Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.066
Filtrar
1.
Clin Sci (Lond) ; 138(18): 1173-1177, 2024 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-39289952

RESUMO

Cardiac functional, morphological, and histological analysis, coupled with liquid chromatography and mass spectrometry, of two transgenic mouse models with cardiomyocyte-specific overexpression of insulin-like growth factor 1 receptor (IGF1R) or a dominant-negative PI3K mutant (DCM-dnPI3K) revealed distinctive functional and molecular profiles during physiological (driven by IGF1R overexpression) and pathological (driven by dn-PI3K overexpression) atrial remodeling. The current study confirmed previously reported findings, including ventricular dilatation and enhanced systolic function with no evidence of arrhythmia in IGF1R model, as well as ventricular hypertrophy and decreased systolic function with intermittent atrial fibrillation in DCM-dnPI3K model. Novel findings obtained from the left atrial (LA) characterization of female mice revealed that physiological atrial enlargement resulted from increased atrial myocyte size and was associated with preserved atrial function, as determined by maintained LA ejection fraction (EF). The proteomic profile of IGF1R transgenic (Tg) mice was enriched for metabolic remodeling and showed a protein expression pattern similar to that of healthy human atria; on the other hand, pathological atrial enlargement resulted from increased atrial fibrosis with normal myocyte size and was associated with impaired atrial function due to a reduced LA EF. The proteomic profile of DCM-dnPI3K mice was enriched to both metabolic and structural remodeling and showed a protein expression pattern similar to that of human AF atria.


Assuntos
Remodelamento Atrial , Átrios do Coração , Camundongos Transgênicos , Miócitos Cardíacos , Medicina de Precisão , Receptor IGF Tipo 1 , Animais , Receptor IGF Tipo 1/metabolismo , Receptor IGF Tipo 1/genética , Átrios do Coração/metabolismo , Átrios do Coração/fisiopatologia , Átrios do Coração/patologia , Feminino , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Humanos , Proteômica/métodos , Fibrose , Fibrilação Atrial/fisiopatologia , Fibrilação Atrial/metabolismo , Fibrilação Atrial/patologia , Fibrilação Atrial/genética , Modelos Animais de Doenças , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatologia , Cardiomegalia/patologia , Cardiomegalia/genética , Masculino , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais , Função do Átrio Esquerdo
2.
Int J Mol Sci ; 25(17)2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-39273251

RESUMO

Insulin-like Growth Factor-1 (IGF-1) is a crucial mitogenic factor with important functions in the mammary gland, mainly through its interaction with the IGF-1 receptor (IGF-1R). This interaction activates a complex signaling network that promotes cell proliferation, epithelial to mesenchymal transition (EMT) and inhibits apoptosis. Despite extensive research, the precise molecular pathways and intracellular mechanisms activated by IGF-1, in cancer, remain poorly understood. Recent evidence highlights the essential roles of IGF-1 and its isoforms in breast cancer (BC) development, progression, and metastasis. The peptides that define the IGF-1 isoforms-IGF-1Ea, IGF-1Eb, and IGF-1Ec-act as key points of convergence for various signaling pathways that influence the growth, metastasis and survival of BC cells. The aim of this review is to provide a detailed exami-nation of the role of the mature IGF-1 and its isoforms in BC biology and their potential use as possible therapeutical targets.


Assuntos
Neoplasias da Mama , Fator de Crescimento Insulin-Like I , Isoformas de Proteínas , Humanos , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Fator de Crescimento Insulin-Like I/metabolismo , Isoformas de Proteínas/metabolismo , Feminino , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais , Transição Epitelial-Mesenquimal , Animais , Proliferação de Células , Peptídeos Semelhantes à Insulina
3.
Int J Mol Sci ; 25(18)2024 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-39337253

RESUMO

In the present study, the PC12 cells as a bioassay system were used to screen the small molecules with nerve growth factor (NGF)- mimic effect from Lavandula angustifolia Mill. The ß-Cyclocitral (ß-cyc) as an active compound was discovered, and its chemical structure was also determined. Furthermore, we focused on the bioactive and action mechanism of this compound to do an intensive study with specific protein inhibitors and Western blotting analysis. The ß-cyc had novel NGF-mimic and NGF-enhancer effects on PC12 cells, while the insulin-like growth factor-1 receptor (IGF-1R)/phosphatidylinositol 3 kinase, (PI3K)/serine/threonine-protein kinase (AKT), and glucocorticoid receptor (GR)/phospholipase C (PLC)/protein kinase C (PKC) signaling pathways were involved in the bioactivity of ß-cyc. In addition, the important role of the rat sarcoma (Ras)/protooncogene serine-threonine protein kinase (Raf) signaling pathway was observed, although it was independent of tyrosine kinase (Trk) receptors. Moreover, the non-label target protein discovery techniques, such as the cellular thermal shift assay (CETSA) and drug affinity responsive target stability (DARTS), were utilized to make predictions of its target protein. The stability of IGF-R and GR, proteins for temperature and protease, was dose-dependently increased after treatment of ß-cyc compared with control groups, respectively. These findings indicated that ß-cyc promoted the neuron differentiation of PC12 cells via targeting IGF-1R and GR and modification of downstream signaling pathways.


Assuntos
Fator de Crescimento Insulin-Like I , Lavandula , Fator de Crescimento Neural , Receptores de Glucocorticoides , Transdução de Sinais , Células PC12 , Ratos , Animais , Fator de Crescimento Neural/metabolismo , Fator de Crescimento Neural/farmacologia , Transdução de Sinais/efeitos dos fármacos , Receptores de Glucocorticoides/metabolismo , Lavandula/química , Fator de Crescimento Insulin-Like I/metabolismo , Receptor IGF Tipo 1/metabolismo , Extratos Vegetais/farmacologia , Extratos Vegetais/química
4.
J Headache Pain ; 25(1): 156, 2024 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-39304806

RESUMO

BACKGROUND: Chronic migraine is a severe and common neurological disorder, yet its precise physiological mechanisms remain unclear. The IGF1/IGF1r signaling pathway plays a crucial role in pain modulation. Studies have shown that IGF1, by binding to its receptor IGF1r, activates a series of downstream signaling cascades involved in neuronal survival, proliferation, autophagy and functional regulation. The activation of these pathways can influence nociceptive transmission. Furthermore, alterations in IGF1/IGF1r signaling are closely associated with the development of various chronic pain conditions. Therefore, understanding the specific mechanisms by which this pathway contributes to pain is of significant importance for the development of novel pain treatment strategies. In this study, we investigated the role of IGF1/IGF1r and its potential mechanisms in a mouse model of chronic migraine. METHODS: Chronic migraine was induced in mice by repeated intraperitoneal injections of nitroglycerin. Mechanical and thermal hypersensitivity responses were assessed using Von Frey filaments and radiant heat, respectively. To determine the role of IGF1/IGF1r in chronic migraine (CM), we examined the effects of the IGF1 receptor antagonist ppp (Picropodophyllin) on pain behaviors and the expression of calcitonin gene-related peptide (CGRP) and c-Fos. RESULT: In the nitroglycerin-induced chronic migraine model in mice, neuronal secretion of IGF1 is elevated within the trigeminal nucleus caudalis (TNC). Increased phosphorylation of the IGF1 receptor occurs, predominantly co-localizing with neurons. Treatment with ppp alleviated basal mechanical hypersensitivity and acute mechanical allodynia. Furthermore, ppp ameliorated autophagic dysfunction and reduced the expression of CGRP and c-Fos. CONCLUSION: Our findings demonstrate that in the chronic migraine (CM) model in mice, there is a significant increase in IGF1 expression in the TNC region. This upregulation of IGF1 leads to enhanced phosphorylation of IGF1 receptors on neurons. Targeting and inhibiting this signaling pathway may offer potential preventive strategies for mitigating the progression of chronic migraine.


Assuntos
Autofagia , Modelos Animais de Doenças , Fator de Crescimento Insulin-Like I , Transtornos de Enxaqueca , Nitroglicerina , Receptor IGF Tipo 1 , Transdução de Sinais , Animais , Transtornos de Enxaqueca/induzido quimicamente , Transtornos de Enxaqueca/metabolismo , Transtornos de Enxaqueca/fisiopatologia , Transtornos de Enxaqueca/tratamento farmacológico , Fator de Crescimento Insulin-Like I/metabolismo , Receptor IGF Tipo 1/metabolismo , Autofagia/efeitos dos fármacos , Autofagia/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Camundongos , Masculino , Nitroglicerina/toxicidade , Nitroglicerina/farmacologia , Camundongos Endogâmicos C57BL
5.
Sci Rep ; 14(1): 22661, 2024 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-39349545

RESUMO

Unlike adult mammals, the hearts of neonatal mice possess the ability to completely regenerate from myocardial infarction (MI). This observation has sparked vast interest in deciphering the potentially lifesaving and morbidity-reducing mechanisms involved in neonatal cardiac regeneration. In mice, the regenerative potential is lost within the first week of life and coincides with a reduction of Insulin-like growth factor 1 receptor (Igf1r) expression in the heart. Igf1r is a well-known regulator of cardiomyocyte maturation and proliferation in neonatal mice. To test the role of Igf1r as a pivotal factor in cardiac regeneration, we knocked down (KD) Igf1r specifically in cardiomyocytes using recombinant adeno-associated virus (rAAV) delivery and troponin T promotor driven shRNAmirs. Cardiomyocyte specific Igf1r KD versus control mice were subjected to experimental MI by permanent ligation of the left anterior descending artery (LAD). Cardiac functional and morphological data were analyzed over a 21-day period. Neonatal Igf1r KD mice showed reduced systolic cardiac function and increased fibrotic cardiac remodeling 21 days post injury. This cardiac phenotype was associated with reduced cardiomyocyte nuclei mitosis and decreased AKT and ERK phosphorylation in Igf1r KD, compared to control neonatal mouse hearts. Our in vivo murine data show that Igf1r KD shifts neonatal cardiac regeneration to a more adult-like scarring phenotype, identifying cardiomyocyte-specific Igf1r signaling as a crucial component of neonatal cardiac regeneration.


Assuntos
Animais Recém-Nascidos , Infarto do Miocárdio , Miócitos Cardíacos , Receptor IGF Tipo 1 , Regeneração , Transdução de Sinais , Animais , Receptor IGF Tipo 1/metabolismo , Receptor IGF Tipo 1/genética , Miócitos Cardíacos/metabolismo , Camundongos , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/genética , Coração/fisiologia , Peptídeos Semelhantes à Insulina
6.
Ann Clin Lab Sci ; 54(4): 510-518, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39293845

RESUMO

OBJECTIVE: LIN28B and insulin-like growth factor-1 receptor (IGF-1R) play important roles in the growth and metastasis of colorectal cancer (CRC). The relationship between LIN28B and IGF-1R and their co-expression role in CRC is unknown. This study aims to assess whether the combined detection of LIN28B and IGF-1R is a better predictor of prognosis in CRC than either marker alone. METHODS: We used immunohistochemistry to detect LIN28B and IGF-1R protein. The correlation between LIN28B and IGF-1R expression in CRC was determined by the Spearman correlation analysis. The association between LIN28B/IGF-1R expression and clinicopathological features was analyzed, and overall survivals were also performed. RESULTS: The expression of LIN28B and IGF-1R in CRC tissues were significantly higher than that in non-cancerous tissues (P<0.001 and P<0.001, respectively). LIN28B over-expression positively correlated with IGF-1R over-expression (r=0.283, P=0.007). The increased expression of LIN28B and IGF-1R were both significantly correlated with TNM stage, lymph node metastasis and distant metastasis (all P<0.05). IGF-1R, not LIN28B, was significantly associated with vascular invasion (P=0.007). Overall survival appears to be lower in patients with high LIN28B expression or/and high IGF-1R expression than those with low LIN28B expression or/and low IGF-1R expression. In addition, the predictive sensitivity of LIN28B combined with IGF-1R was stronger than that of either one alone. CONCLUSIONS: The combined over-expression of LIN28B and IGF-1R in patients with colorectal cancer may provide a more powerful predictor for CRC prognosis.


Assuntos
Biomarcadores Tumorais , Neoplasias Colorretais , Proteínas de Ligação a RNA , Receptor IGF Tipo 1 , Humanos , Neoplasias Colorretais/patologia , Neoplasias Colorretais/metabolismo , Receptor IGF Tipo 1/metabolismo , Masculino , Feminino , Proteínas de Ligação a RNA/metabolismo , Prognóstico , Pessoa de Meia-Idade , Idoso , Biomarcadores Tumorais/metabolismo , Adulto
7.
J Transl Med ; 22(1): 823, 2024 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-39232805

RESUMO

BACKGROUND: Breast cancer (BC) is the most common malignant tumor in women worldwide, and further elucidation of the molecular mechanisms involved in BC pathogenesis is essential to improve the prognosis of BC patients. RNA Binding Motif Protein 8 A (RBM8A), with high affinity to a myriad of RNA transcripts, has been shown to play a crucial role in genesis and progression of multiple cancers. We attempted to explore its functional significance and molecular mechanisms in BC. METHODS: Bioinformatics analysis was performed on publicly available BC datasets. qRT-PCR was used to determine the expression of RBM8A in BC tissues. MTT assay, clone formation assay and flow cytometry were employed to examine BC cell proliferation and apoptosis in vitro. RNA immunoprecipitation (RIP) and RIP-seq were used to investigate the binding of RBM8A/EIF4A3 to the mRNA of IGF1R/IRS-2. RBM8A and EIF4A3 interactions were determined by co-immunoprecipitation (Co-IP) and immunofluorescence. Chromatin immunoprecipitation (Ch-IP) and dual-luciferase reporter assay were carried out to investigate the transcriptional regulation of RBM8A by TEAD4. Xenograft model was used to explore the effects of RBM8A and TEAD4 on BC cell growth in vivo. RESULTS: In this study, we showed that RBM8A is abnormally highly expressed in BC and knockdown of RBM8A inhibits BC cell proliferation and induces apoptosis in vitro. EIF4A3, which phenocopy RBM8A in BC, forms a complex with RBM8A in BC. Moreover, EIF4A3 and RBM8A complex regulate the expression of IGF1R and IRS-2 to activate the PI3K/AKT signaling pathway, thereby promoting BC progression. In addition, we identified TEAD4 as a transcriptional activator of RBM8A by Ch-IP, dual luciferase reporter gene and a series of functional rescue assays. Furthermore, we demonstrated the in vivo pro-carcinogenic effects of TEAD4 and RBM8A by xenograft tumor experiments in nude mice. CONCLUSION: Collectively, these findings suggest that TEAD4 novel transcriptional target RBM8A interacts with EIF4A3 to increase IGF1R and IRS-2 expression and activate PI3K/AKT signaling pathway, thereby further promoting the malignant phenotype of BC cells.


Assuntos
Neoplasias da Mama , Proteínas de Ligação a DNA , Regulação Neoplásica da Expressão Gênica , Proteínas Musculares , Proteínas de Ligação a RNA , Receptor IGF Tipo 1 , Fatores de Transcrição de Domínio TEA , Animais , Feminino , Humanos , Camundongos , Apoptose/genética , Neoplasias da Mama/patologia , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Progressão da Doença , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Camundongos Nus , Proteínas Musculares/metabolismo , Proteínas Musculares/genética , Ligação Proteica , Receptor IGF Tipo 1/metabolismo , Receptor IGF Tipo 1/genética , Receptores de Somatomedina/metabolismo , Receptores de Somatomedina/genética , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/genética , Transdução de Sinais , Fatores de Transcrição de Domínio TEA/metabolismo
8.
Obesity (Silver Spring) ; 32(10): 1923-1933, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39165088

RESUMO

OBJECTIVE: This study aimed to identify whether cord blood DNA methylation at specific loci is associated with neonatal adiposity, a key risk factor for childhood obesity. METHODS: An epigenome-wide association study was conducted using the Hyperglycemia and Adverse Pregnancy Outcome (HAPO) Study as a discovery sample. Linear regression models adjusted for maternal and offspring covariates and cell counts were used to analyze associations between neonatal adiposity as measured by sum of three skinfold thicknesses and cord blood DNA methylation. Assays were performed with Illumina EPIC arrays (791,359 CpG sites after quality control). Replication was performed in an independent cohort, Genetics of Glucose regulation in Gestation and Growth (Gen3G). RESULTS: In 2740 HAPO samples, significant associations were identified at 89 CpG sites after accounting for multiple testing (Bonferroni-adjusted p < 0.05). Replication analyses conducted in 139 Gen3G participants confirmed associations for seven CpG sites. These included IGF1R, which encodes a transmembrane receptor involved in cell growth and survival that binds insulin-like growth factor I and insulin, and KLF7, which encodes a regulator of cell proliferation and inhibitor of adipogenesis; both are key regulators of growth during fetal life. CONCLUSIONS: These findings support epigenetic mechanisms in the developmental origins of neonatal adiposity and as potential biomarkers of metabolic disease risk.


Assuntos
Adiposidade , Metilação de DNA , Sangue Fetal , Fatores de Transcrição Kruppel-Like , Receptor IGF Tipo 1 , Humanos , Sangue Fetal/metabolismo , Feminino , Adiposidade/genética , Recém-Nascido , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Masculino , Gravidez , Estudo de Associação Genômica Ampla , Adulto , Ilhas de CpG , Epigênese Genética , Obesidade Infantil/genética , Obesidade Infantil/sangue
9.
J Transl Med ; 22(1): 719, 2024 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-39103832

RESUMO

With the increasing age of the population worldwide, the incidence rate of Parkinson's disease (PD) is increasing annually. Currently, the treatment strategy for PD only improves clinical symptoms. No effective treatment strategy can slow down the progression of the disease. In the present study, whole transcriptome sequencing was used to obtain the mRNA and miRNA expression profiles in a PD mouse model, which revealed the pathogenesis of PD. The transcription factor RUNX3 upregulated the miR-186-3p expression in the PD model. Furthermore, the high miR-186-3p expression in PD can be targeted to inhibit the DAT expression, resulting in a decrease in the dopamine content of dopaminergic neurons. Moreover, miR-186-3p can be targeted to inhibit the IGF1R expression and prevent the activation of the IGF1R-P-PI3K-P-AKT pathway, thus increasing the apoptosis of dopaminergic neurons by regulating the cytochrome c-Bax-cleaved caspase-3 pathway. Our research showed that the RUNX3-miR-186-3p-DAT-IGF1R axis plays a key role in the pathogenesis of PD, and miR-186-3p is a potential target for the treatment of PD.


Assuntos
Subunidade alfa 3 de Fator de Ligação ao Core , Modelos Animais de Doenças , MicroRNAs , Doença de Parkinson , Receptor IGF Tipo 1 , MicroRNAs/genética , MicroRNAs/metabolismo , Animais , Receptor IGF Tipo 1/metabolismo , Receptor IGF Tipo 1/genética , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Subunidade alfa 3 de Fator de Ligação ao Core/metabolismo , Subunidade alfa 3 de Fator de Ligação ao Core/genética , Camundongos Endogâmicos C57BL , Masculino , Apoptose/genética , Transdução de Sinais , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Camundongos , Sequência de Bases
10.
Artigo em Inglês | MEDLINE | ID: mdl-39097140

RESUMO

Muscle and bone are cooperatively preserved in Daurian ground squirrels (Spermophilus dauricus) during hibernation. As such, we hypothesized that IGF-1 and myostatin may contribute to musculoskeletal maintenance during this period. Thus, we systematically assessed changes in the protein expression levels of IGF-1 and myostatin, as well as their corresponding downstream targets, in the vastus medialis (VM) muscle and femur in Daurian ground squirrels during different stages. Group differences were determined using one-way analysis of variance (ANOVA). Results indicated that the co-localization levels of IGF-1 and its receptor (IGF-1R) increased by 50% during the pre-hibernation period (PRE) and by 35% during re-entry into torpor (RET) compared to the summer active period (SA). The phosphorylation level of FOXO1 in the VM muscle increased by 50% in the torpor (TOR) group and by 82% in the inter-bout arousal (IBA) group compared to the PRE group. The phosphorylation level of SGK-1 increased by 54% in the IBA group and by 62% in the RET group compared to the SA group. In contrast, the protein expression of IGF-1 and phosphorylation levels of PI3K, Akt, mTOR, and GSK3ß in the VM muscle showed no obvious differences among the different groups. ß-catenin protein expression was up-regulated by 84% in the RET group compared to the SA group, while the content of IGF-1 protein, correlation coefficients of IGF-1 and IGF-1R, and phosphorylation levels of PI3K, Akt, and GSK3ß in the femur showed no significant differences among groups. Regarding myostatin and its downstream targets, myostatin protein expression decreased by 70% in the RET group compared to the SA group, whereas ActRIIB protein expression and Smad2/3 phosphorylation in the VM muscle showed no obvious differences among groups. Furthermore, Smad2/3 phosphorylation decreased by 58% in the TOR group and 53% in the RET group compared to the SA group, whereas ActRIIB protein expression in the femur showed no obvious differences among groups. Overall, the observed changes in IGF-1 and myostatin expression and their downstream targets may be involved in musculoskeletal preservation during hibernation in Daurian ground squirrels.


Assuntos
Hibernação , Fator de Crescimento Insulin-Like I , Músculo Esquelético , Miostatina , Sciuridae , Animais , Miostatina/metabolismo , Miostatina/genética , Hibernação/fisiologia , Sciuridae/fisiologia , Sciuridae/metabolismo , Músculo Esquelético/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Fosforilação , Receptor IGF Tipo 1/metabolismo , Receptor IGF Tipo 1/genética , Osso e Ossos/metabolismo , Transdução de Sinais , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fêmur/metabolismo
11.
Life Sci ; 353: 122939, 2024 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-39094905

RESUMO

AIMS: Transactivation of insulin-growth-factor-receptor (IGF-1R) by angiotensin-II-type-1-receptor (AT-1R) was only demonstrated in vascular-smooth-muscle cells and has never been tested in breast-cancer (BC). This investigation addressed the impact of chronic AT-1R blockade by valsartan (Val) on possible concurrent AT-1R/IGF-1R signaling inhibition, regressing BC-tumor-microenvironment (TME) cellular components activation, and hindering BC development. MAIN METHODS: The effect of different Val doses (10, 20, 40 & 80 mg/kg/day for 490 days) was tested on dimethylbenz(a)anthracene (DMBA)-induced progesterone-promoted-BC in rats. The influence on intratumoral/circulating angiotensin-II (ANG-II) levels and AT-1R/Mas-R immunofluorescent-expression were assessed. The potential AT-1R/IGF-1R crosstalk within TME-BC-stem-cells (BCSCs) and cancer-associated-fibroblasts (CAFs) was evaluated by fluorescently marking these cells and locating the immunofluorescently-stained AT-1R/IGF-1R in them using confocal-laser-microscopy and further quantified by flow cytometry. In addition, the molecular alterations following blocking AT-1R were inspected including determining Src; crucial for IGF-1R transactivation by AT-1R, Notch-1; IGF-IR transcriptional-regulator, and PI3K/Akt &IL-6/STAT expression. Further, the suppression of CSCs' capabilities to maintain pluripotency, stemness features, epithelial-to-mesenchymal-transition (EMT), and angiogenesis was evaluated by assessing NANOG gene, aldehyde-dehydrogenase (ALDH), N-cadherin and vascular-endothelial-growth-factor (VEGF), respectively. Furthermore, the proliferative marker; Ki-67, was detected by immunostaining, and tumors were histologically graded using Elston-Ellis-modified-Scarff-Bloom-Richardson method. KEY FINDINGS: Prophylactic Val significantly reduced tumor size, prolonged latency, reduced tumor histopathologic grade, decreased circulating/intratumoral-ANG-II levels, increased Mas-R, and decreased AT1R expression. AT-1R/IGF-1R were co-expressed with a high correlation coefficient on CAFs/BCSCs. Moreover, Val significantly attenuated IGF-1R transactivation and transcriptional regulation via Src and Notch-1 genes' downregulation and reduced Src/IGF-IR-associated PI3K/Akt and IL-6/STAT3 signaling. Further, Val significantly decreased intratumoral NANOG, ALDH, N-cadherin, VEGF, and Ki-67 levels. SIGNIFICANCE: Chronic Val administration carries a potential for repurposing as adjuvant or conjunct therapy for patients at high risk for BC.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II , Neoplasias da Mama , Receptor Tipo 1 de Angiotensina , Receptor IGF Tipo 1 , Microambiente Tumoral , Valsartana , Animais , Feminino , Ratos , Valsartana/farmacologia , Receptor Tipo 1 de Angiotensina/metabolismo , Receptor IGF Tipo 1/metabolismo , Microambiente Tumoral/efeitos dos fármacos , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Neoplasias da Mama/patologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Ratos Sprague-Dawley
12.
J Exp Clin Cancer Res ; 43(1): 211, 2024 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-39075581

RESUMO

BACKGROUND: Early cutaneous squamous cell carcinomas (cSCCs) generally show epithelial differentiation features and good prognosis, whereas advanced cSCCs present mesenchymal traits associated with tumor relapse, metastasis, and poor survival. Currently, the mechanisms involved in cSCC progression are unclear, and the established markers are suboptimal for accurately predicting the clinical course of the disease. METHODS: Using a mouse model of cSCC progression, expression microarray analysis, immunofluorescence and flow cytometry assays, we have identified a prognostic biomarker of tumor relapse, which has been evaluated in a cohort of cSCC patient samples. Phosphoproteomic analysis have revealed signaling pathways induced in epithelial plastic cancer cells that promote epithelial-mesenchymal plasticity (EMP) and tumor progression. These pathways have been validated by genetic and pharmacological inhibition assays. RESULTS: We show that the emergence of epithelial cancer cells expressing integrin αV (ITGAV) promotes cSCC progression to a mesenchymal state. Consistently, ITGAV expression allows the identification of patients at risk of cSCC relapse above the currently employed clinical histopathological parameters. We also demonstrate that activation of insulin-like growth factor-1 receptor (IGF1R) pathway in epithelial cancer cells is necessary to induce EMP and mesenchymal state acquisition in response to tumor microenvironment-derived factors, while promoting ITGAV expression. Likewise, ITGAV knockdown in epithelial plastic cancer cells also blocks EMP acquisition, generating epithelial tumors. CONCLUSIONS: Our results demonstrate that ITGAV is a prognostic biomarker of relapse in cSCCs that would allow improved patient stratification. ITGAV also collaborates with IGF1R to induce EMP in epithelial cancer cells and promotes cSCC progression, revealing a potential therapeutic strategy to block the generation of advanced mesenchymal cSCCs.


Assuntos
Transição Epitelial-Mesenquimal , Receptor IGF Tipo 1 , Transdução de Sinais , Neoplasias Cutâneas , Animais , Humanos , Camundongos , Biomarcadores Tumorais/metabolismo , Biomarcadores Tumorais/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/genética , Linhagem Celular Tumoral , Prognóstico , Receptor IGF Tipo 1/metabolismo , Receptor IGF Tipo 1/genética , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/genética , Microambiente Tumoral , Integrina alfaV/genética , Integrina alfaV/metabolismo
13.
Growth Horm IGF Res ; 77: 101607, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39033666

RESUMO

Type 2 diabetes is characterised by the disruption of insulin and insulin-like growth factor (IGF) signalling. The key hubs of these signalling cascades - the Insulin receptor (IR) and Insulin-like growth factor 1 receptor (IGF1R) - are known to form functional IR-IGF1R hybrid receptors which are insulin resistant. However, the mechanisms underpinning IR-IGF1R hybrid formation are not fully understood, hindering the ability to modulate this for future therapies targeting this receptor. To pinpoint suitable sites for intervention, computational hotspot prediction was utilised to identify promising epitopes for targeting with point mutagenesis. Specific IGF1R point mutations F450A, R391A and D555A show reduced affinity of the hybrid receptor in a BRET based donor-saturation assay, confirming hybrid formation could be modulated at this interface. These data provide the basis for rational design of more effective hybrid receptor modulators, supporting the prospect of identifying a small molecule that specifically interacts with this target.


Assuntos
Mutagênese Sítio-Dirigida , Receptor IGF Tipo 1 , Receptor de Insulina , Receptor de Insulina/genética , Receptor de Insulina/química , Receptor de Insulina/metabolismo , Humanos , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/química , Receptor IGF Tipo 1/metabolismo , Multimerização Proteica , Peptídeos Semelhantes à Insulina , Antígenos CD
14.
J Cell Mol Med ; 28(13): e18471, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38984951

RESUMO

Diabetes mellitus is a major cause of blindness and chronic ulcers in the working-age population worldwide. Wound healing is deeply dependent on neovascularization to restore blood flow. Former research has found that differentially expressed circular RNAs (circRNAs) are associated with hyperglycaemia-induced endothelial cell damage, and hypoxia-pretreated adipose-derived stem cells (ADSCs)-extracellular vesicle (HEV) transplants have a more therapeutic effect to enhance wound healing in diabetic mice by delivery circRNA. The current investigation employed high-throughput sequencing to identify circRNAs that are abnormally expressed between EV and HEV. The regulatory mechanism and predicted targets of one differentially expressed circRNA, circ-IGF1R, were investigated utilizing bioinformatics analyses, luciferase reporter assays, angiogenic differentiation assays, flow cytometric apoptosis analysis and RT-qPCR. Circ-IGF1R expression increased in HEV, and downregulation of circ-IGF1R suppressed and reversed the promotion effect of HEV on angiogenesis in ulcerated tissue. Bioinformatics analyses and luciferase reporter assays confirmed that miR-503-5p was the downstream target of circ-IGF1R, and inhibiting miR-503-5p restored the promotion effect of HEV on angiogenesis after circ-IGF1R silence. The study also found that miR-503-5p can interact with 3'-UTR of both HK2 and VEGFA. Overexpression of HK2 or VEGFA restored the promotion effect of HExo on angiogenesis after circ-IGF1R silence. Overexpression miR-503-5p or silence HK2/VEGFA reversed the protective effect of circ-IGF1R to MLMECs angiogenic differentiation. Overexpression of circ-IGF1R increased the protective effect of HEV on the promotion of wound healing in mice with diabetes. Circ-IGF1R promotes HIF-1α expression through miR-503-5p sponging. Our data demonstrate that circ-IGF1R overexpression EVs from ADSCs suppress high glucose-induced endothelial cell damage by regulating miR-503-5p/HK2/VEGFA axis.


Assuntos
Vesículas Extracelulares , MicroRNAs , RNA Circular , Receptor IGF Tipo 1 , Fator A de Crescimento do Endotélio Vascular , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Circular/genética , RNA Circular/metabolismo , Animais , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/transplante , Camundongos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Receptor IGF Tipo 1/metabolismo , Receptor IGF Tipo 1/genética , Humanos , Células-Tronco/metabolismo , Masculino , Regulação da Expressão Gênica , Cicatrização/genética , Hipóxia Celular/genética , Transdução de Sinais , Regulação para Cima/genética , Neovascularização Fisiológica/genética
15.
Ecotoxicol Environ Saf ; 283: 116804, 2024 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-39083871

RESUMO

Bisphenol A (BPA), a typical environmental endocrine disruptor, has raised concerns among researchers due to its toxicological effects. Whether neohesperidin (NEO) can intervene in the toxic effects of BPA remains unknown. This study aims to investigate the effects and mechanisms of NEO on the myogenic differentiation of umbilical cord-derived mesenchymal stem cells (UC-MSCs) exposed to BPA. Sheep UC-MSCs were isolated, characterized, and induced to myogenic differentiation. BPA decreased cell viability, cell migration, and the expressions of myogenic marker genes, leading to myogenic differentiation inhibition, which were reversed by NEO. Network pharmacology suggested the IGF1R/AKT1/RHOA pathway as potential targets of BPA and NEO regulating muscle development. Western blot results showed that NEO could reverse the down-regulation of the pathway proteins induced by BPA, and counteract the effects of picropodophyllin (PPP) or MK-2206 dihydrochloride (MK-2206) in the myogenic differentiation of sheep UC-MSCs. Additionally, the expression levels of (p-) IGF1R, AKT1, and RHOA were positively correlated. Taken together, the mechanisms of NEO resistance to BPA involved the IGF1R/AKT1/RHOA signaling pathway. These findings provide a scientific basis for alleviating BPA toxicity, preventing and treating muscular dysplasia, and promoting muscle damage repair.


Assuntos
Compostos Benzidrílicos , Diferenciação Celular , Hesperidina , Células-Tronco Mesenquimais , Fenóis , Proteínas Proto-Oncogênicas c-akt , Receptor IGF Tipo 1 , Transdução de Sinais , Células-Tronco Mesenquimais/efeitos dos fármacos , Compostos Benzidrílicos/toxicidade , Fenóis/toxicidade , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Transdução de Sinais/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Receptor IGF Tipo 1/metabolismo , Receptor IGF Tipo 1/genética , Hesperidina/farmacologia , Hesperidina/análogos & derivados , Proteína rhoA de Ligação ao GTP/metabolismo , Cordão Umbilical/citologia , Cordão Umbilical/efeitos dos fármacos , Disruptores Endócrinos/toxicidade , Ovinos , Desenvolvimento Muscular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos
16.
Cell Signal ; 122: 111307, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39048037

RESUMO

The kinase domains of receptor tyrosine kinases (RTKs) are highly conserved, yet they are able to discriminate among potential substrates to selectively activate downstream signaling pathways. In this study, we tested the importance of catalytic domain specificity by creating two series of chimeric RTKs. In one set, the kinase domain of insulin-like growth factor I receptor (IGF1R) was replaced by the kinase domains from insulin receptor (IR), macrophage stimulating protein 1 receptor/Ron (Ron) or Src. In the other set of chimeras, the kinase domain of epidermal growth factor receptor (EGFR) was similarly replaced by the kinase domains of IR, Ron, or Src. We expressed the wild-type and chimeric forms of the receptors in mammalian cells. For some signaling events, such as recognition of IRS1, the identity of the tyrosine kinase catalytic domain did not appear to be crucial. In contrast, recognition of some sites, such as the C-terminal autophosphorylation sites on EGFR, did depend on the identity of the kinase domain. Our data also showed that ligand dependence was lost when the native kinase domains were replaced by Src, suggesting that the identity of the kinase domains could be important for proper receptor regulation. Overall, the results are consistent with the idea that the fidelity of RTK signaling depends on co-localization and targeting with substrates, as well as on the intrinsic specificity of the kinase domain.


Assuntos
Receptores ErbB , Receptores Proteína Tirosina Quinases , Receptor IGF Tipo 1 , Humanos , Receptores ErbB/metabolismo , Receptor IGF Tipo 1/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Fosforilação , Transdução de Sinais , Animais , Receptor de Insulina/metabolismo , Especificidade por Substrato , Quinases da Família src/metabolismo , Domínio Catalítico , Domínios Proteicos , Células HEK293 , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Substratos do Receptor de Insulina/metabolismo
17.
Nutrients ; 16(14)2024 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-39064775

RESUMO

The most widely used synthetic glucocorticoid, dexamethasone (DEX), causes stunted growth in children when used excessively or for long periods of time; however, there are still plenty of pediatric patients require long-term treatment with DEX. As an alternative, growth hormone is used in combination, but it has side effects, a high cost, and psychological factors, and it is not satisfactory in terms of effectiveness. It is necessary to develop a safe and affordable treatment that can replace it. The Korean Food and Drug Administration approved HT042, a standardized functional food ingredient, with the claim that it can help height growth of children. In this study, it was found that HT042 activated the Indian hedgehog/parathyroid hormone-related protein signaling pathway and enhanced the number of growth hormone receptors and insulin-like growth factor-1 receptors on the growth plate surface, which were reduced by DEX treatment, and restored growth retardation. In metatarsal bone and primary chondrocyte models, it was found that HT042 can promote the length of growth plate and recover DEX-induced growth retardation. It was also found that HT042 promotes cell proliferation using bromodeoxyuridine and terminal deoxynucleotidyl transferase dUTP nick end labeling assays; moreover, we verified increased expression of GHR/IGF-1R and Ihh/PTHrP pathway activity using qRT-PCR, western blotting, and siRNA analyses to verify its direct action on the growth plate. The anti-apoptotic effect of HT042 was identified by regulating the expression of apoptotic factors such as caspase-3, Bcl2, Bclx, and Bax. These results were identified using both ex vivo and in vitro models. Our study verified that co-administration of HT042 could recover the DEX induced growth retardation.


Assuntos
Proliferação de Células , Dexametasona , Lâmina de Crescimento , Ossos do Metatarso , Extratos Vegetais , Transdução de Sinais , Animais , Dexametasona/farmacologia , Ossos do Metatarso/efeitos dos fármacos , Lâmina de Crescimento/efeitos dos fármacos , Ratos , Proliferação de Células/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Extratos Vegetais/farmacologia , Masculino , Proteínas Hedgehog/metabolismo , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Ratos Sprague-Dawley , Apoptose/efeitos dos fármacos , Receptores da Somatotropina/metabolismo , Receptores da Somatotropina/genética , Receptor IGF Tipo 1/metabolismo , Receptor IGF Tipo 1/genética , Desenvolvimento Ósseo/efeitos dos fármacos , Transtornos do Crescimento/induzido quimicamente
18.
FASEB J ; 38(13): e23797, 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-38963344

RESUMO

The role of N-glycosylation in the myogenic process remains poorly understood. Here, we evaluated the impact of N-glycosylation inhibition by Tunicamycin (TUN) or by phosphomannomutase 2 (PMM2) gene knockdown, which encodes an enzyme essential for catalyzing an early step of the N-glycosylation pathway, on C2C12 myoblast differentiation. The effect of chronic treatment with TUN on tibialis anterior (TA) and extensor digitorum longus (EDL) muscles of WT and MLC/mIgf-1 transgenic mice, which overexpress muscle Igf-1Ea mRNA isoform, was also investigated. TUN-treated and PMM2 knockdown C2C12 cells showed reduced ConA, PHA-L, and AAL lectin binding and increased ER-stress-related gene expression (Chop and Hspa5 mRNAs and s/uXbp1 ratio) compared to controls. Myogenic markers (MyoD, myogenin, and Mrf4 mRNAs and MF20 protein) and myotube formation were reduced in both TUN-treated and PMM2 knockdown C2C12 cells. Body and TA weight of WT and MLC/mIgf-1 mice were not modified by TUN treatment, while lectin binding slightly decreased in the TA muscle of WT (ConA and AAL) and MLC/mIgf-1 (ConA) mice. The ER-stress-related gene expression did not change in the TA muscle of WT and MLC/mIgf-1 mice after TUN treatment. TUN treatment decreased myogenin mRNA and increased atrogen-1 mRNA, particularly in the TA muscle of WT mice. Finally, the IGF-1 production and IGF1R signaling pathways activation were reduced due to N-glycosylation inhibition in TA and EDL muscles. Decreased IGF1R expression was found in TUN-treated C2C12 myoblasts which was associated with lower IGF-1-induced IGF1R, AKT, and ERK1/2 phosphorylation compared to CTR cells. Chronic TUN-challenge models can help to elucidate the molecular mechanisms through which diseases associated with aberrant N-glycosylation, such as Congenital Disorders of Glycosylation (CDG), affect muscle and other tissue functions.


Assuntos
Diferenciação Celular , Chaperona BiP do Retículo Endoplasmático , Músculo Esquelético , Mioblastos , Receptor IGF Tipo 1 , Transdução de Sinais , Tunicamicina , Animais , Camundongos , Glicosilação , Mioblastos/metabolismo , Chaperona BiP do Retículo Endoplasmático/metabolismo , Tunicamicina/farmacologia , Receptor IGF Tipo 1/metabolismo , Receptor IGF Tipo 1/genética , Músculo Esquelético/metabolismo , Desenvolvimento Muscular/fisiologia , Linhagem Celular , Camundongos Transgênicos , Estresse do Retículo Endoplasmático , Fator de Crescimento Insulin-Like I/metabolismo , Fator de Crescimento Insulin-Like I/genética
19.
Exp Gerontol ; 194: 112512, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38971545

RESUMO

OBJECTIVE: This study investigated sex-specific pathogenesis mechanisms in Alzheimer's disease (AD) using single-nucleus RNA sequencing (snRNA-seq) data. METHODS: Data from the Gene Expression Omnibus (GEO) were searched using terms "Alzheimer's Disease", "single cell", and "Homo sapiens". Studies excluding APOE E4 and including comprehensive gender information with 10× sequencing methods were selected, resulting in GSE157827 and GSE174367 datasets from human prefrontal cortex samples. Sex-stratified analyses were conducted on these datasets, and the outcomes of the analysis for GSE157827 were compared with those of GSE174367. The findings were validated using expression profiling from the mouse dataset GSE85162. Furthermore, real-time PCR experiments in mice further confirmed these findings. The Seurat R package was used to identify cell types, and batch effects were mitigated using the Harmony R package. Cell proportions by sex were compared using the Mann-Whitney-Wilcoxon test, and gene expression variability was displayed with an empirical cumulative distribution plot. Differentially expressed genes were identified using the FindMarkers function with the MAST test. Transcription factors were analyzed using the RcisTarget R package. RESULTS: Seven cell types were identified: astrocytes, endothelial cells, excitatory neurons, inhibitory neurons, microglia, oligodendrocytes, and oligodendrocyte progenitor cells. Additionally, five distinct subpopulations of both endothelial and microglial cells were also identified, respectively. Key findings included: (1) In endothelial cells, genes involved in synapse organization, such as Insulin Like Growth Factor 1 Receptor (IGF1R) and Fms Related Receptor Tyrosine Kinase 1(FLT1), showed higher expression in females with AD. (2) In microglial cells, genes in the ribosome pathway exhibited higher expression in males without AD compared to females (with or without AD) and males with AD. (3) Chromodomain Helicase DNA Binding Protein 2 (CHD2) negatively regulated gene expression in the ribosome pathway in male microglia, suppressing AD, this finding was further validated in mice. (4) Differences between Asians and Caucasians were observed based on sex and disease status stratification. CONCLUSIONS: IGF1R and FLT1 in endothelial cells contribute to AD in females, while CHD2 negatively regulates ribosome pathway gene expression in male microglia, suppressing AD in humans and mice.


Assuntos
Doença de Alzheimer , Células Endoteliais , Microglia , Receptor IGF Tipo 1 , Receptor 1 de Fatores de Crescimento do Endotélio Vascular , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Feminino , Animais , Masculino , Microglia/metabolismo , Humanos , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Camundongos , Células Endoteliais/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Fatores Sexuais , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Córtex Pré-Frontal/metabolismo , Camundongos Endogâmicos C57BL
20.
J Zhejiang Univ Sci B ; 25(7): 568-580, 2024 Jul 10.
Artigo em Inglês, Chinês | MEDLINE | ID: mdl-39011677

RESUMO

Diabetes mellitus (DM) is a disease syndrome characterized by chronic hyperglycaemia. A long-term high-glucose environment leads to reactive oxygen species (ROS) production and nuclear DNA damage. Human umbilical cord mesenchymal stem cell (HUcMSC) infusion induces significant antidiabetic effects in type 2 diabetes mellitus (T2DM) rats. Insulin-like growth factor 1 (IGF1) receptor (IGF1R) is important in promoting glucose metabolism in diabetes; however, the mechanism by which HUcMSC can treat diabetes through IGF1R and DNA damage repair remains unclear. In this study, a DM rat model was induced with high-fat diet feeding and streptozotocin (STZ) administration and rats were infused four times with HUcMSC. Blood glucose, interleukin-6 (IL-6), IL-10, glomerular basement membrane, and renal function were examined. Proteins that interacted with IGF1R were determined through coimmunoprecipitation assays. The expression of IGF1R, phosphorylated checkpoint kinase 2 (p-CHK2), and phosphorylated protein 53 (p-p53) was examined using immunohistochemistry (IHC) and western blot analysis. Enzyme-linked immunosorbent assay (ELISA) was used to determine the serum levels of 8-hydroxydeoxyguanosine (8-OHdG). Flow cytometry experiments were used to detect the surface markers of HUcMSC. The identification of the morphology and phenotype of HUcMSC was performed by way of oil red "O" staining and Alizarin red staining. DM rats exhibited abnormal blood glucose and IL-6/10 levels and renal function changes in the glomerular basement membrane, increased the expression of IGF1 and IGF1R. IGF1R interacted with CHK2, and the expression of p-CHK2 was significantly decreased in IGF1R-knockdown cells. When cisplatin was used to induce DNA damage, the expression of p-CHK2 was higher than that in the IGF1R-knockdown group without cisplatin treatment. HUcMSC infusion ameliorated abnormalities and preserved kidney structure and function in DM rats. The expression of IGF1, IGF1R, p-CHK2, and p-p53, and the level of 8-OHdG in the DM group increased significantly compared with those in the control group, and decreased after HUcMSC treatment. Our results suggested that IGF1R could interact with CHK2 and mediate DNA damage. HUcMSC infusion protected against kidney injury in DM rats. The underlying mechanisms may include HUcMSC-mediated enhancement of diabetes treatment via the IGF1R-CHK2-p53 signalling pathway.


Assuntos
Quinase do Ponto de Checagem 2 , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Nefropatias Diabéticas , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Ratos Sprague-Dawley , Receptor IGF Tipo 1 , Transdução de Sinais , Proteína Supressora de Tumor p53 , Cordão Umbilical , Animais , Masculino , Ratos , Receptor IGF Tipo 1/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Humanos , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/terapia , Cordão Umbilical/citologia , Quinase do Ponto de Checagem 2/metabolismo , Células-Tronco Mesenquimais/metabolismo , Nefropatias Diabéticas/terapia , Nefropatias Diabéticas/metabolismo , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Experimental/metabolismo , Dieta Hiperlipídica , Dano ao DNA , Glicemia/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...