Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Michelle R. Jones is active.

Publication


Featured researches published by Michelle R. Jones.


Journal of Medical Genetics | 2012

Replication of association of DENND1A and THADA variants with polycystic ovary syndrome in European cohorts

Mark O. Goodarzi; Michelle R. Jones; Xiaohui Li; Angela K. Chua; Obed A. Garcia; Yii-Der I. Chen; Ronald M. Krauss; Jerome I. Rotter; Wendy Ankener; Richard S. Legro; Ricardo Azziz; Jerome F. Strauss; Andrea Dunaif; Margrit Urbanek

Background Polycystic ovary syndrome (PCOS) is a complex endocrine disorder with a strong familial component. PCOS is characterised by hyperandrogenaemia and irregular menses. A recent genome-wide association study (GWAS) of PCOS in a Chinese cohort identified three reproducible PCOS susceptibility loci mapping to 2p16.3 (luteinising hormone/choriogonadotropin receptor; LHCGR), 2p21 (thyroid associated protein; THADA), and 9q33.3 (DENN/MADD domain containing 1A; DENNDIA). The impact of these loci in non-Chinese PCOS cohorts remains to be determined. Methods and results The study tested association with PCOS of seven single nucleotide polymorphisms mapping to the three Chinese PCOS loci in two European derived PCOS cohorts (cohort A = 939 cases and 957 controls; cohort B = 535 cases and 845 controls). Cases fulfilled the National Institute of Child Health & Human Development criteria for PCOS. Variation in DENND1A was strongly associated with PCOS in the study cohort (pcombined cohorts=10−8); multiple variants in THADA were also associated with PCOS, while there was no significant evidence for association of LHCGR variation with PCOS. The present study had >80% power to detect an effect of similar size as was observed by Chen et al for DENND1A and THADA, but reduced power (at <40%) for LHCGR at p=0.0001. The study had sufficient power (57–88%) for LHCGR at p=0.01. Conclusions At least two of the PCOS susceptibility loci identified in the Chinese PCOS GWAS (DENND1A and THADA) are also associated with PCOS in European derived populations, and are therefore likely to be important in the aetiology of PCOS regardless of ethnicity. The analysis of the LHCGR gene was not sufficiently powered to detect modest effects.


PLOS Genetics | 2012

A genome-wide scan of ashkenazi jewish crohn's disease suggests novel susceptibility loci

Eimear E. Kenny; Itsik Pe'er; Amir Karban; Laurie J. Ozelius; Adele A. Mitchell; Sok Meng Ng; Monica Erazo; Harry Ostrer; Clara Abraham; Maria T. Abreu; Gil Atzmon; Nir Barzilai; Steven R. Brant; Susan Bressman; Edward R. Burns; Yehuda Chowers; Lorraine N. Clark; Ariel Darvasi; Dana Doheny; Richard H. Duerr; Rami Eliakim; Nir Giladi; Peter K. Gregersen; Hakon Hakonarson; Michelle R. Jones; Karen Marder; Dermot McGovern; Jennifer G. Mulle; Avi Orr-Urtreger; Deborah D. Proctor

Crohns disease (CD) is a complex disorder resulting from the interaction of intestinal microbiota with the host immune system in genetically susceptible individuals. The largest meta-analysis of genome-wide association to date identified 71 CD–susceptibility loci in individuals of European ancestry. An important epidemiological feature of CD is that it is 2–4 times more prevalent among individuals of Ashkenazi Jewish (AJ) descent compared to non-Jewish Europeans (NJ). To explore genetic variation associated with CD in AJs, we conducted a genome-wide association study (GWAS) by combining raw genotype data across 10 AJ cohorts consisting of 907 cases and 2,345 controls in the discovery stage, followed up by a replication study in 971 cases and 2,124 controls. We confirmed genome-wide significant associations of 9 known CD loci in AJs and replicated 3 additional loci with strong signal (p<5×10−6). Novel signals detected among AJs were mapped to chromosomes 5q21.1 (rs7705924, combined p = 2×10−8; combined odds ratio OR = 1.48), 2p15 (rs6545946, p = 7×10−9; OR = 1.16), 8q21.11 (rs12677663, p = 2×10−8; OR = 1.15), 10q26.3 (rs10734105, p = 3×10−8; OR = 1.27), and 11q12.1 (rs11229030, p = 8×10−9; OR = 1.15), implicating biologically plausible candidate genes, including RPL7, CPAMD8, PRG2, and PRG3. In all, the 16 replicated and newly discovered loci, in addition to the three coding NOD2 variants, accounted for 11.2% of the total genetic variance for CD risk in the AJ population. This study demonstrates the complementary value of genetic studies in the Ashkenazim.


Nature | 2010

CRTC3 links catecholamine signalling to energy balance

Youngsup Song; Judith Y. Altarejos; Mark O. Goodarzi; Hiroshi Inoue; Xiuqing Guo; Rebecca Berdeaux; Jeong Ho Kim; Jason Goode; Motoyuki Igata; José C. Paz; Meghan F. Hogan; Pankaj K. Singh; Naomi Goebel; Lili Vera; Nina Miller; Jinrui Cui; Michelle R. Jones; Yii-Der I. Chen; Kent D. Taylor; Willa A. Hsueh; Jerome I. Rotter; Marc Montminy

The adipose-derived hormone leptin maintains energy balance in part through central nervous system-mediated increases in sympathetic outflow that enhance fat burning. Triggering of β-adrenergic receptors in adipocytes stimulates energy expenditure by cyclic AMP (cAMP)-dependent increases in lipolysis and fatty-acid oxidation. Although the mechanism is unclear, catecholamine signalling is thought to be disrupted in obesity, leading to the development of insulin resistance. Here we show that the cAMP response element binding (CREB) coactivator Crtc3 promotes obesity by attenuating β-adrenergic receptor signalling in adipose tissue. Crtc3 was activated in response to catecholamine signals, when it reduced adenyl cyclase activity by upregulating the expression of Rgs2, a GTPase-activating protein that also inhibits adenyl cyclase activity. As a common human CRTC3 variant with increased transcriptional activity is associated with adiposity in two distinct Mexican-American cohorts, these results suggest that adipocyte CRTC3 may play a role in the development of obesity in humans.


PLOS ONE | 2011

FTO and MC4R gene variants are associated with obesity in polycystic ovary syndrome.

Kathryn G. Ewens; Michelle R. Jones; Wendy Ankener; Douglas R. Stewart; Margrit Urbanek; Andrea Dunaif; Richard S. Legro; Angela Chua; Ricardo Azziz; Richard S. Spielman; Mark O. Goodarzi; Jerome F. Strauss

Polycystic ovary syndrome (PCOS) is the leading cause of anovulatory infertility in women. It is also associated with metabolic disturbances that place women at increased risk for obesity and type 2 diabetes. There is strong evidence for familial clustering of PCOS and a genetic predisposition. However, the gene(s) responsible for the PCOS phenotypes have not been elucidated. This two-phase family-based and case-control genetic study was designed to address the question of whether SNPs identified as susceptibility loci for obesity in genome-wide association studies (GWAS) are also associated with PCOS and elevated BMI. Members of 439 families having at least one offspring with PCOS were genotyped for 15 SNPs previously shown to be associated with obesity. Linkage and association with PCOS was assessed using the transmission/disequilibrium test (TDT). These SNPs were also analyzed in an independent case-control study involving 395 women with PCOS and 176 healthy women with regular menstrual cycles. Only one of these 15 SNPs (rs2815752 in NEGR1) was found to have a nominally significant association with PCOS (χ2 = 6.11, P = 0.013), but this association failed to replicate in the case-control study. While not associated with PCOS itself, five SNPs in FTO and two in MC4R were associated with BMI as assessed with a quantitative-TDT analysis, several of which replicated association with BMI in the case-control cohort. These findings demonstrate that certain SNPs associated with obesity contribute to elevated BMI in PCOS, but do not appear to play a major role in PCOS per se. These findings support the notion that PCOS phenotypes are a consequence of an oligogenic/polygenic mechanism.


The Journal of Clinical Endocrinology and Metabolism | 2015

Further Investigation in Europeans of Susceptibility Variants for Polycystic Ovary Syndrome Discovered in Genome-Wide Association Studies of Chinese Individuals

M. Brower; Michelle R. Jones; Jerome I. Rotter; Ronald M. Krauss; Richard S. Legro; Ricardo Azziz; Mark O. Goodarzi

CONTEXT Two genome-wide association studies (GWAS) of polycystic ovary syndrome (PCOS) have identified 11 susceptibility loci in Chinese individuals. Some of the risk loci identified in Chinese cohorts, mostly from the first GWAS, have been replicated in Europeans. Replication of the loci from the second GWAS in European cohorts is necessary to determine whether the same variants confer risk for PCOS in multiple ethnicities. OBJECTIVE The objective of the study was to determine the effects of the Chinese GWAS loci in European-origin individuals. DESIGN This was a genetic association study. SETTING The study was conducted at a tertiary care academic center. PATIENTS Eight hundred forty-five European subjects with PCOS and 845 controls participated in the study. INTERVENTIONS INTERVENTIONS included blood sampling and genotyping. MAIN OUTCOME MEASURE The association between PCOS and 12 independent single-nucleotide polymorphisms mapping to seven of the Chinese GWAS loci in a European cohort was measured. RESULTS Variants in DENND1A (P = .0002), THADA (P = .035), FSHR (P = .007), and INSR (P = .046) were associated with PCOS in Europeans. The genetic risk score, generated for each subject based on the total number of risk alleles, was associated with the diagnosis of PCOS (P < .0001) and remained associated (P = .02), even after exclusion of the four variants individually associated with PCOS. CONCLUSIONS At least four of the PCOS susceptibility loci identified in the Chinese GWAS are associated with PCOS in Europeans. The overall genetic burden for PCOS, as demonstrated by the risk score, is also associated with the diagnosis of PCOS in Europeans. The PCOS susceptibility loci identified in the Chinese GWAS are thus likely to play an important role in the etiology of PCOS across ethnicities.


Fertility and Sterility | 2011

Replication of association of a novel insulin receptor gene polymorphism with polycystic ovary syndrome

Mark O. Goodarzi; Yvonne V. Louwers; Kent D. Taylor; Michelle R. Jones; Jinrui Cui; Soonil Kwon; Yii-Der I. Chen; Xiuqing Guo; Lisette Stolk; André G. Uitterlinden; Joop S.E. Laven; Ricardo Azziz

OBJECTIVE To evaluate association with polycystic ovary syndrome (PCOS) of 295 variants in 39 genes central to metabolic insulin signaling and glycogen synthase kinase 3β (GSK-3β) regulation, followed by replication efforts. DESIGN Case-control association study, with discovery and replication cohorts. SETTING Subjects were recruited from reproductive endocrinology clinics, and controls were recruited from communities surrounding the University of Alabama at Birmingham and Erasmus Medical Center, Rotterdam. PATIENT(S) A total of 273 cases with PCOS and 173 control subjects in the discovery cohort; and 526 cases and 3,585 control subjects in the replication cohort. All subjects were caucasian. INTERVENTION(S) Phenotypic and genotypic assessment. MAIN OUTCOME MEASURE(S) Detection of 295 single-nucleotide polymorphisms (SNPs), PCOS status. RESULT(S) Several SNPs were associated with PCOS in the discovery cohort. Four insulin receptor (INSR) SNPs and three insulin receptor substrate 2 (IRS2) SNPs associated with PCOS were genotyped in the replication cohort. One INSR SNP (rs2252673) replicated association with PCOS. The minor allele conferred increased odds of PCOS in both cohorts, independent of body mass index. CONCLUSION(S) A pathway-based tagging SNP approach allowed us to identify novel INSR SNPs associated with PCOS, one of which confirmed association in a large replication cohort.


British Journal of Obstetrics and Gynaecology | 2010

Association study of four key folliculogenesis genes in polycystic ovary syndrome.

Kari Sproul; Michelle R. Jones; Ruchi Mathur; Ricardo Azziz; Mark O. Goodarzi

Please cite this paper as: Sproul K, Jones M, Mathur R, Azziz R, Goodarzi M. Association study of four key folliculogenesis genes in polycystic ovary syndrome. BJOG 2010; DOI: 10.1111/j.1471‐0528.2010.02527.x.


Diabetes Care | 2008

First Evidence of Genetic Association Between AKT2 and Polycystic Ovary Syndrome

Mark O. Goodarzi; Michelle R. Jones; Yii-Der Ida Chen; Ricardo Azziz

OBJECTIVE—Insulin resistance has been reported in up to 70% of women with polycystic ovary syndrome (PCOS). Physiologic and genetic data currently implicate post–insulin receptor signaling defects in substrates such as glycogen synthase kinase 3β (GSK3β). The AKT2 gene was chosen as a candidate for PCOS because its product affects glucose metabolism and mitogenic signaling, interacts with GSK3β, and mediates cell survival in the ovary. RESEARCH DESIGN AND METHODS—Subjects were recruited from the reproductive endocrinology clinic at the University of Alabama at Birmingham, and control subjects were recruited from the surrounding community; 287 white women with PCOS and 187 white control subjects were genotyped for four single nucleotide polymorphisms (SNPs) in AKT2. Genotyping took place at Cedars-Sinai Medical Center in Los Angeles. SNPs and haplotypes were tested for association with PCOS risk and phenotypic markers of PCOS. RESULTS—Minor allele carriers of SNPs rs3730051 and rs8100018 had increased odds of PCOS (odds ratio [OR] 2.2, P = 0.004, and 2.4, P = 0.001, respectively). The haplotype T-G-C-T was significantly associated with PCOS (OR 2.0, P = 0.01). Carriers of the risk haplotypes for both AKT2 and GSK3B had a further increased odds of PCOS (OR 3.1, P = 0.005). CONCLUSIONS—These data suggest that polymorphisms in two components of the insulin signaling pathway, AKT2 and GSK3B, are associated with PCOS. The presence of multiple lesions in a single pathway may confer increased risk.


Fertility and Sterility | 2016

Genetic determinants of polycystic ovary syndrome: progress and future directions

Michelle R. Jones; Mark O. Goodarzi

The field of the genetics of polycystic ovary syndrome (PCOS) has relatively recently moved into the era of genome-wide association studies. This has led to the discovery of 16 robust loci for PCOS. Some loci contain genes with clear roles in reproductive (LHCGR, FSHR, and FSHB) and metabolic (INSR and HMGA2) dysfunction in the syndrome. The next challenge facing the field is the identification of causal variants and genes and the role they play in PCOS pathophysiology. The potential for gene discovery to improve diagnosis and treatment of PCOS is promising, though there is much to be done in the field before the current findings can be translated to the clinic.


PLOS Genetics | 2015

Systems Genetics Reveals the Functional Context of PCOS Loci and Identifies Genetic and Molecular Mechanisms of Disease Heterogeneity

Michelle R. Jones; M. Brower; Ning Xu; Jinrui Cui; Emebet Mengesha; Yii-Der I. Chen; Kent D. Taylor; Ricardo Azziz; Mark O. Goodarzi

Genome wide association studies (GWAS) have revealed 11 independent risk loci for polycystic ovary syndrome (PCOS), a common disorder in young women characterized by androgen excess and oligomenorrhea. To put these risk loci and the single nucleotide polymorphisms (SNPs) therein into functional context, we measured DNA methylation and gene expression in subcutaneous adipose tissue biopsies to identify PCOS-specific alterations. Two genes from the LHCGR region, STON1-GTF2A1L and LHCGR, were overexpressed in PCOS. In analysis stratified by obesity, LHCGR was overexpressed only in non-obese PCOS women. Although not differentially expressed in the entire PCOS group, INSR was underexpressed in obese PCOS subjects only. Alterations in gene expression in the LHCGR, RAB5B and INSR regions suggest that SNPs in these loci may be functional and could affect gene expression directly or indirectly via epigenetic alterations. We identified reduced methylation in the LHCGR locus and increased methylation in the INSR locus, changes that are concordant with the altered gene expression profiles. Complex patterns of meQTL and eQTL were identified in these loci, suggesting that local genetic variation plays an important role in gene regulation. We propose that non-obese PCOS women possess significant alterations in LH receptor expression, which drives excess androgen secretion from the ovary. Alternatively, obese women with PCOS possess alterations in insulin receptor expression, with underexpression in metabolic tissues and overexpression in the ovary, resulting in peripheral insulin resistance and excess ovarian androgen production. These studies provide a genetic and molecular basis for the reported clinical heterogeneity of PCOS.

Collaboration


Dive into the Michelle R. Jones's collaboration.

Top Co-Authors

Avatar

Mark O. Goodarzi

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Ricardo Azziz

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

Jerome I. Rotter

Los Angeles Biomedical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Kent D. Taylor

Los Angeles Biomedical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Yii-Der I. Chen

Los Angeles Biomedical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Xiuqing Guo

Los Angeles Biomedical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Richard S. Legro

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Jinrui Cui

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ronald M. Krauss

Children's Hospital Oakland Research Institute

View shared research outputs
Top Co-Authors

Avatar

Talin Haritunians

Cedars-Sinai Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge