Network


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

Hotspot


Dive into the research topics where Jeffrey W. Streb is active.

Publication


Featured researches published by Jeffrey W. Streb.


Circulation Research | 2004

Thioredoxin-2 Inhibits Mitochondria-Located ASK1-Mediated Apoptosis in a JNK-Independent Manner

Rong Zhang; Rafia S. Al-Lamki; Lanfang Bai; Jeffrey W. Streb; Joseph M. Miano; John R. Bradley; Wang Min

Apoptosis signal-regulating kinase 1 (ASK1) mediates cytokines and oxidative stress (ROS)–induced apoptosis in a mitochondria-dependent pathway. However, the underlying mechanism has not been defined. In this study, we show that ASK1 is localized in both cytoplasm and mitochondria of endothelial cells (ECs) where it binds to cytosolic (Trx1) and mitochondrial thioredoxin (Trx2), respectively. Cys-250 and Cys-30 in the N-terminal domain of ASK1 are critical for binding of Trx1 and Trx2, respectively. Mutation of ASK1 at C250 enhanced ASK1-induced JNK activation and apoptosis, whereas mutation of ASK1 at C30 specifically increased ASK1-induced apoptosis without effects on JNK activation. We further show that a JNK-specific inhibitor SP600125 completely blocks TNF induced JNK activation, Bid cleavage, and Bax mitochondrial translocation, but only partially inhibits cytochrome c release and EC death, suggesting that TNF induces both JNK-dependent and JNK-independent apoptotic pathways in EC. Mitochondria-specific expression of a constitutively active ASK1 strongly induces EC apoptosis without JNK activation, Bid cleavage, and Bax mitochondrial translocation. These data suggest that mitochondrial ASK1 mediates a JNK-independent apoptotic pathway induced by TNF. To determine the role of Trx2 in regulation of mitochondrial ASK1 activity, we show that overexpression of Trx2 inhibits ASK1-induced apoptosis without effects on ASK1-induced JNK activation. Moreover, specific knockdown of Trx2 in EC increases TNF/ASK1-induced cytochrome c release and cell death without increase in JNK activation, Bid cleavage, and Bax translocation. Our data suggest that ASK1 in cytoplasm and mitochondria mediate distinct apoptotic pathways induced by TNF, and Trx1 and Trx2 cooperatively inhibit ASK1 activities.


Nature Cell Biology | 2009

SRF and myocardin regulate LRP-mediated amyloid-beta clearance in brain vascular cells.

Robert D. Bell; Rashid Deane; Nienwen Chow; Xiaochun Long; Abhay P. Sagare; Itender Singh; Jeffrey W. Streb; Huang Guo; Anna Rubio; William E. Van Nostrand; Joseph M. Miano; Berislav V. Zlokovic

Amyloid β-peptide (Aβ) deposition in cerebral vessels contributes to cerebral amyloid angiopathy (CAA) in Alzheimers disease (AD). Here, we report that in AD patients and two mouse models of AD, overexpression of serum response factor (SRF) and myocardin (MYOCD) in cerebral vascular smooth muscle cells (VSMCs) generates an Aβ non-clearing VSMC phenotype through transactivation of sterol regulatory element binding protein-2, which downregulates low density lipoprotein receptor-related protein-1, a key Aβ clearance receptor. Hypoxia stimulated SRF/MYOCD expression in human cerebral VSMCs and in animal models of AD. We suggest that SRF and MYOCD function as a transcriptional switch, controlling Aβ cerebrovascular clearance and progression of AD.


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

Serum response factor and myocardin mediate arterial hypercontractility and cerebral blood flow dysregulation in Alzheimer's phenotype

Nienwen Chow; Robert D. Bell; Rashid Deane; Jeffrey W. Streb; Jiyuan Chen; Andrew I. Brooks; William E. Van Nostrand; Joseph M. Miano; Berislav V. Zlokovic

Cerebral angiopathy contributes to cognitive decline and dementia in Alzheimers disease (AD) through cerebral blood flow (CBF) reductions and dysregulation. We report vascular smooth muscle cells (VSMC) in small pial and intracerebral arteries, which are critical for CBF regulation, express in AD high levels of serum response factor (SRF) and myocardin (MYOCD), two interacting transcription factors that orchestrate a VSMC-differentiated phenotype. Consistent with this finding, AD VSMC overexpressed several SRF-MYOCD-regulated contractile proteins and exhibited a hypercontractile phenotype. MYOCD overexpression in control human cerebral VSMC induced an AD-like hypercontractile phenotype and diminished both endothelial-dependent and -independent relaxation in the mouse aorta ex vivo. In contrast, silencing SRF normalized contractile protein content and reversed a hypercontractile phenotype in AD VSMC. MYOCD in vivo gene transfer to mouse pial arteries increased contractile protein content and diminished CBF responses produced by brain activation in wild-type mice and in two AD models, the Dutch/Iowa/Swedish triple mutant human amyloid β-peptide (Aβ)-precursor protein (APP)- expressing mice and APPsw+/− mice. Silencing Srf had the opposite effect. Expression of SRF did not change in VSMC subjected to Alzheimers neurotoxin, Aβ. Thus, SRF-MYOCD overexpression in small cerebral arteries appears to initiate independently of Aβ a pathogenic pathway mediating arterial hypercontractility and CBF dysregulation, which are associated with Alzheimers dementia.


Journal of Biological Chemistry | 2004

Multiple promoters direct expression of three AKAP12 isoforms with distinct subcellular and tissue distribution profiles.

Jeffrey W. Streb; Chad M. Kitchen; Irwin H. Gelman; Joseph M. Miano

A Kinase Anchoring Protein 12 (AKAP12; also known as src-suppressed C kinase substrate (SSeCKS) and Gravin) is a multivalent anchoring protein with tumor suppressor activity. Although expression of AKAP12 has been examined in a number of contexts, its expression control remains to be elucidated. Herein, we characterize the genomic organization of the AKAP12 locus, its regulatory regions, and the spatial distribution of the proteins encoded by the AKAP12 gene. Using comparative genomics and various wet-lab assays, we show that the AKAP12 locus is organized as three separate transcription units that are governed by non-redundant promoters coordinating distinct tissue expression profiles. The proteins encoded by the three AKAP12 isoforms (designated α, β, and γ) share >95% amino acid sequence identity but differ at their N termini. Analysis of the targeting of each isoform reveals distinct spatial distribution profiles. An N-terminal myristoylation motif present in AKAP12α is shown to be necessary and sufficient for targeted expression of this AKAP12 isoform to the endoplasmic reticulum, a novel subcellular compartment for AKAP12. Our results demonstrate heretofore unrecognized complexity within the AKAP12 locus and suggest a mechanism for genetic control of signaling specificity through distinct regulation of alternately targeted anchoring protein isoforms.


Current Drug Targets - Cardiovascular & Hematological Disorders | 2003

Retinoids: pleiotropic agents of therapy for vascular diseases?

Jeffrey W. Streb; Joseph M. Miano

Retinoids, the natural and synthetic derivatives of vitamin A, exert broad biological effects and have been used clinically to treat a variety of dermatological and neoplastic diseases. The principal mode of action of many retinoids is through the binding and activation of a family of nuclear receptors that modulate gene transcription. Recent evidence demonstrates that retinoids effectively attenuate experimental vessel wall narrowing due to atherosclerosis, post-balloon injury stenosis, and bypass graft failure. Moreover, retinoids promote a differentiated phenotype in smooth muscle cells (SMC) which, unlike other muscle types, is not fixed and is subject to considerable modulation in disease states. A growing number of in vitro studies have reported desirable effects of retinoids on cell migration, proliferation, apoptosis, matrix remodeling, fibrinolysis, coagulation, and inflammation, all of which impinge on vascular disease. Since vascular SMC and endothelial cells (EC) express most retinoid receptors, the mechanisms underlying retinoid-mediated events in these cells and the vessel wall likely relate to an altered transcriptome. In fact, there is a growing list of retinoid-response genes encoding proteins that likely mediate the actions of retinoids. Retinoid-response genes, therefore, represent promising targets of therapy for the refined treatment of vascular diseases. The purpose of this review is to summarize the emerging importance of retinoids in the control of vascular cell responses with special emphasis on potential mechanisms underlying retinoid-induced changes in the vessel wall following injury. Given the similarities in the pathogenesis of neoplasia and vascular disease, it is reasonable to consider testing the efficacy of retinoids for the treatment of human vascular disease.


Journal of Biological Chemistry | 2005

Cross-species sequence analysis reveals multiple charged residue-rich domains that regulate nuclear/cytoplasmic partitioning and membrane localization of A kinase anchoring protein 12 (SSeCKS/Gravin)

Jeffrey W. Streb; Joseph M. Miano

A kinase anchoring proteins (AKAPs) assemble and compartmentalize multiprotein signaling complexes at discrete subcellular locales and thus confer specificity to transduction cascades using ubiquitous signaling enzymes, such as protein kinase A. Intrinsic targeting domains in each AKAP determine the subcellular localization of these complexes and, along with protein-protein interaction domains, form the core of AKAP function. As a foundational step toward elucidating the relationship between location and function, we have used cross-species sequence analysis and deletion mapping to facilitate the identification of the targeting determinants of AKAP12 (also known as SSeCKS or Gravin). Three charged residue-rich regions were identified that regulate two aspects of AKAP12 localization, nuclear/cytoplasmic partitioning and perinuclear/cell periphery targeting. Using deletion mapping and green fluorescent protein chimeras, we uncovered a heretofore unrecognized nuclear localization potential. Five nuclear localization signals, including a novel class of this type of signal termed X2-NLS, are found in the central region of AKAP12 and are important for nuclear targeting. However, this nuclear localization is suppressed by the negatively charged C terminus that mediates nuclear exclusion. In this condition, the distribution of AKAP12 is regulated by an N-terminal targeting domain that simultaneously directs perinuclear and peripheral AKAP12 localization. Three basic residue-rich regions in the N-terminal targeting region have similarity to the MARCKS proteins and were found to control AKAP12 localization to ganglioside-rich regions at the cell periphery. Our data suggest that AKAP12 localization is regulated by a hierarchy of targeting domains and that the localization of AKAP12-assembled signaling complexes may be dynamically regulated.


Journal of Histochemistry and Cytochemistry | 2006

Tissue Expression of the Novel Serine Carboxypeptidase Scpep1

Ting-Hein D. Lee; Jeffrey W. Streb; Mary A. Georger; Joseph M. Miano

We previously identified a novel gene designated retinoid-inducible serine carboxypeptidase (RISC or Scpep1). Here we characterize a polyclonal antibody raised to Scpep1 and assess its localization in mouse cells and tissues. Western blot analysis revealed an immunospecific ∼35-kDa protein corresponding to endogenous Scpep1. This protein is smaller than the predicted ∼51-kDa, suggesting that Scpep1 is proteolytically cleaved to a mature enzyme. Immunohistochemical studies demonstrate Scpep1 expression in embryonic heart and vasculature as well as in adult aortic smooth muscle cells and endothelial cells. Scpep1 displays a broad expression pattern in adult tissues with detectable levels in epithelia of digestive tract and urinary bladder, islet of Langerhans, type II alveolar cells and macrophages of lung, macrophage-like cells of lymph nodes and spleen, Leydig cells of testis, and nerve fibers in brain and ganglia. Consistent with previous mRNA studies in kidney, Scpep1 protein is restricted to proximal convoluted tubular epithelium (PCT). Immunoelectron microscopy shows enriched Scpep1 within lysosomes of the PCT, and immunofluorescence microscopy colocalizes Scpep1 with lysosomal-associated membrane protein-2. These results suggest that Scpep1 is a widely distributed lysosomal protease requiring proteolytic cleavage for activity. The highly specific Scpep1 antibody characterized herein provides a necessary reagent for elucidating Scpep1 function.


PLOS ONE | 2011

Retinoid-Induced Expression and Activity of an Immediate Early Tumor Suppressor Gene in Vascular Smooth Muscle Cells

Jeffrey W. Streb; Xiaochun Long; Ting Hein Lee; Qiang Sun; Chad M. Kitchen; Mary A. Georger; Orazio J. Slivano; William S. Blaner; Daniel W. Carr; Irwin H. Gelman; Joseph M. Miano

Retinoids are used clinically to treat a number of hyper-proliferative disorders and have been shown in experimental animals to attenuate vascular occlusive diseases, presumably through nuclear receptors bound to retinoic acid response elements (RARE) located in target genes. Here, we show that natural or synthetic retinoids rapidly induce mRNA and protein expression of a specific isoform of A-Kinase Anchoring Protein 12 (AKAP12β) in cultured smooth muscle cells (SMC) as well as the intact vessel wall. Expression kinetics and actinomycin D studies indicate Akap12β is a retinoid-induced, immediate-early gene. Akap12β promoter analyses reveal a conserved RARE mildly induced with atRA in a region that exhibits hyper-acetylation. Immunofluorescence microscopy and protein kinase A (PKA) regulatory subunit overlay assays in SMC suggest a physical association between AKAP12β and PKA following retinoid treatment. Consistent with its designation as a tumor suppressor, inducible expression of AKAP12β attenuates SMC growth in vitro. Further, immunohistochemistry studies establish marked decreases in AKAP12 expression in experimentally-injured vessels of mice as well as atheromatous lesions in humans. Collectively, these results demonstrate a novel role for retinoids in the induction of an AKAP tumor suppressor that blocks vascular SMC growth thus providing new molecular insight into how retiniods may exert their anti-proliferative effects in the injured vessel wall.


Journal of Molecular and Cellular Cardiology | 2002

Myocardin: A Component of a Molecular Switch for Smooth Muscle Differentiation

Jiyuan Chen; Chad M. Kitchen; Jeffrey W. Streb; Joseph M. Miano


Genome Research | 2005

Defining the mammalian CArGome

Qiang Sun; Guang Chen; Jeffrey W. Streb; Xiaochun Long; Yumei Yang; Christian J. Stoeckert; Joseph M. Miano

Collaboration


Dive into the Jeffrey W. Streb's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jiyuan Chen

University of Rochester Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Berislav V. Zlokovic

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Irwin H. Gelman

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nienwen Chow

University of Rochester

View shared research outputs
Top Co-Authors

Avatar

Qiang Sun

University of Rochester

View shared research outputs
Top Co-Authors

Avatar

Rashid Deane

University of Rochester

View shared research outputs
Researchain Logo
Decentralizing Knowledge