Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.072
Filtrar
1.
Best Pract Res Clin Endocrinol Metab ; 38(2): 101851, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38087658

RESUMEN

After identification of fibroblast growth factor (FGF) 23 as the pivotal regulator of chronic serum inorganic phosphate (Pi) levels, the etiology of disorders causing hypophosphatemic rickets/osteomalacia has been clarified, and measurement of intact FGF23 serves as a potent tool for differential diagnosis of chronic hypophosphatemia. Additionally, measurement of bone-specific alkaline phosphatase (BAP) is recommended to differentiate acute and subacute hypophosphatemia from chronic hypophosphatemia. This article divides the etiology of chronic hypophosphatemia into 4 groups: A. FGF23 related, B. primary tubular dysfunction, C. disturbance of vitamin D metabolism, and D. parathyroid hormone 1 receptor (PTH1R) mediated. Each group is further divided into its inherited form and acquired form. Topics for each group are described, including "ectopic FGF23 syndrome," "alcohol consumption-induced FGF23-related hypophosphatemia," "anti-mitochondrial antibody associated hypophosphatemia," and "vitamin D-dependent rickets type 3." Finally, a flowchart for differential diagnosis of chronic hypophosphatemia is introduced.


Asunto(s)
Raquitismo Hipofosfatémico Familiar , Hipofosfatemia , Osteomalacia , Humanos , Hipofosfatemia/diagnóstico , Hipofosfatemia/etiología , Raquitismo Hipofosfatémico Familiar/diagnóstico , Raquitismo Hipofosfatémico Familiar/complicaciones , Fosfatos/metabolismo , Factores de Crecimiento de Fibroblastos/fisiología , Osteomalacia/etiología , Osteomalacia/complicaciones , Vitamina D
2.
Sci Signal ; 16(777): eadh8118, 2023 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-36943923

RESUMEN

FGF21-mediated activation of noradrenergic neurons enables recovery from ethanol intoxication in mice.


Asunto(s)
Etanol , Factores de Crecimiento de Fibroblastos , Animales , Ratones , Factores de Crecimiento de Fibroblastos/fisiología , Ratones Endogámicos C57BL
3.
Vitam Horm ; 120: 47-78, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35953117

RESUMEN

Phosphate homeostasis is dependent on the interaction and coordination of four main organ systems: thyroid/parathyroids, gastrointestinal tract, bone and kidneys, and three key hormonal regulators, 1,25-hydroxyvitamin D3, parathyroid hormone and FGF23 with its co- factor klotho. Phosphorus is a critical nutritional element for normal cellular function, but in excess can be toxic to tissues, particularly the vasculature. As phosphate, it also has an important interaction and inter-dependence with calcium and calcium homeostasis sharing some of the same controlling hormones, although this is not covered in our article. We have chosen to provide a current overview of phosphate homeostasis only, focusing on the role of two major organ systems, the gastrointestinal tract and kidneys, and their contribution to the control of phosphate balance. We describe in some detail the mechanisms of intestinal and renal phosphate transport, and compare and contrast their regulation. We also consider a significant example of phosphate imbalance, with phosphate retention, which is chronic kidney disease; why consequent hyperphosphatemia is important, and some of the newer means of managing it.


Asunto(s)
Calcio , Vitamina D , Factores de Crecimiento de Fibroblastos/fisiología , Glucuronidasa/genética , Homeostasis , Humanos , Riñón , Hormona Paratiroidea/fisiología , Fosfatos , Vitamina D/fisiología
4.
Pediatr Radiol ; 52(12): 2290-2305, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35536416

RESUMEN

Phosphorus, predominantly in the form of inorganic phosphate PO4-3, has many essential physiological functions. In the skeleton, phosphate and calcium form the mineral component and phosphate is also essential in regulating function of skeletal cells. Considerable advances have been made in our understanding of phosphate homeostasis since the recognition of fibroblast growth factor-23 (FGF23) as a bone-derived phosphaturic hormone. This second part of a two-part review of disorders of phosphate homeostasis in children covers hypophosphatemic and hyperphosphatemic disorders that are of interest to the pediatric radiologist, emphasizing, but not limited to, those related to abnormalities of FGF23 signaling.


Asunto(s)
Hipofosfatemia , Niño , Humanos , Hipofosfatemia/diagnóstico por imagen , Factores de Crecimiento de Fibroblastos/fisiología , Homeostasis/fisiología , Fosfatos , Huesos
5.
Adv Exp Med Biol ; 1362: 1-6, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35288867

RESUMEN

Systemic phosphate homeostasis is tightly controlled by the delicate cross-organ talk among intestine, kidney, bone, and parathyroid glands. The endocrine regulation of phosphate homeostasis is primarily mediated by fibroblast growth factor 23 (FGF23), vitamin D, and parathyroid hormone (PTH). Bone-derived FGF23 acts on the proximal tubular epithelial cells of the kidney to partly maintain the homeostatic balance of the phosphate. FGF23, through binding with its cell surface receptors in the presence of klotho, can activate downstream signaling kinases to reduce the functionality of the sodium-phosphate (NaPi) co-transporters of the kidney to influence the systemic phosphate homeostasis. Given the complexity of molecular regulation of phosphate homeostasis, providing information on all aspects of its homeostatic control in a single volume of a book is an overwhelming task. As the Editor, I have organized the chapters that I believe will provide necessary information on the physiologic regulation and pathologic dysregulation of phosphate in health and diseases. Readers will be able to use this volume as a quick reference for updated information on phosphate metabolism without prior acquaintance with the field.


Asunto(s)
Factores de Crecimiento de Fibroblastos , Homeostasis , Fosfatos , Factor-23 de Crecimiento de Fibroblastos/fisiología , Factores de Crecimiento de Fibroblastos/fisiología , Homeostasis/fisiología , Humanos , Proteínas Klotho/fisiología , Fosfatos/metabolismo
6.
Adv Exp Med Biol ; 1362: 37-46, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35288871

RESUMEN

Vitamin D plays an essential role in calcium and inorganic phosphate (Pi) homeostasis, maintaining their optimal levels to assure adequate bone mineralization. Vitamin D, as calcitriol (1,25(OH)2D), not only increases intestinal calcium and phosphate absorption but also facilitates their renal reabsorption, leading to elevated serum calcium and phosphate levels. The interaction of 1,25(OH)2D with its receptor (VDR) increases the efficiency of intestinal absorption of calcium to 30-40% and phosphate to nearly 80%. Serum phosphate levels can also influence 1,25(OH)2D and fibroblast growth factor 23 (FGF23) levels, i.e., higher phosphate concentrations suppress vitamin D activation and stimulate parathyroid hormone (PTH) release, while a high FGF23 serum level leads to reduced vitamin D synthesis. In the vitamin D-deficient state, the intestinal calcium absorption decreases and the secretion of PTH increases, which in turn causes the stimulation of 1,25(OH)2D production, resulting in excessive urinary phosphate loss. Maintenance of phosphate homeostasis is essential as hyperphosphatemia is a risk factor of cardiovascular calcification, chronic kidney diseases (CKD), and premature aging, while hypophosphatemia is usually associated with rickets and osteomalacia. This chapter elaborates on the possible interactions between vitamin D and phosphate in health and disease.


Asunto(s)
Fosfatos , Vitamina D , Factores de Crecimiento de Fibroblastos/fisiología , Humanos , Hormona Paratiroidea/metabolismo , Fosfatos/metabolismo , Vitamina D/metabolismo , Vitaminas
7.
Adv Exp Med Biol ; 1362: 151-160, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35288879

RESUMEN

The endocrine regulator proteins, fibroblast growth factor 23 (FGF23) and Klotho have been well studied as mediators of phosphate metabolism. FGF23 has been implicated in the renal excretion of phosphate by limiting the docking of sodium-dependent phosphate transporters, Npt2a and Npt2c, into the luminal side of renal proximal tubular epithelial cells. By limiting Npt2a/c activity in the renal tubular epithelial cells, phosphate is reabsorbed at lower rates and is excreted at higher rates. The action of Klotho is relatively less understood but has been implicated as an FGF23 cofactor in receptor binding. Klotho is mostly synthesized in the distal tubules of the nephron relative to FGF23's activity in proximal renal tubules. The neurological sequelae due to alterations in the FGF23-Klotho axis may be explained by the direct effects of these phosphate-regulating proteins on neuronal tissues or by the roles of these proteins in phosphate metabolism. Hyperphosphatemia has been associated with vascular wall stiffness that may alter blood flow and weakenvessels in the brain. In contrast, hypophosphatemia may alter ATP usage and metabolism in the central nervous system (CNS), leading to neurological compromise. Altered levels of FGF23 and Klotho have both been associated with neurocognitive decline, clinical dementia, memory loss, and poor executive function in humans. Furthermore, FGF23 and Klotho dysregulation has been linked to structural and functional changes of the cardiovascular system with an increased risk of stroke. Subsequent research should focus on characterizing the neuropathology associated with alterations in the FGF23-Klotho system and dysregulated phosphate metabolism.


Asunto(s)
Factor-23 de Crecimiento de Fibroblastos , Hiperfosfatemia , Proteínas Klotho , Fosfatos , Factores de Crecimiento de Fibroblastos/fisiología , Glucuronidasa/genética , Glucuronidasa/metabolismo , Humanos , Fosfatos/metabolismo
8.
Endocrinology ; 163(2)2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34977930

RESUMEN

Luminal breast cancer (BrCa) has a favorable prognosis compared with other tumor subtypes. However, with time, tumors may evolve and lead to disease progression; thus, there is a great interest in unraveling the mechanisms that drive tumor metastasis and endocrine resistance. In this review, we focus on one of the many pathways that have been involved in tumor progression, the fibroblast growth factor/fibroblast growth factor receptor (FGFR) axis. We emphasize in data obtained from in vivo experimental models that we believe that in luminal BrCa, tumor growth relies in a crosstalk with the stromal tissue. We revisited the studies that illustrate the interaction between hormone receptors and FGFR. We also highlight the most frequent alterations found in BrCa cell lines and provide a short review on the trials that use FGFR inhibitors in combination with endocrine therapies. Analysis of these data suggests there are many players involved in this pathway that might be also targeted to decrease FGF signaling, in addition to specific FGFR inhibitors that may be exploited to increase their efficacy.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Factores de Crecimiento de Fibroblastos/fisiología , Receptores de Factores de Crecimiento de Fibroblastos/fisiología , Receptores de Esteroides/fisiología , Transducción de Señal/fisiología , Animales , Inhibidores de la Aromatasa/uso terapéutico , Neoplasias de la Mama/química , Neoplasias de la Mama/terapia , Línea Celular Tumoral , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Receptor alfa de Estrógeno/análisis , Femenino , Factores de Crecimiento de Fibroblastos/genética , Amplificación de Genes , Humanos , Ratones , Mutación , Receptor Cross-Talk/fisiología , Receptores de Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptores de Factores de Crecimiento de Fibroblastos/genética
9.
Mol Biol Rep ; 49(2): 1413-1427, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34731369

RESUMEN

Alzheimer's disease (AD), acknowledged as the most common progressive neurodegenerative disorder, is the leading cause of dementia in the elderly. The characteristic pathologic hallmarks of AD-including the deposition of extracellular senile plaques (SP) formation, intracellular neurofibrillary tangles, and synaptic loss, along with prominent vascular dysfunction and cognitive impairment-have been observed in patients. Fibroblast growth factors (FGFs), originally characterized as angiogenic factors, are a large family of signaling molecules that are implicated in a wide range of biological functions in brain development, maintenance and repair, as well as in the pathogenesis of brain-related disorders including AD. Many studies have focused on the implication of FGFs in AD pathophysiology. In this review, we will provide a summary of recent findings regarding the role of FGFs and their receptors in the pathogenesis of AD, and discuss the possible opportunities for targeting these molecules as novel treatment strategies in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/fisiología , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Disfunción Cognitiva/metabolismo , Femenino , Factores de Crecimiento de Fibroblastos/genética , Humanos , Masculino , Persona de Mediana Edad , Ovillos Neurofibrilares/metabolismo , Ovillos Neurofibrilares/patología , Placa Amiloide/metabolismo , Placa Amiloide/patología
10.
Biomed Pharmacother ; 146: 112524, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34906775

RESUMEN

Human fibroblast growth factor 19 (FGF19) has become a potential therapeutic target for metabolic-related diseases. However, the effects of FGF19 on obesity-induced bone loss have not been completely elucidated. The aim of this study was to investigate the protective effects of FGF19 in high-fat diet (HFD)-fed obese mice and palmitic acid (PA)-treated osteoblasts and to further explore its underlying mechanisms. In vivo, we found that FGF19 alleviated the decreased bone mineral density (BMD) induced by HFD. Micro-CT analysis of femur samples and histological analysis indicated that FGF19 alleviated HFD-induced loss of bone trabeculae and damage to the bone trabecular structure. In vitro, the results suggested that FGF19 ameliorated the PA-induced decline in osteoblast proliferation, increased cell death and impaired cell morphology. Additionally, FGF19 protected against the decline in activation of alkaline phosphatase (ALP) and protein expression of Collagen-1, Runx-2, and osteopontin (OPN) induced by PA. Furthermore, FGF19 might enhance osteogenic differentiation via the Wnt/ß-catenin pathway and inhibit osteoclastogenesis by regulating the osteoprotegerin (OPG)/receptor activator of NF-κB ligand (RANKL) axis, thus attenuating the negative effect of PA in osteoblasts. In conclusion, our results suggested that FGF19 might promote osteogenic differentiation partially through activation of the Wnt/ß-catenin pathway and alleviate obesity-induced bone loss.


Asunto(s)
Factores de Crecimiento de Fibroblastos , Obesidad , Osteogénesis , Osteoporosis , Animales , Diferenciación Celular , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/fisiología , Ratones , Obesidad/complicaciones , Osteoblastos , Osteoporosis/etiología , Osteoporosis/genética , Ligando RANK/metabolismo , Vía de Señalización Wnt
11.
Neuropharmacology ; 202: 108858, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34715121

RESUMEN

Because of increased opioid misuse, there is a need to identify new targets for minimizing opioid tolerance, and physical and psychological dependence. Previous studies showed that fibroblast growth factor 21 (FGF21) decreased alcohol and sweet preference in mice. In this study, FGF21-transgenic (FGF21-Tg) mice, expressing high FGF21 serum levels, and wildtype (WT) C57BL/6J littermates were treated with morphine and saline to determine if differences exist in their physiological and behavioral responses to opioids. FGF21-Tg mice displayed reduced preference for morphine in the conditioned place preference assay compared to WT littermates. Similarly, FGF21-Tg mice had an attenuation of the magnitude and rate of acute morphine antinociceptive tolerance development, and acute and chronic morphine physical dependence, but exhibited no change in chronic morphine antinociceptive tolerance. The ED50 values for morphine-induced antinociception in the 55 °C hot plate and the 55 °C warm-water tail withdrawal assays were similar in both strains of mice. Likewise, FGF21-Tg and WT littermates had comparable responses to morphine-induced respiratory depression. Overall, FGF21-Tg mice had a decrease in the development of acute analgesic tolerance, and the development of physical dependence, and morphine preference. FGF21 and its receptor have therapeutic potential for reducing opioid withdrawal symptoms and craving, and augmenting opioid therapeutics for acute pain patients to minimize tolerance development.


Asunto(s)
Tolerancia a Medicamentos/genética , Factores de Crecimiento de Fibroblastos/sangre , Factores de Crecimiento de Fibroblastos/fisiología , Dependencia de Morfina/genética , Morfina/efectos adversos , Nocicepción/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Transgénicos , Insuficiencia Respiratoria/inducido químicamente , Insuficiencia Respiratoria/genética , Síndrome de Abstinencia a Sustancias/genética , Síndrome de Abstinencia a Sustancias/terapia
12.
Front Endocrinol (Lausanne) ; 12: 744868, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34721299

RESUMEN

Diabetic foot ulcer (DFU) is a combination of neuropathy and various degrees of peripheral vasculopathy in diabetic patients resulting in lower extremity infection, ulcer formation, and deep-tissue necrosis. The difficulty of wound healing in diabetic patients is caused by a high glucose environment and various biological factors in the patient. The patients' skin local microenvironment changes and immune chemotactic response dysfunction. Wounds are easy to be damaged and ulcerated repeatedly, but difficult to heal, and eventually develop into chronic ulcers. DFU is a complex biological process in which many cells interact with each other. A variety of growth factors released from wounds are necessary for coordination and promotion of healing. Fibroblast growth factor (FGF) is a family of cell signaling proteins, which can mediate various processes such as angiogenesis, wound healing, metabolic regulation and embryonic development through its specific receptors. FGF can stimulate angiogenesis and proliferation of fibroblasts, and it is a powerful angiogenesis factor. Twenty-three subtypes have been identified and divided into seven subfamilies. Traditional treatments for DFU can only remove necrotic tissue, delay disease progression, and have a limited ability to repair wounds. In recent years, with the increasing understanding of the function of FGF, more and more researchers have been applying FGF-1, FGF-2, FGF-4, FGF-7, FGF-21 and FGF-23 topically to DFU with good therapeutic effects. This review elaborates on the recently developed FGF family members, outlining their mechanisms of action, and describing their potential therapeutics in DFU.


Asunto(s)
Pie Diabético/tratamiento farmacológico , Factores de Crecimiento de Fibroblastos/uso terapéutico , Animales , Factores de Crecimiento de Fibroblastos/fisiología , Humanos , Transducción de Señal/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos
13.
Life Sci Alliance ; 4(11)2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34548382

RESUMEN

FGFs and their high-affinity receptors (FGFRs) play key roles in development, tissue repair, and disease. Because FGFRs bind overlapping sets of ligands, their individual functions cannot be determined using ligand stimulation. Here, we generated a light-activated FGFR2 variant (OptoR2) to selectively activate signaling by the major FGFR in keratinocytes. Illumination of OptoR2-expressing HEK 293T cells activated FGFR signaling with remarkable temporal precision and promoted cell migration and proliferation. In murine and human keratinocytes, OptoR2 activation rapidly induced the classical FGFR signaling pathways and expression of FGF target genes. Surprisingly, multi-level counter-regulation occurred in keratinocytes in vitro and in transgenic mice in vivo, including OptoR2 down-regulation and loss of responsiveness to light activation. These results demonstrate unexpected cell type-specific limitations of optogenetic FGFRs in long-term in vitro and in vivo settings and highlight the complex consequences of transferring optogenetic cell signaling tools into their relevant cellular contexts.


Asunto(s)
Queratinocitos/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Animales , Femenino , Factores de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/fisiología , Células HEK293 , Humanos , Queratinocitos/fisiología , Ligandos , Luz , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/genética , Receptores de Factores de Crecimiento de Fibroblastos/fisiología , Transducción de Señal
14.
Cancer Sci ; 112(11): 4655-4668, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34533854

RESUMEN

Platinum-based regimens are the most widely used chemotherapy regimens, but cancer cells often develop resistance, which impedes therapy outcome for patients. Previous studies have shown that fibroblast growth factor 13 (FGF13) is associated with resistance to platinum drugs in HeLa cells. However, the mechanism and universality of this effect have not been clarified. Here, we found that FGF13 was associated with poor platinum-based chemotherapy outcomes in a variety of cancers, such as lung, endometrial, and cervical cancers, through bioinformatics analysis. We then found that FGF13 simultaneously regulates the expression and distribution of hCTR1 and ATP7A in cancer cells, causes reduced platinum influx, and promotes platinum sequestration and efflux upon cisplatin exposure. We subsequently observed that FGF13-mediated platinum resistance requires the microtubule-stabilizing effect of FGF13. Only overexpression of FGF13 with the -SMIYRQQQ- tubulin-binding domain could induce the platinum resistance effect. This phenomenon was also observed in SK-MES-1 cells, KLE cells, and 5637 cells. Our research reveals the mechanism of FGF13-induced platinum drug resistance and suggests that FGF13 can be a sensibilization target and prognostic biomarker for chemotherapy.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Transportador de Cobre 1/metabolismo , ATPasas Transportadoras de Cobre/metabolismo , Resistencia a Antineoplásicos , Factores de Crecimiento de Fibroblastos/fisiología , Células A549 , Antineoplásicos/metabolismo , Línea Celular Tumoral , Cisplatino/metabolismo , Neoplasias Endometriales/tratamiento farmacológico , Neoplasias Endometriales/metabolismo , Femenino , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Microtúbulos/efectos de los fármacos , Compuestos de Platino/metabolismo , Compuestos de Platino/farmacología , Moduladores de Tubulina/metabolismo , Moduladores de Tubulina/farmacología , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/metabolismo
15.
J Exp Zool B Mol Dev Evol ; 336(7): 529-539, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34387925

RESUMEN

Amphibians have a very high capacity for regeneration among tetrapods. This superior regeneration capability in amphibians can be observed in limbs, the tail, teeth, external gills, the heart, and some internal organs. The mechanisms underlying the superior organ regeneration capability have been studied for a long time. Limb regeneration has been investigated as the representative phenomenon for organ-level regeneration. In limb regeneration, a prominent difference between regenerative and nonregenerative animals after limb amputation is blastema formation. A regeneration blastema requires the presence of nerves in the stump region. Thus, nerve regulation is responsible for blastema induction, and it has received much attention. Nerve regulation in regeneration has been investigated using the limb regeneration model and newly established alternative experimental model called the accessory limb model. Previous studies have identified some candidate genes that act as neural factors in limb regeneration, and these studies also clarified related events in early limb regeneration. Consistent with the nervous regulation and related events in limb regeneration, similar regeneration mechanisms in other organs have been discovered. This review especially focuses on the role of nerve-mediated fibroblast growth factor in the initiation phase of organ regeneration. Comparison of the initiation mechanisms for regeneration in various amphibian organs allows speculation about a fundamental regenerative process.


Asunto(s)
Anfibios , Extremidades , Factores de Crecimiento de Fibroblastos/fisiología , Regeneración , Animales , Extremidades/inervación , Cola (estructura animal)
16.
Placenta ; 112: 81-88, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34329971

RESUMEN

INTRODUCTION: This study aimed to evaluate whether FGF19 can alleviate insulin resistance and change the expression of placental IRS1/GLUTs. METHODS: Mice transgenic for Fgf15 (the murine homologue of human FGF19) were constructed, and human recombinant FGF19 was administered to pregnant high-fat diet mice. Then, glycolipid metabolism parameters and the weight of foetus and placenta were observed. The expression levels of key molecules of the insulin signalling pathway and glucose transporters in placentae were detected by qRT-PCR and western blotting. Primary trophoblasts and JAR cells were cultured in high-glucose medium, and FGF19 was added to observe its regulatory effects on IRS1/GLUTs. RESULTS: Overexpressing FGF15 or exogenously administering FGF19 reduced the levels of fasting blood glucose, HOMA-IR, triglycerides, and free fatty acids in pregnant high-fat diet mice compared to control mice (P < 0.05). FGF15/FGF19 did not significantly affect placental weight, foetal weight or litter size (P > 0.05). In addition, FGF15/FGF19 upregulated the expression of p-IRS1 and GLUT4 in the placentae of high-fat diet mice and upregulated GLUT1 levels in the placentae of normal diet-fed mice (P < 0.05), while it did not significantly alter total IRS1 and GLUT3 levels (P > 0.05). Consistent with the results of the animal experiments, FGF19 increased the expression of p-IRS1 and GLUT4 in trophoblast cells cultured in high-glucose medium (P < 0.05). DISCUSSION: Overexpressing FGF15 or administering FGF19 to pregnant high-fat diet mice can improve glycolipid metabolism and alleviate systemic and local insulin resistance. The possible underlying mechanism may involve upregulation of placental expression of p-IRS1 and GLUT4.


Asunto(s)
Factores de Crecimiento de Fibroblastos/fisiología , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Resistencia a la Insulina , Placenta/metabolismo , Animales , Dieta Alta en Grasa , Femenino , Humanos , Ratones Endogámicos C57BL , Ratones Transgénicos , Embarazo , Trofoblastos/metabolismo
17.
PLoS Comput Biol ; 17(6): e1009077, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34161317

RESUMEN

The vertebrate hindbrain is segmented into rhombomeres (r) initially defined by distinct domains of gene expression. Previous studies have shown that noise-induced gene regulation and cell sorting are critical for the sharpening of rhombomere boundaries, which start out rough in the forming neural plate (NP) and sharpen over time. However, the mechanisms controlling simultaneous formation of multiple rhombomeres and accuracy in their sizes are unclear. We have developed a stochastic multiscale cell-based model that explicitly incorporates dynamic morphogenetic changes (i.e. convergent-extension of the NP), multiple morphogens, and gene regulatory networks to investigate the formation of rhombomeres and their corresponding boundaries in the zebrafish hindbrain. During pattern initiation, the short-range signal, fibroblast growth factor (FGF), works together with the longer-range morphogen, retinoic acid (RA), to specify all of these boundaries and maintain accurately sized segments with sharp boundaries. At later stages of patterning, we show a nonlinear change in the shape of rhombomeres with rapid left-right narrowing of the NP followed by slower dynamics. Rapid initial convergence improves boundary sharpness and segment size by regulating cell sorting and cell fate both independently and coordinately. Overall, multiple morphogens and tissue dynamics synergize to regulate the sizes and boundaries of multiple segments during development.


Asunto(s)
Tipificación del Cuerpo/fisiología , Modelos Biológicos , Pez Cebra/embriología , Animales , Tipificación del Cuerpo/genética , Biología Computacional , Desarrollo Embrionario/genética , Desarrollo Embrionario/fisiología , Factores de Crecimiento de Fibroblastos/fisiología , Regulación del Desarrollo de la Expresión Génica , Sustancias de Crecimiento/fisiología , Rombencéfalo/citología , Rombencéfalo/embriología , Transducción de Señal , Procesos Estocásticos , Tretinoina/fisiología , Pez Cebra/genética
18.
FASEB J ; 35(6): e21663, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34042217

RESUMEN

cAMP responsive element-binding protein H (CREBH) is a hepatic transcription factor to be activated during fasting. We generated CREBH knock-in flox mice, and then generated liver-specific CREBH transgenic (CREBH L-Tg) mice in an active form. CREBH L-Tg mice showed a delay in growth in the postnatal stage. Plasma growth hormone (GH) levels were significantly increased in CREBH L-Tg mice, but plasma insulin-like growth factor 1 (IGF1) levels were significantly decreased, indicating GH resistance. In addition, CREBH overexpression significantly increased hepatic mRNA and plasma levels of FGF21, which is thought to be as one of the causes of growth delay. However, the additional ablation of FGF21 in CREBH L-Tg mice could not correct GH resistance at all. CREBH L-Tg mice sustained GH receptor (GHR) reduction and the increase of IGF binding protein 1 (IGFBP1) in the liver regardless of FGF21. As GHR is a first step in GH signaling, the reduction of GHR leads to impairment of GH signaling. These data suggest that CREBH negatively regulates growth in the postnatal growth stage via various pathways as an abundant energy response by antagonizing GH signaling.


Asunto(s)
Composición Corporal , Índice de Masa Corporal , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/fisiología , Factores de Crecimiento de Fibroblastos/fisiología , Regulación del Desarrollo de la Expresión Génica , Hormona del Crecimiento/metabolismo , Hígado/metabolismo , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Transducción de Señal
19.
Diabetes ; 70(7): 1443-1457, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33883213

RESUMEN

The counterregulatory response to hypoglycemia is an essential survival function. It is controlled by an integrated network of glucose-responsive neurons, which trigger endogenous glucose production to restore normoglycemia. The complexity of this glucoregulatory network is, however, only partly characterized. In a genetic screen of a panel of recombinant inbred mice we previously identified Fgf15, expressed in neurons of the dorsomedial hypothalamus (DMH), as a negative regulator of glucagon secretion. Here, we report on the generation of Fgf15CretdTomato mice and their use to further characterize these neurons. We show that they were glutamatergic and comprised glucose-inhibited and glucose-excited neurons. When activated by chemogenetics, Fgf15 neurons prevented the increase in vagal nerve firing and the secretion of glucagon normally triggered by insulin-induced hypoglycemia. On the other hand, they increased the activity of the sympathetic nerve in the basal state and prevented its silencing by glucose overload. Higher sympathetic tone increased hepatic Creb1 phosphorylation, Pck1 mRNA expression, and hepatic glucose production leading to glucose intolerance. Thus, Fgf15 neurons of the DMH participate in the counterregulatory response to hypoglycemia by a direct adrenergic stimulation of hepatic glucose production while suppressing vagally induced glucagon secretion. This study provides new insights into the complex neuronal network that prevents the development of hypoglycemia.


Asunto(s)
Factores de Crecimiento de Fibroblastos/fisiología , Glucagón/metabolismo , Gluconeogénesis/fisiología , Hipotálamo/metabolismo , Hígado/metabolismo , Neuronas/fisiología , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/fisiología , Femenino , Hipoglucemia/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Sistema Nervioso Simpático/fisiología
20.
J Cell Mol Med ; 25(7): 3585-3600, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33751819

RESUMEN

Obesity is associated with biological dysfunction in skeletal muscle. As a condition of obesity accompanied by muscle mass loss and physical dysfunction, sarcopenic obesity (SO) has become a novel public health problem. Human fibroblast growth factor 19 (FGF19) plays a therapeutic role in metabolic diseases. However, the protective effects of FGF19 on skeletal muscle in obesity and SO are still not completely understood. Our results showed that FGF19 administration improved muscle loss and grip strength in young and aged mice fed a high-fat diet (HFD). Increases in muscle atrophy markers (FOXO-3, Atrogin-1, MuRF-1) were abrogated by FGF19 in palmitic acid (PA)-treated C2C12 myotubes and in the skeletal muscle of HFD-fed mice. FGF19 not only reduced HFD-induced body weight gain, excessive lipid accumulation and hyperlipidaemia but also promoted energy expenditure (PGC-1α, UCP-1, PPAR-γ) in brown adipose tissue (BAT). FGF19 treatment restored PA- and HFD-induced hyperglycaemia, impaired glucose tolerance and insulin resistance (IRS-1, GLUT-4) and mitigated the PA- and HFD-induced decrease in FNDC-5/irisin expression. However, these beneficial effects of FGF19 on skeletal muscle were abolished by inhibiting AMPK, SIRT-1 and PGC-1α expression. Taken together, this study suggests that FGF19 protects skeletal muscle against obesity-induced muscle atrophy, metabolic derangement and abnormal irisin secretion partially through the AMPK/SIRT-1/PGC-α signalling pathway, which might be a potential therapeutic target for obesity and SO.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Factores de Crecimiento de Fibroblastos/fisiología , Fibronectinas/metabolismo , Atrofia Muscular/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Sirtuina 1/metabolismo , Tejido Adiposo Pardo/efectos de los fármacos , Animales , Línea Celular , Dieta Alta en Grasa , Factores de Crecimiento de Fibroblastos/farmacología , Hiperglucemia/metabolismo , Hiperlipidemias/metabolismo , Resistencia a la Insulina , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Obesidad/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA