Network


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

Hotspot


Dive into the research topics where Kimberly Lezon-Geyda is active.

Publication


Featured researches published by Kimberly Lezon-Geyda.


Blood | 2011

PR-domain-containing Mds1-Evi1 is critical for long-term hematopoietic stem cell function

Yi Zhang; Sandra Stehling-Sun; Kimberly Lezon-Geyda; Subhash C. Juneja; Lucie Coillard; Gouri Chatterjee; Charles A. Wuertzer; Fernando D. Camargo; Archibald S. Perkins

The Mds1 and Evi1 complex locus (Mecom) gives rise to several alternative transcripts implicated in leukemogenesis. However, the contribution that Mecom-derived gene products make to normal hematopoiesis remains largely unexplored. To investigate the role of the upstream transcription start site of Mecom in adult hematopoiesis, we created a mouse model with a lacZ knock-in at this site, termed ME(m1), which eliminates Mds1-Evi1 (ME), the longer, PR-domain-containing isoform produced by the gene (also known as PRDM3). β-galactosidase-marking studies revealed that, within hematopoietic cells, ME is exclusively expressed in the stem cell compartment. ME deficiency leads to a reduction in the number of HSCs and a complete loss of long-term repopulation capacity, whereas the stem cell compartment is shifted from quiescence to active cycling. Genetic exploration of the relative roles of endogenous ME and EVI1 isoforms revealed that ME preferentially rescues long-term HSC defects. RNA-seq analysis in Lin(-)Sca-1(+)c-Kit(+) cells (LSKs) of ME(m1) documents near complete silencing of Cdkn1c, encoding negative cell-cycle regulator p57-Kip2. Reintroduction of ME into ME(m1) LSKs leads to normalization of both p57-Kip2 expression and growth control. Our results clearly demonstrate a critical role of PR-domain-containing ME in linking p57-kip2 regulation to long-term HSC function.


Blood | 2015

Mutations in the Gardos channel (KCNN4) are associated with hereditary xerocytosis

Edyta Glogowska; Kimberly Lezon-Geyda; Yelena Maksimova; Vincent P. Schulz; Patrick G. Gallagher

Hereditary xerocytosis (HX; MIM 194380) is an autosomal-dominant hemolytic anemia characterized by primary erythrocyte dehydration. In many patients, heterozygous mutations associated with delayed channel inactivation have been identified in PIEZO1. This report describes patients from 2 well-phenotyped HX kindreds, including from one of the first HX kindreds described, who lack predicted heterozygous PIEZO1-linked variants. Whole-exome sequencing identified novel, heterozygous mutations affecting the Gardos channel, encoded by the KCNN4 gene, in both kindreds. Segregation analyses confirmed transmission of the Gardos channel mutations with disease phenotype in affected individuals. The KCNN4 variants were different mutations in the same residue, which is highly conserved across species and within members of the small-intermediate family of calcium-activated potassium channel proteins. Both mutations were predicted to be deleterious by mutation effect algorithms. In sickle erythrocytes, the Gardos channel is activated under deoxy conditions, leading to cellular dehydration due to salt and water loss. The identification of KCNN4 mutations in HX patients supports recent studies that indicate it plays a critical role in normal erythrocyte deformation in the microcirculation and participates in maintenance of erythrocyte volume homeostasis.


PLOS ONE | 2013

Molecular Phenotypes in Triple Negative Breast Cancer from African American Patients Suggest Targets for Therapy

Robert Lindner; Catherine Sullivan; Onyinye Offor; Kimberly Lezon-Geyda; Kyle Halligan; Neal Fischbach; Mansi Shah; Veerle Bossuyt; Vincent P. Schulz; David Tuck; Lyndsay Harris

Triple negative breast cancer (TNBC) is characterized by high proliferation, poor differentiation and a poor prognosis due to high rates of recurrence. Despite lower overall incidence African American (AA) patients suffer from higher breast cancer mortality in part due to the higher proportion of TNBC cases among AA patients compared to European Americans (EA). It was recently shown that the clinical heterogeneity of TNBC is reflected by distinct transcriptional programs with distinct drug response profiles in preclinical models. In this study, gene expression profiling and immunohistochemistry were used to elucidate potential differences between TNBC tumors of EA and AA patients on a molecular level. In a retrospective cohort of 136 TNBC patients, a major transcriptional signature of proliferation was found to be significantly upregulated in samples of AA ethnicity. Furthermore, transcriptional profiles of AA tumors showed differential activation of insulin-like growth factor 1 (IGF1) and a signature of BRCA1 deficiency in this cohort. Using signatures derived from the meta-analysis of TNBC gene expression carried out by Lehmann et al., tumors from AA patients were more likely of basal-like subtypes whereas transcriptional features of many EA samples corresponded to mesenchymal-like or luminal androgen receptor driven subtypes. These results were validated in The Cancer Genome Atlas mRNA and protein expression data, again showing enrichment of a basal-like phenotype in AA tumors and mesenchymal subtypes in EA tumors. In addition, increased expression of VEGF-activated genes together with elevated microvessel area determined by the AQUA method suggest that AA patients exhibit higher tumor vascularization. This study confirms the existence of distinct transcriptional programs in triple negative breast cancer in two separate cohorts and that these programs differ by racial group. Differences in TNBC subtypes and levels of tumor angiogenesis in AA versus EA patients suggest that targeted therapy choices should be considered in the context of race.


Biochemistry | 2011

Targeting a DNA binding motif of the EVI1 protein by a pyrrole-imidazole polyamide

Yi Zhang; Géraldine Sicot; Xiaohui Cui; Marion Vogel; Charles A. Wuertzer; Kimberly Lezon-Geyda; John Wheeler; Daniel A. Harki; Katy A. Muzikar; Daniel A. Stolper; Peter B. Dervan; Archibald S. Perkins

The zinc finger protein EVI1 is causally associated with acute myeloid leukemogenesis, and inhibition of its function with a small molecule therapeutic may provide effective therapy for EVI1-expressing leukemias. In this paper we describe the development of a pyrrole-imidazole polyamide to specifically block EVI1 binding to DNA. We first identify essential domains for leukemogenesis through structure-function studies on both EVI1 and the t(3;21)(q26;q22)-derived RUNX1-MDS1-EVI1 (RME) protein, which revealed that DNA binding to the cognate motif GACAAGATA via the first of two zinc finger domains (ZF1, encompassing fingers 1-7) is essential transforming activity. To inhibit DNA binding via ZF1, we synthesized a pyrrole-imidazole polyamide 1, designed to bind to a subsite within the GACAAGATA motif and thereby block EVI1 binding. DNase I footprinting and electromobility shift assays revealed a specific and high affinity interaction between polyamide 1 and the GACAAGATA motif. In an in vivo CAT reporter assay using NIH-3T3-derived cell line with a chromosome-embedded tet-inducible EVI1-VP16 as well as an EVI1-responsive reporter, polyamide 1 completely blocked EVI1-responsive reporter activity. Growth of a leukemic cell line bearing overexpressed EVI1 was also inhibited by treatment with polyamide 1, while a control cell line lacking EVI1 was not. Finally, colony formation by RME was attenuated by polyamide 1 in a serial replating assay. These studies provide evidence that a cell permeable small molecule may effectively block the activity of a leukemogenic transcription factor and provide a valuable tool to dissect critical functions of EVI1 in leukemogenesis.


Blood | 2014

Global transcriptome analysis and enhancer landscape of human primary T follicular helper and T effector lymphocytes

Jason S. Weinstein; Kimberly Lezon-Geyda; Yelena Maksimova; Samuel Craft; Yaoping Zhang; Mack Su; Vincent P. Schulz; Joe Craft; Patrick G. Gallagher

T follicular helper (Tfh) cells are a subset of CD4(+) T helper cells that migrate into germinal centers and promote B-cell maturation into memory B and plasma cells. Tfh cells are necessary for promotion of protective humoral immunity following pathogen challenge, but when aberrantly regulated, drive pathogenic antibody formation in autoimmunity and undergo neoplastic transformation in angioimmunoblastic T-cell lymphoma and other primary cutaneous T-cell lymphomas. Limited information is available on the expression and regulation of genes in human Tfh cells. Using a fluorescence-activated cell sorting-based strategy, we obtained primary Tfh and non-Tfh T effector cells from tonsils and prepared genome-wide maps of active, intermediate, and poised enhancers determined by chromatin immunoprecipitation-sequencing, with parallel transcriptome analyses determined by RNA sequencing. Tfh cell enhancers were enriched near genes highly expressed in lymphoid cells or involved in lymphoid cell function, with many mapping to sites previously associated with autoimmune disease in genome-wide association studies. A group of active enhancers unique to Tfh cells associated with differentially expressed genes was identified. Fragments from these regions directed expression in reporter gene assays. These data provide a significant resource for studies of T lymphocyte development and differentiation and normal and perturbed Tfh cell function.


Blood | 2017

Novel mechanisms of PIEZO1 dysfunction in hereditary xerocytosis

Edyta Glogowska; Eve R. Schneider; Yelena Maksimova; Vincent P. Schulz; Kimberly Lezon-Geyda; John Wu; Kottayam Radhakrishnan; Siobán B. Keel; Donald H. Mahoney; Alison M. Freidmann; Rachel A. Altura; Elena O. Gracheva; Sviatoslav N. Bagriantsev; Theodosia A. Kalfa; Patrick G. Gallagher

Mutations in PIEZO1 are the primary cause of hereditary xerocytosis, a clinically heterogeneous, dominantly inherited disorder of erythrocyte dehydration. We used next-generation sequencing-based techniques to identify PIEZO1 mutations in individuals from 9 kindreds referred with suspected hereditary xerocytosis (HX) and/or undiagnosed congenital hemolytic anemia. Mutations were primarily found in the highly conserved, COOH-terminal pore-region domain. Several mutations were novel and demonstrated ethnic specificity. We characterized these mutations using genomic-, bioinformatic-, cell biology-, and physiology-based functional assays. For these studies, we created a novel, cell-based in vivo system for study of wild-type and variant PIEZO1 membrane protein expression, trafficking, and electrophysiology in a rigorous manner. Previous reports have indicated HX-associated PIEZO1 variants exhibit a partial gain-of-function phenotype with generation of mechanically activated currents that inactivate more slowly than wild type, indicating that increased cation permeability may lead to dehydration of PIEZO1-mutant HX erythrocytes. In addition to delayed channel inactivation, we found additional alterations in mutant PIEZO1 channel kinetics, differences in response to osmotic stress, and altered membrane protein trafficking, predicting variant alleles that worsen or ameliorate erythrocyte hydration. These results extend the genetic heterogeneity observed in HX and indicate that various pathophysiologic mechanisms contribute to the HX phenotype.


Bone | 2014

Deletion of mecom in mouse results in early-onset spinal deformity and osteopenia

Subhash C. Juneja; Alin Vonica; Caroline Zeiss; Kimberly Lezon-Geyda; Bogdan Yatsula; David R. Sell; Vincent M. Monnier; Sharon Lin; Thomas Ardito; David R. Eyre; David G. Reynolds; Zhenqiang Yao; Hani A. Awad; Hongbo Yu; Michael Wilson; Sylvie Honnons; Brendan F. Boyce; Lianping Xing; Yi Zhang; Archibald S. Perkins

Recent studies have indicated a role for a MECOM allele in susceptibility to osteoporotic fractures in humans. We have generated a mutation in Mecom in mouse (termed ME(m1)) via lacZ knock-in into the upstream transcription start site for the gene, resulting in disruption of Mds1 and Mds1-Evi1 transcripts, but not of Evi1 transcripts. We demonstrate that ME(m1/m1) mice have severe kyphoscoliosis that is reminiscent of human congenital or primary kyphoscoliosis. ME(m1/m1) mice appear normal at birth, but by 2weeks, they exhibit a slight lumbar lordosis and narrowed intervertebral space. This progresses to severe lordosis with disc collapse and synostosis, together with kyphoscoliosis. Bone formation and strength testing show that ME(m1/m1) mice have normal bone formation and composition but are osteopenic. While endochondral bone development is normal, it is markedly dysplastic in its organization. Electron micrographs of the 1week postnatal intervertebral discs reveals marked disarray of collagen fibers, consistent with an inherent weakness in the non-osseous connective tissue associated with the spine. These findings indicate that lack of ME leads to a complex defect in both osseous and non-osseous musculoskeletal tissues, including a marked vertebral osteopenia, degeneration of the IVD, and disarray of connective tissues, which is likely due to an inherent inability to establish and/or maintain components of these tissues.


Blood | 2018

Clinical spectrum of pyruvate kinase deficiency: Data from the pyruvate kinase deficiency natural history study

Rachael F. Grace; Paola Bianchi; Eduard J. van Beers; Stefan Eber; Bertil Glader; Hassan M. Yaish; Jenny M. Despotovic; Jennifer A. Rothman; Mukta Sharma; Melissa Mcnaull; Elisa Fermo; Kimberly Lezon-Geyda; D. Holmes Morton; Ellis J. Neufeld; Satheesh Chonat; Nina Kollmar; Christine M. Knoll; Kevin H.M. Kuo; Janet L. Kwiatkowski; Dagmar Pospisilova; Yves Pastore; Alexis A. Thompson; Peter E. Newburger; Yaddanapudi Ravindranath; Winfred C. Wang; Marcin W. Wlodarski; Heng Wang; Susanne Holzhauer; Vicky R. Breakey; Joachim B. Kunz

An international, multicenter registry was established to collect retrospective and prospective clinical data on patients with pyruvate kinase (PK) deficiency, the most common glycolytic defect causing congenital nonspherocytic hemolytic anemia. Medical history and laboratory and radiologic data were retrospectively collected at enrollment for 254 patients with molecularly confirmed PK deficiency. Perinatal complications were common, including anemia that required transfusions, hyperbilirubinemia, hydrops, and prematurity. Nearly all newborns were treated with phototherapy (93%), and many were treated with exchange transfusions (46%). Children age 5 years and younger were often transfused until splenectomy. Splenectomy (150 [59%] of 254 patients) was associated with a median increase in hemoglobin of 1.6 g/dL and a decreased transfusion burden in 90% of patients. Predictors of a response to splenectomy included higher presplenectomy hemoglobin (P = .007), lower indirect bilirubin (P = .005), and missense PKLR mutations (P = .0017). Postsplenectomy thrombosis was reported in 11% of patients. The most frequent complications included iron overload (48%) and gallstones (45%), but other complications such as aplastic crises, osteopenia/bone fragility, extramedullary hematopoiesis, postsplenectomy sepsis, pulmonary hypertension, and leg ulcers were not uncommon. Overall, 87 (34%) of 254 patients had both a splenectomy and cholecystectomy. In those who had a splenectomy without simultaneous cholecystectomy, 48% later required a cholecystectomy. Although the risk of complications increases with severity of anemia and a genotype-phenotype relationship was observed, complications were common in all patients with PK deficiency. Diagnostic testing for PK deficiency should be considered in patients with apparent congenital hemolytic anemia and close monitoring for iron overload, gallstones, and other complications is needed regardless of baseline hemoglobin. This trial was registered at www.clinicaltrials.gov as #NCT02053480.


International Journal of Surgical Pathology | 2011

Chromosome 7 Aneusomy in Metaplastic Breast Carcinomas With Chondroid, Squamous, and Spindle-Cell Differentiation

Katja Gwin; Kimberly Lezon-Geyda; Lyndsay Harris; Fattaneh A. Tavassoli

Metaplastic breast carcinomas (MBCs) are basal-like tumors that frequently express epidermal growth factor receptor (EGFR) via an unknown underlying genetic mechanism. In this study, the EGFR/CEP7 gene copy number in 17 MBCs with chondroid, squamous, and spindle-cell differentiation showing EGFR expression by immunohistochemistry was analyzed using fluorescence in situ hybridization. All cases had a balanced EGFR/CEP7 ratio. EGFR gene amplification was not observed in any case. Monosomy was found in 25% and polysomy in 12.5% of carcinomas with chondroid differentiation. All spindle-cell carcinomas and 50% of squamous carcinomas showed trisomy. Comparison with CEP7 copy number revealed aneusomy of chromosome 7, as opposed to increases or decreases specific to the EGFR gene or 7p. Although no direct correlation between EGFR expression by immunohistochemistry and aneusomy was observed, cases with a score of 3+ showed a higher frequency of EGFR gene copy gain. In the absence of EGFR amplification, chromosome 7 aneusomy might be a useful criterion for the determination of potential candidates for EGFR tyrosine kinase inhibitor clinical trials.


International Journal of Cancer | 2016

Brief‐exposure to preoperative bevacizumab reveals a TGF‐β signature predictive of response in HER2‐negative breast cancers

Vinay Varadan; Sitharthan Kamalakaran; Hannah Gilmore; Nilanjana Banerjee; Angel Janevski; Kristy Miskimen; Nicole Williams; Ajay Basavanhalli; Anant Madabhushi; Kimberly Lezon-Geyda; Veerle Bossuyt; Donald R. Lannin; Maysa Abu-Khalaf; William M. Sikov; Nevenka Dimitrova; Lyndsay Harris

To best define biomarkers of response, and to shed insight on mechanism of action of certain clinically important agents for early breast cancer, we used a brief‐exposure paradigm in the preoperative setting to study transcriptional changes in patient tumors that occur with one dose of therapy prior to combination chemotherapy. Tumor biopsies from breast cancer patients enrolled in two preoperative clinical trials were obtained at baseline and after one dose of bevacizumab (HER2‐negative), trastuzumab (HER2‐positive) or nab‐paclitaxel, followed by treatment with combination chemo‐biologic therapy. RNA‐Sequencing based PAM50 subtyping at baseline of 46 HER2‐negative patients revealed a strong association between the basal‐like subtype and pathologic complete response (pCR) to chemotherapy plus bevacizumab (p ≤ 0.0027), but did not provide sufficient specificity to predict response. However, a single dose of bevacizumab resulted in down‐regulation of a well‐characterized TGF‐β activity signature in every single breast tumor that achieved pCR (p ≤ 0.004). The TGF‐β signature was confirmed to be a tumor‐specific read‐out of the canonical TGF‐β pathway using pSMAD2 (p ≤ 0.04), with predictive power unique to brief‐exposure to bevacizumab (p ≤ 0.016), but not trastuzumab or nab‐paclitaxel. Down‐regulation of TGF‐β activity was associated with reduction in tumor hypoxia by transcription and protein levels, suggesting therapy‐induced disruption of an autocrine‐loop between tumor stroma and malignant cells. Modulation of the TGF‐β pathway upon brief‐exposure to bevacizumab may provide an early functional readout of pCR to preoperative anti‐angiogenic therapy in HER2‐negative breast cancer, thus providing additional avenues for exploration in both preclinical and clinical settings with these agents.

Collaboration


Dive into the Kimberly Lezon-Geyda's collaboration.

Top Co-Authors

Avatar

Lyndsay Harris

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Vinay Varadan

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Archibald S. Perkins

University of Rochester Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge