Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Cureus ; 16(3): e56406, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38638771

RESUMEN

In the realm of obstetric care, discerning the subtle signs of primary hyperparathyroidism (PHPT) amidst common pregnancy symptoms remains a formidable challenge. Our exploration into a case of gestational hypercalcemia peels back the layers of this complexity, revealing the clinical conundrum posed by overlapping gastrointestinal manifestations. The journey from diagnosis through surgical intervention to the resolution of symptoms underscores the importance of vigilance for PHPT in pregnant patients. This case further prompts consideration of gamma-aminobutyric acid (GABA) as a potential piece in the puzzle of persistent symptoms post-calcium normalization, inviting a broader dialogue on the intricacies of parathyroid pathology in pregnancy.

2.
QJM ; 2024 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-38336882
3.
JCEM Case Rep ; 1(6): luad150, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-38045869

RESUMEN

Postprandial hyperinsulinemic hypoglycemia, although rare, is a well-documented complication that can manifest after upper gastrointestinal surgery. Despite its potential for severe morbidity, the underlying pathogenesis and optimal treatment strategies for this condition remain insufficiently understood. This report presents a compelling case of postprandial hypoglycemia following Billroth-II gastrojejunostomy, characterized by a marked increase in postprandial insulin levels, accompanied by the exaggerated response of incretin hormones. The incretin effect in this patient was found to be exceptionally high, measuring at approximately 90%. While nutritional interventions proved ineffective in alleviating the patient's symptoms, the administration of octreotide significantly attenuated the exaggerated postprandial insulin and incretin response, substantially ameliorating both the symptoms and postprandial hypoglycemia. Monthly subcutaneous injections of long-acting repeatable octreotide were initiated, resulting in the complete resolution of symptomatic postprandial hypoglycemia. Although the patient developed acalculous cholecystitis and gallstone cholangitis 2 years after commencing octreotide therapy, she has remained free from symptomatic postprandial hypoglycemia for more than 4 years. Our case underscores the efficacy of somatostatin analogs in the management of postprandial hyperinsulinemia after gastrointestinal surgery, shedding light on the potential involvement of incretin hormones in the pathophysiology of this condition.

4.
JCEM Case Rep ; 1(2): luad020, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37908468

RESUMEN

Lymphocytic infundibulo-neurohypophysitis (LINH) is a rare autoimmune inflammatory process that selectively affects the neurohypophysis and the pituitary stalk, typically presenting with central diabetes insipidus (CDI). LINH is considered underdiagnosed because the definitive diagnosis requires invasive pituitary surgery with a high risk of complications. We present a case of CDI resulting from LINH, which was treated with conservative management, eschewing both glucocorticoid treatment and pituitary surgery. At presentation, the hormonal assessment indicated the presence of CDI without anterior pituitary dysfunction. Magnetic resonance imaging revealed stalk thickening without a posterior pituitary bright spot, and anti-rabphilin-3A antibodies were positive in serum. Collectively, we made a diagnosis of LINH. Considering that the patient did not exhibit any symptoms of mass effect, we chose conservative treatment with desmopressin acetate. One year later, the stalk thickening regressed spontaneously without surgical or glucocorticoid treatment, although the posterior pituitary bright spot remained absent, and CDI did not improve. The inflammatory process of LINH is mostly self-limited and recovers spontaneously, whereas life-long desmopressin treatment may be required because of pituitary stalk fibrosis and atrophy. Our case highlights the importance of noninvasive diagnosis and careful follow-up in preventing unnecessary interventions for patients with LINH.

5.
Eur Heart J Case Rep ; 7(11): ytad487, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37954559

RESUMEN

Background: Heart failure concomitant with prolactinoma is extremely rare. Case summary: We present the case of a 29-year-old man who had acute decompensated heart failure concomitant with visual loss in his right eye. Transthoracic echocardiography indicated severely decreased left ventricular (LV) function. A massive tumour on the sella turcica was detected by brain computed tomography. The findings of the laboratory tests showed hyperprolactinaemia with hypopituitarism, and the antigen test for coronavirus disease 2019 was positive as an incidental finding. Medication for heart failure and cabergoline therapy were started immediately. His LV function significantly improved, and he had no symptoms after a year. Discussion: Prolactinoma in men, which can cause visual loss and hypopituitarism, is frequently substantial when diagnosed. The cardiac manifestation of prolactinoma is uncommon. It is believed that a major contributing component to the pathogenesis of peripartum cardiomyopathy is hyperprolactinaemia. Hyperprolactinaemia may cause endothelial damage and cardiomyocyte dysfunction, eventually resulting in LV dysfunction. The success of LV reverse remodelling may be significantly impacted by heart failure and hormone treatments. Heart failure and endocrine therapy should be administered concurrently to patients who have prolactinoma and congestive heart failure.

6.
Elife ; 122023 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-37732504

RESUMEN

Pancreatic α-cells secrete glucagon, an insulin counter-regulatory peptide hormone critical for the maintenance of glucose homeostasis. Investigation of the function of human α-cells remains a challenge due to the lack of cost-effective purification methods to isolate high-quality α-cells from islets. Here, we use the reaction-based probe diacetylated Zinpyr1 (DA-ZP1) to introduce a novel and simple method for enriching live α-cells from dissociated human islet cells with ~95% purity. The α-cells, confirmed by sorting and immunostaining for glucagon, were cultured up to 10 days to form α-pseudoislets. The α-pseudoislets could be maintained in culture without significant loss of viability, and responded to glucose challenge by secreting appropriate levels of glucagon. RNA-sequencing analyses (RNA-seq) revealed that expression levels of key α-cell identity genes were sustained in culture while some of the genes such as DLK1, GSN, SMIM24 were altered in α-pseudoislets in a time-dependent manner. In conclusion, we report a method to sort human primary α-cells with high purity that can be used for downstream analyses such as functional and transcriptional studies.


Asunto(s)
Células Secretoras de Glucagón , Células Secretoras de Insulina , Islotes Pancreáticos , Humanos , Glucagón/metabolismo , Transcriptoma , Islotes Pancreáticos/metabolismo , Insulina/metabolismo , Células Secretoras de Glucagón/metabolismo , Glucosa/metabolismo , Fluoresceínas/metabolismo , Células Secretoras de Insulina/metabolismo
9.
Cureus ; 14(5): e25548, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35783884

RESUMEN

We report the case of a 49-year-old woman presenting with amenorrhea and progressive visual field defect for one month. Endocrinological workup revealed a high concentration of serum prolactin, and magnetic resonance imaging (MRI) showed pituitary macroadenoma with extrasellar extension as well as compression of optic nerves. Treatment with a dopamine agonist, cabergoline, for eight weeks led to the resolution of the visual field defect accompanied by a rapid decrease in the serum prolactin level. Follow-up MRI three months after the initial diagnosis revealed alleviation of visible mechanical compression of the optic chiasm by the tumor. We considered that the absence of retinal nerve damage and prompt initiation of cabergoline contributed to the rapid recovery of the visual acuity.

10.
Diabetes Ther ; 13(4): 733-746, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35285007

RESUMEN

INTRODUCTION: Previous studies suggested that ß-cell function markers such as fasting and postprandial serum C-peptide and C-peptide increment (FCPR, PCPR, and ΔCPR, respectively) may be useful in estimating glycemic response to glucagon-like peptide-1 receptor agonists. However, it remains elusive whether baseline glycemic control confounds these markers. Here we aimed to identify the least confounded ß-cell function markers and investigate whether these markers could predict glycemic response to dulaglutide. METHODS: We evaluated FCPR, PCPR, and ΔCPR levels in patients with type 2 diabetes who initiated dulaglutide treatment after a standardized meal tolerance test (MTT). We first investigated the confounding effects of baseline HbA1c on ß-cell function markers using Pearson's correlation test. Then, we evaluated the association between each ß-cell function marker and glycemic response (HbA1c change 0-6 months) to dulaglutide using generalized linear model and logistic regression analysis with adjustment for baseline HbA1c. RESULTS: In 141 patients, baseline HbA1c was significantly inversely correlated with PCPR and ΔCPR (P < 0.01 for both) but not with FCPR (r = 0.02; P = 0.853), suggesting that FCPR was the marker least confounded by baseline glycemic control. Of all patients, 59 continued dulaglutide for at least 6 months without initiating any additional glucose-lowering medications. Mean ± SE HbA1c change 0-6 months was - 1.16 ± 0.17% (P < 0.001 vs. baseline). The ß-cell function markers were significantly associated with HbA1c change 0-6 months in the generalized linear model. FCPR was also a significant predictor for achieving a reduction in HbA1c of at least 1% (P = 0.044) with an area under the receiver operating characteristic curve of 0.83 (sensitivity = 0.81 and specificity = 0.79). CONCLUSION: Fasting and meal-induced C-peptide levels are associated with glycemic response to dulaglutide, among which FCPR is least confounded by baseline glycemic control, suggesting its utility as a marker for glycemic response to dulaglutide.

11.
Cell Death Differ ; 28(12): 3199-3213, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34663907

RESUMEN

SARS-CoV-2 vaccinations have greatly reduced COVID-19 cases, but we must continue to develop our understanding of the nature of the disease and its effects on human immunity. Previously, we suggested that a dysregulated STAT3 pathway following SARS-Co-2 infection ultimately leads to PAI-1 activation and cascades of pathologies. The major COVID-19-associated metabolic risks (old age, hypertension, cardiovascular diseases, diabetes, and obesity) share high PAI-1 levels and could predispose certain groups to severe COVID-19 complications. In this review article, we describe the common metabolic profile that is shared between all of these high-risk groups and COVID-19. This profile not only involves high levels of PAI-1 and STAT3 as previously described, but also includes low levels of glutamine and NAD+, coupled with overproduction of hyaluronan (HA). SARS-CoV-2 infection exacerbates this metabolic imbalance and predisposes these patients to the severe pathophysiologies of COVID-19, including the involvement of NETs (neutrophil extracellular traps) and HA overproduction in the lung. While hyperinflammation due to proinflammatory cytokine overproduction has been frequently documented, it is recently recognized that the immune response is markedly suppressed in some cases by the expansion and activity of MDSCs (myeloid-derived suppressor cells) and FoxP3+ Tregs (regulatory T cells). The metabolomics profiles of severe COVID-19 patients and patients with advanced cancer are similar, and in high-risk patients, SARS-CoV-2 infection leads to aberrant STAT3 activation, which promotes a cancer-like metabolism. We propose that glutamine deficiency and overproduced HA is the central metabolic characteristic of COVID-19 and its high-risk groups. We suggest the usage of glutamine supplementation and the repurposing of cancer drugs to prevent the development of severe COVID-19 pneumonia.


Asunto(s)
COVID-19/fisiopatología , Glutamina/deficiencia , Animales , COVID-19/sangre , COVID-19/epidemiología , Comorbilidad , Glutamina/sangre , Humanos , Ácido Hialurónico/sangre , Metaboloma , Inhibidor 1 de Activador Plasminogénico/sangre , Factores de Riesgo , Índice de Severidad de la Enfermedad
12.
J Biol Chem ; 296: 100646, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33839150

RESUMEN

Dysregulated glucagon secretion deteriorates glycemic control in type 1 and type 2 diabetes. Although insulin is known to regulate glucagon secretion via its cognate receptor (insulin receptor, INSR) in pancreatic alpha cells, the role of downstream proteins and signaling pathways underlying insulin's activities are not fully defined. Using in vivo (knockout) and in vitro (knockdown) studies targeting insulin receptor substrate (IRS) proteins, we compared the relative roles of IRS1 and IRS2 in regulating alpha cell function. Alpha cell-specific IRS1-knockout mice exhibited glucose intolerance and inappropriate glucagon suppression during glucose tolerance tests. In contrast, alpha cell-specific IRS2-knockout animals manifested normal glucose tolerance and suppression of glucagon secretion after glucose administration. Alpha cell lines with stable IRS1 knockdown could not repress glucagon mRNA expression and exhibited a reduction in phosphorylation of AKT Ser/Thr kinase (AKT, at Ser-473 and Thr-308). AlphaIRS1KD cells also displayed suppressed global protein translation, including reduced glucagon expression, impaired cytoplasmic Ca2+ response, and mitochondrial dysfunction. This was supported by the identification of novel IRS1-specific downstream target genes, Trpc3 and Cartpt, that are associated with glucagon regulation in alpha cells. These results provide evidence that IRS1, rather than IRS2, is a dominant regulator of pancreatic alpha cell function.


Asunto(s)
Células Secretoras de Glucagón/patología , Glucagón/metabolismo , Intolerancia a la Glucosa/patología , Proteínas Sustrato del Receptor de Insulina/fisiología , Resistencia a la Insulina , Animales , Femenino , Células Secretoras de Glucagón/metabolismo , Intolerancia a la Glucosa/etiología , Intolerancia a la Glucosa/metabolismo , Masculino , Ratones , Ratones Noqueados , Fosforilación , Transducción de Señal
13.
Mod Rheumatol Case Rep ; 4(1): 16-20, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-33086965

RESUMEN

An 18-year-old female was diagnosed with subclinical Cushing's syndrome (CS) due to a left adrenal adenoma. When she was 20 years old, she developed lupus nephritis. She was treated with high-dose prednisolone (PSL) and soon developed the symptoms of CS. When she was 25 years old, we evaluated her serum glucocorticoid level while she continued to take oral PSL. The result suggested her CS was affected by both the oral PSL and the endogenous cortisol secreted by the adrenocortical adenoma, which was therefore resected. Seven months after the operation, the patient's body weight was decreasing, and her SLE remained in clinical remission. CS complicated by SLE is rare, and the decision to surgically remove an adrenal tumor in such a case is even more rare.


Asunto(s)
Síndrome de Cushing/complicaciones , Lupus Eritematoso Sistémico/complicaciones , Lupus Eritematoso Sistémico/diagnóstico , Adolescente , Adenoma Corticosuprarrenal/diagnóstico , Adenoma Corticosuprarrenal/etiología , Adenoma Corticosuprarrenal/cirugía , Toma de Decisiones Clínicas , Síndrome de Cushing/diagnóstico , Manejo de la Enfermedad , Femenino , Glucocorticoides/sangre , Humanos , Nefritis Lúpica/diagnóstico , Nefritis Lúpica/tratamiento farmacológico , Nefritis Lúpica/etiología , Prednisolona/administración & dosificación , Resultado del Tratamiento
14.
J Biol Chem ; 293(16): 5934-5946, 2018 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-29496993

RESUMEN

Chronic low-grade inflammation in the pancreatic islets is observed in individuals with type 2 diabetes, and macrophage levels are elevated in the islets of these individuals. However, the molecular mechanisms underlying the interactions between the pancreatic ß cells and macrophages and their involvement in inflammation are not fully understood. Here, we investigated the role of S100 calcium-binding protein A8 (S100A8), a member of the damage-associated molecular pattern molecules (DAMPs), in ß-cell inflammation. Co-cultivation of pancreatic islets with unstimulated peritoneal macrophages in the presence of palmitate (to induce lipotoxicity) and high glucose (to induce glucotoxicity) synergistically increased the expression and release of islet-produced S100A8 in a Toll-like receptor 4 (TLR4)-independent manner. Consistently, a significant increase in the expression of the S100a8 gene was observed in the islets of diabetic db/db mice. Furthermore, the islet-derived S100A8 induced TLR4-mediated inflammatory cytokine production by migrating macrophages. When human islet cells were co-cultured with U937 human monocyte cells, the palmitate treatment up-regulated S100A8 expression. This S100A8-mediated interaction between islets and macrophages evoked ß-cell apoptosis, which was ameliorated by TLR4 inhibition in the macrophages or S100A8 neutralization in the pancreatic islets. Of note, both glucotoxicity and lipotoxicity triggered S100A8 secretion from the pancreatic islets, which in turn promoted macrophage infiltration of the islets. Taken together, a positive feedback loop between islet-derived S100A8 and macrophages drives ß-cell apoptosis and pancreatic islet inflammation. We conclude that developing therapeutic approaches to inhibit S100A8 may serve to prevent ß-cell loss in patients with diabetes.


Asunto(s)
Apoptosis , Calgranulina A/inmunología , Inflamación/inmunología , Células Secretoras de Insulina/inmunología , Macrófagos/inmunología , Animales , Línea Celular , Células Cultivadas , Glucosa/inmunología , Humanos , Células Secretoras de Insulina/citología , Macrófagos/citología , Masculino , Ratones Endogámicos C57BL , Palmitatos/inmunología , Transducción de Señal , Receptor Toll-Like 4/inmunología
15.
J Diabetes Investig ; 9(1): 25-32, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28429513

RESUMEN

AIMS/INTRODUCTION: Glucagon-like peptide-1 (GLP-1) secreted from enteroendocrine L cells is an incretin that potentiates insulin secretion and is already applied in therapies for type 2 diabetes. However, detailed examination of L cells throughout the gastrointestinal tract remains unclear, because of difficulties in purifying scattered L cells from other cells. In the present study, we identified characteristics of L cells of the upper small intestine (UI), the lower small intestine (LI) and the colon using glucagon-green fluorescent protein-expressing mice that express GFP driven by the proglucagon promoter. MATERIALS AND METHODS: The localization and density of primary L cells were evaluated by anti-green fluorescent protein antibody reactivity. GLP-1 content, messenger ribonucleic acid (mRNA) expression levels and secretion in purified L cells were measured. RESULTS: The number of L cells significantly increased toward the colon. In contrast, the GLP-1 content and secretion from L cells were higher in the UI than in the LI and colon. L cells from the UI and LI expressed notably high mRNA levels of the transcription factor, islet 1. The mRNA expression levels of peptide YY in L cells were higher in the LI than in the UI and colon. The mRNA expression levels of gastric inhibitory polypeptide in L cells from the UI were significantly higher compared with those from the LI and colon. CONCLUSIONS: L cells show different numbers and characteristics throughout the gut, and they express different mRNA levels of transcription factors and gastrointestinal hormones. These results contribute to the therapeutic application of promoting GLP-1 release from L cells for the treatment of type 2 diabetes.


Asunto(s)
Células Enteroendocrinas/metabolismo , Hormonas Gastrointestinales/metabolismo , Tracto Gastrointestinal/metabolismo , Animales , Recuento de Células , Células Enteroendocrinas/citología , Polipéptido Inhibidor Gástrico/metabolismo , Tracto Gastrointestinal/citología , Péptido 1 Similar al Glucagón/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Cultivo Primario de Células , ARN Mensajero/metabolismo , Factores de Transcripción/metabolismo
16.
Medicine (Baltimore) ; 96(9): e6034, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28248860

RESUMEN

RATIONALE: Lymphocytic hypophysitis is a relatively rare autoimmune disease defined by lymphocytic infiltration to the pituitary. Its rarity and wide spectrum of clinical manifestations make clarification of the pathology difficult. Here, we describe a case we examined from the primary diagnosis to final discharge, showing the serial progression of lymphocytic infundibuloneurohypophysitis (LINH) to panhypopituitarism with extrapituitary inflammatory invasion in a short period, and responding favorably to high-dose glucocorticoid treatment. PATIENT CONCERNS: Polyuria, General fatigue and Nausea/Vomiting. DIAGNOSES: Central diabetes insipidus (CDI), Lymphocytic infundibuloneurohypophysitis (LINH). INTERVENTIONS: Desmopressin acetate, High-dose glucocorticoid (GC) treatment. OUTCOMES: He was prescribed desmopressin acetate and subsequently discharged. A month later, he revisited our hospital with general fatigue and nausea/vomiting. A screening test disclosed hypopituitarism with adrenal insufficiency. MRI revealed expanded contrast enhancement to the peripheral extrapituitary lesion. He received high-dose GC treatment and the affected lesion exhibited marked improvement on MRI, along with the recovery of the anterior pituitary function. LESSONS: This case demonstrates the potential for classical LINH to develop into panhypopituitarsim. We consider this is the first documentation of approaching the cause of atypical LINH with progressive clinical course from the pathological viewpoint.


Asunto(s)
Hipofisitis Autoinmune/complicaciones , Hipopituitarismo/etiología , Anciano , Progresión de la Enfermedad , Humanos , Masculino
17.
Endocrinology ; 158(5): 1172-1180, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28324023

RESUMEN

Free fatty acid receptors GPR120 and GPR40 are involved in the secretion of gut hormones. GPR120 and GPR40 are expressed in enteroendocrine K cells, and their activation induces the secretion of the incretin glucose-dependent insulinotropic polypeptide (GIP). However, the role of these receptors in fat-induced GIP secretion in vivo and the associated mechanisms are unclear. In this study, we investigated corn oil-induced GIP secretion in GPR120-knockout (GPR120-/-) and GPR40-knockout (GPR40-/-) mice. Oil-induced GIP secretion was reduced by 50% and 80% in GPR120-/- and GPR40-/- mice, respectively, compared with wild-type mice. This was not associated with a significant difference in K-cell number or GIP content in K cells, nor messenger RNA levels of the lipid receptor GPR119, nor bile acid receptors TGR5 and farnesoid X receptor. GPR120-/- and GPR40-/- mice also exhibited substantially decreased levels of cholecystokinin (CCK), a hormone from I cells that promotes bile and pancreatic lipase secretion, and this decrease was associated with impaired gallbladder contraction. Notably, treatment with a CCK analog resulted in recovery of oil-induced GIP secretion in GPR120-/- mice but not in GPR40-/- mice. These results indicate that corn oil-induced GIP secretion from K cells involves both GPR120 and GPR40 signaling pathways, and GPR120-induced GIP secretion is indirectly mediated by CCK.


Asunto(s)
Colecistoquinina/metabolismo , Aceite de Maíz/farmacología , Polipéptido Inhibidor Gástrico/metabolismo , Receptores Acoplados a Proteínas G/fisiología , Animales , Grasas de la Dieta/farmacología , Células Enteroendocrinas/efectos de los fármacos , Células Enteroendocrinas/metabolismo , Ácidos Grasos no Esterificados/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Acoplados a Proteínas G/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
18.
Diabetes ; 66(4): 868-879, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28096257

RESUMEN

Gastric inhibitory polypeptide receptor (GIPR) directly induces energy accumulation in adipose tissue in vitro. However, the importance of the direct effect of GIPR signaling on adipose tissue in vivo remains unclear. In the current study, we generated adipose tissue-specific GIPR knockout (GIPRadipo-/-) mice and investigated the direct actions of GIP in adipose tissue. Under high-fat diet (HFD)-fed conditions, GIPRadipo-/- mice had significantly lower body weight and lean body mass compared with those in floxed GIPR (GIPRfl/fl) mice, although the fat volume was not significantly different between the two groups. Interestingly, insulin resistance, liver weight, and hepatic steatosis were reduced in HFD-fed GIPRadipo-/- mice. Plasma levels of interleukin-6 (IL-6), a proinflammatory cytokine that induces insulin resistance, were reduced in HFD-fed GIPRadipo-/- mice compared with those in HFD-fed GIPRfl/fl mice. Suppressor of cytokine signaling 3 (SOCS3) signaling is located downstream of the IL-6 receptor and is associated with insulin resistance and hepatic steatosis. Expression levels of SOCS3 mRNA were significantly lower in adipose and liver tissues of HFD-fed GIPRadipo-/- mice compared with those of HFD-fed GIPRfl/fl mice. Thus, GIPR signaling in adipose tissue plays a critical role in HFD-induced insulin resistance and hepatic steatosis in vivo, which may involve IL-6 signaling.


Asunto(s)
Dieta Alta en Grasa , Hígado Graso/genética , Resistencia a la Insulina/genética , Interleucina-6/metabolismo , Hígado/metabolismo , Obesidad/genética , ARN Mensajero/metabolismo , Receptores de la Hormona Gastrointestinal/genética , Células 3T3-L1 , Tejido Adiposo/metabolismo , Animales , Peso Corporal/genética , Metabolismo Energético , Ensayo de Inmunoadsorción Enzimática , Hígado Graso/metabolismo , Prueba de Tolerancia a la Glucosa , Inmunohistoquímica , Metabolismo de los Lípidos/genética , Hígado/diagnóstico por imagen , Locomoción , Ratones , Ratones Noqueados , Obesidad/metabolismo , Transducción de Señal , Proteína 3 Supresora de la Señalización de Citocinas/genética , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Tomografía Computarizada por Rayos X
19.
Medicine (Baltimore) ; 96(51): e9091, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29390437

RESUMEN

RATIONALE: Unilateral adrenalectomy as part of surgical resection of renal cell carcinoma (RCC) is not thought to increase the risk of chronic adrenal insufficiency, as the contralateral adrenal gland is assumed to be capable of compensating for the lost function of the resected gland. However, recent studies have indicated that adrenalectomy might cause irreversible impairment of the adrenocortical reserve. We describe a case of chronic primary adrenal insufficiency in a 68-year-old man who previously underwent unilateral adrenonephrectomy, which was complicated by severe postoperative adrenal stress that involved cardiopulmonary disturbance and systemic infection. PATIENT CONCERNS: A 68-year-old Japanese man presented with weight loss of 6 kg over a 4-month period, and renal biopsy confirmed a diagnosis of RCC. He underwent adrenonephrectomy for the RCC, but developed postoperative septic shock because of a retroperitoneal cystic infection and ventricular fibrillation that was induced by vasospastic angina. The patient was successfully treated using antibiotics and percutaneous coronary intervention, and was subsequently discharged with no apparent complications except decreased appetite and general fatigue. However, his appetite and fatigue did not improve over time and he was readmitted for an examination. DIAGNOSES: The workup revealed a markedly elevated adrenocorticotropic hormone (ACTH) level (151.4 pg/mL, normal: 7-50 pg/mL) and a mildly decreased morning serum cortisol level (6.4 mg/mL, normal: 7-28 mg/mL). In addition to the patient's clinical symptoms and laboratory results, the results from ACTH and corticotropin-releasing hormone stimulation tests were used to make a diagnosis of primary adrenal insufficiency. INTERVENTIONS: Treatment was initiated using oral prednisolone (20 mg), which rapidly resolved his symptoms. At the 1-year follow-up, the patient had a markedly decreased serum cortisol level (2.0 mg/mL) with an ACTH level that was within the normal range (44.1 pg/mL) before his morning dose of prednisolone, which confirmed the diagnosis of chronic primary adrenal insufficiency. LESSONS: Clinicians must be aware of chronic adrenal insufficiency as a possible complication of unilateral adrenalectomy, especially when patients who underwent unilateral adrenalectomy experience severe adrenal stress.


Asunto(s)
Enfermedad de Addison/etiología , Adrenalectomía/efectos adversos , Nefrectomía/efectos adversos , Enfermedad de Addison/diagnóstico , Enfermedad de Addison/tratamiento farmacológico , Anciano , Carcinoma de Células Renales/cirugía , Glucocorticoides/uso terapéutico , Humanos , Hidrocortisona/sangre , Neoplasias Renales/cirugía , Masculino , Prednisolona/uso terapéutico
20.
Endocr J ; 63(3): 249-55, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26727302

RESUMEN

Primary aldosteronism (PA) is associated with a higher rate of cardiovascular events than essential hypertension. Although adrenalectomy has been reported to reduce carotid intima-media thickness (IMT) in patients with PA, the effects of the selective aldosterone blocker, eplerenone, on vascular damage in these patients remains unclear. To evaluate the effects of eplerenone on vascular status in PA patients, we sequentially measured carotid IMT (using computer software to calculate an average IMT for accurate and reproducible evaluation) in 22 patients including 8 patients treated by unilateral adrenalectomy and 14 patients treated with eplerenone for 12 months. Patients who underwent adrenalectomy showed significant reductions in aldosterone concentration (from 345 ± 176 pg/mL to 67 ± 34 pg/mL; P<0.01) and IMT (from 0.67 ± 0.07 mm to 0.63 ± 0.09 mm; P<0.05) 6 months after surgery. Patients treated with eplerenone showed significant reductions in IMT from baseline (0.75 ± 0.10 mm) to 6 (0.71 ± 0.11 mm; P<0.05) and 12 (0.65 ± 0.09 mm; P<0.01) months, although plasma aldosterone level increased significantly, from 141 ± 105 pg/mL to 207 ± 98 pg/mL (P<0.05). Eplerenone treatment of patients with PA reduces blood pressure, increases serum potassium level, and improves vascular status. Carotid IMT may be a useful marker for evaluating the effectiveness of eplerenone in patients with PA.


Asunto(s)
Aterosclerosis/prevención & control , Hiperaldosteronismo/tratamiento farmacológico , Antagonistas de Receptores de Mineralocorticoides/uso terapéutico , Espironolactona/análogos & derivados , Adrenalectomía/efectos adversos , Adulto , Anciano , Aldosterona/sangre , Aterosclerosis/diagnóstico por imagen , Aterosclerosis/epidemiología , Aterosclerosis/etiología , Biomarcadores , Grosor Intima-Media Carotídeo , Monitoreo de Drogas , Eplerenona , Femenino , Estudios de Seguimiento , Humanos , Hiperaldosteronismo/sangre , Hiperaldosteronismo/fisiopatología , Hiperaldosteronismo/cirugía , Hipertensión/etiología , Hipertensión/prevención & control , Hipopotasemia/etiología , Hipopotasemia/prevención & control , Japón/epidemiología , Masculino , Persona de Mediana Edad , Antagonistas de Receptores de Mineralocorticoides/efectos adversos , Potasio/sangre , Reproducibilidad de los Resultados , Riesgo , Espironolactona/efectos adversos , Espironolactona/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...