Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Am J Physiol Renal Physiol ; 311(6): F1182-F1188, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27413197

RESUMO

Connecting tubule glomerular feedback (CTGF) is a mechanism where an increase in sodium (Na) concentration in the connecting tubule (CNT) causes the afferent arteriole (Af-Art) to dilate. We recently reported that aldosterone within the CNT lumen enhances CTGF via a nongenomic effect involving GPR30 receptors and sodium/hydrogen exchanger (NHE), but the signaling pathways of this mechanism are unknown. We hypothesize that aldosterone enhances CTGF via cAMP/protein kinase A (PKA) pathway that activates protein kinase C (PKC) and stimulates superoxide (O2-) production. Rabbit Af-Arts and their adherent CNTs were microdissected and simultaneously perfused. Two consecutive CTGF curves were elicited by increasing the CNT luminal NaCl. We found that the main effect of aldosterone was to sensitize CTGF and we analyzed data by comparing NaCl concentration in the CNT perfusate needed to achieve half of the maximal response (EC50). During the control period, the NaCl concentration that elicited a half-maximal response (EC50) was 37.0 ± 2.0 mmol/l; addition of aldosterone (10-8 mol/l) to the CNT lumen decreased EC50 to 19.3 ± 1.3 mmol/l (P ≤ 0.001 vs. Control). The specific adenylyl cyclase inhibitor 2',3'-dideoxyadenosine (ddA; 2 × 10-4 mol/l) and the PKA inhibitor H-89 dihydrochloride hydrate (H-89; 2 × 10-6 mol/l) prevented the aldosterone effect. The selective PKC inhibitor GF109203X (10-8 mol/l) also prevented EC50 reduction caused by aldosterone. CNT intraluminal addition of O2- scavenger tempol (10-4 mol/l) blocked the aldosterone effect. We conclude that aldosterone inside the CNT lumen enhances CTGF via a cAMP/PKA/PKC pathway and stimulates O2- generation and this process may contribute to renal damage by increasing glomerular capillary pressure.


Assuntos
Aldosterona/farmacologia , Retroalimentação Fisiológica/efeitos dos fármacos , Glomérulos Renais/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Cloreto de Sódio/metabolismo , Animais , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Óxidos N-Cíclicos/farmacologia , Relação Dose-Resposta a Droga , Isoquinolinas/farmacologia , Glomérulos Renais/metabolismo , Masculino , Proteína Quinase C/metabolismo , Coelhos , Marcadores de Spin , Sulfonamidas/farmacologia
2.
Am J Physiol Renal Physiol ; 310(10): F1026-34, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-26962108

RESUMO

N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP) is a natural tetrapeptide with anti-inflammatory and antifibrotic properties. Previously, we have shown that prolyl oligopeptidase (POP) is involved in the Ac-SDKP release from thymosin-ß4 (Tß4). However, POP can only hydrolyze peptides shorter than 30 amino acids, and Tß4 is 43 amino acids long. This indicates that before POP hydrolysis takes place, Tß4 is hydrolyzed by another peptidase that releases NH2-terminal intermediate peptide(s) with fewer than 30 amino acids. Our peptidase database search pointed out meprin-α metalloprotease as a potential candidate. Therefore, we hypothesized that, prior to POP hydrolysis, Tß4 is hydrolyzed by meprin-α. In vitro, we found that the incubation of Tß4 with both meprin-α and POP released Ac-SDKP, whereas no Ac-SDKP was released when Tß4 was incubated with either meprin-α or POP alone. Incubation of Tß4 with rat kidney homogenates significantly released Ac-SDKP, which was blocked by the meprin-α inhibitor actinonin. In addition, kidneys from meprin-α knockout (KO) mice showed significantly lower basal Ac-SDKP amount, compared with wild-type mice. Kidney homogenates from meprin-α KO mice failed to release Ac-SDKP from Tß4. In vivo, we observed that rats treated with the ACE inhibitor captopril increased plasma concentrations of Ac-SDKP, which was inhibited by the coadministration of actinonin (vehicle, 3.1 ± 0.2 nmol/l; captopril, 15.1 ± 0.7 nmol/l; captopril + actinonin, 6.1 ± 0.3 nmol/l; P < 0.005). Similar results were obtained with urinary Ac-SDKP after actinonin treatment. We conclude that release of Ac-SDKP from Tß4 is mediated by successive hydrolysis involving meprin-α and POP.


Assuntos
Rim/metabolismo , Metaloendopeptidases/metabolismo , Oligopeptídeos/metabolismo , Serina Endopeptidases/metabolismo , Timosina/metabolismo , Animais , Pressão Sanguínea , Captopril , Ácidos Hidroxâmicos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Prolil Oligopeptidases , Distribuição Aleatória , Ratos Sprague-Dawley
3.
Am J Physiol Heart Circ Physiol ; 310(9): H1176-83, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-26945075

RESUMO

N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP) is a naturally occurring tetrapeptide that prevents inflammation and fibrosis in hypertension and other cardiovascular diseases. We previously showed that, in angiotensin II-induced hypertension, Ac-SDKP decreased the activation of nuclear transcription factor NF-κB, whereas, in experimental autoimmune myocarditis and hypertension animal models, it also reduced the expression of endothelial leukocyte adhesion molecule ICAM-1. However, the mechanisms by which Ac-SDKP downregulated ICAM-1 expression are still unclear. TNF-α is a proinflammatory cytokine that induces ICAM-1 expression in various cell types via TNF receptor 1 and activation of the classical NF-κB pathway. We hypothesized that in endothelial cells Ac-SDKP suppresses TNF-α-induced ICAM-1 expression by decreasing IKK phosphorylation that as a consequence leads to a decrease of IκB phosphorylation and NF-κB activation. To test this hypothesis, human coronary artery endothelial cells were treated with Ac-SDKP and then stimulated with TNF-α. We found that TNF-α-induced ICAM-1 expression was significantly decreased by Ac-SDKP in a dose-dependent manner. Ac-SDKP also decreased TNF-α-induced NF-κB translocation from cytosol to nucleus, as assessed by electrophoretic mobility shift assay, which correlated with a decrease in IκB phosphorylation. In addition, we found that Ac-SDKP decreased TNF-α-induced IKK phosphorylation and IKK-ß expression. However, Ac-SDKP had no effect on TNF-α-induced phosphorylation of p38 MAP kinase or ERK. Thus we conclude that Ac-SDKP inhibition of TNF-α activation of canonical, i.e., IKK-ß-dependent, NF-κB pathway and subsequent decrease in ICAM-1 expression is achieved via inhibition of IKK-ß.


Assuntos
Anti-Inflamatórios/farmacologia , Células Endoteliais/efeitos dos fármacos , Quinase I-kappa B/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , NF-kappa B/metabolismo , Oligopeptídeos/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Células Endoteliais/enzimologia , Humanos , Fosforilação , Transdução de Sinais , Regulação para Cima
4.
Am J Physiol Heart Circ Physiol ; 311(5): H1287-H1296, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27496875

RESUMO

Galectin-3 (Gal-3), a member of the ß-galactoside lectin family, has an important role in immune regulation. In hypertensive rats and heart failure patients, Gal-3 is considered a marker for an unfavorable prognosis. Nevertheless, the role and mechanism of Gal-3 action in hypertension-induced target organ damage are unknown. We hypothesized that, in angiotensin II (ANG II)-induced hypertension, genetic deletion of Gal-3 prevents left ventricular (LV) adverse remodeling and LV dysfunction by reducing the innate immune responses and myocardial fibrosis. To induce hypertension, male C57BL/6J and Gal-3 knockout (KO) mice were infused with ANG II (3 µg·min-1·kg-1 sc) for 8 wk. We assessed: 1) systolic blood pressure by plethysmography, 2) LV function and remodeling by echocardiography, 3) myocardial fibrosis by histology, 4) cardiac CD68+ macrophage infiltration by histology, 5) ICAM-1 and VCAM-1 expression by Western blotting, 6) plasma cytokines, including interleukin-6 (IL-6), by enzyme-linked immunosorbent assay, and 7) regulatory T (Treg) cells by flow cytometry as detected by their combined expression of CD4, CD25, and FOXP3. Systolic blood pressure and cardiac hypertrophy increased similarly in both mouse strains when infused with ANG II. However, hypertensive C57BL/6J mice suffered impaired ejection and shortening fractions. In these mice, the extent of myocardial fibrosis and macrophage infiltration was greater in histological sections, and cardiac ICAM-1, as well as plasma IL-6, expression was higher as assessed by Western blotting. However, all these parameters were blunted in Gal-3 KO mice. Hypertensive Gal-3 KO mice also had a higher number of splenic Treg lymphocytes. In conclusion, in ANG II-induced hypertension, genetic deletion of Gal-3 prevented LV dysfunction without affecting blood pressure or LV hypertrophy. This study indicates that the ANG II effects are, in part, mediated or triggered by Gal-3 together with the related intercellular signaling (ICAM-1 and IL-6), leading to cardiac inflammation and fibrosis.


Assuntos
Angiotensina II/toxicidade , Cardiomegalia/diagnóstico por imagem , Galectina 3/genética , Hipertensão/genética , Macrófagos/patologia , Miocárdio/patologia , Disfunção Ventricular Esquerda/diagnóstico por imagem , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Pressão Sanguínea , Western Blotting , Cardiomegalia/etiologia , Citocinas/metabolismo , Modelos Animais de Doenças , Ecocardiografia , Ensaio de Imunoadsorção Enzimática , Fibrose , Citometria de Fluxo , Hipertensão/induzido quimicamente , Hipertensão/complicações , Hipertensão/fisiopatologia , Molécula 1 de Adesão Intercelular/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/metabolismo , Pletismografia , Linfócitos T Reguladores , Molécula 1 de Adesão de Célula Vascular/metabolismo , Disfunção Ventricular Esquerda/etiologia , Função Ventricular Esquerda
5.
Am J Physiol Renal Physiol ; 308(9): F1026-31, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25715987

RESUMO

Afferent (Af-Art) and efferent arterioles resistance regulate glomerular capillary pressure. The nephron regulates Af-Art resistance via: 1) vasoconstrictor tubuloglomerular feedback (TGF), initiated in the macula densa via Na-K-2Cl cotransporters (NKCC2) and 2) vasodilator connecting tubuloglomerular feedback (CTGF), initiated in connecting tubules via epithelial Na channels (ENaC). Furosemide inhibits NKCC2 and TGF. Benzamil inhibits ENaC and CTGF. In vitro, CTGF dilates preconstricted Af-Arts. In vivo, benzamil decreases stop-flow pressure (PSF), suggesting that CTGF antagonizes TGF; however, even when TGF is blocked, CTGF does not increase PSF, suggesting there is another mechanism antagonizing CTGF. We hypothesize that in addition to NKCC2, activation of Na/H exchanger (NHE) antagonizes CTGF, and when both are blocked CTGF dilates Af-Arts and this effect is blocked by a CTGF inhibitor benzamil. Using micropuncture, we studied the effects of transport inhibitors on TGF responses by measuring PSF while increasing nephron perfusion from 0 to 40 nl/min. Control TGF response (-7.9 ± 0.2 mmHg) was blocked by furosemide (-0.4 ± 0.2 mmHg; P < 0.001). Benzamil restored TGF in the presence of furosemide (furosemide: -0.2 ± 0.1 vs. furosemide+benzamil: -4.3 ± 0.3 mmHg; P < 0.001). With furosemide and NHE inhibitor, dimethylamiloride (DMA), increase in tubular flow increased PSF (furosemide+DMA: 2.7 ± 0.5 mmHg, n = 6), and benzamil blocked this (furosemide+DMA+benzamil: -1.1 ± 0.2 mmHg; P < 0.01, n = 6). We conclude that NHE in the nephron decreases PSF (Af-Art constriction) when NKCC2 and ENaC are inhibited, suggesting that in the absence of NKCC2, NHE causes a TGF response and that CTGF dilates the Af-Art when TGF is blocked with NKCC2 and NHE inhibitors.


Assuntos
Amilorida/análogos & derivados , Bloqueadores do Canal de Sódio Epitelial/farmacologia , Canais Epiteliais de Sódio/efeitos dos fármacos , Furosemida/farmacologia , Glomérulos Renais/efeitos dos fármacos , Túbulos Renais/efeitos dos fármacos , Néfrons/efeitos dos fármacos , Inibidores de Simportadores de Cloreto de Sódio e Potássio/farmacologia , Trocadores de Sódio-Hidrogênio/antagonistas & inibidores , Sódio/metabolismo , Membro 1 da Família 12 de Carreador de Soluto/antagonistas & inibidores , Amilorida/farmacologia , Animais , Arteríolas/efeitos dos fármacos , Arteríolas/fisiologia , Canais Epiteliais de Sódio/metabolismo , Retroalimentação , Glomérulos Renais/irrigação sanguínea , Glomérulos Renais/metabolismo , Túbulos Renais/irrigação sanguínea , Túbulos Renais/metabolismo , Masculino , Néfrons/metabolismo , Ratos Sprague-Dawley , Circulação Renal/efeitos dos fármacos , Trocadores de Sódio-Hidrogênio/metabolismo , Membro 1 da Família 12 de Carreador de Soluto/metabolismo , Fatores de Tempo , Vasoconstrição/efeitos dos fármacos , Vasodilatação/efeitos dos fármacos
6.
Am J Physiol Renal Physiol ; 308(10): F1146-54, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25740596

RESUMO

Systemic lupus erythematosus is an autoimmune disease characterized by the development of auto antibodies against a variety of self-antigens and deposition of immune complexes that lead to inflammation, fibrosis, and end-organ damage. Up to 60% of lupus patients develop nephritis and renal dysfunction leading to kidney failure. N-acetyl-seryl-aspartyl-lysyl-proline, i.e., Ac-SDKP, is a natural tetrapeptide that in hypertension prevents inflammation and fibrosis in heart, kidney, and vasculature. In experimental autoimmune myocarditis, Ac-SDKP prevents cardiac dysfunction by decreasing innate and adaptive immunity. It has also been reported that Ac-SDKP ameliorates lupus nephritis in mice. We hypothesize that Ac-SDKP prevents lupus nephritis in mice by decreasing complement C5-9, proinflammatory cytokines, and immune cell infiltration. Lupus mice treated with Ac-SDKP for 20 wk had significantly lower renal levels of macrophage and T cell infiltration and proinflammatory chemokine/cytokines. In addition, our data demonstrate for the first time that in lupus mouse Ac-SDKP prevented the increase in complement C5-9, RANTES, MCP-5, and ICAM-1 kidney expression and it prevented the decline of glomerular filtration rate. Ac-SDKP-treated lupus mice had a significant improvement in renal function and lower levels of glomerular damage. Ac-SDKP had no effect on the production of autoantibodies. The protective Ac-SDKP effect is most likely achieved by targeting the expression of proinflammatory chemokines/cytokines, ICAM-1, and immune cell infiltration in the kidney, either directly or via C5-9 proinflammatory arm of complement system.


Assuntos
Modelos Animais de Doenças , Lúpus Eritematoso Sistêmico/complicações , Nefrite Lúpica/etiologia , Nefrite Lúpica/prevenção & controle , Oligopeptídeos/uso terapêutico , Animais , Movimento Celular , Proteínas do Sistema Complemento/metabolismo , Citocinas/metabolismo , Feminino , Taxa de Filtração Glomerular/efeitos dos fármacos , Molécula 1 de Adesão Intercelular/metabolismo , Lúpus Eritematoso Sistêmico/metabolismo , Lúpus Eritematoso Sistêmico/patologia , Nefrite Lúpica/metabolismo , Camundongos , Camundongos Endogâmicos MRL lpr , Oligopeptídeos/farmacologia , Linfócitos T/patologia
7.
Chin J Cancer ; 33(3): 148-58, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24016393

RESUMO

Tumor cell proliferation, infiltration, migration, and neovascularization are known causes of treatment resistance in glioblastoma multiforme (GBM). The purpose of this study was to determine the effect of radiation on the growth characteristics of primary human GBM developed in a nude rat. Primary GBM cells grown from explanted GBM tissues were implanted orthotopically in nude rats. Tumor growth was confirmed by magnetic resonance imaging on day 77 (baseline) after implantation. The rats underwent irradiation to a dose of 50 Gy delivered subcuratively on day 84 postimplantation (n = 8), or underwent no radiation (n = 8). Brain tissues were obtained on day 112 (nonirradiated) or day 133 (irradiated). Immunohistochemistry was performed to determine tumor cell proliferation (Ki-67) and to assess the expression of infiltration marker (matrix metalloproteinase-2, MMP-2) and cell migration marker (CD44). Tumor neovascularization was assessed by microvessel density using von-Willebrand factor (vWF) staining. Magnetic resonance imaging showed well-developed, infiltrative tumors in 11 weeks postimplantation. The proportion of Ki-67-positive cells in tumors undergoing radiation was (71 +/- 15)% compared with (25 +/- 12)% in the nonirradiated group (P = 0.02). The number of MMP-2-positive areas and proportion of CD44-positive cells were also high in tumors receiving radiation, indicating great invasion and infiltration. Microvessel density analysis did not show a significant difference between nonirradiated and irradiated tumors. Taken together, we found that subcurative radiation significantly increased proliferation, invasion, and migration of primary GBM. Our study provides insights into possible mechanisms of treatment resistance following radiation therapy for GBM.


Assuntos
Neoplasias Encefálicas/patologia , Movimento Celular/efeitos da radiação , Proliferação de Células/efeitos da radiação , Glioblastoma/patologia , Tolerância a Radiação , Animais , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/radioterapia , Linhagem Celular Tumoral , Feminino , Glioblastoma/metabolismo , Glioblastoma/radioterapia , Humanos , Receptores de Hialuronatos/metabolismo , Imuno-Histoquímica , Antígeno Ki-67/metabolismo , Imageamento por Ressonância Magnética , Metaloproteinase 2 da Matriz/metabolismo , Microvasos/patologia , Transplante de Neoplasias , Neovascularização Patológica/patologia , Radioterapia de Alta Energia , Ratos , Ratos Nus
8.
BMC Med Imaging ; 13: 17, 2013 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-23758888

RESUMO

BACKGROUND: A major challenge in the development of cell based therapies for glioma is to deliver optimal number of cells (therapeutic dose) to the tumor. Imaging tools such as magnetic resonance imaging (MRI), optical imaging, positron emission tomography (PET) and single-photon emission computed tomography (SPECT) has been used in cell tracking and/or biodistribution studies. In this study, we evaluate the dynamic biodistribution of systemic injected labeled cells [human cord blood derived endothelial progenitor cells (EPCs) and cytotoxic T-cells (CTLs)] in rat glioma model with in vivo SPECT imaging. METHODS: Human cord blood EPCs, T-cells and CD14⁺ cells (monocytes/dendritic cells) were isolated using the MidiMACS system. CD14⁺ cells were converted to dendritic cells (DC) and also primed with U251 tumor cell line lysate. T-cells were co-cultured with irradiated primed DCs at 10:1 ratio to make CTLs. Both EPCs and CTLs were labeled with In-111-oxine at 37°C in serum free DMEM media. Glioma bearing animals were randomly assigned into three groups. In-111 labeled cells or In-111 oxine alone were injected through tail vein and SPECT imaging was performed on day 0, 1, and 3. In-111 oxine activity in various organs and tumor area was determined. Histochemical analysis was performed to further confirm the migration and homing of injected cells at the tumor site. RESULTS: EPCs and CTLs showed an In-111 labeling efficiency of 87.06 ± 7.75% and 70.8 ± 12.9% respectively. Initially cell migration was observed in lung following inravenous administration of In-111 labeled cells and decreased on day 1 and 3, which indicate re-distribution of labeled cells from lung to other organs. Relatively higher In-111 oxine activity was observed in tumor areas at 24 hours in animals received In-111 labeled cells (EPCs or CTLs). Histiological analysis revealed iron positive cells in and around the tumor area in animals that received labeled cells (CTLs and EPCs). CONCLUSION: We observed differential biodistribution of In-111-oxine labeled EPCs and CTLs in different organs and intracranial glioma. This study indicates In-111 oxine based SPECT imaging is an effective tool to study the biodistribution of therapeutically important cells.


Assuntos
Glioma/diagnóstico por imagem , Glioma/cirurgia , Transplante de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/diagnóstico por imagem , Linfócitos T Citotóxicos/diagnóstico por imagem , Linfócitos T Citotóxicos/transplante , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Animais , Linhagem Celular Tumoral , Rastreamento de Células/métodos , Humanos , Ratos , Ratos Nus , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Distribuição Tecidual
9.
Cancers (Basel) ; 15(20)2023 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-37894298

RESUMO

Triple-negative breast cancer (TNBC), as one of the most aggressive forms of breast cancer, is characterized by a poor prognosis and a very low rate of disease-free and overall survival. In recent years, immunotherapeutic approaches targeting T cell checkpoint molecules, such as cytotoxic lymphocyte antigen-4 (CTLA-4), programmed death1 (PD-1) or its ligand, programmed death ligand 1 (PD-L1), have shown great potential and have been used to treat various cancers as single therapies or in combination with other modalities. However, despite this remarkable progress, patients with TNBC have shown a low response rate to this approach, commonly developing resistance to immune checkpoint blockade, leading to treatment failure. Extracellular acidosis within the tumor microenvironment (also known as the Warburg effect) is one of the factors preventing immune cells from mounting effective responses and contributing to immunotherapy treatment failure. Therefore, reducing tumor acidity is important for increasing cancer immunotherapy effectiveness and this has yet to be realized in the TNBC environment. In this study, the oral administration of sodium bicarbonate (NaHCO3) enhanced the antitumor effect of anti-PD-L1 antibody treatment, as demonstrated by generated antitumor immunity, tumor growth inhibition and enhanced survival in 4T1-Luc breast cancer model. Here, we show that NaHCO3 increased extracellular pH (pHe) in tumor tissues in vivo, an effect that was accompanied by an increase in T cell infiltration, T cell activation and IFN-γ, IL2 and IL12p40 mRNA expression in tumor tissues, as well as an increase in T cell activation in tumor-draining lymph nodes. Interestingly, these changes were further enhanced in response to combined NaHCO3 + anti-PD-L1 therapy. In addition, the acidic extracellular conditions caused a significant increase in PD-L1 expression in vitro. Taken together, these results indicate that alkalizing therapy holds potential as a new tumor microenvironment immunomodulator and we hypothesize that NaHCO3 can enhance the antitumor effects of anti-PD-L1 breast cancer therapy. The combination of these treatments may have an exceptional impact on future TNBC immunotherapeutic approaches by providing a powerful personalized medicine paradigm. Therefore, our findings have a great translational potential for improving outcomes in TNBC patients.

10.
J Pharmacol Exp Ther ; 338(2): 421-9, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21527533

RESUMO

Endothelial progenitor cells (EPCs) contribute to physiological and pathological neovascularization. Previous data have suggested that the cytochrome P450 4A/F (CYP4A/F)-20-hydroxyeicosatetraenoic acid (20-HETE) system regulates neovascularization. Therefore, we studied whether the angiogenic effects of the CYP4A/F-20-HETE system involve regulation of EPC function. We extracted human umbilical cord blood and isolated EPCs, which express AC133(+)CD34(+) and kinase insert domain receptor (KDR) surface markers and contain mRNA and protein for CYP4A11 and CYP4A22 enzymes, as opposed to mesenchymal stem cells, which only express negligible amounts of CYP4A11/22. When EPCs were incubated with arachidonic acid, they produced 20-HETE, which stimulated the cells to proliferate and migrate, as did vascular endothelial growth factor. Incubation with 1 µM N-hydroxy-N'-(4-butyl-2-methylphenyl)formamidine (HET0016), a selective inhibitor of 20-HETE synthesis, reduced the proliferative and migratory effects of vascular endothelial growth factor and also significantly abolished EPC migration mediated by stroma-derived factor-1α, as did (6,15) 20-hydroxyeicosadienoic acid. Coculturing EPCs and endothelial cells on a Matrigel matrix led to tube formation, which in turn was inhibited by both HET0016 and 20-hydroxyeicosadienoic acid. We concluded that the CYP4A/F-20-HETE system is expressed in EPCs and can act as both an autocrine and a paracrine regulatory factor.


Assuntos
Citocromo P-450 CYP4A/fisiologia , Células Endoteliais/fisiologia , Sangue Fetal/fisiologia , Células-Tronco Fetais/fisiologia , Ácidos Hidroxieicosatetraenoicos/fisiologia , Movimento Celular/fisiologia , Proliferação de Células , Células Cultivadas , Células Endoteliais/citologia , Sangue Fetal/citologia , Células-Tronco Fetais/citologia , Humanos
11.
BMC Med Imaging ; 11: 21, 2011 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-22165848

RESUMO

BACKGROUND: The current method to determine the efficacy of chemoprevention in TRAMP mouse model of carcinoma of prostate (CaP) is by extracting and weighing the prostate at different time points or by immunohistochemistry analysis. Non-invasive determination of volumes of prostate glands and seminal vesicles before, during and after treatment would be valuable in investigating the efficacy of newer chemopreventive agents in CaP. The purpose of this study was to determine whether in vivo magnetic resonance imaging (MRI) using a 3 tesla clinical MRI system can be used to follow the effect of chemoprevention in TRAMP model of mouse CaP. METHODS: Mice were randomized into control and treated groups. The animals in treated group received 10 µmol/kg of CDDO, 5 days a week for 20 weeks. Animals underwent in vivo MRI of prostate gland and seminal vesicles by a clinical 3 Tesla MRI system just before (at 5 weeks), during and at the end of treatment, at 25 weeks. T1-weighted and fat saturation (FATSAT) multiecho fast spin echo T2-weighted images (T2WI) were acquired. Volume of the prostate glands and seminal vesicles was determined from MR images. T2 signal intensity changes in the seminal vesicles were determined by subtracting higher echo time (TE) from lower TE T2WI. Following treatments all animals were sacrificed, prostate and seminal vesicles collected, and the tissues prepared for histological staining. All data were expressed as mean ± 1 standard deviation. Two-way or multivariate analysis of variance followed by post-hoc test was applied to determine the significant differences. A p-value of <0.05 was considered significant. RESULTS: Histological analysis indicated tumor in 100% of control mice, whereas 10% of the treated mice showed tumor in prostate gland. Both MRI and measured prostate weights showed higher volume/weight in control mouse group. MRI showed significantly higher volume of seminal vesicles in control animals and T2 signal intensity changes in seminal vesicles of control mice indicating higher number of tumor foci, which was also proven by histology. CONCLUSIONS: In vivo MRI is helpful in determining the efficacy of chemoprevention of prostate cancer in TRAMP mice.


Assuntos
Quimioprevenção , Imageamento por Ressonância Magnética , Ácido Oleanólico/análogos & derivados , Neoplasias da Próstata/prevenção & controle , Animais , Progressão da Doença , Masculino , Camundongos , Camundongos Transgênicos , Ácido Oleanólico/uso terapêutico , Próstata/patologia , Neoplasias da Próstata/patologia , Glândulas Seminais/patologia
12.
Cancer Biol Ther ; 22(2): 124-135, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33459132

RESUMO

Gold nanoparticles (AuNPs) have been shown to enhance cancer radiotherapy (RT) gain by localizing the absorption of radiation energy in the tumor while sparing surrounding normal tissue from radiation toxicity. Previously, we showed that AuNPs enhanced RT induced DNA damage and cytotoxicity in MCF7 breast cancer cells. Interestingly, we found that cancer cells exhibited a size-dependent AuNPs intracellular localization (4 nm preferentially in the cytoplasm and 14 nm in the nucleus). We extended those studies to an in vivo model and examined the AuNPs effects on RT cytotoxicity, survival and immunomodulation of tumor microenvironment (TME) in human triple negative breast cancer (TNBC) xenograft mouse model. We also explored the significance of nanoparticle size in these AuNPs' effects. Mice treated with RT and RT plus 4 nm or 14 nm AuNPs showed a significant tumor growth delay, compared to untreated animals, while dual RT plus AuNPs treatment exhibited additive effect compared to either RT or AuNPs treatment alone. Survival log-rank test showed significant RT enhancement with 14 nm AuNP alone; however, 4 nm AuNPs did not exhibit RT enhancement. Both sizes of AuNPs enhanced RT induced immunogenic cell death (ICD) that was coupled with significant macrophage infiltration in mice pretreated with 14 nm AuNPs. These results showing significant AuNP size-dependent RT enhancement, as evident by both tumor growth delay and overall survival, reveal additional underlying immunological mechanisms and provide a platform for studying RT multimodal approaches for TNBC that may be combined with immunotherapies, enhancing their effect.


Assuntos
Ouro/química , Imunomodulação/genética , Nanopartículas Metálicas/química , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/radioterapia , Humanos , Análise de Sobrevida , Neoplasias de Mama Triplo Negativas/mortalidade
13.
ScientificWorldJournal ; 10: 1088-99, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20563532

RESUMO

Although the cellular and molecular mechanisms of tumor growth and metastasis are not completely understood, it is established that formation and growth of new blood vessels is a conditio sine qua non for tumor survival, growth, and expansion. Numerous studies over the past decades demonstrated that neovascularization associated with tumor growth occurs via angiogenic and vasculogenic mechanisms that involve sprouting angiogenesis, intussusceptive angiogenesis, vessel co-option, vasculogenic mimicry, lymphangiogenesis, and the recruitment of endothelial progenitor cells (EPCs). Due to their ability to self-renew, circulate, home to the ischemic sites, and differentiate into mature endothelial cells, EPCs hold enormous potential to be used as a diagnostic and/or therapeutic agent in antitumor therapies. Hence, this review focuses on EPCs and their role in tumor angiogenesis with the emphasis on EPC recruitment/migration, and the potential use of EPCs as a therapeutic tool and imaging probe.


Assuntos
Endotélio/citologia , Neoplasias/irrigação sanguínea , Neovascularização Patológica , Células-Tronco/citologia , Humanos
14.
Med Phys ; 47(9): 4077-4086, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32449176

RESUMO

PURPOSE: Deep learning models have had a great success in disease classifications using large data pools of skin cancer images or lung X-rays. However, data scarcity has been the roadblock of applying deep learning models directly on prostate multiparametric MRI (mpMRI). Although model interpretation has been heavily studied for natural images for the past few years, there has been a lack of interpretation of deep learning models trained on medical images. In this paper, an efficient convolutional neural network (CNN) was developed and the model interpretation at various convolutional layers was systematically analyzed to improve the understanding of how CNN interprets multimodality medical images and the predictive powers of features at each layer. The problem of small sample size was addressed by feeding the intermediate features into a traditional classification algorithm known as weighted extreme learning machine (wELM), with imbalanced distribution among output categories taken into consideration. METHODS: The training data collection used a retrospective set of prostate MR studies, from SPIE-AAPM-NCI PROSTATEx Challenges held in 2017. Three hundred twenty biopsy samples of lesions from 201 prostate cancer patients were diagnosed and identified as clinically significant (malignant) or not significant (benign). All studies included T2-weighted (T2W), proton density-weighted (PD-W), dynamic contrast enhanced (DCE) and diffusion-weighted (DW) imaging. After registration and lesion-based normalization, a CNN with four convolutional layers were developed and trained on tenfold cross validation. The features from intermediate layers were then extracted as input to wELM to test the discriminative power of each individual layer. The best performing model from the tenfolds was chosen to be tested on the holdout cohort from two sources. Feature maps after each convolutional layer were then visualized to monitor the trend, as the layer propagated. Scatter plotting was used to visualize the transformation of data distribution. Finally, a class activation map was generated to highlight the region of interest based on the model perspective. RESULTS: Experimental trials indicated that the best input for CNN was a modality combination of T2W, apparent diffusion coefficient (ADC) and DWIb50 . The convolutional features from CNN paired with a weighted extreme learning classifier showed substantial performance compared to a CNN end-to-end training model. The feature map visualization reveals similar findings on natural images where lower layers tend to learn lower level features such as edges, intensity changes, etc, while higher layers learn more abstract and task-related concept such as the lesion region. The generated saliency map revealed that the model was able to focus on the region of interest where the lesion resided and filter out background information, including prostate boundary, rectum, etc. CONCLUSIONS: This work designs a customized workflow for the small and imbalanced dataset of prostate mpMRI where features were extracted from a deep learning model and then analyzed by a traditional machine learning classifier. In addition, this work contributes to revealing how deep learning models interpret mpMRI for prostate cancer patient stratification.


Assuntos
Imageamento por Ressonância Magnética Multiparamétrica , Imagem de Difusão por Ressonância Magnética , Humanos , Masculino , Redes Neurais de Computação , Próstata/diagnóstico por imagem , Estudos Retrospectivos
15.
BMC Biotechnol ; 9: 28, 2009 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-19327159

RESUMO

BACKGROUND: Despite enormous progress in gene therapy for breast cancer, an optimal systemic vehicle for delivering gene products to the target tissue is still lacking. The purpose of this study was to determine whether AC133+ progenitor cells (APC) can be used as both gene delivery vehicles and cellular probes for magnetic resonance imaging (MRI). In this study, we used superparamagentic iron oxide (SPIO)-labeled APCs to carry the human sodium iodide symporter (hNIS) gene to the sites of implanted breast cancer in mouse model. In vivo real time tracking of these cells was performed by MRI and expression of hNIS was determined by Tc-99m pertechnetate (Tc-99m) scan. RESULTS: Three million human breast cancer (MDA-MB-231) cells were subcutaneously implanted in the right flank of nude mice. APCs, isolated from fresh human cord blood, were genetically transformed to carry the hNIS gene using adenoviral vectors and magnetically labeled with ferumoxides-protamine sulfate (FePro) complexes. Magnetically labeled genetically transformed cells were administered intravenously in tumor bearing mice when tumors reached 0.5 cm in the largest dimension. MRI and single photon emission computed tomography (SPECT) images were acquired 3 and 7 days after cell injection, with a 7 Tesla animal MRI system and a custom built micro-SPECT using Tc-99m, respectively. Expression of hNIS in accumulated cells was determined by staining with anti-hNIS antibody. APCs were efficiently labeled with ferumoxide-protamine sulfate (FePro) complexes and transduced with hNIS gene. Our study showed not only the accumulation of intravenously administered genetically transformed, magnetically labeled APCs in the implanted breast cancer, but also the expression of hNIS gene at the tumor site. Tc-99m activity ratio (tumor/non-tumor) was significantly different between animals that received non-transduced and transduced cells (P < 0.001). CONCLUSION: This study indicates that genetically transformed, magnetically labeled APCs can be used both as delivery vehicles and cellular probes for detecting in vivo migration and homing of cells. Furthermore, they can potentially be used as a gene carrier system for the treatment of tumor or other diseases.


Assuntos
Neoplasias Mamárias Experimentais/metabolismo , Células-Tronco Pluripotentes/metabolismo , Simportadores/genética , Transdução Genética , Antígeno AC133 , Animais , Antígenos CD , Linhagem Celular Tumoral , Movimento Celular , Meios de Contraste , Dextranos , Feminino , Óxido Ferroso-Férrico , Glicoproteínas , Humanos , Ferro , Imageamento por Ressonância Magnética , Nanopartículas de Magnetita , Camundongos , Camundongos Nus , Óxidos , Peptídeos , Pertecnetato Tc 99m de Sódio , Tomografia Computadorizada de Emissão de Fóton Único
16.
FASEB J ; 22(9): 3234-46, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18556461

RESUMO

This study investigated the factors responsible for migration and homing of magnetically labeled AC133(+) cells at the sites of active angiogenesis in tumor. AC133(+) cells labeled with ferumoxide-protamine sulfate were mixed with either rat glioma or human melanoma cells and implanted in flank of nude mice. An MRI of the tumors including surrounding tissues was performed. Tumor sections were stained for Prussian blue (PB), platelet-derived growth factor (PDGF), hypoxia-inducible factor-1alpha (HIF-1alpha), stromal cell derived factor-1 (SDF-1), matrix metalloproteinase-2 (MMP-2), vascular endothelial growth factor (VEGF), and endothelial markers. Fresh snap-frozen strips from the central and peripheral parts of the tumor were collected for Western blotting. MRIs demonstrated hypointense regions at the periphery of the tumors where the PB(+)/AC133(+) cells were positive for endothelial cells markers. At the sites of PB(+)/AC133(+) cells, both HIF-1alpha and SDF-1 were strongly positive and PDGF and MMP-2 showed generalized expression in the tumor and surrounding tissues. There was no significant association of PB(+)/AC133(+) cell localization and VEGF expression in tumor cells. Western blot demonstrated strong expression of the SDF-1, MMP-2, and PDGF at the peripheral parts of the tumors. HIF-1alpha was expressed at both the periphery and central parts of the tumor. This work demonstrates that magnetically labeled cells can be used as probes for MRI and histological identification of administered cells.


Assuntos
Movimento Celular , Mobilização de Células-Tronco Hematopoéticas , Transplante de Células-Tronco Mesenquimais , Neovascularização Patológica/patologia , Antígeno AC133 , Animais , Antígenos CD/metabolismo , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Quimiocina CXCL12/biossíntese , Dextranos , Feminino , Óxido Ferroso-Férrico , Glioma/patologia , Glicoproteínas/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Ferro , Imageamento por Ressonância Magnética/métodos , Nanopartículas de Magnetita , Metaloproteinase 2 da Matriz/biossíntese , Melanoma Amelanótico/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Óxidos , Peptídeos/metabolismo , Fator de Crescimento Derivado de Plaquetas/biossíntese , Protaminas , Ratos , Fator A de Crescimento do Endotélio Vascular/biossíntese
17.
Front Oncol ; 9: 1313, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31850209

RESUMO

Purpose: The aim of this study was to identify and rank discriminant radiomics features extracted from MR multi-modal images to construct an adaptive model for characterization of Dominant Intra-prostatic Lesions (DILs) from normal prostatic gland tissues (NT). Methods and Materials: Two cohorts were retrospectively studied: Group A consisted of 98 patients and Group B 19 patients. Two image modalities were acquired using a 3.0T MR scanner: Axial T2 Weighted (T2W) and axial diffusion weighted (DW) imaging. A linear regression method was used to construct apparent diffusion coefficient (ADC) maps from DW images. DILs and the NT in the mirrored location were drawn on each modality. One hundred and sixty-eight radiomics features were extracted from DILs and NT. A Partial-Least-Squares-Correlation (PLSC) with one-way ANOVA along with bootstrapping ratio techniques were recruited to identify and rank the most discriminant latent variables. An artificial neural network (ANN) was constructed based on the optimal latent variable feature to classify the DILs and NTs. Nineteen patients were randomly chosen to test the contour variability effect on the radiomics analysis and the performance of the ANN. Finally, the trained ANN and a two dimension (2D) convolutional sampling method were combined and used to estimate DIL-NT probability map for two test cases. Results: Among 168 radiomics-based latent variables, only the first four variables of each modality in the PLSC space were found to be significantly different between the DILs and NTs. Area Under Receiver Operating Characteristic (AUROC), Positive Predictive and Negative Predictive values (PPV and NPV) for the conventional method were 94%, 0.95, and 0.92, respectively. When the feature vector was randomly permuted 10,000 times, a very strong permutation-invariant efficiency (p < 0.0001) was achieved. The radiomic-based latent variables of the NTs and DILs showed no statistically significant differences (Fstatistic < Fc = 4.11 with Confidence Level of 95% for all 8 variables) against contour variability. Dice coefficients between DIL-NT probability map and physician contours for the two test cases were 0.82 and 0.71, respectively. Conclusion: This study demonstrates the high performance of combining radiomics information extracted from multimodal MR information such as T2WI and ADC maps, and adaptive models to detect DILs in patients with PCa.

18.
Mol Vis ; 14: 2117-25, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19050766

RESUMO

PURPOSE: Lentivirus-mediated gene transfer is an important approach to modify the function of progenitor cells in ex vivo gene therapy, but may be susceptible to downregulation due to transcriptional silencing. The purpose of this study was to analyze the stability of lentivirus-mediated transgene expression in undifferentiated and differentiated retinal progenitor cells (RPCs), and to characterize the effect of lentivirus transduction on RPC differentiation in vitro. METHODS: RPCs derived from postnatal day 1 mice were expanded in defined serum-free culture medium and transduced with nonprimate lentiviral vector of feline immunodeficiency virus (FIV) expressing yellow fluorescent protein (YFP) reporter. Long-term expression of YFP in undifferentiated and differentiated RPCs was analyzed. Expression of various markers for RPCs and differentiated cells was analyzed by immunochemical staining in lentivirus-transduced and control RPCs. Differentiated postmitotic cells were revealed by negative labeling of bromodeoxyuridine (BrdU). RESULTS: FIV transduction induced long-term expression of YFP reporter in RPCs for up to 53 days (10 passages) with no sign of decrease in expression level. FIV transduction did not alter the expression profile of various markers in retinal spheres, including nestin, microtubule-associated protein 2 (MAP-2), glial fibrillary acidic protein (GFAP), and opsin. However, YFP expression was downregulated in differentiated BrdU-negative postmitotic cells. CONCLUSIONS: FIV-mediated long-term expression of transgene in undifferentiated RPCs is downregulated upon their differentiation. Thus, lentivirus-mediated ex vivo modulation should be cautiously analyzed for transgene expression not only in undifferentiated RPCs, but also in differentiated postmitotic cells.


Assuntos
Expressão Gênica , Vírus da Imunodeficiência Felina/genética , Retina/citologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Transgenes , Animais , Animais Recém-Nascidos , Gatos , Diferenciação Celular/fisiologia , Regulação para Baixo , Técnicas de Transferência de Genes , Vetores Genéticos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mitose , Fatores de Tempo
19.
Biotechniques ; 43(5): 627-8, 630, 632 passim, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18072592

RESUMO

Cell labeling with superparamagnetic iron oxides (SPIO) is becoming a routine procedure in cellular magnetic resonance imaging (MRI). Quantifying the intracellular iron in labeled cells is a prerequisite for determining the number of accumulated cells by quantitative MRI studies. To establish the most sensitive and reproducible method for measuring iron concentration in magnetically labeled cells, we investigated and compared four different methods using an ultraviolet-visible (UV/VIS) spectrophotometer. Background spectra were obtained for 5 and 10 M hydrochloric acids, a mixture of 100 mM citric acid plus ascorbic acid and bathophenanthroline sulphonate (BPS), and a mixture of 5 M hydrochloric acid plus 5% ferrocyanide. Spectra of the same solutions containing either 10 or 5 microg/mL iron oxides were also created to determine the peak absorbance wavelengths for the dissolved iron. In addition, different known iron concentrations were used to obtain calibration lines for each method. Based on the calibration factors, iron was measured in samples with a known amount of iron and in labeled cells. Methods based on the use of 10 M hydrochloric acid underestimated iron concentration in all experiments; for this method to give an accurate measurement, iron concentration in sample needs to be at least 3 microg/mL.


Assuntos
Ferro/análise , Magnetismo , Espectrofotometria Ultravioleta/métodos , Coloração e Rotulagem/métodos , Ácido Ascórbico/análise , Calibragem , Linhagem Celular Tumoral , Ácido Cítrico/análise , Ferrocianetos/análise , Humanos , Ácido Clorídrico/análise , Ácidos Sulfônicos/análise
20.
Physiol Rep ; 5(2)2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28126732

RESUMO

Systemic lupus erythematosus (SLE) is an autoimmune disease with a high prevalence of hypertension. NZBWF1 (SLE-Hyp) mice develop hypertension that can be prevented by modulating T cells. The peptide N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP) decreases renal damage and improves renal function in a model of SLE without hypertension (MRL/lpr). However, it is not known whether Ac-SDKP prevents hypertension in NZBWF1 mice. We hypothesized that in SLE-Hyp, Ac-SDKP prevents hypertension and renal damage by modulating T cells. Animals were divided into four groups: (1) control + vehicle, (2) control + Ac-SDKP, (3) SLE + vehicle, and (4) SLE + Ac-SDKP Systolic blood pressure (SBP), albuminuria, renal fibrosis, and T-cell phenotype were analyzed. SBP was higher in SLE compared to control mice and was not decreased by Ac-SDKP treatment. Half of SLE mice developed an acute and severe form of hypertension accompanied by albuminuria followed by death. Ac-SDKP delayed development of severe hypertension, albuminuria, and early mortality, but this delay did not reach statistical significance. Ac-SDKP prevented glomerulosclerosis, but not interstitial fibrosis in SLE-Hyp mice. SLE-Hyp mice showed a decrease in helper and cytotoxic T cells as well as an increase in double negative lymphocytes and T helper 17 cells, but these cells were unaffected by Ac-SDKP In conclusion, Ac-SDKP prevents kidney damage, without affecting blood pressure in an SLE animal model. However, during the acute relapse of SLE, Ac-SDKP might also delay the manifestation of an acute and severe form of hypertension leading to early mortality. Ac-SDKP is a potential tool to treat renal damage in SLE-Hyp mice.


Assuntos
Hipertensão/imunologia , Nefropatias/imunologia , Lúpus Eritematoso Sistêmico/fisiopatologia , Oligopeptídeos/administração & dosagem , Albuminúria/prevenção & controle , Animais , Pressão Sanguínea/efeitos dos fármacos , Feminino , Fibrose/prevenção & controle , Hipertensão/complicações , Hipertensão/prevenção & controle , Rim/efeitos dos fármacos , Rim/patologia , Nefropatias/complicações , Nefropatias/patologia , Nefropatias/prevenção & controle , Lúpus Eritematoso Sistêmico/complicações , Lúpus Eritematoso Sistêmico/mortalidade , Camundongos , Oligopeptídeos/uso terapêutico , Análise de Sobrevida , Linfócitos T/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA