Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Immunity ; 38(2): 296-308, 2013 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-23333075

RESUMO

Macrophages frequently infiltrate tumors and can enhance cancer growth, yet the origins of the macrophage response are not well understood. Here we address molecular mechanisms of macrophage production in a conditional mouse model of lung adenocarcinoma. We report that overproduction of the peptide hormone Angiotensin II (AngII) in tumor-bearing mice amplifies self-renewing hematopoietic stem cells (HSCs) and macrophage progenitors. The process occurred in the spleen but not the bone marrow, and was independent of hemodynamic changes. The effects of AngII required direct hormone ligation on HSCs, depended on S1P(1) signaling, and allowed the extramedullary tissue to supply new tumor-associated macrophages throughout cancer progression. Conversely, blocking AngII production prevented cancer-induced HSC and macrophage progenitor amplification and thus restrained the macrophage response at its source. These findings indicate that AngII acts upstream of a potent macrophage amplification program and that tumors can remotely exploit the hormone's pathway to stimulate cancer-promoting immunity.


Assuntos
Adenocarcinoma/metabolismo , Angiotensina II/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Neoplasias Pulmonares/metabolismo , Macrófagos/metabolismo , Baço/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/patologia , Adenocarcinoma de Pulmão , Angiotensina II/metabolismo , Animais , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Comunicação Celular , Movimento Celular , Proliferação de Células , Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Lisofosfolipídeos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Transgênicos , Transdução de Sinais , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Baço/patologia , Carga Tumoral
2.
J Proteome Res ; 19(1): 129-143, 2020 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-31661273

RESUMO

Roux-en-Y gastric bypass (RYGB) surgery reduces weight in obese patients. A marked decrease in blood glucose levels occurs before weight loss; however, key molecules that improve the glycemic profile remain largely unknown. Using a murine RYGB surgery model, we performed multiorgan proteomics and bioinformatics to monitor the proteins and molecular pathways that change in this early glycemic response. Multiplexed proteomic kinetics data analysis revealed that the Roux limb, biliopancreatic limb, liver, and pancreas each exhibited unique temporal and molecular responses to the RYGB surgery. In addition, protein-protein network analysis indicated that the changes to the microbial environment in the intestine may play a crucial role in the beneficial effects of RYGB surgery. Furthermore, insulin-like growth factor binding protein 7 (Igfbp7) was identified as an early induced protein in the Roux limb. Known secretory properties of Igfbp7 prompted us to further investigate its role as a remote organ regulator of glucose metabolism. Igfbp7 overexpression decreased blood glucose levels in diet-induced obese mice and attenuated gluconeogenic gene expression in the liver. Secreted Igfbp7 appeared to mediate these beneficial effects. These results demonstrate that organs responded differentially to RYGB surgery and indicate that Igfbp7 may play an important role in improving blood glucose levels.


Assuntos
Derivação Gástrica , Resistência à Insulina , Animais , Glicemia , Gluconeogênese , Humanos , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/genética , Intestinos , Camundongos , Proteômica
3.
World J Surg Oncol ; 16(1): 221, 2018 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-30419915

RESUMO

BACKGROUND: Liposarcoma of the gallbladder is an extremely rare sarcoma, with only five cases reported in the literature according to our knowledge. CASE PRESENTATION: A 71-year-old woman was referred to the Surgical Oncology Division of Napoleão Laureano Hospital (João Pessoa, PB, Brazil) due to a solid mass at the right side of the abdomen and fever, with no signs of jaundice. Abdominal ultrasonography and computed tomography (CT) evidenced an extensive gallbladder lobular formation adhered to the inferior border of the right hepatic lobe and cholelithiasis. The CT report suggested gallbladder liposarcoma. A cholecystectomy associated with resection of segments IV-B and V of the liver were performed. Intraoperative frozen sections were compatible with gallbladder sarcoma. Anatomopathological examination and immunohistochemistry confirmed dedifferentiated liposarcoma with foci of heterologous leiomyosarcomatous differentiation and undifferentiated fusocellular areas of high histological grade. CONCLUSION: This is the first case of dedifferentiated liposarcoma of the gallbladder to be reported.


Assuntos
Colecistectomia/métodos , Neoplasias da Vesícula Biliar/cirurgia , Vesícula Biliar/patologia , Lipossarcoma/cirurgia , Idoso , Brasil , Feminino , Vesícula Biliar/diagnóstico por imagem , Vesícula Biliar/cirurgia , Neoplasias da Vesícula Biliar/diagnóstico , Neoplasias da Vesícula Biliar/patologia , Humanos , Lipossarcoma/diagnóstico , Lipossarcoma/patologia , Tomografia Computadorizada por Raios X , Ultrassonografia
4.
Am J Pathol ; 185(4): 1156-66, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25680278

RESUMO

Chronic renal disease (CRD) accelerates the development of atherosclerosis. The potent protease cathepsin S cleaves elastin and generates bioactive elastin peptides, thus promoting vascular inflammation and calcification. We hypothesized that selective cathepsin S inhibition attenuates atherogenesis in hypercholesterolemic mice with CRD. CRD was induced by 5/6 nephrectomy in high-fat high-cholesterol fed apolipoprotein E-deficient mice. CRD mice received a diet admixed with 6.6 or 60 mg/kg of the potent and selective cathepsin S inhibitor RO5444101 or a control diet. CRD mice had significantly higher plasma levels of osteopontin, osteocalcin, and osteoprotegerin (204%, 148%, and 55%, respectively; P < 0.05), which were inhibited by RO5444101 (60%, 40%, and 36%, respectively; P < 0.05). Near-infrared fluorescence molecular imaging revealed a significant reduction in cathepsin activity in treated mice. RO5444101 decreased osteogenic activity. Histologic assessment in atherosclerotic plaque demonstrated that RO5444101 reduced immunoreactive cathepsin S (P < 0.05), elastin degradation (P = 0.01), plaque size (P = 0.01), macrophage accumulation (P < 0.01), growth differentiation factor-15 (P = 0.0001), and calcification (alkaline phosphatase activity, P < 0.01; osteocalcin, P < 0.05). Furthermore, cathepsin S inhibitor or siRNA significantly decreased expression of growth differentiation factor-15 and monocyte chemotactic protein-1 in a murine macrophage cell line and human primary macrophages. Systemic inhibition of cathepsin S attenuates the progression of atherosclerotic lesions in 5/6 nephrectomized mice, serving as a potential treatment for atherosclerosis in patients with CRD.


Assuntos
Apolipoproteínas E/deficiência , Aterosclerose/patologia , Catepsinas/antagonistas & inibidores , Falência Renal Crônica/enzimologia , Falência Renal Crônica/patologia , Animais , Artérias/enzimologia , Artérias/patologia , Aterosclerose/complicações , Biomarcadores/sangue , Catepsinas/metabolismo , Quimiocina CCL2/metabolismo , Fator 15 de Diferenciação de Crescimento/metabolismo , Humanos , Interferon gama/farmacologia , Falência Renal Crônica/sangue , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Osteogênese/efeitos dos fármacos , Inibidores de Proteases/química , Inibidores de Proteases/farmacologia , Calcificação Vascular/complicações , Calcificação Vascular/patologia
5.
Arterioscler Thromb Vasc Biol ; 35(11): 2343-2353, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26404485

RESUMO

OBJECTIVE: Despite its large clinical impact, the underlying mechanisms for vein graft failure remain obscure and no effective therapeutic solutions are available. We tested the hypothesis that Notch signaling promotes vein graft disease. APPROACH AND RESULTS: We used 2 biotherapeutics for Delta-like ligand 4 (Dll4), a Notch ligand: (1) blocking antibody and (2) macrophage- or endothelial cell (EC)-targeted small-interfering RNA. Dll4 antibody administration for 28 days inhibited vein graft lesion development in low-density lipoprotein (LDL) receptor-deficient (Ldlr(-/-)) mice, and suppressed macrophage accumulation and macrophage expression of proinflammatory M1 genes. Dll4 antibody treatment for 7 days after grafting also reduced macrophage burden at day 28. Dll4 silencing via macrophage-targeted lipid nanoparticles reduced lesion development and macrophage accumulation, whereas EC-targeted Dll4 small-interfering RNA produced no effects. Gain-of-function and loss-of-function studies suggested in vitro that Dll4 induces proinflammatory molecules in macrophages. Macrophage Dll4 also stimulated smooth muscle cell proliferation and migration and suppressed their differentiation. CONCLUSIONS: These results suggest that macrophage Dll4 promotes lesion development in vein grafts via macrophage activation and crosstalk between macrophages and smooth muscle cells, supporting the Dll4-Notch axis as a novel therapeutic target.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Macrófagos/metabolismo , Proteínas de Membrana/metabolismo , Neointima , Veia Safena/transplante , Veia Cava Inferior/transplante , Proteínas Adaptadoras de Transdução de Sinal , Animais , Anticorpos/farmacologia , Proteínas de Ligação ao Cálcio , Artérias Carótidas/cirurgia , Comunicação Celular , Diferenciação Celular , Movimento Celular , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Macrófagos/imunologia , Masculino , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Interferência de RNA , Receptores de LDL/deficiência , Receptores de LDL/genética , Veia Safena/metabolismo , Veia Safena/patologia , Transdução de Sinais , Fatores de Tempo , Transfecção , Remodelação Vascular , Veia Cava Inferior/imunologia , Veia Cava Inferior/metabolismo , Veia Cava Inferior/patologia
6.
Brain ; 138(Pt 6): 1710-21, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25910782

RESUMO

Characterizing clinically relevant brain metastasis models and assessing the therapeutic efficacy in such models are fundamental for the development of novel therapies for metastatic brain cancers. In this study, we have developed an in vivo imageable breast-to-brain metastasis mouse model. Using real time in vivo imaging and subsequent composite fluorescence imaging, we show a widespread distribution of micro- and macro-metastasis in different stages of metastatic progression. We also show extravasation of tumour cells and the close association of tumour cells with blood vessels in the brain thus mimicking the multi-foci metastases observed in the clinics. Next, we explored the ability of engineered adult stem cells to track metastatic deposits in this model and show that engineered stem cells either implanted or injected via circulation efficiently home to metastatic tumour deposits in the brain. Based on the recent findings that metastatic tumour cells adopt unique mechanisms of evading apoptosis to successfully colonize in the brain, we reasoned that TNF receptor superfamily member 10A/10B apoptosis-inducing ligand (TRAIL) based pro-apoptotic therapies that induce death receptor signalling within the metastatic tumour cells might be a favourable therapeutic approach. We engineered stem cells to express a tumour selective, potent and secretable variant of a TRAIL, S-TRAIL, and show that these cells significantly suppressed metastatic tumour growth and prolonged the survival of mice bearing metastatic breast tumours. Furthermore, the incorporation of pro-drug converting enzyme, herpes simplex virus thymidine kinase, into therapeutic S-TRAIL secreting stem cells allowed their eradication post-tumour treatment. These studies are the first of their kind that provide insight into targeting brain metastasis with stem-cell mediated delivery of pro-apoptotic ligands and have important clinical implications.


Assuntos
Neoplasias Encefálicas/secundário , Neoplasias Encefálicas/terapia , Neoplasias da Mama/patologia , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/transplante , Transplante de Células-Tronco/métodos , Ligante Indutor de Apoptose Relacionado a TNF/uso terapêutico , Animais , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Sobrevivência Celular , Feminino , Vetores Genéticos/genética , Humanos , Masculino , Camundongos , Simplexvirus/enzimologia , Simplexvirus/genética , Ligante Indutor de Apoptose Relacionado a TNF/genética , Timidina Quinase/genética , Timidina Quinase/metabolismo , Timidina Quinase/uso terapêutico
7.
Proc Natl Acad Sci U S A ; 109(7): 2491-6, 2012 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-22308361

RESUMO

Tumor-associated macrophages (TAMs) and tumor-associated neutrophils (TANs) can control cancer growth and exist in almost all solid neoplasms. The cells are known to descend from immature monocytic and granulocytic cells, respectively, which are produced in the bone marrow. However, the spleen is also a recently identified reservoir of monocytes, which can play a significant role in the inflammatory response that follows acute injury. Here, we evaluated the role of the splenic reservoir in a genetic mouse model of lung adenocarcinoma driven by activation of oncogenic Kras and inactivation of p53. We found that high numbers of TAM and TAN precursors physically relocated from the spleen to the tumor stroma, and that recruitment of tumor-promoting spleen-derived TAMs required signaling of the chemokine receptor CCR2. Also, removal of the spleen, either before or after tumor initiation, reduced TAM and TAN responses significantly and delayed tumor growth. The mechanism by which the spleen was able to maintain its reservoir capacity throughout tumor progression involved, in part, local accumulation in the splenic red pulp of typically rare extramedullary hematopoietic stem and progenitor cells, notably granulocyte and macrophage progenitors, which produced CD11b(+) Ly-6C(hi) monocytic and CD11b(+) Ly-6G(hi) granulocytic cells locally. Splenic granulocyte and macrophage progenitors and their descendants were likewise identified in clinical specimens. The present study sheds light on the origins of TAMs and TANs, and positions the spleen as an important extramedullary site, which can continuously supply growing tumors with these cells.


Assuntos
Macrófagos/imunologia , Neoplasias/patologia , Neutrófilos/imunologia , Animais , Humanos , Camundongos , Neoplasias/imunologia , Baço/imunologia , Baço/patologia
8.
Proc Natl Acad Sci U S A ; 109(52): 21444-9, 2012 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-23236189

RESUMO

The treatment of diseased vasculature remains challenging, in part because of the difficulty in implanting drug-eluting devices without subjecting vessels to damaging mechanical forces. Implanting materials using adhesive forces could overcome this challenge, but materials have previously not been shown to durably adhere to intact endothelium under blood flow. Marine mussels secrete strong underwater adhesives that have been mimicked in synthetic systems. Here we develop a drug-eluting bioadhesive gel that can be locally and durably glued onto the inside surface of blood vessels. In a mouse model of atherosclerosis, inflamed plaques treated with steroid-eluting adhesive gels had reduced macrophage content and developed protective fibrous caps covering the plaque core. Treatment also lowered plasma cytokine levels and biomarkers of inflammation in the plaque. The drug-eluting devices developed here provide a general strategy for implanting therapeutics in the vasculature using adhesive forces and could potentially be used to stabilize rupture-prone plaques.


Assuntos
Adesivos/química , Vasos Sanguíneos/patologia , Dexametasona/uso terapêutico , Placa Aterosclerótica/tratamento farmacológico , Placa Aterosclerótica/patologia , Adesividade/efeitos dos fármacos , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/metabolismo , Artérias/efeitos dos fármacos , Artérias/patologia , Vasos Sanguíneos/efeitos dos fármacos , Catecóis/química , Dexametasona/farmacologia , Sistemas de Liberação de Medicamentos , Feminino , Géis/química , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Implantes Experimentais , Inflamação/patologia , Camundongos , Camundongos Endogâmicos C57BL , Solubilidade , Estresse Mecânico , Estresse Fisiológico/efeitos dos fármacos
9.
Eur Heart J ; 34(8): 615-24, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22927557

RESUMO

AIMS: Activation of vascular endothelial cells (ECs) contributes importantly to inflammation and atherogenesis. We previously reported that apolipoprotein CIII (apoCIII), found abundantly on circulating triglyceride-rich lipoproteins, enhances adhesion of human monocytes to ECs in vitro. Statins may exert lipid-independent anti-inflammatory effects. The present study examined whether statins suppress apoCIII-induced EC activation in vitro and in vivo. METHODS AND RESULTS: Physiologically relevant concentrations of purified human apoCIII enhanced attachment of the monocyte-like cell line THP-1 to human saphenous vein ECs (HSVECs) or human coronary artery ECs (HCAECs) under both static and laminar shear stress conditions. This process mainly depends on vascular cell adhesion molecule-1 (VCAM-1), as a blocking VCAM-1 antibody abolished apoCIII-induced monocyte adhesion. ApoCIII significantly increased VCAM-1 expression in HSVECs and HCAECs. Pre-treatment with statins suppressed apoCIII-induced VCAM-1 expression and monocyte adhesion, with two lipophilic statins (pitavastatin and atorvastatin) exhibiting inhibitory effects at lower concentration than those of hydrophilic pravastatin. Nuclear factor κB (NF-κB) mediated apoCIII-induced VCAM-1 expression, as demonstrated via loss-of-function experiments, and pitavastatin treatment suppressed NF-κB activation. Furthermore, in the aorta of hypercholesterolaemic Ldlr(-/-) mice, pitavastatin administration in vivo suppressed VCAM-1 mRNA and protein, induced by apoCIII bolus injection. Similarly, in a subcutaneous dorsal air pouch mouse model of leucocyte recruitment, apoCIII injection induced F4/80+ monocyte and macrophage accumulation, whereas pitavastatin administration reduced this effect. CONCLUSIONS: These findings further establish the direct role of apoCIII in atherogenesis and suggest that anti-inflammatory effects of statins could improve vascular disease in the population with elevated plasma apoCIII.


Assuntos
Apolipoproteína C-III/antagonistas & inibidores , Células Endoteliais/fisiologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Leucócitos Mononucleares/efeitos dos fármacos , Quinolinas/farmacologia , Animais , Aorta , Adesão Celular/fisiologia , Células Cultivadas , Humanos , Leucócitos Mononucleares/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Veia Safena , Molécula 1 de Adesão de Célula Vascular/metabolismo
10.
Circulation ; 125(2): 364-74, 2012 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-22144566

RESUMO

BACKGROUND: Atherosclerotic lesions are believed to grow via the recruitment of bone marrow-derived monocytes. Among the known murine monocyte subsets, Ly-6C(high) monocytes are inflammatory, accumulate in lesions preferentially, and differentiate. Here, we hypothesized that the bone marrow outsources the production of Ly-6C(high) monocytes during atherosclerosis. METHODS AND RESULTS: Using murine models of atherosclerosis and fate-mapping approaches, we show that hematopoietic stem and progenitor cells progressively relocate from the bone marrow to the splenic red pulp, where they encounter granulocyte macrophage colony-stimulating factor and interleukin-3, clonally expand, and differentiate to Ly-6C(high) monocytes. Monocytes born in such extramedullary niches intravasate, circulate, and accumulate abundantly in atheromata. On lesional infiltration, Ly-6C(high) monocytes secrete inflammatory cytokines, reactive oxygen species, and proteases. Eventually, they ingest lipids and become foam cells. CONCLUSIONS: Our findings indicate that extramedullary sites supplement the hematopoietic function of the bone marrow by producing circulating inflammatory cells that infiltrate atherosclerotic lesions.


Assuntos
Antígenos Ly , Aterosclerose/patologia , Movimento Celular/imunologia , Hematopoese Extramedular/imunologia , Monócitos/patologia , Animais , Antígenos Ly/biossíntese , Aterosclerose/imunologia , Medula Óssea , Diferenciação Celular , Fator Estimulador de Colônias de Granulócitos e Macrófagos , Células-Tronco Hematopoéticas/patologia , Inflamação , Interleucina-3 , Camundongos , Monócitos/imunologia
11.
J Neurooncol ; 111(2): 153-61, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23242736

RESUMO

The lack of relevant pre-clinical animal models incorporating the clinical scenario of Glioblastoma multiforme (GBM) resection and recurrence has contributed significantly to the inability to successfully treat GBM. A multi-modality imaging approach that allows real-time assessment of tumor resection during surgery and non-invasive detection of post-operative tumor volumes is urgently needed. In this study, we report the development and implementation of an optical imaging and magnetic resonance imaging (MRI) approach to guide GBM resection during surgery and track tumor recurrence at multiple resolutions in mice. Intra-operative fluorescence-guided surgery allowed real-time monitoring of intracranial tumor removal and led to greater than 90 % removal of established intracranial human GBM. The fluorescent signal clearly delineated tumor margins, residual tumor, and correlated closely with the clinically utilized fluorescence surgical marker 5-aminolevulinic acid/porphyrin. Post-operative non-invasive optical imaging and MRI confirmed near-complete tumor removal, which was further validated by immunohistochemistry (IHC). Longitudinal non-invasive imaging and IHC showed rapid recurrence of multi-focal tumors that exhibited a faster growth rate and altered blood-vessel density compared to non-resected tumors. Surgical tumor resection significantly extended long-term survival, however mice ultimately succumbed to the recurrent GBM. This multi-modality imaging approach to GBM resection and recurrence in mice should provide an important platform for investigating multiple aspects of GBM and ultimately evaluating novel therapeutics.


Assuntos
Neoplasias Encefálicas/cirurgia , Glioblastoma/cirurgia , Análise de Variância , Animais , Linhagem Celular Tumoral , Imagem de Difusão por Ressonância Magnética , Modelos Animais de Doenças , Proteínas de Fluorescência Verde/genética , Humanos , Antígeno Ki-67/metabolismo , Medições Luminescentes , Camundongos , Camundongos Nus , Recidiva Local de Neoplasia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Fatores de Tempo , Transfecção , Carga Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Circ Res ; 107(11): 1364-73, 2010 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-20930148

RESUMO

RATIONALE: Monocytes recruited to ischemic myocardium originate from a reservoir in the spleen, and the release from their splenic niche relies on angiotensin (Ang) II signaling. OBJECTIVE: Because monocytes are centrally involved in tissue repair after ischemia, we hypothesized that early angiotensin-converting enzyme (ACE) inhibitor therapy impacts healing after myocardial infarction partly via effects on monocyte traffic. METHODS AND RESULTS: In a mouse model of permanent coronary ligation, enalapril arrested the release of monocytes from the splenic reservoir and consequently reduced their recruitment into the healing infarct by 45%, as quantified by flow cytometry of digested infarcts. Time-lapse intravital microscopy revealed that enalapril reduces monocyte motility in the spleen. In vitro migration assays and Western blotting showed that this was caused by reduced signaling through the Ang II type 1 receptor. We then studied the long-term consequences of blocked splenic monocyte release in atherosclerotic apolipoprotein (apo)E(-/-) mice, in which infarct healing is impaired because of excessive inflammation in the cardiac wound. Enalapril improved histologic healing biomarkers and reduced inflammation in infarcts measured by FMT-CT (fluorescence molecular tomography in conjunction with x-ray computed tomography) of proteolytic activity. ACE inhibition improved MRI-derived ejection fraction by 14% on day 21, despite initially comparable infarct size. In apoE(-/-) mice, ischemia/reperfusion injury resulted in larger infarct size and enhanced monocyte recruitment and was reversible by enalapril treatment. Splenectomy reproduced antiinflammatory effects of enalapril. CONCLUSION: This study suggests that benefits of early ACE inhibition after myocardial infarction can partially be attributed to its potent antiinflammatory impact on the splenic monocyte reservoir.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/farmacologia , Movimento Celular/efeitos dos fármacos , Monócitos/enzimologia , Monócitos/patologia , Infarto do Miocárdio/enzimologia , Infarto do Miocárdio/patologia , Baço/enzimologia , Baço/patologia , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Animais , Movimento Celular/fisiologia , Enalapril/farmacologia , Enalapril/uso terapêutico , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Monócitos/efeitos dos fármacos , Infarto do Miocárdio/tratamento farmacológico , Baço/efeitos dos fármacos
13.
Proc Natl Acad Sci U S A ; 106(30): 12459-64, 2009 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-19620715

RESUMO

There is a growing need for fast, highly sensitive and quantitative technologies to detect and profile unaltered cells in biological samples. Technologies in current clinical use are often time consuming, expensive, or require considerable sample sizes. Here, we report a diagnostic magnetic resonance (DMR) sensor that combines a miniaturized NMR probe with targeted magnetic nanoparticles for detection and molecular profiling of cancer cells. The sensor measures the transverse relaxation rate of water molecules in biological samples in which target cells of interest are labeled with magnetic nanoparticles. We achieved remarkable sensitivity improvements over our prior DMR prototypes by synthesizing new nanoparticles with higher transverse relaxivity and by optimizing assay protocols. We detected as few as 2 cancer cells in 1-microL sample volumes of unprocessed fine-needle aspirates of tumors and profiled the expression of several cellular markers in <15 min.


Assuntos
Biópsia por Agulha Fina , Espectroscopia de Ressonância Magnética/métodos , Neoplasias/patologia , Animais , Técnicas Biossensoriais , Western Blotting , Contagem de Células , Linhagem Celular Tumoral , Citodiagnóstico/instrumentação , Citodiagnóstico/métodos , Feminino , Citometria de Fluxo , Humanos , Leucócitos/citologia , Leucócitos/metabolismo , Espectroscopia de Ressonância Magnética/instrumentação , Magnetismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Técnicas Analíticas Microfluídicas , Microscopia Eletrônica de Transmissão , Nanopartículas/química , Nanopartículas/ultraestrutura , Neoplasias/metabolismo , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Receptor ErbB-2/metabolismo , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
14.
Proc Natl Acad Sci U S A ; 106(12): 4822-7, 2009 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-19264968

RESUMO

The poor prognosis of patients with aggressive and invasive cancers combined with toxic effects and short half-life of currently available treatments necessitate development of more effective tumor selective therapies. Mesenchymal stem cells (MSCs) are emerging as novel cell-based delivery agents; however, a thorough investigation addressing their therapeutic potential and fate in different cancer models is lacking. In this study, we explored the engineering potential, fate, and therapeutic efficacy of human MSCs in a highly malignant and invasive model of glioblastoma. We show that engineered MSC retain their "stem-like" properties, survive longer in mice with gliomas than in the normal brain, and migrate extensively toward gliomas. We also show that MSCs are resistant to the cytokine tumor necrosis factor apoptosis ligand (TRAIL) and, when engineered to express secreted recombinant TRAIL, induce caspase-mediated apoptosis in established glioma cell lines as well as CD133-positive primary glioma cells in vitro. Using highly malignant and invasive human glioma models and employing real-time imaging with correlative neuropathology, we demonstrate that MSC-delivered recombinant TRAIL has profound anti-tumor effects in vivo. This study demonstrates the efficacy of diagnostic and therapeutic MSC in preclinical glioma models and forms the basis for developing stem cell-based therapies for different cancers.


Assuntos
Engenharia Genética , Glioma/terapia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Animais , Efeito Espectador/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Glioma/patologia , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Transdução Genética , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
15.
PLoS One ; 17(5): e0268836, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35617240

RESUMO

BACKGROUND: Recent experimental studies have suggested a potential link between cathepsin S (CTTS) and gastric adenocarcinoma progression. Herein, we aimed to evaluate the expression of CTTS in gastric adenocarcinoma in patients who underwent curative-intent surgical resection. METHODS: This was a cross-sectional study that included two groups: gastric adenocarcinoma (n = 42) and gastritis (n = 50). The gastritis group was then subdivided into H. pylori-positive (n = 25) and H. pylori-negative (n = 25) groups. Gastric tissue samples were analysed to determine CTTS expression through immunohistochemistry. Samples were obtained by oesophagogastroduodenoscopy or surgical specimens. RESULTS: In patients with gastritis, the age ranged from 18 to 78 years. Among them, 34% were male, and 66% were female. In patients with gastric adenocarcinoma, the age ranged from 37 to 85 years. Among them, 50% were male. When comparing the expression of CTTS between the two groups, only 16% of the gastritis samples had an expression higher than 25%. Alternatively, among patients with gastric adenocarcinoma, 19% had expression between 25-50%, 14.3% between 51-75%, and 26.2% had expression higher than 75% (p < 0.001). In the gastritis group, CTTS expression was significantly higher in patients with a positive test for H. pylori than negative test for H. pylori: 87.5% and 38.5%, respectively (p<0.001). There was no statistically significant association between CTTS positivity and clinicopathological variables, including tumour staging, histological type, angiolymphatic invasion, recurrence, current status and death. CONCLUSION: CTTS expression is higher in gastric adenocarcinoma samples. Patients with gastritis due to H. pylori also show a higher expression of CTTS than patients with negative results for this bacterium.


Assuntos
Adenocarcinoma , Gastrite , Infecções por Helicobacter , Helicobacter pylori , Neoplasias Gástricas , Adenocarcinoma/patologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Catepsinas , Estudos Transversais , Feminino , Mucosa Gástrica/patologia , Gastrite/patologia , Infecções por Helicobacter/complicações , Infecções por Helicobacter/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias Gástricas/patologia , Adulto Jovem
16.
Rev Col Bras Cir ; 49: e20223299, 2022.
Artigo em Inglês, Português | MEDLINE | ID: mdl-35858036

RESUMO

INTRODUCTION: to evaluate the long-term impact of bariatric surgery in the elderly population. METHODS: a retrospective study including all patients older than 60 years who underwent Roux-en-Y gastric bypass (RYGB) at our center and maintained a follow-up longer than 1 year. Clinical and laboratory variables were studied to assess remission of obesity and its comorbidities, as well as variables directly related to the surgical procedure itself, including early and late complications. RESULTS: fifty-six patients were studied, mostly female (76,8%), with a mean age of 64.02 ± 3.34. A rate of complications of 37,5% was observed, with 10,7% requiring hospital admission and emergency surgery. The mean excess weight loss (%EWL) was 74.22% ± 26.76. The remission rates of hypertension and diabetes mellitus were 26.08% and 54.54%, respectively. There was significant difference in BMI reduction (12.25 ± 5.42, p<0.001), total cholesterol (31.37 ± 38.89 p<0,001), LDL cholesterol (23.45 ± 34.9, p=0.002), HDL cholesterol (5.14 ± 11.13, p=0,024), triglycerides (48.85 ± 56.15 p<0.001), HbA1C (1,81 ± 1,97, p<0,001) e PCR (1.43 ± 1.96, p<0.001). CONCLUSION: bariatric surgery was effective in weight loss and remission of comorbidities in the elderly obese population within the long term.


Assuntos
Cirurgia Bariátrica , Diabetes Mellitus Tipo 2 , Derivação Gástrica , Obesidade Mórbida , Idoso , Cirurgia Bariátrica/métodos , Diabetes Mellitus Tipo 2/complicações , Feminino , Derivação Gástrica/métodos , Humanos , Masculino , Pessoa de Meia-Idade , Obesidade/complicações , Obesidade/cirurgia , Obesidade Mórbida/complicações , Obesidade Mórbida/epidemiologia , Obesidade Mórbida/cirurgia , Estudos Retrospectivos , Resultado do Tratamento , Redução de Peso
17.
J Invest Surg ; 35(4): 900-909, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34180750

RESUMO

BACKGROUND: Ischemic preconditioning (IPC), either direct (DIPC) or remote (RIPC), is a procedure aimed at reducing the harmful effects of ischemia-reperfusion (I/R) injury. OBJECTIVES: To assess the local and systemic effects of DIPC, RIPC, and both combined, in the pig liver transplant model. MATERIALS AND METHODS: Twenty-four pigs underwent orthotopic liver transplantation and were divided into 4 groups: control, direct donor preconditioning, indirect preconditioning at the recipient, and direct donor with indirect recipient preconditioning. The recorded parameters were: donor and recipient weight, graft-to-recipient weight ratio (GRWR), surgery time, warm and cold ischemia time, and intraoperative hemodynamic values. Blood samples were collected before native liver removal (BL) and at 0 h, 1 h, 3 h, 6 h, 12 h, 18 h, and 24 h post-reperfusion for the biochemical tests: aspartate aminotransferase (AST), alanine aminotransferase (ALT), alkaline phosphatase (ALP), gamma-glutamyl transferase (GGT), creatinine, BUN (blood urea nitrogen), lactate, total and direct bilirubin. Histopathological examination of liver, gut, kidney, and lung fragments were performed, as well as molecular analyses for expression of the apoptosis-related BAX (pro-apoptotic) and Bcl-XL (anti-apoptotic) genes, eNOS (endothelial nitric oxide synthase) gene, and IL-6 gene related to inflammatory ischemia-reperfusion injury, using real-time polymerase chain reaction (RT-PCR). RESULTS: There were no differences between the groups regarding biochemical and histopathological parameters. We found a reduced ratio between the expression of the BAX gene and Bcl-XL in the livers of animals with IPC versus the control group. CONCLUSIONS: DIPC, RIPC or a combination of both, produce beneficial effects at the molecular level without biochemical or histological changes.


Assuntos
Precondicionamento Isquêmico , Transplante de Fígado , Traumatismo por Reperfusão , Animais , Aspartato Aminotransferases , Precondicionamento Isquêmico/métodos , Fígado/patologia , Transplante de Fígado/efeitos adversos , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Suínos
18.
J Clin Invest ; 118(12): 3860-9, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19033657

RESUMO

The esophageal epithelium is a prototypical stratified squamous epithelium that exhibits an exquisite equilibrium between proliferation and differentiation. After basal cells proliferate, they migrate outward toward the luminal surface, undergo differentiation, and eventually slough due to apoptosis. The identification and characterization of stem cells responsible for the maintenance of the esophageal epithelium remains elusive. Here, we employed Hoechst dye extrusion and BrdU label-retaining assays to identify in mice a potential esophageal stem cell population that localizes to the basal cell compartment. The self-renewing capacity of this population was characterized using a clonogenic assay and a 3D organotypic culture model. The putative esophageal stem cells were also capable of epithelial reconstitution in vivo in direct esophageal epithelial injury models. In both the 3D organotypic culture and direct mucosal injury models, the putative stem cells gave rise to undifferentiated and differentiated cells. These studies therefore provide a basis for understanding the regenerative capacity and biology of the esophageal epithelium when it is faced with injurious insults.


Assuntos
Diferenciação Celular/fisiologia , Proliferação de Células , Células Epiteliais/citologia , Esôfago/citologia , Células-Tronco/citologia , Animais , Bromodesoxiuridina/química , Movimento Celular/fisiologia , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Epitélio/metabolismo , Doenças do Esôfago/metabolismo , Esôfago/metabolismo , Corantes Fluorescentes/química , Camundongos , Células-Tronco/metabolismo
19.
J Clin Invest ; 118(12): 4058-66, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19033674

RESUMO

Eosinophils are multifunctional leukocytes that degrade and remodel tissue extracellular matrix through production of proteolytic enzymes, release of proinflammatory factors to initiate and propagate inflammatory responses, and direct activation of mucus secretion and smooth muscle cell constriction. Thus, eosinophils are central effector cells during allergic airway inflammation and an important clinical therapeutic target. Here we describe the use of an injectable MMP-targeted optical sensor that specifically and quantitatively resolves eosinophil activity in the lungs of mice with experimental allergic airway inflammation. Through the use of real-time molecular imaging methods, we report the visualization of eosinophil responses in vivo and at different scales. Eosinophil responses were seen at single-cell resolution in conducting airways using near-infrared fluorescence fiberoptic bronchoscopy, in lung parenchyma using intravital microscopy, and in the whole body using fluorescence-mediated molecular tomography. Using these real-time imaging methods, we confirmed the immunosuppressive effects of the glucocorticoid drug dexamethasone in the mouse model of allergic airway inflammation and identified a viridin-derived prodrug that potently inhibited the accumulation and enzyme activity of eosinophils in the lungs. The combination of sensitive enzyme-targeted sensors with noninvasive molecular imaging approaches permitted evaluation of airway inflammation severity and was used as a model to rapidly screen for new drug effects. Both fluorescence-mediated tomography and fiberoptic bronchoscopy techniques have the potential to be translated into the clinic.


Assuntos
Androstenos/farmacologia , Anti-Inflamatórios/farmacologia , Bacteriocinas/farmacologia , Broncoscopia/métodos , Dexametasona/farmacologia , Pró-Fármacos/farmacologia , Hipersensibilidade Respiratória/tratamento farmacológico , Hipersensibilidade Respiratória/patologia , Tomografia Óptica/métodos , Androstenos/uso terapêutico , Animais , Anti-Inflamatórios/uso terapêutico , Bacteriocinas/uso terapêutico , Dexametasona/uso terapêutico , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos/métodos , Eosinófilos/enzimologia , Eosinófilos/patologia , Inflamação/tratamento farmacológico , Inflamação/enzimologia , Inflamação/patologia , Inflamação/fisiopatologia , Pulmão/enzimologia , Pulmão/patologia , Pulmão/fisiopatologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Mutantes , Contração Muscular/efeitos dos fármacos , Músculo Liso/enzimologia , Músculo Liso/patologia , Pró-Fármacos/uso terapêutico , Hipersensibilidade Respiratória/enzimologia , Hipersensibilidade Respiratória/fisiopatologia
20.
Arterioscler Thromb Vasc Biol ; 30(10): 1933-9, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20689078

RESUMO

OBJECTIVE: To investigate the effects of pioglitazone (PIO), a peroxisome proliferator-activated receptor γ agonist, on plaque matrix metalloproteinase (MMP) and macrophage (Mac) responses in vivo in a molecular imaging study. METHODS AND RESULTS: In vitro, PIO suppressed MMP-9 protein expression in murine peritoneal Macs (P<0.05). To assess PIO's effects on plaque inflammation, nondiabetic apolipoprotein E(-/-) mice receiving a high-cholesterol diet (HCD) were administered an MMP-activatable fluorescence imaging agent and a spectrally distinct Mac-avid fluorescent nanoparticle. After 24 hours, mice underwent survival dual-target intravital fluorescence microscopy of carotid arterial plaques. These mice were then randomized to HCD or HCD plus 0.012% PIO for 8 weeks, followed by a second intravital fluorescence microscopy study of the same carotid plaque. In the HCD group, in vivo MMP and Mac target-to-background ratios increased similarly (P<0.01 versus baseline). In contrast, PIO reduced MMP and Mac target-to-background ratios (P<0.01) versus HCD. Changes in MMP and Mac signals correlated strongly (r ≥0.75). Microscopy demonstrated MMP and Mac reductions in PIO-treated mice and a PIO-modulated increase in plaque collagen. CONCLUSIONS: Serial optical molecular imaging demonstrates that plaque MMP and Mac activity in vivo intensify with hypercholesterolemia and are reduced by PIO therapy.


Assuntos
Doenças das Artérias Carótidas/tratamento farmacológico , PPAR gama/agonistas , Tiazolidinedionas/farmacologia , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Doenças das Artérias Carótidas/patologia , Doenças das Artérias Carótidas/fisiopatologia , Colesterol na Dieta/administração & dosagem , Colágeno/metabolismo , Feminino , Inflamação/tratamento farmacológico , Inflamação/patologia , Inflamação/fisiopatologia , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Metaloproteinase 9 da Matriz , Inibidores de Metaloproteinases de Matriz , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Pioglitazona
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa