Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 223
Filtrar
1.
Med Oncol ; 39(2): 26, 2022 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-34982265

RESUMEN

Among the various histologic subtypes of ovarian cancers (OCs), ovarian clear cell carcinoma (OCCC) represents a great challenge due to its disease aggressiveness and resistance to chemotherapy. IGF1 is overexpressed in epithelial ovarian cancer (EOC), and IGF1 pathway activation is related to the chemoresistance of various cancers. In this study, we found that the expression level of IGF1 was higher in OCCC than in the most common type of OC, high-grade serous adenocarcinoma (HGSC). Then, we investigated the role of IGF1 pathway activation in the progression of OCCC, observing that activation of the IGF1 pathway using IGF1 promoted the proliferation and migration of ES2 cells, while inactivation of the IGF1 pathway using the selective IGF1R inhibitor OSI-906 reversed the alteration mediated by IGF1. Based on the role of the IGF1 pathway in cancer chemoresistance, we proposed that OSI-906 may restore the sensitivity of OCCC to cisplatin. We first validated that IGF1 increased the IC50 value of cisplatin in ES2 cells, while OSI-906 decreased it. Then we confirmed that IGF1 decreased the apoptosis rate of ES2 cells induced by cisplatin, while OSI-906 increased it. Finally, we conducted animal experiments to investigate whether OSI-906 helps cisplatin control the growth of OCCC. As expected, OSI-906 increased the effect of cisplatin in attenuating the growth of OCCC in vivo. Therefore, we conclude that using OSI-906 may be an effective method to restore the sensitivity of OCCC to cisplatin by targeting the IGF1R/AKT pathway.


Asunto(s)
Adenocarcinoma de Células Claras/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Cisplatino/uso terapéutico , Imidazoles/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Pirazinas/farmacología , Receptor IGF Tipo 1/efectos de los fármacos , Adenocarcinoma de Células Claras/metabolismo , Adenocarcinoma de Células Claras/patología , Animales , Apoptosis , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Resistencia a Medicamentos , Femenino , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor IGF Tipo 1/metabolismo , Transducción de Señal/efectos de los fármacos
2.
Brain Res Bull ; 175: 205-212, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34333050

RESUMEN

BACKGROUND: Chemotherapy-induced peripheral neuropathy (CIPN) is a common and intractable complication in chemotherapy-receiving patients. Insulin-like growth factor-1 (IGF-1) is a popular neurotrophin with various functions, such as maintaining neuronal survival and synaptic functioning in the central nervous system. Therefore, we hypothesized that the IGF-1 signaling pathway could be a candidate target for treating CIPN. METHODS: We established the CIPN model by injecting mice intraperitoneally with oxaliplatin and assessed IGF-1 protein expression, its receptor IGF1R, phospho-IGF1R (p-IGF1R), interleukin-17A (IL-17A), tumor necrosis factor-α (TNF-α), and calcitonin gene-related peptide (CGRP) in the lumbar spinal cord with Western blot and immunofluorescence. To examine the effect of IGF-1 signaling on CIPN, we injected mice intrathecally or intraperitoneally with mouse recombinant IGF-1 (rIGF-1). RESULTS: IGF-1 protein expression decreased significantly in the spinal cord on D3 and D10 (the 3rd and 10th days after beginning oxaliplatin chemotherapy) and was co-localized with astrocytes primarily in the lumbar spinal cord, whereas IGF1R was predominantly expressed on neurons. Both intrathecally- and intraperitoneally-administered rIGF-1 relieved the chemotherapy-induced pain-like behavior and reduced IL-17A, TNF-α, and CGRP protein expressions in the spinal cord. CONCLUSION: Our results indicate a vital role for IGF-1 signaling in CIPN. Targeting IGF-1 signaling could be a potent therapeutic strategy for treating CIPN in clinical settings.


Asunto(s)
Antineoplásicos/toxicidad , Astrocitos/metabolismo , Factor I del Crecimiento Similar a la Insulina/biosíntesis , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Médula Espinal/metabolismo , Animales , Astrocitos/efectos de los fármacos , Citocinas/metabolismo , Inyecciones Espinales , Factor I del Crecimiento Similar a la Insulina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas , Oxaliplatino/toxicidad , Dolor/psicología , Enfermedades del Sistema Nervioso Periférico/psicología , Receptor IGF Tipo 1/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
3.
Autophagy ; 17(8): 2046-2047, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34110249

RESUMEN

Induction of macroautophagy (hereafter termed autophagy) is a strategy to improve the outcome of antineoplastic therapies by facilitating the induction of immunogenic cancer cell death and the consequent immune recognition of malignant cells. We analyzed 65,000 distinct compounds by means of a phenotypic discovery platform for autophagy induction and identified the IGF1R (insulin like growth factor 1 receptor) inhibitor picropodophyllin (PPP) as a potent inducer of autophagic flux. We found that PPP acts on-target, as an inhibitor of the tyrosine kinase activity of IGF1R and enhances the release of adenosine triphosphate, ATP, from stressed and dying cancer cells in vitro, thereby improving the therapeutic efficacy of chemoimmunotherapy in cancer-bearing mice. This PPP effect was phenocopied by another IGF1R inhibitor, linsitinib. Moreover, in human triple-negative breast cancer, phosphorylation of IGF1R correlates with reduced autophagy, an unfavorable local immune profile and poor prognosis. In summary, IGF1R inhibition may constitute a novel strategy for the treatment of cancer in the context of chemoimmunotherapy.


Asunto(s)
Autofagia/efectos de los fármacos , Imidazoles/farmacología , Podofilotoxina/análogos & derivados , Pirazinas/farmacología , Receptor IGF Tipo 1/efectos de los fármacos , Animales , Proliferación Celular/efectos de los fármacos , Humanos , Podofilotoxina/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Receptor IGF Tipo 1/metabolismo
4.
Cells ; 10(2)2021 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-33673340

RESUMEN

Complex immunological mechanisms underlie the pathogenesis of thyroid-associated ophthalmopathy (TAO). Historical models of Graves' disease and TAO have focused almost entirely on autoimmune reactivity directed against the thyrotropin receptor (TSHR). The insulin-like growth factor-I receptor (IGF-IR) has been proposed as a second participating antigen in TAO by virtue of its interactions with IGFs and anti-IGF-IR antibodies generated in Graves' disease. Furthermore, the IGF-IR forms with TSHR a physical and functional complex which is involved in signaling downstream from both receptors. Inhibition of IGF-IR activity results in attenuation of signaling initiated at either receptor. Based on the aggregate of findings implicating IGF-IR in TAO, the receptor has become an attractive therapeutic target. Recently, teprotumumab, a human monoclonal antibody IGF-IR inhibitor was evaluated in two clinical trials of patients with moderate to severe, active TAO. Those studies revealed that teprotumumab was safe and highly effective in reducing disease activity and severity. Targeting IGF-IR with specific biologic agents may result in a paradigm shift in the therapy of TAO.


Asunto(s)
Anticuerpos Monoclonales Humanizados/efectos de los fármacos , Anticuerpos Monoclonales/uso terapéutico , Oftalmopatía de Graves/tratamiento farmacológico , Receptor IGF Tipo 1/antagonistas & inhibidores , Enfermedad de Graves/tratamiento farmacológico , Enfermedad de Graves/inmunología , Oftalmopatía de Graves/inmunología , Humanos , Receptor IGF Tipo 1/efectos de los fármacos , Receptor IGF Tipo 1/inmunología
5.
Nucleic Acids Res ; 49(2): 700-712, 2021 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-33410883

RESUMEN

Aptamers are single-stranded oligonucleotides that bind to a specific target with high affinity, and are widely applied in biomedical diagnostics and drug development. However, the use of aptamers has largely been limited to simple binders or inhibitors that interfere with the function of a target protein. Here, we show that an aptamer can also act as a positive allosteric modulator that enhances the activation of a receptor by stabilizing the binding of a ligand to that receptor. We developed an aptamer, named IR-A43, which binds to the insulin receptor, and confirmed that IR-A43 and insulin bind to the insulin receptor with mutual positive cooperativity. IR-A43 alone is inactive, but, in the presence of insulin, it potentiates autophosphorylation and downstream signaling of the insulin receptor. By using the species-specific activity of IR-A43 at the human insulin receptor, we demonstrate that residue Q272 in the cysteine-rich domain is directly involved in the insulin-enhancing activity of IR-A43. Therefore, we propose that the region containing residue Q272 is a hotspot that can be used to enhance insulin receptor activation. Moreover, our study implies that aptamers are promising reagents for the development of allosteric modulators that discriminate a specific conformation of a target receptor.


Asunto(s)
Antígenos CD/efectos de los fármacos , Aptámeros de Nucleótidos/farmacología , Receptor de Insulina/efectos de los fármacos , Regulación Alostérica , Animales , Antígenos CD/química , Antígenos CD/metabolismo , Células Cultivadas , Cricetinae , Glutamina/química , Humanos , Insulina/metabolismo , Ratones , Fosforilación , Unión Proteica , Conformación Proteica , Dominios Proteicos , Procesamiento Proteico-Postraduccional , Ratas , Receptor IGF Tipo 1/química , Receptor IGF Tipo 1/efectos de los fármacos , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/química , Receptor de Insulina/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/efectos de los fármacos , Proteínas Recombinantes/metabolismo , Técnica SELEX de Producción de Aptámeros , Estimulación Química
6.
J Neurochem ; 157(3): 695-709, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33258143

RESUMEN

Glucose hypometabolism is observed in epilepsy and promotes epileptogenesis. Glucose hypometabolism in epilepsy may be attributed to decreased neuronal glucose uptake, but its molecular mechanism remains unclear. Zinc-α2-glycoprotein (ZAG) is related to glucose metabolism and is reported to suppress seizures. The anti-epileptic effect of ZAG may be attributed to its regulation of neuronal glucose metabolism. This study explored the effect of ZAG on neuronal glucose uptake and its molecular mechanism via insulin-like growth factor 1 receptor (IGF1R)-regulated glucose transporter 3 (GLUT-3) expression. The ZAG level was modulated by lentivirus in primary culture neurons. Neuronal seizure models were induced by Mg2+ -free artificial cerebrospinal fluid. We assessed neuronal glucose uptake by the 2-NBDG method and Glucose Uptake Colorimetric Assay Kit. IGF1R was activated by IGF1 and blocked by AXL1717. The expression and distribution of IGF1R and GLUT-3, together with IGF1R phosphorylation, were measured by western blot. The binding between ZAG and IGF1R was determined by coimmunoprecipitation. Neuronal glucose uptake and GLUT-3 expression were significantly decreased by seizure or ZAG knockdown, whereas ZAG over-expression or IGF1 treatment reversed this decrease. The effect of ZAG on neuronal glucose uptake and GLUT-3 expression was blocked by AXL1717. ZAG increased IGF1R distribution and phosphorylation possibly by binding. Additionally, IGF1R increased GLUT-3 activity by increasing GLUT-3 expression. In epilepsy/seizure, neuronal glucose uptake suppression may be attributed to a decrease in ZAG, which suppresses neuronal GLUT-3 expression by regulating the activity of IGF1R. ZAG, IGF1R, and GLUT-3 may be novel potential therapeutic targets of glucose hypometabolism in epilepsy and seizures.


Asunto(s)
Adipoquinas/uso terapéutico , Anticonvulsivantes/uso terapéutico , Transportador de Glucosa de Tipo 3/genética , Glucosa/metabolismo , Neuronas/metabolismo , Receptor IGF Tipo 1/efectos de los fármacos , Convulsiones/tratamiento farmacológico , Convulsiones/metabolismo , Adipoquinas/genética , Animales , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Deficiencia de Magnesio/complicaciones , Podofilotoxina/análogos & derivados , Podofilotoxina/farmacología , Embarazo , Cultivo Primario de Células , Ratas
7.
Life Sci ; 259: 118180, 2020 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-32758622

RESUMEN

AIMS: Bufothionine had been used for gastric cancer (GC) treatment, and this study managed to uncover the underlying mechanisms. MATERIALS AND METHODS: Cell proliferation was determined by CCK-8 assay and colony formation assay. Flow cytometry (FCM) and TUNEL assay were used to measure cell apoptosis ratio. Intracellular ROS was measured by DCFH-DA probes. qRT-PCR was used to determine miRNAs levels. Western Blot was performed to probe proteins. Dual-luciferase reporter gene system was employed to validate the binding sites of miR-133a-3p and 3'UTR regions of IGF1R mRNA. Immunohistochemistry (IHC) was used to determine the expressions of Ki-67 in mice tumor tissues. KEY FINDINGS: Bufothionine inhibited cell viability, triggered ER stress and promoted ROS production in GC cells, and both ER stress inhibitor Salburinal (Sal) and ROS scavenger (NAC) abrogated Bufothionine induced GC cell death. Besides, miR-133a-3p was upregulated by Bufothionine, and Bufothionine-induced cell death was enhanced by miR-133a-3p overexpression while alleviated by miR-133a-3p knockdown. Furthermore, miR-133a-3p inactivated PI3K/Akt signal pathway by sponging IGF1R, and Bufothionine inhibited insulin-like growth factor 1 receptor (IGF1R) and inactivated PI3K/Akt cascade by upregulating miR-133a-3p. Notably, the promoting effects of overexpressed miR-133a-3p on Bufothionine-induced GC cell death were abrogated by overexpressing IGF1R, and aggravated by the PI3K/Akt cascade inhibitor (LY294002). SIGNIFICANCE: Bufothionine promoted GC cell death by triggering miR-133a-3p/IGF1R/PI3K/Akt axis mediated ER stress and ROS production.


Asunto(s)
Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/genética , Alcaloides Indólicos/farmacología , MicroARNs/genética , Compuestos de Quinolinio/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Neoplasias Gástricas/patología , Animales , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Proliferación Celular , Cromonas/farmacología , Humanos , Ratones , Ratones Endogámicos BALB C , MicroARNs/biosíntesis , Morfolinas/farmacología , Proteína Oncogénica v-akt/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Receptor IGF Tipo 1/efectos de los fármacos , Ensayo de Tumor de Célula Madre , Regulación hacia Arriba/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Cells ; 9(5)2020 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-32365498

RESUMEN

The insulin-like growth factor (IGF)-axis was implicated in cancer progression and identified as a clinically important therapeutic target. Several IGF-I receptor (IGF-IR) targeting drugs including humanized monoclonal antibodies have advanced to phase II/III clinical trials, but to date, have not progressed to clinical use, due, at least in part, to interference with insulin receptor signaling and compensatory signaling by the insulin receptor (IR) isoform A that can bind IGF-II and initiate mitogenic signaling. Here we briefly review the current state of IGF-targeting biologicals, discuss some factors that may be responsible for their poor performance in the clinic and outline the stepwise bioengineering and validation of an IGF-Trap-a novel anti-cancer therapeutic that could bypass these limitations. The IGF-Trap is a heterotetramer, consisting of the entire extracellular domain of the IGF-IR fused to the Fc portion of human IgG1. It binds human IGF-I and IGF-II with a three-log higher affinity than insulin and could inhibit IGF-IR driven cellular functions such as survival, proliferation and invasion in multiple carcinoma cell models in vitro. In vivo, the IGF-Trap has favorable pharmacokinetic properties and could markedly reduce metastatic outgrowth of colon and lung carcinoma cells in the liver, outperforming IGF-IR and ligand-binding monoclonal antibodies. Moreover, IGF-Trap dose-response profiles correlate with their bio-availability profiles, as measured by the IGF kinase receptor-activation (KIRA) assay, providing a novel, surrogate biomarker for drug efficacy. Our studies identify the IGF-Trap as a potent, safe, anti-cancer therapeutic that could overcome some of the obstacles encountered by IGF-targeting biologicals that have already been evaluated in clinical settings.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Receptor IGF Tipo 1/efectos de los fármacos , Receptor de Insulina/metabolismo , Transducción de Señal/efectos de los fármacos , Anticuerpos Monoclonales/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/efectos de los fármacos , Preparaciones Farmacéuticas/metabolismo , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/efectos de los fármacos
9.
Front Endocrinol (Lausanne) ; 11: 610337, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33391187

RESUMEN

Thyroid-associated ophthalmopathy (TAO) has remained a vexing and poorly managed autoimmune component of Graves' disease where the tissues surrounding the eye and in the upper face become inflamed and undergo remodeling. This leads to substantial facial disfigurement while in its most severe forms, TAO can threaten eye sight. In this brief paper, I review some of the background investigation that has led to development of teprotumumab as the first and only US FDA approved medical therapy for TAO. This novel treatment was predicated on recognition that the insulin-like growth factor I receptor plays an important role in the pathogenesis of TAO. It is possible that a similar involvement of that receptor in other autoimmune disease may lead to additional indications for this and alternative insulin-like growth factor I receptor-inhibiting strategies.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Oftalmopatía de Graves/tratamiento farmacológico , Oftalmopatía de Graves/genética , Humanos , Receptor IGF Tipo 1/efectos de los fármacos , Receptor IGF Tipo 1/genética
10.
J Alzheimers Dis ; 71(3): 825-832, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31450504

RESUMEN

Administration of sevoflurane (SEVO) may induce learning and memory deficits, which increases the chances of developing Alzheimer's disease. Here, we studied the effects of SEVO exposure on rats with a focus on the role of insulin-like growth factor (IGF) signaling. SEVO exposure significantly increased neuron cell apoptosis, and caused poor performance of the rats in behavior tests, by suppressing IGF-1 receptor (IGF1R). Bioinformatic analysis predicted microRNA(miR)-223-3p as an IGF1R-binding miRNA, the level of which increased in neurons after exposure to SEVO. In vitro, miR-223-3p suppressed the translation of IGF1R in neural cells. Moreover, transfection with antisense of miR-223-3p significantly attenuated SEVO exposure-induced neuron cell apoptosis. Taken together, these data suggest that SEVO-induced miR-223-3p upregulation suppresses IGF1R to impair IGF signaling, which subsequently leads to learning and memory impairments.


Asunto(s)
Anestésicos por Inhalación/efectos adversos , Trastornos de la Memoria/inducido químicamente , Memoria/efectos de los fármacos , Receptor IGF Tipo 1/efectos de los fármacos , Sevoflurano/efectos adversos , Animales , Apoptosis/efectos de los fármacos , Biología Computacional , Femenino , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/psicología , MicroARNs/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Desempeño Psicomotor/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba
11.
Brain Res Bull ; 143: 27-35, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30278200

RESUMEN

Depression is associated with uncontrolled diabetes, which indicates a lack of insulin effect, yet the role of the insulin receptor in mediating depression is not clearly established because insulin receptors are not required for glucose entry into the brain. However, insulin receptors are important for brain function since insulin receptor knockout mice have depressive-like activity. Depression and cognitive problems are also associated with low insulin-like growth factor-1 (IGF-1) in the elderly. Rodent studies showed chronic IGF-1 administration had antidepressant-like (AD) activity. We asked if insulin in the brain might act through the IGF-1 receptor, as it does in some tissues. We used acute insulin or IGF-1 infusions into the 3rd ventricle (icv) in rats and tested animals in a forced swim test. We found that antidepressive-like behavior is mediated by insulin and IGF-1. Further, administration of the IGF-1 receptor antagonist JB-1 blocked the antidepressive-like activity of the insulin and IGF-1, indicating a strong relationship between insulin, IGF-1 and depression. Insulin acts at least partially through the IGF-1 receptor and is responsive to receptor antagonism. The model offers promise for future studies of the mechanism of depression, and therapy to increase insulin sensitivity and IGF-1 action including exercise and nutrition.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/farmacología , Insulina/farmacología , Receptor IGF Tipo 1/efectos de los fármacos , Animales , Antidepresivos/metabolismo , Antidepresivos/farmacología , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Depresión/etiología , Trastorno Depresivo/etiología , Fluoxetina/metabolismo , Fluoxetina/farmacología , Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Actividad Motora/efectos de los fármacos , Oligopéptidos/metabolismo , Oligopéptidos/farmacología , Ratas , Ratas Sprague-Dawley , Receptor IGF Tipo 1/metabolismo , Receptor IGF Tipo 1/farmacología , Receptor de Insulina/efectos de los fármacos , Receptor de Insulina/metabolismo , Natación/psicología
12.
Aging Cell ; 17(1)2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29094448

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disorder associated with synaptic dysfunction, pathological accumulation of ß-amyloid (Aß), and neuronal loss. The self-association of Aß monomers into soluble oligomers seems to be crucial for the development of neurotoxicity (J. Neurochem., 00, 2007 and 1172). Aß oligomers have been suggested to compromise neuronal functions in AD by reducing the expression levels of the CREB target gene and brain-derived neurotrophic factor (BDNF) (J. Neurosci., 27, 2007 and 2628; Neurobiol. Aging, 36, 2015 and 20406 Mol. Neurodegener., 6, 2011 and 60). We previously reported a broad neuroprotective activity of physiological Aß monomers, involving the activation of type-1 insulin-like growth factor receptors (IGF-IRs) (J. Neurosci., 29, 2009 and 10582, Front Cell Neurosci., 9, 2015 and 297). We now provide evidence that Aß monomers, by activating the IGF-IR-stimulated PI3-K/AKT pathway, induce the activation of CREB in neurons and sustain BDNF transcription and release.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Neuronas/efectos de los fármacos , Receptor IGF Tipo 1/efectos de los fármacos , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/genética , Humanos , Neuronas/metabolismo , Fragmentos de Péptidos/metabolismo , Ratas , Receptor IGF Tipo 1/genética , Receptores de Somatomedina/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
13.
Med Princ Pract ; 26(4): 343-350, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28273667

RESUMEN

OBJECTIVE: The aim of this study was to understand whether or not the protective effect of green tea after fasting-induced damage in the jejunal mucosa of rat is dependent on cell proliferation and the stimulation of specific growth factors. MATERIALS AND METHODS: Sixty adult male Wistar rats were used in this study. The animals were divided randomly into 5 groups, with 12 in each group (G1-5). The animals in G1 (control group) were fed a rat chow diet and water ad libitum. The animals in G2 (fasting group) were fasted for 3 days. The animals in the G3, G4, and G5 groups were fasted for 3 days as G2, but were given water (G3), green tea (G4), or a vitamin E (G5) solution, respectively, for another 7 days. The animals were euthanized, and the jejunum was removed and processed for histological and immunohistochemical analysis. RESULTS: Compared to the G3 group, the jejunal mucosa of G4 rats showed a 70.6% higher level (p < 0.001) of expression of proliferating cell nuclear antigen and 98% higher level (p = 0.0001) of the expression of transforming growth factor-ß1 (TGF-ß1), whereas the level of fibroblast growth factor-1 (FGF-1) and insulin-like growth factor-1 (IGF-1) expression was 22 and 11% lower, respectively, in G4 animals as compared to G3 rats. These differences in the expression of FGF-1 and IGF-1 in G4 animals were not statistically significant. CONCLUSION: In this study, green tea repaired the fasting-induced damage in the jejunal mucosa of rats, mainly by inducing a significant expression of TGF-ß1 in the jejunal mucosa.


Asunto(s)
Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/efectos de los fármacos , Receptor IGF Tipo 1/efectos de los fármacos , Té/metabolismo , Factor de Crecimiento Transformador beta1/efectos de los fármacos , Análisis de Varianza , Animales , Proliferación Celular , Eutanasia Animal , Ayuno , Inmunoquímica , Mucosa Intestinal/efectos de los fármacos , Masculino , Distribución Aleatoria , Ratas , Ratas Wistar
14.
Oncotarget ; 7(31): 50258-50276, 2016 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-27384680

RESUMEN

In this work two acetylene alcohols, compound 1 and compound 2, which were isolated and identified from the sponge Cribrochalina vasculum, and which showed anti-tumor effects were further studied with respect to targets and action mechanisms. Gene expression analyses suggested insulin like growth factor receptor (IGF-1R) signaling to be instrumental in controlling anti-tumor efficacy of these compounds in non-small cell lung cancer (NSCLC). Indeed compounds 1 and 2 inhibited phosphorylation of IGF-1Rß as well as reduced its target signaling molecules IRS-1 and PDK1 allowing inhibition of pro-survival signaling. In silico docking indicated that compound 1 binds to the kinase domain of IGF-1R at the same binding site as the well known tyrosine kinase inhibitor AG1024. Indeed, cellular thermal shift assay (CETSA) confirmed that C. vasculum compound 1 binds to IGF-1R but not to the membrane localized tyrosine kinase receptor EGFR. Importantly, we demonstrate that compound 1 causes IGF-1Rß but not Insulin Receptor degradation specifically in tumor cells with no effects seen in normal diploid fibroblasts. Thus, these compounds hold potential as novel therapeutic agents targeting IGF-1R signaling for anti-tumor treatment.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Neoplasias Pulmonares/metabolismo , Poríferos/química , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/metabolismo , Animales , Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Línea Celular Tumoral , Supervivencia Celular , Receptores ErbB/metabolismo , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas Sustrato del Receptor de Insulina/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Fosforilación , Unión Proteica , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor IGF Tipo 1/efectos de los fármacos , Receptor de Insulina/efectos de los fármacos , Transducción de Señal , Tirfostinos/farmacología
15.
Chin J Physiol ; 59(2): 109-18, 2016 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-27080466

RESUMEN

Risk factors for prostate cancer (PCa) include age, hormones, race, family history and diet. Recently, epidemiologic evidence has indicated that history of diabetes mellitus (DM) is inversely associated with risk of PCa. However, epidemiological investigations have yielded inconsistent results. Hence, the exact mechanism of DM-induced reduction in the incidence of PCa has yet to be fully elucidated. The aim of this study was to investigate the effects of DM factors, including glucose, insulin and insulin-like growth factor-1 (IGF-1), on the proliferation of PCa cell lines in vitro. Cell proliferation and expression of hormone receptors was examined in MTT assay and Western blot analysis, respectively. The results showed that DM factors did not affect the viability of androgen receptor (AR)-expressing PCa cell lines. However, cell proliferation increased after treatment with DM factors in androgen-independent PCa cell lines. On PCa tissue arrays, intensities of total AR and nuclear IGF-1R were higher in malignant tissues than in normal prostate glands. In terms of hormonal receptors, androgen-dependent LNCaP cells treated with insulin and IGF-1 in a low-serum medium showed decreased expression of insulin receptor beta (IRß) and elevated expression of IGF-1 receptor beta (IGF-1Rß). Moreover, expression of AR was upregulated after insulin and IGF-1 treatment in LNCaP cells, but not in the other PCa cell lines. Most of the studied antidiabetic drugs promoted the viability of PCa cells. However, metformin decreased the viability of AR-expressing PCa cells. These results suggest that diabetic factors modify the expression of AR, IR and IGF-1R to increase cancer cell proliferation. Moreover, the growth suppressing effects of metformin on PCa may be via the regulation of the AR signaling pathway.


Asunto(s)
Diabetes Mellitus/fisiopatología , Hipoglucemiantes/farmacología , Neoplasias de la Próstata/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Glucosa/farmacología , Humanos , Inmunohistoquímica , Insulina/farmacología , Factor I del Crecimiento Similar a la Insulina/farmacología , Masculino , Neoplasias de la Próstata/fisiopatología , Receptor IGF Tipo 1/biosíntesis , Receptor IGF Tipo 1/efectos de los fármacos , Receptor de Insulina/biosíntesis , Receptor de Insulina/efectos de los fármacos , Receptores Androgénicos/efectos de los fármacos , Receptores Androgénicos/metabolismo , Transducción de Señal/efectos de los fármacos
16.
Gut Liver ; 10(2): 310-7, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26470769

RESUMEN

BACKGROUND/AIMS: Statins act as antineoplastic agents through the inhibition of cell proliferation. This study sought to demonstrate the effects of statins on extrahepatic bile duct cancer cell apoptosis and to document the changes in protein expression involved in tumor growth and suppression. METHODS: Human extrahepatic bile duct cancer cells were cultured. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays were performed to determine the effect of statins on cell proliferation. Apoptosis was measured by a cell death detection enzyme-linked immunosorbent assay and caspase-3 activity assay, and flow cytometry was used to determine the percentage of cells in each phase of the cell cycle. The protein expression of Bax, Bcl-2, insulin-like growth factor 1 (IGF-1) receptor, extracellular signal-regulated kinase 1/2 (ERK1/2), and Akt was measured by Western blot analysis. RESULTS: Simvastatin suppressed cell proliferation by inducing G1 phase cell cycle arrest in bile duct cancer cells. Furthermore, it induced apoptosis via caspase-3 activation, downregulated the expression of the Bcl-2 protein, and enhanced the expression of the Bax protein. Moreover, simvastatin suppressed the expression of the IGF-1 receptor and IGF-1-induced ERK/Akt activation. CONCLUSIONS: Simvastatin induces apoptosis in bile duct cancer cells, which suggests that it could be an antineoplastic agent for bile duct cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Hipolipemiantes/farmacología , Receptor IGF Tipo 1/efectos de los fármacos , Simvastatina/farmacología , Antineoplásicos/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos
17.
Endocrinology ; 156(7): 2395-408, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25885794

RESUMEN

Medulloblastoma (Med) is the most common malignant brain tumor in children. The role of ESR2 [estrogen receptor (ER)-ß] in promoting Med growth was comprehensively examined in three in vivo models and human cell lines. In a novel Med ERß-null knockout model developed by crossing Esr2(-/-) mice with cerebellar granule cell precursor specific Ptch1 conditional knockout mice, the tumor growth rate was significantly decreased in males and females. The absence of Esr2 resulted in increased apoptosis, decreased B-cell lymphoma 2 (BCL2), and IGF-1 receptor (IGF1R) expression, and decreased levels of active MAPKs (ERK1/2) and protein kinase B (AKT). Treatment of Med in Ptch1(+/-) Trp53(-/-) mice with the antiestrogen chemotherapeutic drug Faslodex significantly increased symptom-free survival, which was associated with increased apoptosis and decreased BCL2 and IGF1R expression and signaling. Similar effects were also observed in nude mice bearing D283Med xenografts. In vitro studies in human D283Med cells metabolically stressed by glutamine withdrawal found that 17ß-estradiol and the ERß selective agonist 2,3-bis(4-hydroxyphenyl)-propionitrile dose dependently protected Med cells from caspase-3-dependent cell death. Those effects were associated with increased phosphorylation of IGF1R, long-term increases in ERK1/2 and AKT signaling, and increased expression of IGF-1, IGF1R, and BCL2. Results of pharmacological experiments revealed that the cytoprotective actions of estradiol were dependent on ERß and IGF1R receptor tyrosine kinase activity and independent of ERα and G protein-coupled estrogen receptor 1 (G protein coupled receptor 30). The presented results demonstrate that estrogen promotes Med growth through ERß-mediated increases in IGF1R expression and activity, which induce cytoprotective mechanisms that decrease apoptosis.


Asunto(s)
Apoptosis/genética , Receptor beta de Estrógeno/genética , Regulación Neoplásica de la Expresión Génica/genética , Meduloblastoma/genética , ARN Mensajero/metabolismo , Receptor IGF Tipo 1/genética , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Línea Celular Tumoral , Estradiol/farmacología , Receptor beta de Estrógeno/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Factor I del Crecimiento Similar a la Insulina/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Factor II del Crecimiento Similar a la Insulina/efectos de los fármacos , Factor II del Crecimiento Similar a la Insulina/genética , Factor II del Crecimiento Similar a la Insulina/metabolismo , Masculino , Meduloblastoma/metabolismo , Ratones , Ratones Noqueados , Receptores Patched , Receptor Patched-1 , Proteínas Proto-Oncogénicas c-bcl-2/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Mensajero/efectos de los fármacos , Receptor IGF Tipo 1/efectos de los fármacos , Receptor IGF Tipo 1/metabolismo , Receptores de Superficie Celular/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína p53 Supresora de Tumor/genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
18.
Mol Carcinog ; 54 Suppl 1: E129-37, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25163779

RESUMEN

Insulin-like growth factor-1 receptor (IGF-1R) is a transmembrane heterotetramer that is activated by Insulin-like growth factor 1 and is crucial for tumor transformation and survival of malignant cells. Importantly, IGF-1R overexpression has been reported in many different cancers, implicating this receptor as a potential target for anticancer therapy. Picropodophyllin (PPP) is a potent inhibitor of IGF-1R and has antitumor efficacy in several cancer types. However, the chemopreventive effect of PPP in lung tumorigenesis has not been investigated. In this study, we investigated the chemopreventive activity of PPP in a mouse lung tumor model. Benzo(a)pyrene was used to induce lung tumors, and PPP was given by nasal inhalation to female A/J mice. Lung tumorigenesis was assessed by tumor multiplicity and tumor load. PPP significantly decreased tumor multiplicity and tumor load. Tumor multiplicity and load were decreased by 52% and 78% respectively by 4 mg/ml aerosolized PPP. Pharmacokinetics analysis showed good bioavailability of PPP in lung and plasma. Treatment with PPP increased staining for cleaved caspase-3 and decreased Ki-67 in lung tumors, suggesting that the lung tumor inhibitory effects of PPP were partially through inhibition of proliferation and induction of apoptosis. In human lung cancer cell lines, PPP inhibited cell proliferation, and also inhibited phosphorylation of IGF-1R downstream targets, AKT and MAPK, ultimately resulting in increased apoptosis. PPP also reduced cell invasion in lung cancer cell lines. In view of our data, PPP merits further investigation as a promising chemopreventive agent for human lung cancer.


Asunto(s)
Anticarcinógenos/uso terapéutico , Neoplasias Pulmonares/prevención & control , Podofilotoxina/análogos & derivados , Receptor IGF Tipo 1/efectos de los fármacos , Animales , Anticarcinógenos/farmacología , Apoptosis/efectos de los fármacos , Benzo(a)pireno/toxicidad , Carcinógenos/toxicidad , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Neoplasias Pulmonares/inducido químicamente , Neoplasias Pulmonares/patología , Ratones , Podofilotoxina/farmacología , Podofilotoxina/uso terapéutico
20.
Nat Med ; 20(9): 1027-34, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25173427

RESUMEN

Crizotinib, a selective tyrosine kinase inhibitor (TKI), shows marked activity in patients whose lung cancers harbor fusions in the gene encoding anaplastic lymphoma receptor tyrosine kinase (ALK), but its efficacy is limited by variable primary responses and acquired resistance. In work arising from the clinical observation of a patient with ALK fusion-positive lung cancer who had an exceptional response to an insulin-like growth factor 1 receptor (IGF-1R)-specific antibody, we define a therapeutic synergism between ALK and IGF-1R inhibitors. Similar to IGF-1R, ALK fusion proteins bind to the adaptor insulin receptor substrate 1 (IRS-1), and IRS-1 knockdown enhances the antitumor effects of ALK inhibitors. In models of ALK TKI resistance, the IGF-1R pathway is activated, and combined ALK and IGF-1R inhibition improves therapeutic efficacy. Consistent with this finding, the levels of IGF-1R and IRS-1 are increased in biopsy samples from patients progressing on crizotinib monotherapy. Collectively these data support a role for the IGF-1R-IRS-1 pathway in both ALK TKI-sensitive and ALK TKI-resistant states and provide a biological rationale for further clinical development of dual ALK and IGF-1R inhibitors.


Asunto(s)
Neoplasias Pulmonares/tratamiento farmacológico , Pirazoles/uso terapéutico , Piridinas/uso terapéutico , Proteínas Tirosina Quinasas Receptoras/efectos de los fármacos , Receptor IGF Tipo 1/efectos de los fármacos , Quinasa de Linfoma Anaplásico , Crizotinib , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Proteínas Sustrato del Receptor de Insulina/genética , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/metabolismo , Persona de Mediana Edad , Pirazoles/farmacología , Piridinas/farmacología , Receptor IGF Tipo 1/metabolismo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA