Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.095
Filtrar
1.
Vitam Horm ; 120: 305-343, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35953115

RESUMEN

The number of the patients with chronic kidney disease is now increasing in the world. The pathophysiology of renal hyperparathyroidism is closely associated with Klotho-FGF-endocrine axes, which must be solved definitively as early as possible. It was revealed that the expression of fgf23 is activated by calciprotein particles, which induces vascular ossification. And it is well known that phosphorus overload directly increases parathyroid hormone and hyperparathyroid bone disease develops in those subjects. On the other hand, low turnover bone disease is often recently. Both the patients with chronic kidney disease suffering from hyperparathyroid bone disease or low turnover bone disease are associated with increased fracture risk. Micropetrosis may be one of the causes of increased fracture risk in the subjects with low turnover bone disease. In this chapter, we now describe the diagnosis, pathophysiology and treatments of renal hyperparathyroidism.


Asunto(s)
Enfermedades Óseas , Hiperparatiroidismo , Insuficiencia Renal Crónica , Calcio/metabolismo , Humanos , Hiperparatiroidismo/metabolismo , Hormona Paratiroidea/metabolismo
2.
J Endocrinol ; 254(1): 13-26, 2022 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-35638565

RESUMEN

GS, the stimulatory heterotrimeric G protein, is an essential regulator of osteogenesis and bone turnover. To determine if increasing GαS in osteoblasts alters bone responses to hyperparathyroidism, we used a transgenic mouse line overexpressing GαS in osteoblasts (GS-Tg mice). Primary osteoblasts from GS-Tg mice showed increased basal and parathyroid hormone (PTH)-stimulated cAMP and greater responses to PTH than cells from WT mice. Skeletal responses to 2-week continuous PTH administration (cPTH) in female mice resulted in trabecular bone loss in WT mice but 74% and 34% increase in trabecular bone mass in long bones and vertebrae, respectively, in GS-Tg mice. Vertebral biomechanical strength was compromised by cPTH treatment in WT mice but not in GS-Tg. Increased peritrabecular fibrosis was greatly increased by cPTH in Gs-Tg compared to WT mice and corresponded with greater increases in Wnt pathway proteins in trabecular bone. Cortical bone responded negatively to cPTH in WT and Gs-Tg mice with large increases in porosity, decreased cortical thickness and compromised biomechanical properties. These results demonstrate that hyperparathyroidism can increase trabecular bone when GS expression and cAMP stimulation in osteoblasts are increased but this is not the case in cortical bone where increased GS expression exacerbates cortical bone loss.


Asunto(s)
Hiperparatiroidismo , Osteoblastos , Animales , Huesos/metabolismo , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Hiperparatiroidismo/metabolismo , Ratones , Ratones Transgénicos , Osteoblastos/metabolismo , Hormona Paratiroidea/farmacología , Proteínas Wnt
3.
Nat Commun ; 13(1): 771, 2022 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-35140213

RESUMEN

Parathyroid hormone (PTH) plays crucial role in maintaining calcium and phosphorus homeostasis. In the progression of secondary hyperparathyroidism (SHPT), expression of calcium-sensing receptors (CaSR) in the parathyroid gland decreases, which leads to persistent hypersecretion of PTH. How to precisely manipulate PTH secretion in parathyroid tissue and underlying molecular mechanism is not clear. Here, we establish an optogenetic approach that bypasses CaSR to inhibit PTH secretion in human hyperplastic parathyroid cells. We found that optogenetic stimulation elevates intracellular calcium, inhibits both PTH synthesis and secretion in human parathyroid cells. Long-term pulsatile PTH secretion induced by light stimulation prevented hyperplastic parathyroid tissue-induced bone loss by influencing the bone remodeling in mice. The effects are mediated by light stimulation of opsin expressing parathyroid cells and other type of cells in parathyroid tissue. Our study provides a strategy to regulate release of PTH and associated bone loss of SHPT through an optogenetic approach.


Asunto(s)
Calcio/metabolismo , Hiperparatiroidismo Secundario/metabolismo , Optogenética , Hormona Paratiroidea/metabolismo , Huesos , Homeostasis , Humanos , Hiperparatiroidismo/metabolismo , Hiperparatiroidismo/patología , Hiperparatiroidismo Secundario/patología , Hiperplasia/metabolismo , Glándulas Paratiroides , Receptores Sensibles al Calcio/metabolismo
4.
Eur J Endocrinol ; 186(3): 351-366, 2022 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-35038313

RESUMEN

OBJECTIVE: The aim of this study was to analyze variants of the gene glial cells missing-2 (GCM2), encoding a parathyroid cell-specific transcription factor, in familial hypoparathyroidism and in familial isolated hyperparathyroidism (FIHP) without and with parathyroid carcinoma. DESIGN: We characterized 2 families with hypoparathyroidism and 19 with FIHP in which we examined the mechanism of action of GCM2 variants. METHODS: Leukocyte DNA of hypoparathyroid individuals was Sanger sequenced for CASR, PTH, GNA11 and GCM2 mutations. DNA of hyperparathyroid individuals underwent MEN1, CDKN1B, CDC73, CASR, RET and GCM2 sequencing. The actions of identified GCM2 variants were evaluated by in vitro functional analyses. RESULTS: A novel homozygous p.R67C GCM2 mutation which failed to stimulate transcriptional activity in a luciferase assay was identified in affected members of two hypoparathyroid families. Oligonucleotide pull-down assay and in silico structural modeling indicated that this mutant had lost the ability to bind the consensus GCM recognition sequence of DNA. Two novel (p.I383M and p.T386S) and one previously reported (p.Y394S) heterozygous GCM2 variants that lie within a C-terminal conserved inhibitory domain were identified in three affected individuals of the hyperparathyroid families. One family member, heterozygous for p.I138M, had parathyroid carcinoma (PC), and a heterozygous p.V382M variant was found in another patient affected by sporadic PC. These variants exerted significantly enhanced in vitrotranscriptional activity, including increased stimulation of the PTH promoter. CONCLUSIONS: We provide evidence that two novel GCM2 R67C inactivating mutations with an inability to bind DNA are causative of hypoparathyroidism. Additionally, we provide evidence that two novel GCM2 variants increased transactivation of the PTH promoter in vitro and are associated with FIHP. Furthermore, our studies suggest that activating GCM2 variants may contribute to facilitating more aggressive parathyroid disease.


Asunto(s)
Hiperparatiroidismo/genética , Hipoparatiroidismo/genética , Mutación , Proteínas Nucleares/genética , Neoplasias de las Paratiroides/genética , Factores de Transcripción/genética , Adulto , Anciano , Anciano de 80 o más Años , Animales , Sitios de Unión , Calcio/sangre , Calcio/orina , ADN/sangre , ADN/metabolismo , Femenino , Humanos , Hiperparatiroidismo/metabolismo , Hiperparatiroidismo/patología , Hipoparatiroidismo/sangre , Lactante , Masculino , Ratones , Persona de Mediana Edad , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Glándulas Paratiroides/patología , Glándulas Paratiroides/cirugía , Hormona Paratiroidea/sangre , Hormona Paratiroidea/genética , Neoplasias de las Paratiroides/metabolismo , Neoplasias de las Paratiroides/patología , Linaje , Regiones Promotoras Genéticas , Análisis de Secuencia de ADN , Factores de Transcripción/química , Factores de Transcripción/metabolismo
5.
Curr Mol Pharmacol ; 15(2): 292-305, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-33573587

RESUMEN

Hormonal coordination is tightly regulated within the human body and thus regulates human physiology. The parathyroid hormone (PTH), a member of the endocrine system, regulates the calcium and phosphate level within the human body. Under non-physiological conditions, PTH levels get upregulated (hyperparathyroidism) or downregulated (hypoparathyroidism) due to external or internal factors. In case of hyperparathyroidism, elevated PTH stimulates cellular receptors present in the bones, kidneys, and intestines to increase the blood calcium level, leading to calcium deposition. This eventually causes various symptoms, including kidney stones. Currently, there is no known medication that directly targets PTH in order to suppress its function. Therefore, it is of great interest to find novel small molecules or any other means that can modulate PTH function. The molecular signaling of PTH starts by binding its N-terminus to the G-protein coupled PTH1/2 receptor. Therefore, any intervention that affects the N-terminus of PTH could be a lead candidate for treating hyperparathyroidism. As a proof-of-concept, there are various possibilities to inhibit molecular PTH function by (i) a small molecule, (ii) N-terminal PTH phosphorylation, (iii) fibril formation and (iv) residue-specific mutations. These modifications put PTH into an inactive state, which will be discussed in detail in this review article. We anticipate that exploring small molecules or other means that affect the N-terminus of PTH could be lead candidates in combating hyperparathyroidism.


Asunto(s)
Hiperparatiroidismo , Hormona Paratiroidea , Calcio/metabolismo , Descubrimiento de Drogas , Humanos , Hiperparatiroidismo/tratamiento farmacológico , Hiperparatiroidismo/metabolismo , Riñón/metabolismo , Hormona Paratiroidea/metabolismo
6.
Front Endocrinol (Lausanne) ; 12: 631680, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34054720

RESUMEN

Purpose: Hyperparathyroidism is the third most common endocrine disease. Parathyroid adenoma (PA) accounts for approximately 85% of cases of primary hyperparathyroidism, but the molecular mechanism is not fully understood. Herein, we aimed to investigate the genetic and transcriptomic profiles of sporadic PA. Methods: Whole-exome sequencing (WES) and transcriptome sequencing (RNA-seq) of 41 patients with PA and RNA-seq of 5 normal parathyroid tissues were performed. Gene mutations and characterized expression changes were identified. To elucidate the molecular mechanism underlying PA, unsupervised consensus clustering of RNA-seq data was performed. The correlations between the sequencing data and clinicopathological features of these patients were analyzed. Results: Previously reported PA driver gene mutations, such as MEN1 (9/41), mTOR (4/41), ZFX (3/41), CASR (3/41), EZH2 (2/41) and FAT1 (2/41), were also identified in our cohort. Furthermore, somatic mutation of EZH1, which had not been reported in PA, was found in 4 samples. RNA-seq showed that the expression levels of 84 genes were upregulated and 646 were downregulated in PA samples compared with normal samples. Unsupervised clustering analysis of RNA-seq data clustered these patients into 10 subgroups related to mutation or abnormal expression of a group of potential pathogenic genes. Conclusion: MEN1, EZH2, CASR, EZH1, ZFX, mTOR and FAT1 mutations in PA were revealed. According to the RNA-seq data clustering analysis, cyclin D1, ß-catenin, VDR, CASR and GCM2 may be important factors contributing to the PA gene expression profile.


Asunto(s)
Adenoma/genética , Adenoma/metabolismo , Exoma , Neoplasias de las Paratiroides/genética , Neoplasias de las Paratiroides/metabolismo , Transcriptoma , Adulto , Anciano , Cadherinas/genética , Cadherinas/metabolismo , Análisis por Conglomerados , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Femenino , Regulación de la Expresión Génica , Genómica , Humanos , Hiperparatiroidismo/genética , Hiperparatiroidismo/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Persona de Mediana Edad , Mutación , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , RNA-Seq , Receptores Sensibles al Calcio/genética , Receptores Sensibles al Calcio/metabolismo , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Bancos de Tejidos
7.
Adv Clin Chem ; 101: 41-93, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33706890

RESUMEN

Parathyroid hormone is an essential regulator of extracellular calcium and phosphate. PTH enhances calcium reabsorption while inhibiting phosphate reabsorption in the kidneys, increases the synthesis of 1,25-dihydroxyvitamin D, which then increases gastrointestinal absorption of calcium, and increases bone resorption to increase calcium and phosphate. Parathyroid disease can be an isolated endocrine disorder or part of a complex syndrome. Genetic mutations can account for diseases of parathyroid gland formulation, dysregulation of parathyroid hormone synthesis or secretion, and destruction of the parathyroid glands. Over the years, a number of different options are available for the treatment of different types of parathyroid disease. Therapeutic options include surgical removal of hypersecreting parathyroid tissue, administration of parathyroid hormone, vitamin D, activated vitamin D, calcium, phosphate binders, calcium-sensing receptor, and vitamin D receptor activators to name a few. The accurate assessment of parathyroid hormone also provides essential biochemical information to properly diagnose parathyroid disease. Currently available immunoassays may overestimate or underestimate bioactive parathyroid hormone because of interferences from truncated parathyroid hormone fragments, phosphorylation of parathyroid hormone, and oxidation of amino acids of parathyroid hormone.


Asunto(s)
Calcio/metabolismo , Hormona Paratiroidea/metabolismo , Bicarbonatos/metabolismo , Desarrollo Óseo , Huesos/metabolismo , Calcio/sangre , Regulación de la Expresión Génica , Homeostasis , Humanos , Hiperparatiroidismo/diagnóstico , Hiperparatiroidismo/metabolismo , Hiperparatiroidismo/patología , Hipoparatiroidismo/diagnóstico , Hipoparatiroidismo/metabolismo , Hipoparatiroidismo/patología , Hormona Paratiroidea/genética , Fosfatos/metabolismo , Receptor de Hormona Paratiroídea Tipo 1/metabolismo
8.
Am J Kidney Dis ; 78(4): 582-589, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33774081

RESUMEN

Osteoporosis is defined as a skeletal disorder of compromised bone strength predisposing those affected to an elevated risk of fracture. However, based on bone histology, osteoporosis is only part of a spectrum of skeletal complications that includes osteomalacia and the various forms of renal osteodystrophy of chronic kidney disease-mineral and bone disorder (CKD-MBD). In addition, the label "kidney-induced osteoporosis" has been proposed, even though the changes caused by CKD do not qualify as osteoporosis by the histological diagnosis. It is clear, therefore, that such terminology may not be helpful diagnostically or in making treatment decisions. A new label, "CKD-MBD/osteoporosis" could be a more appropriate term because it brings osteoporosis under the official label of CKD-MBD. Neither laboratory nor noninvasive diagnostic investigations can discriminate osteoporosis from the several forms of renal osteodystrophy. Transiliac crest bone biopsy can make the diagnosis of osteoporosis by exclusion of other kidney-associated bone diseases, but its availability is limited. Recently, a classification of metabolic bone diseases based on bone turnover, from low to high, together with mineralization and bone volume, has been proposed. Therapeutically, no antifracture treatments have been approved by the US Food and Drug Administration for patients with kidney-associated bone disease. Agents that suppress parathyroid hormone (vitamin D analogues and calcimimetics) are used to treat hyperparathyroid bone disease. Antiresorptive and osteoanabolic agents approved for osteoporosis are being used off-label to treat CKD stages 3b-5 in high-risk patients. It has now been suggested that intermittent administration of parathyroid hormone as early as CKD stage 2 could be an effective management strategy. If confirmed in clinical trials, it could mitigate the retention of phosphorus and subsequently the rise in fibroblast growth factor 23 and may be beneficial for coexisting osteoporosis.


Asunto(s)
Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica/epidemiología , Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica/metabolismo , Osteoporosis/epidemiología , Osteoporosis/metabolismo , Anabolizantes/farmacología , Anabolizantes/uso terapéutico , Enfermedades Óseas Metabólicas/epidemiología , Enfermedades Óseas Metabólicas/metabolismo , Enfermedades Óseas Metabólicas/terapia , Remodelación Ósea/efectos de los fármacos , Remodelación Ósea/fisiología , Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica/terapia , Factor-23 de Crecimiento de Fibroblastos , Humanos , Hiperparatiroidismo/sangre , Hiperparatiroidismo/epidemiología , Hiperparatiroidismo/metabolismo , Hiperparatiroidismo/terapia , Osteoporosis/terapia , Hormona Paratiroidea/metabolismo , Vitamina D/farmacología , Vitamina D/uso terapéutico
9.
Am J Otolaryngol ; 42(3): 102886, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33460974

RESUMEN

BACKGROUND: Surgery is currently the only treatment option for patients with primary hyperparathyroidism (PHPT). Recently, minimally invasive parathyroidectomy (MIP) has begun to replace traditional bilateral neck exploration (BNE). OBJECTIVE: The aim of this study is to compare the results of parathyroidectomies performed in our hospital over the past decade that were guided by intra-operative parathyroid hormone (IOPTH) sampling or frozen section (FS) analysis. MATERIAL AND METHODS: Data on 697 patients who underwent parathyroidectomies in the Department of Endocrine Surgery, Dokuz Eylul University between January 2005 and 2018 were included in this study. Patients with malignancies other than thyroid papillary microcarcinoma and parathyroid cancer were excluded from the study. RESULTS: The concomitant use of neck ultrasound (US) and technetium 99m Sestamibi (99mTc MIBI) scintigraphy successfully localized the hyperfunctioning parathyroid glands in nearly 96% of cases. As compared with the IOPTH group, the operation time was longer in the FS group (p < 0.001), and the need for postoperative calcium (Ca) supplementation was higher (p < 0.001). The duration of hospitalization (days) was significantly higher in the FS group (4.2 ± 3.4 vs. 2.6 ± 1.9) as compared with that in the IOPTH group (p < 0.001). In addition, the recurrence rate in the FS group was significantly higher than that in the IPOTH group (p = 0.002). CONCLUSION: IOPTH sampling is a safe and effective method when performed by experienced surgeons and with appropriate preoperative screening. This study emphasizes that IOPTH sampling. We believe that the success in parathyroid surgery is due to three factors: correct indication, accurate localization and experienced surgeon.


Asunto(s)
Secciones por Congelación , Hiperparatiroidismo/diagnóstico , Hiperparatiroidismo/cirugía , Procedimientos Quirúrgicos Mínimamente Invasivos/métodos , Monitoreo Intraoperatorio/métodos , Hormona Paratiroidea/análisis , Paratiroidectomía/métodos , Adulto , Anciano , Femenino , Humanos , Hiperparatiroidismo/metabolismo , Hiperparatiroidismo/patología , Tiempo de Internación/estadística & datos numéricos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/epidemiología , Tempo Operativo , Cintigrafía , Cirugía Asistida por Computador/métodos , Resultado del Tratamiento , Ultrasonografía
10.
Ann Endocrinol (Paris) ; 82(3-4): 149-150, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32192791

RESUMEN

Cardiovascular diseases remain the leading cause of non-communicable chronic diseases, are related to high morbidity and mortality and are associated to a huge impact on healthcare budgets. Biomarkers play an important role for the diagnosis and prognosis of cardiovascular diseases and are recognized tools for value-based care. Parathyroid hormone (PTH) is a major systemic calcium-regulating hormone and an important regulator of bone and mineral homeostasis. PTH 1-84, the biologically active hormone produced by the parathyroid glands and secreted into the systemic circulation, exerts its biological effects through the interaction of its first 34 amino acids with PTH receptors. PTH levels are raised in several cardio-renal disorders and hyperparathyroidism have detrimental effects on the heart and cardiac cells such as cardiac hypertrophy, remodeling and arrhythmias. High circulating PTH levels, through an increase in intracellular calcium, contribute also to the impairment of mitochondrial function and ATP production and to oxidative stress as well as inflammation states and, at the end, to cardiomyocytes necrosis. The interplay between PTH, fibroblast growth factor 23 and aldosterone is also detrimental for cardiovascular system and participate to endothelial dysfunction. Measurement of PTH levels could be therefore relevant in high risk individuals and could provide added value to established cardiac biomarkers for the sub-phenotyping of patients and treatment selection.


Asunto(s)
Factores de Riesgo de Enfermedad Cardiaca , Hormona Paratiroidea/fisiología , Calcio/metabolismo , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/metabolismo , Cardiopatías/etiología , Cardiopatías/metabolismo , Humanos , Hiperparatiroidismo/complicaciones , Hiperparatiroidismo/metabolismo , Factores de Riesgo
11.
Endocrinology ; 161(10)2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32877917

RESUMEN

The protein product of the cyclin D1 oncogene functions by activating partner cyclin-dependent kinases (cdk)4 or cdk6 to phosphorylate, thereby inactivating, the retinoblastoma protein pRB. Nonclassical, cdk-independent, functions of cyclin D1 have been described but their role in cyclin D1-driven neoplasia, with attendant implications for recently approved cdk4/6 chemotherapeutic inhibitors, requires further examination. We investigated whether cyclin D1's role in parathyroid tumorigenesis in vivo is effected primarily through kinase-dependent or kinase-independent mechanisms. Using a mouse model of cyclin D1-driven parathyroid tumorigenesis (PTH-D1), we generated new transgenic lines harboring a mutant cyclin D1 (KE) that is unable to activate its partner kinases. While this kinase-dead KE mutant effectively drove mammary tumorigenesis in an analogous model, parathyroid-overexpressed cyclin D1 KE mice did not develop the characteristic biochemical hyperparathyroidism or parathyroid hypercellularity of PTH-D1 mice. These results strongly suggest that in parathyroid cells, cyclin D1 drives tumorigenesis predominantly through cdk-dependent mechanisms, in marked contrast with the cdk-independence of cyclin D1-driven mouse mammary cancer. These findings highlight crucial tissue-specific mechanistic differences in cyclin D1-driven tumorigenesis, suggest that parathyroid/endocrine cells may be more tumorigenically vulnerable to acquired genetic perturbations in cdk-mediated proliferative control than other tissues, and carry important considerations for therapeutic intervention.


Asunto(s)
Ciclina D1/genética , Quinasa 4 Dependiente de la Ciclina/fisiología , Quinasa 6 Dependiente de la Ciclina/fisiología , Neoplasias de las Paratiroides/genética , Neoplasias de las Paratiroides/metabolismo , Adenoma/genética , Adenoma/metabolismo , Adenoma/patología , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Ciclina D1/metabolismo , Hiperparatiroidismo/genética , Hiperparatiroidismo/metabolismo , Hiperparatiroidismo/patología , Ratones , Ratones Transgénicos , Mutación , Neoplasias de las Paratiroides/patología , Fosforilación/genética , Transducción de Señal/genética
12.
Int J Mol Sci ; 21(15)2020 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-32751307

RESUMEN

Exercise perturbs homeostasis, alters the levels of circulating mediators and hormones, and increases the demand by skeletal muscles and other vital organs for energy substrates. Exercise also affects bone and mineral metabolism, particularly calcium and phosphate, both of which are essential for muscle contraction, neuromuscular signaling, biosynthesis of adenosine triphosphate (ATP), and other energy substrates. Parathyroid hormone (PTH) is involved in the regulation of calcium and phosphate homeostasis. Understanding the effects of exercise on PTH secretion is fundamental for appreciating how the body adapts to exercise. Altered PTH metabolism underlies hyperparathyroidism and hypoparathyroidism, the complications of which affect the organs involved in calcium and phosphorous metabolism (bone and kidney) and other body systems as well. Exercise affects PTH expression and secretion by altering the circulating levels of calcium and phosphate. In turn, PTH responds directly to exercise and exercise-induced myokines. Here, we review the main concepts of the regulation of PTH expression and secretion under physiological conditions, in acute and chronic exercise, and in relation to PTH-related disorders.


Asunto(s)
Calcio/metabolismo , Ejercicio Físico , Hiperparatiroidismo/metabolismo , Hipoparatiroidismo/metabolismo , Hormona Paratiroidea/genética , Fósforo/metabolismo , Huesos/citología , Huesos/metabolismo , Fibronectinas/genética , Fibronectinas/metabolismo , Regulación de la Expresión Génica , Homeostasis/genética , Humanos , Hiperparatiroidismo/genética , Hiperparatiroidismo/patología , Hipoparatiroidismo/genética , Hipoparatiroidismo/patología , Interleucinas/genética , Interleucinas/metabolismo , Riñón/citología , Riñón/metabolismo , Redes y Vías Metabólicas/genética , Contracción Muscular/genética , Unión Neuromuscular/genética , Unión Neuromuscular/metabolismo , Unión Neuromuscular/patología , Hormona Paratiroidea/metabolismo , Transducción de Señal , Vitamina D/metabolismo
13.
J Physiol Sci ; 70(1): 33, 2020 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-32646367

RESUMEN

Maternal-fetal calcium (Ca2+) transport in the placenta plays a critical role in maintaining fetal bone mineralization. Mutations in the gene encoding the transient receptor potential cation channel, subfamily V, member 6 (TRPV6) have been identified as causative mutations of transient neonatal hyperparathyroidism due to insufficient maternal-fetal Ca2+ transport in the placenta. In this study, we found two novel mutations in subjects that have transient neonatal hyperparathyroidism. TRPV6 carrying the mutation p.Arg390His that localizes to the outer edge of the first transmembrane domain (S1) showed impaired trafficking to the plasma membrane, whereas TRPV6 having the mutation p.Gly291Ser in the sixth ankyrin repeat (AR) domain had channel properties that were comparable those of WT channels, although the increases in steady-state intracellular Ca2+ concentration could have led to Ca2+ overload and subsequent death of cells expressing this mutant channel. These results indicate that the AR6 domain contributes to TRPV6-mediated maintenance of intracellular Ca2+ concentrations, and that this region could play a novel role in regulating the activity of TRPV6 Ca2+-selective channels.


Asunto(s)
Canales de Calcio/genética , Hiperparatiroidismo/diagnóstico , Mutación , Diagnóstico Prenatal/métodos , Canales Catiónicos TRPV/genética , Adulto , Calcio/metabolismo , Canales de Calcio/metabolismo , Femenino , Feto/diagnóstico por imagen , Humanos , Hiperparatiroidismo/genética , Hiperparatiroidismo/metabolismo , Recién Nacido , Masculino , Embarazo , Canales Catiónicos TRPV/metabolismo
14.
Endocr J ; 66(4): 319-327, 2019 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-30799315

RESUMEN

Hyperparathyrodism-jaw tumor (HPT-JT) syndrome is an autosomal dominant disorder. Loss of function of the cell division cycle protein 73 homolog (CDC73) gene is responsible for the syndrome. This gene encodes an ubiquitously expressed 531 amino acid protein, parafibromin, that acts as a tumor suppressor. Loss of heterozygosity (LOH) of the CDC73 locus in many HPT-JT associated parathyroid tumors from patients with germline mutation is in accordance with Knudson's "two-hit" model for hereditary cancer. A 41-year-old man with mandible ossifying fibroma suffered from severe hypercalcemia due to parathyroid carcinoma (PC). Genetic analysis was performed to evaluate germinal and somatic CDC73 gene mutation as well as real-time qRT-PCR to quantify CDC73 mRNA, miR-155 and miR-664 expression levels. Immunohistochemistry and Western blotting (WB) assay were carried out to evaluate parafibromin protein expression. A novel heterozygous nonsense mutation, c.191-192 delT, was identified in the CDC73 gene. No CDC73 LOH was found in PC tissue, nor any differences in expression levels for CDC73 gene, miR-155 and miR-664 between PC and parathyroid adenoma control tissues. On the contrary, both immunohistochemistry and WB assay showed an approximate 90% reduction of parafibromin protein expression in PC. In conclusion, this study describes a novel germinal mutation, c.191-192 delT, in the CDC73 gene. Despite normal CDC73 gene expression, we found a significant decrease in parafibromin. We hypothesize that a gene silencing mechanism, possibly induced by microRNA, could play a role in determining somatic post-transcriptional inactivation of the wild type CDC73 allele.


Asunto(s)
Adenoma/genética , Carcinoma/genética , Fibroma/genética , Mutación de Línea Germinal , Hiperparatiroidismo/genética , Neoplasias Maxilomandibulares/genética , Neoplasias de las Paratiroides/genética , Proteínas Supresoras de Tumor/genética , Adenoma/metabolismo , Adenoma/patología , Adulto , Alelos , Carcinoma/metabolismo , Carcinoma/patología , Fibroma/metabolismo , Fibroma/patología , Humanos , Hiperparatiroidismo/metabolismo , Hiperparatiroidismo/patología , Inmunohistoquímica , Neoplasias Maxilomandibulares/metabolismo , Neoplasias Maxilomandibulares/patología , Pérdida de Heterocigocidad , Masculino , Neoplasias de las Paratiroides/metabolismo , Neoplasias de las Paratiroides/patología , Proteínas Supresoras de Tumor/metabolismo
15.
J Bone Miner Res ; 34(5): 955-963, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30690792

RESUMEN

The relationship between impaired calcium sensing, dysregulated parathyroid hormone (PTH) secretion, and parathyroid cell proliferation in parathyroid neoplasia is not understood. We previously reported that a GTPase activating protein, regulator of G-protein signaling 5 (RGS5) is overexpressed in a subset of parathyroid tumors associated with primary hyperparathyroidism (PHPT) and that RGS5 can inhibit signaling from the calcium-sensing receptor (CASR). In vivo, we found that RGS5-null mice have abnormally low PTH levels. To gain a better understanding of the potential role of RGS5 overexpression in parathyroid neoplasia and PHPT and to investigate whether inhibition of CASR signaling can lead to parathyroid neoplasia, we created and characterized a transgenic mouse strain overexpressing RGS5 specifically in the parathyroid gland. These mice develop hyperparathyroidism, bone changes reflective of elevated PTH, and parathyroid neoplasia. Further, expression of exogenous RGS5 in normal human parathyroid cells results in impaired signaling from CASR and negative feedback on PTH secretion. These results provide evidence that RGS5 can modulate signaling from CASR and support a role for RGS5 in the pathogenesis of PHPT through inhibition of CASR signaling. © 2019 American Society for Bone and Mineral Research.


Asunto(s)
Regulación de la Expresión Génica , Hiperparatiroidismo/metabolismo , Proteínas RGS/biosíntesis , Receptores Sensibles al Calcio/metabolismo , Transducción de Señal , Animales , Hiperparatiroidismo/genética , Hiperparatiroidismo/patología , Ratones , Ratones Transgénicos , Proteínas RGS/genética , Receptores Sensibles al Calcio/genética
16.
J Bone Miner Res ; 34(1): 22-37, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30536424

RESUMEN

We review advancing and overlapping stages for our understanding of the expressions of six hyperparathyroid (HPT) syndromes: multiple endocrine neoplasia type 1 (MEN1) or type 4, multiple endocrine neoplasia type 2A (MEN2A), hyperparathyroidism-jaw tumor syndrome, familial hypocalciuric hypercalcemia, neonatal severe primary hyperparathyroidism, and familial isolated hyperparathyroidism. During stage 1 (1903 to 1967), the introduction of robust measurement of serum calcium was a milestone that uncovered hypercalcemia as the first sign of dysfunction in many HPT subjects, and inheritability was reported in each syndrome. The earliest reports of HPT syndromes were biased toward severe or striking manifestations. During stage 2 (1959 to 1985), the early formulations of a syndrome were improved. Radioimmunoassays (parathyroid hormone [PTH], gastrin, insulin, prolactin, calcitonin) were breakthroughs. They could identify a syndrome carrier, indicate an emerging tumor, characterize a tumor, or monitor a tumor. During stage 3 (1981 to 2006), the assembly of many cases enabled recognition of further details. For example, hormone non-secreting skin lesions were discovered in MEN1 and MEN2A. During stage 4 (1985 to the present), new genomic tools were a revolution for gene identification. Four principal genes ("principal" implies mutated or deleted in 50% or more probands for its syndrome) (MEN1, RET, CASR, CDC73) were identified for five syndromes. During stage 5 (1993 to the present), seven syndromal genes other than a principal gene were identified (CDKN1B, CDKN2B, CDKN2C, CDKN1A, GNA11, AP2S1, GCM2). Identification of AP2S1 and GCM2 became possible because of whole-exome sequencing. During stages 4 and 5, the newly identified genes enabled many studies, including robust assignment of the carriers and non-carriers of a mutation. Furthermore, molecular pathways of RET and the calcium-sensing receptor were elaborated, thereby facilitating developments in pharmacotherapy. Current findings hold the promise that more genes for HPT syndromes will be identified and studied in the near future. © 2018 American Society for Bone and Mineral Research.


Asunto(s)
Hiperparatiroidismo , Neoplasia Endocrina Múltiple Tipo 1 , Neoplasia Endocrina Múltiple Tipo 2a , Proteínas de Neoplasias , Neoplasias de las Paratiroides , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Hiperparatiroidismo/clasificación , Hiperparatiroidismo/genética , Hiperparatiroidismo/historia , Hiperparatiroidismo/metabolismo , Neoplasia Endocrina Múltiple Tipo 1/clasificación , Neoplasia Endocrina Múltiple Tipo 1/genética , Neoplasia Endocrina Múltiple Tipo 1/historia , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Neoplasia Endocrina Múltiple Tipo 2a/clasificación , Neoplasia Endocrina Múltiple Tipo 2a/genética , Neoplasia Endocrina Múltiple Tipo 2a/historia , Neoplasia Endocrina Múltiple Tipo 2a/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias de las Paratiroides/clasificación , Neoplasias de las Paratiroides/genética , Neoplasias de las Paratiroides/historia , Neoplasias de las Paratiroides/metabolismo , Síndrome
17.
Cells Tissues Organs ; 206(1-2): 54-61, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30466097

RESUMEN

We developed a novel model for studying hyperparathyroidism by growing ex vivo 3-dimensional human parathyroids as part of a microphysiological system (MPS) that mimics human physiology. The purpose of this study was to validate the parathyroid portion of the MPS. We prospectively collected parathyroid tissue from 46 patients with hyperparathyroidism for growth into pseudoglands. We evaluated pseudogland architecture and calcium responsiveness. Following 2 weeks in culture, dispersed cells successfully coalesced into pseudoglands ∼500-700 µm in diameter that mimicked the appearance of normal parathyroid glands. Functionally, they also appeared similar to intact parathyroids in terms of organization and calcium-sensing receptor expression. Immunohistochemical staining for calcium-sensing receptor revealed 240-450/cell units of mean fluorescence intensity within the pseudoglands. Finally, the pseudoglands showed varying levels of calcium responsiveness, indicated by changes in parathyroid hormone (PTH) levels. In summary, we successfully piloted the development of a novel MPS for studying the effects of hyperparathyroidism on human organ systems. We are currently evaluating the effect of PTH on adverse remodeling of tissue engineered cardiac, skeletal, and bone tissue within the MPS.


Asunto(s)
Hiperparatiroidismo/metabolismo , Técnicas de Cultivo de Órganos/métodos , Organoides/fisiología , Glándulas Paratiroides/fisiología , Calcio/metabolismo , Humanos , Hiperparatiroidismo/patología , Organoides/patología , Organoides/ultraestructura , Glándulas Paratiroides/patología , Glándulas Paratiroides/ultraestructura , Hormona Paratiroidea/metabolismo
18.
Sci Rep ; 8(1): 5310, 2018 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-29593281

RESUMEN

Hypercalcemia and hyperparathyroidism in patients receiving maintenance hemodialysis (MHD) can cause the progression of cardiovascular diseases (CVD) and mineral bone disorders (MBD). The KDIGO recommends the dialysates with a calcium (Ca) concentration of 1.25-1.5 mmol/L for MHD treatments, but the optimal concentration remains controversial. Here, we conducted a systematic review and a meta-analysis of seven randomized controlled trials examining a total of 622 patients to investigate the optimal concentration for MHD for 6 months or longer. The dialysates with a low Ca concentration (1.125 or 1.25 mmol/L) significantly lowered the serum Ca and raised the intact parathyroid hormone levels by 0.52 mg/dL (95% confidence interval, 0.20-0.85) and 39.59 pg/mL (14.80-64.38), respectively, compared with a high Ca concentration (1.50 or 1.75 mmol/L). Three studies showed that a low concentration was preferred for lowering arterial calcifications or atherosclerosis in different arteries, but one study showed that coronary arterial calcifications increased with a low concentration. Two studies showed contradictory outcomes in terms of MBD. Our meta-analysis showed that a dialysate with a low Ca concentration lowered the serum Ca levels in patients receiving long-term MHD, but further studies are needed to determine the optimal Ca concentration in terms of CVD and MBD.


Asunto(s)
Calcio/sangre , Soluciones para Diálisis/farmacología , Soluciones para Hemodiálisis/farmacología , Diálisis Renal/métodos , Enfermedades Óseas/sangre , Enfermedades Óseas/etiología , Enfermedades Cardiovasculares/sangre , Enfermedades Cardiovasculares/etiología , Humanos , Hipercalcemia/sangre , Hipercalcemia/prevención & control , Hiperparatiroidismo/sangre , Hiperparatiroidismo/metabolismo , Hiperparatiroidismo/prevención & control , Hormona Paratiroidea/sangre , Fosfatos/sangre , Ensayos Clínicos Controlados Aleatorios como Asunto , Diálisis Renal/efectos adversos
19.
Sci Rep ; 7(1): 15300, 2017 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-29127344

RESUMEN

The bone catabolic actions of parathyroid hormone (PTH) are seen in patients with hyperparathyroidism, or with infusion of PTH in rodents. We have previously shown that the chemokine, monocyte chemoattractant protein-1 (MCP-1), is a mediator of PTH's anabolic effects on bone. To determine its role in PTH's catabolic effects, we continuously infused female wild-type (WT) and MCP-1-/- mice with hPTH or vehicle. Microcomputed tomography (µCT) analysis of cortical bone showed that hPTH-infusion induced significant bone loss in WT mice. Further, µCT analysis of trabecular bone revealed that, compared with the vehicle-treated group, the PTH-treated WT mice had reduced trabecular thickness and trabecular number. Notably, MCP-1-/- mice were protected against PTH-induced cortical and trabecular bone loss as well as from increases in serum CTX (C-terminal crosslinking telopeptide of type I collagen) and TRACP-5b (tartrate-resistant acid phosphatase 5b). In vitro, bone marrow macrophages (BMMs) from MCP-1-/- and WT mice were cultured with M-CSF, RANKL and/or MCP-1. BMMs from MCP-1-/- mice showed decreased multinucleated osteoclast formation compared with WT mice. Taken together, our work demonstrates that MCP-1 has a role in PTH's catabolic effects on bone including monocyte and macrophage recruitment, osteoclast formation, bone resorption, and cortical and trabecular bone loss.


Asunto(s)
Resorción Ósea/metabolismo , Quimiocina CCL2/metabolismo , Hiperparatiroidismo , Osteoclastos/metabolismo , Hormona Paratiroidea/efectos adversos , Animales , Resorción Ósea/inducido químicamente , Resorción Ósea/genética , Resorción Ósea/patología , Hueso Esponjoso/metabolismo , Hueso Esponjoso/patología , Quimiocina CCL2/genética , Hueso Cortical/metabolismo , Hueso Cortical/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Hiperparatiroidismo/inducido químicamente , Hiperparatiroidismo/genética , Hiperparatiroidismo/metabolismo , Hiperparatiroidismo/patología , Ratones , Ratones Noqueados , Osteoclastos/patología , Hormona Paratiroidea/farmacología , Microtomografía por Rayos X
20.
J Clin Endocrinol Metab ; 102(12): 4417-4420, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29053801

RESUMEN

Context: Parathyroid hormone-related protein (PTH-rP)-induced hypercalcemia or pseudophyperparathyroidism during pregnancy is a condition that can result in serious fetal and maternal complications. Among others, breast tissue might be the cause of this PTH-rP production, in which case medical treatment is possible, as we describe in this case. Setting: A 32-year-old woman presented in the 15th week of pregnancy with massive enlargement of breasts and abdominal pain due to severe hypercalcemia, hypercalciuria, and suppressed PTH. Hematological and solid malignancy were excluded. PTH-rP was found to be fourfold to eightfold increased, which is pathological even for pregnancy term. PTH-rP is produced in mammarian tissue as well as in placental tissue, in reaction to prolactin receptor activation. Prolactin hypersensitivity of breast tissue can cause excessive PTH-rP production during pregnancy. Intervention: Dopamine agonists were applied to decrease prolactin. Results: Calcium levels normalized, and PTH-rP levels became undetectable with bromocriptin treatment. A full-term healthy baby was born without disorders of calcium homeostasis, neither directly after birth nor 2 years after follow-up. After delivery, dopamine agonists could be tapered without recurrence of hypercalcemia. Conclusion: Pseudohyperparathyroidism of pregnancy was caused by increased sensitivity of mammarian tissue for prolactin, which could be treated medically, preventing emergency mastectomy.


Asunto(s)
Agonistas de Dopamina/uso terapéutico , Hipercalcemia/tratamiento farmacológico , Hipercalcemia/genética , Hiperparatiroidismo/genética , Proteína Relacionada con la Hormona Paratiroidea/genética , Complicaciones del Embarazo/tratamiento farmacológico , Complicaciones del Embarazo/genética , Adulto , Bromocriptina/uso terapéutico , Calcio/sangre , Femenino , Humanos , Hiperparatiroidismo/metabolismo , Recién Nacido , Hormona Paratiroidea/sangre , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Embarazo , Resultado del Embarazo , Prolactina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...