Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-37265473

ABSTRACT

Primary aldosteronism (PA) is the most common cause of endocrine hypertension. Unilateral PA can be cured using unilateral adrenalectomy (Adx). PA surgery outcome (PASO) criteria, which include clinical and biochemical outcomes, have been proposed to evaluate PA cure after Adx. However, clinical outcomes are often inconsistent with biochemical outcomes. In addition, although confirmatory tests are included as endpoints of biochemical outcomes in the PASO criteria, their clinical usefulness has not yet been established. We evaluated clinical parameters and confirmatory test results before and after Adx in 16 patients with PA and assessed the usefulness of the confirmatory tests. The following were the clinical outcomes after Adx: 37.5% complete success, 62.5% partial success, and 0% absent success. The ratio of biochemical complete success was as follows: 69% aldosterone/renin ratio and basal plasma aldosterone concentration, 19% as assessed by the captopril challenge test, 47% as assessed by the saline infusion test, 30% as assessed by the furosemide upright test, and 100% urine aldosterone. Of these, biochemical complete success was judged in four cases by aldosterone/renin ratio and basal plasma aldosterone concentration, one case by captopril challenge test, five cases by saline infusion test, and one case by furosemide upright test. Although clinical outcomes and urine aldosterone levels improved after Adx, confirmatory tests failed to improve in some cases. The current criteria are not considered useful for biochemical evaluation after Adx. To determine whether additional treatment with mineralocorticoid receptor antagonists is required, more accurate biochemical criteria should be established after Adx.


Subject(s)
Hyperaldosteronism , Hypertension , Humans , Adrenalectomy/methods , Aldosterone , Hyperaldosteronism/diagnosis , Hyperaldosteronism/surgery , Captopril , Furosemide , Renin , Saline Solution , Retrospective Studies
2.
Sci Rep ; 12(1): 1294, 2022 01 25.
Article in English | MEDLINE | ID: mdl-35079086

ABSTRACT

Immune checkpoint inhibitors (ICIs) treatment can result in endocrine immune-related adverse events (irAEs), including pituitary dysfunction. Quick diagnosis of secondary adrenal insufficiency (AI) is challenging because no universal definition of ICI-induced secondary AI has been agreed. The aim of this study was to clarify the clinical features of ICI-induced secondary AI that can be used for screening in standard clinical practice. This retrospective study was performed using the medical records of patients who received ICIs at Hirosaki University Hospital between 1 September 2014 and 31 January 2021. Longitudinal clinical data of patients who developed AI were analyzed and compared with the data of thyroid irAEs. Regression analysis showed a significant correlation between ICI-induced secondary AI and absolute or relative eosinophil counts at pre-onset of AI, as well as differences or rate of increase in eosinophil counts at baseline and at pre-onset. Absolute eosinophil counts > 198.36/µL or relative eosinophil counts > 5.6% at pre-onset, and a difference of 65.25/µL or a rate of eosinophil count increase of 1.97 between the baseline and at pre-onset showed the best sensitivity and specificity. This is the first report to demonstrate that eosinophil counts can be a predictor of ICI-induced secondary AI.


Subject(s)
Adrenal Insufficiency/chemically induced , Adrenal Insufficiency/immunology , Eosinophils/immunology , Immune Checkpoint Inhibitors/adverse effects , Neoplasms/drug therapy , Adrenal Insufficiency/blood , Aged , Biomarkers , C-Reactive Protein/analysis , Female , Humans , Leukocyte Count , Longitudinal Studies , Male , Middle Aged , Prognosis , Retrospective Studies , Sodium/blood
3.
Endocr J ; 67(3): 353-359, 2020 Mar 28.
Article in English | MEDLINE | ID: mdl-31801917

ABSTRACT

McCune-Albright syndrome (MAS) is a rare disorder. MAS is classically defined by the occurrence of fibrous dysplasia, café-au-lait skin macules, and precocious puberty. In addition to precocious puberty, other hyperfunctioning endocrinopathies may occur. We evaluated hypothalamic-pituitary-adrenal function in two cases of typical MAS associated with fibrous dysplasia and growth hormone excess. Pituitary adenoma or hyperplasia was not detected by magnetic resonance imaging. Hormonal data showed normal or low cortisol levels, despite high ACTH levels in the blood. A high ratio of circulating ACTH to cortisol was found in the two cases. Insulin tolerance and CRH tests showed hyper-responses of ACTH and an insufficient increase in cortisol levels. No involvement of 11ß-HSD1 by GH excess was suggested because basal levels of ACTH and cortisol showed no changes, even after therapy for acromegaly by somatostatin analogues. Patients with Cushing's disease cases of pituitary macroadenoma can have high circulating ACTH precursor levels, and elevated ACTH precursors have been observed in ectopic ACTH syndrome. Autonomous cortisol excess was excluded by the level of midnight cortisol and the level of cortisol after a low-dose dexamethasone suppression test in the two cases. Finally, the gel filtration profiles of immunoreactive ACTH contents showed the presence of aberrant ACTH precursors. To the best of our knowledge, there have been no reports of MAS associated with aberrant ACTH precursors. Our findings in these cases emphasize that attention should be to secretion of inactive ACTH precursors in MAS.


Subject(s)
Adrenocorticotropic Hormone/blood , Fibrous Dysplasia, Polyostotic/blood , Hydrocortisone/blood , Pro-Opiomelanocortin/blood , Adult , Humans , Insulin Resistance/physiology , Male
4.
Endocr J ; 67(2): 177-184, 2020 Feb 28.
Article in English | MEDLINE | ID: mdl-31666445

ABSTRACT

Cushing's disease is primarily caused by autonomic hypersecretion of adrenocorticotropic hormone (ACTH) from a pituitary adenoma. In Cushing's disease, mutations in the ubiquitin-specific protease 8 (USP8) have been detected. These mutations are associated with hyperactivation of USP8 that prevent epidermal growth factor receptor (EGFR) degradation. This leads to increased EGFR stability and results in the maintenance of EGFR signaling in Cushing's disease. USP8 inhibitors can suppress the growth of various tumors. In this study, the effects of a potent USP8 inhibitor, DUBs-IN-2, on ACTH production and cell proliferation were examined in mouse corticotroph tumor (AtT-20) cells. Proopiomelanocortin (Pomc) mRNA levels and ACTH levels were decreased in AtT-20 cells by DUBs-IN-2. Further, cell proliferation was inhibited, and apoptosis was induced by DUBs-IN-2. Transcript levels of pituitary tumor-transforming gene 1 (Pttg1), a pituitary tumor growth marker, were increased; and transcript levels of stress response growth arrest and DNA damage-inducible 45 (Gadd45ß) and Cdk5 and ABL enzyme substrate 1 (Cables1) mRNA levels were increased in response to the drug. Gadd45ß or Cables1 knockdown partially inhibited the DUBs-IN-2-induced decrease in cell proliferation, but not Pomc mRNA levels. Both GADD45ß and CABLES1 may be responsible, at least in part, for the USP8-induced suppression of corticotroph tumor cell proliferation. USP-8 may be a new treatment target in Cushing's disease.


Subject(s)
ACTH-Secreting Pituitary Adenoma/metabolism , Adenoma/metabolism , Adrenocorticotropic Hormone/drug effects , Apoptosis/drug effects , Cell Proliferation/drug effects , Endosomal Sorting Complexes Required for Transport/antagonists & inhibitors , Indenes/pharmacology , Pyrazines/pharmacology , Ubiquitin Thiolesterase/antagonists & inhibitors , Adrenocorticotropic Hormone/metabolism , Animals , Antigens, Differentiation/drug effects , Antigens, Differentiation/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase 5/drug effects , Cyclin-Dependent Kinase 5/genetics , Cyclins/drug effects , Cyclins/genetics , Endopeptidases , ErbB Receptors/drug effects , ErbB Receptors/metabolism , Gene Knockdown Techniques , Mice , Pituitary ACTH Hypersecretion/metabolism , Pro-Opiomelanocortin/drug effects , Pro-Opiomelanocortin/genetics , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Securin/drug effects , Securin/genetics
5.
Intern Med ; 58(24): 3557-3562, 2019 Dec 15.
Article in English | MEDLINE | ID: mdl-31462588

ABSTRACT

Pembrolizumab, or anti-programmed death receptor 1 antibody, is an immune checkpoint inhibitor that can cause immune-related adverse events. We herein report for the first time the progression of hypopituitarism and hypothyroidism after treatment with pembrolizumab in a patient with adrenal metastasis of non-small-cell lung cancer. Severe primary hypothyroidism occurred three weeks after the first administration of pembrolizumab. Four months after the discontinuation of pembrolizumab, isolated adrenocorticotropic hormone (ACTH) deficiency was noted. Corticotropin-releasing hormone and rapid ACTH tests performed repeatedly showed that the patient's pituitary and adrenal function had been gradually deteriorating. It is important to diagnose adrenal insufficiency without delay in order to prevent adrenal crisis.


Subject(s)
Adrenal Gland Neoplasms/drug therapy , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Agents, Immunological/adverse effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Hypopituitarism/chemically induced , Hypothyroidism/chemically induced , Lung Neoplasms/drug therapy , Adrenal Gland Neoplasms/secondary , Adrenal Insufficiency/chemically induced , Adrenocorticotropic Hormone/deficiency , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Carcinoma, Non-Small-Cell Lung/secondary , Corticotropin-Releasing Hormone/analysis , Disease Progression , Humans , Lung Neoplasms/pathology , Male
6.
Endocr J ; 66(6): 515-522, 2019 Jun 28.
Article in English | MEDLINE | ID: mdl-30880293

ABSTRACT

Cushing's disease is almost always caused by hypersecretion of adrenocorticotropic hormone (ACTH) from a pituitary adenoma. A mutation in the deubiquitinase gene USP8 has been found in human ACTH-producing pituitary adenoma cells. This mutational hotspot hyperactivates USP8, rescuing epidermal growth factor receptor (EGFR) from lysosomal degradation and ensuring its sustained signaling in Cushing's disease. An EGFR inhibitor would be an effective anti-tumor agent in EGFR-related tumors. We investigated the effect of a potent dual tyrosine kinase inhibitor, lapatinib, on ACTH production and cell proliferation in AtT-20 mouse corticotroph tumor cells. Lapatinib decreased proopiomelanocortin (Pomc) mRNA levels and ACTH levels in AtT-20 cells and also inhibited cell proliferation, induced apoptosis, and decreased pituitary tumor-transforming gene 1 (Pttg1), a hallmark of pituitary tumors, mRNA levels. KSN/Slc nude mice were subcutaneously inoculated with AtT-20 cells. After 1 week, the mice were randomized either to control or lapatinib groups. The inhibitor decreased the tumor weight of AtT-20 allografts in vivo versus control mice. Lapatinib also significantly decreased Pomc and Pttg1 mRNA levels in the tumor and plasma ACTH and corticosterone levels in vivo. Thus, lapatinib decreases the ACTH production and proliferation of corticotroph tumor cells. An EGFR-targeting therapy could be an important treatment for Cushing's disease.


Subject(s)
Adrenocorticotropic Hormone/metabolism , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Corticotrophs/drug effects , Lapatinib/pharmacology , Protein Kinase Inhibitors/pharmacology , ACTH-Secreting Pituitary Adenoma/drug therapy , ACTH-Secreting Pituitary Adenoma/pathology , Adenoma/drug therapy , Adenoma/pathology , Adrenocorticotropic Hormone/blood , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Corticosterone/blood , Corticotrophs/metabolism , Corticotrophs/pathology , Lapatinib/therapeutic use , Mice , Mice, Nude , Neoplasm Transplantation , Protein Kinase Inhibitors/therapeutic use
7.
Onco Targets Ther ; 10: 4329-4338, 2017.
Article in English | MEDLINE | ID: mdl-28919782

ABSTRACT

PURPOSE: The primary cause of Cushing's disease is adrenocorticotropic hormone (ACTH)-producing pituitary adenomas. EGFR signaling induces POMC mRNA-transcript levels and ACTH secretion from corticotroph tumors. The Jak-STAT pathway is located downstream of EGFR signaling; therefore, a Jak2 inhibitor could be an effective therapy for EGFR-related tumors. In this study, we determined the effect of a potent and selective Jak2 inhibitor, SD1029, on ACTH production and proliferation in mouse AtT20 corticotroph tumor cells. MATERIALS AND METHODS: AtT20 pituitary corticotroph tumor cells were cultured after transfection with PTTG1- or GADD45ß-specific siRNA. Expression levels of mouse POMC, PTTG1, and GADD45ß mRNAs were evaluated using quantitative real-time polymerase chain reaction. ACTH levels were measured using ACTH ELISA. Western blot analysis was performed to examine protein expression of phosphorylated STAT3/STAT3. Viable cells and DNA fragmentation were measured using a cell-proliferation assay and cell-death detection ELISA, respectively. Cellular DNA content was analyzed using fluorescence-activated cell sorting. RESULTS: SD1029 decreased POMC and PTTG1 mRNA and ACTH levels, while increasing GADD45ß levels. The drug also decreased AtT20-cell proliferation and induced apoptosis, but did not alter cell-cycle progression. SD1029 also inhibited STAT3 phosphorylation. PTTG1 knockdown inhibited POMC mRNA levels and cell proliferation. However, combined treatment with PTTG1 knockdown and SD1029 had no additive effect on POMC mRNA levels or cell proliferation. GADD45ß knockdown inhibited the SD1029-induced decrease in POMC mRNA levels and also partially inhibited the decrease in cell proliferation. CONCLUSION: Both PTTG1 and GADD45ß may be responsible, at least in part, for the Jak2-induced suppression of ACTH synthesis and cell proliferation. Accordingly, therapies that target EGFR-dependent Jak2/STAT3 may have clinical applications for treating Cushing's disease.

8.
Endocr J ; 62(12): 1083-90, 2015.
Article in English | MEDLINE | ID: mdl-26497760

ABSTRACT

Cushing's disease is primarily caused by adrenocorticotropic hormone (ACTH)-producing pituitary adenomas. Pituitary tumor-transforming gene 1 (PTTG1) expression, a hallmark of pituitary tumors, stimulates pituitary cell proliferation. Histone deacetylases (HDACs) play an important role in regulating gene transcription and HDAC inhibitors induce cellular differentiation and suppress tumor cell proliferation. HDAC inhibitors also repress PTTG1 mRNA levels. Trichostatin A (TSA) is a potent cell-permeable HDAC inhibitor that blocks cell cycle progression. In the present study, we determined the effect of TSA on ACTH production and cellular proliferation in mouse AtT-20 corticotroph tumor cells. TSA decreased proopiomelanocortin (POMC) mRNA levels in AtT-20 cells and reduced ACTH levels in the culture medium of these cells. The TSA-induced decreases in POMC mRNA levels were not modulated when TSA and dexamethasone were simultaneously administered. Drug treatment also decreased AtT-20 cell proliferation, induced apoptosis, and increased the percentage of cells in G0/G1 phase using flow cytometry. TSA decreased PTTG1 mRNA levels. Furthermore, PTTG1 knockdown inhibited cellular proliferation. Its knockdown also inhibited POMC mRNA and ACTH levels. TSA inhibits ACTH production and corticotroph tumor cell proliferation. TSA may inhibit cellular proliferation, and ACTH synthesis and secretion by decreasing PTTG1 expression.


Subject(s)
ACTH-Secreting Pituitary Adenoma/metabolism , ACTH-Secreting Pituitary Adenoma/pathology , Adrenocorticotropic Hormone/biosynthesis , Cell Proliferation/drug effects , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Adrenocorticotropic Hormone/analysis , Animals , Apoptosis/drug effects , Cell Line, Tumor , Gene Expression/drug effects , Gene Knockdown Techniques , Mice , Pituitary ACTH Hypersecretion , Pro-Opiomelanocortin/genetics , RNA, Messenger/analysis , Securin/genetics
9.
BMC Res Notes ; 8: 268, 2015 Jun 26.
Article in English | MEDLINE | ID: mdl-26111524

ABSTRACT

BACKGROUND: Listeria monocytogenes is a facultative intracellular parasitic bacterium that is Gram positive, catalase positive, oxidase negative, and a facultative anaerobe. It is known to infect humans through food. It is a bacillus with low virulence, but can cause meningitis and sepsis in infants and immunocompromised patients. CASE PRESENTATION: A case of 75-year-old Japanese female with small cell carcinoma of the thymus and pleural dissemination is described. She was treated with carboplatin and etoposide and showed a partial response. However, the tumor recurred 6 months later. Therefore, we again administered carboplatin and etoposide. Though peritoneal dissemination was suspected based on abdominal computed tomography findings after two courses, the assessment was stable disease. She was occasionally treated for constipation. She developed chills, rigor, and diarrhea, necessitating admission on the 7th day of the third course of chemotherapy. We suspected intestinal infection, and cefepime was thus administered. However, her blood pressure dropped and neutropenia manifested on the 4th day of admission. We therefore switched the antibiotic from cefepime to meropenem and also administered granulocyte-colony stimulating factor. Listeria monocytogenes was detected by two blood cultures, and the antimicrobial medication was thus switched to ampicillin, in consideration of sensitivity. Her general condition improved and she was able to leave the hospital on the 19th day after admission. CONCLUSIONS: During chemotherapy, factors such as impaired bowel movements, malnutrition, and myeloablation can contribute to the development of severe infections. It is necessary to comprehensively assess a patient's state and treat all aspects of illness.


Subject(s)
Antineoplastic Agents/adverse effects , Carboplatin/adverse effects , Carcinoma, Small Cell/drug therapy , Etoposide/adverse effects , Listeriosis/etiology , Neoplasm Recurrence, Local/drug therapy , Sepsis/etiology , Aged , Ampicillin/therapeutic use , Anti-Bacterial Agents/therapeutic use , Antineoplastic Agents/administration & dosage , Carboplatin/administration & dosage , Carcinoma, Small Cell/pathology , Cefepime , Cephalosporins/therapeutic use , Etoposide/administration & dosage , Female , Granulocyte Colony-Stimulating Factor/therapeutic use , Humans , Listeria monocytogenes/drug effects , Listeria monocytogenes/pathogenicity , Listeria monocytogenes/physiology , Listeriosis/drug therapy , Listeriosis/microbiology , Listeriosis/pathology , Meropenem , Neoplasm Recurrence, Local/pathology , Sepsis/drug therapy , Sepsis/microbiology , Sepsis/pathology , Thienamycins/therapeutic use , Thymus Gland/drug effects , Thymus Gland/pathology , Thymus Neoplasms/drug therapy , Thymus Neoplasms/pathology , Treatment Outcome
10.
Endocr J ; 62(7): 645-54, 2015.
Article in English | MEDLINE | ID: mdl-25948499

ABSTRACT

Cushing's disease is primarily caused by pituitary corticotroph adenomas, which autonomically secrete adrenocorticotropic hormone (ACTH). ACTH production may be associated with tumor cell proliferation; however, the effects of cell cycle progression on ACTH production and cell proliferation are little known in corticotroph tumor cells. A DNA polymerase inhibitor, aphidicolin, arrests cells at the entrance to the S phase and blocks the cell cycle; aphidicolin also induces apoptosis in tumor cells. In the present study, we determined ACTH production and cell proliferation of AtT-20 corticotroph tumor cells following treatment with aphidicolin. Aphidicolin decreased proopiomelanocortin mRNA levels in AtT-20 cells and the levels of ACTH in the culture medium of these cells. Aphidicolin also decreased cell proliferation and induced apoptosis in AtT-20 cells. Fluorescence-activated cell sorting analyses revealed that this agent increased the percentage of G0/G1 phase cells, and decreased S phase cells. Aphidicolin decreased the phosphorylation of cyclic adenosine monophosphate response element-binding protein and Akt. Aphidicolin increased the levels of tumor protein 27 (p27) and 53 (p53), while it decreased cyclin E levels. Aphidicolin also increased the mRNA levels of the stress response gene growth arrest and DNA damage-inducible 45ß (GADD45ß), a putative downstream target of p53. The p53 knockdown increased GADD45ß mRNA levels. The GADD45ß knockdown inhibited the decreases in cell proliferation. Thus, aphidicolin inhibits cell proliferation via the p53-GADD45ß pathway in AtT-20 cells.


Subject(s)
Antigens, Differentiation/metabolism , Aphidicolin/pharmacology , Cell Proliferation/drug effects , Tumor Suppressor Protein p53/metabolism , Adrenocorticotropic Hormone/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cell Line, Tumor , Cell Proliferation/physiology , Mice , Phosphorylation/drug effects , Pro-Opiomelanocortin/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...