Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Obes Surg ; 33(3): 750-760, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36698049

RESUMO

Nutritional deficiencies following malabsorptive surgeries are a major concern. PURPOSE: To present clinical-based, mid-term nutritional outcomes in single-anastomosis duodeno-ileal bypass with sleeve gastrectomy (SADI-S) patients using a nutritional supplement based on the American Society for Metabolic & Bariatric Surgery (ASMBS) guidelines. SETTING: Single private institute, Australia. MATERIALS AND METHODS: Data from 196 patients who underwent a primary SADI-S by a single surgeon from January 2017 through March 2022 were retrospectively analysed. All patients received either original or altered formulated nutritional supplementation throughout the study. In total, three formulae, slightly different from each other, were used at three different time points to formulate the supplement. RESULTS: In total, 196 patients were included. The average age and preoperative body mass index were 44.9 ± 6.7 years and 43.6 ± 22.5 kg/m2, respectively. Nutritional follow-up was available on 77.5%, 73.2%, 73.4%, and 59.7% of patients at 12, 24, 36, and 48 months, respectively. At baseline, 48.3%, 30%, 14.9%, 13.3%, 12.4%, 3.8%, 2.3%, and 0.5% of the patients had vitamin D, calcium, folic acid, total protein, iron, vitamin B12, copper, and vitamin A deficiencies, respectively. Postoperatively, mild to moderate vitamin deficiencies were noted in 14.2% of the patients in the first 18 months; however, at 4 years, the cohort had zero nutritional deficiencies. There were no long-term complications, revisions/conversions, or mortalities related to nutritional deficiencies. CONCLUSION: Factors, like preoperative and postoperative early, aggressive correction of nutritional deficiencies, regular laboratory monitoring and follow-ups with the multidisciplinary team, and adherence to our formulated nutritional supplement, have contributed to favourable nutritional outcomes at 4 years.


Assuntos
Derivação Gástrica , Desnutrição , Obesidade Mórbida , Humanos , Obesidade Mórbida/cirurgia , Estudos Retrospectivos , Austrália/epidemiologia , Desnutrição/epidemiologia , Desnutrição/etiologia , Anastomose Cirúrgica/efeitos adversos , Gastrectomia/efeitos adversos , Derivação Gástrica/efeitos adversos
2.
Biochim Biophys Acta Gene Regul Mech ; 1860(10): 1025-1036, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28847732

RESUMO

Different genes encode the α-actin isoforms that are predominantly expressed in heart and skeletal muscle. Mutations in the skeletal muscle α-actin gene (ACTA1) cause muscle diseases that are mostly lethal in the early postnatal period. We previously demonstrated that the disease phenotype of ACTA1 mouse models could be rescued by transgenic over-expression of cardiac α-actin (ACTC1). ACTC1 is the predominant striated α-actin isoform in the heart but is also expressed in developing skeletal muscle. To develop a translatable therapy, we investigated the genetic regulation of Actc1 expression. Using strains from The Collaborative Cross (CC) genetic resource, we found that Actc1 varies in expression by up to 24-fold in skeletal muscle. We defined significant expression quantitative trait loci (eQTL) associated with early adult Actc1 expression in soleus and heart. eQTL in both heart and soleus mapped to the Actc1 locus and replicate an eQTL mapped for Actc1 in BXD heart and quadriceps. We built on this previous work by analysing genes within the eQTL peak regions to prioritise likely candidates for modifying Actc1 expression. Additionally we interrogated the CC founder haplotype contributions to enable prioritisation of genetic variants for functional analyses. Methylation around the Actc1 transcriptional start site in early adult skeletal muscle negatively correlated with Actc1 expression in a strain-dependent manner, while other marks of regulatory potential (histone modification and chromatin accessibility) were unaltered. This study provides novel insights into the complex genetic regulation of Actc1 expression in early adult skeletal muscles.


Assuntos
Actinas , Metilação de DNA/fisiologia , Regulação da Expressão Gênica/fisiologia , Músculo Esquelético/metabolismo , Regiões Promotoras Genéticas/fisiologia , Locos de Características Quantitativas , Actinas/biossíntese , Actinas/genética , Animais , Camundongos
3.
Stem Cells ; 35(5): 1341-1354, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28090703

RESUMO

Developmental stage-specific differentiation of stem or progenitor cells into safe and functional cells is of fundamental importance in regenerative medicine, including ß-cell replacement. However, the differentiation of islet progenitor cells (IPCs) into insulin-secreting ß cells remains elusive. Here, we report that the multifunctional molecule nicotinamide (NIC) is a specific differentiation regulator of mouse IPCs. The differentiated cells regulated by NIC exhibited many characteristics of adult ß cells, including ameliorating preclinical diabetes and a highly comparable transcriptome profile. Gene set enrichment analysis showed that during differentiation, numerous IPC transcription factor genes, including Ngn3, Pax4, Fev, and Mycl1, were all down regulated. Pharmacological, biochemical, and gene knockdown analyses collectively demonstrated that NIC regulated the differentiation via inhibiting Sirt1 (silent information regulator transcript 1). Finally, NIC also regulates human IPC differentiation. Thus, our study advances islet developmental biology and impacts on translational research and regenerative therapies to diabetes and other diseases. Stem Cells 2017;35:1341-1354.


Assuntos
Diferenciação Celular , Diabetes Mellitus Experimental/terapia , Células Secretoras de Insulina/transplante , Niacinamida/farmacologia , Células-Tronco/citologia , Transcriptoma/genética , Animais , Diferenciação Celular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/ultraestrutura , Proteínas Luminescentes/metabolismo , Camundongos SCID , Sirtuína 1/metabolismo , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Fatores de Transcrição/metabolismo
4.
Data Brief ; 7: 172-176, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27761488

RESUMO

The data shows results acquired in a large cohort of 5668 ethnic Arabs involved in a common variants association study for coronary artery disease (CAD) and myocardial infarction (MI) using the Affymetrix Axiom Genotyping platform ("A genome-wide association study reveals susceptibility loci for myocardial infarction/coronary artery disease in Saudi Arabs" Wakil et al. (2015) [1] ). Several loci were described that conferred risk for CAD or MI, some of which were validated in an independent set of samples. Principal Component (PCA) analysis suggested that the Saudi Cohort was close to the CEU and TSI populations, thus pointing to similarity with European populations.

5.
J Immunol ; 196(7): 3043-53, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26912320

RESUMO

Genome-wide association studies have found >60 loci that confer genetic susceptibility to type 1 diabetes (T1D). Many of these are defined only by anonymous single nucleotide polymorphisms: the underlying causative genes, as well as the molecular bases by which they mediate susceptibility, are not known. Identification of how these variants affect the complex mechanisms contributing to the loss of tolerance is a challenge. In this study, we performed systematic analyses to characterize these variants. First, all known genes in strong linkage disequilibrium (r(2) > 0.8) with the reported single nucleotide polymorphisms for each locus were tested for commonly occurring nonsynonymous variations. We found only a total of 22 candidate genes at 16 T1D loci with common nonsynonymous alleles. Next, we performed functional studies to examine the effect of non-HLA T1D risk alleles on regulating expression levels of genes in four different cell types: EBV-transformed B cell lines (resting and 6 h PMA stimulated) and purified CD4(+) and CD8(+) T cells. We mapped cis-acting expression quantitative trait loci and found 24 non-HLA loci that affected the expression of 31 transcripts significantly in at least one cell type. Additionally, we observed 25 loci that affected 38 transcripts in trans. In summary, our systems genetics analyses defined the effect of T1D risk alleles on levels of gene expression and provide novel insights into the complex genetics of T1D, suggesting that most of the T1D risk alleles mediate their effect by influencing expression of multiple nearby genes.


Assuntos
Diabetes Mellitus Tipo 1/genética , Predisposição Genética para Doença , Variação Genética , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Transformada , Epistasia Genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Estudos de Associação Genética , Estudo de Associação Genômica Ampla , Genótipo , Humanos , Leucócitos Mononucleares/metabolismo , Desequilíbrio de Ligação , Polimorfismo de Nucleotídeo Único , Locos de Características Quantitativas , Reprodutibilidade dos Testes
6.
Atherosclerosis ; 245: 62-70, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26708285

RESUMO

BACKGROUND: Multiple loci have been identified for coronary artery disease (CAD) by genome-wide association studies (GWAS), but no such studies on CAD incidence has been reported yet for any Middle Eastern population. METHODS: In this study, we performed a GWAS for CAD and myocardial infarction (MI) incidence in 5668 Saudis of Arab descent using the Affymetrix Axiom Genotyping platform. RESULTS: We describe SNPs at 16 loci that showed significant (P < 5 × 10(-8)) or suggestive GWAS association (P < 1 × 10(-5)) with CAD or MI, in the ethnic Saudi Arab population. Among the four variants reaching GWAS significance in the present study, the rs10738607_G [0.78(0.71-0.85); p = 2.17E-08] in CDNK2A/B gene was associated with CAD. Two other SNPs on the same gene, rs10757274_G [0.79(0.73-0.86); p = 2.98E-08] and rs1333045_C [0.79(0.73-0.86); p = 1.15E-08] as well as the rs9982601_T [1.38(1.23-1.55); p = 3.49E-08] on KCNE2 were associated with MI. These variants have been previously described in other populations. Several SNPs, including the rs7421388 (PLCL1) and rs12541758 (TRPA1) displaying a suggestive GWAS association (P < 1 × 10(-5)) with CAD as well as rs41411047 (RNF13), rs32793 (PDZD2) and rs4739066 (YTHDF3), similarly showing weak association with MI, were confirmed in an independent dataset. Furthermore, our estimation of heritability of CAD and MI based on observed genome-wide sharing in unrelated Saudi Arabs was approximately 33% and 44%, respectively. CONCLUSIONS: Our study has identified susceptibility variants for CAD/MI in ethnic Arabs. These findings provide further insights into pathways contributing to the susceptibility for CAD and will enable more comprehensive genetic studies of these diseases in Middle East populations.


Assuntos
Doença da Artéria Coronariana/genética , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla/métodos , Infarto do Miocárdio/genética , Doença da Artéria Coronariana/metabolismo , Feminino , Genótipo , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio/epidemiologia , Infarto do Miocárdio/metabolismo , Fatores de Risco , Arábia Saudita/epidemiologia
7.
Genetics ; 198(1): 75-86, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25236450

RESUMO

The Collaborative Cross (CC) was designed to facilitate rapid gene mapping and consists of hundreds of recombinant inbred lines descended from eight diverse inbred founder strains. A decade in production, it can now be applied to mapping projects. Here, we provide a proof of principle for rapid identification of major-effect genes using the CC. To do so, we chose coat color traits since the location and identity of many relevant genes are known. We ascertained in 110 CC lines six different coat phenotypes: albino, agouti, black, cinnamon, and chocolate coat colors and the white-belly trait. We developed a pipeline employing modifications of existing mapping tools suitable for analyzing the complex genetic architecture of the CC. Together with analysis of the founders' genome sequences, mapping was successfully achieved with sufficient resolution to identify the causative genes for five traits. Anticipating the application of the CC to complex traits, we also developed strategies to detect interacting genes, testing joint effects of three loci. Our results illustrate the power of the CC and provide confidence that this resource can be applied to complex traits for detection of both qualitative and quantitative trait loci.


Assuntos
Mapeamento Cromossômico/métodos , Cor de Cabelo/genética , Locos de Características Quantitativas , Animais , Hibridização Genética , Camundongos
8.
Proc Natl Acad Sci U S A ; 111(28): 10305-10, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24982147

RESUMO

Over 40 susceptibility loci have been identified for type 1 diabetes (T1D). Little is known about how these variants modify disease risk and progression. Here, we combined in vitro and in vivo experiments with clinical studies to determine how genetic variation of the candidate gene cathepsin H (CTSH) affects disease mechanisms and progression in T1D. The T allele of rs3825932 was associated with lower CTSH expression in human lymphoblastoid cell lines and pancreatic tissue. Proinflammatory cytokines decreased the expression of CTSH in human islets and primary rat ß-cells, and overexpression of CTSH protected insulin-secreting cells against cytokine-induced apoptosis. Mechanistic studies indicated that CTSH exerts its antiapoptotic effects through decreased JNK and p38 signaling and reduced expression of the proapoptotic factors Bim, DP5, and c-Myc. CTSH overexpression also up-regulated Ins2 expression and increased insulin secretion. Additionally, islets from Ctsh(-/-) mice contained less insulin than islets from WT mice. Importantly, the TT genotype was associated with higher daily insulin dose and faster disease progression in newly diagnosed T1D patients, indicating agreement between the experimental and clinical data. In line with these observations, healthy human subjects carrying the T allele have lower ß-cell function, which was evaluated by glucose tolerance testing. The data provide strong evidence that CTSH is an important regulator of ß-cell function during progression of T1D and reinforce the concept that candidate genes for T1D may affect disease progression by modulating survival and function of pancreatic ß-cells, the target cells of the autoimmune assault.


Assuntos
Catepsina H/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Células Secretoras de Insulina/metabolismo , Adolescente , Alelos , Animais , Apoptose/genética , Catepsina H/genética , Linhagem Celular , Criança , Pré-Escolar , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 1/terapia , Regulação da Expressão Gênica/genética , Genótipo , Humanos , Células Secretoras de Insulina/patologia , Camundongos , Camundongos Knockout , Ratos
9.
Stem Cell Rev Rep ; 8(3): 803-12, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22278131

RESUMO

Embryonic stem cells (ESCs) have been promised as a renewable source for regenerative medicine, including providing a replacement therapy in type 1 diabetes. However, they have not yet been differentiated into functional insulin-secreting ß cells. This is due partially to the knowledge gap regarding the transcription factors (TFs) required for pancreas development. We hypothesize that, if directed differentiation in vitro recapitulates the developmental process in vivo, ESCs provide a powerful model to discover novel pancreatic TF genes. Guided by knowledge of their normal development and using RT-PCR and immunochemical analyses, we have established protocols for directed differentiation of mouse ESCs into pancreatic progenitors. Microarray analyses of these differentiating ESC cells at days 0, 4, 8 and 15 confirmed their sequential differentiation. By day 15, we found up-regulation of a group of pancreatic progenitor marker genes including Pdx1, Ptf1a, Nkx6.1, Pax4 and Pax6. Consistently, Pdx1-immunoreactive cells were detected on day 15. Most of these Pdx1(+) cells also expressed Nkx6.1. Bioinformatic analyses of sequential datasets allowed identification of over 20 novel TF genes potentially important for pancreas development. The dynamic expression of representative known and novel genes was confirmed by quantitative real time RT-PCR analysis. This strategy may be modified to study novel regulatory molecules for development of other tissue and organ systems.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/metabolismo , Pâncreas/crescimento & desenvolvimento , Fatores de Transcrição/genética , Animais , Linhagem Celular , Técnicas de Cocultura , Células-Tronco Embrionárias/fisiologia , Endoderma/citologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Pâncreas/citologia , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/metabolismo , Transcrição Gênica
10.
Mamm Genome ; 22(9-10): 556-62, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21732193

RESUMO

Iron homeostasis is crucial to many biological functions in nearly all organisms, with roles ranging from oxygen transport to immune function. Disruption of iron homeostasis may result in iron overload or iron deficiency. Iron deficiency may have severe consequences, including anemia or changes in immune or neurotransmitter systems. Here we report on the variability of phenotypic iron tissue loss and splenomegaly and the associated quantitative trait loci (QTLs), polymorphic areas in the mouse genome that may contain one or more genes that play a role in spleen iron concentration or spleen weight under each dietary treatment. Mice from 26 BXD/Ty recombinant inbred strains, including the parent C57BL/6 and DBA/2 strains, were randomly assigned to adequate iron or iron-deficient diets at weaning. After 120 days, splenomegaly was measured by spleen weight, and spleen iron was assessed using a modified spectrophotometry technique. QTL analyses and gene expression comparisons were then conducted using the WebQTL GeneNetwork. We observed wide, genetic-based variability in splenomegaly and spleen iron loss in BXD/Ty recombinant inbred strains fed an iron-deficient diet. Moreover, we identified several suggestive QTLs. Matching our QTLs with gene expression data from the spleen revealed candidate genes. Our work shows that individual differences in splenomegaly response to iron deficiency are influenced at least partly by genetic constitution. We propose mechanistic hypotheses by which splenomegaly may result from iron deficiency.


Assuntos
Deficiências de Ferro , Distúrbios do Metabolismo do Ferro/genética , Locos de Características Quantitativas , Baço/metabolismo , Animais , Feminino , Perfilação da Expressão Gênica , Ferro/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Tamanho do Órgão/genética , Fenótipo , Fatores Sexuais
11.
Diabetes ; 60(3): 1030-40, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21266329

RESUMO

OBJECTIVE: Interactions between genetic and environmental factors lead to immune dysregulation causing type 1 diabetes and other autoimmune disorders. Recently, many common genetic variants have been associated with type 1 diabetes risk, but each has modest individual effects. Familial clustering of type 1 diabetes has not been explained fully and could arise from many factors, including undetected genetic variation and gene interactions. RESEARCH DESIGN AND METHODS: To address this issue, the Type 1 Diabetes Genetics Consortium recruited 3,892 families, including 4,422 affected sib-pairs. After genotyping 6,090 markers, linkage analyses of these families were performed, using a novel method and taking into account factors such as genotype at known susceptibility loci. RESULTS: Evidence for linkage was robust at the HLA and INS loci, with logarithm of odds (LOD) scores of 398.6 and 5.5, respectively. There was suggestive support for five other loci. Stratification by other risk factors (including HLA and age at diagnosis) identified one convincing region on chromosome 6q14 showing linkage in male subjects (corrected LOD = 4.49; replication P = 0.0002), a locus on chromosome 19q in HLA identical siblings (replication P = 0.006), and four other suggestive loci. CONCLUSIONS: This is the largest linkage study reported for any disease. Our data indicate there are no major type 1 diabetes subtypes definable by linkage analyses; susceptibility is caused by actions of HLA and an apparently random selection from a large number of modest-effect loci; and apart from HLA and INS, there is no important susceptibility factor discoverable by linkage methods.


Assuntos
Diabetes Mellitus Tipo 1/genética , Ligação Genética , Análise de Variância , Mapeamento Cromossômico , Feminino , Loci Gênicos , Predisposição Genética para Doença , Genótipo , Humanos , Masculino , Fatores de Risco , Irmãos
12.
Pancreas ; 39(2): 201-8, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19812524

RESUMO

OBJECTIVE: Despite great progress in understanding the transcriptional regulation of the development of insulin-secreting beta cells, the quantitative temporal expression of insulin gene(s) remains largely unknown. We here aimed to quantify insulin gene transcripts during development. METHODS: We described bioinformatics algorithms to quantify (insulin) gene transcript abundance in sequential microarray data sets at the global level. Several molecular techniques were used to confirm our analyses. RESULTS: We demonstrated that the expression of insulin genes was up-regulated at approximately 14-fold, 700- to 2000-fold, and 5000- to 6000-fold in Pdx1- and Ngn3-expressing cells and adult islets compared with definitive endodermal or embryonic stem cells, respectively. The expression of multiple genes encoding molecules involved in posttranslational modifications of insulin and glucose sensing was also elevated in the same period. All islet and associated genes determined with microarray data were confirmed not only to be up-regulated by real-time quantitative reverse transcriptase polymerase chain reaction but also that the magnitude of their increase quantified with these 2 methods was statistically highly correlated. Consistent with the above, green fluorescence protein expression under the control of the mouse insulin 1 promoter could be visualized in the pancreas from embryonic day (E) 11.5, increasing progressively through E13.5 to E15.5. CONCLUSION: Our study provides a novel insight into islet developmental biology.


Assuntos
Biologia Computacional , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica no Desenvolvimento , Insulina/genética , Ilhotas Pancreáticas/crescimento & desenvolvimento , Análise de Sequência com Séries de Oligonucleotídeos , Fatores Etários , Algoritmos , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular/genética , Proliferação de Células , Bases de Dados Genéticas , Células-Tronco Embrionárias/metabolismo , Redes Reguladoras de Genes , Idade Gestacional , Proteínas de Homeodomínio/genética , Ilhotas Pancreáticas/embriologia , Ilhotas Pancreáticas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transativadores/genética
13.
J Autoimmun ; 31(3): 233-6, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18511236

RESUMO

"Systems Genetics" detects variation in phenotypic traits and integrates this with underlying genetic variation. A powerful application of systems genetics is analyzing effects of genome-wide genetic variants on transcriptome-wide variation in gene expression. We see systems genetics as a new powerful technology which will empower research in genetics and in other disciplines. Here, we present preliminary analyses of systems genetics approaches to defining genetic interactions within the thymus, which is the key site for T lymphocyte development and imposition of immunological tolerance.


Assuntos
Autoimunidade/genética , Perfilação da Expressão Gênica , Receptores de Interleucina-7/imunologia , Linfócitos T/imunologia , Timo/metabolismo , Animais , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Receptores de Interleucina-7/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...