Network


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

Hotspot


Dive into the research topics where Anwarul Ferdous is active.

Publication


Featured researches published by Anwarul Ferdous.


Molecular Cell | 2001

The 19S regulatory particle of the proteasome is required for efficient transcription elongation by RNA polymerase II

Anwarul Ferdous; Fernando Gonzalez; Liping Sun; Thomas Kodadek; Stephen Albert Johnston

It is generally thought that the primary or even sole activity of the 19S regulatory particle of the 26S proteasome is to facilitate the degradation of polyubiquitinated proteins by the 20S-core subunit. However, we present evidence that the 19S complex is required for efficient elongation of RNA polymerase II (RNAP II) in vitro and in vivo. First, yeast strains carrying alleles of SUG1 and SUG2, encoding 19S components, exhibit phenotypes indicative of elongation defects. Second, in vitro transcription is inhibited by antibodies raised against Sug1, or by heat-inactivating temperature-sensitive Sug1 mutants with restoration of elongation by addition of immunopurified 19S complex. Finally, Cdc68, a known elongation factor, coimmunoprecipitates with the 19S complex, indicating a physical interaction. Inhibition of the 20S proteolytic core of the proteasome has no effect on elongation. This work defines a nonproteolytic role for the 19S complex in RNAP II transcription.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Nkx2-5 transactivates the Ets-related protein 71 gene and specifies an endothelial/endocardial fate in the developing embryo.

Anwarul Ferdous; Arianna Caprioli; Michelina Iacovino; Cindy M. Martin; Jesse Morris; James A. Richardson; Shuaib Latif; Robert E. Hammer; Richard P. Harvey; Eric N. Olson; Michael Kyba; Daniel J. Garry

Recent studies support the existence of a common progenitor for the cardiac and endothelial cell lineages, but the underlying transcriptional networks responsible for specification of these cell fates remain unclear. Here we demonstrated that Ets-related protein 71 (Etsrp71), a newly discovered ETS family transcription factor, was a novel downstream target of the homeodomain protein, Nkx2–5. Using genetic mouse models and molecular biological techniques, we demonstrated that Nkx2–5 binds to an evolutionarily conserved Nkx2–5 response element in the Etsrp71 promoter and induces the Etsrp71 gene expression in vitro and in vivo. Etsrp71 was transiently expressed in the endocardium/endothelium of the developing embryo (E7.75-E9.5) and was extinguished during the latter stages of development. Using a gene disruption strategy, we found that Etsrp71 mutant embryos lacked endocardial/endothelial lineages and were nonviable. Moreover, using transgenic technologies and transcriptional and chromatin immunoprecipitation (ChIP) assays, we further established that Tie2 is a direct downstream target of Etsrp71. Collectively, our results uncover a novel functional role for Nkx2–5 and define a transcriptional network that specifies an endocardial/endothelial fate in the developing heart and embryo.


Cell | 2014

Spliced X-box binding protein 1 couples the unfolded protein response to hexosamine biosynthetic pathway

Zhao V. Wang; Yingfeng Deng; Ningguo Gao; Zully Pedrozo; Dan L. Li; Cyndi R. Morales; Alfredo Criollo; Xiang Luo; Wei Tan; Nan Jiang; Mark A. Lehrman; Beverly A. Rothermel; Ann Hwee Lee; Sergio Lavandero; Pradeep P.A. Mammen; Anwarul Ferdous; Thomas G. Gillette; Philipp E. Scherer; Joseph A. Hill

The hexosamine biosynthetic pathway (HBP) generates uridine diphosphate N-acetylglucosamine (UDP-GlcNAc) for glycan synthesis and O-linked GlcNAc (O-GlcNAc) protein modifications. Despite the established role of the HBP in metabolism and multiple diseases, regulation of the HBP remains largely undefined. Here, we show that spliced X-box binding protein 1 (Xbp1s), the most conserved signal transducer of the unfolded protein response (UPR), is a direct transcriptional activator of the HBP. We demonstrate that the UPR triggers HBP activation via Xbp1s-dependent transcription of genes coding for key, rate-limiting enzymes. We further establish that this previously unrecognized UPR-HBP axis is triggered in a variety of stress conditions. Finally, we demonstrate a physiologic role for the UPR-HBP axis by showing that acute stimulation of Xbp1s in heart by ischemia/reperfusion confers robust cardioprotection in part through induction of the HBP. Collectively, these studies reveal that Xbp1s couples the UPR to the HBP to protect cells under stress.


Circulation Research | 2008

Hypoxia-Inducible Factor-2α Transactivates Abcg2 and Promotes Cytoprotection in Cardiac Side Population Cells

Cindy M. Martin; Anwarul Ferdous; Teresa D. Gallardo; Caroline G. Humphries; Hesham A. Sadek; Arianna Caprioli; Joseph A. Garcia; Luke I. Szweda; Mary G. Garry; Daniel J. Garry

Stem and progenitor cell populations occupy a specialized niche and are consequently exposed to hypoxic as well as oxidative stresses. We have previously established that the multidrug resistance protein Abcg2 is the molecular determinant of the side population (SP) progenitor cell population. We observed that the cardiac SP cells increase in number more than 3-fold within 3 days of injury. Transcriptome analysis of the SP cells isolated from the injured adult murine heart reveals increased expression of cytoprotective transcripts. Overexpression of Abcg2 results in an increased ability to consume hydrogen peroxide and is associated with increased levels of &agr;-glutathione reductase protein expression. Importantly, overexpression of Abcg2 also conferred a cell survival benefit following exposure to hydrogen peroxide. To further examine the molecular regulation of the Abcg2 gene, we demonstrated that hypoxia-inducible factor (HIF)-2&agr; binds an evolutionary conserved HIF-2&agr; response element in the murine Abcg2 promoter. Transcriptional assays reveal a dose-dependent activation of Abcg2 expression by HIF-2&agr;. These results support the hypothesis that Abcg2 is a direct downstream target of HIF-2&agr; which functions with other factors to initiate a cytoprotective program for this progenitor SP cell population that resides in the adult heart.


Journal of Cardiovascular Translational Research | 2010

FoxO, Autophagy, and Cardiac Remodeling

Anwarul Ferdous; Pavan K. Battiprolu; Yan G. Ni; Beverly A. Rothermel; Joseph A. Hill

In response to changes in workload, the heart grows or shrinks. Indeed, the myocardium is capable of robust and rapid structural remodeling. In the setting of normal, physiological demand, the heart responds with hypertrophic growth of individual cardiac myocytes, a process that serves to maintain cardiac output and minimize wall stress. However, disease-related stresses, such as hypertension or myocardial infarction, provoke a series of changes that culminate in heart failure and/or sudden death. At the other end of the spectrum, cardiac unloading, such as occurs with prolonged bed rest or weightlessness, causes the heart to shrink. In recent years, considerable strides have been made in deciphering the molecular and cellular events governing pro- and anti-growth events in the heart. Prominent among these mechanisms are those mediated by FoxO (Forkhead box-containing protein, O subfamily) transcription factors. In many cell types, these proteins are critical regulators of cell size, viability, and metabolism, and their importance in the heart is just emerging. Also in recent years, evidence has emerged for a pivotal role for autophagy, an evolutionarily conserved pathway of lysosomal degradation of damaged proteins and organelles, in cardiac growth and remodeling. Indeed, evidence for activated autophagy has been detected in virtually every form of myocardial disease. Now, it is clear that FoxO is an upstream regulator of both autophagy and the ubiquitin-proteasome system. Here, we discuss recent advances in our understanding of cardiomyocyte autophagy, its governance by FoxO, and the roles each of these plays in cardiac remodeling.


Journal of Clinical Investigation | 2013

The Xbp1s/GalE axis links ER stress to postprandial hepatic metabolism

Yingfeng Deng; Zhao V. Wang; Caroline Tao; Ningguo Gao; William L. Holland; Anwarul Ferdous; Joyce J. Repa; Guosheng Liang; Jin Ye; Mark A. Lehrman; Joseph A. Hill; Jay D. Horton; Philipp E. Scherer

Postprandially, the liver experiences an extensive metabolic reprogramming that is required for the switch from glucose production to glucose assimilation. Upon refeeding, the unfolded protein response (UPR) is rapidly, though only transiently, activated. Activation of the UPR results in a cessation of protein translation, increased chaperone expression, and increased ER-mediated protein degradation, but it is not clear how the UPR is involved in the postprandial switch to alternate fuel sources. Activation of the inositol-requiring enzyme 1 (IRE1) branch of the UPR signaling pathway triggers expression of the transcription factor Xbp1s. Using a mouse model with liver-specific inducible Xbp1s expression, we demonstrate that Xbp1s is sufficient to provoke a metabolic switch characteristic of the postprandial state, even in the absence of caloric influx. Mechanistically, we identified UDP-galactose-4-epimerase (GalE) as a direct transcriptional target of Xbp1s and as the key mediator of this effect. Our results provide evidence that the Xbp1s/GalE pathway functions as a novel regulatory nexus connecting the UPR to the characteristic postprandial metabolic changes in hepatocytes.


The FASEB Journal | 2004

Targeted gene delivery to sinusoidal endothelial cells: DNA nanoassociate bearing hyaluronan-glycocalyx

Yoshiyuki Takei; Atsushi Maruyama; Anwarul Ferdous; Yoshiya Nishimura; Sunao Kawano; Kenichi Ikejima; Shigetoshi Okumura; Shoichiro Asayama; Masayuki Nogawa; Masao Hashimoto; Yoko Makino; Masahiko Kinoshita; Sumio Watanabe; Toshihiro Akaike; John J. Lemasters; Nobuhiro Sato

Liver sinusoidal endothelial cells (SECs) possess unique receptors that recognize and internalize hyaluronic acid (HA). To develop a system for targeting foreign DNA to SECs, comb‐type polycations having HA side chains were prepared by coupling HA to poly(L‐lysine) (PLL). The HA‐grafted‐PLL copolymer (PLL‐g‐HA) thus formed was mixed with DNA in 154 mM NaCl to form soluble nanoassociates bearing hydrated hyaluronate shells. Agarose gel retardation assays revealed selective interaction of the PLL backbone with DNA despite the presence of polyanionic HA side chains. To determine whether the PLL‐g‐HA/DNA complexes were recognized by SEC HA receptors in vivo, we injected Wistar rats i.v. via the tail vein with PLL‐ g‐HA complexed to a β‐galactosidase expression plasmid (pSV β‐Gal) labeled with 32P. One hour postinjection, >90% of the injected radioactivity remained in the liver. Administration of the PLL‐g‐HA complexed to an FITC‐labeled DNA revealed that the carrier‐DNA complex was distributed exclusively in SECs. A large number of SECs expressing β‐galactosidase was detected along the sinusoidal lining after transfection with PLL‐g‐HA/pSV β‐Gal. Moreover, PLL‐g‐HA effectively stabilized DNA triplex formation. In conclusion, the new PLL‐g‐ HA/DNA carrier system permits targeted transfer of exogenous genes selectively to the SECs.


Development | 2011

ER71 directs mesodermal fate decisions during embryogenesis

Tara L. Rasmussen; Junghun Kweon; Mackenzie A. Diekmann; Fikru Belema-Bedada; Qingfeng Song; Kathy Bowlin; Xiaozhong Shi; Anwarul Ferdous; Tongbin Li; Michael Kyba; Joseph M. Metzger; Naoko Koyano-Nakagawa; Daniel J. Garry

Er71 mutant embryos are nonviable and lack hematopoietic and endothelial lineages. To further define the functional role for ER71 in cell lineage decisions, we generated genetically modified mouse models. We engineered an Er71-EYFP transgenic mouse model by fusing the 3.9 kb Er71 promoter to the EYFP reporter gene. Using FACS and transcriptional profiling, we examined the EYFP+ population of cells in Er71 mutant and wild-type littermates. In the absence of ER71, we observed an increase in the number of EYFP-expressing cells, increased expression of the cardiac molecular program and decreased expression of the hemato-endothelial program, as compared with wild-type littermate controls. We also generated a novel Er71-Cre transgenic mouse model using the same 3.9 kb Er71 promoter. Genetic fate-mapping studies revealed that the ER71-expressing cells give rise to the hematopoietic and endothelial lineages in the wild-type background. In the absence of ER71, these cell populations contributed to alternative mesodermal lineages, including the cardiac lineage. To extend these analyses, we used an inducible embryonic stem/embryoid body system and observed that ER71 overexpression repressed cardiogenesis. Together, these studies identify ER71 as a critical regulator of mesodermal fate decisions that acts to specify the hematopoietic and endothelial lineages at the expense of cardiac lineages. This enhances our understanding of the mechanisms that govern mesodermal fate decisions early during embryogenesis.


Journal of Biological Chemistry | 2008

Physical and Functional Interactions of Monoubiquitylated Transactivators with the Proteasome

Chase T. Archer; Lyle Burdine; Bo Liu; Anwarul Ferdous; Stephen Albert Johnston; Thomas Kodadek

Destabilization of activator-DNA complexes by the proteasomal ATPases can inhibit transcription by limiting activator interaction with DNA. Modification of the activator by monoubiquitylation protects the activator from this destabilization activity. In this study, we probe the mechanism of this protective effect of monoubiquitylation. Using novel label transfer and chemical cross-linking techniques, we show that ubiquitin contacts the ATPase complex directly, apparently via Rpn1 and Rpt1. This interaction results in the dissociation of the activation domain-ATPase complex via an allosteric process. A model is proposed in which activator monoubiquitylation serves to limit the lifetime of the activator-ATPase complex interaction and thus the ability of the ATPases to unfold the activator and dissociate the protein-DNA complex.


Journal of Biological Chemistry | 1999

Poly(L-lysine)-graft-dextran copolymer promotes pyrimidine motif triplex DNA formation at physiological pH. Thermodynamic and kinetic studies.

Hidetaka Torigoe; Anwarul Ferdous; Hiromitsu Watanabe; Toshihiro Akaike; Atsushi Maruyama

Extreme instability of pyrimidine motif triplex DNA at physiological pH severely limits its use for artificial control of gene expression in vivo. Stabilization of the pyrimidine motif triplex at physiological pH is therefore of great importance in improving its therapeutic potential. To this end, isothermal titration calorimetry interaction analysis system and electrophoretic mobility shift assay have been used to explore the thermodynamic and kinetic effects of our previously reported triplex stabilizer, poly (l-lysine)-graft-dextran (PLL-g-Dex) copolymer, on pyrimidine motif triplex formation at physiological pH. Both the thermodynamic and kinetic analyses have clearly indicated that in the presence of the PLL-g-Dex copolymer, the binding constant of the pyrimidine motif triplex formation at physiological pH was about 100 times higher than that observed without any triplex stabilizer. Of importance, the triplex-promoting efficiency of the copolymer was more than 20 times higher than that of physiological concentrations of spermine, a putative intracellular triplex stabilizer. Kinetic data have also demonstrated that the observed copolymer-mediated promotion of the triplex formation at physiological pH resulted from the considerable increase in the association rate constant rather than the decrease in the dissociation rate constant. Our results certainly support the idea that the PLL-g-Dex copolymer could be a key material and may eventually lead to progress in therapeutic applications of the antigene strategy in vivo.

Collaboration


Dive into the Anwarul Ferdous's collaboration.

Top Co-Authors

Avatar

Toshihiro Akaike

Tokyo Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Atsushi Maruyama

Tokyo Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Joseph A. Hill

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Hiromitsu Watanabe

Tokyo Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Michael Kyba

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar

Arianna Caprioli

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Thomas G. Gillette

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Eric N. Olson

University of Texas Southwestern Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge