Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Mediators Inflamm ; 2020: 3185015, 2020.
Article in English | MEDLINE | ID: mdl-33149723

ABSTRACT

Individuals with sickle cell disease (SCD) present both chronic and acute inflammatory events. The TGF-ß pathway is known to play a role in immune response, angiogenesis, inflammation, hematopoiesis, vascular inflammation, and cell proliferation. Polymorphisms in the transforming growth factor-beta receptor 3 (TGFBR3) gene have been linked to several inflammatory diseases. This study investigated associations between two TGFBR3 haplotypes and classical laboratory parameters, as well as clinical manifestations, in SCD. We found that individuals with the GG haplotype presented higher levels of total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), triglycerides, non-HDL cholesterol, total proteins, and globulin than individuals with non-GG haplotypes. In addition, the GG haplotype was associated with a previous history of pneumonia. Individuals with the CGG haplotype presented increased plateletcrit, TC, LDL-C levels, and non-HDL cholesterol. The CCG haplotype was also associated with a previous history of pneumonia. Our findings suggest that individuals with the GG and CGG haplotypes of TGFBR3 present important alterations in lipid profile.


Subject(s)
Anemia, Sickle Cell/blood , Anemia, Sickle Cell/genetics , Haplotypes , Hemoglobins/metabolism , Lipids/chemistry , Polymorphism, Single Nucleotide , Proteoglycans/genetics , Receptors, Transforming Growth Factor beta/genetics , Adolescent , Biomarkers/metabolism , Cell Proliferation , Child , Cholesterol/metabolism , Cholesterol, LDL , Female , Genotype , Humans , Inflammation , Linkage Disequilibrium , Male , Pneumonia/metabolism , Prognosis , Proteoglycans/blood , Receptors, Transforming Growth Factor beta/blood , Young Adult
2.
Mol Biol Rep ; 47(9): 6693-6703, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32803503

ABSTRACT

Depletion of S-adenosyl methionine and 5-methyltetrahydrofolate; and elevation of total plasma homocysteine were documented in CAD patients, which might modulate the gene-specific methylation status and alter their expression. In this study, we have aimed to delineate CAD-specific epigenetic signatures by investigating the methylation and expression of 11 candidate genes i.e. ABCG1, LIPC, PLTP, IL-6, TNF-α, CDKN2A, CDKN2B, F2RL3, FGF2, P66 and TGFBR3. The methylation-specific PCR and qRT-PCR were used to assess the methylation status and the expression of candidate genes, respectively. CAD patients showed the upregulation of IL-6, TNF-α, CDKN2A, CDKN2B, F2RL3, FGF2, P66, and TGFBR3. Hypomethylation of CDKN2A loci was shown to increase risk for CAD by 1.79-folds (95% CI 1.22-2.63). Classification and regression tree (CART) model of gene expression showed increased risk for CAD with F2RL3 > 3.4-fold, while demonstrating risk reduction with F2RL3 < 3.4-fold and IL-6 < 7.7-folds. This CAD prediction model showed the excellent sensitivity (0.98, 95% CI 0.88-1.00), specificity (0.91, 95% CI 0.86-0.92), positive predictive value (0.82, 95% CI 0.75-0.84), and negative predictive value (0.99, 95% CI 0.94-1.00) with an overall accuracy of 92.8% (95% CI 87.0-94.1%). Folate and B12 deficiencies were observed in CAD cases, which were shown to contribute to hypomethylation and upregulation of the prime candidate genes i.e. CDKN2A and F2RL3. Early onset diabetes was associated with IL-6 and TNF-α hypomethylation and upregulation of CDKN2A. The expression of F2RL3 and IL-6 (or) hypomethylation status at CDKN2A locus are potential biomarkers in CAD risk prediction. Early epigenetic imprints of CAD were observed in early onset diabetes. Folate and B12 deficiencies are the contributing factors to these changes in CAD-specific epigenetic signatures.


Subject(s)
Coronary Artery Disease/metabolism , DNA Methylation , Epigenesis, Genetic , Adult , Biomarkers/blood , Coronary Artery Disease/genetics , Correlation of Data , Cyclin-Dependent Kinase Inhibitor p15/blood , Cyclin-Dependent Kinase Inhibitor p16/blood , Demography , Diabetes Mellitus/blood , Female , Fibroblast Growth Factor 2/blood , Folic Acid/blood , Folic Acid Deficiency , Humans , Interleukin-6/blood , Male , Middle Aged , Proteoglycans/blood , Receptors, Thrombin/blood , Receptors, Transforming Growth Factor beta/blood , Regression Analysis , Risk Factors , Tumor Necrosis Factor-alpha/blood
3.
Minerva Pediatr ; 72(4): 278-287, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32418410

ABSTRACT

Distinguishing between constitutional delay of growth and puberty (CDGP) and congenital hypogonadotropic hypogonadism (CHH) may be challenging. CDGP and CHH appear to belong to the same clinical spectrum (with low sex hormones and low LH and FSH), although one is classically transient and known as a self-limited form of delayed puberty (CDGP) while the other is permanent (CHH). Thus, the clinical history and the outcomes of these two conditions require different approaches, and an adequate and timely management for the patients is mandatory. Since the initial presentation of CDGP and CHH is almost identical and given the similarities of CDGP and partial forms of CHH (i.e. patients with partial and early interrupted pubertal development) the scientific community has been struggling to find some diagnostic tests able to allow an accurate differential diagnosis between these two conditions in delayed puberty. In this review we provide an up to date insight on the tests available, their meanings and accuracy, as well as some clues to effectively differentiate between constitutional pubertal delay and pathologic CHH.


Subject(s)
Growth Disorders/diagnosis , Hypogonadism/diagnosis , Puberty, Delayed/diagnosis , Diagnosis, Differential , Female , Follicle Stimulating Hormone/blood , Humans , Hypogonadism/blood , Hypogonadism/congenital , Hypogonadism/genetics , Inhibins/blood , Insulin/blood , Kisspeptins/blood , Luteinizing Hormone/blood , Male , Proteins , Puberty, Delayed/etiology , Puberty, Delayed/genetics , Receptors, Peptide/blood , Receptors, Transforming Growth Factor beta/blood , Sex Factors , Time Factors
4.
Reprod Biomed Online ; 39(6): 1017-1025, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31727499

ABSTRACT

RESEARCH QUESTION: The ectodomain of the anti-Müllerian hormone (AMH) type 2 receptor is shed by proteases under certain conditions, which makes it measurable in the blood. The aim of this study was to identify correlations of soluble anti-Müllerian hormone receptor type 2 (sAMHR2) with other sex hormone concentrations and to assess whether sAMHR2 may serve as a new biomarker in fertility disorders. DESIGN: In a retrospective cross-sectional study of women (n = 186) with different gynaecological-endocrinological disorders, mixed-effect models were used to analyse the correlation with established diagnostic hormone tests. Receiver operating characteristic curve analysis was performed to assess the diagnostic performance. RESULTS: There was a strong correlation of sAMHR2 with LH (r = 0.898) and FSH (r = 0.846) and a moderate correlation of AMH with testosterone (r = 0.666) and androstenedione (r = 0.696) (all P < 0.001). In diagnoses of polycystic ovary syndrome (PCOS), AMH showed the best performance (area under the curve [AUC] 0.981, cut-off 4 ng/ml) with 96% sensitivity and 94% specificity. sAMHR2 concentrations and sAMHR2/AMH ratios were elevated in women with ovarian insufficiency, compared with all other study groups, including post-menopausal women on hormone replacement therapy. Highest sensitivity and specificity (100% and 98.2%, respectively) were achieved with sAMHR2/AMH ratio for the diagnosis of post-menopausal status (cut-off 68.85). The sAMHR2/AMH ratio (AUC 0.997) had a better performance than sAMHR2 (AUC 0.947), FSH (AUC 0.989) and LH (AUC 0.967). CONCLUSIONS: The sAMHR2/AMH ratio may serve as a useful biomarker for infertility diagnostics to identify post-menopausal women.


Subject(s)
Infertility, Female/blood , Receptors, Peptide/blood , Receptors, Transforming Growth Factor beta/blood , Adolescent , Adult , Biomarkers/blood , Cross-Sectional Studies , Female , Humans , Middle Aged , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/diagnosis , Postmenopause/blood , Retrospective Studies , Young Adult
5.
J Assist Reprod Genet ; 36(6): 1281-1289, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31089932

ABSTRACT

OBJECTIVE: Our study aimed to investigate the relationship between polymorphisms (Apa1, Bsm1, Fok1, and Cdx2) in the VDR gene as well as AMH and AMHR2 genes and their influence on AMH and 25(OH)D levels in PCOS women. STUDY DESIGN: Seventy-five patients with PCOS and 23 control women were included. Serum AMH and 25(OH)D levels in patients and controls were measured by enzyme-linked immunosorbent assay (ELISA). Polymorphisms in VDR gene Fok1 C/T (rs2228587), Bsm1 A/G (rs1544410), Apa1 A/C (rs7975232), and Cdx2 A/G (rs11568820) polymorphisms as well as AMH G/T (rs10407022) and AMHR2 A/G (rs2002555) were analyzed using real-time PCR. RESULTS: Analysis of the VDR Cdx2 polymorphism showed a significantly higher frequency of the homozygous GG (mutant) genotype in the PCOS group as compared with the control group (p < 0.05). The analysis revealed a statistically significant correlation between the presence of FokI and ApaI polymorphisms and AMH levels in PCOS women (p < 0.05). The presence of mutant genotypes (CT, TT) in the Fok1 and (CA, CC) in the Apa1 polymorphisms were associated with higher AMH level in PCOS women (p < 0.05). No statistically significant correlations between AMH and AMHR2 polymorphisms and AMH level were found. Moreover, there was no correlation between AMH and 25(OH)D levels in the PCOS or in the control group. CONCLUSION: It seems that the elevated AMH level is associated with VDR Fokl and Apal polymorphisms, but not with 25(OH)D levels in PCOS women. Further research is needed to determine the role of VDR polymorphism in AMH level in PCOS.


Subject(s)
Anti-Mullerian Hormone/blood , Polycystic Ovary Syndrome/blood , Receptors, Calcitriol/blood , Receptors, Peptide/blood , Receptors, Transforming Growth Factor beta/blood , Adult , Anti-Mullerian Hormone/genetics , Female , Genotype , Humans , Ovulation/genetics , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/physiopathology , Polymorphism, Single Nucleotide/genetics , Receptors, Calcitriol/genetics , Receptors, Peptide/genetics , Receptors, Transforming Growth Factor beta/genetics , Vitamin D/blood
6.
Horm Mol Biol Clin Investig ; 38(1)2019 Mar 25.
Article in English | MEDLINE | ID: mdl-30904901

ABSTRACT

Background Infertile women may have underlying genetic abnormalities. There is, at present, a significant number of studies on the relation between the follicle stimulating hormone receptor (FSHR) or anti-Müllerian hormone type II receptor (AMHRII) polymorphisms and response to in-vitro fertilisation (IVF) treatment. However, it is not yet clear which genotype or combination of genotypes is favourable towards a better ovarian stimulation and pregnancy outcome. Materials and methods In this study we assessed the distribution of the genotypes of FSHR Ser680Asn and of AMHRII -482A>G gene polymorphisms in a group of 126 infertile women and a control group of 100 fertile women by using real-time polymerase chain reaction (RT-PCR). Results Statistical analysis showed that the frequency of the genotypes is similar in both control and IVF/ intracytoplasmic sperm injection (ICSI) groups. Further investigation of the frequency of the nine possible combinations of these polymorphisms in the groups revealed no correlation between infertility and combination of the polymorphisms. Women with one polymorphism have on average 5.5 units higher levels of AMH compared to women carrying no polymorphism. In women with no polymorphisms, for each unit of FSH increase, the average concentration of blood AMH is expected to be 72% lower. Conclusion The distribution of the FSHR Ser680Asn and of the AMHRII -482A>G gene polymorphisms, in the Greek population is similar in fertile and infertile women. The study showed that FSH and AMH correlated levels in certain cases could be used to estimate a patient's ovarian reserve.


Subject(s)
Infertility, Female/genetics , Polymorphism, Single Nucleotide , Receptors, FSH/genetics , Receptors, Peptide/genetics , Receptors, Transforming Growth Factor beta/genetics , Adult , Female , Humans , Infertility, Female/therapy , Ovarian Reserve , Receptors, FSH/blood , Receptors, Peptide/blood , Receptors, Transforming Growth Factor beta/blood , Sperm Injections, Intracytoplasmic/statistics & numerical data
7.
Heart Surg Forum ; 22(6): E432-E437, 2019 10 09.
Article in English | MEDLINE | ID: mdl-31895025

ABSTRACT

OBJECTIVE: The aim of this study was to observe the impacts of the specific cyclooxygenase-2 inhibitor celecoxib on cardiac structures, functions, and inflammatory factors during the process of pressure overload-induced myocardial hypertrophy. METHODS: Twenty-four male Sprague Dawley rats were randomly divided into 3 groups: the sham operation group, the surgery group, and the celecoxib group. The model was established according to the abdominal aortic coarctation method. RESULTS: At 16 weeks, rats in the celecoxib group were fed a celecoxib-mixed diet (10 mg/kg) for 8 consecutive weeks. At week 24 after model establishment, the cardiac structures and functions were observed; changes in the levels of tumor necrosis factor (TNF)-α, transforming growth factor (TGF)-ß, prostaglandin E2 (PGE2), C-reactive protein (CRP), and uric acid (UA) were detected; and the contents of Smad1/2/3 proteins (Smad1, Smad2, and Smad3)  were determined. Left ventricular mass index, the heart weight/body weight ratio, and TNF-α, TGF-ß, PGE2, CRP, and UA levels of the celecoxib group were all significantly decreased relative to those of the surgery group (P < .05); moreover, the cardiac functions were significantly improved compared to those of the surgery group (P < .05). CONCLUSIONS: These results show that inflammatory factors are involved in the myocardial hypertrophy process and that celecoxib may reverse myocardial hypertrophy through a variety of pathways.


Subject(s)
Cardiomegaly/pathology , Cardiomegaly/physiopathology , Celecoxib/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Animals , C-Reactive Protein/metabolism , Cardiomegaly/blood , Cardiomegaly/drug therapy , Diet , Dinoprostone/blood , Disease Models, Animal , Drug Administration Schedule , Heart/drug effects , Male , Organ Size , Random Allocation , Rats, Sprague-Dawley , Receptors, Transforming Growth Factor beta/blood , Smad1 Protein/blood , Smad2 Protein/blood , Smad3 Protein/blood , Tumor Necrosis Factor-alpha/blood , Uric Acid/blood
8.
J Clin Endocrinol Metab ; 102(8): 3065-3074, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28633389

ABSTRACT

Context: Preeclampsia (PE) can be classified into early-onset (<34 weeks of gestation) and late-onset (>34 weeks of gestation) subtypes. Soluble endoglin, an auxiliary receptor for transforming growth factor (TGF)-ß ligands, is increased in PE circulation and believed to inhibit TGF-ß action by sequestering the ligands. However, soluble endoglin, with a low affinity to TGF-ß ligands, has been demonstrated to have little effect by itself on TGF-ß action. Objectives: We examined whether multiple soluble TGF-ß receptors are elevated in PE circulation and whether they synergistically block TGF-ß signaling. Design: TGF-ß receptors were measured using enzyme-linked immunosorbent assay in sera collected from preeclamptic pregnancies and gestation-age-matched controls. TGF-ß signaling was assessed using an in vitro bioassay and a tube formation assay. Results: TGF-ß type I, II, and III receptors were all identified in pregnant serum; all were substantially elevated in early-onset but not late-onset PE. Endoglin was increased in both subtypes. At the greatest concentrations detected in PE, none of these soluble TGF-ß receptors alone, including endoglin, inhibited TGF-ß signaling. However, when all four soluble receptors were present, signaling of both TGF-ß1 and TGF-ß2 was substantially reduced. Removal of any one of these soluble receptors alleviated TGF-ß1 inhibition; however, removal of soluble TGFßRIII was necessary to relieve TGF-ß2 inhibition. Conclusions: Multiple soluble TGF-ß receptors are present in pregnant circulation and elevated in early-onset PE; they synergistically inhibit TGF-ß signaling, which might be more likely to occur in early-onset than late-onset PE. Reducing soluble TGFßRIII, rather than endoglin, would be more effective in alleviating the inhibition of both TGF-ß1 and TGF-ß2 signaling in PE.


Subject(s)
Endoglin/blood , Pre-Eclampsia/blood , Protein Serine-Threonine Kinases/blood , Proteoglycans/blood , Receptors, Transforming Growth Factor beta/blood , Transforming Growth Factor beta/metabolism , Adult , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Female , Gestational Age , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , In Vitro Techniques , Pre-Eclampsia/metabolism , Pregnancy , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Signal Transduction , Young Adult
9.
Int J Gynaecol Obstet ; 137(3): 246-252, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28281288

ABSTRACT

OBJECTIVE: To assess circulating levels of transforming growth factor (TGF)-ß superfamily members and their soluble receptors in hypertensive disorders of pregnancy, and to investigate associations with clinical manifestations. METHODS: A retrospective study was conducted using data for women admitted to a center in China for delivery between May 2011 and April 2013. Women with severe pre-eclampsia, mild pre-eclampsia, and gestational hypertension were included, along with a control group. Serum levels of activin A, inhibin A, TGF-ß1, soluble endoglin (sEng), and soluble betaglycan (sBG) were measured. RESULTS: Women with severe pre-eclampsia (n = 17) had higher mean levels of activin A (23.5±2.1 µg/L), inhibin A (1.7±0.2 µg/L), sEng (32.1±3.2 µg/L), and sBG (84.1±9.4 µg/L) than did normotensive controls (n = 18), women with gestational hypertension (n = 15), and those with mild pre-eclampsia (n = 14; all P<0.05). Women with early-onset pre-eclampsia (n = 13) had higher levels of these serum markers than did preterm normotensive controls (n = 8; all P<0.001). Women with severe or early-onset pre-eclampsia had the lowest TGF-ß1 levels. Activin A, inhibin A, sEng, and sBG levels were positively correlated with mean arterial pressure and proteinuria (all P<0.01). CONCLUSION: Pre-eclampsia is associated with an imbalance of members of the TGF-ß superfamily and their soluble receptors, which might contribute to the development of pre-eclampsia and help to predict onset and severity.


Subject(s)
Hypertension, Pregnancy-Induced/blood , Receptors, Transforming Growth Factor beta/blood , Transforming Growth Factor beta/blood , Activins/blood , Adult , Endoglin/blood , Female , Humans , Inhibins/blood , Pre-Eclampsia/blood , Pregnancy , Proteoglycans/blood , Retrospective Studies , Transforming Growth Factor beta1/blood
10.
Pediatr Res ; 80(5): 759-765, 2016 11.
Article in English | MEDLINE | ID: mdl-27603563

ABSTRACT

BACKGROUND: Growth factors (GFs) are milk bioactive components contributing to the regulation of neonatal small intestinal maturation, and their receptors on the small intestinal epithelium play essential roles in mediating the functions of GFs. There is limited data correlating milk GFs and their receptors in the neonatal small intestine during the perinatal period. METHODS: Small intestines of C57BL/6N mouse pups were collected at regular intervals during fetal life and up to postnatal day (PD) 60. Gene expression of GF receptors was determined by real-time qPCR. Milk GF concentrations up to PD21 were analyzed by enzyme-linked immunosorbent assay. RESULTS: The majority of GF receptors showed significantly greater expression in the fetus than in postnatal life, and a sharp decrease occurred from PD14 extending to PD60; solid food restriction (PD14 and PD18) did not affect this decrease. Concentrations of five detected milk GFs demonstrated that GFs and the corresponding small intestinal receptors exhibited different correlations, with only milk transforming growth factor ß1 (TGF-ß1) having a significant positive correlation with TGF-ß receptor 1 mRNA. CONCLUSION: Gene expression of small intestinal GF receptors is likely a process of neonatal intestinal maturation that is affected concurrently by milk GFs and additional endogenous factors.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Intestine, Small/metabolism , Milk/chemistry , Animals , Animals, Newborn , ErbB Receptors/blood , Female , Gene Expression Regulation, Developmental , Intestine, Small/embryology , Intestine, Small/growth & development , Lactation , Male , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-met/blood , Receptor, IGF Type 1/blood , Receptor, Insulin/blood , Receptor, Nerve Growth Factor/blood , Receptors, Fibroblast Growth Factor/blood , Receptors, Platelet-Derived Growth Factor/blood , Receptors, Transforming Growth Factor beta/blood , Vascular Endothelial Growth Factor Receptor-1/blood
11.
J Transl Med ; 14(1): 202, 2016 07 04.
Article in English | MEDLINE | ID: mdl-27378305

ABSTRACT

BACKGROUND: Increased leucine-rich α2-glycoprotein-1 (LRG1) has been observed in plasma of individuals with various diseases. However, the role of LRG1 in allergic airway disease has not been investigated. OBJECTIVE: To explore the involvement of LRG1 in allergy and its cell origins. METHODS: The expression levels of LRG1 and its receptor transforming growth factor-beta receptor II (TGFBR2) in patients with allergic rhinitis (AR) and asthma (AS) were examined by flow cytometry, and enzyme-linked immunosorbent assay (ELISA). RESULTS: LRG1 and soluble TGFBR2 expression in plasma of patients with AR and AS were markedly lower than that of healthy control (HC) subjects. Large proportions of CD123 + HLA-DR-, CD16+, CD4+, CD8+, CD14+, and CD19+ cells expressed LRG1, although the percentages of LRG1+ cells in these cell populations were lower in AR and AS patients. Up to 89.8 and 15.5 % of dispersed mast cells expressed LRG1 and TGFBR2. Moreover, allergen extract exposure significantly reduced LRG1 and TGFBR2 expression in the plasma and leukocytes of patients with AR and AS. CONCLUSIONS: Reduced LRG1 and TGFBR2 levels in patients with allergic airway disorders are likely caused by inhibitory actions of allergens in LRG1 producing cells. Thus, LRG1 may be a key regulatory factor of allergic responses.


Subject(s)
Asthma/blood , Down-Regulation , Glycoproteins/blood , Organ Specificity , Peripheral Blood Stem Cells/metabolism , Rhinitis, Allergic/blood , Adolescent , Adult , Aged , Allergens/immunology , Case-Control Studies , Cell Line , Female , Flow Cytometry , Humans , Male , Mast Cells/metabolism , Middle Aged , Protein Serine-Threonine Kinases/blood , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/blood , Time Factors , Tryptases/blood , Young Adult
12.
Int J Environ Res Public Health ; 13(2): 185, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26848671

ABSTRACT

The objective of this work was the analysis of the relationships between the genotypes of the AMH and AMH receptor type 2 genes, hormone levels and the menstrual cycle in a group of Polish women in the late reproductive stage. The study was conducted using a measurement-based method (body weight and height), laboratory method (serum hormone levels AMH, FSH and E2), and genetic analysis (DNA isolated from whole blood by a salting-out method). The study involved 345 healthy, late-reproductive-stage women from Poland, aged 42.3 ± 4.5 years. The analysis demonstrated that neither the T/T and G/T+G/G genotypes of the AMH Ile(49)Ser polymorphism (rs10407022), nor the A/A and the G/A + G/G genotypes of the AMHR2 2482 A > G polymorphism (rs2002555), nor the C/C and C/T + T/T genotypes of the AMH polymorphism (rs11170547) were statistically significantly related (p > 0.05) to such factors as age, BMI, hormone (FSH and E2) levels and ovarian parameters (AMH) in the follicular phase. No relationships were found between ovarian parameters (FSH, E2, AMH) and genetic variants of AMH (rs10407022) and AMHR2 (rs11170547, rs2002555) in healthy women in the late reproductive stage.


Subject(s)
Anti-Mullerian Hormone/genetics , Follicular Phase/genetics , Polymorphism, Single Nucleotide , Receptors, Peptide/genetics , Receptors, Transforming Growth Factor beta/genetics , Sexual Maturation/genetics , Adult , Anti-Mullerian Hormone/blood , Biomarkers/blood , Female , Follicular Phase/blood , Genetic Markers , Genotype , Humans , Middle Aged , Poland , Receptors, Peptide/blood , Receptors, Transforming Growth Factor beta/blood , Sexual Maturation/physiology
13.
Growth Factors ; 33(3): 200-9, 2015.
Article in English | MEDLINE | ID: mdl-26190421

ABSTRACT

Type III transforming growth factor (TGFß) receptor (TGFßrIII) modulates TGFß superfamily signaling. Its tumor tissue expression is downregulated in human breast cancer. We determined (indirect ELISA) plasma levels of the soluble receptor (sTGFßrIII) in 47 women with breast cancer (AJCC stages 0-IIB) (cases) pre-surgery and over two months after the surgery, and in 36 healthy women (controls). Plasma sTBFßrIII was lower in cases than in the controls (age-adjusted difference -29.7 ng/mL, p < 0.001), and discriminated between disease and health (sensitivity and specificity 100% at 16.6 ng/mL). With adjustment for age, AJCC stage, lymph node involvement, HER2 and hormone receptor status, higher pre-surgery sTBFßrIII was associated with better progression-free survival (HR = 0.68, 95%CI 0.49-0.89, p = 0.004). An increasing trend in plasma sTBFßrIII was observed over 2 months after the surgery (0.6% increase/day, p < 0.001), consistently across the patient subsets. Data suggest a high potential of plasma sTBFßrIII as a novel diagnostic and prognostic biomarker in breast cancer.


Subject(s)
Biomarkers, Tumor/blood , Breast Neoplasms/diagnosis , Breast Neoplasms/mortality , Proteoglycans/blood , Receptor, ErbB-2/metabolism , Receptors, Transforming Growth Factor beta/blood , Adult , Aged , Aged, 80 and over , Breast Neoplasms/therapy , Disease-Free Survival , Enzyme-Linked Immunosorbent Assay , Female , Follow-Up Studies , Humans , Lymphatic Metastasis/pathology , Middle Aged , Proteoglycans/genetics , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction/genetics
14.
Am J Med Genet A ; 167A(10): 2435-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26096872

ABSTRACT

Loeys-Dietz syndrome (LDS) is an autosomal dominant connective tissue disorder, caused by heterozygous mutations in TGFBR1 or TGFBR2 and characterized by vascular complications (cerebral, thoracic, and abdominal arterial aneurysms and/or dissections) and skeletal manifestations. We here report the first patient with LDS presenting with reversible cerebral vasoconstriction syndrome (RCVS), a clinico-radiological condition characterized by recurrent thunderclap headaches, with or without neurological symptoms, and reversible vasoconstriction of cerebral arteries. The patient was a 9-year-old boy with a heterozygous TGFBR2 mutation, manifesting camptodactyly, talipes equinovarus, and lamboid craniosynostosis. He complained of severe recurrent headaches 2 months after total aortic replacement for aortic root dilatation and a massive Stanford type B aortic dissection. A thoracic CT scan revealed a left subclavian artery dissection. Brain MRI and MRA detected bilateral internal carotid artery constriction along with a cortical subarachnoid hemorrhage without intracranial aneurysms. Subsequently, he developed visual disturbance and a generalized seizure associated with multiple legions of cortical and subcortical increased signals including the left posterior lobe, consistent with posterior reversible encephalopathy syndrome (PRES), a condition characterized by headaches, visual disorders, seizures, altered mentation, consciousness disturbances, focal neurological signs, and vasogenic edema predominantly in the white matter of the posterior lobe. Vasoconstriction of the internal carotid artery was undetectable 2 months later, and he was diagnosed as having RCVS. Endothelial dysfunction, associated with impaired TGF-ß signaling, might have been attributable to the development of RCVS and PRES.


Subject(s)
Aortic Dissection/genetics , Loeys-Dietz Syndrome/genetics , Posterior Leukoencephalopathy Syndrome/genetics , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , Subarachnoid Hemorrhage/genetics , Vasoconstriction , Aortic Dissection/blood , Aortic Dissection/pathology , Cerebral Arteries/metabolism , Cerebral Arteries/pathology , Child , Gene Expression , Heterozygote , Humans , Loeys-Dietz Syndrome/blood , Loeys-Dietz Syndrome/pathology , Male , Mutation , Posterior Leukoencephalopathy Syndrome/blood , Posterior Leukoencephalopathy Syndrome/pathology , Protein Serine-Threonine Kinases/blood , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/blood , Signal Transduction , Subarachnoid Hemorrhage/blood , Subarachnoid Hemorrhage/pathology
15.
Pediatr Cardiol ; 36(7): 1476-82, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26022443

ABSTRACT

Transforming growth factor beta receptor 2 (TGFBR2) plays a central role in normal heart development, and we investigated whether TGFBR2 polymorphism confers the risk of congenital ventricular septal defect (CVSD). The case-control study included 115 CVSD children and 188 healthy children in a Chinese population. TGFBR2 rs6785358 polymorphism was genotyped with polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). Enzyme-linked immunoassay (ELISA) was used to detect serum TGFBR2 levels. The genotype and allele frequency of TGFBR2 rs6785358 were significantly higher in the CVSD group than in the controls (all P < 0.05). The G allele carriers were associated with increased CVSD risk compared with the A allele carriers in CVSD group (OR 3.503, 95 % CI 2.670-4.596). Stratified analysis by gender revealed that the TGFBR2 rs6785358 genotype and allele frequency were significantly different between the CVSD case and controls, in both the male subgroup and the female subgroup (all P < 0.001). The G allele carriers were more susceptible to CVSD risk than the A allele carriers in both the male subgroup (OR 9.096, 95 % CI 5.398-15.33) and the female subgroup (OR 3.148, 95 % CI 1.764-5.618). Logistic regression analysis revealed that age, gender and genotype were associated with the risk of CVSD (all P < 0.05). The study findings revealed that TGFBR2 rs6785358 polymorphism contributes to CVSD susceptibility, and the G allele may increase the risk of CVSD.


Subject(s)
Heart Septal Defects, Ventricular/genetics , Polymorphism, Restriction Fragment Length , Polymorphism, Single Nucleotide , Protein Serine-Threonine Kinases/blood , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/blood , Receptors, Transforming Growth Factor beta/genetics , Alleles , Asian People , Case-Control Studies , Child , Child, Preschool , China , Female , Gene Frequency , Genetic Predisposition to Disease , Humans , Infant , Logistic Models , Male , Receptor, Transforming Growth Factor-beta Type II
16.
Endokrynol Pol ; 65(5): 348-56, 2014.
Article in English | MEDLINE | ID: mdl-25301484

ABSTRACT

INTRODUCTION: The first part of this paper is related to healthy people and presents concentrations of TGFß1 and VEGF in blood (with and without dividing data with respect to sex), their single measurement values (at 8 am), Mean Daily Concentrations (MDC), Area Under the Curves (AUC; total daily secretion), and circadian rhythm. The second part of the work is related to Graves' orbitopathy (GO). The aim of the study were: 1) to determine the physiological pattern of TGFß1 and VEGF secretion; 2) to compare the serum TGFß1 and VEGF circadian profile in newly diagnosed thyreotoxic patients with active GO and healthy controls (H); and 3) to estimate the influence of high-dose intravenous methylprednisolone pulse therapy (MP) on TGFb1 and VEGF blood levels in GO. MATERIAL AND METHODS: Twenty-two healthy (H) subjects and 16 hyperthyroid GO patients were treated with MP (6 g/14 days) and followed up by ophthalmological assessment. Blood was collected before and after 2 weeks MP-therapy. TGFß1 and VEGF levels were determined by the ELISA method. RESULTS: No difference was observed in the concentrations of TGFß1 and VEGF in the blood of healthy women and men - in further analysis, a combined healthy male and female cohort was used (H). While the absence of circadian rhythms in the concentrations of TGFß1 and VEGF allows the application of a single measurement approach, MDC and AUC measurements were found to be more precise. There were no differences in TGFß1 MDC/AUC between GO and H. VEGF MDC/AUC in GO were higher than in H. MP-therapy increased TGFb1 MDC/AUC, thus in GO after MP, the TGFß1 MDC/AUC were higher than in H. There were no differences in VEGF MDC/AUC during MP-therapy. MP-therapy was effective in 15/16 patients. CONCLUSIONS: 1. MP-therapy increases MDC and AUC of TGFß1. The effectiveness of MP-therapy in patients with active GO may be related to its influence on TGFß1 concentrations in blood. The results suggest the existence of a new mechanism of glucocorticoids action, consisting of an increase in the secretion of TGFß1.2. The elevated serum VEGF in thyreotoxic patients with active GO may reflect long-standing autoimmune processes in orbital and thyroid tissues and intensified angiogenesis in the thyroid gland.


Subject(s)
Graves Ophthalmopathy/blood , Protein Serine-Threonine Kinases/blood , Receptors, Transforming Growth Factor beta/blood , Vascular Endothelial Growth Factor A/blood , Biomarkers/blood , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Receptor, Transforming Growth Factor-beta Type I , Severity of Illness Index
17.
Neoplasia ; 16(6): 489-500, 2014 Jun.
Article in English | MEDLINE | ID: mdl-25077702

ABSTRACT

Bone morphogenetic proteins (BMPs) are members of the TGF-ß superfamily that are over-expressed in breast cancer, with context dependent effects on breast cancer pathogenesis. The type III TGF-ß receptor (TßRIII) mediates BMP signaling. While TßRIII expression is lost during breast cancer progression, the role of TßRIII in regulating BMP signaling in normal mammary epithelium and breast cancer cells has not been examined. Restoring TßRIII expression in a 4T1 murine syngeneic model of breast cancer suppressed Smad1/5/8 phosphorylation and inhibited the expression of the BMP transcriptional targets, Id1 and Smad6, in vivo. Similarly, restoring TßRIII expression in human breast cancer cell lines or treatment with sTßRIII inhibited BMP-induced Smad1/5/8 phosphorylation and BMP-stimulated migration and invasion. In normal mammary epithelial cells, shRNA-mediated silencing of TßRIII, TßRIII over-expression, or treatment with sTßRIII inhibited BMP-mediated phosphorylation of Smad1/5/8 and BMP induced migration. Inhibition of TßRIII shedding through treatment with TAPI-2 or expression of a non-shedding TßRIII mutant rescued TßRIII mediated inhibition of BMP induced Smad1/5/8 phosphorylation and BMP induced migration and/or invasion in both in normal mammary epithelial cells and breast cancer cells. Conversely, expression of a TßRIII mutant, which exhibited increased shedding, significantly reduced BMP-mediated Smad1/5/8 phosphorylation, migration, and invasion. These data demonstrate that TßRIII regulates BMP-mediated signaling and biological effects, primarily through the ligand sequestration effects of sTßRIII in normal and cancerous mammary epithelial cells and suggest that the ratio of membrane bound versus sTßRIII plays an important role in mediating these effects.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Breast Neoplasms/metabolism , Epithelial Cells/metabolism , Proteoglycans/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Movement/genetics , Disease Models, Animal , Epithelial Cells/pathology , Female , Humans , Mice , Mutation , Proteoglycans/blood , Proteoglycans/genetics , Receptors, Transforming Growth Factor beta/blood , Receptors, Transforming Growth Factor beta/genetics
18.
PLoS One ; 9(5): e97632, 2014.
Article in English | MEDLINE | ID: mdl-24851923

ABSTRACT

BACKGROUND: Despite intensive research, the etiopathogenesis of preeclampsia (PE) remains uncertain. Inflammatory and angiogenic factors are thought to play considerable roles in this disease. The objective of this study was to investigate the association between soluble endoglin (sEng), transforming growth factor beta-1 (TGF-ß1) and tumor necrosis factor alpha soluble receptors (sTNF-Rs) and the clinical manifestations of PE. METHODS: Plasma levels of sEng, TGF-ß1 and sTNF-Rs were determined by ELISA in 23 non-pregnant, 21 normotensive pregnant and 43 PE women. PE women were stratified into subgroups according to the severity [mild (n = 12) and severe (n = 31)] and onset-time of the disease [early (n = 19) and late (n = 24)]. RESULTS: Pregnancy was associated with higher levels of sEng, sTNF-R1 and sTNF-R2 than the non-pregnant state. Moreover, PE women had higher levels of sEng and sTNF-R1 than normotensive pregnant women. No difference was found in TGF-ß1 levels, comparing the three study groups. Late PE had higher levels of sTNF-R1 and sTNF-R2 than early PE. No significant differences were found in sEng and TGF-ß1 comparing early and late PE. sEng levels were higher in severe PE than in mild PE and no difference was found for TGF-ß1, sTNF-R1 and sTNF-R2 levels. There was a positive correlation among sEng, TNF-R1 and sTNF-2 levels. Logistic regression analysis revealed that primiparity and sEng levels are independently associated with the development of PE. Furthermore, sEng levels are independently associated with the disease severity. CONCLUSIONS: These results suggest that pregnancy is a condition associated with higher levels of anti-angiogenic and pro-inflammatory factors than the non-pregnant state and that PE is associated with an imbalance of these factors in the maternal circulation.


Subject(s)
Antigens, CD/blood , Pre-Eclampsia/physiopathology , Protein Serine-Threonine Kinases/blood , Receptors, Cell Surface/blood , Receptors, Transforming Growth Factor beta/blood , Receptors, Tumor Necrosis Factor/blood , Endoglin , Enzyme-Linked Immunosorbent Assay , Female , Humans , Pre-Eclampsia/blood , Pregnancy , Receptor, Transforming Growth Factor-beta Type I
19.
Exp Oncol ; 36(1): 38-43, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24691283

ABSTRACT

UNLABELLED: The elevated anti-GalNAcß IgG level of serum was shown to be associated with the significantly better survival of patients with gastrointestinal cancer. AIM: To characterize the specificity of IgG antibodies to GalNAcß-terminated glycans of long-term gastric cancer survivors. METHODS: Serum antibodies and affinity-isolated antibodies were analysed by the indirect and competitive ELISA using glycan-polyacrylamide (PAA) conjugates as well as by isoelectric focusing and Western blotting. RESULTS: In the serum probes, a partial cross-reactivity of antibodies to GalNAcß, GalNAcß1-3Galß (X2di), GalNAcß1-3GalNAcß (PFdi) and GlcNAcß was observed. The isolated anti-GalNAcß IgGs demonstrated the cross-reactivity to the X2di glycan mainly. The affinity of the X2di-PAA to anti-GalNAcß IgGs was 11-21 times lower than that of the GalNAcß-PAA. Anti-X2di and anti-PFdi IgGs demonstrated monoreactivity to their key glycans-PAA used in isolation. The IC50 values of key glycoconjugates ranged from 1 to 5 · 10(-7) M. No polyreactivity of antibodies to the unrelated antigens (ferritin, casein and DNA) was found. The polyclonal or oligoclonal distribution of IgG bands was established and the monoreactivity of antibodies was not associated with the clonal distribution of bands. CONCLUSION: The cross-reactivity of anti-GalNAcß antibodies to X2di and related glycans deserves attention in the clarification of the role of antibodies in cancer progression and enhancement of the prognostic potential in the combined determination of antibody markers.


Subject(s)
Galactosyltransferases/biosynthesis , Immunoglobulin G/blood , Proteoglycans/blood , Receptors, Transforming Growth Factor beta/blood , Stomach Neoplasms/blood , Adult , Aged , Antibodies/blood , Antibody Specificity/immunology , Female , Galactosyltransferases/immunology , Humans , Male , Middle Aged , Proteoglycans/immunology , Receptors, Transforming Growth Factor beta/immunology , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , Stomach Neoplasms/surgery , Survival Analysis , Survivors
20.
Fertil Steril ; 101(5): 1353-8, 2014 May.
Article in English | MEDLINE | ID: mdl-24613539

ABSTRACT

OBJECTIVE: To evaluate antimüllerian hormone (AMH) and AMH receptor II (AMHRII) mRNA and protein expression in endometrium and in ovarian or deep lesions of women with endometriosis. DESIGN: Prospective study. SETTING: University hospitals in Italy and Brazil. PATIENTS: Patients with endometriosis (n = 55) and healthy women (n = 45). INTERVENTIONS: Specimens of endometrium obtained by hysteroscopy from patients with endometriosis and from healthy control subjects; specimens of ovarian endometriosis (n = 29) or of deep endometriosis (n = 26) were collected by laparoscopy. Serum samples were collected in some endometriotic patients (n = 23) and healthy control subjects (n = 20). MAIN OUTCOME MEASURE(S): AMH and AMHRII mRNA levels were evaluated by quantitative reverse-transcription polymerase chain reaction and protein localization by immunohistochemistry. AMH levels in tissue homogenates and in serum were assessed by ELISA. RESULT(S): Endometrium from women with endometriosis showed higher AMH and AMHRII mRNA levels than control women, with no significant differences between proliferative and secretory phases. Specimens collected from ovarian or deep endometriosis showed the highest AMH and AMHRII mRNA expression. Immunolocalization study confirmed the high AMH and AMHRII protein expression in endometriotic lesions. No difference of serum AMH levels between the groups was found. CONCLUSION(S): The increased AMH and AMHRII mRNA and protein expression in endometrium and in endometriotic lesions suggests a possible involvement of AMH in endometriosis.


Subject(s)
Anti-Mullerian Hormone/biosynthesis , Endometriosis/diagnosis , Endometriosis/metabolism , Gene Expression Regulation , Receptors, Peptide/biosynthesis , Receptors, Transforming Growth Factor beta/biosynthesis , Adult , Anti-Mullerian Hormone/blood , Biomarkers/blood , Biomarkers/metabolism , Endometriosis/surgery , Endometrium/metabolism , Endometrium/pathology , Female , Humans , Prospective Studies , Receptors, Peptide/blood , Receptors, Transforming Growth Factor beta/blood , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...