Network


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

Hotspot


Dive into the research topics where Lalitha Nagarajan is active.

Publication


Featured researches published by Lalitha Nagarajan.


Biochemical and Biophysical Research Communications | 1982

Insulin promotes the growth of F9 embryonal carcinoma cells apparently by acting through its own receptor.

Lalitha Nagarajan; Wayne B. Anderson

Abstract Low concentrations of insulin (20 to 40 ng/ml) stimulate the growth of F9 embryonal carcinoma cells under defined, serum-free conditions. [125I] Insulin binding studies reveal the presence of high and low affinity receptor sites, and insulin does not compete for [125I] multiplication-stimulating activity (MSA) binding to F9 cells. The addition of antibodies to the insulin receptor (anti R) to serum-free growth medium also promotes F9 cell proliferation. Anti R blocks [125I] insulin binding to F9 cells, but does not alter the binding of [125I] MSA, indicating that anti R is exerting its growth-promoting effects by interacting with the insulin receptor. These results demonstrate that F9 cells do possess specific insulin receptors and establish that insulin stimulates the growth of these cells by acting directly through its own receptor and not through the MSA receptor also present on these cells.


Oncogene | 2005

SSBP2, a candidate tumor suppressor gene, induces growth arrest and differentiation of myeloid leukemia cells

Hong Liang; Susmita Samanta; Lalitha Nagarajan

Acute myelogenous leukemia (AML) is the most common leukemia in adults with clonal proliferation of myeloid stem cells. Two or more cooperating mechanisms, namely block in differentiation, enhanced proliferation and resistance to programmed cell death, underlie this neoplastic transformation. Nonrandom, complete and partial deletions of chromosome 5 are common anomalies in AML. Using positional cloning strategies, we characterized an evolutionarily conserved candidate myeloid leukemia suppressor gene encoding sequence-specific single-stranded DNA binding protein 2 (SSBP2) from chromosome 5q13.3, a locus that is frequently deleted in AML. Recent studies in Drosophila and Xenopus demonstrate a pivotal role for SSBPs in embryonic differentiation. In mammals, SSBP2 is one of three highly related and ubiquitously expressed genes. Here, we identify frequent loss of SSBP2 protein expression in human AML cell lines using highly specific antibodies. Furthermore, inducible expression of SSBP2 in the AML cell line U937 leads to loss of clonogenicity, G1 arrest and partial differentiation. Remarkably, inducible expression of SSBP2 is accompanied by downregulation of C-MYC expression. Our findings are consistent with human SSBP2 being a novel regulator of hematopoietic growth and differentiation, whose loss confers a block in differentiation advantage to myeloid leukemic cells.


Leukemia & Lymphoma | 1995

Molecular Analysis of the 5q- Chromosome

Lalitha Nagarajan

Acquired interstitial deletions of the long arm of chromosome 5, are seen in anomalies of the myeloid cells. The refractory anemia (RA) or 5q- syndrome, in which the erythroid and megakaryocytic lineages are predominantly affected, is a relatively indolent clinical entity distinguishable, from the constellation of preneoplastic myelodysplastic syndrome (MDS) and acute myelogenous leukemia (AML) with trilineage involvement. Recent molecular evidence suggests that the critical region of 5q deletion in MDS/AML resides in the D5S89 locus, which is proximal (centromeric) to the minimal region of loss in the 5q- syndrome RA. The invariable loss of the D5S89 locus in MDS/AML qualifies it for the MDS/AML tumor suppressor locus. The telomeric 5q31 gene governs erythroid and megakaryocytic differentiation and can be termed the RA locus. Isolation and characterization of these genes will lead to an understanding of molecular mechanisms underlying normal hematopoiesis and leukemic transformation.


Cancer Genetics and Cytogenetics | 1999

Spectral karyotypic study of the HL-60 cell line: Detection of complex rearrangements involving chromosomes 5, 7, and 16 and delineation of critical region of deletion on 5q31.1

Jan C. Liang; Yi Ning; Rui yu Wang; Hesed Padilla-Nash; Evelin Schröck; Dirk G. Soenksen; Lalitha Nagarajan; Thomas Ried

Interstitial deletions of the q arm of chromosome 5 have been associated with acute myelogenous leukemia (AML); therefore, accurate identification of rearrangements of this chromosome in a model cell line, HL-60, is important for understanding the critical genes involved in this disease. In this study, we employed a newly developed technology termed spectral karyotyping to delineate chromosomal rearrangements in this cell line. Our study revealed a derivative of chromosome 7 that resulted from translocations of chromosome arms 5q and 16q to 7q; that is, der(7)t(5;7)(?;q?)t(5;16)(?;q?). Interestingly, both chromosomes 5 and 7 were also involved in translocations with chromosome 16 in der(16) t(5;16)(q?;q?22-24) and der(16)t(7;16)(?;q?22-24), respectively. Other notable chromosomal abnormalities that were not previously reported in the HL-60 included an insertion of chromosome 8 in the q arm of chromosome 11, a translocation between chromosomes 9 and 14, and a translocation between chromosomes 14 and 15. In an attempt to define the loss of the 5q31.1 region in HL-60, we performed fluorescence in situ hybridization analysis by utilizing bacterial artificial chromosomes BAC1 and BAC2 that spanned the IL9 and EGR1 gene interval, which was previously shown to be a critical region of loss in AML. We showed that a copy of both BAC1 (spanning the D5S399 locus) and BAC2 (spanning the D5S393 locus centromeric to BAC1) were present in the normal chromosome 5, but a second copy of BAC1 was lost and a second copy of BAC2 was inserted in the der(16)t(7;16) chromosome. Thus, not only was this study the first to use the new 24-color karyotyping technique to identify several novel chromosomal rearrangements in HL-60, but it also narrowed the 5q31.1 critical region of deletion to the region represented by BAC1.


Cancer Genetics and Cytogenetics | 1994

5q- chromosome. Evidence for complex interstitial breaks in a case of refractory anemia with excess blasts.

Lalitha Nagarajan; Lian Zhao; Xiaoyan Lu; J.A. Warrington; John J. Wasmuth; Michael J. Siciliano; Albert B. Deisseroth; Jan C. Liang

Interstitial loss of the long arm of chromosome 5 (5q-) is an anomaly frequently seen in myelodysplasia (MDS) and acute myelogenous leukemia (AML). Although the limits of the interstitial deletions vary among patients, there is a critical region of overlap at 5q31 that is consistently deleted in most cases. The order of genes in the critical 5q31 region is centromere, interleukin gene cluster, an anonymous polymorphic locus D5S89, early growth response factor, CSF1 receptor, telomere. Fluorescence in situ hybridization of specific 5q31 probes to metaphases with del(5) (q11q31) from a patient with secondary refractory anemia with excess blasts in transformation demonstrates that the interstitial deletion is not contiguous. The 5q- chromosome has lost the D5S89 and CSF1R loci while retaining some of the sequences in between. A probe derived from a 300-kbp yeast artificial chromosome containing the D5S89 locus is interrupted on the normal chromosome 5 of this patient. Data presented in this report are consistent with (i) presence of a critical gene within the YAC and (ii) more than a single interstitial break within the 5q- chromosome. These results, while pinpointing one of the critical 5q31 loci, also provide evidence for a second telomeric locus.


Oncogene | 2010

SSBP2 is an in vivo tumor suppressor and regulator of LDB1 stability

Yang Wang; Sherry Klumpp; Hesham M. Amin; Hong Liang; June Li; Zeev Estrov; Patrick A. Zweidler-McKay; Stephen J. Brandt; Alan Agulnick; Lalitha Nagarajan

SSBP proteins bind and stabilize transcriptional cofactor LIM domain-binding protein1 (LDB1) from proteosomal degradation to promote tissue-specific transcription through an evolutionarily conserved pathway. The human SSBP2 gene was isolated as a candidate tumor suppressor from a critical region of loss in chromosome 5q14.1. By gene targeting, we show increased predisposition to B-cell lymphomas and carcinomas in Ssbp2−/− mice. Remarkably, loss of Ssbp2 causes increased LDB1 turnover in the thymus, a pathway exploited in Trp53−/−Ssbp2−/− mice to develop highly aggressive, immature thymic lymphomas. Using T-cell differentiation as a model, we report a stage-specific upregulation of Ssbp2 expression, which in turn regulates LDB1 turnover under physiological conditions. Furthermore, transcript levels of pTα, a target of LDB1-containing complex, and a critical regulator T-cell differentiation are reduced in Ssbp2−/− immature thymocytes. Our findings suggest that disruption of the SSBP2-regulated pathways may be an infrequent but critical step in malignant transformation of multiple tissues.


Leukemia & Lymphoma | 1998

The Unexplored 5q13 Locus: A Role in Hematopoietic Malignancies

Patricia D. Castro; Jeffery Fairman; Lalitha Nagarajan

Deletions and translocations at 5q13 point out a locus involved in the development of acute myeloblastic leukemia (AML) and myelodysplastic syndromes (MDS) as well as other neoplasms. The chromosomal rearrangements of 5q13 are well documented, but have not been a primary focus of research. In this report, we provide evidence for a novel critical locus at 5q13.3, encoding gene(s) which may be disrupted by chromosomal translocations or deletions. Rare cases of myeloid neoplasms with t(5q13) as the sole chromosomal anomaly argue for a gene which gives rise to fusion proteins. Our preliminary studies have localized one of the critical genes to a <3 Mb. interval between the polymorphic markers AFMB347yf9 and GATAP18104 at the band 5q13.3. Other results also suggest that the 5q 13.3 locus may span a fragile site which undergoes unbalanced translocations and interstitial deletions accompanied by loss of significant segments of chromosome 5. Molecular reagents generated by the human genome mapping and sequencing initiative will allow us to characterize the critical genes at 5q13.3 and facilitate genotypic analysis of AML and MDS.


Neuromuscular Disorders | 2014

Most expression and splicing changes in myotonic dystrophy type 1 and type 2 skeletal muscle are shared with other muscular dystrophies

Linda L. Bachinski; Keith A. Baggerly; Valerie L. Neubauer; Tamara J. Nixon; Olayinka Raheem; Mario Sirito; Anna K. Unruh; Jiexin Zhang; Lalitha Nagarajan; Lubov Timchenko; Guillaume Bassez; Bruno Eymard; Josep Gamez; Tetsuo Ashizawa; B. Udd; Ralf Krahe

The prevailing pathomechanistic paradigm for myotonic dystrophy (DM) is that aberrant expression of embryonic/fetal mRNA/protein isoforms accounts for most aspects of the pleiotropic phenotype. To identify aberrant isoforms in skeletal muscle of DM1 and DM2 patients, we performed exon-array profiling and RT-PCR validation on the largest DM sample set to date, including Duchenne, Becker and tibial muscular dystrophy (NMD) patients as disease controls, and non-disease controls. Strikingly, most expression and splicing changes in DM patients were shared with NMD controls. Comparison between DM and NMD identified almost no significant differences. We conclude that DM1 and DM2 are essentially identical for dysregulation of gene expression, and DM expression changes represent a subset of broader spectrum dystrophic changes. We found no evidence for qualitative splicing differences between DM1 and DM2. While some DM-specific splicing differences exist, most of the DM splicing differences were also seen in NMD controls. SSBP3 exon 6 missplicing was observed in all diseased muscle and led to reduced protein. We conclude there is no widespread DM-specific spliceopathy in skeletal muscle and suggest that missplicing in DM (and NMD) may not be the driving mechanism for the muscle pathology, since the same pathways show expression changes unrelated to splicing.


Biochemical and Biophysical Research Communications | 2008

Single-stranded DNA-binding proteins regulate the abundance and function of the LIM-homeodomain transcription factor LHX2 in pituitary cells.

Ying Cai; Zhixiong Xu; Lalitha Nagarajan; Stephen J. Brandt

A family of single-stranded DNA-binding proteins (or SSBPs) has been shown to augment the function of LIM-homeodomain (LIM-HD) transcription factors in embryogenesis by interaction with LIM domain-binding protein-1 (LDB1). No DNA-binding complex has been described, however, containing a LIM-HD protein, LDB1, and SSBP, and the mechanism by which SSBPs affect LIM-HD function had not been elucidated. Through use of electrophoretic mobility shift, antibody supershift, and ChIP analyses, we show that an Lhx2-Ldb1-Ssbp3 complex binds a specific element in the Lhx2 target gene Cga (encoding the alpha subunit of glycoprotein hormones) in the alphaT3-1 pituitary cell line. Using overexpression and knockdown approaches, we demonstrate that SSBP3 inhibits Lhx2 and Ldb1 turnover, stimulates assembly of this DNA-binding complex, promotes its recruitment to the Cga promoter, and enhances Cga transcription. These studies provide novel insights into the regulation of pituitary gene expression and LIM-HD function more generally.


Experimental Cell Research | 1983

A new differentiated cell line (Dif 5) derived by retinoic acid treatment of F9 teratocarcinoma cells capable of extracellular matrix production and growth in the absence of serum

Lalitha Nagarajan; Anton M. Jetten; Wayne B. Anderson

Treatment of F9 teratocarcinoma cells with all trans retinoic acid (RA) causes them to differentiate into two or three morphologically distinct cell types. Whereas the majority of these retinoid-derived cells exhibit properties resembling parietal endoderm, a small percentage of this differentiated cell population manifests properties distinct from the parietal endoderm cell type. The isolation and partial characterization of such a non-parietal endoderm cell line (Dif 5) derived from F9 cells following prolonged (44 days) exposure to 1 microM retinoic acid are described. Unlike the retinoid-induced parietal endoderm-like cell population, which exhibits a dramatic, characteristic morphological change upon treatment with 8-bromo cAMP, Dif 5 cells do not show any morphological change with exposure to this cAMP analog. Dif 5 cells synthesize and deposit an extracellular matrix consisting of several components of Reicherts membrane (fibronectin, laminin, and type IV collagen). This new cell line does not synthesize alpha-fetoprotein but does secrete plasminogen activator. An interesting property of these cells is their ability to grow in the absence of serum or other hormonal supplements. Yet the Dif 5 cells do exhibit density-dependent inhibition of growth. Unlike the parent F9 cells or parietal yolk sac (PYS-2) cells, these cells do possess specific cell surface receptors for epidermal growth factor (EGF). The growth-arrested Dif 5 cells can be reinitiated to proliferate by the addition of fetal calf serum (FCS) or EGF. The properties of Dif 5 cells determined fail to fulfill all the characteristics described for either parietal or visceral endodermal cells. This raises the possibility that Dif 5 cells might represent an endodermal cell type which is intermediate in differentiation to either parietal or visceral endoderm but which lacks the biochemical signal to complete this stage of differentiation. This new Dif 5 cell line should be of considerable value in studying the modulation of growth requirements and extracellular matrix formation during early embryonic development.

Collaboration


Dive into the Lalitha Nagarajan's collaboration.

Top Co-Authors

Avatar

Hong Liang

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Stephen J. Brandt

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ying Cai

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jan C. Liang

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jeffery Fairman

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Wei Guo

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

David F. Claxton

Penn State Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Jeff Fairman

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lazar V. Kottickal

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge