Network


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

Hotspot


Dive into the research topics where Kathy L. McGraw is active.

Publication


Featured researches published by Kathy L. McGraw.


Blood | 2012

STAT3 mutations unify the pathogenesis of chronic lymphoproliferative disorders of NK cells and T-cell large granular lymphocyte leukemia

Andres Jerez; Michael J. Clemente; Hideki Makishima; Hanna L M Koskela; Francis LeBlanc; Kwok Peng Ng; Thomas L. Olson; Bartlomiej Przychodzen; Manuel Afable; Inés Gómez-Seguí; Kathryn M Guinta; Lisa Durkin; Eric D. Hsi; Kathy L. McGraw; Dan Zhang; Marcin W. Wlodarski; Kimmo Porkka; Mikkael A. Sekeres; Alan F. List; Satu Mustjoki; Thomas P. Loughran; Jaroslaw P. Maciejewski

Chronic lymphoproliferative disorders of natural killer cells (CLPD-NKs) and T-cell large granular lymphocytic leukemias (T-LGLs) are clonal lymphoproliferations arising from either natural killer cells or cytotoxic T lymphocytes (CTLs). We have investigated for distribution and functional significance of mutations in 50 CLPD-NKs and 120 T-LGL patients by direct sequencing, allele-specific PCR, and microarray analysis. STAT3 gene mutations are present in both T and NK diseases: approximately one-third of patients with each type of disorder convey these mutations. Mutations were found in exons 21 and 20, encoding the Src homology 2 domain. Patients with mutations are characterized by symptomatic disease (75%), history of multiple treatments, and a specific pattern of STAT3 activation and gene deregulation, including increased expression of genes activated by STAT3. Many of these features are also found in patients with wild-type STAT3, indicating that other mechanisms of STAT3 activation can be operative in these chronic lymphoproliferative disorders. Treatment with STAT3 inhibitors, both in wild-type and mutant cases, resulted in accelerated apoptosis. STAT3 mutations are frequent in large granular lymphocytes suggesting a similar molecular dysregulation in malignant chronic expansions of NK and CTL origin. STAT3 mutations may distinguish truly malignant lymphoproliferations involving T and NK cells from reactive expansions.


Blood | 2012

Loss of heterozygosity in 7q myeloid disorders: clinical associations and genomic pathogenesis

Andres Jerez; Yuka Sugimoto; Hideki Makishima; Amit Verma; Anna M. Jankowska; Bartlomiej Przychodzen; Valeria Visconte; Ramon V. Tiu; Christine L. O'Keefe; Azim Mohamedali; Austin Kulasekararaj; Andrea Pellagatti; Kathy L. McGraw; Hideki Muramatsu; Alison R. Moliterno; Mikkael A. Sekeres; Michael A. McDevitt; Seiji Kojima; Alan F. List; Jacqueline Boultwood; Ghulam J. Mufti; Jaroslaw P. Maciejewski

Loss of heterozygosity affecting chromosome 7q is common in acute myeloid leukemia and myelodysplastic syndromes, pointing toward the essential role of this region in disease phenotype and clonal evolution. The higher resolution offered by recently developed genomic platforms may be used to establish more precise clinical correlations and identify specific target genes. We analyzed a series of patients with myeloid disorders using recent genomic technologies (1458 by single-nucleotide polymorphism arrays [SNP-A], 226 by next-generation sequencing, and 183 by expression microarrays). Using SNP-A, we identified chromosome 7q loss of heterozygosity segments in 161 of 1458 patients (11%); 26% of chronic myelomonocytic leukemia patients harbored 7q uniparental disomy, of which 41% had a homozygous EZH2 mutation. In addition, we describe an SNP-A-isolated deletion 7 hypocellular myelodysplastic syndrome subset, with a high rate of progression. Using direct and parallel sequencing, we found no recurrent mutations in typically large deletion 7q and monosomy 7 patients. In contrast, we detected a markedly decreased expression of genes included in our SNP-A defined minimally deleted regions. Although a 2-hit model is present in most patients with 7q uniparental disomy and a myeloproliferative phenotype, haplodeficient expression of defined regions of 7q may underlie pathogenesis in patients with deletions and predominant dysplastic features.


Oncogene | 2013

Lenalidomide Promotes p53 Degradation by Inhibiting MDM2 Auto-ubiquitination in Myelodysplastic Syndrome with Chromosome 5q Deletion

Sheng Wei; Xianghong Chen; Kathy L. McGraw; Lan Min Zhang; Rami S. Komrokji; Justine Clark; Gisela Caceres; D Billingsley; Lubomir Sokol; Jeffrey E. Lancet; Nicole Fortenbery; J Zhou; E A Eksioglu; David Sallman; Hong-Gang Wang; P.K. Epling-Burnette; Julie Y. Djeu; Mikkael A. Sekeres; Jaroslaw P. Maciejewski; Alan F. List

Allelic deletion of the RPS14 gene is a key effector of the hypoplastic anemia in patients with myelodysplastic syndrome (MDS) and chromosome 5q deletion (del(5q)). Disruption of ribosome integrity liberates free ribosomal proteins to bind to and trigger degradation of mouse double minute 2 protein (MDM2), with consequent p53 transactivation. Herein we show that p53 is overexpressed in erythroid precursors of primary bone marrow del(5q) MDS specimens accompanied by reduced cellular MDM2. More importantly, we show that lenalidomide (Len) acts to stabilize MDM2, thereby accelerating p53 degradation. Biochemical and molecular analyses showed that Len inhibits the haplodeficient protein phosphatase 2A catalytic domain alpha (PP2Acα) phosphatase resulting in hyperphosphorylation of inhibitory serine-166 and serine-186 residues on MDM2, and displaces binding of RPS14 to suppress MDM2 autoubiquitination whereas PP2Acα overexpression promotes drug resistance. Bone marrow specimens from del(5q) MDS patients resistant to Len overexpressed PP2Acα accompanied by restored accumulation of p53 in erythroid precursors. Our findings indicate that Len restores MDM2 functionality in the 5q- syndrome to overcome p53 activation in response to nucleolar stress, and therefore may warrant investigation in other disorders of ribosomal biogenesis.


Blood | 2013

GM-CSF–dependent pSTAT5 sensitivity is a feature with therapeutic potential in chronic myelomonocytic leukemia

Eric Padron; Jeffrey S. Painter; Sateesh Kunigal; Adam W. Mailloux; Kathy L. McGraw; Jessica M. McDaniel; Eunhee Kim; Christopher Bebbington; Mark Baer; Geoffrey T. Yarranton; Jeffrey E. Lancet; Rami S. Komrokji; Omar Abdel-Wahab; Alan F. List; P.K. Epling-Burnette

Granulocyte-macrophage-colony-stimulating factor (GM-CSF) hypersensitivity is a hallmark of juvenile myelomonocytic leukemia (JMML) but has not been systematically shown in the related human disease chronic myelomonocytic leukemia (CMML). We find that primary CMML samples demonstrate GM-CSF-dependent hypersensitivity by hematopoietic colony formation assays and phospho-STAT5 (pSTAT5) flow cytometry compared with healthy donors. Among CMML patients, the pSTAT5 hypersensitive response positively correlated with high-risk disease, peripheral leukocytes, monocytes, and signaling-associated mutations. When compared with IL-3 and G-CSF, GM-CSF hypersensitivity was cytokine specific and thus a possible target for intervention in CMML. To explore this possibility, we treated primary CMML cells with KB003, a novel monoclonal anti-GM-CSF antibody, and JAK2 inhibitors. We found that an elevated proportion of immature GM-CSF receptor-α(R) subunit-expressing cells were present in the bone marrow myeloid compartment of CMML. In survival assays, we found that myeloid and monocytic progenitors were sensitive to GM-CSF signal inhibition. Our data indicate that a committed myeloid precursor expressing CD38 may represent the progenitor population with enhanced GM-CSF dependence in CMML, consistent with results in JMML. These preclinical data indicate that GM-CSF signaling inhibitors merit further investigation in CMML and that GM-CSFR expression on myeloid progenitors may be a biomarker for this therapy.


Blood | 2013

STAT3-mutations indicate the presence of subclinical T cell clones in a subset of aplastic anemia and myelodysplastic syndrome patients

Andres Jerez; Michael J. Clemente; Hideki Makishima; Hanna Rajala; Inés Gómez-Seguí; Thomas L. Olson; Kathy L. McGraw; Bartlomiej Przychodzen; Austin Kulasekararaj; Manuel Afable; Holleh D Husseinzadeh; Naoko Hosono; Francis LeBlanc; Sonja Lagström; Dan Zhang; Pekka Ellonen; André Tichelli; Catherine Nissen; Alan E. Lichtin; Aleksandra Wodnar-Filipowicz; Ghulam J. Mufti; Alan F. List; Satu Mustjoki; Thomas P. Loughran; Jaroslaw P. Maciejewski

Large granular lymphocyte leukemia (LGL) is often associated with immune cytopenias and can cooccur in the context of aplastic anemia (AA) and myelodysplastic syndromes (MDS). We took advantage of the recent description of signal transducer and activator of transcription 3 (STAT3) mutations in LGL clonal expansions to test, using sensitive methods, for the presence of these mutations in a large cohort of 367 MDS and 140 AA cases. STAT3 clones can be found not only in known LGL concomitant cases, but in a small proportion of unsuspected ones (7% AA and 2.5% MDS). In STAT3-mutated AA patients, an interesting trend toward better responses of immunosuppressive therapy and an association with the presence of human leukocyte antigen-DR15 were found. MDSs harboring a STAT3 mutant clone showed a lower degree of bone marrow cellularity and a higher frequency of developing chromosome 7 abnormalities. STAT3-mutant LGL clones may facilitate a persistently dysregulated autoimmune activation, responsible for the primary induction of bone marrow failure in a subset of AA and MDS patients.


British Journal of Haematology | 2011

Identification of a risk dependent microRNA expression signature in myelodysplastic syndromes

Lubomir Sokol; Gisela Caceres; Stefano Volinia; Hans Alder; Gerard J. Nuovo; Chang Gong Liu; Kathy L. McGraw; Justine Clark; Celia Sigua; Dung Tsa Chen; Lynn C. Moscinski; Carlo M. Croce; Alan F. List

The myelodysplastic syndromes (MDS) display both haematological and biological heterogeneity with variable leukaemia potential. MicroRNAs play an important role in tumour suppression and the regulation of self‐renewal and differentiation of haematopoietic progenitors. Using a microarray platform, we evaluated microRNA expression from 44 patients with MDS and 17 normal controls. We identified a thirteen microRNA signature with statistically significant differential expression between normal and MDS specimens (P < 0·01), including down‐regulation of members of the leukaemia‐associated MIRLET7 family. A unique signature consisting of 10 microRNAs was closely associated with International Prognostic Scoring System (IPSS) risk category permitting discrimination between lower (Low/Intermediate‐1) and higher risk (Intermediate‐2/High) disease (P < 0·01). Selective overexpression of MIR181 family members was detected in higher risk MDS, indicating pathogenetic overlap with acute myeloid leukaemia. Survival analysis of an independent cohort of 22 IPSS lower risk MDS patients revealed a median survival of 3·5 years in patients with high expression of MIR181 family compared to 9·3 years in patients with low MIR181 expression (P = 0·002). Our pilot study suggested that analysis of microRNA expression profile offers diagnostic utility, and provide pathogenetic and prognostic discrimination in MDS.


Proceedings of the National Academy of Sciences of the United States of America | 2013

TP53 suppression promotes erythropoiesis in del(5q) MDS, suggesting a targeted therapeutic strategy in lenalidomide-resistant patients.

Gisela Caceres; Kathy L. McGraw; Bon Ham Yip; Andrea Pellagatti; Joseph O. Johnson; Ling Zhang; Kenian Liu; Lan Min Zhang; William J. Fulp; Ji-Hyun Lee; Najla Al Ali; Ashley A. Basiorka; Larry J. Smith; F. Joseph Daugherty; Neil Littleton; Richard A. Wells; Lubomir Sokol; Sheng Wei; Rami S. Komrokji; Jacqueline Boultwood; Alan F. List

Significance The hypoplastic anemia characteristic of del(5q) myelodysplastic syndrome (MDS) arises from ribosomal protein insufficiency, resulting in erythroid-specific activation of p53. We found that suppression of p53 by cenersen, an antisense oligonucleotide, markedly improved erythroid colony formation in primary MDS specimens assessed by two-stage colony formation assay. Erythropoietic rescue significantly correlated with the magnitude of reduction in nuclear p53. In addition, in a cohort of eight lower-risk, lenalidomide-refractory del(5q) MDS patients treated with lenalidomide and dexamethasone, a glucocorticoid receptor-dependent antagonist of p53, transfusion independence was restored in five of eight patients, accompanied by in vivo expansion of erythroid precursors without clonal suppression. These results suggest inhibition of p53 may be a unique therapeutic strategy in patients with lenalidomide-resistant del(5q) MDS. Stabilization of p53 in erythroid precursors in response to nucleosomal stress underlies the hypoplastic anemia in myelodysplastic syndromes (MDS) with chromosome 5q deletion [del(5q)]. We investigated whether cenersen, a clinically active 20-mer antisense oligonucleotide complementary to TP53 exon10, could suppress p53 expression and restore erythropoiesis in del(5q) MDS. Cenersen treatment of ribosomal protein S-14-deficient erythroblasts significantly reduced cellular p53 and p53-up-regulated modulator of apoptosis expression compared with controls, accompanied by a significant reduction in apoptosis and increased cell proliferation. In a two-stage erythroid differentiation assay, cenersen significantly suppressed nuclear p53 in bone marrow CD34+ cells isolated from patients with del(5q) MDS, whereas erythroid burst recovery increased proportionally to the magnitude of p53 suppression without evidence of del(5q) clonal suppression (r = −0.6; P = 0.005). To explore the effect of p53 suppression on erythropoiesis in vivo, dexamethasone, a glucocorticoid receptor-dependent p53 antagonist, was added to lenalidomide treatment in eight lower-risk, transfusion-dependent, del(5q) MDS patients with acquired drug resistance. Transfusion independence was restored in five patients accompanied by expansion of erythroid precursors and decreased cellular p53 expression. We conclude that targeted suppression of p53 could support effective erythropoiesis in lenalidomide-resistant del(5q) MDS.


Blood | 2016

The NLRP3 Inflammasome functions as a driver of the myelodysplastic syndrome phenotype.

Ashley A. Basiorka; Kathy L. McGraw; Erika A. Eksioglu; Xianghong Chen; Joseph O. Johnson; Ling Zhang; Qing Zhang; Brittany Irvine; Thomas Cluzeau; David Sallman; Eric Padron; Rami S. Komrokji; Lubomir Sokol; Rebecca C. Coll; Avril A. B. Robertson; Matthew A. Cooper; John L. Cleveland; Luke A. J. O'Neill; Sheng Wei; Alan F. List

Despite genetic heterogeneity, myelodysplastic syndromes (MDSs) share features of cytological dysplasia and ineffective hematopoiesis. We report that a hallmark of MDSs is activation of the NLRP3 inflammasome, which drives clonal expansion and pyroptotic cell death. Independent of genotype, MDS hematopoietic stem and progenitor cells (HSPCs) overexpress inflammasome proteins and manifest activated NLRP3 complexes that direct activation of caspase-1, generation of interleukin-1β (IL-1β) and IL-18, and pyroptotic cell death. Mechanistically, pyroptosis is triggered by the alarmin S100A9 that is found in excess in MDS HSPCs and bone marrow plasma. Further, like somatic gene mutations, S100A9-induced signaling activates NADPH oxidase (NOX), increasing levels of reactive oxygen species (ROS) that initiate cation influx, cell swelling, and β-catenin activation. Notably, knockdown of NLRP3 or caspase-1, neutralization of S100A9, and pharmacologic inhibition of NLRP3 or NOX suppress pyroptosis, ROS generation, and nuclear β-catenin in MDSs and are sufficient to restore effective hematopoiesis. Thus, alarmins and founder gene mutations in MDSs license a common redox-sensitive inflammasome circuit, which suggests new avenues for therapeutic intervention.


Leukemia | 2016

Targeted resequencing analysis of 31 genes commonly mutated in myeloid disorders in serial samples from myelodysplastic syndrome patients showing disease progression

Andrea Pellagatti; Swagata Roy; C Di Genua; Adam Burns; Kathy L. McGraw; Simona Valletta; María José Larrayoz; Marta Fernandez-Mercado; Joanne Mason; Sally Killick; Cristina Mecucci; María José Calasanz; Alan F. List; Anna Schuh; Jacqueline Boultwood

Targeted resequencing analysis of 31 genes commonly mutated in myeloid disorders in serial samples from myelodysplastic syndrome patients showing disease progression


British Journal of Haematology | 2013

JAK2-V617F-mediated signalling is dependent on lipid rafts and statins inhibit JAK2-V617F-dependent cell growth

Lori N. Griner; Kathy L. McGraw; Joseph O. Johnson; Alan F. List; Gary W. Reuther

Aberrant JAK2 signalling plays an important role in the aetiology of myeloproliferative neoplasms (MPNs). JAK2 inhibitors, however, do not readily eliminate neoplastic MPN cells and thus do not induce patient remission. Further understanding JAK2 signalling in MPNs may uncover novel avenues for therapeutic intervention. Recent work has suggested a potential role for cellular cholesterol in the activation of JAK2 by the erythropoietin receptor and in the development of an MPN‐like disorder in mice. Our study demonstrates for the first time that the MPN‐associated JAK2‐V617F kinase localizes to lipid rafts and that JAK2‐V617F‐dependent signalling is inhibited by lipid raft disrupting agents, which target membrane cholesterol, a critical component of rafts. We also show for the first time that statins, 3‐hydroxy‐3‐methyl‐glutaryl coenzyme A (HMG‐CoA) reductase inhibitors, widely used to treat hypercholesterolaemia, induce apoptosis and inhibit JAK2‐V617F‐dependent cell growth. These cells are more sensitive to statin treatment than non‐JAK2‐V617F‐dependent cells. Importantly, statin treatment inhibited erythropoietin‐independent erythroid colony formation of primary cells from MPN patients, but had no effect on erythroid colony formation from healthy individuals. Our study is the first to demonstrate that JAK2‐V617F signalling is dependent on lipid rafts and that statins may be effective in a potential therapeutic approach for MPNs.

Collaboration


Dive into the Kathy L. McGraw's collaboration.

Top Co-Authors

Avatar

Alan F. List

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Rami S. Komrokji

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Lubomir Sokol

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Ashley A. Basiorka

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Sheng Wei

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Eric Padron

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

David Sallman

University of South Florida

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ling Zhang

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Jeffrey E. Lancet

University of South Florida

View shared research outputs
Researchain Logo
Decentralizing Knowledge