Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 988
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
J Pathol ; 259(2): 205-219, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36373776

RESUMO

Colorectal cancer (CRC) is one of the most common cancers worldwide. The tumor microenvironment exerts crucial effects in driving CRC progression. Cancer-associated fibroblasts (CAFs) serve as one of the most important tumor microenvironment components promoting CRC progression. This study aimed to elucidate the novel molecular mechanisms of CAF-secreted insulin-like growth factor (IGF) 2 in colorectal carcinogenesis. Our results indicated that IGF2 was a prominent factor upregulated in CAFs compared with normal fibroblasts. CAF-derived conditioned media (CM) promoted tumor growth, migration, and invasion of HCT 116 and DLD-1 cells. IGF1R expression is significantly increased in CRC, serving as a potent receptor in response to IGF2 stimulation and predicting unfavorable outcomes for CRC patients. Apart from the PI3K-AKT pathway, RNA-seq analysis revealed that the YAP1-target signature serves as a prominent downstream effector to mediate the oncogenic signaling of IGF2-IGF1R. By single-cell RNA sequencing (scRNA-seq) and immunohistochemical validation, IGF2 was found to be predominantly secreted by CAFs, whereas IGF1R was expressed mainly by cancer cells. IGF2 triggers the nuclear accumulation of YAP1 and upregulates YAP1 target signatures; however, these effects were abolished by either IGF1R knockdown or inhibition with picropodophyllin (PPP), an IGF1R inhibitor. Using CRC organoid and in vivo studies, we found that cotargeting IGF1R and YAP1 with PPP and verteporfin (VP), a YAP1 inhibitor, enhanced antitumor effects compared with PPP treatment alone. In conclusion, this study revealed a novel molecular mechanism by which CAFs promote CRC progression. The findings highlight the translational potential of the IGF2-IGF1R-YAP1 axis as a prognostic biomarker and therapeutic target for CRC. © 2022 The Pathological Society of Great Britain and Ireland.


Assuntos
Fibroblastos Associados a Câncer , Neoplasias Colorretais , Humanos , Fibroblastos Associados a Câncer/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Linhagem Celular Tumoral , Transdução de Sinais , Carcinogênese/patologia , Neoplasias Colorretais/patologia , Proliferação de Células , Microambiente Tumoral , Fator de Crescimento Insulin-Like II/genética , Fator de Crescimento Insulin-Like II/metabolismo , Fator de Crescimento Insulin-Like II/farmacologia , Receptor IGF Tipo 1/metabolismo , Receptor IGF Tipo 1/farmacologia
2.
J Am Chem Soc ; 145(44): 24272-24283, 2023 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-37899626

RESUMO

Targeted degradation of the cell-surface and extracellular proteins via the endogenous lysosomal degradation pathways, such as lysosome-targeting chimeras (LYTACs), has recently emerged as an attractive tool to expand the scope of extracellular chemical biology. Herein, we report a series of recombinant proteins genetically fused to insulin-like growth factor 2 (IGF2), which we termed iLYTACs, that can be conveniently obtained in high yield by standard cloning and bacterial expression in a matter of days. We showed that both type-I iLYTACs, in which IGF2 was fused to a suitable affibody or nanobody capable of binding to a specific protein target, and type-II iLYTAC (or IGF2-Z), in which IGF2 was fused to the IgG-binding Z domain that served as a universal antibody-binding adaptor, could be used for effective lysosomal targeting and degradation of various extracellular and membrane-bound proteins-of-interest. These heterobifunctional iLYTACs are fully genetically encoded and can be produced on a large scale from conventional E. coli expression systems without any form of chemical modification. In the current study, we showed that iLYTACs successfully facilitated the cell uptake, lysosomal localization, and efficient lysosomal degradation of various disease-relevant protein targets from different mammalian cell lines, including EGFR, PD-L1, CD20, and α-synuclein. The antitumor properties of iLYTACs were further validated in a mouse xenograft model. Overall, iLYTACs represent a general and modular strategy for convenient and selective targeted protein degradation, thus expanding the potential applications of current LYTACs and related techniques.


Assuntos
Escherichia coli , Proteínas de Membrana , Humanos , Camundongos , Animais , Proteínas de Membrana/metabolismo , Escherichia coli/metabolismo , Transdução de Sinais , Lisossomos/metabolismo , Linhagem Celular , Mamíferos/metabolismo , Fator de Crescimento Insulin-Like II/metabolismo , Fator de Crescimento Insulin-Like II/farmacologia
3.
J Biochem Mol Toxicol ; 37(3): e23271, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36510830

RESUMO

Atherosclerosis (AS) is one of the principal causes of cardiovascular disorder. Reportedly, vascular smooth muscle cells (VSMCs) and human umbilical vein endothelial cells (HUVECs) play key roles in AS development, and microRNAs (miRNAs) regulate their functions. The function of miR-216b-5p in AS remains unknown. Human VSMCs and human HUVECs were treated with ox-LDL to establish the in vitro model of AS. MiR-216b-5p and IGF2 expressions in VSMCs and HUVECs were probed by qRT-PCR and western blot. The viability, cell cycle progression, and apoptosis of VSMCs and HUVECs were evaluated by Cell Counting Kit-8 (CCK-8), 5-ethynyl-2'-deoxyuridine, and flow cytometry assays, respectively. The binding sites between IGF2 3'UTR and miR-216b-5p were validated by dual-luciferase reporter assay. miR-216b-5p expression was declined in ox-LDL-induced VSMCs and HUVECs. In VSMCs, miR-216b-5p overexpression inhibited excessive proliferation and induced apoptosis. MiR-216b-5p could markedly restrain the viabiblity of VSMCs induced by ox-LDL and enhanced the viability of HUVECs. Additionally, IGF2 was confirmed as the direct target of miR-216b-5p and transfection of IGF2 overexpression plasmids rescued the effects of miR-216b-5p on VSMCs and HUVECs. miR-216b-5p alleviates the dysfunction of VSMCs and HUVECs caused by ox-LDL via repressing IGF2, and exerts protective functions to block the development of AS.


Assuntos
Aterosclerose , MicroRNAs , Humanos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Músculo Liso Vascular/metabolismo , MicroRNAs/metabolismo , Lipoproteínas LDL/metabolismo , Aterosclerose/metabolismo , Apoptose , Divisão Celular , Proliferação de Células , Fator de Crescimento Insulin-Like II/metabolismo , Fator de Crescimento Insulin-Like II/farmacologia
4.
Phytother Res ; 37(9): 3964-3981, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37186468

RESUMO

Doxorubicin (DOX), an effective chemotherapeutic drug, has been used to treat various cancers; however, its cardiotoxic side effects restrict its therapeutic efficacy. Fisetin, a flavonoid phytoestrogen derived from a range of fruits and vegetables, has been reported to exert cardioprotective effects against DOX-induced cardiotoxicity; however, the underlying mechanisms remain unclear. This study investigated fisetin's cardioprotective role and mechanism against DOX-induced cardiotoxicity in H9c2 cardiomyoblasts and ovariectomized (OVX) rat models. MTT assay revealed that fisetin treatment noticeably rescued DOX-induced cell death in a dose-dependent manner. Moreover, western blotting and TUNEL-DAPI staining showed that fisetin significantly attenuated DOX-induced cardiotoxicity in vitro and in vivo by inhibiting the insulin-like growth factor II receptor (IGF-IIR) apoptotic pathway through estrogen receptor (ER)-α/-ß activation. The echocardiography, biochemical assay, and H&E staining results demonstrated that fisetin reduced DOX-induced cardiotoxicity by alleviating cardiac dysfunction, myocardial injury, oxidative stress, and histopathological damage. These findings imply that fisetin has a significant therapeutic potential against DOX-induced cardiotoxicity.


Assuntos
Cardiotoxicidade , Fator de Crescimento Insulin-Like II , Ratos , Animais , Cardiotoxicidade/tratamento farmacológico , Fator de Crescimento Insulin-Like II/metabolismo , Fator de Crescimento Insulin-Like II/farmacologia , Fator de Crescimento Insulin-Like II/uso terapêutico , Receptores de Estrogênio/metabolismo , Doxorrubicina/efeitos adversos , Estresse Oxidativo , Miócitos Cardíacos , Apoptose
5.
Growth Factors ; 40(5-6): 254-271, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36206173

RESUMO

Methanolic crude extract of Scoparia dulcis (CESD) was orally administered to female mice during the early gestation (day 4-day 8) at a dose of 500 mg/kg/day. It induces embryo resorption and morphological changes of fetal maternal tissue. Histomorphology was studied by routine hematoxylin eosin stain. In situ immunofluorescence localization of IGF-II using Texas red showed an ordered expression of the growth factor in the maternal decidual cells, trophoblast cells and the embryo. Western blot analysis showed a gradual increase of IGF-II from D4 to D8 of control females. In contrast, the CESD-treated females showed resorption of embryo on D8 with disorganized in situ expression and lowered IGF-II in fetal maternal tissue. The phytocompounds present in the CESD could modulate either the ER or IGF-II receptors causing reduced IGF-II expression in the target tissues which lead to the failure of embryonic growth during periimplantation.


Assuntos
Fator de Crescimento Insulin-Like II , Extratos Vegetais , Trofoblastos , Animais , Feminino , Camundongos , Gravidez , Transtornos do Crescimento/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Fator de Crescimento Insulin-Like II/genética , Fator de Crescimento Insulin-Like II/metabolismo , Fator de Crescimento Insulin-Like II/farmacologia , Trofoblastos/metabolismo , Extratos Vegetais/farmacologia , Scoparia/química
6.
Biol Reprod ; 104(3): 602-610, 2021 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-33348377

RESUMO

Insulin-like growth factors (IGFs) are known for their involvement in endocrine and paracrine regulation of ovarian function. Although IGF2 is the predominant circulating and intraovarian form of IGFs in primate species, the stage-specific follicular expression, action, and regulation of IGF2 are not well defined. Therefore, experiments were conducted to investigate the follicular IGF production in response to steroid hormone regulation and the direct IGF actions on follicular development and function in vitro. Preantral follicles were isolated from rhesus macaque ovaries and cultured to the antral stage in media supplemented with follicle-stimulating hormone and insulin. Follicles were randomly assigned to treatment groups: (a) control, (b) trilostane (a steroid synthesis inhibitor), (c) trilostane + estradiol, (d) trilostane + progesterone, and (e) trilostane + dihydrotestosterone. Media was analyzed for IGF concentrations, which were correlated to follicle growth. Follicles produced IGF2, but not IGF1, at the antral stage. Steroid depletion decreased, whereas steroid replacement increased, IGF2 production by antral follicles. Media IGF2 levels correlated positively with antral follicle diameters. Macaque preantral follicles and granulosa cells were subsequently cultured without (control) and with recombinant human IGF2 supplementation. Follicle survival, growth, and paracrine factor production, as well as granulosa cell proliferation and gonadotropin receptor gene expression, were assessed. IGF2 addition increased follicle survival rates, diameters and inhibin B production, as well as granulosa cell proliferation. These data demonstrate that IGF2 produced by antral follicles, in response to steroid hormone regulation, could act as a paracrine factor that positively impacts preantral follicle development and function in primates.


Assuntos
Fator de Crescimento Insulin-Like II/metabolismo , Fator de Crescimento Insulin-Like II/farmacologia , Folículo Ovariano/crescimento & desenvolvimento , Folículo Ovariano/metabolismo , Animais , Relação Dose-Resposta a Droga , Estradiol/análogos & derivados , Estradiol/farmacologia , Feminino , Hormônio Foliculoestimulante/farmacologia , Fator de Crescimento Insulin-Like II/administração & dosagem , Macaca mulatta , Progesterona/farmacologia , Técnicas de Cultura de Tecidos
7.
Acta Neuropathol ; 140(5): 737-764, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32642868

RESUMO

Impaired neuronal proteostasis is a salient feature of many neurodegenerative diseases, highlighting alterations in the function of the endoplasmic reticulum (ER). We previously reported that targeting the transcription factor XBP1, a key mediator of the ER stress response, delays disease progression and reduces protein aggregation in various models of neurodegeneration. To identify disease modifier genes that may explain the neuroprotective effects of XBP1 deficiency, we performed gene expression profiling of brain cortex and striatum of these animals and uncovered insulin-like growth factor 2 (Igf2) as the major upregulated gene. Here, we studied the impact of IGF2 signaling on protein aggregation in models of Huntington's disease (HD) as proof of concept. Cell culture studies revealed that IGF2 treatment decreases the load of intracellular aggregates of mutant huntingtin and a polyglutamine peptide. These results were validated using induced pluripotent stem cells (iPSC)-derived medium spiny neurons from HD patients and spinocerebellar ataxia cases. The reduction in the levels of mutant huntingtin was associated with a decrease in the half-life of the intracellular protein. The decrease in the levels of abnormal protein aggregation triggered by IGF2 was independent of the activity of autophagy and the proteasome pathways, the two main routes for mutant huntingtin clearance. Conversely, IGF2 signaling enhanced the secretion of soluble mutant huntingtin species through exosomes and microvesicles involving changes in actin dynamics. Administration of IGF2 into the brain of HD mice using gene therapy led to a significant decrease in the levels of mutant huntingtin in three different animal models. Moreover, analysis of human postmortem brain tissue and blood samples from HD patients showed a reduction in IGF2 level. This study identifies IGF2 as a relevant factor deregulated in HD, operating as a disease modifier that buffers the accumulation of abnormal protein species.


Assuntos
Doença de Huntington/metabolismo , Doença de Huntington/patologia , Fator de Crescimento Insulin-Like II/metabolismo , Agregação Patológica de Proteínas/metabolismo , Animais , Humanos , Fator de Crescimento Insulin-Like II/farmacologia , Camundongos , Camundongos Transgênicos , Agregados Proteicos/efeitos dos fármacos
8.
Clin Exp Ophthalmol ; 48(9): 1261-1275, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33026147

RESUMO

BACKGROUND: Retinopathy of prematurity (ROP), a major cause of significant visual morbidity and blindness in preterm infants, is closely related to pathological angiogenesis. The aim of the study is to evaluate the effect of a new 12-aa peptide (named peptide CW-703) from human insulin-like growth factor-2, against angiogenesis in ROP. METHODS: In order to evaluate the inhibitory effect of CW-703 on the proliferation, migration, tube formation and apoptosis of human umbilical vein endothelial cells (ScienCell) in vitro, we used MTS assays, a modified Boyden chamber, Matrigel system and TUNEL assays. Effects in vivo were assayed using chorioallantoic membrane assays and oxygen-induced retinopathy (OIR) models in mice. We also performed eletrophysiological and histologic examinations to evaluate the possible toxicity of the peptide. Real-time PCR, ELISA and western blotting were used to elucidate the mechanism of CW-703. RESULTS: CW-703 inhibited angiogenesis in vitro by suppressing endothelial cell proliferation, migration and tube formation. CW-703 also prevented angiogenesis in chicken chorioallantoic membrane assays and OIR assays in mice. No evident functional or morphologic abnormalities in neuroretina after CW-703 injection were revealed in electrophysiological tests and histological examinations. Moreover, we elucidated that CW-703 competed for binding to IGF-1R and inhibited angiogenesis by inhibiting IGF-1R/PI3K/AKT activation and downregulating vascular endothelial growth factor expression. CONCLUSION: The novel peptide CW-703 may act as an effective inhibitor of ocular pathologic angiogenesis, especially in treating ROP.


Assuntos
Neovascularização Retiniana , Inibidores da Angiogênese/uso terapêutico , Animais , Proliferação de Células , Modelos Animais de Doenças , Células Endoteliais da Veia Umbilical Humana , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Fator de Crescimento Insulin-Like II/farmacologia , Fator de Crescimento Insulin-Like II/uso terapêutico , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica , Oxigênio/uso terapêutico , Oxigênio/toxicidade , Fosfatidilinositol 3-Quinases/farmacologia , Fosfatidilinositol 3-Quinases/uso terapêutico , Neovascularização Retiniana/induzido quimicamente , Neovascularização Retiniana/tratamento farmacológico , Neovascularização Retiniana/prevenção & controle , Fator A de Crescimento do Endotélio Vascular
9.
Environ Toxicol ; 35(10): 1043-1049, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32415908

RESUMO

In this study, we used ICI 182 780 (ICI), an estrogen receptor (ER) antagonist, to investigate the estrogenic activity of Danshen, and to further explored whether Danshen extract can block Leu27IGF-II-induced hypertrophy in H9c2 cardiomyoblast cells. We first used an IGF-II analog Leu27IGF-II, which specifically activates IGF2R signaling cascades and induces H9c2 cardiomyoblast cell hypertrophy. However, Danshen extract completely inhibited Leu27IGF-II-induced cell size increase, ANP and BNP hypertrophic marker expression, and IGF2R induction. We also observed that Danshen extract inhibited calcineurin protein expression and NFAT3 nuclear translocation, leading to suppression of Leu27IGF-II-induced cardiac hypertrophy. Moreover, the anti-Leu27IGF-II-IGF2R signaling effect of Danshen was totally reversed by ICI, which suggest the cardio protective effect of Danshen is mediated through estrogen receptors. Our study suggests that, Danshen exerts estrogenic activity, and thus, it could be used as a selective ER modulator in IGFIIR induced hypertrophy model.


Assuntos
Crescimento Celular/efeitos dos fármacos , Medicamentos de Ervas Chinesas/farmacologia , Fator de Crescimento Insulin-Like II/análogos & derivados , Mioblastos Cardíacos/efeitos dos fármacos , Receptor IGF Tipo 2/metabolismo , Salvia miltiorrhiza/química , Animais , Calcineurina/metabolismo , Cardiomegalia/prevenção & controle , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Medicamentos de Ervas Chinesas/isolamento & purificação , Antagonistas do Receptor de Estrogênio/farmacologia , Fulvestranto/farmacologia , Fator de Crescimento Insulin-Like II/farmacologia , Mioblastos Cardíacos/metabolismo , Mioblastos Cardíacos/patologia , Transporte Proteico , Ratos , Receptores de Estrogênio/metabolismo , Transdução de Sinais
10.
J Neurosci ; 38(4): 1015-1029, 2018 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-29217683

RESUMO

Autism spectrum disorder (ASD) is a developmental disability characterized by impairments in social interaction and repetitive behavior, and is also associated with cognitive deficits. There is no current treatment that can ameliorate most of the ASD symptomatology; thus, identifying novel therapies is urgently needed. We used male BTBR T+Itpr3tf /J (BTBR) mice, a model that reproduces most of the core behavioral phenotypes of ASD, to test the effects of systemic administration of insulin-like growth factor II (IGF-II), a polypeptide that crosses the blood-brain barrier and acts as a cognitive enhancer. We show that systemic IGF-II treatments reverse the typical defects in social interaction, cognitive/executive functions, and repetitive behaviors reflective of ASD-like phenotypes. In BTBR mice, IGF-II, via IGF-II receptor, but not via IGF-I receptor, reverses the abnormal levels of the AMPK-mTOR-S6K pathway and of active translation at synapses. Thus, IGF-II may represent a novel potential therapy for ASD.SIGNIFICANCE STATEMENT Currently, there is no effective treatment for autism spectrum disorder (ASD), a developmental disability affecting a high number of children. Using a mouse model that expresses most of the key core as well as associated behavioral deficits of ASD, that are, social, cognitive, and repetitive behaviors, we report that a systemic administration of the polypeptide insulin-like growth factor II (IGF-II) reverses all these deficits. The effects of IGF-II occur via IGF-II receptors, and not IGF-I receptors, and target both basal and learning-dependent molecular abnormalities found in several ASD mice models, including those of identified genetic mutations. We suggest that IGF-II represents a potential novel therapeutic target for ASD.


Assuntos
Transtorno do Espectro Autista/metabolismo , Fator de Crescimento Insulin-Like II/farmacologia , Serina-Treonina Quinases TOR/metabolismo , Animais , Fator de Crescimento Insulin-Like II/metabolismo , Masculino , Camundongos , Fenótipo , Receptor IGF Tipo 2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
11.
Development ; 143(11): 1859-73, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27068108

RESUMO

We investigate the roles of mTor signaling in the formation of Müller glia-derived progenitor cells (MGPCs) in the chick retina. During embryonic development, pS6 (a readout of active mTor signaling) is present in early-stage retinal progenitors, differentiating amacrine and ganglion cells, and late-stage progenitors or maturing Müller glia. By contrast, pS6 is present at low levels in a few scattered cell types in mature, healthy retina. Following retinal damage, in which MGPCs are known to form, mTor signaling is rapidly activated in Müller glia. Inhibition of mTor in damaged retinas prevented the accumulation of pS6 in Müller glia and reduced numbers of proliferating MGPCs. Inhibition of mTor had no effect on MAPK signaling or on upregulation of the stem cell factor Klf4, whereas Pax6 upregulation was significantly reduced. Inhibition of mTor potently blocked the MGPC-promoting effects of Hedgehog, Wnt and glucocorticoid signaling in damaged retinas. In the absence of retinal damage, insulin, IGF1 and FGF2 induced pS6 in Müller glia, and this was blocked by mTor inhibitor. In FGF2-treated retinas, in which MGPCs are known to form, inhibition of mTor blocked the accumulation of pS6, the upregulation of Pax6 and the formation of proliferating MGPCs. We conclude that mTor signaling is required, but not sufficient, to stimulate Müller glia to give rise to proliferating progenitors, and the network of signaling pathways that drive the formation of MGPCs requires activation of mTor.


Assuntos
Células Ependimogliais/citologia , Neuroglia/citologia , Retina/metabolismo , Transdução de Sinais , Células-Tronco/citologia , Serina-Treonina Quinases TOR/metabolismo , Animais , Movimento Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Galinhas , Células Ependimogliais/efeitos dos fármacos , Células Ependimogliais/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Insulina/farmacologia , Fator de Crescimento Insulin-Like I/farmacologia , Fator de Crescimento Insulin-Like II/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Modelos Biológicos , N-Metilaspartato/farmacologia , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Fator de Transcrição PAX6/metabolismo , PTEN Fosfo-Hidrolase/antagonistas & inibidores , PTEN Fosfo-Hidrolase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Retina/patologia , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo
12.
Cytotherapy ; 21(6): 619-630, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30956007

RESUMO

BACKGROUND: Insulin-like growth factor 2 (IGF2), an essential component of the stem cell niche, has been reported to modulate the proliferation and differentiation of stem cells. Previously, a continuous expression of IGF2 in tissues was reported to maintain the self-renewal ability of several types of stem cells. Therefore, in this study, we investigated the expression of IGF2 in adipose tissues and explored the effects of IGF2 on adipose-derived stromal cells (ADSCs) in vitro. METHODS: The expression pattern of IGF2 in rat adipose tissues was determined by gene expression and protein analyses. The effect of IGF2 on proliferation, stemness-related marker expression and adipogenic and osteogenic differentiation was systematically investigated. Furthermore, antagonists of IGF2-specific receptors-namely, BMS-754807 and picropodophyllin-were added to explore the underlying signal transduction mechanisms. RESULTS: IGF2 levels displayed a tendency to decrease with age in rat adipose tissues. After the addition of IGF2, isolated ADSCs displayed higher proliferation and expression of the stemness-related markers NANOG, OCT4 and SOX2 and greater differentiation potential to adipocytes and osteoblasts. Additionally, both type 1 insulin-like growth factor receptor (IGF-1R) and insulin receptor (IR) participated in the IGF2-mediated promotion of stemness in ADSCs. CONCLUSIONS: Our findings indicate that IGF2 could enhance the stemness of rat ADSCs via IGF-1R and IR and may highlight an effective method for the expansion of ADSCs for clinical application.


Assuntos
Tecido Adiposo/citologia , Fator de Crescimento Insulin-Like II/metabolismo , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/metabolismo , Células Estromais/citologia , Adipócitos/citologia , Adipogenia , Fatores Etários , Animais , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células , Células Cultivadas , Feminino , Fator de Crescimento Insulin-Like II/genética , Fator de Crescimento Insulin-Like II/farmacologia , Osteoblastos/citologia , Osteogênese , Ratos Sprague-Dawley , Receptor IGF Tipo 1/genética , Receptor de Insulina/genética , Transdução de Sinais , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo
13.
Gen Comp Endocrinol ; 283: 113240, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31394085

RESUMO

Leptin is primarily considered a peripheral satiety hormone and is also found to perform important roles in energy homeostasis in vertebrates ranging from fish to mammals. The liver is a major source of leptin production in teleost fish. Using goldfish as a model, a previous report by our group illustrated the positive regulation of leptin mRNA levels by treatment with the hyperglycemic hormone glucagon, and our present study provided evidence for the negative regulation of hepatic leptin-AI and leptin-AII transcripts through the administration of the hypoglycemic hormone insulin. This study is the first to demonstrate changes in the hepatopancreatic insulin, glucagon, leptin-AI and leptin-AII mRNA levels in goldfish during fasting and refeeding. Insulin was found to be effective in suppressing leptin-AI and leptin-AII transcript levels in goldfish liver via both in vivo intraperitoneal injection and in vitro cell incubation approaches. Only the insulin receptor, not the IGF-I receptor, was involved in insulin-inhibited leptin mRNA level. The suppression of leptin levels by insulin was caused by the activation of MKK3/6/p38MAPK and MEK1/2/Erk1/2 cascades. Insulin treatment could eliminate the stimulation of glucagon on leptin mRNA level. Our study describes the regulation and signal transduction mechanism of insulin on leptin mRNA levels in the goldfish liver, suggesting that the leptin function in fish is speculated to be not only an anorexigenic factor but also a metabolic mediator. This also supports the hypothesis that the poikilothermal fish use a passive survival strategy during the periods of food deprivation, which is mediated by the fish-specifically high leptin levels induced by the cooperation of insulin and glucagon.


Assuntos
Privação de Alimentos/fisiologia , Carpa Dourada/genética , Insulina/farmacologia , Leptina/genética , Fígado/metabolismo , Animais , Colforsina/farmacologia , Jejum , Comportamento Alimentar/efeitos dos fármacos , Glucagon/farmacologia , Hepatopâncreas/efeitos dos fármacos , Hepatopâncreas/metabolismo , Humanos , Injeções Intraperitoneais , Insulina/administração & dosagem , Fator de Crescimento Insulin-Like I/farmacologia , Fator de Crescimento Insulin-Like II/farmacologia , Leptina/metabolismo , Fígado/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores para Leptina/metabolismo , Transdução de Sinais/efeitos dos fármacos
14.
J Proteome Res ; 17(9): 2963-2977, 2018 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-30014700

RESUMO

Charcoal-stripped fetal bovine serum (CS-FBS) is commonly used to study androgen responsiveness and androgen metabolism in cultured prostate cancer (CaP) cells. Switching CaP cells from FBS to CS-FBS may reduce the activity of androgen receptor (AR), inhibit cell proliferation, or modulate intracellular androgen metabolism. The removal of proteins by charcoal stripping may cause changes in biological functions and has not yet been investigated. Here we profiled proteins in FBS and CS-FBS using an ion-current-based quantitative platform consisting of reproducible surfactant-aided precipitation/on-pellet digestion, long-column nanoliquid chromatography separation, and ion-current-based analysis. A total of 143 proteins were identified in FBS, among which 14 proteins including insulin-like growth factor 2 (IGF-2) and IGF binding protein (IGFBP)-2 and -6 were reduced in CS-FBS. IGF-1 receptor (IGF1R) and insulin receptor were sensitized to IGFs in CS-FBS. IGF-1 and IGF-2 stimulation fully compensated for the loss of AR activity to maintain cell growth in CS-FBS. Endogenous production of IGF and IGFBPs was verified in CaP cells and clinical CaP specimens. This study provided the most comprehensive protein profiles of FBS and CS-FBS and offered an opportunity to identify new protein regulators and signaling pathways that regulate AR activity, androgen metabolism, and proliferation of CaP cells.


Assuntos
Proteínas Sanguíneas/isolamento & purificação , Células Epiteliais/efeitos dos fármacos , Neoplasias da Próstata/metabolismo , Proteômica/métodos , Testosterona/farmacologia , Adsorção , Animais , Proteínas Sanguíneas/química , Bovinos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Carvão Vegetal/química , Meios de Cultura/química , Meios de Cultura/farmacologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feto , Expressão Gênica , Humanos , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/isolamento & purificação , Proteína 6 de Ligação a Fator de Crescimento Semelhante à Insulina/isolamento & purificação , Fator de Crescimento Insulin-Like I/isolamento & purificação , Fator de Crescimento Insulin-Like I/farmacologia , Fator de Crescimento Insulin-Like II/isolamento & purificação , Fator de Crescimento Insulin-Like II/farmacologia , Masculino , Próstata/efeitos dos fármacos , Próstata/metabolismo , Próstata/patologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Receptor IGF Tipo 1/isolamento & purificação , Receptor de Insulina/isolamento & purificação , Receptores Androgênicos/biossíntese , Receptores Androgênicos/genética , Testosterona/isolamento & purificação
15.
Acta Biochim Biophys Sin (Shanghai) ; 50(10): 976-983, 2018 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-30184089

RESUMO

Embryonic epicardial cells (EPCs) can facilitate cardiomyocyte growth through secreting several essential growth factors, and participate in cardiac development through auto-differentiating into many cardiac cell lineages. Proper proliferation of EPCs is the precondition of these functions, so it is quite necessary to explore the mechanisms involving in EPC proliferation. In this study, we aimed to explore whether insulin-like growth factor 1 receptor (IGF1R) signaling participated in regulating the proliferation of EPCs. Our results showed that the expressions of IGF1R and its ligands IGF1 and IGF2 can be clearly spotted on the epicardium layer from E11.5d to E17.5d. Inhibition of IGF1R signaling using picropodophyllin or NVP-AEW541 significantly decreased the proliferation activity and blocked the cell cycle progression of epicardial cells in vitro. On the contrary, activating IGF1R with recombinant IGF1 and IGF2 promoted epicardial cell proliferation and cell cycle. We also found that decreased expression and phosphorylation of FAK in IGF1R inhibitor-treated cells and use of FAK inhibitor Y15 could significantly inhibit the IGFs-induced EPC proliferation. In conclusion, our results suggest that IGF1R signaling plays an important role in regulating EPC proliferation, and this effect may be mediated by FAK pathway.


Assuntos
Proliferação de Células/fisiologia , Quinase 1 de Adesão Focal/metabolismo , Pericárdio/citologia , Receptor IGF Tipo 1/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Quinase 1 de Adesão Focal/genética , Fator de Crescimento Insulin-Like I/metabolismo , Fator de Crescimento Insulin-Like I/farmacologia , Fator de Crescimento Insulin-Like II/metabolismo , Fator de Crescimento Insulin-Like II/farmacologia , Camundongos Endogâmicos C57BL , Pericárdio/embriologia , Podofilotoxina/análogos & derivados , Podofilotoxina/farmacologia , Pirimidinas/farmacologia , Pirróis/farmacologia , Transdução de Sinais/efeitos dos fármacos
16.
Proc Natl Acad Sci U S A ; 112(6): 1833-8, 2015 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-25624490

RESUMO

The extensive intratumor heterogeneity revealed by sequencing cancer genomes is an essential determinant of tumor progression, diagnosis, and treatment. What maintains heterogeneity remains an open question because competition within a tumor leads to a strong selection for the fittest subclone. Cancer cells also cooperate by sharing molecules with paracrine effects, such as growth factors, and heterogeneity can be maintained if subclones depend on each other for survival. Without strict interdependence between subclones, however, nonproducer cells can free-ride on the growth factors produced by neighboring producer cells, a collective action problem known in game theory as the "tragedy of the commons," which has been observed in microbial cell populations. Here, we report that similar dynamics occur in cancer cell populations. Neuroendocrine pancreatic cancer (insulinoma) cells that do not produce insulin-like growth factor II (IGF-II) grow slowly in pure cultures but have a proliferation advantage in mixed cultures, where they can use the IGF-II provided by producer cells. We show that, as predicted by evolutionary game theory, producer cells do not go extinct because IGF-II acts as a nonlinear public good, creating negative frequency-dependent selection that leads to a stable coexistence of the two cell types. Intratumor cell heterogeneity can therefore be maintained even without strict interdependence between cell subclones. Reducing the amount of growth factors available within a tumor may lead to a reduction in growth followed by a new equilibrium, which may explain relapse in therapies that target growth factors.


Assuntos
Proliferação de Células/efeitos dos fármacos , Fator de Crescimento Insulin-Like II/metabolismo , Insulinoma/metabolismo , Modelos Biológicos , Neoplasias Pancreáticas/metabolismo , Animais , Evolução Biológica , Linhagem da Célula , Simulação por Computador , Citometria de Fluxo , Teoria dos Jogos , Jogos Experimentais , Fator de Crescimento Insulin-Like II/genética , Fator de Crescimento Insulin-Like II/farmacologia , Camundongos , Camundongos Knockout , Seleção Genética
17.
Nature ; 469(7331): 491-7, 2011 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-21270887

RESUMO

We report that, in the rat, administering insulin-like growth factor II (IGF-II, also known as IGF2) significantly enhances memory retention and prevents forgetting. Inhibitory avoidance learning leads to an increase in hippocampal expression of IGF-II, which requires the transcription factor CCAAT enhancer binding protein ß and is essential for memory consolidation. Furthermore, injections of recombinant IGF-II into the hippocampus after either training or memory retrieval significantly enhance memory retention and prevent forgetting. To be effective, IGF-II needs to be administered within a sensitive period of memory consolidation. IGF-II-dependent memory enhancement requires IGF-II receptors, new protein synthesis, the function of activity-regulated cytoskeletal-associated protein and glycogen-synthase kinase 3 (GSK3). Moreover, it correlates with a significant activation of synaptic GSK3ß and increased expression of GluR1 (also known as GRIA1) α-amino-3-hydroxy-5-methyl-4-isoxasolepropionic acid receptor subunits. In hippocampal slices, IGF-II promotes IGF-II receptor-dependent, persistent long-term potentiation after weak synaptic stimulation. Thus, IGF-II may represent a novel target for cognitive enhancement therapies.


Assuntos
Hipocampo/metabolismo , Fator de Crescimento Insulin-Like II/metabolismo , Memória/fisiologia , Animais , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Regulação da Expressão Gênica , Hipocampo/efeitos dos fármacos , Fator de Crescimento Insulin-Like II/farmacologia , Potenciação de Longa Duração/fisiologia , Masculino , Memória/efeitos dos fármacos , Ratos , Ratos Long-Evans , Fatores de Tempo
18.
Am J Physiol Endocrinol Metab ; 310(1): E24-31, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26530156

RESUMO

Enhancing placental insulin-like growth factor (IGF) availability appears to be an attractive strategy for improving outcomes in fetal growth restriction (FGR). Our approach was the novel use of [Leu(27)]IGF-II, a human IGF-II analog that binds the IGF-II clearance receptor IGF-IIR in fetal growth-restricted (FGR) mice. We hypothesized that the impact of [Leu(27)]IGF-II infusion in C57BL/6J (wild-type) and endothelial nitric oxide synthase knockout (eNOS(-/-); FGR) mice would be to enhance fetal growth and investigated this from mid- to late gestation; 1 mg·kg(-1)·day(-1) [Leu(27)]IGF-II was delivered via a subcutaneous miniosmotic pump from E12.5 to E18.5. Fetal and placental weights recorded at E18.5 were used to generate frequency distribution curves; fetuses <5th centile were deemed growth restricted. Placentas were harvested for immunohistochemical analysis of the IGF system, and maternal serum was collected for measurement of exogenously administered IGF-II. In WT pregnancies, [Leu(27)]IGF-II treatment halved the number of FGR fetuses, reduced fetal(P = 0.028) and placental weight variations (P = 0.0032), and increased the numbers of pups close to the mean fetal weight (131 vs. 112 pups within 1 SD). Mixed-model analysis confirmed litter size to be negatively correlated with fetal and placental weight and showed that [Leu(27)]IGF-II preferentially improved fetal weight in the largest litters, as defined by number. Unidirectional (14C)MeAIB transfer per gram placenta (System A amino acid transporter activity) was inversely correlated with fetal weight in [Leu(27)]IGF-II-treated WT animals (P < 0.01). In eNOS(-/-) mice, [Leu(27)]IGF-II reduced the number of FGR fetuses(1 vs. 5 in the untreated group). The observed reduction in FGR pup numbers in both C57 and eNOS(-/-) litters suggests the use of this analog as a means of standardizing and rescuing fetal growth, preferentially in the smallest offspring.


Assuntos
Desenvolvimento Fetal/efeitos dos fármacos , Retardo do Crescimento Fetal/patologia , Fator de Crescimento Insulin-Like II/análogos & derivados , Animais , Modelos Animais de Doenças , Embrião de Mamíferos , Feminino , Retardo do Crescimento Fetal/tratamento farmacológico , Humanos , Fator de Crescimento Insulin-Like II/administração & dosagem , Fator de Crescimento Insulin-Like II/genética , Fator de Crescimento Insulin-Like II/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase Tipo III/genética , Gravidez , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacologia
19.
Cardiovasc Diabetol ; 15(1): 161, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27905925

RESUMO

BACKGROUND: Abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) is a major contributor to the development of atherosclerotic process. In a previous work, we demonstrated that the insulin receptor isoform A (IRA) and its association with the insulin-like growth factor-I receptor (IGF-IR) confer a proliferative advantage to VSMCs. However, the role of IR and IGF-IR in VSMC migration remains poorly understood. METHODS: Wound healing assays were performed in VSMCs bearing IR (IRLoxP+/+ VSMCs), or not (IR-/- VSMCs), expressing IRA (IRA VSMCs) or expressing IRB (IRB VSMCs). To study the role of IR isoforms and IGF-IR in experimental atherosclerosis, we used ApoE-/- mice at 8, 12, 18 and 24 weeks of age. Finally, we analyzed the mRNA expression of total IR, IRB isoform, IGF-IR and IGFs by qRT-PCR in the medial layer of human aortas. RESULTS: IGF-I strongly induced migration of the four cell lines through IGF-IR. In contrast, insulin and IGF-II only caused a significant increase of IRA VSMC migration which might be favored by the formation of IRA/IGF-IR receptors. Additionally, a specific IGF-IR inhibitor, picropodophyllin, completely abolished insulin- and IGF-II-induced migration in IRB, but not in IRA VSMCs. A significant increase of IRA and IGF-IR, and VSMC migration were observed in fibrous plaques from 24-week-old ApoE-/- mice. Finally, we observed a marked increase of IGF-IR, IGF-I and IGF-II in media from fatty streaks as compared with both healthy aortas and fibrolipidic lesions, favoring the ability of medial VSMCs to migrate into the intima. CONCLUSIONS: Our data suggest that overexpression of IGF-IR or IRA isoform, as homodimers or as part of IRA/IGF-IR hybrid receptors, confers a stronger migratory capability to VSMCs as might occur in early stages of atherosclerotic process.


Assuntos
Aterosclerose/metabolismo , Movimento Celular , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Receptor Cross-Talk , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/metabolismo , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Aterosclerose/genética , Aterosclerose/patologia , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Dieta Ocidental , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Insulina/farmacologia , Fator de Crescimento Insulin-Like I/farmacologia , Fator de Crescimento Insulin-Like II/farmacologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/patologia , Isoformas de Proteínas , Receptor Cross-Talk/efeitos dos fármacos , Receptor IGF Tipo 1/agonistas , Receptor IGF Tipo 1/genética , Receptor de Insulina/agonistas , Receptor de Insulina/genética , Receptores de Somatomedina/genética , Receptores de Somatomedina/metabolismo , Transdução de Sinais , Fatores de Tempo
20.
Org Biomol Chem ; 14(39): 9225-9238, 2016 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-27488745

RESUMO

Preptin is a 34-residue pancreatic hormone shown to be anabolic to bone in vitro and in vivo. The bone activity of preptin resides within the (1-16) N-terminal fragment. Due to its peptidic nature, the truncated fragment of preptin is enzymatically unstable; however it provides an attractive framework for the creation of stable analogues using various peptidomimetic techniques. An alanine scan of preptin (1-16) was undertaken which showed that substitution of Ser at position 3 or Pro at position 14 did not inhibit the proliferative activity of preptin in primary rat osteoblasts (bone-forming cells). Importantly, Ser-3 to Ala substitution also showed a significant activity on osteoblast differentiation in vitro and increased the formation of mineralised bone matrix. Additional modifications with non-proteinogenic amino acids at position 3 improved the stability in liver microsomes, but diminished the osteoblast proliferative activity. In addition, to provide greater structural diversity, a series of macrocyclic preptin (1-16) analogues was synthesised using head-to-tail and head-to-side chain macrolactamisation as well as ring-closing metathesis. However, a detrimental effect on osteoblast activity was observed upon macrocyclisation.


Assuntos
Anabolizantes/química , Anabolizantes/farmacologia , Osso e Ossos/efeitos dos fármacos , Fator de Crescimento Insulin-Like II/química , Fator de Crescimento Insulin-Like II/farmacologia , Osteoporose/tratamento farmacológico , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/farmacologia , Sequência de Aminoácidos , Anabolizantes/metabolismo , Anabolizantes/uso terapêutico , Animais , Osso e Ossos/patologia , Proliferação de Células/efeitos dos fármacos , Fator de Crescimento Insulin-Like II/metabolismo , Fator de Crescimento Insulin-Like II/uso terapêutico , Microssomos Hepáticos/metabolismo , Osteoblastos/efeitos dos fármacos , Osteoblastos/patologia , Osteoporose/patologia , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/uso terapêutico , Estabilidade Proteica , Ratos , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA