Network


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

Hotspot


Dive into the research topics where Alexander H. Stegh is active.

Publication


Featured researches published by Alexander H. Stegh.


Nature | 2011

SMAD4-dependent barrier constrains prostate cancer growth and metastatic progression

Zhihu Ding; Chang Jiun Wu; Gerald C. Chu; Yonghong Xiao; Jingfang Zhang; Samuel R. Perry; Emma S. Labrot; Xiaoqiu Wu; Rosina T. Lis; Yujin Hoshida; David Hiller; Baoli Hu; Shan Jiang; Hongwu Zheng; Alexander H. Stegh; Kenneth L. Scott; Sabina Signoretti; Nabeel Bardeesy; Y. Alan Wang; David E. Hill; Todd R. Golub; Meir J. Stampfer; Wing Hung Wong; Massimo Loda; Lorelei A. Mucci; Lynda Chin; Ronald A. DePinho

Effective clinical management of prostate cancer (PCA) has been challenged by significant intratumoural heterogeneity on the genomic and pathological levels and limited understanding of the genetic elements governing disease progression. Here, we exploited the experimental merits of the mouse to test the hypothesis that pathways constraining progression might be activated in indolent Pten-null mouse prostate tumours and that inactivation of such progression barriers in mice would engender a metastasis-prone condition. Comparative transcriptomic and canonical pathway analyses, followed by biochemical confirmation, of normal prostate epithelium versus poorly progressive Pten-null prostate cancers revealed robust activation of the TGFβ/BMP–SMAD4 signalling axis. The functional relevance of SMAD4 was further supported by emergence of invasive, metastatic and lethal prostate cancers with 100% penetrance upon genetic deletion of Smad4 in the Pten-null mouse prostate. Pathological and molecular analysis as well as transcriptomic knowledge-based pathway profiling of emerging tumours identified cell proliferation and invasion as two cardinal tumour biological features in the metastatic Smad4/Pten-null PCA model. Follow-on pathological and functional assessment confirmed cyclin D1 and SPP1 as key mediators of these biological processes, which together with PTEN and SMAD4, form a four-gene signature that is prognostic of prostate-specific antigen (PSA) biochemical recurrence and lethal metastasis in human PCA. This model-informed progression analysis, together with genetic, functional and translational studies, establishes SMAD4 as a key regulator of PCA progression in mice and humans.


Molecular and Cellular Biology | 2000

Identification of the Cytolinker Plectin as a Major Early In Vivo Substrate for Caspase 8 during CD95- and Tumor Necrosis Factor Receptor-Mediated Apoptosis

Alexander H. Stegh; Harald Herrmann; Stefan Lampel; Dieter Weisenberger; Kerstin Andrä; Martin Seper; Gerhard Wiche; Peter H. Krammer; Marcus E. Peter

ABSTRACT Caspase 8 plays an essential role in the execution of death receptor-mediated apoptosis. To determine the localization of endogenous caspase 8, we used a panel of subunit-specific anti-caspase 8 monoclonal antibodies in confocal immunofluorescence microscopy. In the human breast carcinoma cell line MCF7, caspase 8 predominantly colocalized with and bound to mitochondria. After induction of apoptosis through CD95 or tumor necrosis factor receptor I, active caspase 8 translocated to plectin, a major cross-linking protein of the three main cytoplasmic filament systems, whereas the caspase 8 prodomain remained bound to mitochondria. Plectin was quantitatively cleaved by caspase 8 at Asp 2395 in the center of the molecule in all cells tested. Cleavage of plectin clearly preceded that of other caspase substrates such as poly(ADP-ribose) polymerase, gelsolin, cytokeratins, or lamin B. In primary fibroblasts from plectin-deficient mice, apoptosis-induced reorganization of the actin cytoskeleton, as seen in wild-type cells, was severely impaired, suggesting that during apoptosis, plectin is required for the reorganization of the microfilament system.


The EMBO Journal | 1998

DEDD, a novel death effector domain‐containing protein, targeted to the nucleolus

Alexander H. Stegh; Olaf Schickling; Andreas Ehret; Carsten Scaffidi; Christoph Peterhänsel; Thomas G. Hofmann; Ingrid Grummt; Peter H. Krammer; Marcus E. Peter

The CD95 signaling pathway comprises proteins that contain one or two death effector domains (DED), such as FADD/Mort1 or caspase‐8. Here we describe a novel 37 kDa protein, DEDD, that contains an N‐terminal DED. DEDD is highly conserved between human and mouse (98.7% identity) and is ubiquitously expressed. Overexpression of DEDD in 293T cells induced weak apoptosis, mainly through its DED by which it interacts with FADD and caspase‐8. Endogenous DEDD was found in the cytoplasm and translocated into the nucleus upon stimulation of CD95. Immunocytological studies revealed that overexpressed DEDD directly translocated into the nucleus, where it co‐localizes in the nucleolus with UBF, a basal factor required for RNA polymerase I transcription. Consistent with its nuclear localization, DEDD contains two nuclear localization signals and the C‐terminal part shares sequence homology with histones. Recombinant DEDD binds to both DNA and reconstituted mononucleosomes and inhibits transcription in a reconstituted in vitro system. The results suggest that DEDD is a final target of a chain of events by which the CD95‐induced apoptotic signal is transferred into the nucleolus to shut off cellular biosynthetic activities.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Bcl2L12-mediated inhibition of effector caspase-3 and caspase-7 via distinct mechanisms in glioblastoma

Alexander H. Stegh; Santosh Kesari; John E. Mahoney; Harry T. Jenq; Kristin L. Forloney; Alexei Protopopov; David N. Louis; Lynda Chin; Ronald A. DePinho

Glioblastoma multiforme (GBM) is a highly aggressive brain cancer that is characterized by the paradoxical features of intense apoptosis resistance yet a marked propensity to undergo necrosis. Bcl2L12 (for Bcl2-Like12) is a nuclear and cytoplasmic oncoprotein that is universally overexpressed in primary GBM and functions to block postmitochondrial apoptosis signaling by neutralizing effector caspase-3 and caspase-7 maturation. This postmitochondrial block in apoptosis engenders the alternate cell fate of cellular necrosis, thus providing a molecular explanation for GBMs classical features. Whereas Bcl2L12-mediated neutralization of caspase-7 maturation involves physical interaction, the mechanism governing Bcl2L12-mediated inhibition of caspase-3 activity is not known. The nuclear localization of Bcl2L12 prompted expression profile studies of primary astrocytes engineered to overexpress Bcl2L12. The Bcl2L12 transcriptome revealed a striking induction of the small heat shock protein α-basic-crystallin (αB-crystallin/HspB5), a link reinforced by robust αB-crystallin expression in Bcl2L12-expressing orthotopic glioma and strong coexpression of αB-crystallin and Bcl2L12 proteins in human primary GBMs. On the functional level, enforced αB-crystallin or Bcl2L12 expression enhances orthotopic tumor growth. Conversely, RNAi-mediated knockdown of αB-crystallin in Bcl2L12-expressing astrocytes and glioma cell lines with high endogenous αB-crystallin showed enhanced apoptosis, yet decreased necrotic cell death with associated increased caspase-3 but not caspase-7 activation. Mirroring this specific effect on effector caspase-3 activation, αB-crystallin selectively binds pro-caspase-3 and its cleavage intermediates in vitro and in vivo. Thus, αB-crystallin is a Bcl2L12-induced oncoprotein that enables Bcl2L12 to block the activation of both effector caspases via distinct mechanisms, thereby contributing to GBM pathogenesis and its hallmark biological properties.


Expert Opinion on Therapeutic Targets | 2012

Targeting the p53 signaling pathway in cancer therapy – the promises, challenges and perils

Alexander H. Stegh

Introduction: Research over the past three decades has identified p53 as a multi-functional transcription factor. p53 influences myriad, highly diverse cellular processes, and represents one of the most important and extensively studied tumor suppressors. Activated by various stresses, p53 blocks cancer progression by provoking transient or permanent growth arrest, by enabling DNA repair, or by advancing cellular death programs. This anti-cancer activity profile, together with genomic and mutational analyses documenting inactivation of p53 in more than 50% of human cancers, motivated drug development efforts to (re-) activate p53 in established tumors. Areas covered: The complexities of p53 signaling in cancer are summarized, including current strategies and challenges to restore p53s tumor suppressive function in established tumors, to inactivate p53 inhibitors, and to restore wild type function of p53 mutant proteins. Expert opinion: p53 represents an attractive target for the development of anti-cancer therapies. Whether p53 is ‘druggable’, however, remains an area of active research and discussion, as p53 has pro-survival functions and chronic p53 activation accelerates aging, which may compromise the long-term homeostasis of an organism. The complex biology and dual functions of p53 in cancer prevention and age-related cellular responses pose significant challenges to the development of p53-targeting cancer therapies.


Genes & Development | 2015

miR-182 integrates apoptosis, growth, and differentiation programs in glioblastoma

Fotini M. Kouri; Lisa A. Hurley; Weston L. Daniel; Emily S. Day; Youjia Hua; Liangliang Hao; Chian Yu Peng; Timothy J. Merkel; Markus A. Queisser; Carissa Ritner; Hailei Zhang; C. David James; Jacob I. Sznajder; Lynda Chin; David A. Giljohann; John A. Kessler; Marcus E. Peter; Chad A. Mirkin; Alexander H. Stegh

Glioblastoma multiforme (GBM) is a lethal, therapy-resistant brain cancer consisting of numerous tumor cell subpopulations, including stem-like glioma-initiating cells (GICs), which contribute to tumor recurrence following initial response to therapy. Here, we identified miR-182 as a regulator of apoptosis, growth, and differentiation programs whose expression level is correlated with GBM patient survival. Repression of Bcl2-like12 (Bcl2L12), c-Met, and hypoxia-inducible factor 2α (HIF2A) is of central importance to miR-182 anti-tumor activity, as it results in enhanced therapy susceptibility, decreased GIC sphere size, expansion, and stemness in vitro. To evaluate the tumor-suppressive function of miR-182 in vivo, we synthesized miR-182-based spherical nucleic acids (182-SNAs); i.e., gold nanoparticles covalently functionalized with mature miR-182 duplexes. Intravenously administered 182-SNAs penetrated the blood-brain/blood-tumor barriers (BBB/BTB) in orthotopic GBM xenografts and selectively disseminated throughout extravascular glioma parenchyma, causing reduced tumor burden and increased animal survival. Our results indicate that harnessing the anti-tumor activities of miR-182 via safe and robust delivery of 182-SNAs represents a novel strategy for therapeutic intervention in GBM.


Journal of Cell Biology | 2002

DEDD regulates degradation of intermediate filaments during apoptosis.

Justine C. Lee; Olaf Schickling; Alexander H. Stegh; Robert G. Oshima; David Dinsdale; Gerald M. Cohen; Marcus E. Peter

Apoptosis depends critically on regulated cytoskeletal reorganization events in a cell. We demonstrate that death effector domain containing DNA binding protein (DEDD), a highly conserved and ubiquitous death effector domain containing protein, exists predominantly as mono- or diubiquitinated, and that diubiquitinated DEDD interacts with both the K8/18 intermediate filament network and pro–caspase-3. Early in apoptosis, both cytosolic DEDD and its close homologue DEDD2 formed filaments that colocalized with and depended on K8/18 and active caspase-3. Subsequently, these filamentous structures collapsed into intracellular inclusions that migrated into cytoplasmic blebs and contained DEDD, DEDD2, active caspase-3, and caspase-3–cleaved K18 late in apoptosis. Biochemical studies further confirmed that DEDD coimmunoprecipitated with both K18 and pro–caspase-3, and kinetic analyses placed apoptotic DEDD staining prior to caspase-3 activation and K18 cleavage. In addition, both caspase-3 activation and K18 cleavage was inhibited by expression of DEDDΔNLS1-3, a cytosolic form of DEDD that cannot be ubiquitinated. Finally, siRNA mediated DEDD knockdown cells exhibited inhibition of staurosporine-induced DNA degradation. Our data suggest that DEDD represents a novel scaffold protein that directs the effector caspase-3 to certain substrates facilitating their ordered degradation during apoptosis.


Genes & Development | 2010

Glioma oncoprotein Bcl2L12 inhibits the p53 tumor suppressor

Alexander H. Stegh; Cameron Brennan; John A. Mahoney; Kristin L. Forloney; Harry T. Jenq; Janina P. Luciano; Alexei Protopopov; Lynda Chin; Ronald A. DePinho

Glioblastoma multiforme (GBM) is a lethal brain tumor characterized by intense apoptosis resistance and extensive necrosis. Bcl2L12 (for Bcl2-like 12) is a cytoplasmic and nuclear protein that is overexpressed in primary GBM and functions to inhibit post-mitochondrial apoptosis signaling. Here, we show that nuclear Bcl2L12 physically and functionally interacts with the p53 tumor suppressor, as evidenced by the capacity of Bcl2L12 to (1) enable bypass of replicative senescence without concomitant loss of p53 or p19 (Arf), (2) inhibit p53-dependent DNA damage-induced apoptosis, (3) impede the capacity of p53 to bind some of its target gene promoters, and (4) attenuate endogenous p53-directed transcriptomic changes following genotoxic stress. Correspondingly, The Cancer Genome Atlas profile and tissue protein analyses of human GBM specimens show significantly lower Bcl2L12 expression in the setting of genetic p53 pathway inactivation. Thus, Bcl2L12 is a multifunctional protein that contributes to intense therapeutic resistance of GBM through its ability to operate on two key nodes of cytoplasmic and nuclear signaling cascades.


Cell Death & Differentiation | 2001

Nuclear localization of DEDD leads to caspase-6 activation through its death effector domain and inhibition of RNA polymerase I dependent transcription.

Olaf Schickling; Alexander H. Stegh; J Byrd; Marcus E. Peter

The death effector domain (DED) is a protein/protein interaction domain only found in proteins that are involved in apoptosis signaling. DEDD is a novel apoptosis signaling molecule that carries an N-terminal DED with complete sequence identity between the murine, rat, bovine and human domains. We previously identified two nuclear localization signals (NLS) responsible for DEDDs nuclear localization when transiently expressed. Using a new anti-DEDD antibody that allows us to stain endogenous DEDD in immunofluorescence microscopy we now detect a significant amount of DEDD in nucleoli of all cells tested. When overexpressed, DEDD localizes to nucleoli-like structures, activates caspase-6 and specifically inhibits RNA polymerase I (Pol I) dependent transcription in vivo as shown by blockage of BrUTP incorporation. The DED in DEDD is sufficient for its DNA binding, caspase-6 activating and Pol I specific transcriptional repressor activity. We have identified a third NLS in DEDD and only mutation of all three NLS generated a protein, DEDDΔNLS1-3, that mainly localized to the cytoplasm. This protein no longer induced apoptosis, indicating that in contrast to other DED proteins, such as FADD, caspase-8 or c-FLIP, DEDD induces apoptosis from within the nucleus. This effect is abolished when specific point mutations are made within the DED. The DED in DEDD therefore represents a novel domain that is structurally similar to other DEDs but functionally different from classical DEDs found in FADD or caspase-8. Cell Death and Differentiation (2001) 8, 1157–1168


Cardiology Clinics | 2001

APOPTOSIS AND CASPASES

Alexander H. Stegh; Marcus E. Peter

The expedition into the apoptosis signaling pathway, although it has just begun, has resulted in the discovery of a significant number of remarkable signaling molecules at all levels of this novel pathway After the pinnacle of this frenetic cloning effort has been reached, however, it is important to put this pathway and its constituents into a biological and pathophysiological context. It has become clear that cell death does not automatically mean activation of caspases. The recent discovery of a function of effector caspases of the apoptosis pathway outside of apoptosis is currently revolutionizing our view of these seemingly unrelated and rather counteracting processes, cell death and cell proliferation. It appears that caspases play a much more fundamental role in cells than originally expected.

Collaboration


Dive into the Alexander H. Stegh's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Samuel A. Jensen

Children's Memorial Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cameron Brennan

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge