Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
World J Clin Cases ; 11(10): 2290-2300, 2023 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-37122511

RESUMEN

BACKGROUND: Hypoparathyroidism, which can be sporadic or a component of an inherited syndrome, is the most common cause of hypocalcemia. If hypocalcemia is accompanied by other electrolyte disturbances, such as hypokalemia and hypomagnesemia, then the cause, such as renal tubular disease, should be carefully identified. CASE SUMMARY: An 18-year-old female visited our clinic because of short stature and facial deformities, including typical phenotypes, such as low ear position, depression of the nasal bridge, small hands and feet, and loss of dentition. The lab results suggested normal parathyroid hormone but hypocalcemia. In addition, multiple electrolyte disturbances were found, including hypokalemia, hypocalcemia and hypomagnesemia. The physical signs showed a short fourth metatarsal bone of both feet. The X-ray images showed cortical thickening of long bones and narrowing of the medulla of the lumen. Cranial computed tomography indicated calcification in the bilateral basal ganglia. Finally, the genetic investigation showed a de novo heterogenous mutation of "FAM111A" (c. G1706A:p.R569H). Through a review of previously reported cases, the mutation was found to be the most common mutation site in Kenny-Caffey syndrome type 2 (KCS2) cases reported thus far (16/23, 69.6%). The mutation was slightly more prevalent in females than in males (11/16, 68.8%). Except for hypocalcemia, other clinical manifestations are heterogeneous. CONCLUSION: As a rare autosomal dominant genetic disease of hypoparathyroidism, the clinical manifestations of KCS2 are atypical and diverse. This girl presented with short stature, facial deformities and skeletal deformities. The laboratory results revealed hypocalcemia as the main electrolyte disturbance. Even though her family members showed normal phenotypes, gene detection was performed to find the mutation of the FAM111A gene and confirmed the diagnosis of KCS2.

2.
Ann Anat ; 248: 152070, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36801365

RESUMEN

BACKGROUND: As a systemic skeletal disorder, osteoporosis can increase fracture risk. This study wants to discuss the mechanism of osteoporosis and find possible molecular therapy. Bone morphogenetic protein 2 (BMP2) was utilized to stimulate MC3T3-E1 to establish a cellular osteoporosis model in vitro. METHODS: Initially, the viability of BMP2-induced MC3T3-E1 was assessed with a Cell counting kit-8 (CCK-8) assay. By real-time quantitative PCR (RT-qPCR) and western blot, Robo2 expression after roundabout (Robo) silencing or overexpression was estimated. Besides, alkaline phosphatase (ALP) expression, mineralization level and LC3II green fluorescent protein (GFP) expression were evaluated using ALP assay, Alizarin red staining and immunofluorescence staining, separately. Additionally, the expression of proteins related to osteoblast differentiation and autophagy was analyzed by RT-qPCR and western blot. Then, following autophagy inhibitor 3-methyladenine (3-MA) treatment, osteoblast differentiation and mineralization were measured again. RESULTS: MC3T3-E1 cells were differentiated into osteoblasts under BMP2 induction and Robo2 expression was greatly ascended. After Robo2 silencing, Robo2 expression was markedly diminished. ALP activity and mineralization level in BMP2-induced MC3T3-E1 cells were declined after depleting Robo2. Robo2 expression was conspicuously enhanced after overexpressing Robo2. Robo2 overexpression promoted the differentiation and mineralization of BMP2-induced MC3T3-E1 cells. Rescue experiments revealed that Robo2 silence and its overexpression could regulate the autophagy of BMP2-stimulated MC3T3-E1 cells. After 3-MA treatment, the increased ALP activity and mineralization level of BMP2-induced MC3T3-E1 cells with Robo2 upregulation were reduced. Furthermore, parathyroid hormone 1-34 (PTH1-34) treatment enhanced the expression of ALP, Robo2, LC3II and Beclin-1 and reduced the levels of LC3I and p62 of MC3T3-E1 cells concentration-dependently. CONCLUSION: Collectively, Robo2, which was activated by PTH1-34, promoted osteoblast differentiation and mineralization through autophagy.


Asunto(s)
Autofagia , Osteogénesis , Receptores Inmunológicos , Diferenciación Celular/fisiología , Línea Celular , Osteoblastos , Hormona Paratiroidea , Animales , Ratones
3.
Mol Neurobiol ; 58(7): 3158-3174, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33634377

RESUMEN

Histone deacetylase (HDAC) inhibitors can protect the brain from ischemic injury. This study aimed to identify the regulation of HDAC3 in cerebral ischemic injury. Middle cerebral artery occlusion (MCAO) was performed to establish a mouse model with cerebral ischemic injury, in which expression of HDAC3 and miR-19a was evaluated using RT-qPCR. In MCAO mice with silencing of HDAC3, infarct volume was determined using 2,3,5-triphenyl tetrazolium chloride (TTC) staining, and serum levels of TNF-α, IL-6, and IL-8 were measured using ELISA. An in vitro model was constructed in human umbilical vein endothelial cells (HUVECs) with oxygen-glucose deprivation/reoxygenation (OGD/R), followed by gain- and loss-of-function experiments. Relationships among miR-19a, HDAC3, and syndecan-1 (SDC1) were explored using RIP, ChIP, and dual-luciferase reporter assays. The expression of HDAC3, SDC1, JAK1, and STAT3 along with the extent of JAK1 and STAT3 phosphorylation was measured by Western blot analysis. HUVEC viability, apoptosis, and angiogenesis were assessed by CCK-8, flow cytometry, and angiogenesis assays in vitro separately. We found elevated HDAC3 and downregulated miR-19a expression in the MCAO mice. Decreased TNF-α, IL-6, and IL-8 serum levels were observed in response to silencing of HDAC3. HDAC3 inhibited the expression of miR-19a, which in turn targeted SDC1, leading to JAK1/STAT3 signaling pathway activation. HDAC3 overexpression or miR-19a inhibition repressed HUVEC viability and angiogenesis but enhanced HUVEC apoptosis. Our data unraveled the mechanism whereby HDAC3 inhibition ameliorated cerebral ischemic injury by activating the JAK1/STAT3 signaling pathway through miR-19a-mediated SDC1 inhibition.


Asunto(s)
Isquemia Encefálica/metabolismo , Histona Desacetilasas/biosíntesis , Janus Quinasa 1/metabolismo , MicroARNs/biosíntesis , Factor de Transcripción STAT3/metabolismo , Sindecano-1/metabolismo , Animales , Isquemia Encefálica/patología , Isquemia Encefálica/prevención & control , Regulación hacia Abajo/fisiología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Janus Quinasa 1/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/antagonistas & inhibidores , Factor de Transcripción STAT3/antagonistas & inhibidores , Regulación hacia Arriba/fisiología
4.
Folia Histochem Cytobiol ; 59(2): 114-123, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35411930

RESUMEN

INTRODUCTION: Isoflurane anesthesia at the period of brain development can lead to neurotoxicity and long-term cognitive impairment. This study aimed to investigate the role of miR-497 on isoflurane-induced neurotoxicity. MATERIAL AND METHODS: Neonatal rats (P7) were subject to isoflurane for 2 h at P7, P9, and P11. MiR-497 and neuron apoptosis were evaluated in hippocampal tissue by qRT-PCR and western blot. Fear conditioning test and Morris water maze were performed to determine cognitive function. The cell viability of isolated hippocampal neuronal cells exposed to isoflurane was measured using MTT test. The regulation of phospholipase D1 (PLD1) by miR-497 in isolated hippocampal neuronal cells was evaluated by luciferase reporter assays and western blot. Immunohistochemistry and TUNEL staining were employed to examine the PLD1 expression and neuronal cell apoptosis in hippocampus of neonatal rats, respectively. RESULTS: Repeated isoflurane anesthesia led to neurons' apoptosis and long-term cognitive impairment. Isoflurane exposure led to apoptosis and viability reduction in hippocampal neuronal cells. MiR-497 was observed to be upregulated after isoflurane exposure both in vivo and in vitro. Knockdown of miR-497 attenuated isoflurane-induced neuronal cells apoptosis and viability reduction. Furthermore, PLD1 was predicted and then validated as a novel target of miR-497. miR-497 could negatively regulate PLD1 by binding to its 3'-untranslated region. Downregulation of PLD1 was also observed after isoflurane exposure in neonatal rat hippocampus and hippocampal primary neuronal cell cultures. CONCLUSIONS: Induction of miR-497 was involved in isoflurane anesthesia-induced cognitive impairment and neuronal cell apoptosis by targeting PLD1. miR-497 may be a novel potential mechanism in isoflurane-induced neurotoxicity so that our findings provide new insight into a better and understanding of the clinical application of isoflurane.


Asunto(s)
Disfunción Cognitiva , Isoflurano , MicroARNs , Animales , Animales Recién Nacidos , Apoptosis , Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/genética , Disfunción Cognitiva/metabolismo , Hipocampo , Isoflurano/metabolismo , Isoflurano/toxicidad , MicroARNs/genética , MicroARNs/metabolismo , Ratas
5.
Brain Res Bull ; 152: 1-10, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31185248

RESUMEN

Hypoxic-ischemic brain damage (HIBD) is a common neurological disorder. Emerging reports reveal that long non-coding RNAs and microRNAs (miRs) are implicated in the progress of HIBD. In this study we tried to ascertain whether lncRNA MALAT1, with the involvement of miR-429 and WNT1, affects HIBD. Initially, a HIBD mouse model was established. Then, we treated HIBD mice with dexmedetomidine (DEX) and then up- or down-regulated the expression of MALAT1, miR-429 and WNT1 in HIBD mice and neurons. Meanwhile, brain injury and hippocampal neuronal apoptosis were evaluated. Moreover, the interaction among MALAT1, miR-429 and WNT1 in HIBD was investigated. MALAT1 and WNT1 were high-expressed in brain tissues of HIBD mice while miR-429 was low-expressed in brain tissues from HIBD mice. Interestingly, MALAT1 silencing was observed to enhance the cerebral protection of DEX against HIBD. In addition, it was confirmed that MALAT1 sponged miR-429 downregulating expression of miR-429, thereby promoting apoptosis of hippocampal neurons. This effect was achieved through up-regulating the level of WNT1. Taken together, this study demonstrates that silencing of MALAT1 enhances the cerebral protection of DEX against HIBD by suppressing WNT1 expression through miR-429.


Asunto(s)
Hipocampo/irrigación sanguínea , Hipocampo/patología , Hipoxia-Isquemia Encefálica/metabolismo , MicroARNs/metabolismo , Neuronas/patología , ARN Largo no Codificante/metabolismo , Proteína Wnt1/metabolismo , Animales , Apoptosis/fisiología , Dexmedetomidina/farmacología , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Hipoxia-Isquemia Encefálica/genética , Hipoxia-Isquemia Encefálica/patología , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Neuronas/metabolismo , ARN Largo no Codificante/genética , Proteína Wnt1/genética
6.
Artículo en Inglés | MEDLINE | ID: mdl-28511119

RESUMEN

Herba Erigerontis has long been used to cure apoplexy hemiplegia and precordial pain in China. In addition, the bioactivities of its total flavonoids-breviscapine included inhibiting amyloid beta (Aß) fibril formation, antioxidation and metal chelating, which are beneficial to treat Alzheimer's disease (AD). Hence, A HPLC-QTOF-MS based plasma metabonomics approach was applied to investigate the neuroprotective effects of breviscapine on intracerebroventricular injection of aggregated Aß 1-42 induced AD mice for the first time in the study. Ten potential biomarkers were screened out by multivariate statistical analysis, eight of which were further identified as indoleacrylic acid, C16 sphinganine, LPE (22:6), sulfolithocholic acid, LPC (16:0), PA (22:1/0:0), taurodeoxycholic acid, and PC (0:0/18:0). According to their metabolic pathways, it was supposed that breviscapine ameliorated the learning and memory deficits of AD mice predominantly by regulating phospholipids metabolism, elevating serotonin level and lowering cholesterols content in vivo.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Colesterol/metabolismo , Flavonoides/farmacología , Fármacos Neuroprotectores/farmacología , Fosfolípidos/metabolismo , Serotonina/sangre , Enfermedad de Alzheimer/sangre , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/psicología , Péptidos beta-Amiloides , Animales , Conducta Animal/efectos de los fármacos , Biomarcadores/sangre , Cromatografía Líquida de Alta Presión/métodos , Modelos Animales de Enfermedad , Flavonoides/administración & dosificación , Flavonoides/uso terapéutico , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Redes y Vías Metabólicas/efectos de los fármacos , Metabolómica , Ratones , Ratones Endogámicos ICR , Ratones Transgénicos , Fármacos Neuroprotectores/administración & dosificación , Fragmentos de Péptidos , Espectrometría de Masas en Tándem/métodos , Triptófano/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...