Network


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

Hotspot


Dive into the research topics where Jay A. Tischfield is active.

Publication


Featured researches published by Jay A. Tischfield.


The EMBO Journal | 2016

SIRT7 promotes genome integrity and modulates non-homologous end joining DNA repair.

Berta N. Vazquez; Joshua K. Thackray; Nicolas G. Simonet; Noriko Kane-Goldsmith; Paloma Martínez-Redondo; Trang Nguyen; Samuel F. Bunting; Alejandro Vaquero; Jay A. Tischfield; Lourdes Serrano

Sirtuins, a family of protein deacetylases, promote cellular homeostasis by mediating communication between cells and environment. The enzymatic activity of the mammalian sirtuin SIRT7 targets acetylated lysine in the N‐terminal tail of histone H3 (H3K18Ac), thus modulating chromatin structure and transcriptional competency. SIRT7 deletion is associated with reduced lifespan in mice through unknown mechanisms. Here, we show that SirT7‐knockout mice suffer from partial embryonic lethality and a progeroid‐like phenotype. Consistently, SIRT7‐deficient cells display increased replication stress and impaired DNA repair. SIRT7 is recruited in a PARP1‐dependent manner to sites of DNA damage, where it modulates H3K18Ac levels. H3K18Ac in turn affects recruitment of the damage response factor 53BP1 to DNA double‐strand breaks (DSBs), thereby influencing the efficiency of non‐homologous end joining (NHEJ). These results reveal a direct role for SIRT7 in DSB repair and establish a functional link between SIRT7‐mediated H3K18 deacetylation and the maintenance of genome integrity.


JAMA Psychiatry | 2017

Polygenic Scores for Major Depressive Disorder and Risk of Alcohol Dependence

Allan M. Andersen; Robert H. Pietrzak; Henry R. Kranzler; Li Ma; Hang Zhou; Xiaoming Liu; John Kramer; Samuel Kuperman; Howard J. Edenberg; John I. Nurnberger; John P. Rice; Jay A. Tischfield; Alison Goate; Tatiana Foroud; Jacquelyn L. Meyers; Bernice Porjesz; Danielle M. Dick; Victor Hesselbrock; Eric Boerwinkle; Steven M. Southwick; John H. Krystal; Myrna M. Weissman; Douglas F. Levinson; James B. Potash; Joel Gelernter; Shizhong Han

Importance Major depressive disorder (MDD) and alcohol dependence (AD) are heritable disorders with significant public health burdens, and they are frequently comorbid. Common genetic factors that influence the co-occurrence of MDD and AD have been sought in family, twin, and adoption studies, and results to date have been promising but inconclusive. Objective To examine whether AD and MDD overlap genetically, using a polygenic score approach. Design, Settings, and Participants Association analyses were conducted between MDD polygenic risk score (PRS) and AD case-control status in European ancestry samples from 4 independent genome-wide association study (GWAS) data sets: the Collaborative Study on the Genetics of Alcoholism (COGA); the Study of Addiction, Genetics, and Environment (SAGE); the Yale-Penn genetic study of substance dependence; and the National Health and Resilience in Veterans Study (NHRVS). Results from a meta-analysis of MDD (9240 patients with MDD and 9519 controls) from the Psychiatric Genomics Consortium were applied to calculate PRS at thresholds from Pu2009<u2009.05 to Pu2009⩽u2009.99 in each AD GWAS data set. Main Outcomes and Measures Association between MDD PRS and AD. Results Participants analyzed included 788 cases (548 [69.5%] men; mean [SD] age, 38.2 [10.8] years) and 522 controls (151 [28.9.%] men; age [SD], 43.9 [11.6] years) from COGA; 631 cases (333 [52.8%] men; age [SD], 35.0 [7.7] years) and 756 controls (260 [34.4%] male; age [SD] 36.1 [7.7] years) from SAGE; 2135 cases (1375 [64.4%] men; age [SD], 39.4 [11.5] years) and 350 controls (126 [36.0%] men; age [SD], 43.5 [13.9] years) from Yale-Penn; and 317 cases (295 [93.1%] men; age [SD], 59.1 [13.1] years) and 1719 controls (1545 [89.9%] men; age [SD], 64.5 [13.3] years) from NHRVS. Higher MDD PRS was associated with a significantly increased risk of AD in all samples (COGA: best Pu2009=u20091.7u2009×u200910−6, R2u2009=u20090.026; SAGE: best Pu2009=u2009.001, R2u2009=u20090.01; Yale-Penn: best Pu2009=u2009.035, R2u2009=u20090.0018; and NHRVS: best Pu2009=u2009.004, R2u2009=u20090.0074), with stronger evidence for association after meta-analysis of the 4 samples (best Pu2009=u20093.3u2009×u200910−9). In analyses adjusted for MDD status in 3 AD GWAS data sets, similar patterns of association were observed (COGA: best Pu2009=u20097.6u2009×u200910−6, R2u2009=u20090.023; Yale-Penn: best Pu2009=u2009.08, R2u2009=u20090.0013; and NHRVS: best Pu2009=u2009.006, R2u2009=u20090.0072). After recalculating MDD PRS using MDD GWAS data sets without comorbid MDD-AD cases, significant evidence was observed for an association between the MDD PRS and AD in the meta-analysis of 3 GWAS AD samples without MDD cases (best Pu2009=u2009.007). Conclusions and Relevance These results suggest that shared genetic susceptibility contributes modestly to MDD and AD comorbidity. Individuals with elevated polygenic risk for MDD may also be at risk for AD.


Molecular Brain | 2016

Ethanol-mediated activation of the NLRP3 inflammasome in iPS cells and iPS cells-derived neural progenitor cells

Lidia De Filippis; Apoorva Halikere; Heather McGowan; Jennifer C. Moore; Jay A. Tischfield; Ronald P. Hart; Zhiping P. Pang

BackgroundAlcohol abuse produces an enormous impact on health, society, and the economy. Currently, there are very limited therapies available, largely due to the poor understanding of mechanisms underlying alcohol use disorders (AUDs) in humans. Oxidative damage of mitochondria and cellular proteins aggravates the progression of neuroinflammation and neurological disorders initiated by alcohol abuse.ResultsHere we show that ethanol exposure causes neuroinflammation in both human induced pluripotent stem (iPS) cells and human neural progenitor cells (NPCs). Ethanol exposure for 24 hours or 7 days does not affect the proliferation of iPS cells and NPCs, but primes an innate immune-like response by activating the NLR family pyrin domain containing 3 (NLRP3) inflammasome pathway. This leads to an increase of microtubule-associated protein 1A/1B-light chain 3+ (LC3B+) autophagic puncta and impairment of the mitochondrial and lysosomal distribution. In addition, a decrease of mature neurons derived from differentiating NPCs is evident in ethanol pre-exposed compared to control NPCs. Moreover, a second insult of a pro-inflammatory factor in addition to ethanol preexposure enhances innate cellular inflammation in human iPS cells.ConclusionsThis study provides strong evidence that neuronal inflammation contributes to the pathophysiology of AUDs through the activation of the inflammasome pathway in human cellular models.


Journal of Psychiatric Research | 2016

Pre- and perinatal complications in relation to Tourette syndrome and co-occurring obsessive-compulsive disorder and attention-deficit/hyperactivity disorder

Mohamed Abdulkadir; Jay A. Tischfield; Robert A. King; Thomas V. Fernandez; Lawrence W. Brown; Keun Ah Cheon; Barbara J. Coffey; Sebastian F T M de Bruijn; Lonneke Elzerman; Blanca Garcia-Delgar; Donald L. Gilbert; Dorothy E. Grice; Julie Hagstrøm; Tamasine Hedderly; Isobel Heyman; Hyun Ju Hong; Chaim Huyser; Laura Ibanez-Gomez; Young Key Kim; Young Shin Kim; Yun Joo Koh; Sodahm Kook; Samuel Kuperman; Andreas Lamerz; Bennett L. Leventhal; Andrea G. Ludolph; Marcos Madruga-Garrido; Athanasios Maras; Marieke D. Messchendorp; Pablo Mir

Pre- and perinatal complications have been implicated in the onset and clinical expression of Tourette syndrome albeit with considerable inconsistencies across studies. Also, little is known about their role in co-occurring obsessive-compulsive disorder (OCD) and attention-deficit/hyperactivity disorder (ADHD) in individuals with a tic disorder. Therefore, we aimed to investigate the role of pre- and perinatal complications in relation to the presence and symptom severity of chronic tic disorder and co-occurring OCD and ADHD using data of 1113 participants from the Tourette International Collaborative Genetics study. This study included 586 participants with a chronic tic disorder and 527 unaffected family controls. We controlled for age and sex differences by creating propensity score matched subsamples for both case-control and within-case analyses. We found that premature birth (ORxa0=xa01.72) and morning sickness requiring medical attention (ORxa0=xa02.57) were associated with the presence of a chronic tic disorder. Also, the total number of pre- and perinatal complications was higher in those with a tic disorder (ORxa0=xa01.07). Furthermore, neonatal complications were related to the presence (ORxa0=xa01.46) and severity (bxa0=xa02.27) of co-occurring OCD and also to ADHD severity (bxa0=xa01.09). Delivery complications were only related to co-occurring OCD (ORxa0=xa01.49). We conclude that early exposure to adverse situations during pregnancy is related to the presence of chronic tic disorders. Exposure at a later stage, at birth or during the first weeks of life, appears to be associated with co-occurring OCD and ADHD.


Scientific Reports | 2016

Increased nicotine response in iPSC-derived human neurons carrying the CHRNA5 N398 allele

Eileen N. Oni; Apoorva Halikere; Guohui Li; Alana J. Toro-Ramos; Mavis R. Swerdel; Jessica L. Verpeut; Jennifer C. Moore; Nicholas T. Bello; Laura J. Bierut; Alison Goate; Jay A. Tischfield; Zhiping P. Pang; Ronald P. Hart

Genetic variation in nicotinic receptor alpha 5 (CHRNA5) has been associated with increased risk of addiction-associated phenotypes in humans yet little is known the underlying neural basis. Induced pluripotent stem cells (iPSCs) were derived from donors homozygous for either the major (D398) or the minor (N398) allele of the nonsynonymous single nucleotide polymorphism (SNP), rs16969968, in CHRNA5. To understand the impact of these nicotinic receptor variants in humans, we differentiated these iPSCs to dopamine (DA) or glutamatergic neurons and then tested their functional properties and response to nicotine. Results show that N398 variant human DA neurons differentially express genes associated with ligand receptor interaction and synaptic function. While both variants exhibited physiological properties consistent with mature neuronal function, the N398 neuronal population responded more actively with an increased excitatory postsynaptic current response upon the application of nicotine in both DA and glutamatergic neurons. Glutamatergic N398 neurons responded to lower nicotine doses (0.1u2009μM) with greater frequency and amplitude but they also exhibited rapid desensitization, consistent with previous analyses of N398-associated nicotinic receptor function. This study offers a proof-of-principle for utilizing human neurons to study gene variants contribution to addiction.


Journal of Medicinal Chemistry | 2016

l-Cystine Diamides as l-Cystine Crystallization Inhibitors for Cystinuria

Longqin Hu; Yanhui Yang; Herve Aloysius; Haifa Albanyan; Min Yang; Jian Jie Liang; Anthony C. Yu; Alexander G. Shtukenberg; Laura N. Poloni; Vladyslav Kholodovych; Jay A. Tischfield; David S. Goldfarb; Michael D. Ward; Amrik Sahota

l-Cystine bismorpholide (1a) and l-cystine bis(N-methylpiperazide) (1b) were seven and twenty-four times more effective than l-cystine dimethyl ester (CDME) in increasing the metastable supersaturation range of l-cystine, respectively, effectively inhibiting l-cystine crystallization. This behavior can be attributed to inhibition of crystal growth at microscopic length scale, as revealed by atomic force microscopy. Both 1a and 1b are more stable than CDME, and 1b was effective in vivo in a knockout mouse model of cystinuria.


Frontiers in Neuroscience | 2016

Targeted Re-Sequencing Approach of Candidate Genes Implicates Rare Potentially Functional Variants in Tourette Syndrome Etiology

John Alexander; Hera Potamianou; Jinchuan Xing; Li Deng; Iordanis Karagiannidis; Fotis Tsetsos; Petros Drineas; Zsanett Tarnok; Renata Rizzo; Tomasz Wolańczyk; Luca Farkas; Péter Nagy; Urszula Szymanska; Christos Androutsos; Vaia Tsironi; Anastasia Koumoula; Csaba Barta; TSGeneSEE; Paul Sandor; Cathy L. Barr; Jay A. Tischfield; Peristera Paschou; Gary A. Heiman; Marianthi Georgitsi

Although the genetic basis of Tourette Syndrome (TS) remains unclear, several candidate genes have been implicated. Using a set of 382 TS individuals of European ancestry we investigated four candidate genes for TS (HDC, SLITRK1, BTBD9, and SLC6A4) in an effort to identify possibly causal variants using a targeted re-sequencing approach by next generation sequencing technology. Identification of possible disease causing variants under different modes of inheritance was performed using the algorithms implemented in VAAST. We prioritized variants using Variant ranker and validated five rare variants via Sanger sequencing in HDC and SLITRK1, all of which are predicted to be deleterious. Intriguingly, one of the identified variants is in linkage disequilibrium with a variant that is included among the top hits of a genome-wide association study for response to citalopram treatment, an antidepressant drug with off-label use also in obsessive compulsive disorder. Our findings provide additional evidence for the implication of these two genes in TS susceptibility and the possible role of these proteins in the pathobiology of TS should be revisited.


Nature Medicine | 2017

α-Lipoic acid treatment prevents cystine urolithiasis in a mouse model of cystinuria

Tiffany Zee; Neelanjan Bose; Jarcy Zee; Jennifer Beck; See Yang; Jaspreet Parihar; Min Yang; Sruthi Damodar; David Hall; Monique N. O'Leary; Arvind Ramanathan; Roy Gerona; David W. Killilea; Thomas Chi; Jay A. Tischfield; Amrik Sahota; Arnold Kahn; Marshall L. Stoller; Pankaj Kapahi

Cystinuria is an incompletely dominant disorder characterized by defective urinary cystine reabsorption that results in the formation of cystine-based urinary stones. Current treatment options are limited in their effectiveness at preventing stone recurrence and are often poorly tolerated. We report that the nutritional supplement α-lipoic acid inhibits cystine stone formation in the Slc3a1−/− mouse model of cystinuria by increasing the solubility of urinary cystine. These findings identify a novel therapeutic strategy for the clinical treatment of cystinuria.


Translational Psychiatry | 2016

Genome-wide polygenic scores for age at onset of alcohol dependence and association with alcohol-related measures

Manav Kapoor; Yi-Ling Chou; Howard J. Edenberg; Tatiana Foroud; Nicholas G. Martin; P. A. F. Madden; Jen-Chyong Wang; Sarah Bertelsen; Leah Wetherill; Andrew I. Brooks; Grace Chan; Victor Hesselbrock; Samuel Kuperman; Sarah E. Medland; G. W. Montgomery; Jay A. Tischfield; John Whitfield; Laura J. Bierut; A. C. Heath; K. K. Bucholz; Alison Goate; Arpana Agrawal

Age at onset of alcohol dependence (AO-AD) is a defining feature of multiple drinking typologies. AO-AD is heritable and likely shares genetic liability with other aspects of alcohol consumption. We examine whether polygenic variation in AO-AD, based on a genome-wide association study (GWAS), was associated with AO-AD and other aspects of alcohol consumption in two independent samples. Genetic risk scores (GRS) were created based on AO-AD GWAS results from a discovery sample of 1788 regular drinkers from extended pedigrees from the Collaborative Study of the Genetics of Alcoholism (COGA). GRS were used to predict AO-AD, AD and Alcohol dependence symptom count (AD-SX), age at onset of intoxication (AO-I), as well as maxdrinks in regular drinking participants from two independent samples—the Study of Addictions: Genes and Environment (SAGE; n=2336) and an Australian sample (OZ-ALC; n=5816). GRS for AO-AD from COGA explained a modest but significant proportion of the variance in all alcohol-related phenotypes in SAGE. Despite including effect sizes associated with large numbers of single nucleotide polymorphisms (SNPs; >110u2009000), GRS explained, at most, 0.7% of the variance in these alcohol measures in this independent sample. In OZ-ALC, significant but even more modest associations were noted with variance estimates ranging from 0.03 to 0.16%. In conclusion, there is modest evidence that genetic variation in AO-AD is associated with liability to other aspects of alcohol involvement.


Frontiers in Psychiatry | 2016

Functional Evaluations of Genes Disrupted in Patients with Tourette’s Disorder

Nawei Sun; Jay A. Tischfield; Robert A. King; Gary A. Heiman

Tourette’s disorder (TD) is a highly heritable neurodevelopmental disorder with complex genetic architecture and unclear neuropathology. Disruptions of particular genes have been identified in subsets of TD patients. However, none of the findings have been replicated, probably due to the complex and heterogeneous genetic architecture of TD that involves both common and rare variants. To understand the etiology of TD, functional analyses are required to characterize the molecular and cellular consequences caused by mutations in candidate genes. Such molecular and cellular alterations may converge into common biological pathways underlying the heterogeneous genetic etiology of TD patients. Herein, we review specific genes implicated in TD etiology, discuss the functions of these genes in the mammalian central nervous system and the corresponding behavioral anomalies exhibited in animal models, and importantly, review functional analyses that can be performed to evaluate the role(s) that the genetic disruptions might play in TD. Specifically, the functional assays include novel cell culture systems, genome editing techniques, bioinformatics approaches, transcriptomic analyses, and genetically modified animal models applied or developed to study genes associated with TD or with other neurodevelopmental and neuropsychiatric disorders. By describing methods used to study diseases with genetic architecture similar to TD, we hope to develop a systematic framework for investigating the etiology of TD and related disorders.

Collaboration


Dive into the Jay A. Tischfield's collaboration.

Top Co-Authors

Avatar

Samuel Kuperman

Roy J. and Lucille A. Carver College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alison Goate

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Bernice Porjesz

SUNY Downstate Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jacquelyn L. Meyers

SUNY Downstate Medical Center

View shared research outputs
Top Co-Authors

Avatar

John I. Nurnberger

Indiana University – Purdue University Indianapolis

View shared research outputs
Top Co-Authors

Avatar

Laura J. Bierut

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge