Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Eye (Lond) ; 38(6): 1176-1182, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38049626

RESUMO

BACKGROUND/OBJECTIVES: CLN2 Batten Disease is a fatal neurodegenerative condition of childhood associated with retinal dystrophy and blindness. Intracerebroventricular infusion of rhTPP1 greatly slows the rate of neurodegenerative decline but not retinopathy. Intravitreal rhTPP1 is known to slow retinal degeneration in a canine model of CLN2. We report a first-in-man controlled clinical trial of intravitreal rhTPP1 for CLN2 associated retinal dystrophy. SUBJECTS/METHODS: 8 children aged 5-9 with CLN2 Batten Disease were prospectively enroled. Severely affected patients were preferentially selected, provided that vision was better than no perception of light. Children underwent 8 weekly intravitreal injections of rhTPP1 (0.2 mg in 0.05 ml) into the right eye for 12-18 months. The left eye was untreated and acts as a paired control. The primary outcome was safety based on the clinical detection of complications. A secondary outcome was paracentral macular volume (PMV) measured by spectral domain OCT. Linear regression/paired t tests were used to compare rates of decline. RESULTS: No severe adverse reactions (uveitis, raised IOP, media opacity) occurred. The mean baseline PMV was 1.28 mm3(right), 1.27 mm3(left). 3 of the youngest patients exhibited bilateral progressive retinal thinning (p < 0.05), whereas retinal volume was stable in the remaining 5 patients. In the 3 patients undergoing retinal degeneration, the rate of PMV loss was slower in the treated vs. untreated eye (p = 0.000042, p = 0.0011, p = 0.00022). CONCLUSIONS: Intravitreal rhTPP1 appears to be a safe and effective treatment for CLN2 related retinopathy however commencement of treatment early in the course of disease is more likely to be efficacious.


Assuntos
Lipofuscinoses Ceroides Neuronais , Distrofias Retinianas , Criança , Humanos , Animais , Cães , Tripeptidil-Peptidase 1 , Aminopeptidases/efeitos adversos , Serina Proteases/efeitos adversos , Dipeptidil Peptidases e Tripeptidil Peptidases/uso terapêutico , Lipofuscinoses Ceroides Neuronais/tratamento farmacológico , Terapia de Reposição de Enzimas , Injeções Intravítreas , Distrofias Retinianas/induzido quimicamente , Distrofias Retinianas/complicações , Distrofias Retinianas/tratamento farmacológico
2.
Orphanet J Rare Dis ; 17(1): 19, 2022 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-35045884

RESUMO

BACKGROUND: Cerliponase alfa, an enzyme replacement therapy for neuronal ceroid lipofuscinosis type 2 (CLN2), is currently available in England through a managed access agreement (MAA). It is administered every 2 weeks via an intracerebroventricular device. Here we report qualitative research with families of children with CLN2 disease and healthcare professionals (HCPs) who run the MAA, to understand how access to cerliponase alfa via the MAA at Great Ormond Street Hospital (GOSH) in London, and the overall management of CLN2 disease, was affected during the coronavirus disease 2019 (COVID-19) pandemic. METHODS: Telephone interviews were conducted with nine families, representing 11 children with CLN2 disease, and two HCPs in November and December 2020. RESULTS: Children had received cerliponase alfa treatment for a mean (SD) of 23.1 ± 24.7 months (7.1 ± 4.6 months in the MAA). Families travelled 7-398 km for treatment (mean 210 ± 111 km). Treatment with cerliponase alfa was designated "essential" by GOSH and continued as normal during the pandemic but with extra safety precautions, and no children missed any treatments. Families were highly motivated to continue treatment, despite considerable anxiety about the risk of coronavirus infection from travelling and staying overnight but were reassured by communications from GOSH and the safety precautions put in place. Support therapy services were widely compromised, causing families concern about deterioration in their children's condition. Families were confused about COVID-19 testing and shielding, and were unclear whether children with CLN2 disease were vulnerable to COVID-19. CONCLUSIONS: Looking forward, advice for children with CLN2 disease should be specific and tailored, taking into account the family unit. Support therapies should be considered essential alongside cerliponase alfa treatment.


Assuntos
COVID-19 , Lipofuscinoses Ceroides Neuronais , Aminopeptidases , Teste para COVID-19 , Criança , Dipeptidil Peptidases e Tripeptidil Peptidases , Humanos , Pandemias , Proteínas Recombinantes , SARS-CoV-2 , Serina Proteases , Tripeptidil-Peptidase 1
3.
Orphanet J Rare Dis ; 16(1): 217, 2021 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-33980287

RESUMO

BACKGROUND: Utility studies enable preference-based quantification of a disease's impact on patients' health-related quality of life (HRQoL). It is often difficult to obtain utility values for rare, neurodegenerative conditions due to cognitive burden of direct elicitation methods, and the limited size of patient/caregiver populations. CLN2 disease (neuronal ceroid lipofuscinosis type 2) is an ultra-rare, progressive condition, for which there are no published utility data fully capturing all disease stages. This case study demonstrates how utility values can be estimated for ultra-rare paediatric diseases by asking clinicians to complete EQ-5D-5L questionnaires based on vignettes describing the stages of CLN2 disease. METHODS: An indirect elicitation method using proxy-reporting by clinical experts was adopted. Eighteen vignettes were developed, describing nine progressive disease stages as defined by motor and language domain scores of the CLN2 Clinical Rating Scale, in individuals treated with cerliponase alfa or standard care. Eight clinical experts with experience of treating CLN2 disease with cerliponase alfa and current standard care completed the proxy version 2 EQ-5D-5L online after reading these vignettes. Resulting scores were converted to EQ-5D-5L utility values for each disease stage, using UK, German and Spanish value sets. RESULTS: Utility values, which are typically anchored by 0 (equivalent to death) and 1 (full health), decreased with CLN2 disease progression (results spanned the maximum range of the utility scale). Assigned utility values were consistently higher for patients receiving cerliponase alfa than standard care; differences were statistically significant for the 6 most severe disease stages (p < 0.05). Analysis of the individual dimensions of the EQ-5D-5L showed that greatest differences between patients treated with cerliponase alfa and standard care occurred in the pain dimension (differences in mean scores ranged between no difference and 1.8), with notable differences also observed in the anxiety/depression dimension (differences in mean scores ranged between 0.1 and 1.0). CONCLUSIONS: This study demonstrates a feasible methodology for eliciting utility values in CLN2 disease, indicating HRQoL declines with disease progression. Vignettes describing patients receiving cerliponase alfa were consistently assigned higher utility values for the same disease state, suggesting this treatment improves HRQoL compared with standard care. Trial registration NCT01907087, NCT02485899.


Assuntos
Lipofuscinoses Ceroides Neuronais , Qualidade de Vida , Criança , Ensaios Clínicos como Assunto , Depressão , Nível de Saúde , Humanos , Doenças Raras , Inquéritos e Questionários , Tripeptidil-Peptidase 1
4.
Endocrinology ; 162(7)2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33837405

RESUMO

Targeted oncogenesis is the process of driving tumor formation by engineering transgenic mice that express an oncogene under the control of a cell-type specific promoter. Such tumors can be adapted to cell culture, providing immortalized cell lines. To make it feasible to follow the process of tumorigenesis and increase the opportunity for generating cell lines, we developed a mouse strain that expresses SV40 T antigens in response to Cre-recombinase. Using CRISPR/Cas9 we inserted a cassette with coding sequences for SV40 T antigens and an internal ribosome entry site with green fluorescent protein cassette (IRES-GFP) into the Rosa26 locus, downstream from a stop sequence flanked by loxP sites: Rosa26LSL-SV40-GFP. These mice were mated with previously established Prop1-cre and Tshb-cre transgenic lines. Both the Rosa26LSL-SV40-GFP/+; Prop1-cre and Rosa26LSL-SV40-GFP/+; Tshb-cre mice developed fully penetrant dwarfism and large tumors by 4 weeks. Tumors from both of these mouse lines were adapted to growth in cell culture. We have established a progenitor-like cell line (PIT-P1) that expresses Sox2 and Pitx1, and a thyrotrope-like cell line (PIT-T1) that expresses Pou1f1 and Cga. These studies demonstrate the utility of the novel, Rosa26LSL-SV40-GFP mouse line for reliable targeted oncogenesis and development of unique cell lines.


Assuntos
Antígenos Transformantes de Poliomavirus/genética , Expressão Gênica/efeitos dos fármacos , Integrases/farmacologia , Neoplasias Hipofisárias/genética , Animais , Carcinogênese , Linhagem Celular Tumoral , Cruzamentos Genéticos , Técnicas de Introdução de Genes , Proteínas de Homeodomínio/genética , Hiperplasia , Camundongos , Camundongos Transgênicos , Hipófise/metabolismo , Hipófise/patologia , Tireotropina Subunidade beta/genética
5.
Proc Natl Acad Sci U S A ; 118(15)2021 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-33827924

RESUMO

Methylation of histone H3K9 is a hallmark of epigenetic silencing in eukaryotes. Nucleosome modifications often rely on positive feedback where enzymes are recruited by modified nucleosomes. A combination of local and global feedbacks has been proposed to account for some dynamic properties of heterochromatin, but the range at which the global feedbacks operate and the exact mode of heterochromatin propagation are not known. We investigated these questions in fission yeast. Guided by mathematical modeling, we incrementally increased the size of the mating-type region and profiled heterochromatin establishment over time. We observed exponential decays in the proportion of cells with active reporters, with rates that decreased with domain size. Establishment periods varied from a few generations in wild type to >200 generations in the longest region examined, and highly correlated silencing of two reporters located outside the nucleation center was observed. On a chromatin level, this indicates that individual regions are silenced in sudden bursts. Mathematical modeling accounts for these bursts if heterochromatic nucleosomes facilitate a deacetylation or methylation reaction at long range, in a distance-independent manner. A likely effector of three-dimensional interactions is the evolutionarily conserved Swi6HP1 H3K9me reader, indicating the bursting behavior might be a general mode of heterochromatin propagation.


Assuntos
Regulação Fúngica da Expressão Gênica , Inativação Gênica , Heterocromatina/metabolismo , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Genes Fúngicos Tipo Acasalamento , Heterocromatina/genética , Modelos Genéticos , Schizosaccharomyces , Proteínas de Schizosaccharomyces pombe/genética , Proteínas de Schizosaccharomyces pombe/metabolismo
6.
J Clin Endocrinol Metab ; 106(7): 1956-1976, 2021 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-33729509

RESUMO

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.


Assuntos
Doenças do Sistema Endócrino/genética , Testes Genéticos/estatística & dados numéricos , Hipopituitarismo/genética , Mutação/genética , Adolescente , Adulto , Argentina , Criança , Pré-Escolar , Feminino , Heterogeneidade Genética , Humanos , Lactente , Proteínas com Homeodomínio LIM/genética , Masculino , Fenótipo , Polimorfismo de Nucleotídeo Único , Fatores de Transcrição/genética , Adulto Jovem
7.
Mamm Genome ; 30(1-2): 5-22, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30778664

RESUMO

Maternal and fetal sources of thyroid hormone are important for the development of many organ systems. Thyroid hormone deficiency causes variable intellectual disability and hearing impairment in mouse and man, but the basis for this variation is not clear. To explore this variation, we studied two thyroid hormone-deficient mouse mutants with mutations in pituitary-specific transcription factors, POU1F1 and PROP1, that render them unable to produce thyroid stimulating hormone. DW/J-Pou1f1dw/dw mice have profound deafness and both neurosensory and conductive hearing impairment, while DF/B-Prop1df/df mice have modest elevations in hearing thresholds consistent with developmental delay, eventually achieving normal hearing ability. The thyroid glands of Pou1f1 mutants are more severely affected than those of Prop1df/df mice, and they produce less thyroglobulin during the neonatal period critical for establishing hearing. We previously crossed DW/J-Pou1f1dw/+ and Cast/Ei mice and mapped a major locus on Chromosome 2 that protects against hypothyroidism-induced hearing impairment in Pou1f1dw/dw mice: modifier of dw hearing (Mdwh). Here we refine the location of Mdwh by genotyping 196 animals with 876 informative SNPs, and we conduct novel mapping with a DW/J-Pou1f1dw/+ and 129/P2 cross that reveals 129/P2 mice also have a protective Mdwh locus. Using DNA sequencing of DW/J and DF/B strains, we determined that the genes important for thyroid gland function within Mdwh vary in amino acid sequence between strains that are susceptible or resistant to hypothyroidism-induced hearing impairment. These results suggest that the variable effects of congenital hypothyroidism on the development of hearing ability are attributable to genetic variation in postnatal thyroid gland folliculogenesis and function.


Assuntos
Suscetibilidade a Doenças , Variação Genética , Perda Auditiva/etiologia , Hipotireoidismo/complicações , Glândula Tireoide/embriologia , Glândula Tireoide/metabolismo , Alelos , Animais , Animais Recém-Nascidos , Biomarcadores , Tronco Encefálico/efeitos dos fármacos , Tronco Encefálico/metabolismo , Modelos Animais de Doenças , Orelha Média/embriologia , Orelha Média/metabolismo , Perda Auditiva/diagnóstico , Perda Auditiva/tratamento farmacológico , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Mutação , Glândula Tireoide/patologia , Hormônios Tireóideos/farmacologia
8.
Artigo em Inglês | MEDLINE | ID: mdl-29739035

RESUMO

BACKGROUND: Congenital Hypopituitarism is caused by genetic and environmental factors. Over 30 genes have been implicated in isolated and/or combined pituitary hormone deficiency. The etiology remains unknown for up to 80% of the patients, but most cases have been analyzed by limited candidate gene screening. Mutations in the PROP1 gene are the most common known cause, and the frequency of mutations in this gene varies greatly by ethnicity. We designed a custom array to assess the frequency of mutations in known hypopituitarism genes and new candidates, using single molecule molecular inversion probes sequencing (smMIPS). METHODS: We used this panel for the first systematic screening for causes of hypopituitarism in children. Molecular inversion probes were designed to capture 693 coding exons of 30 known genes and 37 candidate genes. We captured genomic DNA from 51 pediatric patients with CPHD (n = 43) or isolated GH deficiency (IGHD) (n = 8) and their parents and conducted next generation sequencing. RESULTS: We obtained deep coverage over targeted regions and demonstrated accurate variant detection by comparison to whole-genome sequencing in a control individual. We found a dominant mutation GH1, p.R209H, in a three-generation pedigree with IGHD. CONCLUSIONS: smMIPS is an efficient and inexpensive method to detect mutations in patients with hypopituitarism, drastically limiting the need for screening individual genes by Sanger sequencing.

9.
Endocr Rev ; 37(6): 636-675, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27828722

RESUMO

The genetic basis for combined pituitary hormone deficiency (CPHD) is complex, involving 30 genes in a variety of syndromic and nonsyndromic presentations. Molecular diagnosis of this disorder is valuable for predicting disease progression, avoiding unnecessary surgery, and family planning. We expect that the application of high throughput sequencing will uncover additional contributing genes and eventually become a valuable tool for molecular diagnosis. For example, in the last 3 years, six new genes have been implicated in CPHD using whole-exome sequencing. In this review, we present a historical perspective on gene discovery for CPHD and predict approaches that may facilitate future gene identification projects conducted by clinicians and basic scientists. Guidelines for systematic reporting of genetic variants and assigning causality are emerging. We apply these guidelines retrospectively to reports of the genetic basis of CPHD and summarize modes of inheritance and penetrance for each of the known genes. In recent years, there have been great improvements in databases of genetic information for diverse populations. Some issues remain that make molecular diagnosis challenging in some cases. These include the inherent genetic complexity of this disorder, technical challenges like uneven coverage, differing results from variant calling and interpretation pipelines, the number of tolerated genetic alterations, and imperfect methods for predicting pathogenicity. We discuss approaches for future research in the genetics of CPHD.


Assuntos
Genômica/métodos , Hipopituitarismo/genética , Animais , Humanos
10.
PLoS One ; 11(9): e0160068, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27685990

RESUMO

Cocaine-and Amphetamine Regulated Transcript (CART) peptide is expressed in the brain, endocrine and neuroendocrine systems and secreted into the serum. It is thought to play a role in regulation of hypothalamic pituitary functions. Here we report a spatial and temporal analysis of Cart expression in the pituitaries of adult and developing normal and mutant mice with hypopituitarism. We found that Prop1 is not necessary for initiation of Cart expression in the fetal pituitary at e14.5, but it is required indirectly for maintenance of Cart expression in the postnatal anterior pituitary gland. Pou1f1 deficiency has no effect on Cart expression before or after birth. There is no 1:1 correspondence between CART and any particular cell type. In neonates, CART is detected primarily in non-proliferating, POU1F1-positive cells. CART is also found in some cells that express TSH and GH suggesting a correspondence with committed progenitors of the POU1F1 lineage. In summary, we have characterized the normal temporal and cell specific expression of CART in mouse development and demonstrate that postnatal CART expression in the pituitary gland requires PROP1.

11.
Elife ; 52016 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-27351100

RESUMO

Mutations in PROP1 are the most common cause of hypopituitarism in humans; therefore, unraveling its mechanism of action is highly relevant from a therapeutic perspective. Our current understanding of the role of PROP1 in the pituitary gland is limited to the repression and activation of the pituitary transcription factor genes Hesx1 and Pou1f1, respectively. To elucidate the comprehensive PROP1-dependent gene regulatory network, we conducted genome-wide analysis of PROP1 DNA binding and effects on gene expression in mutant mice, mouse isolated stem cells and engineered mouse cell lines. We determined that PROP1 is essential for stimulating stem cells to undergo an epithelial to mesenchymal transition-like process necessary for cell migration and differentiation. Genomic profiling reveals that PROP1 binds to genes expressed in epithelial cells like Claudin 23, and to EMT inducer genes like Zeb2, Notch2 and Gli2. Zeb2 activation appears to be a key step in the EMT process. Our findings identify PROP1 as a central transcriptional component of pituitary stem cell differentiation.


Assuntos
Diferenciação Celular , Transição Epitelial-Mesenquimal , Proteínas de Homeodomínio/metabolismo , Células-Tronco/fisiologia , Animais , Linhagem Celular , Camundongos , Camundongos Knockout
12.
BMC Dev Biol ; 16(1): 16, 2016 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-27184910

RESUMO

BACKGROUND: The pituitary gland is a highly vascularized tissue that requires coordinated interactions between the neural ectoderm, oral ectoderm, and head mesenchyme during development for proper physiological function. The interactions between the neural ectoderm and oral ectoderm, especially the role of the pituitary organizer in shaping the pituitary precursor, Rathke's pouch, are well described. However, less is known about the role of head mesenchyme in pituitary organogenesis. The head mesenchyme is derived from definitive mesoderm and neural crest, but the relative contributions of these tissues to the mesenchyme adjacent to the pituitary are not known. RESULTS: We carried out lineage tracing experiments using two neural crest-specific mouse cre lines, Wnt1-cre and P0-cre, and determined that the head mesenchyme rostral to the pituitary gland is neural crest derived. To assess the role of the neural crest in pituitary development we ablated it, using Wnt1-cre to delete Ctnnb1 (ß-catenin), which is required for neural crest development. The Wnt1-cre is active in the neural ectoderm, principally in the mesencephalon, but also in the posterior diencephalon. Loss of ß-catenin in this domain causes a rostral shift in the ventral diencephalon, including the pituitary organizer, resulting in pituitary dysmorphology. The neural crest deficient embryos have abnormally dilated pituitary vasculature due to a loss of neural crest derived pericytes. CONCLUSIONS: ß-catenin in the Wnt1 expression domain, including the neural crest, plays a critical role in regulation of pituitary gland growth, development, and vascularization.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Mesencéfalo/metabolismo , Crista Neural/metabolismo , Organogênese/genética , Hipófise/metabolismo , beta Catenina/genética , Animais , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Feminino , Imuno-Histoquímica , Hibridização In Situ , Masculino , Mesencéfalo/embriologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microscopia de Fluorescência , Proteína P0 da Mielina/genética , Proteína P0 da Mielina/metabolismo , Crista Neural/embriologia , Hipófise/embriologia , Proteína Wnt1/genética , Proteína Wnt1/metabolismo , beta Catenina/metabolismo
13.
Hum Mol Genet ; 24(4): 939-53, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25315894

RESUMO

OTX2 is a homeodomain transcription factor that is necessary for normal head development in mouse and man. Heterozygosity for loss-of-function alleles causes an incompletely penetrant, haploinsufficiency disorder. Affected individuals exhibit a spectrum of features that range from developmental defects in eye and/or pituitary development to acephaly. To investigate the mechanism underlying the pituitary defects, we used different cre lines to inactivate Otx2 in early head development and in the prospective anterior and posterior lobes. Mice homozygous for Otx2 deficiency in early head development and pituitary oral ectoderm exhibit craniofacial defects and pituitary gland dysmorphology, but normal pituitary cell specification. The morphological defects mimic those observed in humans and mice with OTX2 heterozygous mutations. Mice homozygous for Otx2 deficiency in the pituitary neural ectoderm exhibited altered patterning of gene expression and ablation of FGF signaling. The posterior pituitary lobe and stalk, which normally arise from neural ectoderm, were extremely hypoplastic. Otx2 expression was intact in Rathke's pouch, the precursor to the anterior lobe, but the anterior lobe was hypoplastic. The lack of FGF signaling from the neural ectoderm was sufficient to impair anterior lobe growth, but not the differentiation of hormone-producing cells. This study demonstrates that Otx2 expression in the neural ectoderm is important intrinsically for the development of the posterior lobe and pituitary stalk, and it has significant extrinsic effects on anterior pituitary growth. Otx2 expression early in head development is important for establishing normal craniofacial features including development of the brain, eyes and pituitary gland.


Assuntos
Ectoderma/embriologia , Ectoderma/metabolismo , Deleção de Genes , Organogênese/genética , Fatores de Transcrição Otx/genética , Adeno-Hipófise/embriologia , Adeno-Hipófise/metabolismo , Animais , Proliferação de Células , Feminino , Fatores de Crescimento de Fibroblastos/metabolismo , Técnicas de Inativação de Genes , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipotálamo-Hipofisário/fisiologia , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , Mutação , Fatores de Transcrição Otx/metabolismo , Fenótipo , Adeno-Hipófise/patologia , Transdução de Sinais
14.
Curr Top Dev Biol ; 106: 1-47, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24290346

RESUMO

Many aspects of pituitary development have become better understood in the past two decades. The signaling pathways regulating pituitary growth and shape have emerged, and the balancing interactions between the pathways are now appreciated. Markers for multipotent progenitor cells are being identified, and signature transcription factors have been discovered for most hormone-producing cell types. We now realize that pulsatile hormone secretion involves a 3D integration of cellular networks. About a dozen genes are known to cause pituitary hypoplasia when mutated due to their essential roles in pituitary development. Similarly, a few genes are known that predispose to familial endocrine neoplasia, and several genes mutated in sporadic pituitary adenomas are documented. In the next decade, we anticipate gleaning a deeper appreciation of these processes at the molecular level, insight into the development of the hypophyseal portal blood system, and evolution of better therapeutics for congenital and acquired hormone deficiencies and for common craniopharyngiomas and pituitary adenomas.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Mutação , Doenças da Hipófise/genética , Hipófise/metabolismo , Animais , Humanos , Modelos Genéticos , Doenças da Hipófise/metabolismo , Doenças da Hipófise/fisiopatologia , Hipófise/crescimento & desenvolvimento , Hormônios Hipofisários/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Células-Tronco/citologia , Células-Tronco/metabolismo
15.
PLoS One ; 6(12): e28355, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22145038

RESUMO

Humans with PROP1 mutations have multiple pituitary hormone deficiencies (MPHD) that typically advance from growth insufficiency diagnosed in infancy to include more severe growth hormone (GH) deficiency and progressive reduction in other anterior pituitary hormones, eventually including adrenocorticotropic hormone (ACTH) deficiency and hypocortisolism. Congenital deficiencies of GH, prolactin, and thyroid stimulating hormone have been reported in the Prop1(null) (Prop1(-/-)) and the Ames dwarf (Prop1(df/df)) mouse models, but corticotroph and pituitary adrenal axis function have not been thoroughly investigated. Here we report that the C57BL6 background sensitizes mutants to a wasting phenotype that causes approximately one third to die precipitously between weaning and adulthood, while remaining homozygotes live with no signs of illness. The wasting phenotype is associated with severe hypoglycemia. Circulating ACTH and corticosterone levels are elevated in juvenile and aged Prop1 mutants, indicating activation of the pituitary-adrenal axis. Despite this, young adult Prop1 deficient mice are capable of responding to restraint stress with further elevation of ACTH and corticosterone. Low blood glucose, an expected side effect of GH deficiency, is likely responsible for the elevated corticosterone level. These studies suggest that the mouse model differs from the human patients who display progressive hormone loss and hypocortisolism.


Assuntos
Hormônio Adrenocorticotrópico/sangue , Envelhecimento/patologia , Nanismo/genética , Hormônio do Crescimento/deficiência , Proteínas de Homeodomínio/fisiologia , Animais , Glicemia/metabolismo , Corticosterona/sangue , Feminino , Homozigoto , Hipopituitarismo/genética , Técnicas Imunoenzimáticas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/genética , Fenótipo , Prolactina/deficiência , Radioimunoensaio
16.
Genetics ; 189(2): 665-73, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21840860

RESUMO

Thyroid hormone has pleiotropic effects on cochlear development, and genomic variation influences the severity of associated hearing deficits. DW/J-Pou1f1dw/dw mutant mice lack pituitary thyrotropin, which causes severe thyroid hormone deficiency and profound hearing impairment. To assess the genetic complexity of protective effects on hypothyroidism-induced hearing impairment, an F1 intercross was generated between DW/J-Pou1f1dw/+ carriers and an inbred strain with excellent hearing derived from Mus castaneus, CAST/EiJ. Approximately 24% of the (DW/J×CAST/EiJ) Pou1f1dw/dw F2 progeny had normal hearing. A genome scan revealed a locus on chromosome 2, named modifier of dw hearing, or Mdwh, that rescues hearing despite persistent hypothyroidism. This chromosomal region contains the modifier of tubby hearing 1 (Moth1) locus that encodes a protective allele of the microtubule-associated protein MTAP1A. DW/J-Pou1f1dw/+ carriers were crossed with the AKR strain, which also carries a protective allele of Mtap1a, and we found that AKR is not protective for hearing in the (DW/J×AKR) Pou1f1dw/dw F2 progeny. Thus, protective alleles of Mtap1a are not sufficient to rescue DW/J-Pou1f1dw/dw hearing. We expect that identification of protective modifiers will enhance our understanding of the mechanisms of hypothyroidism-induced hearing impairment.


Assuntos
Genes Modificadores/genética , Perda Auditiva/genética , Hipotireoidismo/complicações , Fator de Transcrição Pit-1/genética , Alelos , Animais , Sequência de Bases , Mapeamento Cromossômico , Cromossomos de Mamíferos/genética , Cruzamentos Genéticos , Feminino , Predisposição Genética para Doença/genética , Perda Auditiva/etiologia , Masculino , Camundongos , Camundongos Endogâmicos AKR , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Camundongos Mutantes , Proteínas Associadas aos Microtúbulos/genética , Dados de Sequência Molecular , Polimorfismo de Nucleotídeo Único , Locos de Características Quantitativas/genética , Fator de Transcrição Pit-1/deficiência
17.
Physiol Genomics ; 43(19): 1105-16, 2011 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-21828248

RESUMO

Mutations in the transcription factors PROP1 and PIT1 (POU1F1) lead to pituitary hormone deficiency and hypopituitarism in mice and humans. The dysmorphology of developing Prop1 mutant pituitaries readily distinguishes them from those of Pit1 mutants and normal mice. This and other features suggest that Prop1 controls the expression of genes besides Pit1 that are important for pituitary cell migration, survival, and differentiation. To identify genes involved in these processes we used microarray analysis of gene expression to compare pituitary RNA from newborn Prop1 and Pit1 mutants and wild-type littermates. Significant differences in gene expression were noted between each mutant and their normal littermates, as well as between Prop1 and Pit1 mutants. Otx2, a gene critical for normal eye and pituitary development in humans and mice, exhibited elevated expression specifically in Prop1 mutant pituitaries. We report the spatial and temporal regulation of Otx2 in normal mice and Prop1 mutants, and the results suggest Otx2 could influence pituitary development by affecting signaling from the ventral diencephalon and regulation of gene expression in Rathke's pouch. The discovery that Otx2 expression is affected by Prop1 deficiency provides support for our hypothesis that identifying molecular differences in mutants will contribute to understanding the molecular mechanisms that control pituitary organogenesis and lead to human pituitary disease.


Assuntos
Perfilação da Expressão Gênica/métodos , Proteínas de Homeodomínio/metabolismo , Hipopituitarismo/metabolismo , Fatores de Transcrição Otx/metabolismo , Fator de Transcrição Pit-1/metabolismo , Animais , Feminino , Genótipo , Proteínas de Homeodomínio/genética , Hipopituitarismo/genética , Imuno-Histoquímica , Hibridização In Situ , Masculino , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Fatores de Transcrição Otx/genética , Reação em Cadeia da Polimerase , Gravidez , Fator de Transcrição Pit-1/genética
18.
Dev Biol ; 352(2): 215-27, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21262217

RESUMO

The intermediate and anterior lobes of the pituitary gland are derived from an invagination of oral ectoderm that forms Rathke's pouch. During gestation proliferating cells are enriched around the pouch lumen, and they appear to delaminate as they exit the cell cycle and differentiate. During late mouse gestation and the postnatal period, anterior lobe progenitors re-enter the cell cycle and expand the populations of specialized, hormone-producing cells. At birth, all cell types are present, and their localization appears stratified based on cell type. We conducted a birth dating study of Rathke's pouch derivatives to determine whether the location of specialized cells at birth is correlated with the timing of cell cycle exit. We find that all of the anterior lobe cell types initiate differentiation concurrently with a peak between e11.5 and e13.5. Differentiation of intermediate lobe melanotropes is delayed relative to anterior lobe cell types. We discovered that specialized cell types are not grouped together based on birth date and are dispersed throughout the anterior lobe. Thus, the apparent stratification of specialized cells at birth is not correlated with cell cycle exit. Thus, the currently popular model of cell specification, dependent upon timing of extrinsic, directional gradients of signaling molecules, needs revision. We propose that signals intrinsic to Rathke's pouch are necessary for cell specification between e11.5 and e13.5 and that cell-cell communication likely plays an important role in regulating this process.


Assuntos
Modelos Neurológicos , Hipófise/embriologia , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Ciclo Celular , Diferenciação Celular , Proliferação de Células , Células-Tronco Embrionárias/citologia , Feminino , Fatores de Crescimento de Fibroblastos/metabolismo , Idade Gestacional , Camundongos , Hipófise/citologia , Hipófise/fisiologia , Adeno-Hipófise/citologia , Adeno-Hipófise/embriologia , Adeno-Hipófise/fisiologia , Hormônios Hipofisários/biossíntese , Gravidez , Transdução de Sinais
19.
Horm Res ; 71 Suppl 2: 101-15, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19407506

RESUMO

Genetic cases of congenital pituitary hormone deficiency are common and many are caused by transcription factor defects. Mouse models with orthologous mutations are invaluable for uncovering the molecular mechanisms that lead to problems in organ development and typical patient characteristics. We are using mutant mice defective in the transcription factors PROP1 and POU1F1 for gene expression profiling to identify target genes for these critical transcription factors and candidates for cases of pituitary hormone deficiency of unknown aetiology. These studies reveal critical roles for Wnt signalling pathways, including the TCF/LEF transcription factors and interacting proteins of the groucho family, bone morphogenetic protein antagonists and targets of notch signalling. Current studies are investigating the roles of novel homeobox genes and pathways that regulate the transition from proliferation to differentiation, cell adhesion and cell migration. Pituitary adenomas are a common human health problem, yet most cases are sporadic, necessitating alternative approaches to traditional Mendelian genetic studies. Mouse models of adenoma formation offer the opportunity for gene expression profiling during progressive stages of hyperplasia, adenoma and tumorigenesis. This approach holds promise for the identification of relevant pathways and candidate genes as risk factors for adenoma formation, understanding mechanisms of progression, and identifying drug targets and clinically relevant biomarkers.


Assuntos
Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Regulação Neoplásica da Expressão Gênica/genética , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Hipófise/metabolismo , Neoplasias Hipofisárias/genética , Neoplasias Hipofisárias/metabolismo , Animais , Biologia Computacional/métodos , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Camundongos , Camundongos Mutantes , Hipófise/patologia , Hormônios Hipofisários/deficiência , Hormônios Hipofisários/genética , Hormônios Hipofisários/metabolismo , Neoplasias Hipofisárias/patologia
20.
Genesis ; 46(10): 507-14, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18802953

RESUMO

This report introduces a gonadotrope-specific cre transgenic mouse capable of ablating floxed genes in mature pituitary gonadotropes. Initial analysis of this transgenic line, Tg(Lhb-cre)1Sac, reveals that expression is limited to the pituitary cells that produce luteinizing hormone beta, beginning appropriately at e17.5. Cre activity is detectable by a reporter gene in nearly every LHbeta-producing cell, but the remaining hormone-producing cell types and other organs exhibit little to no activity. We used the Tg(Lhb-cre)1Sac strain to assess the role Pitx2 in gonadotrope function. The gonadotrope-specific Pitx2 knockout mice exhibit normal expression of LHbeta, sexual maturation, and fertility, suggesting that Pitx2 is not required for gonadotrope maintenance or for regulated production of gonadotropins.


Assuntos
Fertilidade/genética , Deleção de Genes , Gonadotrofos/metabolismo , Gônadas/embriologia , Proteínas de Homeodomínio/genética , Adeno-Hipófise/metabolismo , Fatores de Transcrição/genética , Animais , Feminino , Gônadas/metabolismo , Integrases/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Adeno-Hipófise/citologia , Hormônios Hipofisários/biossíntese , Proteína Homeobox PITX2
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA