Network


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

Hotspot


Dive into the research topics where Stephen J. Brandt is active.

Publication


Featured researches published by Stephen J. Brandt.


Journal of Clinical Investigation | 1990

Dysregulated interleukin 6 expression produces a syndrome resembling Castleman's disease in mice.

Stephen J. Brandt; David M. Bodine; Cynthia E. Dunbar; Arthur W. Nienhuis

Interleukin 6 (IL-6) is an important regulator of the acute phase response, T cell function, and terminal B cell differentiation. Excessive or inappropriate production of this cytokine may be involved in a variety of autoimmune and neoplastic disorders. To investigate the consequences of dysregulated synthesis of IL-6 in vivo, a high-titer recombinant retroviral vector produced in psi-2 packaging cells was used to introduce the coding sequences of murine IL-6 into mouse hematopoietic cells. Congenitally anemic W/Wv mice reconstituted with bone marrow cells transduced with the retroviral vector developed a syndrome characterized by anemia, transient granulocytosis, hypoalbuminemia, and polyclonal hypergammaglobulinemia, with marked splenomegaly and peripheral lymphadenopathy. Extensive plasma cell infiltration of lymph nodes, spleen, liver, and lung was noted. The similarity of these findings to those of multicentric Castlemans disease, taken together with the observation that lymph nodes from these patients elaborate large amounts of this cytokine, suggest that the inappropriate synthesis of IL-6 has a primary role in the pathogenesis of this systemic lymphoproliferative disorder.


Cell | 1987

Distinct patterns of expression of different protein kinase C mRNAs in rat tissues

Stephen J. Brandt; James E. Niedel; Robert M. Bell; W.Scott Young

The identification of multiple protein kinase C (PKC) cDNA sequences from rat, bovine, and human tissues has led to the discovery of a family of PKC genes. Using probes complementary to three cDNA sequences from rat brain, designated PKC-I, -II, and -III, the distribution of PKC transcripts in rat tissues was studied by in situ hybridization histochemistry. In brain, PKC-II and PKC-III transcripts colocalized except in the brain stem and spinal cord, where some cells appeared to contain only PKC-III message. PKC-I mRNA had a different, but partially overlapping, pattern of expression. In spleen, PKC-II and PKC-III, but not PKC-I, transcripts were concentrated in the marginal zone of white pulp, the major B-lymphocyte compartment of rat spleen. The distinct distributions of rat PKC mRNAs suggest that different members of the PKC family have specialized functions.


Current Opinion in Hematology | 2002

New insights into erythropoiesis.

Mark J. Koury; Stephen T. Sawyer; Stephen J. Brandt

Commitment of hematopoietic cells to the erythroid lineage involves the actions of several transcription factors, including TAL1, LMO2, and GATA-2. The differentiation of committed erythroid progenitor cells involves other transcription factors, including NF-E2 and EKLF. Upon binding erythropoietin, the principal regulator of erythropoiesis, cell surface erythropoietin receptors dimerize and activate specific intracellular kinases, including Janus family tyrosine protein kinase 2, phosphoinositol-3 kinase, and mitogen-activated protein kinase. Important substrates of these kinases are tyrosines in the erythropoietin receptors themselves and the signal transducer and transcription activator proteins. Erythropoietin prevents erythroid cell apoptosis. Some of the apoptotic tendency of erythroid cells can be attributed to proapoptotic molecules produced by hematopoietic cells, macrophages, and stromal cells. Cell divisions accompanying terminal erythroid differentiation are finely controlled by cell cycle regulators, and disruption of these terminal divisions causes erythroid cell apoptosis. In reticulocyte maturation, regulated degradation of internal organelles involves a lipoxygenase, whereas survival requires the antiapoptotic protein Bcl-x.


Molecular and Cellular Biology | 2003

Identification of a TAL1 target gene reveals a positive role for the LIM domain-binding protein Ldb1 in erythroid gene expression and differentiation.

Zhixiong Xu; Suming Huang; Long-Sheng Chang; Alan Agulnick; Stephen J. Brandt

ABSTRACT The TAL1 (or SCL) gene, originally identified from its involvement by a recurrent chromosomal translocation, encodes a basic helix-loop-helix transcription factor essential for erythropoiesis. Although presumed to regulate transcription, its target genes are largely unknown. We show here that a nuclear complex containing TAL1, its DNA-binding partner E47, zinc finger transcription factor GATA-1, LIM domain protein LMO2, and LIM domain-binding protein Ldb1 transactivates the protein 4.2 (P4.2) gene through two E box GATA elements in its proximal promoter. Binding of this complex to DNA was dependent on the integrity of both E box and GATA sites and was demonstrated to occur on the P4.2 promoter in cells. Maximal transcription in transiently transfected cells required both E box GATA elements and expression of all five components of the complex. This complex was shown, in addition, to be capable of linking in solution double-stranded oligonucleotides corresponding to the two P4.2 E box GATA elements. This DNA-linking activity required Ldb1 and increased with dimethyl sulfoxide-induced differentiation of murine erythroleukemia (MEL) cells. In contrast, enforced expression in MEL cells of dimerization-defective mutant Ldb1, as well as wild-type Ldb1, significantly decreased E box GATA DNA-binding activities, P4.2 promoter activity, and accumulation of P4.2 and β-globin mRNAs. These studies define a physiologic target for a TAL1- and GATA-1-containing ternary complex and reveal a positive role for Ldb1 in erythroid gene expression and differentiation.


Journal of Clinical Investigation | 1989

Constitutive production of macrophage colony-stimulating factor by human ovarian and breast cancer cell lines.

S. Ramakrishnan; Fengji Xu; Stephen J. Brandt; James E. Niedel; Robert C. Bast; E. L. Brown

Many nonhematologic tumors produce growth factors that may influence cellular proliferation either by autocrine or by paracrine mechanisms. In the current study, human tumor cell lines were investigated for the constitutive production of macrophage colony-stimulating factor (M-CSF). Culture supernatants obtained from cell lines were analyzed using a radioimmunoassay and a radioreceptor assay specific for M-CSF. Among the various cell types analyzed, all the ovarian cell lines and a majority of the breast cancer cell lines secreted significant amount of an M-CSF-like factor. Treatment of mouse bone marrow cultures with culture supernatants from ovarian cancer cells stimulated the production of macrophage colonies. Analysis of total cellular RNA obtained from the ovarian cell lines by Northern blot showed multiple sizes of M-CSF transcripts with an abundance of a 4.2-kb message. The relative amount of M-CSF transcripts correlated with the level of immunoreactive material seen in the culture supernatants.


Circulation | 1999

Retroviral Gene Therapy in ApoE-Deficient Mice ApoE Expression in the Artery Wall Reduces Early Foam Cell Lesion Formation

Alyssa H. Hasty; MacRae F. Linton; Stephen J. Brandt; Vladimir R. Babaev; Linda A. Gleaves; Sergio Fazio

BACKGROUND Apolipoprotein E (apoE) has long been known to play an important role in the clearance of plasma lipoproteins. More recently, a direct role for apoE in delaying atherogenesis has been proposed. Macrophage production of apoE in the artery wall has been demonstrated to provide protection against atherosclerotic lesion development independently from its role in lipoprotein clearance. However, whether macrophage apoE can affect lesion growth at all stages of atherogenesis remains to be established. METHODS AND RESULTS To evaluate the role of macrophage apoE in different stages of atherogenesis, as well as to establish a novel gene therapy approach to atherosclerotic vascular disease, we used an apoE-expressing retrovirus to transduce apoE-deficient (-/-) bone marrow for transplantation into apoE(-/-) recipient mice. Three weeks after bone marrow transplantation, apoE was expressed from arterial macrophages and was detectable in plasma associated with lipoproteins at 0.5% to 1% of normal levels but did not affect plasma cholesterol levels. We used 2 groups of recipient mice: younger mice with lesions consisting primarily of foam cells and older mice with more advanced lesions. When either the mouse or human apoE transgenes were expressed in mice from 5 to 13 weeks of age, there was a significant reduction in lesion area, whereas no effects were detected in mice that expressed apoE from 10 to 26 weeks of age. CONCLUSIONS We demonstrate that arterial macrophage apoE secretion can delay atherogenesis if expressed during foam cell formation but is not beneficial during the later stages of atherogenesis. These data also provide evidence that apoE transgene expression from arterial macrophages may have therapeutic applications.


Molecular and Cellular Biology | 2000

mSin3A Regulates Murine Erythroleukemia Cell Differentiation through Association with the TAL1 (or SCL) Transcription Factor

Suming Huang; Stephen J. Brandt

ABSTRACT Activation of the TAL1 (or SCL) gene is the most frequent gain-of-function mutation in T-cell acute lymphoblastic leukemia (T-ALL). TAL1 belongs to the basic helix-loop-helix (HLH) family of transcription factors that bind as heterodimers with theE2A and HEB/HTF4 gene products to a nucleotide sequence motif termed the E-box. Reported to act both as an activator and as a repressor of transcription, the mechanisms underlying TAL1-regulated gene expression are poorly understood. We report here that the corepressor mSin3A is associated with TAL1 in murine erythroleukemia (MEL) and human T-ALL cells. Interaction mapping showed that the basic-HLH domain of TAL1 was both necessary and sufficient for TAL1-mSin3A interaction. TAL1 was found, in addition, to interact with the histone deacetylase HDAC1 in vitro and in vivo, and a specific histone deacetylase inhibitor, trichostatin A (TSA), relieved TAL1-mediated repression of an E-box-containing promoter and a GAL4 reporter linked to a thymidine kinase minimal promoter. Further, TAL1 association with mSin3A and HDAC1 declined during dimethyl sulfoxide-induced differentiation of MEL cells in parallel with a decrease in mSin3A abundance. Finally, TSA had a synergistic effect with enforced TAL1 expression in stimulating MEL cells to differentiate, while constitutive expression of mSin3A inhibited MEL cell differentiation. These results demonstrate that a corepressor complex containing mSin3A and HDAC1 interacts with TAL1 and restricts its function in erythroid differentiation. This also has implications for this transcription factors actions in leukemogenesis.


The EMBO Journal | 2000

P/CAF-mediated acetylation regulates the function of the basic helix-loop-helix transcription factor TAL1/SCL.

Suming Huang; Yi Qiu; Yubin Shi; Zhixiong Xu; Stephen J. Brandt

The basic helix–loop–helix transcription factor TAL1 (or SCL) is a critical regulator of hematopoietic and vascular development and is misexpressed in the majority of patients with T‐cell acute lymphoblastic leukemia. We found previously that TAL1 could interact with transcriptional co‐activator and co‐repressor complexes possessing histone acetyltransferase and deacetylase activities, respectively. Here, we report that TAL1 is subject to acetylation in vivo and can be acetylated by p300 and the p300/CBP‐associated factor P/CAF in vitro. P/CAF‐mediated acetylation, which mapped to a lysine‐rich motif in the loop region, increased TAL1 binding to DNA while selectively inhibiting its interaction with the transcriptional co‐repressor mSin3A. Furthermore, P/CAF protein, TAL1–P/CAF interaction and TAL1 acetylation increased significantly in murine erythroleukemia cells induced to differentiate in culture, while enforced expression of an acetylation‐defective P/CAF mutant inhibited endogenous TAL1 acetylation, TAL1 DNA‐binding activity, TAL1‐directed transcription and terminal differentiation of these cells. These results reveal a novel mechanism by which TAL1 activity is regulated and implicate acetylation of this transcription factor in promotion of erythroid differentiation.


Clinical Cancer Research | 2008

Histone deacetylase inhibitor romidepsin has differential activity in core binding factor acute myeloid leukemia.

Olatoyosi Odenike; Serhan Alkan; Dorie Sher; John E. Godwin; Dezheng Huo; Stephen J. Brandt; Margaret Green; Jingping Xie; Yanming Zhang; David H. Vesole; Patrick J. Stiff; John J. Wright; Richard A. Larson; Wendy Stock

Purpose: Recruitment of histone deacetylases (HDAC) is a mechanism of transcriptional repression implicated in the differentiation block in acute myeloid leukemia (AML). We hypothesized that the HDAC inhibitor romidepsin could cause transcriptional derepression, up-regulation of specific target genes in AML, and differentiation of the leukemic clone. The primary objectives of the study were to evaluate the safety and efficacy of romidepsin in advanced AML. Experimental Design: Twenty patients were stratified into cohort A or B based on the absence or presence of chromosomal abnormalities known to recruit HDACs, including those involving core binding factor (CBF). Romidepsin was administered i.v. at 13 mg/m2/d on days 1, 8, and 15 of a 28-day cycle. Pharmacodynamic endpoints were evaluated at serial time points. Results: Common adverse effects noted were grade 1 to 2 nausea, anorexia, and fatigue. No objective evidence of antileukemic activity was seen in cohort A. In cohort B, although there were no clinical responses by standard criteria, antileukemic activity was observed in 5 of 7 patients. Two patients had clearance of bone marrow blasts and 3 patients had a >50% decrease in bone marrow blasts. Furthermore, in cohort B, at 24 h, there was a significant increase in MDR1 (P = 0.005), p15 (P = 0.01), and p14 (P < 0.0001) expression. In cohort A, although there was a trend toward up-regulation of MDR1, p15, and p14 expression, these changes were not statistically significant. Conclusion: Romidepsin has differential antileukemic and molecular activity in CBF AML. Development of this agent in CBF AML should focus on combinations that target related mechanisms of gene silencing such as DNA methylation.


Oncogene | 2007

Histone deacetylase inhibitors induce the degradation of the t(8;21) fusion oncoprotein

Gong Yang; Mary Ann Thompson; Stephen J. Brandt; Scott W. Hiebert

The t(8;21) chromosomal translocation that generates the fusion oncoprotein RUNX1-ETO predominates in leukemia patients of the French-American-British (FAB) class M2 subtype. The oncoprotein has the capacity to promote expansion of hematopoietic stem/progenitor cells and induces leukemia in association with other genetic alterations. Here, we show that RUNX1-ETO undergoes degradation in response to treatment with histone deacetylase inhibitors, one of which, depsipeptide (DEP), is currently undergoing phase II clinical testing in a variety of malignancies. These compounds induce turnover of RUNX1-ETO without affecting the stability of RUNX1-ETO partner proteins. In addition, RUNX1-ETO physically interacts with heat shock protein 90 (HSP90). DEP treatment interrupts the association of RUNX1-ETO with HSP90 and induces proteasomal degradation of RUNX1-ETO. DEP and the HSP90 antagonist 17-allylamino-geldanamycin (17-AAG) both triggered RUNX1-ETO degradation, but without any additive or cooperative effects. These findings may stimulate the development of more rational and effective approaches for treating t(8;21) patients using histone deacetylase inhibitors or HSP90 inhibitors.

Collaboration


Dive into the Stephen J. Brandt's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

David Morgan

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar

John P. Greer

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ying Cai

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Lalitha Nagarajan

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Zhixiong Xu

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Madan Jagasia

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Wichai Chinratanalab

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Adetola A. Kassim

Vanderbilt University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge