Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Am J Hum Genet ; 108(8): 1526-1539, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34270938

ABSTRACT

Pituitary hormone deficiency occurs in ∼1:4,000 live births. Approximately 3% of the cases are due to mutations in the alpha isoform of POU1F1, a pituitary-specific transcriptional activator. We found four separate heterozygous missense variants in unrelated individuals with hypopituitarism that were predicted to affect a minor isoform, POU1F1 beta, which can act as a transcriptional repressor. These variants retain repressor activity, but they shift splicing to favor the expression of the beta isoform, resulting in dominant-negative loss of function. Using a high-throughput splicing reporter assay, we tested 1,070 single-nucleotide variants in POU1F1. We identified 96 splice-disruptive variants, including 14 synonymous variants. In separate cohorts, we found two additional synonymous variants nominated by this screen that co-segregate with hypopituitarism. This study underlines the importance of evaluating the impact of variants on splicing and provides a catalog for interpretation of variants of unknown significance in POU1F1.


Subject(s)
High-Throughput Screening Assays/methods , Hypopituitarism/pathology , Mutation , Pituitary Hormones/deficiency , RNA Splicing/genetics , Transcription Factor Pit-1/genetics , Adolescent , Adult , Child , Child, Preschool , Humans , Hypopituitarism/etiology , Hypopituitarism/metabolism , Male , Pedigree
2.
Genet Med ; 26(4): 101059, 2024 04.
Article in English | MEDLINE | ID: mdl-38158857

ABSTRACT

PURPOSE: Oral-facial-digital (OFD) syndromes are genetically heterogeneous developmental disorders, caused by pathogenic variants in genes involved in primary cilia formation and function. We identified a previously undescribed type of OFD with brain anomalies, ranging from alobar holoprosencephaly to pituitary anomalies, in 6 unrelated families. METHODS: Exome sequencing of affected probands was supplemented with alternative splicing analysis in patient and control lymphoblastoid and fibroblast cell lines, and primary cilia structure analysis in patient fibroblasts. RESULTS: In 1 family with 2 affected males, we identified a germline variant in the last exon of ZRSR2, NM_005089.4:c.1211_1212del NP_005080.1:p.(Gly404GlufsTer23), whereas 7 affected males from 5 unrelated families were hemizygous for the ZRSR2 variant NM_005089.4:c.1207_1208del NP_005080.1:p.(Arg403GlyfsTer24), either occurring de novo or inherited in an X-linked recessive pattern. ZRSR2, located on chromosome Xp22.2, encodes a splicing factor of the minor spliceosome complex, which recognizes minor introns, representing 0.35% of human introns. Patient samples showed significant enrichment of minor intron retention. Among differentially spliced targets are ciliopathy-related genes, such as TMEM107 and CIBAR1. Primary fibroblasts containing the NM_005089.4:c.1207_1208del ZRSR2 variant had abnormally elongated cilia, confirming an association between defective U12-type intron splicing, OFD and abnormal primary cilia formation. CONCLUSION: We introduce a novel type of OFD associated with elongated cilia and differential splicing of minor intron-containing genes due to germline variation in ZRSR2.


Subject(s)
Alternative Splicing , Orofaciodigital Syndromes , Male , Humans , Alternative Splicing/genetics , Orofaciodigital Syndromes/genetics , RNA Splicing , Introns , Spliceosomes/genetics , Ribonucleoproteins/genetics
3.
J Clin Immunol ; 43(2): 271-285, 2023 02.
Article in English | MEDLINE | ID: mdl-36251205

ABSTRACT

Patients with inborn errors of immunity (IEI) in Argentina were encouraged to receive licensed Sputnik, AstraZeneca, Sinopharm, Moderna, and Pfizer vaccines, even though most of the data of humoral and cellular responses combination on available vaccines comes from trials conducted in healthy individuals. We aimed to evaluate the safety and immunogenicity of the different vaccines in IEI patients in Argentina. The study cohort included adults and pediatric IEI patients (n = 118) and age-matched healthy controls (HC) (n = 37). B cell response was evaluated by measuring IgG anti-spike/receptor binding domain (S/RBD) and anti-nucleocapsid(N) antibodies by ELISA. Neutralization antibodies were also assessed with an alpha-S protein-expressing pseudo-virus assay. The T cell response was analyzed by IFN-γ secretion on S- or N-stimulated PBMC by ELISPOT and the frequency of S-specific circulating T follicular-helper cells (TFH) was evaluated by flow cytometry.No moderate/severe vaccine-associated adverse events were observed. Anti-S/RBD titers showed significant differences in both pediatric and adult IEI patients versus the age-matched HC cohort (p < 0.05). Neutralizing antibodies were also significantly lower in the patient cohort than in age-matched HC (p < 0.01). Positive S-specific IFN-γ response was observed in 84.5% of IEI patients and 82.1% presented S-specific TFH cells. Moderna vaccines, which were mainly administered in the pediatric population, elicited a stronger humoral response in IEI patients, both in antibody titer and neutralization capacity, but the cellular immune response was similar between vaccine platforms. No difference in humoral response was observed between vaccinated patients with and without previous SARS-CoV-2 infection.In conclusion, COVID-19 vaccines showed safety in IEI patients and, although immunogenicity was lower than HC, they showed specific anti-S/RBD IgG, neutralizing antibody titers, and T cell-dependent cellular immunity with IFN-γ secreting cells. These findings may guide the recommendation for a vaccination with all the available vaccines in IEI patients to prevent COVID-19 disease.


Subject(s)
COVID-19 , Vaccines , Adult , Humans , Child , COVID-19 Vaccines , Leukocytes, Mononuclear , COVID-19/prevention & control , SARS-CoV-2 , Vaccination , Antibodies, Neutralizing , Enzyme-Linked Immunospot Assay , Immunoglobulin G , Antibodies, Viral , Immunity, Cellular
4.
Am J Physiol Endocrinol Metab ; 311(6): E974-E988, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27802964

ABSTRACT

We studied the impact of high prolactin titers on liver and adipocyte gene expression related to glucose and insulin homeostasis in correlation with obesity onset. To that end we used mutant female mice that selectively lack dopamine type 2 receptors (D2Rs) from pituitary lactotropes (lacDrd2KO), which have chronic high prolactin levels associated with increased body weight, marked increments in fat depots, adipocyte size, and serum lipids, and a metabolic phenotype that intensifies with age. LacDrd2KO mice of two developmental ages, 5 and 10 mo, were used. In the first time point, obesity and increased body weight are marginal, although mice are hyperprolactinemic, whereas at 10 mo there is marked adiposity with a 136% increase in gonadal fat and a 36% increase in liver weight due to lipid accumulation. LacDrd2KO mice had glucose intolerance, hyperinsulinemia, and impaired insulin response to glucose already in the early stages of obesity, but changes in liver and adipose tissue transcription factors were time and tissue dependent. In chronic hyperprolactinemic mice liver Prlr were upregulated, there was liver steatosis, altered expression of the lipogenic transcription factor Chrebp, and blunted response of Srebp-1c to refeeding at 5 mo of age, whereas no effect was observed in the glycogenesis pathway. On the other hand, in adipose tissue a marked decrease in lipogenic transcription factor expression was observed when morbid obesity was already settled. These adaptive changes underscore the role of prolactin signaling in different tissues to promote energy storage.


Subject(s)
Adipocytes/metabolism , Hepatocytes/metabolism , Hyperprolactinemia/genetics , Liver/metabolism , Obesity/genetics , Receptors, Dopamine D2/genetics , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Enzyme-Linked Immunosorbent Assay , Fatty Liver/genetics , Fatty Liver/metabolism , Female , Gene Expression , Glucose/metabolism , Glucose Tolerance Test , Homeostasis/genetics , Hyperprolactinemia/metabolism , Immunohistochemistry , Insulin/metabolism , Lactotrophs/metabolism , Lipogenesis/genetics , Mice , Mice, Knockout , Nuclear Proteins/genetics , Obesity/metabolism , Radioimmunoassay , Real-Time Polymerase Chain Reaction , Receptors, Prolactin/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Transcription Factors/genetics , Up-Regulation
5.
Nat Rev Endocrinol ; 20(2): 77-92, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38102391

ABSTRACT

Pituitary cells that express the transcription factor SOX2 are stem cells because they can self-renew and differentiate into multiple pituitary hormone-producing cell types as organoids. Wounding and physiological challenges can activate pituitary stem cells, but cell numbers are not fully restored, and the ability to mobilize stem cells decreases with increasing age. The basis of these limitations is still unknown. The regulation of stem cell quiescence and activation involves many different signalling pathways, including those mediated by WNT, Hippo and several cytokines; more research is needed to understand the interactions between these pathways. Pituitary organoids can be formed from human or mouse embryonic stem cells, or from human induced pluripotent stem cells. Human pituitary organoid transplantation is sufficient to induce corticosterone release in hypophysectomized mice, raising the possibility of therapeutic applications. Today, pituitary organoids have the potential to assess the role of individual genes and genetic variants on hormone production ex vivo, providing an important tool for the advancement of exciting frontiers in pituitary stem cell biology and pituitary organogenesis. In this article, we provide an overview of notable discoveries in pituitary stem cell function and highlight important areas for future research.


Subject(s)
Induced Pluripotent Stem Cells , Humans , Animals , Mice , Induced Pluripotent Stem Cells/metabolism , Pituitary Gland/metabolism , Transcription Factors/metabolism , Signal Transduction , Cell Differentiation
6.
Article in English | MEDLINE | ID: mdl-38717911

ABSTRACT

CONTEXT: The pituitary gland is key for childhood growth, puberty, and metabolism. Pituitary dysfunction is associated with a spectrum of phenotypes, from mild to severe. Congenital Hypopituitarism (CH) is the most commonly reported pediatric endocrine dysfunction with an incidence of 1:4000, yet low rates of genetic diagnosis have been reported. OBJECTIVE: We aimed to unveil the genetic etiology of CH in a large cohort of patients from Argentina. METHODS: We performed whole exome sequencing of 137 unrelated cases of CH, the largest cohort examined with this method to date. RESULTS: Of the 137 cases, 19.1% and 16% carried pathogenic or likely pathogenic variants in known and new genes, respectively, while 28.2% carried variants of uncertain significance. This high yield was achieved through the integration of broad gene panels (genes described in animal models and/or other disorders), an unbiased candidate gene screen with a new bioinformatics pipeline (including genes high loss of function intolerance), and analysis of copy number variants. Three novel findings emerged. First, the most prevalent affected gene encodes the cell adhesion factor ROBO1. Affected children had a spectrum of phenotypes, consistent with a role beyond pituitary stalk interruption syndrome. Second, we found that CHD7 mutations also produce a phenotypic spectrum, not always associated with full CHARGE syndrome. Third, we add new evidence of pathogenicity in the genes PIBF1 and TBC1D32, and report 13 novel candidate genes associated with CH (e.g. PTPN6, ARID5B). CONCLUSION: Overall, these results provide an unprecedented insight into the diverse genetic etiology of hypopituitarism.

7.
Genesis ; 51(11): 785-92, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23996951

ABSTRACT

Tissue-specific expression of cre recombinase is a well-established genetic tool to analyze gene function, and it is limited only by the efficiency and specificity of available cre mouse strains. Here, we report the generation of a transgenic line containing a cre cassette with codon usage optimized for mammalian cells (iCre) under the control of a mouse glycoprotein hormone α-subunit (αGSU) regulatory sequences in a bacterial artificial chromosome genomic clone. Initial analysis of this transgenic line, Tg(αGSU-iCre), with cre reporter strains reveals onset of cre activity in the differentiating cells of the developing anterior pituitary gland at embryonic day 12.5, with a pattern characteristic of endogenous αGSU. In adult mice, αGSU-iCre was active in the anterior lobe of the pituitary gland and in the cells that produce αGSU (gonadotropes and thyrotropes) with high penetrance. Little or no activity was observed in other tissues, including skeletal and cardiac muscle, brain, kidney, lungs, testis, ovary, and liver. This αGSU-iCre line is suitable for efficient, specific, and developmentally regulated deletion of floxed alleles in anterior pituitary gonadotropes and thyrotropes.


Subject(s)
Glycoprotein Hormones, alpha Subunit/genetics , Gonadotrophs/metabolism , Integrases/metabolism , Recombination, Genetic , Thyrotrophs/metabolism , Alleles , Animals , Chromosomes, Artificial, Bacterial , Cloning, Molecular , Embryo, Mammalian , Female , Genotype , Glycoprotein Hormones, alpha Subunit/metabolism , Integrases/genetics , Male , Mice , Mice, Transgenic , Organ Specificity , Regulatory Sequences, Nucleic Acid
8.
Pituitary ; 16(3): 303-10, 2013 Sep.
Article in English | MEDLINE | ID: mdl-22886682

ABSTRACT

The role of angiogenesis in human pituitary tumor progression is questioned. Our aim was to characterize the morphologic changes that occur in the vasculature of pituitary adenomas, in correlation with the expression of nestin, a protein found in endothelial cells of newly formed vessels of developing organs. We also evaluated the relation of angiogenic markers and nestin with Ki-67 index. Immunohistochemical studies were performed on paraffin embedded samples of 47 pituitary adenomas and six normal pituitaries. We determined microvessel density (number of CD31+ or CD34+ vessels per square millimetre), vascular area (cumulative area occupied by vessels), average vessel size, and further classified vessels as small (< 100 µm2) or large (> 100 µm2). We correlated the above parameters with nestin expression and Ki-67 index. Lower vascular area compared to normal tissue was found in adenomas (p < 0.05). Interestingly, pituitary adenomas had significantly more small vessels than control pituitaries (p < 0.04 for CD31 and CD34). In tumors many capillaries were positive for nestin, while scarce staining was detected in controls, so that nestin positive area was significantly higher in tumors. Furthermore, nestin area correlated positively with the % of small vessels. Ki-67 correlated neither with vascular area nor with nestin expression. In human pituitary tumors there was a predominance of small capillaries in correlation with increased expression of the progenitor marker nestin. We suggest that angiogenesis is an active process in these tumors, in spite of their low total vascular area when compared to normal pituitaries.


Subject(s)
Adenoma/metabolism , Nestin/metabolism , Pituitary Neoplasms/metabolism , Adult , Aged , Blood Vessels/metabolism , Humans , Immunohistochemistry , In Vitro Techniques , Ki-67 Antigen/metabolism , Middle Aged , Neovascularization, Pathologic , Young Adult
9.
Front Endocrinol (Lausanne) ; 14: 1132787, 2023.
Article in English | MEDLINE | ID: mdl-36843573

ABSTRACT

Prokineticin receptor 2 (PROKR2) encodes for a G-protein-coupled receptor that can bind PROK1 and PROK2. Mice lacking Prokr2 have been shown to present abnormal olfactory bulb formation as well as defects in GnRH neuron migration. Patients carrying mutations in PROKR2 typically present hypogonadotropic hypogonadism, anosmia/hyposmia or Kallmann Syndrome. More recently variants in PROKR2 have been linked to several other endocrine disorders. In particular, several patients with pituitary disorders have been reported, ranging from mild phenotypes, such as isolated growth hormone deficiency, to more severe ones, such as septo-optic dysplasia. Here we summarize the changing landscape of PROKR2-related disease, the variants reported to date, and discuss their origin, classification and functional assessment.


Subject(s)
Kallmann Syndrome , Neuropeptides , Mice , Animals , Neuropeptides/metabolism , Kallmann Syndrome/genetics , Genotype , Receptors, G-Protein-Coupled/genetics , Phenotype , Receptors, Peptide/genetics
10.
J Pharmacol Exp Ther ; 337(3): 766-74, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21406548

ABSTRACT

Prolactin-secreting adenomas are the most frequent type among pituitary tumors, and pharmacological therapy with dopamine agonists remains the mainstay of treatment. But some adenomas are resistant, and a decrease in the number or function of dopamine D2 receptors (D2Rs) has been described in these cases. D2R knockout [Drd2(-/-)] mice have chronic hyperprolactinemia and pituitary hyperplasia and provide an experimental model for dopamine agonist-resistant prolactinomas. We described previously that disruption of D2Rs increases vascular endothelial growth factor (VEGF) expression. We therefore designed two strategies of antiangiogenesis using prolactinomas generated in Drd2(-/-) female mice: direct intra-adenoma mVEGF R1 (Flt-1)/Fc chimera (VEGF-TRAP) injection for 3 weeks [into subcutaneously transplanted pituitaries from Drd2(-/-) mice] and systemic VEGF neutralization with the specific monoclonal antibody G6-31. Both strategies resulted in substantial decrease of prolactin content and lactotrope area, and a reduction in tumor size was observed in in situ prolactinomas. There were significant decreases in vascularity, evaluated by cluster of differentiation molecule 31 vessel staining, and proliferation (proliferating cell nuclear antigen staining) in response to both anti-VEGF treatments. These data demonstrate that the antiangiogenic approach was effective in inhibiting the growth of in situ dopamine-resistant prolactinomas as well as in the transplanted adenomas. No differences in VEGF protein expression were observed after either anti-VEGF treatment, and, although serum VEGF was increased in G6-31-treated mice, pituitary activation of the VEGF receptor 2 signaling pathway was reduced. Our results indicate that, even though the role of angiogenesis in pituitary adenomas is contentious, VEGF might contribute to adequate vascular supply and represent a supplementary therapeutic target in dopamine agonist-resistant prolactinomas.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Pituitary Neoplasms/drug therapy , Prolactinoma/drug therapy , Recombinant Fusion Proteins/therapeutic use , Vascular Endothelial Growth Factor Receptor-1/metabolism , Animals , Antibodies, Monoclonal/metabolism , Cell Proliferation/drug effects , Dopamine/metabolism , Female , Hyperplasia , Mice , Mice, Inbred C57BL , Mice, Knockout , Microvessels/drug effects , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Pituitary Gland/blood supply , Pituitary Gland/metabolism , Pituitary Gland/pathology , Pituitary Neoplasms/blood supply , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/pathology , Prolactin/blood , Prolactinoma/blood supply , Prolactinoma/metabolism , Prolactinoma/pathology , Receptors, Dopamine D2/genetics , Receptors, Vascular Endothelial Growth Factor , Vascular Endothelial Growth Factor A/immunology , Vascular Endothelial Growth Factor A/metabolism
11.
J Clin Endocrinol Metab ; 106(7): 1956-1976, 2021 06 16.
Article in English | MEDLINE | ID: mdl-33729509

ABSTRACT

PURPOSE: Congenital hypopituitarism (CH) can present in isolation or with other birth defects. Mutations in multiple genes can cause CH, and the use of a genetic screening panel could establish the prevalence of mutations in known and candidate genes for this disorder. It could also increase the proportion of patients that receive a genetic diagnosis. METHODS: We conducted target panel genetic screening using single-molecule molecular inversion probes sequencing to assess the frequency of mutations in known hypopituitarism genes and new candidates in Argentina. We captured genomic deoxyribonucleic acid from 170 pediatric patients with CH, either alone or with other abnormalities. We performed promoter activation assays to test the functional effects of patient variants in LHX3 and LHX4. RESULTS: We found variants classified as pathogenic, likely pathogenic, or with uncertain significance in 15.3% of cases. These variants were identified in known CH causative genes (LHX3, LHX4, GLI2, OTX2, HESX1), in less frequently reported genes (FOXA2, BMP4, FGFR1, PROKR2, PNPLA6) and in new candidate genes (BMP2, HMGA2, HNF1A, NKX2-1). CONCLUSION: In this work, we report the prevalence of mutations in known CH genes in Argentina and provide evidence for new candidate genes. We show that CH is a genetically heterogeneous disease with high phenotypic variation and incomplete penetrance, and our results support the need for further gene discovery for CH. Identifying population-specific pathogenic variants will improve the capacity of genetic data to predict eventual clinical outcomes.


Subject(s)
Endocrine System Diseases/genetics , Genetic Testing/statistics & numerical data , Hypopituitarism/genetics , Mutation/genetics , Adolescent , Adult , Argentina , Child , Child, Preschool , Female , Genetic Heterogeneity , Humans , Infant , LIM-Homeodomain Proteins/genetics , Male , Phenotype , Polymorphism, Single Nucleotide , Transcription Factors/genetics , Young Adult
12.
Neuroendocrinology ; 92(4): 207-14, 2010.
Article in English | MEDLINE | ID: mdl-20975260

ABSTRACT

Dopamine D2 receptor (D2R) participation in prolactin regulation is well documented, but the role of D2Rs in the control of other hormones involved in growth, food intake and glucose metabolism has not been extensively studied. The study of D2R knockout mice (Drd2(-/-)) puts forward new insights into the role of the D2R in growth hormone (GH)-releasing hormone-GH regulation, peptides involved in food intake, glucose homeostasis, as well as in prolactinoma development. The expected phenotype of chronic hyperprolactinemia and prolactinoma development was found in the Drd2(-/-) mouse, and this model constitutes a valuable tool in the study of dopamine-resistant prolactinomas. Unexpectedly, these mice were growth retarded, and the importance of functional hypothalamic D2Rs in the neonatal period was revealed. In the Drd2(-/-) mouse there was a failure of high neonatal GH levels and therefore the expansion of pituitary somatotropes was permanently altered. These mice also had increased food intake, and a sexually dimorphic participation of the D2R in food intake regulation is suggested. The effect described is probably secondary to D2R regulation of prolactin secretion. Furthermore, the negative modulation of D2Rs on α-melanocyte-stimulating hormone release and positive action on the hypothalamic expression of orexins reveals the complex D2R regulation of food intake. Finally, pancreatic D2Rs inhibit glucose-stimulated insulin release. Lack of dopaminergic inhibition throughout development in the Drd2(-/-) mouse may exert a gradual deteriorating effect on insulin homeostasis, so that eventually glucose intolerance develops. These results highlight the complex endocrine actions of the D2Rs at different levels, hypothalamus, pituitary or pancreas, which function to improve fitness, reproductive success and survival.


Subject(s)
Endocrine System/physiology , Metabolism/genetics , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/physiology , Animals , Eating/genetics , Eating/physiology , Endocrine System/metabolism , Growth Hormone/metabolism , Growth Hormone-Releasing Hormone/metabolism , Mice , Mice, Knockout , Prolactin/metabolism , Receptors, Dopamine D2/metabolism
13.
Front Horm Res ; 38: 59-69, 2010.
Article in English | MEDLINE | ID: mdl-20616496

ABSTRACT

The role of dopaminergic receptors in the control of GH release remains controversial. The dopamine receptor 2 (D2R) knockout mouse represents a useful model to study the participation of the D2R on growth and GHRH-GH regulation. These knockout mice have hyperprolactinemia and lactotrope hyperplasia, but unexpectedly, they are also growth retarded. In D2R knockout mice there is a significant decrease in somatotrope population, which is paralleled by decreased GH content and output from pituitary cells. The sensitivity of GHRH-induced GH and cAMP release is similar between genotypes, even though the response amplitude is lower in knockouts. We point to an involvement of D2R signaling at the hypothalamic level as dopamine did not release GH acting at the pituitary level, and both somatostatin and GHRH mRNA expression are altered in knockout mice. The similarity of the pituitary defect in the D2R knockout mouse to that of GHRH deficient models suggests a probable mechanism. Loss of dopamine signaling via hypothalamic D2Rs at a critical age may cause inadequate GHRH secretion subsequently leading to inappropriate somatotrope lineage development. Furthermore, GH pulsatility, which depends on a regulated temporal balance between GHRH and somatostatin output might be compromised in D2R knockout mice, leading to lower IGF-I, and growth retardation.


Subject(s)
Dopamine/physiology , Growth Hormone-Releasing Hormone/physiology , Growth Hormone/physiology , Neurotransmitter Agents/physiology , Acromegaly/drug therapy , Animals , Growth , Humans , Mice , Receptors, Dopamine D2/physiology
14.
Front Endocrinol (Lausanne) ; 11: 614999, 2020.
Article in English | MEDLINE | ID: mdl-33542708

ABSTRACT

The anterior pituitary gland is comprised of specialized cell-types that produce and secrete polypeptide hormones in response to hypothalamic input and feedback from target organs. These specialized cells arise during embryonic development, from stem cells that express SOX2 and the pituitary transcription factor PROP1, which is necessary to establish the stem cell pool and promote an epithelial to mesenchymal-like transition, releasing progenitors from the niche. Human and mouse embryonic stem cells can differentiate into all major hormone-producing cell types of the anterior lobe in a highly plastic and dynamic manner. More recently human induced pluripotent stem cells (iPSCs) emerged as a viable alternative due to their plasticity and high proliferative capacity. This mini-review gives an overview of the major advances that have been achieved to develop protocols to generate pituitary hormone-producing cell types from stem cells and how these mechanisms are regulated. We also discuss their application in pituitary diseases, such as pituitary hormone deficiencies.


Subject(s)
Cell Differentiation/physiology , Induced Pluripotent Stem Cells/physiology , Induced Pluripotent Stem Cells/transplantation , Pituitary Gland/physiology , Pituitary Gland/transplantation , Regenerative Medicine/methods , Animals , Humans , Induced Pluripotent Stem Cells/cytology , Pituitary Diseases/pathology , Pituitary Diseases/therapy , Pituitary Gland/cytology , Regenerative Medicine/trends
16.
Endocr Pathol ; 20(1): 35-40, 2009.
Article in English | MEDLINE | ID: mdl-19172414

ABSTRACT

The present study reports the case of a 70-year-old Caucasian man who was referred to the Military Hospital of Buenos Aires for evaluation of a giant sellar-extrasellar mass with extension in the right temporal lobe and compression of the third ventricle. Patient was initially responsive to cabergoline with reduction of prolactin levels and shrinkage of tumor burden for at least 36 months. Thereafter, prolactin levels and tumor size increased even though cabergoline dosage was increased. Transcraneal surgery was performed at 56 months of treatment. Prolactin levels and tumor proliferation did not subside and the patient died 14 months later. High GH and IGF-I levels were observed in the late stages of tumor development, with no evidence of acromegalic features. Immunohistochemistry of the excised tumor revealed strong immunoreactivity for VEGF and FGF-2, two potent angiogenic factors, and CD31 (an endothelial marker) indicating high vascularization of the adenoma.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm/genetics , Ergolines/therapeutic use , Neovascularization, Pathologic/metabolism , Pituitary Neoplasms/pathology , Prolactinoma/pathology , Aged , Biomarkers, Tumor/analysis , Cabergoline , Humans , Hypothyroidism , Immunohistochemistry , Magnetic Resonance Imaging , Male , Pituitary Neoplasms/drug therapy , Pituitary Neoplasms/metabolism , Prolactin/blood , Prolactinoma/drug therapy , Prolactinoma/metabolism
19.
Mol Cell Endocrinol ; 445: 14-26, 2017 04 15.
Article in English | MEDLINE | ID: mdl-27650955

ABSTRACT

The anterior pituitary gland is comprised of specialized cell-types that produce and secrete polypeptide hormones in response to hypothalamic input and feedback from target organs. These specialized cells arise from stem cells that express SOX2 and the pituitary transcription factor PROP1, which is necessary to establish the stem cell pool and promote an epithelial to mesenchymal-like transition, releasing progenitors from the niche. The adult anterior pituitary responds to physiological challenge by mobilizing the SOX2-expressing progenitor pool and producing additional hormone-producing cells. Knowledge of the role of signaling pathways and extracellular matrix components in these processes may lead to improvements in the efficiency of differentiation of embryonic stem cells or induced pluripotent stem cells into hormone producing cells in vitro. Advances in our basic understanding of pituitary stem cell regulation and differentiation may lead to improved diagnosis and treatment for patients with hypopituitarism.


Subject(s)
Homeodomain Proteins/metabolism , Pituitary Gland, Anterior/cytology , SOXB1 Transcription Factors/metabolism , Stem Cells/cytology , Animals , Cell Differentiation , Epithelial-Mesenchymal Transition , Extracellular Matrix/metabolism , Humans , Pituitary Gland, Anterior/metabolism , Signal Transduction , Stem Cells/metabolism
20.
Medicina (B Aires) ; 66(6): 574-82, 2006.
Article in Spanish | MEDLINE | ID: mdl-17240634

ABSTRACT

Stem cells are defined by virtue of their functional attributes: absence of tissue specific differentitated markers, capable of proliferation, able to self-maintain the population, able to produce a large number of differentiated, functional progeny, able to regenerate the tissue after injury. Cell therapy is an alternative for the treatment of several diseases, like cardiac diseases (cell cardiomyoplasty). A variety of stem cells could be used for cardiac repair: from cardiac and extracardiac sources. Each cell type has its own profile of advantages, limitations, and practicability issues in specific clinical settings. Differentiation of bone marrow stem cells to cardiomyocyte-like cells have been observed under different culture conditions. The presence of resident cardiac stem cell population capable of differentiation into cardiomyocyte or vascular lineage suggests that these cells could be used for cardiac tissue repair, and represent a great promise for clinical application. Stem cells mobilization by cytokines may also offer a strategy for cardiac regeneration. The use of stem cells (embryonic and adult) may hold the key to replacing cells lost in many devastating diseases. This potential benefit is a major focus for stem cell research.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation , Heart/physiology , Regeneration/physiology , Stem Cells/cytology , Adult , Adult Stem Cells/cytology , Bone Marrow Cells/cytology , Embryonic Stem Cells/cytology , Guided Tissue Regeneration , Humans , Myocytes, Cardiac/cytology , Stem Cell Transplantation , Tissue Engineering/methods
SELECTION OF CITATIONS
SEARCH DETAIL