Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 312
Filter
1.
J Neuroendocrinol ; 35(6): e13287, 2023 06.
Article in English | MEDLINE | ID: mdl-37322808

ABSTRACT

Deficient anterior pituitary with common variable immune deficiency (DAVID) syndrome is a rare condition characterized by adrenocorticotropic hormone (ACTH) deficiency and primary hypogammaglobulinemia. It is due to heterozygous mutations of the nuclear factor kappa-B subunit 2 (NFKB2) gene. Only a few isolated cases have been reported since its first description by our team. Through the international multicenter GENHYPOPIT network, we identified a new case of DAVID syndrome. We then conducted an extensive review of the DAVID syndrome cases published from 2012 to 2022. A 7-year-old boy was diagnosed with symptomatic hypoglycemia revealing ACTH deficiency. Laboratory tests showed asymptomatic hypogammaglobulinemia. He harbored a heterozygous point mutation in NFKB2 gene (c.2600C > T, p.Ala867Val). His management included hydrocortisone replacement treatment, and he also received subcutaneous immunoglobulins during the Covid-19 pandemic. We analyzed 28 cases of DAVID syndrome with ACTH deficiency. ACTH deficiency was the only hormone deficiency in 79% of patients, but some patients harbored growth hormone (GH) and thyroid stimulating hormone (TSH) deficiencies. The first presenting symptoms were sinus/pulmonary infections (82%, mean age of 3 years) and alopecia (mean age of 4.7 years). ACTH deficiency was the third presenting condition (mean age at diagnosis of 8.6 years). All patients had hypogammaglobulinemia (decreased IgA and IgM levels), and 57% of patients had at least one autoimmune manifestation. Heterozygous mutations at the 3'end of the NFKB2 gene, coding for the C-terminal domain of the protein, were identified in all cases. Better knowledge of DAVID syndrome will help clinicians make an early diagnosis to avoid life-threatening complications.


Subject(s)
Common Variable Immunodeficiency , Pituitary Hormones, Anterior , Adult , Child , Female , Humans , Male , Adrenocorticotropic Hormone/deficiency , Agammaglobulinemia/complications , Autoimmunity , Common Variable Immunodeficiency/complications , Common Variable Immunodeficiency/genetics , Common Variable Immunodeficiency/immunology , Common Variable Immunodeficiency/physiopathology , Heterozygote , Human Growth Hormone/deficiency , Infections/complications , Mothers , Mutation , Phenotype , Pituitary Hormones, Anterior/deficiency , Syndrome , Thyrotropin/deficiency
2.
Endocr Pract ; 28(7): 711-718, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35569735

ABSTRACT

OBJECTIVE: To review the diagnosis and management of hypothyroidism during pregnancy, in the preconception period, and in the postpartum period. METHODS: A literature review of English-language papers published between 1982 and 2022, focusing on the most recent literature. RESULTS: During pregnancy, thyroid function laboratory tests need to be interpreted with regard to gestational age. Overt hypothyroidism, regardless of the thyroid-stimulating hormone (TSH) level, should always be promptly treated when it is diagnosed before conception or during pregnancy or lactation. Most women with pre-existing treated hypothyroidism require an increase in levothyroxine (LT4) dosing to maintain euthyroidism during gestation. LT4-treated pregnant patients need close monitoring of their serum TSH levels to avoid overtreatment or undertreatment. There is no consensus about whether to initiate LT4 in women with mild forms of gestational thyroid hypofunction. However, in light of current evidence, it is reasonable to treat women with subclinical hypothyroidism with LT4, particularly if the TSH level is >10 mIU/L or thyroperoxidase antibodies are present. Women who are not treated need to be followed up to ensure that treatment is initiated promptly if thyroid failure progresses. Additional studies are needed to better understand the effects of the initiation of LT4 in early gestation in women with subclinical hypothyroidism and hypothyroxinemia and determine optimal strategies for thyroid function screening in the preconception period and during pregnancy. CONCLUSION: The diagnosis and management of hypothyroidism in the peripregnancy period present specific challenges. While making management decisions, it is essential to weigh the risks and benefits of treatments for not just the mother but also the fetus.


Subject(s)
Hypothyroidism , Pregnancy Complications , Drug Monitoring , Female , Humans , Hypothyroidism/blood , Hypothyroidism/diagnosis , Hypothyroidism/drug therapy , Postnatal Care , Preconception Care , Pregnancy , Pregnancy Complications/blood , Pregnancy Complications/diagnosis , Pregnancy Complications/drug therapy , Prenatal Care , Thyroid Function Tests , Thyrotropin/blood , Thyrotropin/deficiency , Thyroxine/administration & dosage , Thyroxine/blood , Thyroxine/deficiency , Thyroxine/therapeutic use
3.
J Clin Endocrinol Metab ; 106(12): 3655-3667, 2021 11 19.
Article in English | MEDLINE | ID: mdl-34302730

ABSTRACT

CONTEXT: Because subclinical hyperthyroidism increases the risk of osteoporosis and fractures, concerns are growing about the long-term skeletal safety of TSH suppression therapy after total thyroidectomy in patients with differentiated thyroid cancer (DTC). OBJECTIVE: We aimed to determine the effect of TSH suppression therapy on bone mineral density (BMD) in DTC patients. METHODS: We searched PubMed, Embase, the Cochrane library, and other sources. Eligible observational studies included DTC patients who underwent TSH suppression therapy and BMD measurement. Two independent reviewers extracted data on the studies' characteristics and outcomes and determined their risk of bias. Data were extracted from each study for postmenopausal/premenopausal women's and men's lumbar spine (LS), femoral neck (FN), and total hip (TH) BMD and summed using a random-effects meta-analysis model. The weighted mean differences with 95% CIs are expressed for the differences in outcome measurements between groups. RESULTS: Seventeen studies (739 patients and 1085 controls) were included for quantitative analysis. In postmenopausal women, TSH suppression therapy showed a significant decrease in LS BMD (-0.03; -0.05, -0.02), and a similar trend was seen in TH. In premenopausal women, TSH suppression therapy significantly increased LS BMD (0.04; 0.02, 0.06) and FN BMD (0.02; 0.01, 0.04). In men, there was no significant association between TSH suppression therapy and BMD at any site compared with the controls. CONCLUSION: Evidence from observational studies suggests that postmenopausal women treated with TSH suppression therapy are at risk for lower BMD. Attention should be paid to long-term skeletal safety in DTC survivors.


Subject(s)
Adenocarcinoma/surgery , Bone Density , Hyperthyroidism/drug therapy , Osteoporosis/pathology , Thyroid Neoplasms/surgery , Thyroidectomy/adverse effects , Thyrotropin/adverse effects , Adenocarcinoma/pathology , Humans , Hyperthyroidism/etiology , Hyperthyroidism/pathology , Osteoporosis/etiology , Osteoporosis/metabolism , Prognosis , Thyroid Neoplasms/pathology , Thyrotropin/deficiency
4.
BMC Endocr Disord ; 21(1): 111, 2021 May 27.
Article in English | MEDLINE | ID: mdl-34044831

ABSTRACT

BACKGROUND: Low free triiodothyronine (FT3) levels are related to a poor prognosis deterioration in patients with COVID-19 presenting with non-thyroidal illness syndrome (NTI). This study was designed to explore whether free thyroxin (FT4) or thyroid stimulating hormone (TSH) levels affected the mortality of patients with COVID-19 presenting with NTI. METHODS: Patients with COVID-19 complicated with NTI who were treated at our hospital were included in this retrospective study. Patients were divided into low TSH and normal TSH groups, as well as low and normal-high FT4 group, according to the reference range of TSH or FT4 levels. The 90-day mortality and critical illness rates were compared among patients with low and normal TSH levels, as well as among patients with low FT4 levels and normal-high FT4 levels; in addition, differences in demographic and laboratory data were compared. A Kaplan-Meier analysis and Cox proportional hazards models were used to assess the associations of TSH and FT4 levels with mortality. RESULTS: One hundred fifty patients with low FT3 levels and without a history of thyroid disease were included, 68% of whom had normal FT4 and TSH levels. Critical illness rates (74.07% VS 37.40%, P = 0.001) and mortality rates (51.85% VS 22.76%, P = 0.002) were significantly higher in the low TSH group than in the normal TSH group. Although no significant difference in the critical illness rate was found (P = 0.296), the mortality rate was significantly higher in the low FT4 group (P = 0.038). Low TSH levels were independently related to 90-day mortality (hazard ratio = 2.78, 95% CI:1.42-5.552, P = 0.003). CONCLUSIONS: Low FT4 and TSH concentrations were associated with mortality in patients with COVID-19 presenting with NTI; moreover, low TSH levels were an independent risk factor for mortality in these patients.


Subject(s)
COVID-19/epidemiology , COVID-19/mortality , Euthyroid Sick Syndromes/epidemiology , SARS-CoV-2 , Thyrotropin/blood , Thyroxine/blood , Adult , Aged , Aged, 80 and over , COVID-19/blood , Cohort Studies , Comorbidity , Euthyroid Sick Syndromes/blood , Female , Humans , Male , Middle Aged , Prognosis , Proportional Hazards Models , Retrospective Studies , Risk Factors , Thyrotropin/deficiency , Thyroxine/deficiency
7.
Endocr J ; 66(12): 1117-1123, 2019 Dec 25.
Article in English | MEDLINE | ID: mdl-31484850

ABSTRACT

A 14-year-old girl was referred to our department because of headache and visual impairment following the resection of recurrent cardiac myxoma. Head magnetic resonance imaging (MRI) scan detected an intra- and supra-sellar tumor. Moreover, the patient showed the presence of spotty skin pigmentations on her cheeks and lower lip. Blood examination revealed hypothyrotropinemia, and ultrasonography results revealed multiple thyroid nodules. She was diagnosed with Carney complex (CNC). Her pituitary tumor was suspected as growth hormone (GH)-secreting adenoma, because overgrowth was observed in the patient. However, biochemical examinations, including oral glucose tolerance test, failed to show the characteristic findings of GH-secreting adenoma. In contrast, insulin tolerance test showed GH deficiency. Her visual impairment improved without performing decompression surgery, and the tumor size decreased, as per the MRI findings. Based on clinical course, the patient was diagnosed with pituitary apoplexy in pituitary adenoma, following which she was discharged. At 3 months after discharge, thyrotropin-releasing hormone loading test performed revealed low thyrotropin-stimulating hormone and thyroid hormone levels, and the patient was in a depressed mood. Therefore, l-T4 replacement was initiated, following which her GH secretory capacity gradually improved. Here, we report, to the best of our knowledge, the first case of a patient with pituitary apoplexy in CNC. Such condition must be identified in young patients with recurrent cardiac myxoma, and examinations, such as head MRI, must be performed.


Subject(s)
Cardiac Surgical Procedures/adverse effects , Carney Complex/complications , Heart Neoplasms/surgery , Myxoma/surgery , Pituitary Apoplexy/etiology , Pituitary Neoplasms/complications , Adolescent , Carney Complex/diagnosis , Female , Growth Hormone-Secreting Pituitary Adenoma/complications , Humans , Neoplasm Recurrence, Local/surgery , Pituitary Neoplasms/diagnosis , Pituitary Neoplasms/surgery , Thyrotropin/deficiency
8.
J Clin Endocrinol Metab ; 104(12): 6229-6237, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31504637

ABSTRACT

CONTEXT: Congenital isolated TSH deficiency (i-TSHD) is a rare form of congenital hypothyroidism. Five genes (IGSF1, IRS4, TBL1X, TRHR, and TSHB) responsible for the disease have been identified, although their relative frequencies and hypothalamic/pituitary unit phenotypes have remained to be clarified. OBJECTIVES: To define the relative frequencies and hypothalamic/pituitary unit phenotypes of congenital i-TSHD resulting from single gene mutations. PATIENTS AND METHODS: Thirteen Japanese patients (11 boys and 2 girls) with congenital i-TSHD were enrolled. IGSF1, IRS4, TBL1X, TRHR, and TSHB were sequenced. For a TBL1X mutation (p.Asn382del), its pathogenicity was verified in vitro. For a literature review, published clinical data derived from 74 patients with congenital i-TSHD resulting from single-gene mutations were retrieved and analyzed. RESULTS: Genetic screening of the 13 study subjects revealed six mutation-carrying patients (46%), including five hemizygous IGSF1 mutation carriers and one hemizygous TBL1X mutation carrier. Among the six mutation carriers, one had intellectual disability and the other one had obesity, but the remaining four did not show nonendocrine phenotypes. Loss of function of the TBL1X mutation (p.Asn382del) was confirmed in vitro. The literature review demonstrated etiology-specific relationship between serum prolactin (PRL) levels and TRH-stimulated TSH levels with some degree of overlap. CONCLUSIONS: The mutation screening study covering the five causative genes of congenital i-TSHD was performed, showing that the IGSF1 defect was the leading genetic cause of the disease. Assessing relationships between serum PRL levels and TRH-stimulated TSH levels would contribute to predict the etiologies of congenital i-TSHD.


Subject(s)
Congenital Hypothyroidism/genetics , Congenital Hypothyroidism/pathology , Immunoglobulins/genetics , Mass Screening/methods , Membrane Proteins/genetics , Mutation , Thyrotropin/deficiency , Adolescent , Adult , Biomarkers/analysis , Child , Child, Preschool , DNA Mutational Analysis/methods , Female , Follow-Up Studies , Humans , Infant , Infant, Newborn , Insulin Receptor Substrate Proteins/genetics , Male , Pedigree , Prognosis , Receptors, Thyrotropin-Releasing Hormone/genetics , Thyrotropin/blood , Thyrotropin/genetics , Transducin/genetics , Young Adult
9.
Int J Dermatol ; 56(9): 952-956, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28436021

ABSTRACT

BACKGROUND: Burning mouth syndrome (BMS) is a disorder characterized by chronic mouth pain in the absence of objective clinical abnormalities. Vitamin or mineral deficiencies may have a role in BMS, but data regarding the prevalence and relevance of hematinic deficiencies are conflicting. We aimed to determine the frequency of specific laboratory abnormalities in patients with BMS. METHODS: We retrospectively reviewed the results of screening blood tests in patients with BMS at our institution between January 2003 and December 2013. RESULTS: Among 659 patients with BMS, the most common decreased values or deficiencies were vitamin D3 (15%), vitamin B2 (15%), vitamin B6 (5.7%), zinc (5.7%), vitamin B1 (5.3%), thyrotropin (TSH) (3.2%), vitamin B12 (0.8%), and folic acid (0.7%). Laboratory values for fasting blood glucose and TSH were increased in 23.7% and 5.2%, respectively. CONCLUSIONS: In patients with symptoms of BMS, our results suggest it is reasonable to screen for fasting blood glucose, vitamin D (D2 and D3 ), vitamin B6 , zinc, vitamin B1 , and TSH. Deficiencies of vitamin B12 and folic acid were rare (<1% abnormal).


Subject(s)
Avitaminosis/blood , Burning Mouth Syndrome/blood , Burning Mouth Syndrome/complications , Adult , Aged , Aged, 80 and over , Avitaminosis/complications , Blood Glucose/metabolism , Cholecalciferol/blood , Cholecalciferol/deficiency , Female , Folic Acid Deficiency/blood , Folic Acid Deficiency/complications , Humans , Male , Middle Aged , Retrospective Studies , Riboflavin Deficiency/blood , Riboflavin Deficiency/complications , Thiamine Deficiency/blood , Thiamine Deficiency/complications , Thyrotropin/blood , Thyrotropin/deficiency , Vitamin B 12 Deficiency/blood , Vitamin B 12 Deficiency/complications , Vitamin B 6 Deficiency/blood , Vitamin B 6 Deficiency/complications , Vitamin D Deficiency/blood , Vitamin D Deficiency/complications , Young Adult , Zinc/blood , Zinc/deficiency
10.
J Clin Endocrinol Metab ; 102(7): 2301-2309, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28368540

ABSTRACT

Introduction and Aim: Cumulative time-dependent excess mortality in hyperthyroid patients has been suggested. However, the effect of antithyroid treatment on mortality, especially in subclinical hyperthyroidism, remains unclarified. We investigated the association between hyperthyroidism and mortality in both treated and untreated hyperthyroid individuals. Patients and Methods: Register-based cohort study of 235,547 individuals who had at least one serum thyroid-stimulating hormone (TSH) measurement in the period 1995 to 2011 (7.3 years median follow-up). Hyperthyroidism was defined as at least two measurements of low serum TSH. Mortality rates for treated and untreated hyperthyroid subjects compared with euthyroid controls were calculated using multivariate Cox regression analyses, controlling for age, sex, and comorbidities. Cumulative periods of decreased serum TSH were analyzed as a time-dependent covariate. Results: Hazard ratio (HR) for mortality was increased in untreated [1.23; 95% confidence interval (CI), 1.12 to 1.37; P < 0.001], but not in treated, hyperthyroid patients. When including cumulative periods of TSH in the Cox regression analyses, HR for mortality per every 6 months of decreased TSH was 1.11 (95% CI, 1.09 to 1.13; P < 0.0001) in untreated hyperthyroid patients (n = 1137) and 1.13 (95% CI, 1.11 to 1.15; P < 0.0001) in treated patients (n = 1656). This corresponds to a 184% and 239% increase in mortality after 5 years of decreased TSH in untreated and treated hyperthyroidism, respectively. Conclusions: Mortality is increased in hyperthyroidism. Cumulative periods of decreased TSH increased mortality in both treated and untreated hyperthyroidism, implying that excess mortality may not be driven by lack of therapy, but rather inability to keep patients euthyroid. Meticulous follow-up during treatment to maintain biochemical euthyroidism may be warranted.


Subject(s)
Hyperthyroidism/blood , Hyperthyroidism/mortality , Thyrotropin/blood , Adult , Aged , Aged, 80 and over , Antithyroid Agents/therapeutic use , Denmark/epidemiology , Female , Humans , Hyperthyroidism/therapy , Male , Middle Aged , Registries , Thyroidectomy , Thyrotropin/deficiency , Thyroxine/blood , Triiodothyronine/blood
11.
J Med Genet ; 54(3): 166-175, 2017 03.
Article in English | MEDLINE | ID: mdl-27627987

ABSTRACT

BACKGROUND: Levels of serum thyroid-stimulating hormone (TSH) indicate thyroid function, because thyroid hormone negatively controls TSH release. Genetic variants in the vascular endothelial growth factor A (VEGFA) gene are associated with TSH levels. The aim of this study was to characterise the association of VEGFA variants with TSH in a Danish cohort and to identify and characterise functional variants. METHODS: We performed an association study of the VEGFA locus for circulating TSH levels in 8445 Danish individuals. Lead variants were tested for allele-specific effects in vitro using luciferase reporter and gel-shift assays. RESULTS: Four SNPs in VEGFA were associated with circulating TSH (rs9472138, rs881858, rs943080 and rs4711751). For rs881858, the presence of each G-allele was associated with a corresponding decrease in TSH levels of 2.3% (p=8.4×10-9) and an increase in circulating free T4 levels (p=0.0014). The SNP rs881858 is located in a binding site for CHOP (C/EBP homology protein) and c/EBPß (ccaat enhancer binding protein ß). Reporter-gene analysis showed increased basal enhancer activity of the rs881858 A-allele versus the G-allele (34.5±9.9% (average±SEM), p=0.0012), while co-expression of CHOP effectively suppressed the rs881858 A-allele activity. The A-allele showed stronger binding to CHOP in gel-shift assays. CONCLUSIONS: VEGF is an important angiogenic signal required for tissue expansion. We show that VEGFA variation giving allele-specific response to transcription factors with overlapping binding sites associate closely with circulating TSH levels. Because CHOP is induced by several types of intracellular stress, this indicates that cellular stress could be involved in the normal or pathophysiological response of the thyroid to TSH. TRIAL REGISTRATION NUMBER: NCT00289237, NCT00316667; Results.


Subject(s)
Myocardial Ischemia/genetics , Thyrotropin/blood , Transcription Factor CHOP/genetics , Vascular Endothelial Growth Factor A/genetics , Denmark , Enhancer Elements, Genetic , Female , Genetic Association Studies , Genetic Predisposition to Disease , Heterozygote , Humans , Male , Middle Aged , Myocardial Ischemia/blood , Myocardial Ischemia/pathology , Polymorphism, Single Nucleotide , Protein Binding/genetics , Thyroid Gland/metabolism , Thyroid Gland/pathology , Thyrotropin/deficiency , Thyrotropin/genetics
12.
Curr Treat Options Oncol ; 17(9): 50, 2016 09.
Article in English | MEDLINE | ID: mdl-27476159

ABSTRACT

OPINION STATEMENT: Survival rates of childhood cancer have improved markedly, and today more than 80 % of those diagnosed with a pediatric malignancy will become 5-year survivors. Nevertheless, survivors exposed to cranial radiotherapy (CRT) are at particularly high risk for long-term morbidity, such as endocrine insufficiencies, metabolic complications, and cardiovascular morbidity. Deficiencies of one or more anterior pituitary hormones have been described following therapeutic CRT for primary brain tumors, nasopharyngeal tumors, and following prophylactic CRT for childhood acute lymphoblastic leukemia (ALL). Studies have consistently shown a strong correlation between the total radiation dose and the development of pituitary deficits. Further, age at treatment and also time since treatment has strong implications on pituitary hormone deficiencies. There is evidence that the hypothalamus is more radiosensitive than the pituitary and is damaged by lower doses of CRT. With doses of CRT <50 Gy, the primary site of radiation damage is the hypothalamus and this usually causes isolated GH deficiency (GHD). Higher doses (>50 Gy) may produce direct anterior pituitary damage, which contributes to multiple pituitary deficiencies. The large group of ALL survivors treated with CRT in the 70-80-ties has now reached adulthood, and these survivors were treated mainly with 24 Gy, and the vast majority of these patients suffer from GHD. Further, after long-term follow-up, insufficiencies in prolactin (PRL) and thyroid stimulating hormone (TSH) have also been reported and a proportion of these patients were also adrenocoticotrophic hormone (ACTH) deficient. CRT to the hypothalamus causes neuroendocrine dysfunction, which means that the choice of GH test is crucial for the diagnosis of GHD.


Subject(s)
Brain Neoplasms/secondary , Cranial Irradiation/adverse effects , Hypothalamus/radiation effects , Pituitary Gland/radiation effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Adrenocorticotropic Hormone/blood , Adrenocorticotropic Hormone/deficiency , Brain Neoplasms/complications , Brain Neoplasms/radiotherapy , Child , Child, Preschool , Gonadotropins/blood , Gonadotropins/deficiency , Human Growth Hormone/blood , Human Growth Hormone/deficiency , Humans , Hypothalamus/metabolism , Pituitary Gland/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Precursor Cell Lymphoblastic Leukemia-Lymphoma/radiotherapy , Prolactin/blood , Prolactin/deficiency , ROC Curve , Thyrotropin/blood , Thyrotropin/deficiency , Time Factors , Treatment Outcome
13.
Lancet ; 388(10058): 2403-2415, 2016 Nov 12.
Article in English | MEDLINE | ID: mdl-27041067

ABSTRACT

Hypopituitarism refers to deficiency of one or more hormones produced by the anterior pituitary or released from the posterior pituitary. Hypopituitarism is associated with excess mortality, a key risk factor being cortisol deficiency due to adrenocorticotropic hormone (ACTH) deficiency. Onset can be acute or insidious, and the most common cause in adulthood is a pituitary adenoma, or treatment with pituitary surgery or radiotherapy. Hypopituitarism is diagnosed based on baseline blood sampling for thyroid stimulating hormone, gonadotropin, and prolactin deficiencies, whereas for ACTH, growth hormone, and antidiuretic hormone deficiency dynamic stimulation tests are usually needed. Repeated pituitary function assessment at regular intervals is needed for diagnosis of the predictable but slowly evolving forms of hypopituitarism. Replacement treatment exists in the form of thyroxine, hydrocortisone, sex steroids, growth hormone, and desmopressin. If onset is acute, cortisol deficiency should be replaced first. Modifications in replacement treatment are needed during the transition from paediatric to adult endocrine care, and during pregnancy.


Subject(s)
Adenoma/therapy , Hormone Replacement Therapy/methods , Hypophysectomy/adverse effects , Hypopituitarism , Pituitary Gland/metabolism , Pituitary Hormones, Anterior/administration & dosage , Pituitary Hormones, Anterior/deficiency , Pituitary Irradiation/adverse effects , Pituitary Neoplasms/therapy , Acute Disease , Adenoma/blood , Adenoma/radiotherapy , Adenoma/surgery , Adrenocorticotropic Hormone/administration & dosage , Adrenocorticotropic Hormone/deficiency , Chronic Disease , Deamino Arginine Vasopressin/administration & dosage , Gonadal Steroid Hormones/administration & dosage , Gonadal Steroid Hormones/deficiency , Gonadotropins, Pituitary/administration & dosage , Gonadotropins, Pituitary/deficiency , Human Growth Hormone/administration & dosage , Human Growth Hormone/deficiency , Humans , Hydrocortisone/administration & dosage , Hydrocortisone/deficiency , Hypopituitarism/blood , Hypopituitarism/diagnosis , Hypopituitarism/drug therapy , Hypopituitarism/etiology , Pituitary Neoplasms/blood , Pituitary Neoplasms/radiotherapy , Pituitary Neoplasms/surgery , Prolactin/administration & dosage , Prolactin/deficiency , Radiotherapy/adverse effects , Thyrotropin/administration & dosage , Thyrotropin/deficiency , Thyroxine/administration & dosage , Thyroxine/deficiency , Vasopressins/administration & dosage , Vasopressins/deficiency
14.
Pituitary ; 19(4): 399-406, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27083162

ABSTRACT

PURPOSE: To evaluate the utility of Insulin-like growth factor I (IGF-I) standard deviation score (SDS) as a surrogate marker of severity of hypopituitarism in adults with pituitary pathology. METHODS: We performed a retrospective data analysis, including 269 consecutive patients with pituitary disease attending a tertiary endocrine clinic in 1990-2015. The medical files were reviewed for the complete pituitary hormone profile, including IGF-I, and clinical data. Age-adjusted assay reference ranges of IGF-I were used to calculate IGF-I SDS for each patient. The main outcome measures were positive and negative predictive values of low and high IGF-I SDS, respectively, for the various pituitary hormone deficiencies. RESULTS: IGF-I SDS correlated negatively with the number of altered pituitary axes (p < 0.001). Gonadotropin was affected in 76.6 % of cases, followed by thyrotropin (58.4 %), corticotropin (49.1 %), and prolactin (22.7 %). Positive and negative predictive values yielded a clear trend for the probability of low/high IGF-I SDS for all affected pituitary axes. Rates of diabetes insipidus correlated with IGF-I SDS values both for the full study population, and specifically for patients with non-functioning pituitary adenomas. CONCLUSIONS: IGF-I SDS can be used to evaluate the somatotroph function, as a valid substitute to absolute IGF-I levels. Moreover, IGF-I SDS predicted the extent of hypopituitarism in adults with pituitary disease, and thus can serve as a marker of hypopituitarism severity.


Subject(s)
Adenoma/metabolism , Hypopituitarism/metabolism , Insulin-Like Growth Factor I/metabolism , Pituitary Neoplasms/metabolism , Adenoma/complications , Adenoma/therapy , Adrenocorticotropic Hormone/deficiency , Adrenocorticotropic Hormone/metabolism , Adult , Aged , Cranial Irradiation , Female , Follicle Stimulating Hormone/deficiency , Follicle Stimulating Hormone/metabolism , Gonadotropins, Pituitary/deficiency , Gonadotropins, Pituitary/metabolism , Humans , Hypopituitarism/etiology , Luteinizing Hormone/deficiency , Luteinizing Hormone/metabolism , Male , Middle Aged , Pituitary Diseases/complications , Pituitary Diseases/metabolism , Pituitary Gland/surgery , Pituitary Neoplasms/complications , Pituitary Neoplasms/therapy , Prolactin/deficiency , Prolactin/metabolism , Retrospective Studies , Severity of Illness Index , Testosterone/metabolism , Thyrotropin/deficiency , Thyrotropin/metabolism , Thyroxine/metabolism
15.
Neurobiol Aging ; 40: 22-40, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26973101

ABSTRACT

APP/PS1 double transgenic mice expressing human mutant amyloid precursor protein (APP) and presenilin-1 (PS1) demonstrate robust brain amyloid beta (Aß) peptide containing plaque deposition, increased markers of oxidative stress, behavioral dysfunction, and proinflammatory gliosis. On the other hand, lack of growth hormone, prolactin, and thyroid-stimulating hormone due to a recessive mutation in the Prop 1 gene (Prop1df) in Ames dwarf mice results in a phenotype characterized by potentiated antioxidant mechanisms, improved learning and memory, and significantly increased longevity in homozygous mice. Based on this, we hypothesized that a similar hormone deficiency might attenuate disease changes in the brains of APP/PS1 mice. To test this idea, APP/PS1 mice were crossed to the Ames dwarf mouse line. APP/PS1, wild-type, df/+, df/df, df/+/APP/PS1, and df/df/APP/PS1 mice were compared at 6 months of age through behavioral testing and assessing amyloid burden, reactive gliosis, and brain cytokine levels. df/df mice demonstrated lower brain growth hormone and insulin-like growth factor 1 concentrations. This correlated with decreased astrogliosis and microgliosis in the df/df/APP/PS1 mice and, surprisingly, reduced Aß plaque deposition and Aß 1-40 and Aß 1-42 concentrations. The df/df/APP/PS1 mice also demonstrated significantly elevated brain levels of multiple cytokines in spite of the attenuated gliosis. These data indicate that the df/df/APP/PS1 line is a unique resource in which to study aging and resistance to disease and suggest that the affected pituitary hormones may have a role in regulating disease progression.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Brain/metabolism , Growth Hormone/deficiency , Homeodomain Proteins/genetics , Mutation , Phenotype , Presenilin-1/genetics , Presenilin-1/metabolism , Prolactin/deficiency , Thyrotropin/deficiency , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Animals , Brain/pathology , Cells, Cultured , Cytokines/metabolism , Gene Expression , Gliosis , Insulin-Like Growth Factor I/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Plaque, Amyloid/metabolism
16.
Eur J Endocrinol ; 174(5): 669-79, 2016 May.
Article in English | MEDLINE | ID: mdl-26888628

ABSTRACT

OBJECTIVE: To determine characteristics of children initially diagnosed with isolated growth hormone deficiency (IGHD) of organic aetiology, who later developed multiple pituitary hormone deficiencies (MPHD). DESIGN: Data were analysed for 716 growth hormone-treated children with organic IGHD, who were growth hormone-naïve at baseline in the multinational, observational Genetics and Neuroendocrinology of Short Stature International Study. METHODS: Development of MPHD was ascertained from investigator-provided diagnoses, adverse events and concomitant medications. Analyses were performed for all patients and separately for those who developed MPHD within 4.5 years or had >3.5 years follow-up and continued to have IGHD (4-year cohort). RESULTS: MPHD developed in 71/716 (9.9%) children overall, and in 60/290 (20.7%) in the 4-year cohort. The most frequent additional deficiencies were thyroid-stimulating hormone (47 patients) and gonadotropins (23 patients). Compared with those who remained with IGHD, children who developed MPHD had more severe GHD at study entry, significantly lower baseline insulin-like growth factor1, peak stimulated growth hormone, and more frequent diagnosis of intracranial tumour or mutation of gene(s) controlling hypothalamic-pituitary development and/or function. Multivariate logistic regression analyses identified female gender, longer follow-up, higher baseline age and lower peak stimulated growth hormone as predictors of MPHD development. CONCLUSIONS: MPHD is more likely to develop in patients with severe organic IGHD, especially those with history of intracranial tumour or mutation of gene(s) controlling hypothalamic-pituitary development and/or function. Older baseline age, female gender and longer follow-up duration were also associated with higher incidence of MPHD. Long-term monitoring of pituitary function is recommended, irrespective of the aetiology of GHD.


Subject(s)
Disease Progression , Gonadotropins/deficiency , Human Growth Hormone/deficiency , Hypopituitarism/epidemiology , Pituitary Hormones/deficiency , Adolescent , Age Factors , Child , Congenital Hypothyroidism/epidemiology , Female , Follow-Up Studies , Humans , Hypothyroidism , Male , Sex Factors , Thyrotropin/deficiency
17.
J Clin Endocrinol Metab ; 101(1): 12-5, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26529631

ABSTRACT

BACKGROUND: Pituitary stalk interruption syndrome (PSIS) and holoprosencephaly (HPE) are congenital midline defects. Rare mutations in the sonic hedgehog (SHH) signaling gene CDON have recently been reported in patients with HPE. OBJECTIVE: To report a unique case of PSIS with a maternally inherited nonsense mutation in the SHH signaling protein CDON. METHOD: We performed exome sequencing on a case of PSIS. Control databases (1000 Genomes, dbSNP, Exome Variant Server, ExAC Browser) and an ancestry-matched control panel were screened upon identification of CDON mutation. RESULTS: We identified a novel heterozygous nonsense mutation (c.2764T>C, Glu922Ter) in a case of PSIS without HPE who presented with neonatal hypoglycemia and cholestasis associated with GH, TSH, and ACTH deficiencies. This mutation was absent in all control databases and from 400 healthy ancestry-matched control subjects. The mutation was inherited from the patient's mother, who was operated on in childhood for strabismus. The absence of this variant in control samples suggests that it is likely to be responsible for the phenotype. CONCLUSION: We report for the first time a mutation in the CDON gene associated with PSIS.


Subject(s)
Cell Adhesion Molecules/genetics , Pituitary Diseases/genetics , Pituitary Diseases/pathology , Pituitary Gland/pathology , Tumor Suppressor Proteins/genetics , Adrenocorticotropic Hormone/deficiency , Codon, Nonsense/genetics , Databases, Genetic , Exons/genetics , Holoprosencephaly/genetics , Hormone Replacement Therapy , Human Growth Hormone/deficiency , Humans , Infant, Newborn , Male , Syndrome , Thyrotropin/deficiency
18.
Minerva Pediatr ; 68(2): 152-4, 2016 Apr.
Article in English | MEDLINE | ID: mdl-24825480

ABSTRACT

In a short girl with celiac disease and Hashimoto's thyroiditis (HT), suspicion of an associated pituitary lesion was suggested by the finding of a thyroid function pattern that was not compatible with HT-related hypothyroidism (low FT4 with normal TSH). This case report reinforces the view that the finding of a normal TSH in presence of a low FT4 should always alert pediatricians and raise suspicion of central hypothyroidism, even when a primary thyroid disease has been already identified. In this case TSH deficiency played a critical role in disclosing diagnosis of craniopharyngioma (CP). Therefore, the subsequent work-up was directed towards investigating pituitary function and morphology. Endocrinological investigations evidenced a picture of TSH and other pituitary hormone deficiency, whereas magnetic resonance imaging revealed an intrasellar CP. Therefore, in this case TSH deficiency played a key-role in disclosing CP diagnosis.


Subject(s)
Craniopharyngioma/diagnostic imaging , Hashimoto Disease/diagnosis , Pituitary Neoplasms/diagnostic imaging , Thyrotropin/deficiency , Body Height , Celiac Disease/complications , Child , Craniopharyngioma/pathology , Female , Follow-Up Studies , Humans , Magnetic Resonance Imaging , Pituitary Neoplasms/pathology
19.
Horm Res Paediatr ; 84(5): 349-54, 2015.
Article in English | MEDLINE | ID: mdl-26302767

ABSTRACT

BACKGROUND: Recent reports have indicated that loss-of-function mutations in the immunoglobulin superfamily member 1 gene (IGSF1, OMIM 300888) cause congenital central hypothyroidism with macroorchidism. METHODS: We conducted a next-generation sequencing-based comprehensive mutation screening for pituitary hormone deficiencies to elucidate molecular mechanisms other than anatomical abnormalities of the pituitary that might be responsible for multiple anterior hormone deficiency in a male patient who originally visited our institute complaining of short stature. He was born large for gestational age (4,370 g, +3.0 SD) after an obstructed labour. Endocrinological evaluation revealed growth hormone and thyroid-stimulating hormone deficiency. Magnetic resonance imaging showed a discontinuity of the pituitary stalk with an ectopic posterior lobe and a hypoplastic anterior lobe, likely explaining multiple anterior pituitary hormone deficiency. RESULT: We identified a novel hemizygous IGSF1 mutation (c.1137_1138delCA, p.Asn380Glnfs*6) in the patient. In reviewing the literature, we noticed that all reported Japanese male IGSF1 mutation carriers were born larger than mean standards for gestational age (mean birth weight SD score of +2.0, 95% confidence interval 1.0-3.0). CONCLUSION: This case suggests that more attention should be paid to intrauterine growth and birth history when patients are suspected of having an IGSF1 mutation.


Subject(s)
Human Growth Hormone/deficiency , Immunoglobulins/genetics , Membrane Proteins/genetics , Thyrotropin/deficiency , Adolescent , Birth Weight/genetics , Congenital Hypothyroidism/genetics , Female , Fetus/pathology , Frameshift Mutation/genetics , Humans , Hypothalamo-Hypophyseal System/pathology , Hypothyroidism , Magnetic Resonance Imaging , Male , Obstetric Labor Complications , Pituitary Gland/pathology , Pregnancy
20.
PLoS One ; 10(5): e0126648, 2015.
Article in English | MEDLINE | ID: mdl-25955177

ABSTRACT

LHX4 is a LIM homeodomain transcription factor involved in the early steps of pituitary ontogenesis. To date, 8 heterozygous LHX4 mutations have been reported as responsible of combined pituitary hormone deficiency (CPHD) in Humans. We identified 4 new LHX4 heterozygous allelic variants in patients with congenital hypopituitarism: W204X, delK242, N271S and Q346R. Our objective was to determine the role of LHX4 variants in patients' phenotypes. Heterologous HEK293T cells were transfected with plasmids encoding for wild-type or mutant LHX4. Protein expression was analysed by Western Blot, and DNA binding by electro-mobility shift assay experiments. Target promoters of LHX4 were cotransfected with wild type or mutant LHX4 to test the transactivating abilities of each variant. Our results show that the W204X mutation was associated with early GH and TSH deficiencies and later onset ACTH deficiency. It led to a truncated protein unable to bind to alpha-Gsu promoter binding consensus sequence. W204X was not able to activate target promoters in vitro. Cotransfection experiments did not favour a dominant negative effect. In contrast, all other mutants were able to bind the promoters and led to an activation similar as that observed with wild type LHX4, suggesting that they were likely polymorphisms. To conclude, our study underlines the need for functional in vitro studies to ascertain the role of rare allelic variants of LHX4 in disease phenotypes. It supports the causative role of the W204X mutation in CPHD and adds up childhood onset ACTH deficiency to the clinical spectrum of the various phenotypes related to LHX4 mutations.


Subject(s)
Hypopituitarism/genetics , LIM-Homeodomain Proteins/genetics , Mutation , Transcription Factors/genetics , Adrenocorticotropic Hormone/deficiency , Alleles , Child , Child, Preschool , Female , Growth Hormone/deficiency , HEK293 Cells , Heterozygote , Humans , Hypopituitarism/complications , Infant , Male , Thyrotropin/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...