Network


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

Hotspot


Dive into the research topics where Suvarnamala Pushkaran is active.

Publication


Featured researches published by Suvarnamala Pushkaran.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Unrestrained erythroblast development in Nix−/− mice reveals a mechanism for apoptotic modulation of erythropoiesis

Abhinav Diwan; Andrew G. Koesters; Amy Odley; Suvarnamala Pushkaran; Christopher P. Baines; Benjamin T. Spike; Diedre Daria; Anil G. Jegga; Hartmut Geiger; Bruce J. Aronow; Jeffery D. Molkentin; Kay F. Macleod; Theodosia A. Kalfa; Gerald W. Dorn

Normal production of RBCs requires that the antiapoptotic protein Bcl-xl be induced at end stages of differentiation in response to erythropoietin (Epo) signaling. The critical proapoptotic pathways inhibited by Bcl-xl in erythroblasts are unknown. We used gene targeting in the mouse to evaluate the BH3-only factor Nix, which is transcriptionally up-regulated during Epo-stimulated in vitro erythrocyte differentiation. Nix null mice are viable and fertile. Peripheral blood counts revealed a profound reticulocytosis and thrombocytosis despite normal serum Epo levels and blood oxygen tension. Nix null mice exhibited massive splenomegaly, with splenic and bone marrow erythroblastosis and reduced apoptosis in vivo during erythrocyte maturation. Hematopoietic progenitor populations were unaffected. Cultured Nix null erythroid cells were hypersensitive to Epo and resistant to apoptosis stimulated by cytokine deprivation and calcium ionophore. Transcriptional profiling of Nix null spleens revealed increased expression of cell cycle and erythroid genes, including Bcl-xl, and diminished expression of cell death and B cell-related genes. Thus, cell-autonomous Nix-mediated apoptosis in opposition to the Epo-induced erythroblast survival pathway appears indispensable for regulation of erythrocyte production and maintenance of hematological homeostasis. These results suggest that physiological codependence and coordinated regulation of pro- and antiapoptotic Bcl2 family members may represent a general regulatory paradigm in hematopoiesis.


Blood | 2012

Signaling and cytoskeletal requirements in erythroblast enucleation

Diamantis G. Konstantinidis; Suvarnamala Pushkaran; James F. Johnson; Jose A. Cancelas; Stefanos Manganaris; Chad E. Harris; David A. Williams; Yi Zheng; Theodosia A. Kalfa

To understand the role of cytoskeleton and membrane signaling molecules in erythroblast enucleation, we developed a novel analysis protocol of multiparameter high-speed cell imaging in flow. This protocol enabled us to observe F-actin and phosphorylated myosin regulatory light chain (pMRLC) assembled into a contractile actomyosin ring (CAR) between nascent reticulocyte and nucleus, in a population of enucleating erythroblasts. CAR formation and subsequent enucleation were not affected in murine erythroblasts with genetic deletion of Rac1 and Rac2 GTPases because of compensation by Rac3. Pharmacologic inhibition or genetic deletion of all Rac GTPases altered the distribution of F-actin and pMRLC and inhibited enucleation. Erythroblasts treated with NSC23766, cytochalasin-D, colchicine, ML7, or filipin that inhibited Rac activity, actin or tubulin polymerization, MRLC phosphorylation, or lipid raft assembly, respectively, exhibited decreased enucleation efficiency, as quantified by flow cytometry. As assessed by high-speed flow-imaging analysis, colchicine inhibited erythroblast polarization, implicating microtubules during the preparatory stage of enucleation, whereas NSC23766 led to absence of lipid raft assembly in the reticulocyte-pyrenocyte border. In conclusion, enucleation is a multistep process that resembles cytokinesis, requiring establishment of cell polarity through microtubule function, followed by formation of a contractile actomyosin ring, and coalescence of lipid rafts between reticulocyte and pyrenocyte.


Haematologica | 2010

Rac1 and Rac2 GTPases are necessary for early erythropoietic expansion in the bone marrow but not in the spleen.

Theodosia A. Kalfa; Suvarnamala Pushkaran; Xiaoling Zhang; James F. Johnson; Dao Pan; Deidre Daria; Hartmut Geiger; Jose A. Cancelas; David A. Williams; Yi Zheng

Background The small Rho GTPases Rac1 and Rac2 have both overlapping and distinct roles in actin organization, cell survival, and proliferation in various hematopoietic cell lineages. The role of these Rac GTPases in erythropoiesis has not yet been fully elucidated. Design and Methods Cre-recombinase-induced deletion of Rac1 genomic sequence was accomplished on a Rac2-null genetic background, in mouse hematopoietic cells in vivo. The erythroid progenitors and precursors in the bone marrow and spleen of these genetically engineered animals were evaluated by colony assays and flow cytometry. Apoptosis and proliferation of the different stages of erythroid progenitors and precursors were evaluated by flow cytometry. Results Erythropoiesis in Rac1−/−;Rac2−/− mice is characterized by abnormal burst-forming unit-erythroid colony morphology and decreased numbers of megakaryocyte-erythrocyte progenitors, erythroid colony-forming units, and erythroblasts in the bone marrow. In contrast, splenic erythropoiesis is increased. Combined Rac1 and Rac2 deficiency compromises proliferation of the megakaryocyte-erythrocyte progenitor population in the bone marrow, while it allows increased survival and proliferation of megakaryocyte-erythrocyte progenitors in the spleen. Conclusions These data suggest that Rac1 and Rac2 GTPases are essential for normal bone marrow erythropoiesis but that they are dispensable for erythropoiesis in the spleen, implying different signaling pathways for homeostatic and stress erythropoiesis.


Blood Cells Molecules and Diseases | 2010

Altered phosphorylation of cytoskeleton proteins in sickle red blood cells: The role of protein kinase C, Rac GTPases, and reactive oxygen species

Alex George; Suvarnamala Pushkaran; Lina Li; Xiuli An; Yi Zheng; Narla Mohandas; Clinton H. Joiner; Theodosia A. Kalfa

The small Rho GTPases Rac1 and Rac2 regulate actin structures and mediate reactive oxygen species (ROS) production via NADPH oxidase in a variety of cells. We have demonstrated that deficiency of Rac1 and Rac2 GTPases in mice disrupts the normal hexagonal organization of the RBC cytoskeleton and reduces erythrocyte deformability. This is associated with increased phosphorylation of adducin at Ser-724, (corresponding to Ser-726 in human erythrocytes), a domain target of protein kinase C (PKC). PKC phosphorylates adducin and leads to decreased F-actin capping and dissociation of spectrin from actin, implicating a significant role of such phosphorylation in cytoskeletal remodeling. We evaluated adducin phosphorylation in erythrocytes from patients with sickle cell disease and found it consistently increased at Ser-726. In addition, ROS concentration is elevated in sickle erythrocytes by 150-250% compared to erythrocytes from normal control individuals. Here, we review previous studies demonstrating that altered phosphorylation of erythrocyte cytoskeletal proteins and increased ROS production result in disruption of cytoskeleton stability in healthy and sickle cell erythrocytes. We discuss in particular the known and potential roles of protein kinase C and the Rac GTPases in these two processes.


Blood | 2015

Cytokinesis failure in RhoA-deficient mouse erythroblasts involves actomyosin and midbody dysregulation and triggers p53 activation

Diamantis G. Konstantinidis; Katie M. Giger; Mary A. Risinger; Suvarnamala Pushkaran; Ping Zhou; Phillip Dexheimer; Satwica Yerneni; Paul R. Andreassen; Ursula Klingmüller; James Palis; Yi Zheng; Theodosia A. Kalfa

RhoA GTPase has been shown in vitro in cell lines and in vivo in nonmammalian organisms to regulate cell division, particularly during cytokinesis and abscission, when 2 daughter cells partition through coordinated actomyosin and microtubule machineries. To investigate the role of this GTPase in the rapidly proliferating mammalian erythroid lineage, we developed a mouse model with erythroid-specific deletion of RhoA. This model was proved embryonic lethal as a result of severe anemia by embryonic day 16.5 (E16.5). The primitive red blood cells were enlarged, poikilocytic, and frequently multinucleated, but were able to sustain life despite experiencing cytokinesis failure. In contrast, definitive erythropoiesis failed and the mice died by E16.5, with profound reduction of maturing erythroblast populations within the fetal liver. RhoA was required to activate myosin-regulatory light chain and localized at the site of the midbody formation in dividing wild-type erythroblasts. Cytokinesis failure caused by RhoA deficiency resulted in p53 activation and p21-transcriptional upregulation with associated cell-cycle arrest, increased DNA damage, and cell death. Our findings demonstrate the role of RhoA as a critical regulator for efficient erythroblast proliferation and the p53 pathway as a powerful quality control mechanism in erythropoiesis.


Pediatric Blood & Cancer | 2011

Cooperating G6PD Mutations Associated with Severe Neonatal Hyperbilirubinemia and Cholestasis

Benjamin Mizukawa; Alex George; Suvarnamala Pushkaran; Lana Weckbach; KarenAnn Kalinyak; James E. Heubi; Theodosia A. Kalfa

We report a novel glucose‐6‐phosphate dehydrogenase (G6PD) mutation, which we propose to name G6PD Cincinnati (c.1037A > T, p.N346I), found in combination with G6PD Gastonia (c.637G > T, p.V213L) in an infant who presented with neonatal cholestasis. The G6PD Cincinnati mutation results in a non‐conservative amino acid substitution at the tetramer interface disturbing its formation, as seen by native gel electrophoresis and immunoblotting. G6PD Gastonia disrupts dimerization of the enzyme and by itself causes chronic non‐spherocytic hemolytic anemia. The G6PD Cincinnati mutation may have aggravated the clinical picture of G6PD Gastonia with the result of severe perinatal hemolysis causing cholestasis and associated liver injury. Pediatr Blood Cancer 2011;56:840–842.


Blood | 2006

Rac GTPases regulate the morphology and deformability of the erythrocyte cytoskeleton

Theodosia A. Kalfa; Suvarnamala Pushkaran; Narla Mohandas; John H. Hartwig; Velia M. Fowler; James F. Johnson; Clinton H. Joiner; David A. Williams; Yi Zheng


Blood | 2007

Cdc42 critically regulates the balance between myelopoiesis and erythropoiesis.

Linda Yang; Lei Wang; Theodosia A. Kalfa; Jose A. Cancelas; Xun Shang; Suvarnamala Pushkaran; Jun Mo; David A. Williams; Yi Zheng


Journal of Visualized Experiments | 2014

Identification of a Murine Erythroblast Subpopulation Enriched in Enucleating Events by Multi-spectral Imaging Flow Cytometry

Diamantis G. Konstantinidis; Suvarnamala Pushkaran; Katie M. Giger; Stefanos Manganaris; Yi Zheng; Theodosia A. Kalfa


Blood | 2013

Development Of a Comprehensive Rapid Next-Generation Sequencing Assay For The Diagnosis Of Inherited Hemolytic Anemia

Neha Dagaonkar; Suvarnamala Pushkaran; Katie M. Giger; Ammar Husami; Diane Kissell; Clinton H. Joiner; Mehdi Keddache; Kejian Zhang; Theodosia A. Kalfa

Collaboration


Dive into the Suvarnamala Pushkaran's collaboration.

Top Co-Authors

Avatar

Theodosia A. Kalfa

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Yi Zheng

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

David A. Williams

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

James F. Johnson

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jose A. Cancelas

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Alex George

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Diamantis G. Konstantinidis

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Narla Mohandas

Lawrence Berkeley National Laboratory

View shared research outputs
Top Co-Authors

Avatar

Katie M. Giger

University of Cincinnati

View shared research outputs
Researchain Logo
Decentralizing Knowledge