL-13 (P < .05), whereas highPAC increased both IL-4 and IL-13 (P < .05) compared with EEN. All EEN diets reduced (P < .05) crypt depth compared with the chow group. Compared with standard chow, GC numbers and luminal MUC2 were reduced with EEN (P < .05). These effects were attenuated (P < .05) with midPAC and highPAC. No changes were observed in tissue Th1 cytokines. CONCLUSIONS: Adding PACs to EEN reverses impaired intestinal barrier function following EEN by improving the gut mucous layer and function through increased GC size and number as well as levels of MUC2 and ileal IL-4 and IL-13.


Assuntos
Alimentos Formulados/análise , Trato Gastrointestinal/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Extratos Vegetais/farmacologia , Proantocianidinas/farmacologia , Vaccinium macrocarpon/química , Animais , Proliferação de Células/efeitos dos fármacos , Nutrição Enteral , Ensaio de Imunoadsorção Enzimática , Trato Gastrointestinal/metabolismo , Células Caliciformes/efeitos dos fármacos , Células Caliciformes/metabolismo , Interleucina-13/sangue , Interleucina-1beta/sangue , Interleucina-4/sangue , Interleucina-6/sangue , Mucosa Intestinal/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos ICR , Mucina-2/sangue , Fator de Necrose Tumoral alfa/sangue
18.
J Speech Lang Hear Res ; 54(4): 1060-6, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21330647

RESUMO

PURPOSE: Epithelial homeostasis is critical for vocal fold health, yet little is known about the cells that support epithelial self-renewal. As a known characteristic of stem cells is that they are slow-cycling in vivo, the purpose of this prospective controlled study was to identify and quantify slow-cycling cells or putative stem cells in murine vocal fold epithelium. METHOD: Twelve mice were administered daily intraperitoneal injections of a nucleotide dye, bromodeoxyuridine (BrdU), over 7 consecutive days. Under this pulse-chase paradigm, slow-cycling cells retain the dye (label-retaining cells; LRCs) while more rapidly cycling cells lose dye to dilution during multiple cell divisions. The percentage of label-retaining cells (%LRCs) was calculated following a chase period of 2, 4, and 8 weeks postinjections. RESULTS: The %LRCs decreased significantly from 9.4% at 2 weeks to 3.1% at 8 weeks following injections (p < .05). No statistically significant differences in the quantity of BrdU-positive cells were measured between the anterior, mid-membranous, or cartilaginous regions of the vocal fold (p > .05). CONCLUSION: These findings are consistent with the presence and first report of a small population of putative stem cells along the length of murine vocal fold epithelium.


Assuntos
Células Epiteliais/citologia , Laringe/citologia , Nicho de Células-Tronco/fisiologia , Prega Vocal/citologia , Animais , Movimento Celular , Rastreamento de Células , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL
19.
PLoS One ; 6(6): e21701, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21738769

RESUMO

CD1 molecules are glycoproteins that present lipids and glycolipids for recognition by T cells. CD1-dependent immune activation has been implicated in a wide range of immune responses, however, our understanding of the role of this pathway in human disease remains limited because of species differences between humans and other mammals: whereas humans express five different CD1 gene products (CD1a, CD1b, CD1c, CD1d, and CD1e), muroid rodents express only one CD1 isoform (CD1d). Here we report that immune deficient mice engrafted with human fetal thymus, liver, and CD34(+) hematopoietic stem cells develop a functional human CD1 compartment. CD1a, b, c, and d isoforms were highly expressed by human thymocytes, and CD1a(+) cells with a dendritic morphology were present in the thymic medulla. CD1(+) cells were also detected in spleen, liver, and lungs. APCs from spleen and liver were capable of presenting bacterial glycolipids to human CD1-restricted T cells. ELISpot analyses of splenocytes demonstrated the presence of CD1-reactive IFN-γ producing cells. CD1d tetramer staining directly identified human iNKT cells in spleen and liver samples from engrafted mice, and injection of the glycolipid antigen α-GalCer resulted in rapid elevation of human IFN-γ and IL-4 levels in the blood indicating that the human iNKT cells are biologically active in vivo. Together, these results demonstrate that the human CD1 system is present and functionally competent in this humanized mouse model. Thus, this system provides a new opportunity to study the role of CD1-related immune activation in infections to human-specific pathogens.


Assuntos
Antígenos CD1/metabolismo , Animais , Antígenos CD1/genética , Citometria de Fluxo , Humanos , Camundongos , Camundongos SCID
20.
Transplantation ; 90(10): 1071-8, 2010 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-20885339

RESUMO

BACKGROUND: Lacto-N-fucopentaose III (LNFPIII) is a pentasaccharide containing the Lewis(x) trisaccharide that is found on schistosome eggs and in breast milk. LNFPIII conjugates suppress host immune responses and have therapeutic efficacy in mouse models of psoriasis and type 1 diabetes. METHODS: We used nonvascularized neonatal ear-heart transplantation and heterotopic vascularized heart transplantation models to evaluate immunosuppressive effects of LNFPIII and subsequently analyzed the mechanism. RESULTS: We found that administration of LNFPIII conjugates prolonged median graft survival by 80% when 1-day-old DBA/2 hearts were transplanted into ears of B6 mice. A similar graft prolongation was observed in a fully vascularized heterotopic heart transplantation model (DBA/2 into B6), No prolongation was observed with carrier protein (human serum albumin [HSA] or dextran) alone. We found increased programmed death ligand 1 (PD-L1) expression on F4/80 macrophages, CD4+ T cells, and CD11b+ CD11c+ (myeloid) dendritic cells, and increased arginase1 and Ym1 expression, typical of alternatively activated macrophages, in the draining (cervical) lymph node cells. We found accumulation of Foxp3+ regulatory T cells (Tregs) in the lymph nodes draining donor hearts, suggesting a possible role of Treg induction in graft prolongation. Anti-PD-L1 antibody treatment abrogated LNFPIII-mediated the graft survival benefit and Treg accumulation. LNFPIII-treated macrophages had increased PD-L1 expression and significantly prolonged DBA/2 allograft survival when injected intraperitoneally into B6 recipient mice. CONCLUSIONS: LNFPIII prolongs fully allogeneic graft survival in both vascularized and nonvascularized allograft transplantation models. The mechanism of graft prolongation seems to involve both alternatively activated PDL-1 macrophages and recruitment of Foxp3+ Treg cells.


Assuntos
Amino Açúcares/farmacologia , Sobrevivência de Enxerto/efeitos dos fármacos , Sobrevivência de Enxerto/imunologia , Transplante de Coração/imunologia , Imunossupressores/farmacologia , Polissacarídeos/farmacologia , Animais , Animais Recém-Nascidos , Antígeno B7-1/imunologia , Antígeno B7-H1 , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Feminino , Fatores de Transcrição Forkhead/metabolismo , Transplante de Coração/patologia , Ativação de Macrófagos , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/imunologia , Masculino , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Peptídeos/antagonistas & inibidores , Peptídeos/imunologia , Gravidez , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Fatores de Tempo , Transplante Homólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA