Network


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

Hotspot


Dive into the research topics where Mark A. Kimak is active.

Publication


Featured researches published by Mark A. Kimak.


Nature Genetics | 2000

Missense mutations interfere with VEGFR-3 signalling in primary lymphoedema.

Marika J. Karkkainen; Robert E. Ferrell; Elizabeth C. Lawrence; Mark A. Kimak; Kara L. Levinson; Michele A. McTigue; Kari Alitalo; David N. Finegold

Primary lymphoedema is a rare, autosomal dominant disorder that leads to a disabling and disfiguring swelling of the extremities and, when untreated, tends to worsen with time. Here we link primary human lymphoedema to the FLT4 locus, encoding vascular endothelial growth factor receptor-3 (VEGFR-3), in several families. All disease-associated alleles analysed had missense mutations and encoded proteins with an inactive tyrosine kinase, preventing downstream gene activation. Our study establishes that VEGFR-3 is important for normal lymphatic vascular function and that mutations interfering with VEGFR-3 signal transduction are a cause of primary lymphoedema.


Molecular Psychiatry | 2009

Genetic variation in components of dopamine neurotransmission impacts ventral striatal reactivity associated with impulsivity

Erika E. Forbes; Sarah M. Brown; Mark A. Kimak; Robert E. Ferrell; Stephen B. Manuck; Ahmad R. Hariri

Individual differences in traits such as impulsivity involve high reward sensitivity and are associated with risk for substance use disorders. The ventral striatum (VS) has been widely implicated in reward processing, and individual differences in its function are linked to these disorders. Dopamine (DA) plays a critical role in reward processing and is a potent neuromodulator of VS reactivity. Moreover, altered DA signaling has been associated with normal and pathological reward-related behaviors. Functional polymorphisms in DA-related genes represent an important source of variability in DA function that may subsequently impact VS reactivity and associated reward-related behaviors. Using an imaging genetics approach, we examined the modulatory effects of common, putatively functional DA-related polymorphisms on reward-related VS reactivity associated with self-reported impulsivity. Genetic variants associated with relatively increased striatal DA release (DRD2 −141C deletion) and availability (DAT1 9-repeat), as well as diminished inhibitory postsynaptic DA effects (DRD2 −141C deletion and DRD4 7-repeat), predicted 9–12% of the interindividual variability in reward-related VS reactivity. In contrast, genetic variation directly affecting DA signaling only in the prefrontal cortex (COMT Val158Met) was not associated with variability in VS reactivity. Our results highlight an important role for genetic polymorphisms affecting striatal DA neurotransmission in mediating interindividual differences in reward-related VS reactivity. They further suggest that altered VS reactivity may represent a key neurobiological pathway through which these polymorphisms contribute to variability in behavioral impulsivity and related risk for substance use disorders.


Biological Psychiatry | 2009

Divergent Effects of Genetic Variation in Endocannabinoid Signaling on Human Threat- and Reward-Related Brain Function

Ahmad R. Hariri; Adam Gorka; Luke W. Hyde; Mark A. Kimak; Indrani Halder; Francesca Ducci; Robert E. Ferrell; David Goldman; Stephen B. Manuck

BACKGROUND Fatty acid amide hydrolase (FAAH) is a key enzyme in regulating endocannabinoid (eCB) signaling. A common single nucleotide polymorphism (C385A) in the human FAAH gene has been associated with increased risk for addiction and obesity. METHODS Using imaging genetics in 82 healthy adult volunteers, we examined the effects of FAAH C385A on threat- and reward-related human brain function. RESULTS Carriers of FAAH 385A, associated with reduced enzyme and possibly increased eCB signaling, had decreased threat-related amygdala reactivity but increased reward-related ventral striatal reactivity in comparison with C385 homozygotes. Similarly divergent effects of FAAH C385A genotype were manifest at the level of brain-behavior relationships. The 385A carriers showed decreased correlation between amygdala reactivity and trait anxiety but increased correlation between ventral striatal reactivity and delay discounting, an index of impulsivity. CONCLUSIONS Our results parallel pharmacologic and genetic dissection of eCB signaling, are consistent with the psychotropic effects of Delta(9)-tetrahydrocannabinol, and highlight specific neural mechanisms through which variability in eCB signaling impacts complex behavioral processes related to risk for addiction and obesity.


Archives of General Psychiatry | 2009

Effects of HTR1A C(-1019)G on amygdala reactivity and trait anxiety.

E. Fakra; Luke W. Hyde; Adam Gorka; Patrick M. Fisher; Karen E. Munoz; Mark A. Kimak; Indrani Halder; Robert E. Ferrell; Stephen B. Manuck; Ahmad R. Hariri

CONTEXT Serotonin 1A (5-hydroxytryptamine 1A [5-HT(1A)]) autoreceptors mediate negative feedback inhibition of serotonergic neurons and play a critical role in regulating serotonin signaling involved in shaping the functional response of major forebrain targets, such as the amygdala, supporting complex behavioral processes. A common functional variation (C[-1019]G) in the human 5-HT(1A) gene (HTR1A) represents 1 potential source of such interindividual variability. Both in vitro and in vivo, -1019G blocks transcriptional repression, leading to increased autoreceptor expression. Thus, -1019G may contribute to relatively decreased serotonin signaling at postsynaptic forebrain target sites via increased negative feedback. OBJECTIVES To evaluate the effects of HTR1A C(-1019)G on amygdala reactivity and to use path analyses to explore the impact of HTR1A-mediated variability in amygdala reactivity on individual differences in trait anxiety. We hypothesized that -1019G, which potentially results in decreased serotonin signaling, would be associated with relatively decreased amygdala reactivity and related trait anxiety. DESIGN Imaging genetics in participants from an archival database. PARTICIPANTS Eighty-nine healthy adults. RESULTS Consistent with prior findings, -1019G was associated with significantly decreased threat-related amygdala reactivity. Importantly, this effect was independent of that associated with another common functional polymorphism that affects serotonin signaling, 5-HTTLPR. While there were no direct genotype effects on trait anxiety, HTR1A C(-1019)G indirectly predicted 9.2% of interindividual variability in trait anxiety through its effects on amygdala reactivity. CONCLUSIONS Our findings further implicate relatively increased serotonin signaling, associated with a genetic variation that mediates increased 5-HT(1A) autoreceptors, in driving amygdala reactivity and trait anxiety. Moreover, they provide empirical documentation of the basic premise that genetic variation indirectly affects emergent behavioral processes related to psychiatric disease risk by biasing the response of underlying neural circuitries.


American Journal of Human Genetics | 2010

GJC2 Missense Mutations Cause Human Lymphedema

Robert E. Ferrell; Catherine J. Baty; Mark A. Kimak; Jenny M. Karlsson; Elizabeth C. Lawrence; Marlise Franke-Snyder; Eleanor Feingold; David N. Finegold

Lymphedema is the clinical manifestation of defects in lymphatic structure or function. Mutations identified in genes regulating lymphatic development result in inherited lymphedema. No mutations have yet been identified in genes mediating lymphatic function that result in inherited lymphedema. Survey microarray studies comparing lymphatic and blood endothelial cells identified expression of several connexins in lymphatic endothelial cells. Additionally, gap junctions are implicated in maintaining lymphatic flow. By sequencing GJA1, GJA4, and GJC2 in a group of families with dominantly inherited lymphedema, we identified six probands with unique missense mutations in GJC2 (encoding connexin [Cx] 47). Two larger families cosegregate lymphedema and GJC2 mutation (LOD score = 6.5). We hypothesize that missense mutations in GJC2 alter gap junction function and disrupt lymphatic flow. Until now, GJC2 mutations were only thought to cause dysmyelination, with primary expression of Cx47 limited to the central nervous system. The identification of GJC2 mutations as a cause of primary lymphedema raises the possibility of novel gap-junction-modifying agents as potential therapy for some forms of lymphedema.


Clinical Cancer Research | 2012

Connexin 47 mutations increase risk for secondary lymphedema following breast cancer treatment

David N. Finegold; Catherine J. Baty; Kelly Z. Knickelbein; Shelley Perschke; Sarah E. Noon; Diana Campbell; Jenny M. Karlsson; Diana Huang; Mark A. Kimak; Elizabeth C. Lawrence; Eleanor Feingold; Adam Brufsky; Robert E. Ferrell

Purpose: Secondary lymphedema is a frequent complication of breast cancer associated with surgery, chemotherapy, or radiation following breast cancer treatment. The potential contribution of genetic susceptibility to risk of developing secondary lymphedema following surgical trauma, radiation, and other tissue insults has not been studied. Experimental Design: To determine whether women with breast cancer and secondary lymphedema had mutations in candidate lymphedema genes, we undertook a case–control study of 188 women diagnosed with breast cancer recruited from the University of Pittsburgh Breast Cancer Program (http://www.upmccancercenter.com/breast/index.cfm) between 2000 and 2010. Candidate lymphedema genes, GJC2 (encoding connexin 47 [Cx47]), FOXC2, HGF, MET, and FLT4 (encoding VEGFR3), were sequenced for mutation. Bioinformatics analysis and in vitro functional assays were used to confirm significance of novel mutations. Results: Cx47 mutations were identified in individuals having secondary lymphedema following breast cancer treatment but not in breast cancer controls or normal women without breast cancer. These novel mutations are dysfunctional as assessed through in vitro assays and bioinformatics analysis and provide evidence that altered gap junction function leads to lymphedema. Conclusions: Our findings challenge the view that secondary lymphedema is solely due to mechanical trauma and support the hypothesis that genetic susceptibility is an important risk factor for secondary lymphedema. A priori recognition of genetic risk (i) raises the potential for early detection and intervention for a high-risk group and (ii) allows the possibility of altering surgical approach and/or chemo- and radiation therapy, or direct medical treatment of secondary lymphedema with novel connexin-modifying drugs. Clin Cancer Res; 18(8); 2382–90. ©2012 AACR.


American Journal of Ophthalmology | 2002

Lymphedema-distichiasis syndrome and FOXC2 gene mutation ☆

Elias I. Traboulsi; Khouloud Al-Khayer; Masayuki Matsumoto; Mark A. Kimak; Susan Crowe; Steven E. Wilson; David N. Finegold; Robert E. Ferrell; David M. Meisler

PURPOSE To describe the clinical characteristics of a family with autosomal dominant lymphedema-distichiasis syndrome and to report the results of analysis of the FOXC2 gene DESIGN Observational and experimental study. METHODS The setting was a clinical practice. The study population was 17 members of a family with lymphedema-distichiasis. Observation procedures were complete ophthalmologic examinations and collection of blood samples. DNA was extracted. Mutation analysis of the coding region of the FOXC2 gene was performed using direct sequencing of polymerase chain reaction (PCR) product and a restriction enzyme assay. The main outcome measure was inheritance of mutation in FOXC2 gene. RESULTS Nine patients had distichiasis or lymphedema or both and eight did not. Sequencing of the coding region of the only translated exon of the FOXC2 gene revealed a C to A transversion at position 939 resulting in a Tyr313Stop codon with premature termination of translation and a truncated protein product. The mutation was present in all nine affected individuals and in an asymptomatic 9-year-old boy. CONCLUSIONS Distichiasis-lymphedema syndrome results from mutations in FOXC2, a member of the forkhead/winged family of transcription factors. There is intrafamilial variation in the clinical expression of the mutation.


Human Molecular Genetics | 1998

Hereditary Lymphedema: Evidence for Linkage and Genetic Heterogeneity

Robert E. Ferrell; Kara L. Levinson; Judith H. Esman; Mark A. Kimak; Elizabeth C. Lawrence; M. Michael Barmada; David N. Finegold


Human Molecular Genetics | 2001

Truncating mutations in FOXC2 cause multiple lymphedema syndromes.

David N. Finegold; Mark A. Kimak; Elizabeth C. Lawrence; Kara L. Levinson; Elizabeth M. Cherniske; Barbara R. Pober; Jean W. Dunlap; Robert E. Ferrell


The FASEB Journal | 2001

VEGFR3 gene structure, regulatory region, and sequence polymorphisms

Kristiina Iljin; Marika J. Karkkainen; Elizabeth C. Lawrence; Mark A. Kimak; Marko Uutela; Jussi Taipale Katri Pajusola Leena Alhonen; Maria Halmekytö; David N. Finegold; Robert E. Ferrell; Kari Alitalo

Collaboration


Dive into the Mark A. Kimak's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Indrani Halder

University of Pittsburgh

View shared research outputs
Researchain Logo
Decentralizing Knowledge