Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
2.
J Clin Endocrinol Metab ; 107(9): e3654-e3660, 2022 08 18.
Article in English | MEDLINE | ID: mdl-35775692

ABSTRACT

CONTEXT: The syndrome of adrenal insufficiency, obesity, and red hair is a rare autosomal recessive disorder. The majority of disease-causing variants associated with the syndrome are located in the coding region of the POMC gene. OBJECTIVE: This work describes 7 unrelated patients who shared a novel homozygous mutation in the 5'-untranslated region (UTR) of the POMC gene and functionally characterize this novel variant. METHODS: Whole-exome sequencing (WES) with autozygosity mapping, Sanger sequencing, model expression system studies, and RNA sequencing were used for identification of the disease-causing variant and its subsequent functional characterization. Seven unrelated patients of the Perm Tatar ethnic group presented with hypoglycemia and excessive weight gain, low plasma adrenocorticotropin, and cortisol. Five of 7 children had red hair; 6 of 7 patients also showed signs of bronchial obstruction. RESULTS: WES showed shared autozygosity regions overlapping the POMC gene. Sanger sequencing of the POMC 5'-UTR detected a homozygous variant chr2:25391366C > T (hg19) at the splice donor site of intron 1. As demonstrated by the model expression system, the variant led to a significant decrease in the POMC messenger RNA level. Analyses of the patients' haplotypes were suggestive of the founder effect. We estimate that the mutation must have occurred at least 4.27 generations ago (95% CI, 0.86-7.67). CONCLUSION: This report presents a new molecular mechanism of POMC deficiency and contributes to the information on phenotypic variability in patients with this disorder.


Subject(s)
Adrenal Insufficiency , Pro-Opiomelanocortin , 5' Untranslated Regions , Adrenal Insufficiency/diagnosis , Child , Founder Effect , Humans , Mutation , Obesity/complications , Pro-Opiomelanocortin/deficiency , Pro-Opiomelanocortin/genetics , RNA Splicing , RNA, Messenger/genetics
3.
Mol Metab ; 53: 101312, 2021 11.
Article in English | MEDLINE | ID: mdl-34329773

ABSTRACT

OBJECTIVE: Proopiomelanocortin (POMC) neurons of the hypothalamic arcuate nucleus are essential regulators of energy balance. Selective loss of POMC production in these cells results in extreme obesity and metabolic comorbidities. Neurogenesis occurs in the adult hypothalamus, but it remains uncertain whether functional POMC neurons emerge in physiologically significant numbers during adulthood. Here, we tested whether Rax-expressing precursors generate POMC neurons in adult mice and rescue the metabolic phenotype caused by congenital hypothalamic POMC deficiency. METHODS: Initially, we identified hypothalamic Rax-expressing cell types using wild-type and Rax-CreERT2:Ai34D mice. Then we generated compound Rax-CreERT2:ArcPomcloxTB/loxTB mice in which endogenous hypothalamic Pomc expression is silenced, but can be restored by tamoxifen administration selectively in neurons derived from Rax+ progenitors. The number of POMC neurons generated by Rax+ progenitors in adult mice and their axonal projections was determined. The metabolic effects of these neurons were assessed by measuring food intake, bodyweight, and body composition, along with glucose and insulin levels. RESULTS: We found that Rax is expressed by tanycytes and a previously unrecognized cell type in the hypothalamic parenchyma of adult mice. Rax+ progenitors generated ~10% of the normal adult hypothalamic POMC neuron population within two weeks of tamoxifen treatment. The same rate and steady state of POMC neurogenesis persisted from young adult to aged mice. These new POMC neurons established terminal projections to brain regions that were involved in energy homeostasis. Mice with Rax+ progenitor-derived POMC neurons had reduced body fat mass, improved glucose tolerance, increased insulin sensitivity, and decreased bodyweight in proportion to the number of new POMC neurons. CONCLUSIONS: These data demonstrate that Rax+ progenitors generate POMC neurons in sufficient numbers during adulthood to mitigate the metabolic abnormalities of hypothalamic POMC-deficient mice. The findings suggest that adult hypothalamic neurogenesis is a robust phenomenon in mice that can significantly impact energy homeostasis.


Subject(s)
Adrenal Insufficiency/metabolism , Eye Proteins/metabolism , Homeodomain Proteins/metabolism , Hypothalamus/metabolism , Neurons/metabolism , Obesity/metabolism , Pro-Opiomelanocortin/deficiency , Pro-Opiomelanocortin/metabolism , Transcription Factors/metabolism , Animals , Eye Proteins/genetics , Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Transcription Factors/genetics
4.
Front Endocrinol (Lausanne) ; 12: 689387, 2021.
Article in English | MEDLINE | ID: mdl-34177811

ABSTRACT

Proopiomelanocortin (POMC) deficiency is an extremely rare inherited autosomal recessive disorder characterized by severe obesity, adrenal insufficiency, skin hypopigmentation, and red hair. It is caused by pathogenic variants in the POMC gene that codes the proopiomelanocortin polypeptide which is cleaved to several peptides; the most notable ones are adrenocorticotropic hormone (ACTH), alpha- and beta-melanocyte-stimulating hormones (α-MSH and ß-MSH); the latter two are crucial in melanogenesis and the energy balance by regulating feeding behavior and energy homeostasis through melanocortin receptor 4 (MC4R). The lack of its regulation leads to polyphagia and early onset severe obesity. A novel MC4R agonist, setmelanotide, has shown promising results regarding weight loss in patients with POMC deficiency. A systematic review on previously published clinical and genetic characteristics of patients with POMC deficiency and additional data obtained from two unrelated patients in our care was performed. A 25-year-old male patient, partly previously reported, was remarkable for childhood developed type 1 diabetes (T1D), transient growth hormone deficiency, and delayed puberty. The second case is a girl with an unusual presentation with central hypothyroidism and normal pigmentation of skin and hair. Of all evaluated cases, only 50% of patients had characteristic red hair, fair skin, and eye phenotype. Central hypothyroidism was reported in 36% of patients; furthermore, scarce adolescent data indicate possible growth axis dysbalance and central hypogonadism. T1D was unexpectedly prevalent in POMC deficiency, reported in 14% of patients, which could be an underestimation. POMC deficiency reveals to be a syndrome with several endocrinological abnormalities, some of which may become apparent with time. Apart from timely diagnosis, careful clinical follow-up of patients through childhood and adolescence for possible additional disease manifestations is warranted.


Subject(s)
Adrenal Insufficiency/genetics , Diabetes Mellitus, Type 1/genetics , Obesity/genetics , Pro-Opiomelanocortin/deficiency , Pro-Opiomelanocortin/genetics , Skin Pigmentation/genetics , Adult , Child, Preschool , Female , Humans , Male
5.
J Paediatr Child Health ; 57(4): 484-490, 2021 04.
Article in English | MEDLINE | ID: mdl-33666293

ABSTRACT

Proopiomelanocortin (POMC) deficiency is a rare monogenic disorder characterised by adrenocorticotropic hormone (ACTH) deficiency, red hair and hyperphagic obesity. Two unrelated cases presented with hypoglycaemia due to isolated ACTH deficiency in the neonatal period. POMC deficiency was suspected at age 2 years (c.133-2A>C) and at age 9 months (c.64del) due to infantile hyperphagic obesity. Neither patient had a convincing red hair phenotype at the time of diagnostic suspicion, illustrating the importance of suspecting POMC deficiency in isolated ACTH deficiency. Both patients have normal psychomotor development, whereas the only other reported case of c.64del had significant delay. This suggests, if ACTH deficiency is treated early in the neonatal period, that psychomotor retardation is not a part of the phenotype. We review 24 reported cases of POMC deficiency published to date. Although there is no current specific treatment for obesity in POMC deficiency, we anticipate that setmelanotide may be a useful future treatment option.


Subject(s)
Adrenal Insufficiency , Pro-Opiomelanocortin , Adrenal Insufficiency/diagnosis , Adrenocorticotropic Hormone , Child, Preschool , Humans , Infant , Male , Obesity , Pro-Opiomelanocortin/deficiency , Pro-Opiomelanocortin/genetics
6.
Nat Commun ; 12(1): 426, 2021 01 18.
Article in English | MEDLINE | ID: mdl-33462216

ABSTRACT

Painful neuropathy is a frequent complication in diabetes. Proopiomelanocortin (POMC) is an endogenous opioid precursor peptide, which plays a protective role against pain. Here, we report dysfunctional POMC-mediated antinociception in sensory neurons in diabetes. In streptozotocin-induced diabetic mice the Pomc promoter is repressed due to increased binding of NF-kB p50 subunit, leading to a loss in basal POMC level in peripheral nerves. Decreased POMC levels are also observed in peripheral nervous system tissue from diabetic patients. The antinociceptive pathway mediated by POMC is further impaired due to lysosomal degradation of µ-opioid receptor (MOR). Importantly, the neuropathic phenotype of the diabetic mice is rescued upon viral overexpression of POMC and MOR in the sensory ganglia. This study identifies an antinociceptive mechanism in the sensory ganglia that paves a way for a potential therapy for diabetic neuropathic pain.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetic Neuropathies/pathology , Nociception/physiology , Pro-Opiomelanocortin/deficiency , Sensory Receptor Cells/pathology , Aged , Aged, 80 and over , Animals , Diabetes Mellitus, Experimental/chemically induced , Diabetic Neuropathies/etiology , Female , Ganglia, Spinal/cytology , Ganglia, Spinal/pathology , Humans , Lysosomes , Male , Mice , Mice, Knockout , Pro-Opiomelanocortin/genetics , Proteolysis , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Streptozocin/toxicity
7.
Lancet Diabetes Endocrinol ; 8(12): 960-970, 2020 12.
Article in English | MEDLINE | ID: mdl-33137293

ABSTRACT

BACKGROUND: The melanocortin 4 receptor (MC4R), a component of the leptin-melanocortin pathway, plays a part in bodyweight regulation. Severe early-onset obesity can be caused by biallelic variants in genes that affect the MC4R pathway. We report the results from trials of the MC4R agonist setmelanotide in individuals with severe obesity due to either pro-opiomelanocortin (POMC) deficiency obesity or leptin receptor (LEPR) deficiency obesity. METHODS: These single-arm, open-label, multicentre, phase 3 trials were done in ten hospitals across Canada, the USA, Belgium, France, Germany, the Netherlands, and the UK. Participants aged 6 years or older with POMC or LEPR deficiency obesity received open-label setmelanotide for 12 weeks. Participants with at least 5 kg weight loss (or ≥5% if weighing <100 kg at baseline) entered an 8-week placebo-controlled withdrawal sequence (including 4 weeks each of blinded setmelanotide and placebo treatment) followed by 32 additional weeks of open-label treatment. The primary endpoint, which was assessed in participants who received at least one dose of study medication and had a baseline assessment (full analysis set), was the proportion of participants with at least 10% weight loss compared with baseline at approximately 1 year. A key secondary endpoint was mean percentage change in the most hunger score of the 11-point Likert-type scale at approximately 1 year on the therapeutic dose, which was assessed in a subset of participants aged 12 years or older in the full analysis set who demonstrated at least 5 kg weight loss (or ≥5% in paediatric participants if baseline bodyweight was <100 kg) over the 12-week open-label treatment phase and subsequently proceeded into the placebo-controlled withdrawal sequence, regardless of later disposition. These studies are registered with ClinicalTrials.gov, NCT02896192 and NCT03287960. FINDINGS: Between Feb 14, 2017, and Sept 7, 2018, ten participants were enrolled in the POMC trial and 11 participants were enrolled in the LEPR trial, and included in the full analysis and safety sets. Eight (80%) participants in the POMC trial and five (45%) participants in the LEPR trial achieved at least 10% weight loss at approximately 1 year. The mean percentage change in the most hunger score was -27·1% (n=7; 90% CI -40·6 to -15·0; p=0·0005) in the POMC trial and -43·7% (n=7; -54·8 to -29·1; p<0·0001) in the LEPR trial. The most common adverse events were injection site reaction and hyperpigmentation, which were reported in all ten participants in the POMC trial; nausea was reported in five participants and vomiting in three participants. In the LEPR trial, the most commonly reported treatment-related adverse events were injection site reaction in all 11 participants, skin disorders in five participants, and nausea in four participants. No serious treatment-related adverse events occurred in both trials. INTERPRETATION: Our results support setmelanotide for the treatment of obesity and hyperphagia caused by POMC or LEPR deficiency. FUNDING: Rhythm Pharmaceuticals.


Subject(s)
Adrenal Insufficiency/complications , Anti-Obesity Agents/therapeutic use , Obesity/drug therapy , Pro-Opiomelanocortin/deficiency , Receptor, Melanocortin, Type 4/agonists , Receptors, Leptin/deficiency , alpha-MSH/analogs & derivatives , Adolescent , Adult , Child , Double-Blind Method , Female , Follow-Up Studies , Humans , Male , Obesity/complications , Obesity/etiology , Obesity/pathology , Prognosis , Young Adult , alpha-MSH/therapeutic use
8.
Endocrinology ; 161(8)2020 08 01.
Article in English | MEDLINE | ID: mdl-32591776

ABSTRACT

TBR-760 (formerly BIM-23A760) is a chimeric dopamine (DA)-somatostatin (SST) compound with potent agonist activity at both DA type 2 (D2R) and SST type 2 (SSTR2) receptors. Studies have shown that chimeric DA-SST compounds are more efficacious than individual DA and/or SST analogues, either alone or combined, in inhibiting secretion from primary cultures of human somatotroph and lactotroph tumor cells. Nonfunctioning pituitary adenomas (NFPAs) express both D2R and SSTR2 and, consequently, may respond to TBR-760. We used a mouse model with the pro-opiomelanocortin (POMC) gene knocked out that spontaneously develops aggressive NFPAs. Genomic microarray and DA and SST receptor messenger RNA expression analysis indicate that POMC KO mouse tumors and human NFPAs have similar expression profiles, despite arising from different cell lineages, establishing POMC KO mice as a model for study of NFPAs. Treatment with TBR-760 for 8 weeks resulted in nearly complete inhibition of established tumor growth, whereas tumors from vehicle-treated mice increased in size by 890 ± 0.7%. Comparing TBR-760 with its individual DA and SST components, TBR-760 arrested tumor growth. Treatment with equimolar or 10×-higher doses of the individual SST or DA agonists, either alone or in combination, had no significant effect. One exception was the lower dose of DA agonist that induced modest suppression of tumor growth. Only the chimeric compound TBR-760 arrested tumor growth in this mouse model of NFPA. Further, significant tumor shrinkage was observed in 20% of the mice treated with TBR-760. These results support the development of TBR-760 as a therapy for patients with NFPA.


Subject(s)
Adenoma/drug therapy , Adenoma/pathology , Cell Proliferation/drug effects , Dopamine/analogs & derivatives , Pituitary Neoplasms/drug therapy , Pituitary Neoplasms/pathology , Somatostatin/analogs & derivatives , Adenoma/genetics , Animals , Cell Proliferation/genetics , Disease Models, Animal , Dopamine/pharmacology , Dopamine/therapeutic use , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Mice , Mice, Knockout , Microarray Analysis , Neoplasm Invasiveness , Pituitary Neoplasms/genetics , Pro-Opiomelanocortin/deficiency , Pro-Opiomelanocortin/genetics , Somatostatin/pharmacology , Somatostatin/therapeutic use
9.
Mol Metab ; 35: 100957, 2020 05.
Article in English | MEDLINE | ID: mdl-32244188

ABSTRACT

OBJECTIVE: The steep rise in the prevalence of obesity and its related metabolic syndrome have become a major worldwide health concerns. Melanocortin peptides from hypothalamic arcuate nucleus (Arc) POMC neurons induce satiety to limit food intake. Consequently, Arc Pomc-deficient mice (ArcPomc-/-) exhibit hyperphagia and obesity. Previous studies demonstrated that the circulating levels of adiponectin, a protein abundantly produced and secreted by fat cells, negatively correlate with obesity in both rodents and humans. However, we found that ArcPomc-/- mice have increased circulating adiponectin levels despite obesity. Therefore, we investigated the physiological function and underlying mechanisms of hypothalamic POMC in regulating systemic adiponectin levels. METHODS: Circulating adiponectin was measured in obese ArcPomc-/- mice at ages 4-52 weeks. To determine whether increased adiponectin was a direct result of ArcPomc deficiency or a secondary effect of obesity, we examined plasma adiponectin levels in calorie-restricted mice with or without a history of obesity and in ArcPomc-/- mice before and after genetic restoration of Pomc expression in the hypothalamus. To delineate the mechanisms causing increased adiponectin in ArcPomc-/- mice, we determined sympathetic outflow to adipose tissue by assessing epinephrine, norepinephrine, and tyrosine hydroxylase protein levels and measured the circulating adiponectin in the mice after acute norepinephrine or propranolol treatments. In addition, adiponectin mRNA and protein levels were measured in discrete adipose tissue depots to ascertain which fat depots contributed the most to the high level of adiponectin in the ArcPomc-/- mice. Finally, we generated compound Adiopoq-/-:ArcPomc-/- mice and compared their growth, body composition, and glucose homeostasis to the individual knockout mouse strains and their wild-type controls. RESULTS: Obese ArcPomc-/- female mice had unexpectedly increased plasma adiponectin compared to wild-type siblings at all ages greater than 8 weeks. Despite chronic calorie restriction to achieve normal body weights, higher adiponectin levels persisted in the ArcPomc-/- female mice. Genetic restoration of Pomc expression in the Arc or acute treatment of the ArcPomc-/- female mice with melanotan II reduced adiponectin levels to control littermate values. The ArcPomc-/- mice had defective thermogenesis and decreased epinephrine, norepinephrine, and tyrosine hydroxylase protein levels in their fat pads, indicating reduced sympathetic outflow to adipose tissue. Injections of norepinephrine into the ArcPomc-/- female mice reduced circulating adiponectin levels, whereas injections of propranolol significantly increased adiponectin levels. Despite the beneficial effects of adiponectin on metabolism, the deletion of adiponectin alleles in the ArcPomc-/- mice did not exacerbate their metabolic abnormalities. CONCLUSION: In summary, to the best of our knowledge, this study provides the first evidence that despite obesity, the ArcPomc-/- mouse model has high circulating adiponectin levels, which demonstrated that increased fat mass is not necessarily correlated with hypoadiponectinemia. Our investigation also found a previously unknown physiological pathway connecting POMC neurons via the sympathetic nervous system to circulating adiponectin, thereby shedding light on the biological regulation of adiponectin.


Subject(s)
Adiponectin/blood , Arcuate Nucleus of Hypothalamus/metabolism , Neurons/metabolism , Obesity/blood , Pro-Opiomelanocortin/deficiency , Adiponectin/deficiency , Adiponectin/metabolism , Adipose Tissue/metabolism , Animals , Caloric Restriction , Disease Models, Animal , Female , Melanocortins/metabolism , Metabolism, Inborn Errors/metabolism , Mice , Mice, Knockout , Peptides, Cyclic/pharmacology , Pro-Opiomelanocortin/genetics , Signal Transduction/drug effects , Sympathetic Nervous System/metabolism , alpha-MSH/analogs & derivatives , alpha-MSH/pharmacology
10.
J Mol Cell Biol ; 12(4): 291-304, 2020 05 18.
Article in English | MEDLINE | ID: mdl-31237951

ABSTRACT

The endocrine regulatory roles of the hypothalamic-pituitary-adrenocortical axis on anxiety-like behavior and metabolic status have been found throughout animal taxa. However, the precise effects of the balancing adrenal corticosteroid biosynthesis under the influence of adrenocorticotrophic hormone (ACTH), a pro-opiomelanocortin (POMC)-derived peptide, on animal energy expenditure and somatic growth remain unknown. POMC has also been identified as one of the candidate loci for polycystic ovary syndrome, which features hyperandrogenism and some prevalence of obesity in patients. Here we show that zebrafish lacking functional POMCa exhibit similar phenotypes of stress response and body weight gain but not obesity as observed in mammalian models. In contrast with the impaired anorexigenic signaling cascade of melanocyte-stimulating hormones and leptin, which are responsible for their obesity-prone weight gain observed in various pomc mutant mammals, analyses with our pomca mutant series indicate that ACTH is the key regulator for the phenotype with enhanced somatic growth without obesity in pomca-deficient zebrafish. Hypocortisolism associated with hyperandrogenism has been observed in the pomca-deficient zebrafish, with enhanced activation of mammalian target of rapamycin complex 1; reutilization of amino acids and fatty acid ß-oxidation are observed in the muscle tissue of the pomca-deficient fish. After reducing hyperandrogenism by crossing our pomca mutant fish with a cyp17a1-deficient background, the phenotype of enhanced somatic growth in pomca-deficient fish was no longer observed. Thus, our work also demonstrated that the role of POMCa in stress response seems to be conserved in vertebrates, whereas its effect on adipostasis is unique to teleosts.


Subject(s)
Adiposity , Hyperandrogenism/metabolism , Pro-Opiomelanocortin/deficiency , Zebrafish/growth & development , Zebrafish/metabolism , Adrenocorticotropic Hormone/metabolism , Animals , Anxiety/metabolism , Base Sequence , Behavior, Animal , Cyclic AMP/metabolism , Darkness , Fatty Acids/metabolism , Hydrocortisone/metabolism , Larva/metabolism , Lipids/chemistry , Melanosomes/metabolism , Muscles/metabolism , Mutation/genetics , Obesity/genetics , Oxidation-Reduction , Oxygen Consumption , Pro-Opiomelanocortin/metabolism , Protein Biosynthesis , Receptor, Melanocortin, Type 2/metabolism , Signal Transduction , Testosterone/metabolism
11.
Mol Metab ; 20: 194-204, 2019 02.
Article in English | MEDLINE | ID: mdl-30503832

ABSTRACT

OBJECTIVE: Life-threatening hypoglycemia is a major limiting factor in the management of diabetes. While it is known that counterregulatory responses to hypoglycemia are impaired in diabetes, molecular mechanisms underlying the reduced responses remain unclear. Given the established roles of the hypothalamic proopiomelanocortin (POMC)/melanocortin 4 receptor (MC4R) circuit in regulating sympathetic nervous system (SNS) activity and the SNS in stimulating counterregulatory responses to hypoglycemia, we hypothesized that hypothalamic POMC as well as MC4R, a receptor for POMC derived melanocyte stimulating hormones, is required for normal hypoglycemia counterregulation. METHODS: To test the hypothesis, we induced hypoglycemia or glucopenia in separate cohorts of mice deficient in either POMC or MC4R in the arcuate nucleus (ARC) or the paraventricular nucleus of the hypothalamus (PVH), respectively, and measured their circulating counterregulatory hormones. In addition, we performed a hyperinsulinemic-hypoglycemic clamp study to further validate the function of MC4R in hypoglycemia counterregulation. We also measured Pomc and Mc4r mRNA levels in the ARC and PVH, respectively, in the streptozotocin-induced type 1 diabetes mouse model and non-obese diabetic (NOD) mice to delineate molecular mechanisms by which diabetes deteriorates the defense systems against hypoglycemia. Finally, we treated diabetic mice with the MC4R agonist MTII, administered stereotaxically into the PVH, to determine its potential for restoring the counterregulatory response to hypoglycemia in diabetes. RESULTS: Stimulation of epinephrine and glucagon release in response to hypoglycemia or glucopenia was diminished in both POMC- and MC4R-deficient mice, relative to their littermate controls. Similarly, the counterregulatory response was impaired in association with decreased hypothalamic Pomc and Mc4r expression in the diabetic mice, a phenotype that was not reversed by insulin treatment which normalized glycemia. In contrast, infusion of an MC4R agonist in the PVH restored the counterregulatory response in diabetic mice. CONCLUSION: In conclusion, hypothalamic Pomc as well as Mc4r, both of which are reduced in type 1 diabetic mice, are required for normal counterregulatory responses to hypoglycemia. Therefore, enhancing MC4R function may improve hypoglycemia counterregulation in diabetes.


Subject(s)
Hypoglycemia/metabolism , Hypothalamus/metabolism , Pro-Opiomelanocortin/metabolism , Receptor, Melanocortin, Type 4/metabolism , Animals , Epinephrine/metabolism , Glucagon/metabolism , Homeostasis , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Pro-Opiomelanocortin/deficiency , Pro-Opiomelanocortin/genetics , Receptor, Melanocortin, Type 4/deficiency , Receptor, Melanocortin, Type 4/genetics
12.
J Pediatr Endocrinol Metab ; 31(7): 815-819, 2018 Jul 26.
Article in English | MEDLINE | ID: mdl-29858905

ABSTRACT

Background Proopiomelanocortin (POMC) is a complex polypeptide that produces a variety of biologically active substances via cleavage in a tissue-specific manner [Challis BG, Millington GW. Proopiomelanocortin deficiency. GeneReviews® [Internet]. Seattle (WA): University of Washington, Seattle, 1993-2018], yielding several products including adrenocorticotrophic (ACTH) and melanocyte stimulating hormones (MSH). These peptides have roles in the regulation of food intake, energy homeostasis, adrenal steroidogenesis, melanocyte stimulation and immune modulation. Rare mutations in the POMC gene can lead to ACTH deficiency and thus isolated hypocortisolism. The first cases of POMC mutation were documented by Krude et al. in 1998 [Krude H, Biebermann H, Luck W, Horn R, Brabant G, et al. Severe early-onset obesity, adrenal insufficiency and red hair pigmentation caused by POMC mutations in humans. Nat Genet 1998;19:155-7]. Mutations in the POMC gene were linked with a clinical phenotype of adrenal insufficiency, red hair pigmentation, early onset and rapidly progressive obesity, early onset type 2 diabetes, hypothyroidism, hypogonadism and growth hormone deficiency. Case presentation We describe a prepubertal Hispanic boy with a novel homozygous POMC mutation with severe obesity, hypothyroidism, adrenal insufficiency and abnormal reddish hair pigmentation. The patient presented as a 2-year-old with exponential weight gain, abnormal thyroid labs and speech delay. Laboratory testing demonstrated central adrenal insufficiency and genetic testing confirmed a homozygous mutation (nucleotide change c.20_21ins25) in exon 3 of the POMC gene. Replacement therapy with thyroid hormone and hydrocortisone was coupled to a slight decrease in the rate of weight gain, although hyperphagia persisted. Parent-directed nutrition and activity education as well as attempts to restrict access to food resulted in a plateau of the body mass index (BMI). At 4 years of age, metformin treatment was initiated with the patient showing evolving signs of insulin resistance and failure of lifestyle/dietary intervention to adequately decrease the BMI. Over a 3-year metformin treatment span, the BMI decreased from 34.9 kg/m2 to 32.9 kg/m2. Conclusions We demonstrate a possible role for metformin in stemming progressive weight gain, thereby impacting the early onset obesity due to hyperphagia.


Subject(s)
Adrenal Insufficiency/drug therapy , Hypothyroidism/drug therapy , Metformin/therapeutic use , Mutation , Obesity, Morbid/drug therapy , Obesity/drug therapy , Pro-Opiomelanocortin/deficiency , Pro-Opiomelanocortin/genetics , Skin Pigmentation/drug effects , Adrenal Insufficiency/genetics , Adrenal Insufficiency/pathology , Adult , Body Mass Index , Child , Child, Preschool , Female , Hispanic or Latino , Homozygote , Humans , Hypoglycemic Agents/therapeutic use , Hypothyroidism/enzymology , Hypothyroidism/genetics , Male , Obesity/genetics , Obesity/pathology , Obesity, Morbid/enzymology , Obesity, Morbid/genetics , Pedigree , Phenotype , Prognosis
13.
J Clin Res Pediatr Endocrinol ; 10(1): 68-73, 2018 Mar 01.
Article in English | MEDLINE | ID: mdl-28739551

ABSTRACT

Proopiomelanocortin (POMC) deficiency is a rare monogenic disorder with early-onset obesity. Investigation of this entity have increased our insight into the important role of the leptin-melanocortin pathway in energy balance. Here, we present a patient with POMC deficiency due to a homozygous c.206delC mutation in the POMC gene. We discuss the pathogenesis of this condition with emphasis on the crosstalk between hypothalamic and peripheral signals in the development of obesity and the POMC-melanocortin 4 receptors system as a target for therapeutic intervention.


Subject(s)
Adrenal Insufficiency/diagnosis , Adrenal Insufficiency/metabolism , Obesity/diagnosis , Obesity/metabolism , Pro-Opiomelanocortin/deficiency , Adrenal Insufficiency/genetics , Child, Preschool , Female , Humans , Obesity/genetics , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism
14.
Mol Metab ; 6(10): 1274-1285, 2017 10.
Article in English | MEDLINE | ID: mdl-29031726

ABSTRACT

OBJECTIVE: Hypothalamic arcuate nucleus-specific pro-opiomelanocortin deficient (ArcPomc-/-) mice exhibit improved glucose tolerance despite massive obesity and insulin resistance. We demonstrated previously that their improved glucose tolerance is due to elevated glycosuria. However, the underlying mechanisms that link glucose reabsorption in the kidney with ArcPomc remain unclear. Given the function of the hypothalamic melanocortin system in controlling sympathetic outflow, we hypothesized that reduced renal sympathetic nerve activity (RSNA) in ArcPomc-/- mice could explain their elevated glycosuria and consequent enhanced glucose tolerance. METHODS: We measured RSNA by multifiber recording directly from the nerves innervating the kidneys in ArcPomc-/- mice. To further validate the function of RSNA in glucose reabsorption, we denervated the kidneys of WT and diabetic db/db mice before measuring their glucose tolerance and urine glucose levels. Moreover, we performed western blot and immunohistochemistry to determine kidney GLUT2 and SGLT2 levels in either ArcPomc-/- mice or the renal-denervated mice. RESULTS: Consistent with our hypothesis, we found that basal RSNA was decreased in ArcPomc-/- mice relative to their wild type (WT) littermates. Remarkably, both WT and db/db mice exhibited elevated glycosuria and improved glucose tolerance after renal denervation. The elevated glycosuria in obese ArcPomc-/-, WT and db/db mice was due to reduced renal GLUT2 levels in the proximal tubules. Overall, we show that renal-denervated WT and diabetic mice recapitulate the phenotype of improved glucose tolerance and elevated glycosuria associated with reduced renal GLUT2 levels observed in obese ArcPomc-/- mice. CONCLUSION: Hence, we conclude that ArcPomc is essential in maintaining basal RSNA and that elevated glycosuria is a possible mechanism to explain improved glucose tolerance after renal denervation in drug resistant hypertensive patients.


Subject(s)
Glycosuria/physiopathology , Hypothalamus/metabolism , Kidney/innervation , Pro-Opiomelanocortin/deficiency , Sympathetic Nervous System/physiology , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Diabetes Mellitus, Experimental , Female , Glucose/metabolism , Glucose Tolerance Test , Glucose Transporter Type 2/metabolism , Glycosuria/metabolism , Glycosuria/urine , Insulin/metabolism , Insulin Resistance/physiology , Kidney/metabolism , Male , Mice , Mice, Knockout , Mice, Obese , Obesity/metabolism , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism
15.
Mol Metab ; 6(10): 1321-1329, 2017 10.
Article in English | MEDLINE | ID: mdl-29031731

ABSTRACT

OBJECTIVE: Pro-opiomelanocortin (POMC)-derived peptides act on neurons expressing the Melanocortin 4 receptor (MC4R) to reduce body weight. Setmelanotide is a highly potent MC4R agonist that leads to weight loss in diet-induced obese animals and in obese individuals with complete POMC deficiency. While POMC deficiency is very rare, 1-5% of severely obese individuals harbor heterozygous mutations in MC4R. We sought to assess the efficacy of Setmelanotide in human MC4R deficiency. METHODS: We studied the effects of Setmelanotide on mutant MC4Rs in cells and the weight loss response to Setmelanotide administration in rodent studies and a human clinical trial. We annotated the functional status of 369 published MC4R variants. RESULTS: In cells, we showed that Setmelanotide is significantly more potent at MC4R than the endogenous ligand alpha-melanocyte stimulating hormone and can disproportionally rescue signaling by a subset of severely impaired MC4R mutants. Wild-type rodents appear more sensitive to Setmelanotide when compared to MC4R heterozygous deficient mice, while MC4R knockout mice fail to respond. In a 28-day Phase 1b clinical trial, Setmelanotide led to weight loss in obese MC4R variant carriers. Patients with POMC defects upstream of MC4R show significantly more weight loss with Setmelanotide than MC4R deficient patients or obese controls. CONCLUSIONS: Setmelanotide led to weight loss in obese people with MC4R deficiency; however, further studies are justified to establish whether Setmelanotide can elicit clinically meaningful weight loss in a subset of the MC4R deficient obese population.


Subject(s)
Receptor, Melanocortin, Type 4/agonists , Receptor, Melanocortin, Type 4/deficiency , alpha-MSH/analogs & derivatives , Adrenal Insufficiency/drug therapy , Adrenal Insufficiency/metabolism , Adult , Amino Acid Sequence , Animals , Female , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , Middle Aged , Obesity/drug therapy , Obesity/metabolism , Pro-Opiomelanocortin/deficiency , Pro-Opiomelanocortin/metabolism , Receptor, Melanocortin, Type 4/genetics , Receptor, Melanocortin, Type 4/metabolism , alpha-MSH/pharmacology
16.
J Pediatr Endocrinol Metab ; 30(10): 1137-1140, 2017 Oct 26.
Article in English | MEDLINE | ID: mdl-28915118

ABSTRACT

BACKGROUND: Proopiomelanocortin (POMC) protein, encoded by the POMC gene, is the precursor of adrenocorticotropic hormone (ACTH) that is released from the anterior pituitary gland. Homozygous mutations in the POMC gene is associated with hyperphagia, severe and early-onset obesity, adrenal insufficiency, hypopigmentation of the skin and red hair. CASE PRESENTATION: A 9-year-old girl from a consanguineous family of Iraqi origin was diagnosed with type 1 diabetes. She also had a tall stature. Her laboratory assessment showed low cortisol and ACTH concentrations, normal renin and poor response to ACTH stimulation. Genetic testing revealed a novel biallelic mutation in the POMC gene. Her sibling who had severe obesity and central adrenal insufficiency was found to be a carrier of the same mutation. Both siblings had alabaster-colored skin and brown hair. CONCLUSIONS: POMC deficiency results in significant morbidity due to obesity, and it is also a potentially life threatening disease because of adrenal insufficiency. Therefore any suggestive symptom or sign of POMC deficiency warrants detailed investigations.


Subject(s)
Adrenal Insufficiency/diagnosis , Diabetes Mellitus, Type 1/complications , Mutation , Obesity/diagnosis , Pro-Opiomelanocortin/deficiency , Adrenal Insufficiency/complications , Adrenal Insufficiency/genetics , Child , Delayed Diagnosis , Female , Humans , Infant , Obesity/complications , Obesity/genetics , Pro-Opiomelanocortin/genetics
17.
J Clin Endocrinol Metab ; 102(2): 359-362, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27906547

ABSTRACT

Context: Loss-of-function mutations in the POMC gene are associated with a syndrome with the characteristics of adrenal insufficiency, obesity, and red hair. We describe here a case of pro-opiomelanocortin (POMC) deficiency in which adrenal insufficiency was not treated until the fourth year of life. One of the disease-causative POMC mutations was characterized in vitro using a unique approach. Case Description: A boy presented in the first year of life with red hair, growth acceleration, moderate obesity, and recurrent cholestasis, which was followed by 2 episodes of hypoglycemia at the ages of 1.5 and 3 years. The diagnosis was suspected at the age of 3.6 years after documentation of undetectable levels of plasma adrenocorticotropic hormone and serum cortisol, after which replacement with hydrocortisone was initiated. Sequencing of the POMC gene revealed compound heterozygosity for c.-11C>A/p.W84X mutations. The p.W84X mutation is predicted to result in a marked truncation of preprohormone. Using a messenger RNA transfection approach followed by an in vitro translation assay, we could directly demonstrate that the transcript with c.-11C>A substitution is predominantly translated within a new open reading frame; however, translation of the POMC main reading frame is preserved, with translation efficiency being ∼17% of the wild-type transcript. Conclusions: The current report provides important information on the natural course of POMC deficiency. In vitro translation studies demonstrated residual translation of the main coding region from an allele with the c.-11C>A mutation, which at least partially explains a relatively late presentation of adrenal insufficiency in the patient.


Subject(s)
Adrenal Insufficiency/diagnosis , Adrenal Insufficiency/genetics , Mutation , Obesity/diagnosis , Obesity/genetics , Pro-Opiomelanocortin/deficiency , Pro-Opiomelanocortin/genetics , Alleles , DNA Mutational Analysis/methods , Humans , Infant , Male , Protein Biosynthesis
18.
Genes Brain Behav ; 16(4): 449-461, 2017 04.
Article in English | MEDLINE | ID: mdl-27870313

ABSTRACT

Opioid receptor antagonist naltrexone reduces alcohol consumption and relapse in both humans and rodents. This study investigated whether hypothalamic proopiomelanocortin (POMC) neurons (producing beta-endorphin and melanocortins) play a role in alcohol drinking behaviors. Both male and female mice with targeted deletion of two neuronal Pomc enhancers nPE1 and nPE2 (nPE-/-), resulting in hypothalamic-specific POMC deficiency, were studied in short-access (4-h/day) drinking-in-the-dark (DID, alcohol in one bottle, intermittent access (IA, 24-h cycles of alcohol access every other day, alcohol vs. water in a two-bottle choice) and alcohol deprivation effect (ADE) models. Wild-type nPE+/+ exposed to 1-week DID rapidly established stable alcohol drinking behavior with more intake in females, whereas nPE-/- mice of both sexes had less intake and less preference. Although nPE-/- showed less saccharin intake and preference than nPE+/+, there was no genotype difference in sucrose intake or preference in the DID paradigm. After 3-week IA, nPE+/+ gradually escalated to high alcohol intake and preference, with more intake in females, whereas nPE-/- showed less escalation. Pharmacological blockade of mu-opioid receptors with naltrexone reduced intake in nPE+/+ in a dose-dependent manner, but had blunted effects in nPE-/- of both sexes. When alcohol was presented again after 1-week abstinence from IA, nPE+/+ of both sexes displayed significant increases in alcohol intake (ADE or relapse-like drinking), with more pronounced ADE in females, whereas nPE-/- did not show ADE in either sex. Our results suggest that neuronal POMC is involved in modulation of alcohol 'binge' drinking, escalation and 'relapse', probably via hypothalamic-mediated mechanisms, with sex differences.


Subject(s)
Adrenal Insufficiency/metabolism , Alcohol Drinking/metabolism , Obesity/metabolism , Pro-Opiomelanocortin/deficiency , Adrenal Insufficiency/genetics , Alcohol Drinking/drug therapy , Alcohol Drinking/genetics , Animals , Ethanol/pharmacology , Female , Genotype , Hypothalamus/drug effects , Hypothalamus/metabolism , Male , Mice , Mice, Knockout , Naltrexone/pharmacology , Obesity/genetics , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism
20.
N Engl J Med ; 375(3): 240-6, 2016 Jul 21.
Article in English | MEDLINE | ID: mdl-27468060

ABSTRACT

Patients with rare defects in the gene encoding proopiomelanocortin (POMC) have extreme early-onset obesity, hyperphagia, hypopigmentation, and hypocortisolism, resulting from the lack of the proopiomelanocortin-derived peptides melanocyte-stimulating hormone and corticotropin. In such patients, adrenal insufficiency must be treated with hydrocortisone early in life. No effective pharmacologic treatments have been available for the hyperphagia and obesity that characterize the condition. In this investigator-initiated, open-label study, two patients with proopiomelanocortin deficiency were treated with setmelanotide, a new melanocortin-4 receptor agonist. The patients had a sustainable reduction in hunger and substantial weight loss (51.0 kg after 42 weeks in Patient 1 and 20.5 kg after 12 weeks in Patient 2).


Subject(s)
Hyperphagia/drug therapy , Metabolism, Inborn Errors/drug therapy , Obesity/drug therapy , Pro-Opiomelanocortin/deficiency , Receptor, Melanocortin, Type 4/agonists , alpha-MSH/analogs & derivatives , Adult , Blood Pressure , Female , Humans , Hyperphagia/genetics , Metabolism, Inborn Errors/genetics , Obesity/genetics , Phenotype , Pilot Projects , Pro-Opiomelanocortin/genetics , Young Adult , alpha-MSH/adverse effects , alpha-MSH/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...