Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Cell Rep Med ; 5(5): 101558, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38733986

ABSTRACT

The investigation of the mechanisms behind p53 mutations in acute myeloid leukemia (AML) has been limited by the lack of suitable mouse models, which historically have resulted in lymphoma rather than leukemia. This study introduces two new AML mouse models. One model induces mutant p53 and Mdm2 haploinsufficiency in early development, showing the role of Mdm2 in myeloid-biased hematopoiesis and AML predisposition, independent of p53. The second model mimics clonal hematopoiesis by inducing mutant p53 in adult hematopoietic stem cells, demonstrating that the timing of p53 mutation determines AML vs. lymphoma development. In this context, age-related changes in hematopoietic stem cells (HSCs) collaborate with mutant p53 to predispose toward myeloid transformation rather than lymphoma development. Our study unveils new insights into the cooperative impact of HSC age, Trp53 mutations, and Mdm2 haploinsufficiency on clonal hematopoiesis and the development of myeloid malignancies.


Subject(s)
Clonal Hematopoiesis , Hematopoietic Stem Cells , Leukemia, Myeloid, Acute , Mutation , Proto-Oncogene Proteins c-mdm2 , Tumor Suppressor Protein p53 , Animals , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Clonal Hematopoiesis/genetics , Mice , Mutation/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Mice, Inbred C57BL , Haploinsufficiency/genetics , Disease Models, Animal , Hematopoiesis/genetics
2.
Blood Cancer J ; 13(1): 101, 2023 06 29.
Article in English | MEDLINE | ID: mdl-37386016

ABSTRACT

In TP53 wild-type acute myeloid leukemia (AML), inhibition of MDM2 can enhance p53 protein expression and potentiate leukemic cell apoptosis. MDM2 inhibitor (MDM2i) monotherapy in AML has shown modest responses in clinical trials but combining options of MDM2i with other potent AML-directed agents like cytarabine and venetoclax could improve its efficacy. We conducted a phase I clinical trial (NCT03634228) to study the safety and efficacy of milademetan (an MDM2i) with low-dose cytarabine (LDAC)±venetoclax in adult patients with relapsed refractory (R/R) or newly diagnosed (ND; unfit) TP53 wild-type AML and performed comprehensive CyTOF analyses to interrogate multiple signaling pathways, the p53-MDM2 axis and the interplay between pro/anti-apoptotic molecules to identify factors that determine response and resistance to therapy. Sixteen patients (14 R/R, 2 N/D treated secondary AML) at a median age of 70 years (range, 23-80 years) were treated in this trial. Two patients (13%) achieved an overall response (complete remission with incomplete hematological recovery). Median cycles on trial were 1 (range 1-7) and at a median follow-up of 11 months, no patients remained on active therapy. Gastrointestinal toxicity was significant and dose-limiting (50% of patients ≥ grade 3). Single-cell proteomic analysis of the leukemia compartment revealed therapy-induced proteomic alterations and potential mechanisms of adaptive response to the MDM2i combination. The response was associated with immune cell abundance and induced the proteomic profiles of leukemia cells to disrupt survival pathways and significantly reduced MCL1 and YTHDF2 to potentiate leukemic cell death. The combination of milademetan, LDAC±venetoclax led to only modest responses with recognizable gastrointestinal toxicity. Treatment-induced reduction of MCL1 and YTHDF2 in an immune-rich milieu correlate with treatment response.


Subject(s)
Cytarabine , Leukemia, Myeloid, Acute , Adult , Humans , Young Adult , Middle Aged , Aged , Aged, 80 and over , Tumor Suppressor Protein p53 , Myeloid Cell Leukemia Sequence 1 Protein , Proteomics , Leukemia, Myeloid, Acute/drug therapy
3.
Cell Death Dis ; 14(6): 371, 2023 06 23.
Article in English | MEDLINE | ID: mdl-37353528

ABSTRACT

Mesenchymal stromal cells (MSCs) are a key component of the bone marrow (BM) niche, providing essential support required for the maintenance of hematopoietic stem cells. To advance our understanding of physiological functions of p53 and Mdm2 in BM-MSCs, we developed traceable conditional mouse models targeting Mdm2 and/or Trp53 in vivo. We demonstrate that Mdm2 is essential for the emergence, maintenance, and hematopoietic support of BM-MSCs. Mdm2 haploinsufficiency in BM-MSCs resulted in genotoxic stress-associated thrombocytopenia, suggesting a functional role for Mdm2 in hematopoiesis. In a syngeneic mouse model of acute myeloid leukemia (AML), Trp53 deletion in BM-MSCs improved survival, and protected BM against hematopoietic toxicity from a murine Mdm2i, DS-5272. The transcriptional changes were associated with dysregulation of glycolysis, gluconeogenesis, and Hif-1α in BM-MSCs. Our results reveal a physiologic function of Mdm2 in BM-MSC, identify a previously unknown role of p53 pathway in BM-MSC-mediated support in AML and expand our understanding of the mechanism of hematopoietic toxicity of MDM2is.


Subject(s)
Mesenchymal Stem Cells , Proto-Oncogene Proteins c-mdm2 , Thrombocytopenia , Tumor Suppressor Protein p53 , Animals , Mice , Bone Marrow , Bone Marrow Cells , Disease Models, Animal , DNA Damage , Tumor Suppressor Protein p53/genetics , Proto-Oncogene Proteins c-mdm2/genetics
4.
Cancer Res ; 83(14): 2297-2311, 2023 07 14.
Article in English | MEDLINE | ID: mdl-37205631

ABSTRACT

Missense mutations in the DNA binding domain of p53 are characterized as structural or contact mutations based on their effect on the conformation of the protein. These mutations show gain-of-function (GOF) activities, such as promoting increased metastatic incidence compared with p53 loss, often mediated by the interaction of mutant p53 with a set of transcription factors. These interactions are largely context specific. To understand the mechanisms by which p53 DNA binding domain mutations drive osteosarcoma progression, we created mouse models, in which either the p53 structural mutant p53R172H or the contact mutant p53R245W are expressed specifically in osteoblasts, yielding osteosarcoma tumor development. Survival significantly decreased and metastatic incidence increased in mice expressing p53 mutants compared with p53-null mice, suggesting GOF. RNA sequencing of primary osteosarcomas revealed vastly different gene expression profiles between tumors expressing the missense mutants and p53-null tumors. Further, p53R172H and p53R245W each regulated unique transcriptomes and pathways through interactions with a distinct repertoire of transcription factors. Validation assays showed that p53R245W, but not p53R172H, interacts with KLF15 to drive migration and invasion in osteosarcoma cell lines and promotes metastasis in allogeneic transplantation models. In addition, analyses of p53R248W chromatin immunoprecipitation peaks showed enrichment of KLF15 motifs in human osteoblasts. Taken together, these data identify unique mechanisms of action of the structural and contact mutants of p53. SIGNIFICANCE: The p53 DNA binding domain contact mutant p53R245W, but not the structural mutant p53R172H, interacts with KLF15 to drive metastasis in somatic osteosarcoma, providing a potential vulnerability in tumors expressing p53R245W mutation.


Subject(s)
Bone Neoplasms , Osteosarcoma , Mice , Humans , Animals , Tumor Suppressor Protein p53/genetics , Osteosarcoma/pathology , Mutation , Mice, Knockout , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Transcription Factors/metabolism , DNA , Cell Line, Tumor
5.
Res Sq ; 2023 Mar 02.
Article in English | MEDLINE | ID: mdl-36909480

ABSTRACT

Mesenchymal stromal cells (MSCs) are a key component of the bone marrow (BM) niche, providing essential support required for maintenance of hematopoietic stem cells. To advance our understanding of physiological functions of p53 and Mdm2 in BM-MSCs, we developed traceable conditional mouse models targeting Mdm2 and/or Trp53 in vivo . We demonstrate that Mdm2 is essential for the emergence, maintenance and hematopoietic support of BM-MSCs. Mdm2 haploinsufficiency in BM-MSCs resulted in genotoxic stress-associated thrombocytopenia, suggesting a functional role for Mdm2 in hematopoiesis. In a syngeneic mouse model of acute myeloid leukemia (AML), Trp53 deletion in BM-MSCs improved survival, and protected BM against hematopoietic toxicity from a murine Mdm2i, DS-5272. The transcriptional changes were associated with dysregulation of glycolysis, gluconeogenesis, and Hif-1α in BM-MSCs. Our results reveal a physiologic function of Mdm2 in BM-MSC, identify a previously unknown role of p53 pathway in BM-MSC-mediated support in AML and expand our understanding of the mechanism of hematopoietic toxicity of MDM2is.

6.
Mol Cancer Res ; 21(6): 548-563, 2023 06 01.
Article in English | MEDLINE | ID: mdl-36787422

ABSTRACT

Despite effective new therapies, adaptive resistance remains the main obstacle in acute myelogenous leukemia (AML) therapy. Autophagy induction is a key mechanism for adaptive resistance. Leukemic blasts at diagnosis express higher levels of the apical autophagy kinase ULK1 compared with normal hematopoietic cells. Exposure to chemotherapy and targeted agents upregulate ULK1, hence we hypothesize that developing ULK1 inhibitors may present the unique opportunity for clinical translation of autophagy inhibition. Accordingly, we demonstrate that ULK1 inhibition, by genetic and pharmacologic means, suppresses treatment-induced autophagy, overcomes adaptive drug-resistance, and synergizes with chemotherapy and emerging antileukemia agents like venetoclax (ABT-199). The study next aims at exploring the underlying mechanisms. Mechanistically, ULK1 inhibition downregulates MCL1 antiapoptotic gene, impairs mitochondrial function and downregulates components of the CD44-xCT system, resulting in impaired reactive oxygen species (ROS) mitigation, DNA damage, and apoptosis. For further validation, several mouse models of AML were generated. In these mouse models, ULK1 deficiency impaired leukemic cell homing and engraftment, delayed disease progression, and improved survival. Therefore, in the study, we validated our hypothesis and identified ULK1 as an important mediator of adaptive resistance to therapy and an ideal candidate for combination therapy in AML. Therefore, we propose ULK1 inhibition as a therapeutically relevant treatment option to overcome adaptive drug-resistance in AML. IMPLICATIONS: ULK1 drives a cell-intrinsic adaptive resistance in AML and targeting ULK1-mediated autophagy can synergize with existing and emerging AML therapies to overcome drug-resistance and induce apoptosis.


Subject(s)
Antineoplastic Agents , Leukemia, Myeloid, Acute , Animals , Mice , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Antineoplastic Agents/pharmacology , Autophagy , Drug Resistance, Neoplasm , Apoptosis
7.
Cancer ; 127(20): 3772-3781, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34255353

ABSTRACT

BACKGROUND: TP53 mutation (TP53mut ) confers an adverse prognosis in acute myeloid leukemia (AML). Venetoclax with hypomethylating agents is a current standard for older patients; however, recent reports suggest that TP53mut confers resistance to venetoclax. The authors investigated the outcomes of patients with TP53mut AML who were treated with a 10-day decitabine and venetoclax (DEC10-VEN) (ClinicalTrials.gov identifier NCT03404193). METHODS: Patients with newly diagnosed AML received decitabine 20 mg/m2 for 10 days every 4 to 6 weeks for induction, followed by decitabine for 5 days after response. The venetoclax dose was 400 mg daily. TP53mut was identified in bone marrow samples using next-generation sequencing, with sensitivity of 5%. Outcomes were analyzed according to European LeukemiaNet 2017 guidelines. RESULTS: Among 118 patients (median age, 72 years; age range, 49-89 years), 63 (53%) had secondary AML, 39 (33%) had AML with complex karyotype, and 35 (30%) had TP53mut AML. The median TP53 variant allele frequency was 32% (interquartile range, 16%-65%), 8 patients (23%) had only a single TP53 mutation, 15 (43%) had multiple mutations, and 12 (34%) had mutation and deletion. Outcomes were significantly worse in patients who had TP53mut AML compared with those who had wild-type TP53 AML, with an overall response rate of 66% vs 89% (P = .002), a complete response/complete response with incomplete hematologic recovery rate of 57% vs 77% (P = .029), and a 60-day mortality of 26% vs 4% (P < .001), respectively. Patients with TP53mut versus wild-type TP53 had shorter overall survival at 5.2 versus 19.4 months, respectively (hazard ratio, 4.67; 95% CI, 2.44-8.93; P < .0001), and shorter relapse-free survival at 3.4 versus 18.9 months (hazard ratio, 4.80; 95% CI, 1.97-11.69; P < .0001), respectively. Outcomes with DEC10-VEN in patients with TP53mut AML were comparable to historical results with 10-day decitabine alone. CONCLUSIONS: Patients with TP53mut AML have lower response rates and shorter survival with DEC10-VEN.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Leukemia, Myeloid, Acute , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Decitabine/adverse effects , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Middle Aged , Sulfonamides , Tumor Suppressor Protein p53/genetics
8.
BMC Cancer ; 20(1): 3, 2020 Jan 02.
Article in English | MEDLINE | ID: mdl-31898537

ABSTRACT

BACKGROUND: Ewing sarcoma is a malignancy of primitive cells, possibly of mesenchymal origin. It is probable that genetic perturbations other than EWS-FLI1 cooperate with it to produce the tumor. Sequencing studies identified STAG2 mutations in approximately 15% of cases in humans. In the present study, we hypothesize that loss of Stag2 cooperates with EWS-FLI1 in generating sarcomas derived from murine mesenchymal stem cells (MSCs). METHODS: Mice bearing an inducible EWS-FLI1 transgene were crossed to p53-/- mice in pure C57/Bl6 background. MSCs were derived from the bone marrow of the mice. EWS-FLI1 induction and Stag2 knockdown were achieved in vitro by adenovirus-Cre and shRNA-bearing pGIPZ lentiviral infection, respectively. The cells were then treated with ionizing radiation to 10 Gy. Anchorage independent growth in vitro was assessed by soft agar assays. Cellular migration and invasion were evaluated by transwell assays. Cells were injected with Matrigel intramuscularly into C57/Bl6 mice to test for tumor formation. RESULTS: Primary murine MSCs with the genotype EWS-FLI1 p53-/- were resistant to transformation and did not form tumors in syngeneic mice without irradiation. Stag2 inhibition increased the efficiency and speed of sarcoma formation significantly in irradiated EWS-FLI1 p53-/- MSCs. The efficiency of tumor formation was 91% for cells in mice injected with Stag2-repressed cells and 22% for mice receiving cells without Stag2 inhibition (p < .001). Stag2 knockdown reduced survival of mice in Kaplan-Meier analysis (p < .001). It also increased MSC migration and invasion in vitro but did not affect proliferation rate or aneuploidy. CONCLUSION: Loss of Stag2 has a synergistic effect with EWS-FLI1 in the production of sarcomas from murine MSCs, but the mechanism may not relate to increased proliferation or chromosomal instability. Primary murine MSCs are resistant to transformation, and the combination of p53 null mutation, EWS-FLI1, and Stag2 inhibition does not confer immediate conversion of MSCs to sarcomas. Irradiation is necessary in this model, suggesting that perturbations of other genes beside Stag2 and p53 are likely to be essential in the development of EWS-FLI1-driven sarcomas from MSCs.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Mesenchymal Stem Cells/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Protein c-fli-1/genetics , Proto-Oncogene Protein c-fli-1/metabolism , RNA-Binding Protein EWS/genetics , RNA-Binding Protein EWS/metabolism , Animals , Cell Cycle/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Chromosome Aberrations , Disease Models, Animal , Gene Expression , Genes, p53 , Mice , Mice, Knockout , Mice, Transgenic , RNA Interference , Sarcoma, Ewing/etiology , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/pathology
9.
Genes Dev ; 31(18): 1847-1857, 2017 09 15.
Article in English | MEDLINE | ID: mdl-29021240

ABSTRACT

TP53 is the most frequently mutated gene in human cancer. Many mutant p53 proteins exert oncogenic gain-of-function (GOF) properties that contribute to metastasis, but the mechanisms mediating these functions remain poorly defined in vivo. To elucidate how mutant p53 GOF drives metastasis, we developed a traceable somatic osteosarcoma mouse model that is initiated with either a single p53 mutation (p53R172H) or p53 loss in osteoblasts. Our study confirmed that p53 mutant mice developed osteosarcomas with increased metastasis as compared with p53-null mice. Comprehensive transcriptome RNA sequencing (RNA-seq) analysis of 16 tumors identified a cluster of small nucleolar RNAs (snoRNAs) that are highly up-regulated in p53 mutant tumors. Regulatory element analysis of these deregulated snoRNA genes identified strong enrichment of a common Ets2 transcription factor-binding site. Homozygous deletion of Ets2 in p53 mutant mice resulted in strong down-regulation of snoRNAs and reversed the prometastatic phenotype of mutant p53 but had no effect on osteosarcoma development, which remained 100% penetrant. In summary, our studies identify Ets2 inhibition as a potential therapeutic vulnerability in p53 mutant osteosarcomas.


Subject(s)
Bone Neoplasms/genetics , Bone Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Osteosarcoma/genetics , Osteosarcoma/secondary , Proto-Oncogene Protein c-ets-2/genetics , RNA, Small Nucleolar/genetics , Tumor Suppressor Protein p53/genetics , Animals , Down-Regulation , Gene Expression Profiling , Humans , Liver Neoplasms/genetics , Liver Neoplasms/secondary , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Mice , Mice, Knockout , Mutation , Neoplasm Metastasis , Oligonucleotide Array Sequence Analysis , Osteoblasts/metabolism , Osteoblasts/pathology , Up-Regulation
10.
Stem Cell Reports ; 7(2): 139-48, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27396937

ABSTRACT

Wiskott-Aldrich syndrome (WAS) is an X-linked primary immunodeficiency disease caused by mutations in the gene encoding the WAS protein (WASp). Here, induced pluripotent stem cells (iPSCs) were derived from a WAS patient (WAS-iPSC) and the endogenous chromosomal WAS locus was targeted with a wtWAS-2A-eGFP transgene using zinc finger nucleases (ZFNs) to generate corrected WAS-iPSC (cWAS-iPSC). WASp and GFP were first expressed in the earliest CD34(+)CD43(+)CD45(-) hematopoietic precursor cells and later in all hematopoietic lineages examined. Whereas differentiation to non-lymphoid lineages was readily obtained from WAS-iPSCs, in vitro T lymphopoiesis from WAS-iPSC was deficient with few CD4(+)CD8(+) double-positive and mature CD3(+) T cells obtained. T cell differentiation was restored for cWAS-iPSCs. Similarly, defects in natural killer cell differentiation and function were restored on targeted correction of the WAS locus. These results demonstrate that the defects exhibited by WAS-iPSC-derived lymphoid cells were fully corrected and suggests the potential therapeutic use of gene-corrected WAS-iPSCs.


Subject(s)
Genetic Therapy , Induced Pluripotent Stem Cells/pathology , Lymphopoiesis , Wiskott-Aldrich Syndrome/pathology , Wiskott-Aldrich Syndrome/therapy , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Killer Cells, Natural/metabolism , T-Lymphocytes/immunology , Wiskott-Aldrich Syndrome Protein/genetics
11.
J Biol Chem ; 286(43): 37732-40, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-21908606

ABSTRACT

The Zic transcription factors play critical roles during embryonic development. Mutations in the ZIC2 gene are associated with human holoprosencephaly, but the etiology is still unclear. Here, we report a novel function for ZIC2 as a regulator of ß-catenin·TCF4-mediated transcription. We show that ZIC2 can bind directly to the DNA-binding high mobility group box of TCF4 via its zinc finger domain and inhibit the transcriptional activity of the ß-catenin·TCF4 complex. However, the binding of TCF4 to DNA was not affected by ZIC2. Zic2 RNA injection completely inhibited ß-catenin-induced axis duplication in Xenopus embryos and strongly blocked the ability of ß-catenin to induce expression of known Wnt targets in animal caps. Moreover, Zic2 knockdown in transgenic Xenopus Wnt reporter embryos led to ectopic Wnt signaling activity mainly at the midbrain-hindbrain boundary. Together, our results demonstrate a previously unknown role for ZIC2 as a transcriptional regulator of the ß-catenin·TCF4 complex.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Nuclear Proteins/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Transcription, Genetic/physiology , Wnt Proteins/metabolism , beta Catenin/metabolism , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/microbiology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Embryo, Nonmammalian/metabolism , HEK293 Cells , Humans , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Nuclear Proteins/genetics , Transcription Factor 4 , Transcription Factors/genetics , Wnt Proteins/genetics , Xenopus laevis , beta Catenin/genetics
12.
FEBS Lett ; 581(26): 5122-6, 2007 Oct 30.
Article in English | MEDLINE | ID: mdl-17936758

ABSTRACT

RNA and protein analysis revealed the consistent upregulation of the neural transcription factors ZIC1 and ZIC4 in desmoid tumors and other fibroproliferative disorders. The 5' flanking region of the ZIC1 promoter was unmethylated in desmoid tumor fibroblasts, while a hypermethylated ZIC1 promoter was found in human and mouse cell lines not expressing the gene. In addition, expressing cells showed a H3K4me2 at the ZIC1 promoter, whereas non-expressing cells showed higher levels of H3K9me2 in the same region. To our knowledge, this is the first report describing ZIC1 expression in mesenchymal proliferations and a role for DNA methylation in the control of ZIC1 expression.


Subject(s)
DNA Methylation , Fibromatosis, Aggressive/genetics , Gene Expression Regulation , Histones/metabolism , Transcription Factors/genetics , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Base Sequence , Cell Proliferation , DNA Modification Methylases/antagonists & inhibitors , Decitabine , Fibromatosis, Aggressive/pathology , Humans , Mesoderm/chemistry , Mesoderm/metabolism , Mesoderm/pathology , Methylation , Mice , Molecular Sequence Data , NIH 3T3 Cells , Promoter Regions, Genetic , Transcription Factors/analysis
13.
Eur J Cardiovasc Prev Rehabil ; 13(2): 229-35, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16575277

ABSTRACT

BACKGROUND: A parental history of cardiovascular disease has a strong relationship with risk factor clusters in the offspring. This study was performed to identify major cardiovascular risk factors in middle school-aged children and their parents in both high and low-risk families. DESIGN: A school-based, cross-sectional study. METHODS: The middle schools of the 6th district of Tehran were divided randomly into two groups. A total of 169 high-risk children with their families were recruited from the first group and 105 low-risk children with their families were recruited from the second group of schools. Anthropometric and metabolic measurements were performed. RESULTS: The means of the waist circumference and waist-to-hip ratio were significantly higher in high-risk fathers. The means of total and low-density lipoprotein (LDL) cholesterol were significantly higher in both parents and children of the high-risk group. The means of the fasting plasma glucose were significantly higher in fathers and offspring of high-risk families. More fathers in high-risk families were smokers. The prevalence of increased total cholesterol, LDL-cholesterol and hyperglycemia (> or = 100 mg/dl) were higher in high-risk parents and children. The prevalence of increased body mass index (> or = 25 kg/m for parents and 85th percentile for children) was higher in fathers and children of high-risk families. CONCLUSIONS: Cardiovascular risk factors are more prevalent and clustered in high-risk families. The screening of high-risk families is essential to prevent the progression of atherosclerosis from childhood and reduce the burden of cardiovascular disease in adulthood.


Subject(s)
Cardiovascular Diseases/epidemiology , Adolescent , Adult , Body Size , Cardiovascular Diseases/prevention & control , Child , Cross-Sectional Studies , Dietary Fats , Female , Humans , Iran/epidemiology , Lipids/blood , Male , Middle Aged , Nuclear Family , Risk Factors
14.
BMC Public Health ; 6: 29, 2006 Feb 13.
Article in English | MEDLINE | ID: mdl-16472406

ABSTRACT

BACKGROUND: Elevated plasma total homocysteine is an independent risk factor for cardiovascular disease and a sensitive marker of the inadequate vitamin B12 and folate insufficiency. Folate and vitamin B12 have a protective effect on cardiovascular disease. This population based study was conducted to evaluate the plasma total homocysteine, folate, and vitamin B12 in healthy Iranian individuals. METHODS: This study was a part of the Cardiovascular Risk Factors Survey in the Population Lab Region of Tehran University has been designed and conducted based on the methodology of MONICA/WHO Project. A total of 1214 people aged 25-64 years, were recruited and assessed regarding demographic characteristics, homocysteine, folate, and vitamin B12 levels with interview, questionnaires, examination and blood sampling. Blood samples were gathered and analyzed according to standard methods. RESULTS: The variables were assessed in 1214 participants including 428 men (35.3%) and 786 women (64.7%). Age-adjusted prevalence of hyperhomocysteinemia (Hcy > or = 15 micromol/L) was 73.1% in men and 41.07% in women (P < 0.0001). Geometric mean of plasma homocysteine was 19.02 +/- 1.46 micromol/l in men and 14.05 +/- 1.45 micromol/l in women (P < 0.004) which increased by ageing. Age-adjusted prevalence of low serum folate level was 98.67% in men and 97.92% in women. Age-adjusted prevalence of low serum vitamin B12 level was 26.32% in men and 27.2% in women. Correlation coefficients (Pearson's r) between log tHcy and serum folate, and vitamin B12 indicated an inverse correlation (r = -0.27, r = -0.19, P < 0.0001, respectively). CONCLUSION: These results revealed that the prevalence of hyperhomocysteinemia, low folate and vitamin B12 levels are considerably higher than other communities. Implementation of preventive interventions such as food fortification with folic acid is necessary.


Subject(s)
Folic Acid Deficiency/epidemiology , Folic Acid/blood , Homocysteine/blood , Hyperhomocysteinemia/epidemiology , Nutrition Surveys , Vitamin B 12 Deficiency/epidemiology , Vitamin B 12/blood , Adult , Age Factors , Cardiovascular Diseases/etiology , Cardiovascular Diseases/prevention & control , Female , Humans , Hyperhomocysteinemia/complications , Incidence , Iran/epidemiology , Male , Middle Aged , Prevalence , Sex Factors
15.
Metab Syndr Relat Disord ; 4(3): 172-8, 2006.
Article in English | MEDLINE | ID: mdl-18370735

ABSTRACT

Insulin resistance syndrome is a cluster of metabolic abnormalities that is accompanied with an increased risk of diabetes and cardiovascular diseases. This has become an important problem in urban children due to their increasing levels of obesity. A total of 535 obese 7-11- year-old students of all the primary schools of the 6th district of Tehran were screened according to their waist circumference and then confirmed according to the International Obesity Task Force (IOTF) criteria. Waist circumference, fasting serum triglycerides, high-density lipoprotein (HDL) cholesterol, blood pressure, fasting plasma glucose, and insulin levels were measured. Response rate of the study was 96.3%. The crude prevalence rate of metabolic syndrome in these children was 20.6%. There was no significant difference between genders. Only 8.2% of the studied children were without any of the criteria of the metabolic syndrome. The most common metabolic abnormality was hypertriglyceridemia, and the less common one was low HDL levels. Fasting blood sugar, triglyceride, HDL, blood pressure, and waist circumference were all related to the metabolic syndrome with odds ratio of 9.6, 18.71, 6.12, 17.64, and 13.68, respectively. Moreover, insulin levels were significantly higher in these children (12.25 +/- 5.25 vs. 10.75 +/- 4.25 AmicroIU/mL, p = 0.019). This difference was significant in girls with and without metabolic syndrome, but such a difference was not detected in boys. The prevalence of metabolic syndrome is high in Iranian obese children. Early intervention in this population who will become our future obese adults is needed, not only to increase their life quality, but also to decrease the future burden of diabetes and cardiovascular diseases on the society.

16.
Metab Syndr Relat Disord ; 4(1): 28-34, 2006.
Article in English | MEDLINE | ID: mdl-18370767

ABSTRACT

BACKGROUND: Metabolic syndrome includes obesity, hypertriglyceridemia, glucose intolerance, hypertension, and lipid profile abnormalities. The risk of cardiovascular diseases with this syndrome is higher than with each of its components alone. Currently, cardiovascular disorders have a great burden and a high mortality rate in Iran. This study was performed to determine prevalence of the metabolic syndrome and its underlying components in an urban population of Tehran. METHODS: The 25-64-year-old individuals in the Population Laboratory of Tehran University of Medical Sciences were studied through a single-stage cluster sampling; 1573 participants were studied. The response rate was 94.08%. The study was designed according to the World Health Organization (WHO) MONICA (Multinational Monitoring of Trends and Determinants in Cardiovascular Disease) project using the Adult Treatment Panel III (ATPIII) criteria. The parameters used for the risk analysis were waist circumference, fasting serum triglycerides, high density lipoprotein (HDL)-cholesterol, blood pressure, and fasting plasma glucose. RESULTS: The crude prevalence rate of the metabolic syndrome was 29.9% (age-adjusted: 27.5%). The prevalence was significantly higher in women than in men (35.9% vs. 20.3%). The prevalence increased with age in both genders. In the whole population, 88% met at least one of the metabolic syndrome criteria. The most prevalent metabolic abnormalities were hypertriglyceridemia and hypertension that were present in more than half of the population. Also, means of HDL was significantly higher in metabolic syndrome positive group. By further adjustment for age and sex in multivariate analysis, the difference disappeared and even HDL-C level was slightly lower in people with metabolic syndrome. CONCLUSIONS: RESULTS demonstrate high metabolic syndrome rate among target population especially in women. In view of correlation between metabolic syndrome and cardiovascular disease, it must be the priority for interventional preventive measures.

SELECTION OF CITATIONS
SEARCH DETAIL
...