Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
bioRxiv ; 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39211113

RESUMEN

Cancer cell proliferation requires precise control of E2F1 activity; excess activity promotes apoptosis. Here, we developed cell-permeable and bioavailable macrocycles that selectively kill small cell lung cancer (SCLC) cells with inherent high E2F1 activity by blocking RxL-mediated interactions of cyclin A and cyclin B with select substrates. Genome-wide CRISPR/Cas9 knockout and random mutagenesis screens found that cyclin A/B RxL macrocyclic inhibitors (cyclin A/Bi) induced apoptosis paradoxically by cyclin B- and Cdk2-dependent spindle assembly checkpoint activation (SAC). Mechanistically, cyclin A/Bi hyperactivate E2F1 and cyclin B by blocking their RxL-interactions with cyclin A and Myt1, respectively, ultimately leading to SAC activation and mitotic cell death. Base editor screens identified cyclin B variants that confer cyclin A/Bi resistance including several variants that disrupted cyclin B:Cdk interactions. Unexpectedly but consistent with our base editor and knockout screens, cyclin A/Bi induced the formation of neo-morphic Cdk2-cyclin B complexes that promote SAC activation and apoptosis. Finally, orally-bioavailable cyclin A/Bi robustly inhibited tumor growth in chemotherapy-resistant patient-derived xenograft models of SCLC. This work uncovers gain-of-function mechanisms by which cyclin A/Bi induce apoptosis in cancers with high E2F activity, and suggests cyclin A/Bi as a therapeutic strategy for SCLC and other cancers driven by high E2F activity.

2.
Cell Rep Med ; 4(1): 100878, 2023 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-36599350

RESUMEN

Although immune checkpoint inhibitors (ICIs) are established as effective cancer therapies, overcoming therapeutic resistance remains a critical challenge. Here we identify interleukin 6 (IL-6) as a correlate of poor response to atezolizumab (anti-PD-L1) in large clinical trials of advanced kidney, breast, and bladder cancers. In pre-clinical models, combined blockade of PD-L1 and the IL-6 receptor (IL6R) causes synergistic regression of large established tumors and substantially improves anti-tumor CD8+ cytotoxic T lymphocyte (CTL) responses compared with anti-PD-L1 alone. Circulating CTLs from cancer patients with high plasma IL-6 display a repressed functional profile based on single-cell RNA sequencing, and IL-6-STAT3 signaling inhibits classical cytotoxic differentiation of CTLs in vitro. In tumor-bearing mice, CTL-specific IL6R deficiency is sufficient to improve anti-PD-L1 activity. Thus, based on both clinical and experimental evidence, agents targeting IL-6 signaling are plausible partners for combination with ICIs in cancer patients.


Asunto(s)
Antineoplásicos , Interleucina-6 , Neoplasias , Animales , Ratones , Antineoplásicos/uso terapéutico , Antígeno B7-H1/inmunología , Antígeno B7-H1/uso terapéutico , Linfocitos T CD8-positivos/metabolismo , Inmunoterapia , Interleucina-6/metabolismo , Neoplasias/inmunología , Neoplasias/terapia
3.
Nat Commun ; 12(1): 3969, 2021 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-34172722

RESUMEN

Immune checkpoint inhibitors targeting the PD-1/PD-L1 axis lead to durable clinical responses in subsets of cancer patients across multiple indications, including non-small cell lung cancer (NSCLC), urothelial carcinoma (UC) and renal cell carcinoma (RCC). Herein, we complement PD-L1 immunohistochemistry (IHC) and tumor mutation burden (TMB) with RNA-seq in 366 patients to identify unifying and indication-specific molecular profiles that can predict response to checkpoint blockade across these tumor types. Multiple machine learning approaches failed to identify a baseline transcriptional signature highly predictive of response across these indications. Signatures described previously for immune checkpoint inhibitors also failed to validate. At the pathway level, significant heterogeneity is observed between indications, in particular within the PD-L1+ tumors. mUC and NSCLC are molecularly aligned, with cell cycle and DNA damage repair genes associated with response in PD-L1- tumors. At the gene level, the CDK4/6 inhibitor CDKN2A is identified as a significant transcriptional correlate of response, highlighting the association of non-immune pathways to the outcome of checkpoint blockade. This cross-indication analysis reveals molecular heterogeneity between mUC, NSCLC and RCC tumors, suggesting that indication-specific molecular approaches should be prioritized to formulate treatment strategies.


Asunto(s)
Biomarcadores de Tumor/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Inhibidores de Puntos de Control Inmunológico/farmacología , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Antígeno B7-H1/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Transicionales/tratamiento farmacológico , Carcinoma de Células Transicionales/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Mutación , Resultado del Tratamiento , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/genética , Secuenciación Completa del Genoma
5.
Neural Regen Res ; 16(9): 1677-1685, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33510055

RESUMEN

Transplantation of neural stem cells (NSCs) can protect neurons in animal stroke models; however, their low rates of survival and neuronal differentiation limit their clinical application. Glial niches, an important location of neural stem cells, regulate survival, proliferation and differentiation of neural stem cells. However, the effects of activated glial cells on neural stem cells remain unclear. In the present study, we explored the effects of activated astrocytes and microglia on neural stem cells in vitro stroke models. We also investigated the effects of combined transplantation of neural stem cells and glial cells after stroke in rats. In a Transwell co-culture system, primary cultured astrocytes, microglia or mixed glial cells were exposed to glutamate or H2O2 and then seeded in the upper inserts, while primary neural stem cells were seeded in the lower uncoated wells and cultured for 7 days. Our results showed that microglia were conducive to neurosphere formation and had no effects on apoptosis within neurospheres, while astrocytes and mixed glial cells were conducive to neurosphere differentiation and reduced apoptosis within neurospheres, regardless of their pretreatment. In contrast, microglia and astrocytes induced neuronal differentiation of neural stem cells in differentiation medium, regardless of their pretreatment, with an exception of astrocytes pretreated with H2O2. Rat models of ischemic stroke were established by occlusion of the middle cerebral artery. Three days later, 5 × 105 neural stem cells with microglia or astrocytes were injected into the right lateral ventricle. Neural stem cell/astrocyte-treated rats displayed better improvement of neurological deficits than neural stem cell only-treated rats at 4 days after cell transplantation. Moreover, neural stem cell/microglia-, and neural stem cell/astrocyte-treated rats showed a significant decrease in ischemic volume compared with neural stem cell-treated rats. These findings indicate that microglia and astrocytes exert different effects on neural stem cells, and that co-transplantation of neural stem cells and astrocytes is more conducive to the recovery of neurological impairment in rats with ischemic stroke. The study was approved by the Animal Ethics Committee of Tongji University School of Medicine, China (approval No. 2010-TJAA08220401) in 2010.

6.
Cancer Cell ; 38(6): 803-817.e4, 2020 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-33157048

RESUMEN

Integrated multi-omics evaluation of 823 tumors from advanced renal cell carcinoma (RCC) patients identifies molecular subsets associated with differential clinical outcomes to angiogenesis blockade alone or with a checkpoint inhibitor. Unsupervised transcriptomic analysis reveals seven molecular subsets with distinct angiogenesis, immune, cell-cycle, metabolism, and stromal programs. While sunitinib and atezolizumab + bevacizumab are effective in subsets with high angiogenesis, atezolizumab + bevacizumab improves clinical benefit in tumors with high T-effector and/or cell-cycle transcription. Somatic mutations in PBRM1 and KDM5C associate with high angiogenesis and AMPK/fatty acid oxidation gene expression, while CDKN2A/B and TP53 alterations associate with increased cell-cycle and anabolic metabolism. Sarcomatoid tumors exhibit lower prevalence of PBRM1 mutations and angiogenesis markers, frequent CDKN2A/B alterations, and increased PD-L1 expression. These findings can be applied to molecularly stratify patients, explain improved outcomes of sarcomatoid tumors to checkpoint blockade versus antiangiogenics alone, and develop personalized therapies in RCC and other indications.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor/genética , Carcinoma de Células Renales/tratamiento farmacológico , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias Renales/tratamiento farmacológico , Inhibidores de la Angiogénesis/farmacología , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Bevacizumab/farmacología , Bevacizumab/uso terapéutico , Carcinoma de Células Renales/genética , Ensayos Clínicos Fase III como Asunto , Biología Computacional/métodos , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Neoplasias Renales/genética , Pronóstico , Ensayos Clínicos Controlados Aleatorios como Asunto , Análisis de Secuencia de ARN , Sunitinib/farmacología , Sunitinib/uso terapéutico , Resultado del Tratamiento , Aprendizaje Automático no Supervisado
7.
Nat Med ; 26(5): 693-698, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32405063

RESUMEN

Although elevated plasma interleukin-8 (pIL-8) has been associated with poor outcome to immune checkpoint blockade 1, this has not been comprehensively evaluated in large randomized studies. Here we analyzed circulating pIL-8 and IL8 gene expression in peripheral blood mononuclear cells and tumors of patients treated with atezolizumab (anti-PD-L1 monoclonal antibody) from multiple randomized trials representing 1,445 patients with metastatic urothelial carcinoma (mUC) and metastatic renal cell carcinoma. High levels of IL-8 in plasma, peripheral blood mononuclear cells and tumors were associated with decreased efficacy of atezolizumab in patients with mUC and metastatic renal cell carcinoma, even in tumors that were classically CD8+ T cell inflamed. Low baseline pIL-8 in patients with mUC was associated with increased response to atezolizumab and chemotherapy. Patients with mUC who experienced on-treatment decreases in pIL-8 exhibited improved overall survival when treated with atezolizumab but not with chemotherapy. Single-cell RNA sequencing of the immune compartment showed that IL8 is primarily expressed in circulating and intratumoral myeloid cells and that high IL8 expression is associated with downregulation of the antigen-presentation machinery. Therapies that can reverse the impacts of IL-8-mediated myeloid inflammation will be essential for improving outcomes of patients treated with immune checkpoint inhibitors.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Antígeno B7-H1/antagonistas & inhibidores , Interleucina-8/metabolismo , Neoplasias/diagnóstico , Neoplasias/tratamiento farmacológico , Adulto , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antígeno B7-H1/inmunología , Biomarcadores Farmacológicos/sangre , Biomarcadores Farmacológicos/metabolismo , Biomarcadores de Tumor/sangre , Biomarcadores de Tumor/metabolismo , Carcinoma de Células Renales/diagnóstico , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/mortalidad , Carcinoma de Células Transicionales/diagnóstico , Carcinoma de Células Transicionales/tratamiento farmacológico , Carcinoma de Células Transicionales/metabolismo , Carcinoma de Células Transicionales/mortalidad , Resistencia a Antineoplásicos , Femenino , Humanos , Interleucina-8/sangre , Neoplasias Renales/diagnóstico , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/metabolismo , Neoplasias Renales/mortalidad , Masculino , Neoplasias/metabolismo , Neoplasias/mortalidad , Pronóstico , Análisis de Supervivencia , Insuficiencia del Tratamiento , Neoplasias Urológicas/diagnóstico , Neoplasias Urológicas/tratamiento farmacológico , Neoplasias Urológicas/metabolismo , Neoplasias Urológicas/mortalidad
8.
Polymers (Basel) ; 10(4)2018 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-30966381

RESUMEN

In the current study, the poly (amide-urethane) (PAUt) membranes were successfully fabricated by interfacial polymerization of m-phenylenediamine (MPD) and 5-choroformyloxyisophaloyl chloride (CFIC) on the polysulfone substrates. Two modification methods based on layer-by-layer assembly were applied to modify the PAUt membrane surface to achieve antifouling property: 1. Chitosan (CS) was directly self-assembled on the PAUt membrane (i.e., PAUt-CS); and 2. polydimethyl diallyl ammonium chloride (PDDA), polystyrene sulfonate (PSS), and CS were successively self-assembled on the membrane surface (i.e., PAUt-PDDA/PSS/CS). The resultant membranes were symmetrically characterized by Attenuated Total Reflection Fourier Transform Infrared Spectroscopy (ATR-FTIR), X-ray Photoelectron Spectroscopy (XPS), Scanning Electron Microscopy (SEM), Atomic Force Microscopy (AFM) and Contact Angle Meter (CAM), respectively. The results indicated that the modified membranes had much smoother and more hydrophilic surfaces as compared to the nascent PAUt membrane. Meanwhile, the modified membranes exhibited better reverse osmosis performance in terms of water permeability and salt rejection. After the modified membranes were fouled by lake water, the PAUt-PDDA/PSS/CS membrane presented the best antifouling performance among the three types of membranes. Combining the reverse osmosis performance with the anti-fouling property obviously, the PAUt-PDDA/PSS/CS membrane behaved as a promising candidate to be used in real applications.

9.
RSC Adv ; 8(66): 37817-37827, 2018 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-35558596

RESUMEN

A novel reverse osmosis (RO) composite membrane, poly(amide-urethane-imide@Ag) (PAUI@Ag), was prepared on a polysulfone supporting film through two-step interfacial polymerization. First, in the 1st interfacial polymerization procedure, a new tri-functional crosslinking agent with -OCOCl and -COCl groups, 5-choroformyloxyisophaloyl chloride (CFIC), was reacted with 4-methyl-phenylenediamine (MMPD) without curing treatment to obtain the poly(amide-urethane) base membrane with a CFIC-MMPD precursor separation layer. And then N,N'-dimethyl-m-phenylenediamine (DMMPD) with nano-Ag particle dispersion was introduced onto the base membrane to further construct a CFIC-DMMPD modified ultrathin separation layer via the 2nd interfacial polymerization. Thus, the PAUI@Ag RO membrane with poly(amide-urethane-imide) bi-layer skin was obtained. The membrane was characterized for the chemical composition of separation layer, the membrane cross-section structure and the membrane surface morphology. Permeation experiment was employed to evaluate the PAUI@Ag membrane performance including salt rejection rate and water flux. The results revealed that the PAUI@Ag membrane composed the highly cross-linked separation layer with entire ridges and valleys, small surface roughness, and well dispersed nano-Ag particles. Upon exposure of the membranes to high concentration of free chlorine solutions, the PAUI@Ag RO membrane showed a slightly less chlorine-resistant property compared with the nascent PAUI RO membrane, but was still superior to the conventional polyamide MPD-TMC RO membrane, meanwhile it processed higher anti-biofouling property.

10.
Diabetes Obes Metab ; 20(2): 352-361, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28776922

RESUMEN

AIM: To evaluate the efficacy, pharmacokinetic (PK) profile and tolerability of subcutaneous (s.c.). exendin 9-39 (Ex-9) injection in patients with post-bariatric hypoglycaemia (PBH). METHODS: Nine women who had recurrent symptomatic hypoglycaemia after undergoing Roux-en-Y gastric bypass were enrolled in this 2-part, single-blind, single-ascending-dose study. In Part 1, a single participant underwent equimolar low-dose intravenous (i.v.) vs s.c. Ex-9 administration; in Part 2, 8 participants were administered single ascending doses of s.c. Ex-9 during an oral glucose tolerance test (OGTT). Glycaemic, hormonal, PK and symptomatic responses were compared with those obtained during the baseline OGTT. RESULTS: Although an exposure-response relationship was observed, all doses effectively prevented hyperinsulinaemic hypoglycaemia and improved associated symptoms. On average, the postprandial glucose nadir was increased by 66%, peak insulin was reduced by 57%, and neuroglycopenic symptoms were reduced by 80%. All doses were well tolerated with no treatment-emergent adverse events observed. CONCLUSIONS: Injection s.c. of Ex-9 appears to represent a safe, effective and targeted therapeutic approach for treatment of PBH. Further investigation involving multiple doses with chronic dosing is warranted.


Asunto(s)
Derivación Gástrica/efectos adversos , Receptor del Péptido 1 Similar al Glucagón/antagonistas & inhibidores , Hiperinsulinismo/prevención & control , Hipoglucemia/prevención & control , Hipoglucemiantes/administración & dosificación , Fragmentos de Péptidos/administración & dosificación , Complicaciones Posoperatorias/prevención & control , Adulto , Área Bajo la Curva , Estudios de Cohortes , Relación Dosis-Respuesta a Droga , Femenino , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Prueba de Tolerancia a la Glucosa/efectos adversos , Semivida , Humanos , Hiperinsulinismo/sangre , Hiperinsulinismo/etiología , Hiperinsulinismo/metabolismo , Hipoglucemia/sangre , Hipoglucemia/etiología , Hipoglucemia/metabolismo , Hipoglucemiantes/efectos adversos , Hipoglucemiantes/farmacocinética , Hipoglucemiantes/uso terapéutico , Infusiones Intravenosas , Inyecciones Subcutáneas , Insulina/sangre , Persona de Mediana Edad , Fragmentos de Péptidos/efectos adversos , Fragmentos de Péptidos/farmacocinética , Fragmentos de Péptidos/uso terapéutico , Proyectos Piloto , Complicaciones Posoperatorias/sangre , Complicaciones Posoperatorias/etiología , Complicaciones Posoperatorias/metabolismo , Periodo Posprandial , Método Simple Ciego
11.
PLoS One ; 12(2): e0170728, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28151993

RESUMEN

AIM: The physiologic mechanisms underlying the relationship between obesity and insulin resistance are not fully understood. Impaired adipocyte differentiation and localized inflammation characterize adipose tissue from obese, insulin-resistant humans. The directionality of this relationship is not known, however. The aim of the current study was to investigate whether adipose tissue inflammation is causally-related to impaired adipocyte differentiation. METHODS: Abdominal subcutaneous(SAT) and visceral(VAT) adipose tissue was obtained from 20 human participants undergoing bariatric surgery. Preadipocytes were isolated, and cultured in the presence or absence of CD14+ macrophages obtained from the same adipose tissue sample. Adipocyte differentiation was quantified after 14 days via immunofluorescence, Oil-Red O, and adipogenic gene expression. Cytokine secretion by mature adipocytes cultured with or without CD14+macrophages was quantified. RESULTS: Adipocyte differentiation was significantly lower in VAT than SAT by all measures (p<0.001). With macrophage removal, SAT preadipocyte differentiation increased significantly as measured by immunofluorescence and gene expression, whereas VAT preadipocyte differentiation was unchanged. Adipocyte-secreted proinflammatory cytokines were higher and adiponectin lower in media from VAT vs SAT: macrophage removal reduced inflammatory cytokine and increased adiponectin secretion from both SAT and VAT adipocytes. Differentiation of preadipocytes from SAT but not VAT correlated inversely with systemic insulin resistance. CONCLUSIONS: The current results reveal that proinflammatory immune cells in human SAT are causally-related to impaired preadipocyte differentiation, which in turn is associated with systemic insulin resistance. In VAT, preadipocyte differentiation is poor even in the absence of tissue macrophages, pointing to inherent differences in fat storage potential between the two depots.


Asunto(s)
Adipocitos/citología , Adipogénesis/fisiología , Tejido Adiposo/citología , Resistencia a la Insulina/fisiología , Macrófagos/inmunología , Obesidad/patología , Adipoquinas/metabolismo , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Femenino , Regulación de la Expresión Génica , Humanos , Inflamación/patología , Receptores de Lipopolisacáridos/metabolismo , Masculino , Persona de Mediana Edad
12.
Diabetologia ; 60(3): 531-540, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27975209

RESUMEN

AIMS/HYPOTHESIS: Post-bariatric hypoglycaemia (PBH) is a rare, but severe, metabolic disorder arising months to years after bariatric surgery. It is characterised by symptomatic postprandial hypoglycaemia, with inappropriately elevated insulin concentrations. The relative contribution of exaggerated incretin hormone signalling to dysregulated insulin secretion and symptomatic hypoglycaemia is a subject of ongoing inquiry. This study was designed to test the hypothesis that PBH and associated symptoms are primarily mediated by glucagon-like peptide-1 (GLP-1). METHODS: We conducted a double-blinded crossover study wherein eight participants with confirmed PBH were assigned in random order to intravenous infusion of the GLP-1 receptor (GLP-1r) antagonist. Exendin (9-39) (Ex-9), or placebo during an OGTT on two separate days at the Stanford University Clinical and Translational Research Unit. Metabolic, symptomatic and pharmacokinetic variables were evaluated. Results were compared with a cohort of BMI- and glucose-matched non-surgical controls (NSCs). RESULTS: Infusion of Ex-9 decreased the time to peak glucose and rate of glucose decline during OGTT, and raised the postprandial nadir by over 70%, normalising it relative to NSCs and preventing hypoglycaemia in all PBH participants. Insulin AUC and secretion rate decreased by 57% and 71% respectively, and peak postprandial insulin was normalised relative to NSCs. Autonomic and neuroglycopenic symptoms were significantly reduced during Ex-9 infusion. CONCLUSIONS/INTERPRETATION: GLP-1r blockade prevented hypoglycaemia in 100% of individuals, normalised beta cell function and reversed neuroglycopenic symptoms, supporting the conclusion that GLP-1 plays a primary role in mediating hyperinsulinaemic hypoglycaemia in PBH. Competitive antagonism at the GLP-1r merits consideration as a therapeutic strategy. TRIAL REGISTRATION: ClinicalTrials.gov NCT02550145.


Asunto(s)
Péptido 1 Similar al Glucagón/metabolismo , Hipoglucemia/metabolismo , Adulto , Estudios Cruzados , Método Doble Ciego , Femenino , Derivación Gástrica , Polipéptido Inhibidor Gástrico/metabolismo , Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/antagonistas & inhibidores , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Humanos , Insulina/metabolismo , Masculino , Persona de Mediana Edad , Fragmentos de Péptidos/farmacología , Encuestas y Cuestionarios
13.
BMC Neurosci ; 17(1): 37, 2016 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-27296974

RESUMEN

BACKGROUND: Glucagon-like peptide 1 (GLP-1) analogs protect a variety of cell types against oxidative damage and vascular and neuronal injury via binding to GLP-1 receptors. This study aimed to investigate the effects of the GLP-1 analogs exendin-4 and liraglutide on cerebral blood flow, reactive oxygen species production, expression of oxidative stress-related proteins, cognition, and pelvic sympathetic nerve-mediated bladder contraction after middle cerebral artery occlusion (MCAO) injury in the db/db mouse model of diabetes. RESULTS: Sixty minutes of MCAO increased blood and brain reactive oxygen species counts in male db/db mice, as revealed by dihydroethidium staining. MCAO also increased nuclear factor-κB and intercellular adhesion molecule-1 expression and decreased cerebral microcirculation. These effects were attenuated by treatment with exendin-4 or liraglutide. MCAO did not affect basal levels of phosphorylated Akt (p-Akt) or endothelial nitric oxide synthase (p-eNOS); however, exendin-4 and liraglutide treatments significantly enhanced p-Akt and p-eNOS levels, indicating activation of the p-Akt/p-eNOS signaling pathway. MCAO-induced motor and cognitive deficits and micturition dysfunction, indicated by reduced pelvic nerve-mediated voiding contractions and increased nonvoiding contractions, were also partially attenuated by exendin-4 treatment. CONCLUSIONS: The above data indicate that treatment with GLP-1 agonists exerts protective effects against oxidative, inflammatory, and apoptotic damage in brain areas that control parasympathetic/pelvic nerve-mediated voiding contractions and cognitive and motor behaviors in a diabetic mouse model.


Asunto(s)
Trastornos del Conocimiento/tratamiento farmacológico , Diabetes Mellitus Experimental/tratamiento farmacológico , Receptor del Péptido 1 Similar al Glucagón/agonistas , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Trastornos Urinarios/tratamiento farmacológico , Animales , Circulación Cerebrovascular/efectos de los fármacos , Circulación Cerebrovascular/fisiología , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/fisiopatología , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/fisiopatología , Exenatida , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Hipoglucemiantes/farmacología , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/fisiopatología , Liraglutida/farmacología , Masculino , Ratones , Trastornos del Movimiento/tratamiento farmacológico , Trastornos del Movimiento/etiología , Trastornos del Movimiento/fisiopatología , Nootrópicos/farmacología , Estrés Oxidativo/fisiología , Péptidos/farmacología , Sustancias Protectoras/farmacología , Trastornos Urinarios/etiología , Trastornos Urinarios/fisiopatología , Ponzoñas/farmacología
14.
Diabetes ; 65(5): 1245-54, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26884438

RESUMEN

Obesity is associated with insulin resistance, but significant variability exists between similarly obese individuals, pointing to qualitative characteristics of body fat as potential mediators. To test the hypothesis that obese, insulin-sensitive (IS) individuals possess adaptive adipose cell/tissue responses, we measured subcutaneous adipose cell size, insulin suppression of lipolysis, and regional fat responses to short-term overfeeding in BMI-matched overweight/obese individuals classified as IS or insulin resistant (IR). At baseline, IR subjects exhibited significantly greater visceral adipose tissue (VAT), intrahepatic lipid (IHL), plasma free fatty acids, adipose cell diameter, and percentage of small adipose cells. With weight gain (3.1 ± 1.4 kg), IR subjects demonstrated no significant change in adipose cell size, VAT, or insulin suppression of lipolysis and only 8% worsening of insulin-mediated glucose uptake (IMGU). Alternatively, IS subjects demonstrated significant adipose cell enlargement; decrease in the percentage of small adipose cells; increase in VAT, IHL, and lipolysis; 45% worsening of IMGU; and decreased expression of lipid metabolism genes. Smaller baseline adipose cell size and greater enlargement with weight gain predicted decline in IMGU, as did increase in IHL and VAT and decrease in insulin suppression of lipolysis. Weight gain in IS humans causes maladaptive changes in adipose cells, regional fat distribution, and insulin resistance. The correlation between development of insulin resistance and changes in adipose cell size, VAT, IHL, and insulin suppression of lipolysis highlight these factors as potential mediators between obesity and insulin resistance.


Asunto(s)
Adiposidad , Hiperfagia/patología , Resistencia a la Insulina , Grasa Intraabdominal/patología , Modelos Biológicos , Sobrepeso/patología , Grasa Subcutánea/patología , Adulto , Índice de Masa Corporal , Tamaño de la Célula/efectos de los fármacos , Estudios de Cohortes , Femenino , Regulación Enzimológica de la Expresión Génica , Humanos , Hiperfagia/metabolismo , Hiperfagia/fisiopatología , Hipoglucemiantes/farmacología , Insulina/farmacología , Grasa Intraabdominal/efectos de los fármacos , Grasa Intraabdominal/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Masculino , Persona de Mediana Edad , Obesidad/etiología , Obesidad/metabolismo , Obesidad/patología , Sobrepeso/etiología , Sobrepeso/metabolismo , Grasa Subcutánea/efectos de los fármacos , Grasa Subcutánea/metabolismo , Aumento de Peso
15.
Diabetologia ; 58(7): 1579-86, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25952479

RESUMEN

AIMS/HYPOTHESIS: Proinflammatory immune cell infiltration in human adipose tissue is associated with the development of insulin resistance. We previously identified, via a gene expression-based genome-wide association study, the cell-surface immune cell receptor CD44 as a functionally important gene associated with type 2 diabetes. We then showed that, compared with controls, Cd44 knockout mice were protected from insulin resistance and adipose tissue inflammation during diet-induced obesity. We thus sought to test whether CD44 is associated with adipose tissue inflammation and insulin resistance in humans. METHODS: Participants included 58 healthy, overweight/moderately obese white adults who met predetermined criteria for insulin resistance or insulin sensitivity based on the modified insulin-suppression test. Serum was collected from 43 participants to measure circulating concentrations of CD44. Subcutaneous adipose tissue was obtained from 17 participants to compare CD44, its ligand osteopontin (OPN, also known as SPP1) and pro-inflammatory gene expression. CD44 expression on adipose tissue macrophage (ATM) surfaces was determined by flow cytometry. RESULTS: Serum CD44 concentrations were significantly increased in insulin-resistant (IR) participants. CD44 gene expression in subcutaneous adipose tissue was threefold higher in the IR subgroup. The expression of OPN, CD68 and IL6 was also significantly elevated in IR individuals. CD44 gene expression correlated significantly with CD68 and IL6 expression. CD44 density on ATMs was associated with proinflammatory M1 polarisation. CONCLUSIONS/INTERPRETATION: CD44 and OPN in human adipose tissue are associated with localised inflammation and systemic insulin resistance. This receptor-ligand pair is worthy of further research as a potentially modifiable contributor to human insulin resistance and type 2 diabetes.


Asunto(s)
Tejido Adiposo/metabolismo , Receptores de Hialuranos/metabolismo , Inflamación/metabolismo , Resistencia a la Insulina/fisiología , Macrófagos/metabolismo , Tejido Adiposo/citología , Estudios de Cohortes , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Dieta , Femenino , Expresión Génica/genética , Estudio de Asociación del Genoma Completo , Humanos , Receptores de Hialuranos/genética , Hipoglucemiantes/farmacología , Insulina/farmacología , Resistencia a la Insulina/genética , Masculino , Persona de Mediana Edad , Osteopontina/metabolismo , Sobrepeso/genética , Sobrepeso/metabolismo , Grasa Subcutánea/metabolismo
16.
Oncotarget ; 5(6): 1475-90, 2014 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-24721996

RESUMEN

Celecoxib, a COX-2 inhibitor and non-steroidal anti-inflammatory drug, can prevent several types of cancer, including hepatocellular carcinoma (HCC). Here we show that celecoxib suppressed the self-renewal and drug-pumping functions in HCC cells. Besides, celecoxib depleted CD44+/CD133+ hepatic cancer stem cells (hCSC). Prostaglandin E2 (PGE2) and CD133 overexpression did not reverse the celecoxib-induced depletion of hCSC. Also, celecoxib inhibited progression of rat Novikoff hepatoma. Moreover, a 60-day celecoxib program increased the survival rate of rats with hepatoma. Histological analysis revealed that celecoxib therapy reduced the abundance of CD44+/CD133+ hCSCs in hepatoma tissues. Besides, the hCSCs depletion was associated with elevated apoptosis and blunted proliferation and angiogenesis in hepatoma. Celecoxib therapy activated peroxisome proliferator-activated receptor γ (PPARγ) and up-regulated PTEN, thereby inhibiting Akt and disrupting hCSC expansion. PTEN gene delivery by adenovirus reduced CD44/CD133 expression in vitro and hepatoma formation in vivo. This study suggests that celecoxib suppresses cancer stemness and progression of HCC via activation of PPARγ/PTEN signaling.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Inhibidores de la Ciclooxigenasa 2/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Fosfohidrolasa PTEN/metabolismo , Pirazoles/farmacología , Sulfonamidas/farmacología , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Celecoxib , Proliferación Celular/efectos de los fármacos , Ciclooxigenasa 2/química , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Progresión de la Enfermedad , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Receptores de Hialuranos/metabolismo , Técnicas para Inmunoenzimas , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , PPAR gamma/genética , PPAR gamma/metabolismo , Fosfohidrolasa PTEN/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Activación Transcripcional , Células Tumorales Cultivadas , Regulación hacia Arriba
17.
PLoS One ; 8(10): e78040, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24205079

RESUMEN

BACKGROUND: Given the emerging roles of miRNAs as potential posttranscriptional/posttranslational regulators of the steroidogenic process in adrenocortical and gonadal cells, we sought to determine miRNA profiles in rat adrenals from animals treated with vehicle, ACTH, 17α-E2 or dexamethasone. Key observations were also confirmed using hormone (Bt2cAMP)-treated mouse Leydig tumor cells, MLTC-1, and primary rat ovarian granulosa cells. METHODOLOGY: RNA was extracted from rat adrenal glands and miRNA profiles were established using microarray and confirmed with qRT-PCR. The expression of some of the hormone-sensitive miRNAs was quantified in MLTC-1 and granulosa cells after stimulation with Bt2cAMP. Targets of hormonally altered miRNAs were explored by qRT-PCR and Western blotting in adrenals and granulosa cells. RESULTS: Adrenals from ACTH, 17α-E2 and dexamethasone treated rats exhibited miRNA profiles distinct from control animals. ACTH up-regulated the expression of miRNA-212, miRNA-182, miRNA-183, miRNA-132, and miRNA-96 and down-regulated the levels of miRNA-466b, miRNA-214, miRNA-503, and miRNA-27a. The levels of miR-212, miRNA-183, miRNA-182, miRNA-132, miRNA-370, miRNA-377, and miRNA-96 were up-regulated, whereas miR-125b, miRNA-200b, miR-122, miRNA-466b, miR-138, miRNA-214, miRNA-503 and miRNA27a were down-regulated in response to 17α-E2 treatment. Dexamethasone treatment decreased miRNA-200b, miR-122, miR-19a, miRNA-466b and miRNA27a levels, but increased miRNA-183 levels. Several adrenal miRNAs are subject to regulation by more than one hormone. Significant cAMP-induced changes in certain miRNAs were also noted in MLTC-1 and granulosa cells. Some of the hormone-induced miRNAs in steroidogenic cells were predicted to target proteins involved in lipid metabolism/steroidogenesis. We also obtained evidence that miR-132 and miRNA-214 inhibit the expression of SREBP-1c and LDLR, respectively. CONCLUSION: Our results demonstrate that expression of a number of miRNAs in steroidogenic cells of the testis, ovary and adrenal glands is subject to hormonal regulation and that miRNAs and their regulation by specific hormones are likely to play a key role in posttranscriptional/posttranslational regulation of steroidogenesis.


Asunto(s)
Glándulas Suprarrenales/metabolismo , MicroARNs/genética , Ovario/metabolismo , Testículo/metabolismo , Glándulas Suprarrenales/efectos de los fármacos , Hormona Adrenocorticotrópica/farmacología , Animales , Western Blotting , Línea Celular Tumoral , Dexametasona/farmacología , Femenino , Masculino , Ratones , Ovario/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Testículo/efectos de los fármacos
18.
PLoS One ; 8(8): e72367, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23967297

RESUMEN

This study aimed to characterize and compare the effects of obesity on gene expression profiles in two distinct adipose depots, epididymal and bone marrow, at two different ages in mice. Alterations in gene expression were analyzed in adipocytes isolated from diet-induced obese (DIO) C57BL/6J male mice at 6 and 14 months of age and from leptin deficient mice (ob/ob) at 6 months of age using microarrays. DIO affected gene expression in both depots at 6 and 14 months, but more genes were altered in epididymal than bone marrow adipocytes at each age and younger mice displayed more changes than older animals. In epididymal adipocytes a total of 2789 (9.6%) genes were differentially expressed at 6-months with DIO, whereas 952 (3.3%) were affected at 14-months. In bone marrow adipocytes, 347 (1.2%) genes were differentially expressed at 6-months with DIO, whereas only 189 (0.66%) were changed at 14-months. 133 genes were altered by DIO in both fat depots at 6-months, and 37 genes at 14-months. Only four genes were altered in both depots at both ages with DIO. Bone marrow adipocytes are less responsive to DIO than epididymal adipocytes and the response of both depots to DIO declines with age. This loss of responsiveness with age is likely due to age-associated changes in expression of genes related to adipogenesis, inflammation and mitochondrial function that are similar to and obscure the changes commonly associated with DIO. Patterns of gene expression were generally similar in epididymal adipocytes from ob/ob and DIO mice; however, several genes were differentially expressed in bone marrow adipocytes from ob/ob and DIO mice, perhaps reflecting the importance of leptin signaling for bone metabolism. In conclusion, obesity affects age-associated alterations in gene expression in both epididymal and bone marrow adipocytes regardless of diet or genetic background.


Asunto(s)
Adipocitos/patología , Envejecimiento/genética , Envejecimiento/patología , Células de la Médula Ósea/patología , Epidídimo/patología , Obesidad/fisiopatología , Transcriptoma , Adipocitos/metabolismo , Envejecimiento/metabolismo , Animales , Dieta Alta en Grasa/efectos adversos , Leptina/deficiencia , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/genética , Obesidad/metabolismo , Obesidad/patología , Análisis de Secuencia por Matrices de Oligonucleótidos
19.
Biochem Biophys Res Commun ; 425(2): 169-76, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22819846

RESUMEN

Hepatoma-derived growth factor (HDGF) stimulates the migration, invasion and metastasis in several types of cancer cells. However, the mechanism underlying HDGF-stimulated migration remains unclear. In this study, we investigated the influence of HDGF on cytoskeleton remodeling and phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway in non-transformed NIH/3T3 cells. Exogenous HDGF promoted the migration and the formation of dorsal ruffles and podosome rosettes. Besides, HDGF supply increased the PI3K expression and Akt phosphorylation in dose- and time-dependent manners. Application of LY294002, a PI3K inhibitor, attenuated the HDGF-induced migration, dorsal ruffles and podosome rosettes formation. Consistently, the HDGF-overexpressing NIH/3T3 transfectants exhibited significantly increased motility and elevated PI3K/Akt activities, which were repressed by LY294002 or adenovirus-mediated overexpression of endogenous PI3K antagonist, PTEN. In summary, HDGF elicits the activation of PI3K/Akt signaling cascade, thereby promoting cytoskeleton remodeling to stimulate cellular migration.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/fisiología , Fosfatidilinositol 3-Quinasa/biosíntesis , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Formación de Roseta , Animales , Movimiento Celular , Citoesqueleto/genética , Citoesqueleto/metabolismo , Activación Enzimática , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Células 3T3 NIH , Transfección
20.
FASEB J ; 25(8): 2722-30, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21566206

RESUMEN

We tested the hypothesis that the actions of hormone-sensitive lipase (HSL) affect the microenvironment of the bone marrow and that removal of HSL function by gene deletion maintains high bone mass in aging mice. We compared littermate control wild-type (WT) and HSL(-/-) mice during aging for changes in serum biochemical values, trabecular bone density using micro-computed tomography, bone histomorphometry, and characteristics of primary bone marrow cells and preosteoblasts. There is a regulated expression of HSL and genes involved in lipid metabolism in the bone marrow during aging. HSL(-/-) mice have increased serum levels of insulin and osteocalcin with decreased leptin levels. Compared with the marked adipocyte infiltration in WT bone marrow (65% by area) at 14 mo, HSL(-/-) mice have fewer (16%, P<0.05) and smaller adipocytes in bone marrow. While peak bone density is similar, HSL(-/-) mice maintain a higher bone density (bone volume/total volume 6.1%) with age than WT mice (2.6%, P<0.05). Primary osteoblasts from HSL(-/-) mice show increased growth rates and higher osteogenic potential, manifested by increased expression of Runx2 (3.5-fold, P<0.05) and osteocalcin (4-fold, P<0.05). The absence of HSL directs cells within the bone marrow toward osteoblast differentiation and favors the maintenance of bone density with aging.


Asunto(s)
Envejecimiento/metabolismo , Densidad Ósea/fisiología , Esterol Esterasa/deficiencia , Adipocitos/citología , Adipocitos/metabolismo , Envejecimiento/genética , Envejecimiento/patología , Animales , Densidad Ósea/genética , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Remodelación Ósea/genética , Remodelación Ósea/fisiología , Recuento de Células , Diferenciación Celular , Tamaño de la Célula , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Femenino , Regulación Enzimológica de la Expresión Génica , Insulina/sangre , Leptina/sangre , Metabolismo de los Lípidos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoblastos/citología , Osteoblastos/metabolismo , Osteocalcina/sangre , Osteocalcina/genética , Osteocalcina/metabolismo , Esterol Esterasa/genética , Esterol Esterasa/metabolismo , Células del Estroma/citología , Células del Estroma/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA