• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 30
  • 22
  • 9
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • Tagged with
  • 64
  • 48
  • 42
  • 20
  • 18
  • 17
  • 15
  • 14
  • 14
  • 14
  • 12
  • 12
  • 11
  • 10
  • 10
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
21

Studies of immunological risk factors in type 1 diabetes /

Walldén, Jenny, January 2008 (has links) (PDF)
Diss. (sammanfattning) Linköping : Linköpings universitet, 2008. / Härtill 4 uppsatser.
22

The role of endoplasmic reticulum stress signaling in isolated islet apoptosis

Park, Soon Hyang. January 1900 (has links)
Thesis (M.Sc.). / Written for the Dept. of Surgery, Division of Surgical Research. Title from title page of PDF (viewed 2009/06/30). Includes bibliographical references.
23

ExpressÃo dos genes GNAS e BTG2 e de um painel de microRNAs em somatotrofinomas esporÃdicos com e sem mutaÃÃo no gene GNAS

Ana Rosa Pinto Quidute 18 October 2013 (has links)
nÃo hà / IntroduÃÃo: MutaÃÃes nos genes GNAS e AIP estÃo presentes em 35% e 3%, respectivamente, dos somatotrofinomas esporÃdicos. Recentemente, observa-se importÃncia biolÃgica crescente dos microRNAs (miRNAs) na tumorigÃnese hipofisÃria. Entretanto, mecanismos moleculares envolvidos na patogÃnese de 60% desses tumores permanecem nÃo elucidados. Objetivos: Identificar a prevalÃncia de mutaÃÃes nos genes GNAS e AIP em um grupo de somatotrofinomas esporÃdicos. Comparar parÃmetros clÃnicos e bioquÃmicos ao diagnÃstico como idade, tamanho tumoral e agressividade (escore Hardy), hormÃnio do crescimento (GH), prolactina (PRL) e Fator de Crescimento Insulin-Like I (IGF-1) e resposta as terapÃuticas entre os grupos com (gsp+) e sem (gsp-) mutaÃÃo no GNAS. Analisar a expressÃo dos genes GNAS e BTG2 e miRNAs entre somatotrofinomas e hipÃfises normais (HN) e a associaÃÃo entre a expressÃo com agressividade, a resposta à cirurgia e a todas as terapÃuticas adjuvantes disponÃveis. Material e MÃtodos: 26 pacientes com diagnÃstico de acromegalia. Tamanho tumoral foi avaliado por RNM/CT e o grau de invasibilidade pelo escore de Hardy (I a IV). GH basal ≤2.5μg/L ou nadir de GH apÃs o GTT≤1μg/L e IGF-1 normal para idade e sexo foram utilizados como critÃrio de cura apÃs cirurgia transesfenoidal (CTE). Como controle com o anÃlogo da somatostatina (AS), adotamos a normalizaÃÃo dos nÃveis de IGF-1 para idade e sexo. As amostras tumorais (n=26) foram obtidas durante a CTE, realizado histopatolÃgico e armazenadas a -70 ÂC, para estudos moleculares. HN (07) foram obtidas durante autÃpsias. RNA e DNA total foram extraÃdos pelo TRIzolÂ. Os cÃdons 201 e 227 do gene GNAS e o AIP completo foram sequenciados. ExpressÃo relativa dos genes GNAS e BTG2 e dos miRNAs let-7a, miR-16a, miR-21, miR-141, miR-143, miR-15a, miR-145, miR-23a, miR-23b e miR-24-2 foi avaliada por qPCR (sondas TaqMan), pelo mÃtodo 2-ΔΔCt. Resultados: A frequÃncia de mutaÃÃes no GNAS foi de 35% e no AIP 3,8%. NÃo houve diferenÃa entre as mÃdias de idade (39,0Â11,5 vs 43,6Â9,0 anos; p=0,32), nas concentraÃÃes plasmÃticas basais de GH (62,4Â128,1 vs 39,9Â48,3Âg/L; p=0,39), IGF-1 (435,5Â230,8 vs 556,9 238,3 %ULNR; p=0,32), PRL (25,7Â29,8 vs 30,9Â32,8 ng/L; p=0,69) e agressividade tumoral entre os gsp+ e gsp-(p=1,00). Ao analisar o uso do AS como terapÃutica adjuvante à CTE, observamos que 04/05 (80%) dos indivÃduos com somatotrofinoma gsp+ obtiveram controle da doenÃa, enquanto que no grupo gsp- 02/06 (33%) obtiveram controle (p=0,08). Quando associamos ao AS, os agonistas dopaminÃrgicos e/ou radioterapia externa, observamos que 05/05 (100%) dos pacientes gsp+ tiveram critÃrio de controle da doenÃa, contra (04/09) 44% no grupo gsp- (p=0,09). NÃo houve diferenÃa na expressÃo de GNAS entre os somatotrofinomas e as HN (1,07Â0,55 vs 0,98Â0,28; p=0,97), e entre os gsp+ e gsp- (1,04Â0,59 vs 1,10Â0,55; p=0,97, respectivamente). Os tumores Hardy I / II apresentaram maior expressÃo do GNAS do que os tumores classificados como III / IV (p=0,02). NÃo houve associaÃÃo entre a expressÃo do GNAS e o controle da doenÃa com cirurgia isolada ou com o uso de todas as terapÃuticas adjuvantes. Observamos hipoexpressÃo do BTG2 e dos miR-16a e miR-141 em somatotrofinomas quando foram comparados com as HN (p=0,002, fold=-6,63; p=0,01, fold=-10,00; p=0,0003, fold=-50,00, respectivamente) sem diferenÃas entre os gsp+ e gsp-. Houve hiperexpressÃo do miR-21 (p=0,02;fold=10,18) em somatotrofinomas (20,16Â18,48) quando comparado com as HN (2,52 Â3,56), sem diferenÃa entre os gsp + e gsp-. NÃo houve diferenÃa na expressÃo entre os grupos gsp+ e gsp- para os miRNAs let-7a, miR-21, miR-143, miR-15a, miR-23a e miR-24-2. Entretanto, miR-145 e miR-23b foram mais hipoexpressos no grupo gsp+ quando comparados ao gsp- (p=0,03, fold=-4,83 e p=0,02, fold=-2,77, respectivamente). NÃo houve associaÃÃo entre a expressÃo do BTG2 e o painel de miRNAs com agressividade e com o controle da doenÃa. ConclusÃo: Na presente sÃrie de somatotrofinomas, assumidos como esporÃdicos, a frequÃncia de mutaÃÃes nos genes GNAS (35%) e AIP (3,8%) foram semelhantes aos relatados na literatura. NÃo houve diferenÃas nas caracterÃsticas clÃnicas e bioquÃmicas, agressividade, resposta Ãs terapÃuticas, e na expressÃo diferencial do GNAS entre os pacientes com tumores gsp+ e gsp-. HipoexpressÃo de BTG2 (gene supressor tumoral relacionado Ãs vias de sinalizaÃÃo do p53 e do Rb), baixa expressÃo de miRNAs (supressores tumorais) e alta expressÃo de oncomirs em somatotrofinomas sugerem um papel desses na tumorigÃnese somatotrÃfica. / Introduction: Mutations in GNAS and AIP genes are present in 35% and 3%, respectively, of the sporadic somatotropinomas. Recently, increased biological importance of microRNAs (miRNAs) has been observed in pituitary tumorigenesis. However, the molecular mechanisms involved in the pathogenesis of 60% of these tumors remain to be elucidated. Objectives: To identify the prevalence of mutations in GNAS and AIP genes in a series of sporadic somatotropinomas. Compare clinical, bioquimical parametrer at diagnosis as age, tumor size and theirs aggressiveness, pre-operative growth hormone (GH), prolactin (PRL) and insulin-like growth factor-I (IGF-1) levels and treatment responsiveness between somatotropinomas with (gsp+) and without (gsp-) GNAS mutation.To analyze the expression of GNAS and BTG2 genes and a panel of miRNAs between somatotrofinomas and normal pituitaries (NP) and the association between the expression of these genes and miRNAs with aggressiveness, as well as disease control with surgery or control with all adjuvant therapeutic approaches. Material and Methods: 26 patients with acromegaly. GH basal ≤2.5μg/L or nadir after OGTT ≤1μg/L and normal IGF-I matched for age and sex were used as diagnosis and for cure criteria after transsphenoidal surgery (TS). As control after somatostatin analogues (SA), we adopted the normalization of IGF-I matched for age and sex. Tumor size was evaluated by MRI/CT and the degree of invasiveness by Hardy score (I to IV).Tumor samples (26) were obtained during TS, processed for histopathology and stored at -70ÂC for molecular studies. NP (07) were obtained during autopsy. Total DNA and RNA were extracted by TRIzolÂ. Codons 201 and 227 of the GNAS gene and the whole AIP gene were sequenced. Relative expression of BTG2 and GNAS genes and miRNAs let-7a, miR-16a, miR-21, miR-141, miR-143, miR-15a, miR-145, miR-23a, miR-23b, and miR-24-2 was measured by qPCR (TaqMan probes) using 2-ΔΔCt method. Results: Frequencies of GNAS and AIP mutations were 35% and 3.8%, respectively. There was no difference between the mean age (39.0  11.5 vs 43.6  9.0 years, p=0.32), basal GH (62.4Â128.1 vs 39.9  48.3 μg/L; p=0.39), IGF-I (435.5  230.8 vs. 556.9  238.3; p=0.32) and PRL (25.7  29.8 vs. 30.9  32.8 ng/L, p=0.69) in plasma concentration, and tumor aggressiveness (p=1.00) between (gsp+) and (gsp-) groups. We observed that 80% (04/05) of gsp+ whereas 33% (02/06) of the gsp- achieved control (p=0.07) after SA therapy adjuvant to TS. When SA, dopamine agonists and/or external radiotherapy were associated 100% (05/05) of gsp+ group and 44% (04/09) of gsp- group (p=0.08) showed disease control.There was no difference in GNAS expression between somatotropinomas and NP (1.07  0.55 vs 0.98  0.28, p=0.97) as well as between somatotropinomasgsp+ and gsp- (1.04  0.59 vs 1.10  0.55, p=0.97, respectively). Hardy I/II tumors showed higher GNAS expression than Hardy III/IV (p=0.02), but there was no association between GNAS expression and disease control with surgery alone or associated with other adjuvant therapies. We observed hypoexpression of BTG2 and miR-16a and miR-141 in somatotropinomas compared with NP (-6.6 fold, p=0.002; -10.0 fold, p=0.01; and -50.0 fold, p=0.0003, respectively) with no difference between gsp+ and gsp- somatotropinomas. There was miR-21 overexpression in somatotropinomas compared with NP (20.2  18.5 vs 2.5  3.6; 10.2 fold, p=0.02), with no difference between gsp+ and gsp- somatotropinomas. However, miR-145 and miR-23b were more hipoexpressed in gsp+ compared to gsp- (-4.8fold, p=0.03 and-2.7 fold, p=0.02). There was no association between the expression of BTG2 and a panel of miRNAs with aggressiveness or disease control. Conclusion: In this series of assumed sporadic somatotopinomas, the frequencies of mutations in GNAS (35%) and AIP (3.8%) were similar to the literature. There were no differences in clinical and biochemical characteristics, aggressiveness, response to therapy, and GNAS expression in patients with gsp+ and gsp- somatotropinomas. Hypoexpression of BTG2, a tumor suppressor gene related to p53 and Rb signaling pathways, low expression of tumor suppressor miRNAs and high expression of oncomirs in somatotropinomas suggest a role in the somatotrophic tumorigenesis.
24

The role of the growth hormone/IGF-I system on islet cell growth and insulin action /

Robertson, Katherine. January 2007 (has links)
No description available.
25

Pathogenicity of IgG-Fc desialylation and its association with Th17 cells in an animal model of systemic lupus erythematosus / 全身性エリテマトーデスの動物モデルにおけるIgG-Fc脱シアル化の病原性とTh17細胞との関連

Nishida, Yuri 23 January 2024 (has links)
京都大学 / 新制・課程博士 / 博士(医学) / 甲第24994号 / 医博第5028号 / 新制||医||1069(附属図書館) / 京都大学大学院医学研究科医学専攻 / (主査)教授 上野 英樹, 教授 椛島 健治, 教授 濵﨑 洋子 / 学位規則第4条第1項該当 / Doctor of Medical Science / Kyoto University / DFAM
26

Estudo molecular dos genes GNAS, PTTG, AIP, CDKN1B e MEG3 em adenomas hipofisários esporádicos / Molecular study of GNAS, PTTG, AIP, CDKN1B and MEG3 genes in sporadic pituitary adenomas

Foltran, Renata Kikuchi 26 February 2016 (has links)
INTRODUÇÃO: Os adenomas hipofisários são neoplasias benignas que representam cerca de 15% das neoplasias intracranianas. Em sua maioria ocorre de forma esporádica. Estudos moleculares desses adenomas identificaram anormalidades genéticas que podem ter um papel na sua tumorigênese. Dentre alguns desses genes foram descritos os oncogenes GNAS e PTTG e os genes supressores tumorais AIP, CDKN1B e MEG3. OBJETIVO: realizar estudo molecular dos genes associados a tumorigênese através da pesquisa de mutações nos genes GNAS, AIP e CDKN1B e o estudo de expressão gênica de CDKN1B, PTTG e MEG3 em adenomas aparentemente esporádicos, correlacionando com os dados clínicos e laboratoriais, em pacientes acompanhados no serviço de Endocrinologia do HCFMUSP. CASUÍSTICA E MÉTODOS: Compreendeu 96 adenomas hipofisários aparentemente esporádicos: 41 somatotropinomas, 27 corticotropinomas, 21 adenomas clinicamente não funcionantes (ACNF) e 7 prolactinomas. Foi realizada avaliação restrospectiva dos dados clínicos e laboratoriais ao diagnóstico. Após a análise histológica por hematoxilinaeosina, foi realizada análise imunohistoquímica das proteínas Ki-67 e p53 e molecular do DNA genômico e RNA, extraídos do tecido tumoral. Análise mutacional das regiões codificantes de AIP e CDKN1B e dos hotspots de GNAS nos éxons 8 e 9 foi realizada através de amplificação por PCR e sequenciamento automático. A quantificação relativa do RNAm de CDKN1B, MEG3 e PTTG foi avaliada pelo método de 2-??Ct por PCR em tempo real. RESULTADOS: Presença de mutações somáticas no gene GNAS (gsp+) em 14,5% dos adenomas. Não houve diferenças significativas clínicas e laboratoriais entre os adenomas gsp+ e gsp-. Variantes com potencial patogênico não foram identificadas nos genes AIP e CDKN1B. A análise imunohistoquímica do Ki-67 apresentou média de 1,32% (0,9-4,5) e do p53 média de 1,04 (1,0-1,8). O gene CDKN1B apresentou expressão média de ,12 ± 0,74 (0,1-3,1), com expressão mais baixa nos corticotropinomas. O gene PTTG apresentou expressão média de 2,49 ± 3,10 (0,2-19,0), com maior expressão nos corticotropinomas. O gene MEG3 apresentou expressão média de 0,95 ± 1,38 (0,0-8,8), com valores mais baixos nos ACNF. Três padrões de cluster nos níveis de expressão de RNAm dos genes CDKN1B, PTTG e MEG3 foram identificados: cluster A = CDKN1B >= 1,85/ PTTG >= 1,25/ MEG3 >= 0,65 foi observado em 100% dos corticotropinomas; cluster B= CDKN1B >= 0,95/ PTTG >= 2,25/ MEG3 >= 0,65 observado apenas nos somatotropinomas (32%) e o cluster C= CDKN1B >= 0,95/ PTTG >= 1,25/ MEG3 >= 0,05 observado na maioria dos ACNF (73%). CONCLUSÕES: A maioria dos adenomas apresentaram índices de Ki-67 menor do que 3%. Em conformidade com este achado, a imunohistoquímica para p53 não se mostrou estatisticamente significativa. A mutação ativadora na proteína Gs? (gsp+) foi a mutação mais frequente em adenomas hipofisários esporádicos, principalmente em somatotropinomas. Não foram identificadas variantes com potencial patogênico nos genes AIP e CDKN1B, portanto, parece ser um evento raro em adenomas esporádicos. A expressão gênica aumentada do gene PTTG foi identificada principalmente nos corticotropinomas. No entanto, ela não foi preditiva de subtipo de adenoma. A expressão gênica do CDKN1B estava diminuída na maioria dos corticotropinomas e normal na maioria dos somatotropinomas e ACNF. A expressão gênica do MEG3 estava diminuída na maioria dos adenomas ACNF e corticotropinomas e normal na maioria dos somatotropinomas. Na análise de cluster hierárquico, foram identificados três padrões de expressão gênica que se correlacionaram com subtipo de adenoma hipofisário / BACKGROUND: Pituitary adenomas are benign tumors that account for about 15% of intracranial tumors. Mostly occurs sporadically. Molecular studies of these adenomas identified genetic abnormalities that may have a role in tumorigenesis. Some of these genes have been described as the oncogenes GNAS and PTTG and tumor suppressor genes AIP, CDKN1B and MEG3. OBJECTIVE: perform a molecular study of genes related in tumorigenesis to evaluate presence of mutations in GNAS, AIP and CDKN1B genes and gene expression analysis of CDKN1B, PTTG and MEG3 genes in apparently sporadic adenomas, correlating with the clinical and laboratory data from patients treated at the Endocrinology service of HCFMUSP.SUBJECTS AND METHODS: 96 apparently sporadic adenomas was included: 41 somatotropinomas, 27 corticotropinomas, 21 clinically nonfunctioning pituitary adenomas (NFPA) and seven prolactinomas. Retrospective evaluation of clinical and laboratory data from diagnosis. After histological analysis by hematoxylin-eosin staining, it was performed immunohistochemical analysis of Ki -67 and p53 proteins and molecular analysis of genomic DNA and RNA extracted from tumor tissue. Mutational analysis of coding regions of AIP and CDKN1B and hotspots exons 8 and 9 of GNAS was performed by PCR and automatic sequencing. Relative quantification of mRNA CDKN1B, MEG3 and PTTG was evaluated by 2-??Ct method using Real Time PCR. RESULTS: Presence of somatic mutations on GNAS gene (gsp+) in 14,5% of pituitary adenomas. There were no clinical and laboratorial differences between gsp+ and gsp- somatotropinomas. Variants with pathogenic potencial were not identified in AIP and CDKN1B genes. Imunohistochemical analysis showed mean of 1,32% (0,9-4,5) for Ki-67 and mean of 1,04% (1,0-1,8) for p53. Gene expression of CDKN1B presented a mean of 1,12 ± 0,74 (0,1-3,1) with lower expression in corticotropinomas. Gene expression of PTTG presented a mean of 2,49 ± 3,10 (0,2-19,0) with higher expression in corticotropinomas. Gene expression of MEG3 presented a mean of 0,95 ± 1,38 (0,0-8,8) with lower expression in NFPA. Three cluster patterns in the levels of mRNA expression of genes CDKN1B, PTTG and MEG3 were identified: cluster A = CDKN1B >= 1,85/ PTTG >= 1,25/ MEG3 >= 0,65 observed in 100% of corticotropinomas; cluster B= CDKN1B >= 0,95/ PTTG >= 2,25/ MEG3 >= 0,65 observed only in somatotropinomas (32%) and cluster C= CDKN1B >= 0,95/ PTTG >= 1,25/ MEG3 >= 0,05 observed in most of NFPA (73%). CONCLUSIONS: Most of the adenomas showed Ki -67 index lower than 3%. In accordance with this finding, immunohistochemistry for p53 was not statistically significant. The activating mutation in the Gs? protein (gsp+) was the most common mutation in sporadic pituitary adenomas, particularly in somatotropinomas. Variants with pathogenic potential have not been identified in the AIP and CDKN1B gene therefore seems to be a rare event in sporadic adenomas. Increased gene expression of PTTG was primarily identified in corticotropinomas. However, it was not predictive of adenoma subtype. The gene expression of CDKN1B was decreased in most corticotropinomas and normal in most somatotropinomas and NFPA. The gene expression of MEG3 was decreased in most of NFPA and corticotropinomas, and normal in most somatotropinomas. In hierarchical cluster analysis was identified three patterns of gene expression that correlated with pituitary adenoma subtype
27

Estudo molecular dos genes GNAS, PTTG, AIP, CDKN1B e MEG3 em adenomas hipofisários esporádicos / Molecular study of GNAS, PTTG, AIP, CDKN1B and MEG3 genes in sporadic pituitary adenomas

Renata Kikuchi Foltran 26 February 2016 (has links)
INTRODUÇÃO: Os adenomas hipofisários são neoplasias benignas que representam cerca de 15% das neoplasias intracranianas. Em sua maioria ocorre de forma esporádica. Estudos moleculares desses adenomas identificaram anormalidades genéticas que podem ter um papel na sua tumorigênese. Dentre alguns desses genes foram descritos os oncogenes GNAS e PTTG e os genes supressores tumorais AIP, CDKN1B e MEG3. OBJETIVO: realizar estudo molecular dos genes associados a tumorigênese através da pesquisa de mutações nos genes GNAS, AIP e CDKN1B e o estudo de expressão gênica de CDKN1B, PTTG e MEG3 em adenomas aparentemente esporádicos, correlacionando com os dados clínicos e laboratoriais, em pacientes acompanhados no serviço de Endocrinologia do HCFMUSP. CASUÍSTICA E MÉTODOS: Compreendeu 96 adenomas hipofisários aparentemente esporádicos: 41 somatotropinomas, 27 corticotropinomas, 21 adenomas clinicamente não funcionantes (ACNF) e 7 prolactinomas. Foi realizada avaliação restrospectiva dos dados clínicos e laboratoriais ao diagnóstico. Após a análise histológica por hematoxilinaeosina, foi realizada análise imunohistoquímica das proteínas Ki-67 e p53 e molecular do DNA genômico e RNA, extraídos do tecido tumoral. Análise mutacional das regiões codificantes de AIP e CDKN1B e dos hotspots de GNAS nos éxons 8 e 9 foi realizada através de amplificação por PCR e sequenciamento automático. A quantificação relativa do RNAm de CDKN1B, MEG3 e PTTG foi avaliada pelo método de 2-??Ct por PCR em tempo real. RESULTADOS: Presença de mutações somáticas no gene GNAS (gsp+) em 14,5% dos adenomas. Não houve diferenças significativas clínicas e laboratoriais entre os adenomas gsp+ e gsp-. Variantes com potencial patogênico não foram identificadas nos genes AIP e CDKN1B. A análise imunohistoquímica do Ki-67 apresentou média de 1,32% (0,9-4,5) e do p53 média de 1,04 (1,0-1,8). O gene CDKN1B apresentou expressão média de ,12 ± 0,74 (0,1-3,1), com expressão mais baixa nos corticotropinomas. O gene PTTG apresentou expressão média de 2,49 ± 3,10 (0,2-19,0), com maior expressão nos corticotropinomas. O gene MEG3 apresentou expressão média de 0,95 ± 1,38 (0,0-8,8), com valores mais baixos nos ACNF. Três padrões de cluster nos níveis de expressão de RNAm dos genes CDKN1B, PTTG e MEG3 foram identificados: cluster A = CDKN1B >= 1,85/ PTTG >= 1,25/ MEG3 >= 0,65 foi observado em 100% dos corticotropinomas; cluster B= CDKN1B >= 0,95/ PTTG >= 2,25/ MEG3 >= 0,65 observado apenas nos somatotropinomas (32%) e o cluster C= CDKN1B >= 0,95/ PTTG >= 1,25/ MEG3 >= 0,05 observado na maioria dos ACNF (73%). CONCLUSÕES: A maioria dos adenomas apresentaram índices de Ki-67 menor do que 3%. Em conformidade com este achado, a imunohistoquímica para p53 não se mostrou estatisticamente significativa. A mutação ativadora na proteína Gs? (gsp+) foi a mutação mais frequente em adenomas hipofisários esporádicos, principalmente em somatotropinomas. Não foram identificadas variantes com potencial patogênico nos genes AIP e CDKN1B, portanto, parece ser um evento raro em adenomas esporádicos. A expressão gênica aumentada do gene PTTG foi identificada principalmente nos corticotropinomas. No entanto, ela não foi preditiva de subtipo de adenoma. A expressão gênica do CDKN1B estava diminuída na maioria dos corticotropinomas e normal na maioria dos somatotropinomas e ACNF. A expressão gênica do MEG3 estava diminuída na maioria dos adenomas ACNF e corticotropinomas e normal na maioria dos somatotropinomas. Na análise de cluster hierárquico, foram identificados três padrões de expressão gênica que se correlacionaram com subtipo de adenoma hipofisário / BACKGROUND: Pituitary adenomas are benign tumors that account for about 15% of intracranial tumors. Mostly occurs sporadically. Molecular studies of these adenomas identified genetic abnormalities that may have a role in tumorigenesis. Some of these genes have been described as the oncogenes GNAS and PTTG and tumor suppressor genes AIP, CDKN1B and MEG3. OBJECTIVE: perform a molecular study of genes related in tumorigenesis to evaluate presence of mutations in GNAS, AIP and CDKN1B genes and gene expression analysis of CDKN1B, PTTG and MEG3 genes in apparently sporadic adenomas, correlating with the clinical and laboratory data from patients treated at the Endocrinology service of HCFMUSP.SUBJECTS AND METHODS: 96 apparently sporadic adenomas was included: 41 somatotropinomas, 27 corticotropinomas, 21 clinically nonfunctioning pituitary adenomas (NFPA) and seven prolactinomas. Retrospective evaluation of clinical and laboratory data from diagnosis. After histological analysis by hematoxylin-eosin staining, it was performed immunohistochemical analysis of Ki -67 and p53 proteins and molecular analysis of genomic DNA and RNA extracted from tumor tissue. Mutational analysis of coding regions of AIP and CDKN1B and hotspots exons 8 and 9 of GNAS was performed by PCR and automatic sequencing. Relative quantification of mRNA CDKN1B, MEG3 and PTTG was evaluated by 2-??Ct method using Real Time PCR. RESULTS: Presence of somatic mutations on GNAS gene (gsp+) in 14,5% of pituitary adenomas. There were no clinical and laboratorial differences between gsp+ and gsp- somatotropinomas. Variants with pathogenic potencial were not identified in AIP and CDKN1B genes. Imunohistochemical analysis showed mean of 1,32% (0,9-4,5) for Ki-67 and mean of 1,04% (1,0-1,8) for p53. Gene expression of CDKN1B presented a mean of 1,12 ± 0,74 (0,1-3,1) with lower expression in corticotropinomas. Gene expression of PTTG presented a mean of 2,49 ± 3,10 (0,2-19,0) with higher expression in corticotropinomas. Gene expression of MEG3 presented a mean of 0,95 ± 1,38 (0,0-8,8) with lower expression in NFPA. Three cluster patterns in the levels of mRNA expression of genes CDKN1B, PTTG and MEG3 were identified: cluster A = CDKN1B >= 1,85/ PTTG >= 1,25/ MEG3 >= 0,65 observed in 100% of corticotropinomas; cluster B= CDKN1B >= 0,95/ PTTG >= 2,25/ MEG3 >= 0,65 observed only in somatotropinomas (32%) and cluster C= CDKN1B >= 0,95/ PTTG >= 1,25/ MEG3 >= 0,05 observed in most of NFPA (73%). CONCLUSIONS: Most of the adenomas showed Ki -67 index lower than 3%. In accordance with this finding, immunohistochemistry for p53 was not statistically significant. The activating mutation in the Gs? protein (gsp+) was the most common mutation in sporadic pituitary adenomas, particularly in somatotropinomas. Variants with pathogenic potential have not been identified in the AIP and CDKN1B gene therefore seems to be a rare event in sporadic adenomas. Increased gene expression of PTTG was primarily identified in corticotropinomas. However, it was not predictive of adenoma subtype. The gene expression of CDKN1B was decreased in most corticotropinomas and normal in most somatotropinomas and NFPA. The gene expression of MEG3 was decreased in most of NFPA and corticotropinomas, and normal in most somatotropinomas. In hierarchical cluster analysis was identified three patterns of gene expression that correlated with pituitary adenoma subtype
28

Análogos da somatostatina na acromegalia: comparação da resposta clínica, laboratorial e do volume tumoral com a expressão dos subtipos dos receptores de somatostatina no tumor somatotrófico / Somatostatin analogs in acromegaly: comparison of clinic response, laboratory and tumor volume with expression of somatostatin receptor subtype in somatotroph tumor

Casarini, Ana Paula Malinosk 13 August 2008 (has links)
Este estudo analisa a expressão dos subtipos de receptores da somatostatina (SSTR) em 39 adenomas secretores de GH. Em 19 pacientes acromegálicos, a resposta clínica, laboratorial e radiológica ao análogo da somatostatina (AS) octreotide-LAR foi comparada à expressão dos SSTR. O SSTR mais freqüentemente expresso foi o SSTR5, seguido pelos SSTR3, SSTR2, SSTR1 e SSTR4. O SSTR1 e SSTR2 foram mais expressos nos pacientes que normalizaram GH e IGF-I. Houve correlação positiva entre o grau de redução tumoral e a expressão dos SSTR1, SSTR2 e SSTR3. Portanto, AS específicos para os SSTR´s poderão contribuir para o tratamento de acromegálicos resistentes aos AS atualmente disponíveis / This study aimed to analyze the expression of somatostatin receptor subtypes (SSTR) in 39 GH-secreting pituitary adenomas. In 19 acromegalics the clinical, laboratorial and radiological responses to the somatostatin analog (SA) octreotide-LAR were compared to SSTR´s expression. The most expressed SSTR was SSTR5, followed by SSTR3, SSTR2, SSTR1 and SSTR4. SSTR1 and SSTR2 were more expressed in patients who achieved GH and IGF-I normalization. There was a positive correlation between the degree of tumor reduction with SSTR1, SSTR2 and SSTR3 expression. Therefore, the development of specific SA could contribute to treatment improvement in resistant acromegalics patients to available SA
29

Genetic association of islet amyloid polypeptide (IAPP) encoding pathways in pancreatic beta-cells with type 2 diabetes complemented by functional study. / CUHK electronic theses & dissertations collection

January 2011 (has links)
Lam, Kwok Lim. / "October 2010." / Thesis (Ph.D.)--Chinese University of Hong Kong, 2011. / Includes bibliographical references (leaves 142-173). / Electronic reproduction. Hong Kong : Chinese University of Hong Kong, [2012] System requirements: Adobe Acrobat Reader. Available via World Wide Web. / Abstract also in Chinese.
30

Regulation of Phospholipase C and Plasma Membrane Phosphatidylinositol 4,5-bisphosphate in Insulin-Secreting Cells

Thore, Sophia January 2006 (has links)
The membrane phospholipid phosphatidylinositol 4,5-bisphosphate (PIP2) is an important signaling molecule as substrate for the phospholipase C (PLC)-catalyzed formation of inositol 1,4,5-trisphosphate (IP3) and diacylglycerol, and by directly regulating e.g. ion-channels, the cytoskeleton and vesicle trafficking in various types of cells. The present studies provide insights into the regulation of PLC activity and the plasma membrane concentration of PIP2 in individual insulin-secreting cells. Real-time monitoring of plasma membrane PIP2 was performed with evanescent wave microscopy and the PIP2/IP3-binding pleckstrin-homology-domain from PLC-δ1 fused to GFP. It was demonstrated that membrane depolarization and voltage-dependent Ca2+ influx are sufficient to activate PLC. Rise of the glucose concentration triggered Ca2+-dependent activation of PLC. Simultaneous measurements of the cytoplasmic Ca2+ concentration ([Ca2+]i) demonstrated that oscillations of [Ca2+]i resulting from periodic influx induced cyclic activation of PLC. Activation of muscarinic receptors caused a biphasic PLC response with an initial peak enhanced by positive feedback by Ca2+ mobilized from intracellular stores, followed by sustained activity depending on store-operated Ca2+-entry. Activation of PLC by Ca2+ mobilized from intracellular stores was part of the Ca2+-induced Ca2+ release mechanism by which glucagon stimulates primary mouse pancreatic β-cells. Experiments in permeabilized cells demonstrated rapid turnover of PIP2 with t1/2 ~ 16s. ATP stimulated concentration-dependent synthesis of plasma membrane PIP2, counteracted by the ADP analogue ADPβS. RT-PCR analysis identified transcripts of 10 different phosphoinositide-kinases. The ATP-stimulated PIP2 formation was mediated by type II and III PI4-kinases as well as by PIP5-kinase Iβ. It is concluded that the PIP2 concentration in the plasma membrane is regulated by the ATP/ADP ratio and that its hydrolysis by PLC is tightly controlled by [Ca2+]i in insulin-secreting cells.

Page generated in 0.0823 seconds