Academic literature on the topic 'TGFBR1/2 polymorphisms'

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the lists of relevant articles, books, theses, conference reports, and other scholarly sources on the topic 'TGFBR1/2 polymorphisms.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Journal articles on the topic "TGFBR1/2 polymorphisms"

1

KAKLAMANI, VIRGINIA G., MAUREEN SADIM, YVONI KOUMANTAKI, PHEDON KAKLAMANIS, and BORIS PASCHE. "Role of Polymorphisms in Adamantiades-Behçet’s Disease." Journal of Rheumatology 35, no. 12 (December 2008): 2376–78. http://dx.doi.org/10.3899/jrheum.080676.

Full text
Abstract:
ObjectiveWe previously showed that Adamantiades-Behçet’s disease (A-BD) is associated with a lower incidence of malignancy compared with the general population. Transforming growth factor-β (TGF-β) has been shown to play a role in cartilage regeneration and is increased in patients with A-BD. We also found 2 functional polymorphisms of the TGF-β pathway, TGFBR1*6A and TGFB1*CC, that are associated with risk of malignancy. We tested whether incidence of these polymorphisms would differ in patients with A-BD compared with healthy controls of similar age and geographic location.MethodsWe performed a case-control study including 139 cases and 128 controls from Greece. Cases and controls were genotyped for TGFBR1*6A and TGFB1*CC.ResultsWe found that cases had lower incidence of TGFBR1*6A compared with controls (11.3% vs 13.3%, respectively). Also, the incidence of TGFB1*CC was lower in cases than controls (24.6% vs 27.0%, respectively). These differences were not statistically significant.ConclusionAlthough there is a suggestion that the lower incidence of TGFBR1*6A in A-BD patients may play a protective role against development of malignancy, larger studies would be needed to fully evaluate the role of TGF-β and its polymorphisms in A-BD.
APA, Harvard, Vancouver, ISO, and other styles
2

Guhlich, Manuel, Laura Hubert, Caroline Patricia Nadine Mergler, Margret Rave-Fraenk, Leif Hendrik Dröge, Martin Leu, Heinz Schmidberger, Stefan Rieken, Andrea Hille, and Markus Anton Schirmer. "Identification of Risk Loci for Radiotoxicity in Prostate Cancer by Comprehensive Genotyping of TGFB1 and TGFBR1." Cancers 13, no. 21 (November 8, 2021): 5585. http://dx.doi.org/10.3390/cancers13215585.

Full text
Abstract:
Genetic variability in transforming growth factor beta pathway (TGFB) was suggested to affect adverse events of radiotherapy. We investigated comprehensive variability in TGFB1 (gene coding for TGFβ1 ligand) and TGFBR1 (TGFβ receptor-1) in relation to radiotoxicity. Prostate cancer patients treated with primary radiotherapy (n = 240) were surveyed for acute and late toxicity. Germline polymorphisms (n = 40) selected to cover the common genetic variability in TGFB1 and TGFBR1 were analyzed in peripheral blood cells. Human lymphoblastoid cell lines (LCLs) were used to evaluate a possible impact of TGFB1 and TGFBR1 genetic polymorphisms to DNA repair capacity following single irradiation with 3 Gy. Upon adjustment for multiplicity testing, rs10512263 in TGFBR1 showed a statistically significant association with acute radiation toxicity. Carriers of the Cytosine (C)-variant allele (n = 35) featured a risk ratio of 2.17 (95%-CI 1.41–3.31) for acute toxicity ≥ °2 compared to Thymine/Thymine (TT)-wild type individuals (n = 205). Reduced DNA repair capacity in the presence of the C-allele of rs10512263 might be a mechanistic explanation as demonstrated in LCLs following irradiation. The risk for late radiotoxicity was increased by carrying at least two risk genotypes at three polymorphic sites, including Leu10Pro in TGFB1. Via comprehensive genotyping of TGFB1 and TGFBR1, promising biomarkers for radiotoxicity in prostate cancer were identified.
APA, Harvard, Vancouver, ISO, and other styles
3

Puchenkova, Olesya A., Vladislav O. Soldatov, Andrei E. Belykh, OlgaYu Bushueva, Gennadii A. Piavchenko, Artem A. Venediktov, Nikolay K. Shakhpazyan, Alexey V. Deykin, Mikhail V. Korokin, and Mikhail V. Pokrovskiy. "Cytokines in Abdominal Aortic Aneurysm: Master Regulators With Clinical Application." Biomarker Insights 17 (January 2022): 117727192210956. http://dx.doi.org/10.1177/11772719221095676.

Full text
Abstract:
Abdominal aortic aneurysm (AAA) is a potentially life-threatening disorder with a mostly asymptomatic course where the abdominal aorta is weakened and bulged. Cytokines play especially important roles (both positive and negative) among the molecular actors of AAA development. All the inflammatory cascades, extracellular matrix degradation and vascular smooth muscle cell apoptosis are driven by cytokines. Previous studies emphasize an altered expression and a changed epigenetic regulation of key cytokines in AAA tissue samples. Such cytokines as IL-6, IL-10, IL-12, IL-17, IL-33, IL-1β, TGF-β, TNF-α, IFN-γ, and CXCL10 seem to be crucial in AAA pathogenesis. Some data obtained in animal studies show a protective function of IL-10, IL-33, and canonical TGF-β signaling, as well as a dual role of IL-4, IFN-γ and CXCL10, while TNF-α, IL-1β, IL-6, IL-12/IL-23, IL-17, CCR2, CXCR2, CXCR4 and the TGF-β noncanonical pathway are believed to aggravate the disease. Altogether data highlight significance of cytokines as informative markers and predictors of AAA. Pathologic serum/plasma concentrations of IL-1β, IL-2, IL-6, TNF-α, IL-10, IL-8, IL-17, IFN-γ, and PDGF have been already found in AAA patients. Some of the changes correlate with the size of aneurysms. Moreover, the risk of AAA is associated with polymorphic variants of genes encoding cytokines and their receptors: CCR2 (rs1799864), CCR5 (Delta-32), IL6 (rs1800796 and rs1800795), IL6R (rs12133641), IL10 (rs1800896), TGFB1 (rs1800469), TGFBR1 (rs1626340), TGFBR2 (rs1036095, rs4522809, rs1078985), and TNFA (rs1800629). Finally, 5 single-nucleotide polymorphisms in gene coding latent TGF-β-binding protein ( LTBP4) and an allelic variant of TGFB3 are related to a significantly slower AAA annual growth rate.
APA, Harvard, Vancouver, ISO, and other styles
4

Puchenkova, Olesya A., Vladislav O. Soldatov, Andrei E. Belykh, OlgaYu Bushueva, Gennadii A. Piavchenko, Artem A. Venediktov, Nikolay K. Shakhpazyan, Alexey V. Deykin, Mikhail V. Korokin, and Mikhail V. Pokrovskiy. "Cytokines in Abdominal Aortic Aneurysm: Master Regulators With Clinical Application." Biomarker Insights 17 (January 2022): 117727192210956. http://dx.doi.org/10.1177/11772719221095676.

Full text
Abstract:
Abdominal aortic aneurysm (AAA) is a potentially life-threatening disorder with a mostly asymptomatic course where the abdominal aorta is weakened and bulged. Cytokines play especially important roles (both positive and negative) among the molecular actors of AAA development. All the inflammatory cascades, extracellular matrix degradation and vascular smooth muscle cell apoptosis are driven by cytokines. Previous studies emphasize an altered expression and a changed epigenetic regulation of key cytokines in AAA tissue samples. Such cytokines as IL-6, IL-10, IL-12, IL-17, IL-33, IL-1β, TGF-β, TNF-α, IFN-γ, and CXCL10 seem to be crucial in AAA pathogenesis. Some data obtained in animal studies show a protective function of IL-10, IL-33, and canonical TGF-β signaling, as well as a dual role of IL-4, IFN-γ and CXCL10, while TNF-α, IL-1β, IL-6, IL-12/IL-23, IL-17, CCR2, CXCR2, CXCR4 and the TGF-β noncanonical pathway are believed to aggravate the disease. Altogether data highlight significance of cytokines as informative markers and predictors of AAA. Pathologic serum/plasma concentrations of IL-1β, IL-2, IL-6, TNF-α, IL-10, IL-8, IL-17, IFN-γ, and PDGF have been already found in AAA patients. Some of the changes correlate with the size of aneurysms. Moreover, the risk of AAA is associated with polymorphic variants of genes encoding cytokines and their receptors: CCR2 (rs1799864), CCR5 (Delta-32), IL6 (rs1800796 and rs1800795), IL6R (rs12133641), IL10 (rs1800896), TGFB1 (rs1800469), TGFBR1 (rs1626340), TGFBR2 (rs1036095, rs4522809, rs1078985), and TNFA (rs1800629). Finally, 5 single-nucleotide polymorphisms in gene coding latent TGF-β-binding protein ( LTBP4) and an allelic variant of TGFB3 are related to a significantly slower AAA annual growth rate.
APA, Harvard, Vancouver, ISO, and other styles
5

Kirschneck, Margarita, Nermien Zbidat, Eva Paddenberg, Caio Luiz Bitencourt Reis, Isabela Ribeiro Madalena, Maria Angélica Hueb de Menezes-Oliveira, César Penazzo Lepri, Peter Proff, Christian Kirschneck, and Erika Calvano Küchler. "Transforming Growth Factor Beta Receptor 2 (TGFBR2) Promoter Region Polymorphisms May Be Involved in Mandibular Retrognathism." BioMed Research International 2022 (June 15, 2022): 1–7. http://dx.doi.org/10.1155/2022/1503052.

Full text
Abstract:
Skeletal malocclusions are common phenotypes in humans and have a strong influence on genetic factors. Transforming growth factor beta (TGFβ) controls numerous functions of the human body, including cell proliferation, differentiation, and migration. Thus, this study is aimed at evaluating whether genetic polymorphisms in TGFB1 and its receptor TGFBR2 are associated with mandibular retrognathism in German children and adolescents. Children and teenagers older than 8 years in the mixed or permanent dentition were included in this study. Patients with syndromes and facial trauma and patients with congenital alterations were excluded. Digital cephalometric tracings were performed using the anatomical landmarks point A, point B, sella (S), and nasion (N). Patients that have a retrognathic mandible ( SNB < 78 °) were selected as case group, and the patients with an orthognathic mandible ( SNB = 78 °– 82°) were selected as the control group. Genomic deoxyribonucleic acid (DNA) from saliva was used to evaluate four genetic polymorphisms in TGFB1 (rs1800469 and rs4803455) and TGBR2 (rs3087465 and rs764522) using real-time PCR. Chi-square or Fisher exact tests were used to compare gender, genotype, and allele distribution among groups. Genotype distribution was calculated in an additive and recessive model. Haplotype analysis was also performed. The established alpha of this study was 5%. A total of 146 patients (age ranging from 8 to 18 years) were included in this epidemiological genetic study. The genetic polymorphism rs3087465 in TGFBR2 was associated with mandibular retrognathism. Carrying the AA genotype in the rs3087465 polymorphism decreased the chance of having mandibular retrognathism ( odds ratio = 0.25 , confidence interval 95 % = 0.06 to 0.94, p = 0.045 ). None of the haplotypes was associated with mandibular retrognathism ( p > 0.05 ). In conclusion, we found that the genetic polymorphism rs3087465 in the promoter region of the TGFBR2 was associated with mandibular retrognathism in Germans.
APA, Harvard, Vancouver, ISO, and other styles
6

Pauly, M., G. Mahon, M. A. Dicato, B. Metzger, and A. Menzel. "Single Nucleotide Polymorphisms (SNP'S) in the P53, SMAD7 and TGFBR1 Genes Associated with Advanced Colorectal Cancer in Caucasian Patients Compared to Healthy Controls." Annals of Oncology 23 (September 2012): ix209. http://dx.doi.org/10.1016/s0923-7534(20)33231-2.

Full text
APA, Harvard, Vancouver, ISO, and other styles
7

Kim, Dong Hwan (Dennis), Jina Yun, Jee Hyun Kong, Chul Won Jung, Ahmed Galal, Vikas Gupta, John Kuruvilla, Hans A. Messner, and Jeffrey H. Lipton. "Single Nucleotide Polymorphism (SNP) Approach of Multiple Candidate Pathways Predicting the Risk of Acute / Chronic Graft-Versus-Host Disease or Transplant Outcomes Following Allogeneic Hematopoietic Stem Cell Transplantation: Potential Involvement of Nuclear Factor Kappa-B (NFKB), Platelet-Derived Growth Factor (PDGF) and Transforming Growth Factor-Beta (TGF-β) Pathway with Chronic Graft-Versus-Host Disease Graft-Versus-Host Disease." Blood 114, no. 22 (November 20, 2009): 2221. http://dx.doi.org/10.1182/blood.v114.22.2221.2221.

Full text
Abstract:
Abstract Abstract 2221 Poster Board II-198 Background: Acute graft-versus-host disease (GVHD) was known to be involved in the Th1 cytokine activation and alloreactive T-cell cytotoxicity, while the pathogenesis of chronic GVHD is yet revealed fully although in which Th2 cytokine activation or transforming growth factor (TGF) mediated pathway was suggested to be involved. The current study is a hypothesis generating study in order to identify potential predictive surrogate associated with the risk of acute or chronic GVHD in addition with transplant outcomes after allogeneic hematopoietic stem cell transplantation (HSCT). Methods: The current study was performed to identify genetic surrogates predicting the risk of acute / chronic GVHD, relapse free survival, non-relapse mortality and overall survival in 394 pairs transplanted at the Princess Margaret Hospital, Toronto, ON, Canada. In addition, the predictive markers for organ specific incidence of acute / chronic GVHD were also evaluated (i.e. for skin/liver/gut acute GVHD or skin, eye, oral, lung or liver chronic GVHD). Total of 261 single nucleotide polymorphisms (SNPs) in 56 genes were determined for donor/recipients' genotypes using MALDI-TOF based platform, involving in the pathways of 1) cytokines (i.e. IL1A, IL1B and its receptor, IL1R1, IL2 & IL2RA, IL4 & IL4R, IL6 & IL6R, IL8, IL10 & IL10RA, RB, IL12A/BandIL12RB1, IFNG & IFNGR1/2, TNFTI/II/II), 2) NFKB (NFKB1/2/A, NFKBIA/B, IKB, IKK1, IKBKB, RelB), 3) apoptosis (FAS, TRAIL & TRAILR1), 4) endothelium nitric oxide regulation (EDN1, NOS1/2A/3), 5) PDGF (PDGFB/C/D & PDGFRA/B), 6) TGF-β (TGFB1/2 & TGFBR1/2/3, TGFRB1), 7) Toll-like receptor (TLR4/5), 8) NOD2/CARD15 and 9) prostaglandin-endoperoxide synthase (PTGS1/2). The candidate genotypes have been selected by choosing the SNPs in non-synonymous SNPs in exon region with minor allele frequency of > 0.05 to 0.1. Results: Followings are the lists of recipients' and donors' genotypes with p-value<0.05 thus associating with clinical outcomes following allogeneic HSCT: In summary, the risk of chronic GVHD was significantly associated with SNP of the genes involved in the pathway of NFKB, PDGF, TGF-β, and some of cytokines (esp. type II, IL6 & IL4), while that of acute GVHD associates with the genotypes in the pathway of TNF and apoptosis. In addition, survival after allogeneic transplantation was associated with the genotypes in NOS (nitric oxide synthase, endothelial nitric oxide synthesis pathway), IL-2 and TGF pathway. Conclusion: Because of complex nature of GVHD pathogenesis, multiple candidate pathway SNPs has been explored targeting SNPs in the pathway of cytokines, NFKB, apoptosis, endothelium nitric oxide regulation, NOD2/CARD15, PDGF, PTGS1/2, TGF-β and TLR. Different involvements were noted of TGF-β, PDGF or NFKB with chronic GVHD versus TNF and apoptosis-associated SNPs with acute GVHD. Further study will help us to reach more clear conclusion which genotype is the predictor of the risk of GVHD. Disclosures: No relevant conflicts of interest to declare.
APA, Harvard, Vancouver, ISO, and other styles
8

Chen, Ruo-Xi, Wen-Min Lu, Mei-Ping Lu, Mei-Lin Wang, Xin-Jie Zhu, Zhong-Fei Wu, Hui-Qin Tian, Lu-Ping Zhu, Zheng-Dong Zhang, and Lei Cheng. "Polymorphisms in MicroRNA Target Sites of TGF-β Signaling Pathway Genes and Susceptibility to Allergic Rhinitis." International Archives of Allergy and Immunology 182, no. 5 (2021): 399–407. http://dx.doi.org/10.1159/000511975.

Full text
Abstract:
<b><i>Background:</i></b> The polymorphisms inside microRNA target sites locating in the 3′-UTR region may introduce the micro­RNA-binding changes, which may regulate the gene expression and correlate with the potential diseases. <b><i>Objectives:</i></b> We aimed to investigate whether the polymorphisms in microRNA target sites of transforming growth factor beta (TGF-β) signaling pathway genes are associated with the susceptibility of mite-sensitized allergic rhinitis (AR) in a Han Chinese population. <b><i>Methods:</i></b> In this case-control study, 454 AR patients and 448 healthy controls were recruited. Three HapMap single-nucleotide polymorphisms (SNPs) were mapped to putative microRNA recognition sites and genotyped by TaqMan allelic discrimination assay. <b><i>Results:</i></b> The genotype and allele frequencies of 3 SNPs (rs1590 in <i>TGFBR1</i>; rs1434536 and rs17023107 in <i>BMPR1B</i>) showed lack of significant association with AR. However, in the subgroup analysis, the TG, GG, and TG/GG genotypes of rs1590 exhibited significantly increased risk of AR in the male subgroup (TG: adjusted OR = 1.57, 95% CI = 1.08–2.31; GG: adjusted OR = 1.76, 95% CI = 1.09–2.86; TG/GG: adjusted OR = 1.62, 95% CI = 1.13–2.33). The CT genotypes of rs17023107 might have potential to protect against AR in the patients age of &#x3c;15 years (adjusted OR = 0.37, 95% CI = 0.14–0.95) and the males (adjusted OR = 0.48, 95% CI = 0.25–0.95). No significant association was found between SNPs and the total serum IgE level. <b><i>Conclusions:</i></b> In a Han Chinese population, stratified by age and gender, susceptibility to mite-sensitized AR may be associated with 2 SNPs (rs1590 and rs17023107) in microRNA target sites of TGF-β signaling pathway genes.
APA, Harvard, Vancouver, ISO, and other styles
9

Kim, Dennis Dong Hwan, Hong-Hee Won, Wei Xu, Jieun Uhm, Vikas Gupta, John Kuruvilla, Hans A. Messner, and Jeffrey H. Lipton. "The Risk of Organ Specific Graft-Versus-Host Disease Can Be Predicted by the Multiple Single Nucleotide Polymorphism Based Predictive Models." Blood 120, no. 21 (November 16, 2012): 3056. http://dx.doi.org/10.1182/blood.v120.21.3056.3056.

Full text
Abstract:
Abstract Abstract 3056 Background: The pathogenesis of GVHD is not fully understood. Alloreactive T-lymphocytes are believed to be key mediators of GVHD. However, it is not clear if the pathobiology of GHVD is similar in each target organ GVHD. We aimed to identify predictive single nucleotide polymorphisms (SNP) markers associated with the risk of acute or chronic graft versus host disease (GVHD) as well as organ specific GVHD in 394 transplant recipients and donors. Methods: A total of 259 SNPs were genotyped in 53 genes, and evaluated for the risk of acute/chronic GVHD and organ specific GVHD. Predictive models were generated using both clinical factors and genetic SNP markers confirmed by multivariate analyses. Patients were stratified by quartile (25%) according to their risk score, and the risk of overall and organ specific GVHD were compared among the 3 risk groups (low, intermediate and high risk). C-statistic analysis was also performed to compare the stratification power of the predictive model generated using clinical and genetic factors with a model obtained using only clinical factors. Results: Several SNP markers in the cytokine-, apoptosis-, TGF-¥â or PDGF-mediated pathways were identified as predictive markers of acute/chronic GVHD. The risk of acute GVHD was associated with clinical factors such as HLA disparity and patient age. In addition, recipient FAS genotype (rs2234978), EDN1 genotype (rs4714384), and TGFB genotype (rs1800469), and donor TNFRII genotype (rs3397) were also strong predictive markers for acute GVHD. Significant predictive risk factors forchronic GVHD were the source of stem cells, a previous episode of acute GVHD and the donor IL1R1 genotype (rs3917225). Each organ specific GVHD shared common biologic pathways such as cytokine, TGF-¥â or PDGF-mediated pathways. However, different SNP markers were identified as predictive for individual organ-specific GVHD. Multivariate analyses identified several SNP markers may predict the risk of organ specific acute GVHD in combination with clinical factors. For skin acute GVHD, recipient PDGFD (rs10895534), donor NOS2A (rs3730017), TNFRII (rs3397) and TGFB1 (rs1800469) genotypes were predictive together with clinical factors such as HLA disparity. Donor's genotype for TNFRII (rs3397) was predictive not only for overall acute GVHD but also for skin acute GVHD. No clinical factors were identified for the risk of liver or gut acute GVHD, but several SNP markers were found including recipient PDGFRB (rs2302273), IFNGR1 (rs2234711) and donor PTGS1 (rs10306114), NOS1 (rs9658254), IL1R1 (rs2192752) genotypes for liver acute GVHD and recipient IL4 (rs2243248), donor PDGFD (rs1053861), TGFBR1 (rs420549), IL12A (rs2243115) genotypes for gut acute GVHD. In summary, there are no overlapping SNP markers for the risk prediction of organ specific acute GVHD. For organ specific chronic GVHD, 2 clinical risk factors were predictive including source of stem cells and a preceding history of acute GVHD. In addition, several SNP markers were also identified: recipient PDGFC (rs1425486), donor NFKB1 (rs1805034) and NOS2A (rs3730017) for skin chronic GVHD; recipient IL10RB (rs8178561) and PDGFRB (rs22229562), and donor TGFBR1 (rs868) for eye chronic GVHD; recipient IL12RB1 (rs3746190) and donor FCGR2A (rs1801274) for oral chronic GVHD; and donor IL4R (rs2057768), FAS (rs2234767) and TGFB1 (rs1800469) for lung chronic GVHD. Again, In no overlapping SNP markers were observed for organ-specific chronic GVHD risk. Although this predictive model could not stratify patients according to their risk of overall chronic GVHD (p=0.0763), the predictive models per each organ specific chronic GVHD enabled to stratify the patients according to their risks of each organ specific GVHD (p<0.0001 for skin chronic GVHD, p=0.0033 for eye chronic GVHD, p=0.003 for oral chronic GVHD and p=0.0036 for lung chronic GVHD).Predictive models incorporating clinical and genetic factors improved the stratification power by 11.1% compared to models only including clinical factors. Conclusion: Our study suggests that SNP based approaches can predict the risk of organ-specific GVHD. These SNP markers need to be validated in other series. These SNPs may help focus studies into pathobiology and targeted therapy of GVHD in the future. Disclosures: No relevant conflicts of interest to declare.
APA, Harvard, Vancouver, ISO, and other styles
10

Thorne, Jacob W., Reid Redden, Scott A. Bowdridge, Gabrielle M. Becker, Morgan R. Stegemiller, and Brenda M. Murdoch. "PSV-B-21 Genome-Wide Analysis of Sheep Artificially or Naturally Infected with Gastrointestinal Nematodes." Journal of Animal Science 100, Supplement_3 (September 21, 2022): 307–8. http://dx.doi.org/10.1093/jas/skac247.560.

Full text
Abstract:
Abstract Gastrointestinal nematodes (GIN) are detrimental to the health and productivity of sheep across the world, necessitating genetic selection for improved GIN resistance. In this study, we used genomic analyses across- and within- two important breeds in the United States (US), Rambouillet and Dorper, to investigate physiological mechanisms associated with GIN resistance. Genomic data were evaluated from two experiments where lambs were challenged with GIN, either in a natural environment or artificially. Lambs were genotyped with the Axiom™ Ovine Genotyping Array (50K) with 42,608 single nucleotide polymorphisms (SNP) remaining following quality control for minor allele frequency and variant call rate. In experiment 1, a fecal egg count (FEC) was collected from four- to eight-month-old Dorper and Rambouillet lambs (n=188 total) exposed to GIN on pasture. Using Plink v1.9, a genome-wide association study (GWAS) identified significant SNP (P &lt; 6.0e-5) associated with either an increase or decrease of FEC on chromosomes 1, 2, 3, 4, 12, 15 and 20. In Experiment 2, Rambouillet lambs (n=77) were inoculated with 10,000 Haemonchus contortus L3 larvae and FEC and packed-cell volume (PCV) were recorded. An ensuing GWAS identified significant SNP (P &lt; 2.0e-5) on chromosomes 1, 2, 3, 5, 7, 16, 17 and 23 associated with high and low FEC and SNP on chromosome 10 associated with decreased PCV. Multiple genes were in proximity to the identified SNP, including PIK3 subunits in both experiments. Pathway analysis of reported genes, including PIK3, IMP4, NDST3, MRPL22, HSPA2, TGFA, TGFBR1 and RUNX1 revealed involvement in tissue repair, T-cell differentiation and cytokine signaling in Rambouillet and Dorper sheep more resistant to GIN. Results from this study contribute to the genetic underpinnings of host response to GIN in two important breeds in the US, providing a foundation for future selection of animals more resistant to H. contortus infection.
APA, Harvard, Vancouver, ISO, and other styles

Dissertations / Theses on the topic "TGFBR1/2 polymorphisms"

1

DE, CARIO ROSINA. "Genetic bases of aortopathies: from Marfan syndrome to bicuspid aortic valve." Doctoral thesis, Università di Siena, 2017. http://hdl.handle.net/11365/1005832.

Full text
Abstract:
INTRODUCTION: Aortic aneurysms might represent life-threatening diseases, even though they are less common with respect to many other cardiovascular conditions. The global clinical awareness of aortic aneurysms, along with the latest methods of diagnosis, will definitely help reducing the morbidity and mortality associated with this condition. Genetic screening represents, at present, an emerging and helpful tool in supporting the clinical diagnosis of thoracic aortic aneurysms and/or dissection (TAA/D), a potential lethal condition with a rising incidence. Its clinical impact, as well as the therapeutic management, differs according to the syndromic or non-syndromic manifestations of the disease. TAA/D represents a characteristic clinical feature common to different connective tissue disorders, e.g., Marfan syndrome (MFS), Loeys-Dietz syndrome (LDS), and bicuspid aortic valve (BAV). Analysis of genes associated with these disorders helps in early detection of the disease and in differential diagnosis with important implications for the management of the individuals affected by these complex traits. PART I BACKGROUND: Genetic variants in transforming growth factor beta (TGF-β) receptors type 1 (TGFBR1) and type 2 (TGFBR2) genes have been associated with different hereditary connective tissue disorders sharing TAA/D. Mutations in both TGFBR1/2 genes have been described in patients with TAA/D and MFS, and they are consistently associated with LDS. Existing literature shows discordant data resulting from mutational screening of TGFBR1/2 genes in MFS patients. Aim of the first part of the PhD thesis was to investigate the role of TGFBR1/2 genetic variants in determining and/or modulating MFS clinical phenotype. MATERIALS AND METHODS: 75 unrelated patients who underwent FBN1 mutational screening (47 patients with pathogenetic and 28 without pathogenetic FBN1 mutation) were subjected to direct sequencing for TGFBR2/1genes. RESULTS: Forty-seven MFS patients (63%) carried a pathogenetic FBN1 mutation. No pathogenetic mutations were detected in TGFBR1/2 genes. Ten common polymorphisms were identified in TGFBR2 and 6 in TGFBR1. Their association with cardiovascular (CV) manifestations was evaluated. Carriers of A allele of rs11466512, delA allele of c.383delA or delT allele of c.1256-15del1T polymorphisms had a trend towards or significantly reduced Z-scores [2.2 (1.13-4.77); 2.1 (1.72-3.48); 2.5 (1.85-3.86)] with respect to homozygous wild-type MFS patients [4.20 (2.39-7.25); 3.9 (2.19-7.00); 3.9 (2.14-6.93)]. Carriers of A allele of rs2276767 polymorphism showed a trend towards increased Z-score [4.9 (2.14-7.16)] with respect to wild-type MFS patients [3.3 (1.75-5.45)]. The protective effect of TGFBR1/2 genetic score including all the 4 variants was also evaluated. MFS patients with 2 or more protective alleles included in the score had statistically significant reduced aortic Z-scores [2.20 (1.48-3.37)] with respect to patients with 1 or no protective alleles [4.20 (2.48-7.12)], p=0.007. Patients with severe aortic manifestations (aortic Z-score ≥2 or aortic surgery) showed a significantly lower prevalence of subjects with 2 or more protective alleles included in the genetic score (29.7%) than patients with no or milder CV involvement (63.6%, p=0.029). The genetic score protective effect on global aortic manifestations severity (aortic Z-score ≥2 or aortic surgery) was also observed at the logistic regression analysis adjusted for the presence of FBN1gene mutations [OR=0.21 (95% CI 0.05-0.84), p=0.028]. CONCLUSIONS: our data reappraise the role of TGFBR1 and TGFBR2 as major genes in MFS patients, while suggest that TGFBR1/2 genetic variants (in particular when evaluated as a burden by score) might play a role in modulating CV manifestation severity in MFS. PART II BACKGROUND: BAV is the most common congenital heart defect. Aortic dilatation is a common feature of BAV and an increased risk of aortic dissection has been reported, raising concerns for the proper timing of aortic surgery in these patients. Familial clustering has also been reported, mostly with an autosomal pattern of inheritance with reduced penetrance and variable expressivity. Actually, the heritability of BAV has been estimated to be 89%, thus suggesting the presence of a relevant genetic contribution. Furthermore, BAV has been described as an isolated trait or associated with other clinical manifestations in syndromic conditions, thus the identification of a syndromic condition in a BAV patient is clinically relevant in order to personalize indication to aortic surgery. Differences in the anatomic site of aortic wall vulnerability in MFS and BAV, were evidenced: the maximal aortic dilatation is observed at the level of the proximal thoracic ascending aorta in BAV patients, while it is mainly referred to the aortic root in MFS patients. Therefore, due to differences and similarities between BAV and MFS aortic wall pathology, the pathogenetic mechanisms underlying these conditions are still matter of debate. MATERIAL AND METHODS: 79 BAV patients were enrolled during the three years of the project. Fifteen out of the 79 BAV patients were analyzed by next generation sequencing (NGS). The NGS analysis available in the Center is a targeted sequencing of 91 genes known or with plausibility to be associated with connective tissue disorders. Whenever possible, we performed mutational screening on the first-degree family members of the probands (3 families). RESULTS: Data of this thesis on a cohort of 79 consecutive BAV patients confirmed that patients have a strong male predominance (~75%) and come to the attention of the cardiologists and have BAV diagnosis at a median age of 42 years. This datum underlines the usefulness in this moment of the patients clinical history for a differential diagnosis of the aortopathy that eventually accompanies the BAV diagnosis. In fact the identification of the presence of BAV in a patients with diagnosis of MFS as well as other related connective tissue disorders results in a different management of patients, in particular concerning the timing of aortic surgery. This issue is particular importance due to the fact that 1) in agreement with our and literature previous data, 46.7% of BAV patients in this study have Z-score ≥2 at the aortic root level; and 2) even if patients with ≥7 points of systemic score [calculated according to revised Ghent criteria] were significantly lower in BAV patients than in MFS patients or BAV/MFS patients, the analysis of systemic features showed that BAV patients have at least in part common systemic manifestations with MFS patients furtherly complicating, together with the possible presence of aortic aneurysm, the differential diagnosis between MFS and BAV. The targeted NGS approach, allowing the sequencing of coding region and adiacent regions of consensus for exon splicing of 91 genes (associated with MFS and related disorders, bicuspid aortic valve, aortopathies and aortcic wall remodeling), on the 15 BAV patients who underwent mutational screening identified 78 genetic variants with MAF<0.05 in the European populations (where available): 43 with a MAF<0.01 and 34 called damaging with at least 1 in silico prediction tool. Genes previously suspected to be associated with the disease (NOTCH1, FBN1, TGFB1) and genes not previously associated with the disease (LTBP1, 4) were identified, not all of them classified as damaging according to the applied in silico prediction tools. Family members of 3 probands were also analysed. In some cases, the pathogenetic mutation was detected in apparently healthy family members but not in the affected ones. CONCLUSIONS:data obtained in these first 3 analyzed families suggest that no major pathogenetic mutations in genes previously associated with BAV as well as in novel genes of the 91 included in our NGS panel are associated as major genetic determinants with this clinical phenotype in its isolated form as well as BAV/TAA. This datum together with those obtained in our 15 BAV patients and other previously commented literature data suggest that BAV often perceived as monogenic or oligogenic disorder might be declined as a complex multifactorial genetic trait due to the interaction of individual genetic profile, epigenetic effects, somatic mutations, and environmental factors.
APA, Harvard, Vancouver, ISO, and other styles
2

Rocha, Tatiana Marques Ferreira da. "Associação entre polimorfismos nos genes SLC2A1, SLC2A2, HNF1A, TGFB1 e DCP1A e nefropatia em portadores de diabetes mellitus tipo 1." Universidade de São Paulo, 2013. http://www.teses.usp.br/teses/disponiveis/5/5135/tde-26032013-141528/.

Full text
Abstract:
A nefropatia diabética (ND) decorre da hiperglicemia crônica, de fatores de risco como a hipertensão arterial e a dislipidemia e de uma susceptibilidade genética já evidenciada em inúmeros estudos clínicos. Uma das características histológicas da ND é o acúmulo de proteínas de matriz extracelular no mesângio, para o qual contribuem várias vias bioquímicas. O GLUT-1, codificado pelo gene SLC2A1, é o principal transportador de glucose da célula mesangial e sua expressão está aumentada no glomérulo de animais diabéticos, o que constitui uma alça de feedback positivo pela qual a glicose extracelular aumentada estimula ainda mais sua própria captação, piorando a lesão mesangial. O GLUT-2, codificado pelo gene SLC2A2, é expresso nas células tubulares e nos podócitos e sua expressão também está aumentada na ND. A expressão deste transportador de glicose é regulada pelo fator de transcrição HNF-1. Participa, ainda, da lesão renal induzida pela hiperglicemia o fator de crescimento transformante - (TGF-), que exerce vários efeitos deletérios, tais como diminuir a atividade de metaloproteinases de matriz e promover fibrose renal. Esse fator de crescimento determina a ativação transcricional de genes-alvo, mas necessita de outros ativadores e co-ativadores da transcrição, tais como a proteína SMIF, codificada pelo gene DCP1A. Tendo em vista a participação das proteínas mencionadas acima na patogênese da ND, o presente estudo teve o objetivo de avaliar a associação de polimorfismos de um único nucleotídeo (SNPs) nos genes SLC2A1, SLC2A2, HNF1A, TGFB1 e DCP1A com a doença renal em portadores de diabetes mellitus tipo 1 (DM1). Um total de 449 pacientes (56,4% do sexo feminino, idade média de 36,0±11,0 anos) com mais de 10 anos de doença foram incluídos e classificados de acordo com o estágio de ND: (1) Ausência de ND: excreção urinária de albumina (EUA) normal (< 30 mg/24h ou < 20 g/min) e creatinina plasmática < 1,7 mg/dL sem tratamento anti-hipertensivo; (2) ND incipiente: microalbuminúria (EUA de 30 299 mg/24h ou 20 199 g/min) e creatinina plasmática < 1,7 mg/dL sem tratamento anti-hipertensivo e (3) ND Franca: macroalbuminúria (EUA > 300 mg/24h ou > 200 g/min) ou proteinúria ou tratamento para reposição renal. Também foram avaliadas as associações dos SNPs com o ritmo de filtração glomerular estimado (RFGe). Os SNPs foram genotipados pela metodologia de reação em cadeia da polimerase em tempo real, com o uso de sondas fluorescentes. As associações dos SNPs com a ND foram avaliadas por análise de regressão logística e os odds ratios (OR) e respectivos intervalos de confiança (IC) de 95% foram calculados após ajuste para possíveis confundidores, que foram incluídos como co-variáveis no modelo de regressão. Valores de P < 0.05 (bicaudal) foram considerados estatisticamente significantes. As seguintes associações foram observadas: (1) gene SLC2A1: genótipos CT+TT do SNP rs841848 conferiram risco para a ND incipiente na população global (OR 1,88; CI95% 1,06-3,34; P= 0,03) e nos pacientes do sexo masculino (OR 2,67; CI95% 1,13-6,35; P=0,0247) e para a ND franca (OR 2,70; CI95% 1,18-6,31; e P= 0,0197) apenas nos pacientes do sexo masculino; genótipos GA+AA do SNP rs1385129 conferiram risco para a ND franca na população do sexo masculino (OR 3,09; CI95% 1,34-7,25; P=0,0085); genótipos AT + TT do SNP rs3820589, conferiram proteção contra a ND incipiente na população global (OR 0,36; CI95% 0,16-0,78; P=0,0132) e na população do sexo feminino (OR 0,14; CI95% 0,02-0,52; P=0,0122). (2) gene SLC2A2: genótipos GA+GG do SNP rs5396 conferiram proteção contra ND franca nos pacientes do sexo masculino (OR 0,29; CI95% 0,12-0,69; P=0,0052); os genótipos AG+GG do SNP rs6800180 conferiram proteção contra a ND franca nos pacientes do sexo masculino (OR 0,16; CI95% 0,14-0,90; P=0,0324). (3) gene HNF1A: genótipos AC + CC do SNP rs1169288 conferiram risco para ND franca na população global (OR 2,23; CI95% 1,16-4,38; P=0,0175); genótipos CG+GG do SNP rs1169289 conferiram risco para ND franca na população global (OR 3,43; CI95% 1,61-7,73; P=0,002); (4) Gene TGFB1: genótipos CT + TT do SNP 1800468 conferiram risco para ND incipiente na população total (OR 2,99; CI95% 1,26-7,02; P 0,0116) e o alelo polimórfico T do SNP rs1800469 conferiu risco para um menor RFGe (p=0,0271). (5) gene DCP1A: o alelo polimórfico A do SNP rs11925433 também se associou com um menor RFGe (p=0,0075). Em conclusão, SNPs em genes que codificam as proteínas envolvidas na patogênese da ND GLUT-1, GLUT-2, HNF-1, TGF- e SMIF conferem susceptibilidade para essa complicação crônica nos portadores de DM1 avaliados no presente estudo
Diabetic nephropathy (DN) results from chronic hyperglycemia, risk factors such as hypertension and dyslipidemia as well as from genetic susceptibility, already demonstrated in numerous clinical studies. A histological feature of DN is the accumulation of extracellular matrix proteins in the mesangium after activation of multiple biochemical pathways. GLUT-1, encoded by gene SLC2A1, is the major glucose transporter in mesangial cell and its expression is increased in the glomeruli of diabetic animals, comprising a positive feedback loop whereby high extracellular glucose stimulates its own uptake and worsening mesangial injury. GLUT-2, encoded by SLC2A2 gene, is expressed in podocytes and tubular cells and its expression is also increased in DN. The expression of this glucose transporter is regulated by the transcription factor HNF-1. Transforming growth factor - (TGF-) also participates in renal injury induced by hyperglycemia, exerting several deleterious effects, such as to decrease the activity of matrix metalloproteinases and to promote renal fibrosis. This growth factor determines the transcriptional activation of target genes, but needs other activators and co-activators, such as the protein named SMIF, encoded by the gene DCP1A. Given the involvement of the aforementioned proteins in the pathogenesis of DN, the present study aimed to evaluate the association of single nucleotide polymorphisms (SNPs) in the genes SLC2A1, SLC2A2, HNF1A, TGFB1 e DCP1A with renal disease in patients with type 1 diabetes mellitus (T1DM). A total of 449 patients (56.4% female, mean age 36.0±11.0 years) with disease duration > 10 years were included and grouped according to DN stages: (1) absence of DN: normal urinary albumin excretion (UAE) (< 30 mg/24h or < 20 g/min) and plasmatic creatinine < 1.7 mg/dL without antihypertensive treatment; (2) incipient DN: microalbuminuria (UAE 30 299 mg/24h or 20 199 g/min) and plasmatic creatinine < 1.7 mg/dL without antihypertensive treatment and (3) overt DN: macroalbuminúria (UAE > 300 mg/24h or > 200 g/min) or proteinuria or renal replacement therapy. Associations of SNPs with estimated glomerular filtration rate (eGFR) were also evaluated. All SNPs were genotyped by real time polymerase chain reaction using fluorescent-labelled probes. Associations of the SNPs with DN were assessed by logistic regression analyses and odds ratios (OR) were calculated after adjustments for possible confounders included as covariables in the regressive model. P values <0.05 (two-tails) were considered significant. The following associations were observed: (1) SLC2A1: genotypes CT+TT from rs841848 conferred risk to incipient DN in the overall population (OR 1.88; 95%IC 1.06-3.34; P= 0.03) and in the male patients (OR 2.67; CI95% 1.13-6.35; P=0.0247) and to overt DN (OR 2.70; CI95% 1.18-6.31; e P= 0.0197) only in the male patients; genotypes GA+AA from rs1385129 conferred risk to overt DN in the male population (OR 3.09; CI95% 1.34-7.25; P=0.0085); genotypes AT + TT from rs3820589 conferred protection against incipient DN in the overall population (OR 0.36; CI95% 0.16-0.78; P=0.0132) and in the female population (OR 0.14; CI95% 0.02-0.52; P=0.0122). (2) SLC2A2: genotypes GA+GG from rs5396 conferred protection against overt DN in the male patients (OR 0.29; CI95% 0.12-0.69; P=0.0052); genotypes AG+GG from rs6800180 conferred protection against overt DN in the male patients (OR 0.16; CI95% 0.14-0.90; P=0.0324). (3) HNF1A: genotypes AC + CC from rs1169288 conferred risk to overt DN in the overall population (OR 2.23; CI95% 1.16-4.38; P=0.0175); genotypes CG+GG from rs1169289 conferred risk to overt DN in the overall population (OR 3.43; CI95% 1.61-7.73; P=0.002); (4) TGFB1: genotypes CT + TT from 1800468 conferred risk to incipient DN in the overall population (OR 2.99; CI95% 1.26-7.02; P=0.0116) and the polymorphic allele T from SNP rs1800469 conferred risk to a lower eGFR (p=0.0271). (5) DCP1A: the polymorphic allele A from SNP rs11925433 was also associated with a lower eGFR (p=0.0075). In conclusion, SNPs in the genes encoding proteins GLUT-1, GLUT-2, HNF-1, TGF- e SMIF, all involved in the pathogenesis of DN, conferred susceptibility to this chronic complication in the T1DM patients evaluated in the present study
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography