Network


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

Hotspot


Dive into the research topics where Abena B. Redwood is active.

Publication


Featured researches published by Abena B. Redwood.


The EMBO Journal | 2009

Novel roles for A-type lamins in telomere biology and the DNA damage response pathway

Ignacio Gonzalez-Suarez; Abena B. Redwood; Stephanie M. Perkins; Bart Vermolen; Daniel Lichtensztejin; David A. Grotsky; Lucia Morgado-Palacin; Eric J. Gapud; Barry P. Sleckman; Teresa Sullivan; Julien Sage; Colin L. Stewart; Sabine Mai; Susana Gonzalo

A‐type lamins are intermediate filament proteins that provide a scaffold for protein complexes regulating nuclear structure and function. Mutations in the LMNA gene are linked to a variety of degenerative disorders termed laminopathies, whereas changes in the expression of lamins are associated with tumourigenesis. The molecular pathways affected by alterations of A‐type lamins and how they contribute to disease are poorly understood. Here, we show that A‐type lamins have a key role in the maintenance of telomere structure, length and function, and in the stabilization of 53BP1, a component of the DNA damage response (DDR) pathway. Loss of A‐type lamins alters the nuclear distribution of telomeres and results in telomere shortening, defects in telomeric heterochromatin, and increased genomic instability. In addition, A‐type lamins are necessary for the processing of dysfunctional telomeres by non‐homologous end joining, putatively through stabilization of 53BP1. This study shows new functions for A‐type lamins in the maintenance of genomic integrity, and suggests that alterations of telomere biology and defects in DDR contribute to the pathogenesis of lamin‐related diseases.


Nature Communications | 2015

Loss of lamin A function increases chromatin dynamics in the nuclear interior

Irena Bronshtein; Eldad Kepten; Itamar Kanter; Shirly Berezin; Moshe Lindner; Abena B. Redwood; Sabine Mai; Susana Gonzalo; Roland Foisner; Yaron Shav-Tal; Yuval Garini

Chromatin is organized in a highly ordered yet dynamic manner in the cell nucleus, but the principles governing this organization remain unclear. Similarly, it is unknown whether, and how, various proteins regulate chromatin motion and as a result influence nuclear organization. Here by studying the dynamics of different genomic regions in the nucleus of live cells, we show that the genome has highly constrained dynamics. Interestingly, depletion of lamin A strikingly alters genome dynamics, inducing a dramatic transition from slow anomalous diffusion to fast and normal diffusion. In contrast, depletion of LAP2α, a protein that interacts with lamin A and chromatin, has no such effect on genome dynamics. We speculate that chromosomal inter-chain interactions formed by lamin A throughout the nucleus contribute to chromatin dynamics, and suggest that the molecular regulation of chromatin diffusion by lamin A in the nuclear interior is critical for the maintenance of genome organization.


The EMBO Journal | 2011

A new pathway that regulates 53BP1 stability implicates Cathepsin L and vitamin D in DNA repair

Ignacio Gonzalez-Suarez; Abena B. Redwood; David A. Grotsky; Martin Neumann; Emily H. Cheng; Colin L. Stewart; Adriana Dusso; Susana Gonzalo

Genomic instability due to telomere dysfunction and defective repair of DNA double‐strand breaks (DSBs) is an underlying cause of ageing‐related diseases. 53BP1 is a key factor in DNA DSBs repair and its deficiency is associated with genomic instability and cancer progression. Here, we uncover a novel pathway regulating the stability of 53BP1. We demonstrate an unprecedented role for the cysteine protease Cathepsin L (CTSL) in the degradation of 53BP1. Overexpression of CTSL in wild‐type fibroblasts leads to decreased 53BP1 protein levels and changes in its cellular distribution, resulting in defective repair of DNA DSBs. Importantly, we show that the defects in DNA repair associated with 53BP1 deficiency upon loss of A‐type lamins are due to upregulation of CTSL. Furthermore, we demonstrate that treatment with vitamin D stabilizes 53BP1 and promotes DNA DSBs repair via inhibition of CTSL, providing an as yet unsuspected link between vitamin D action and DNA repair. Given that CTSL upregulation is a hallmark of cancer and progeria, regulation of this pathway could be of great therapeutic significance for these diseases.


Cell Cycle | 2011

A dual role for A-type lamins in DNA double-strand break repair

Abena B. Redwood; Stephanie M. Perkins; Robert P. Vanderwaal; Zhihui Feng; Kenneth J. Biehl; Ignacio Gonzalez-Suarez; Lucia Morgado-Palacin; Wei Shi; Julien Sage; Joseph Roti-Roti; Colin L. Stewart; Junran Zhang; Susana Gonzalo

A-type lamins are emerging as regulators of nuclear organization and function. Changes in their expression are associated with cancer and mutations are linked to degenerative diseases -laminopathies-. Although a correlation exists between alterations in lamins and genomic instability, the molecular mechanisms remain largely unknown. We previously found that loss of A-type lamins leads to degradation of 53BP1 protein and defective long-range non-homologous end-joining (NHEJ) of dysfunctional telomeres. Here, we determined how loss of A-type lamins affects the repair of short-range DNA double-strand breaks (DSBs) induced by ionizing radiation (IR). We find that lamins deficiency allows activation of the DNA damage response, but compromises the accumulation of 53BP1 at IR-induced foci (IRIF), hindering the fast phase of repair corresponding to classical-NHEJ. Importantly, reconstitution of 53BP1 is sufficient to rescue long-range and short-range NHEJ. Moreover, we demonstrate an unprecedented role for A-type lamins in the maintenance of homologous recombination (HR). Depletion of lamins compromises HR by a mechanism involving transcriptional downregulation of BRCA1 and RAD51 by the repressor complex formed by the Rb family member p130 and E2F4. In line with the DNA repair defects, lamins-deficient cells exhibit increased radiosensitivity. This study demonstrates that A-type lamins promote genomic stability by maintaining the levels of proteins with key roles in DNA DSBs repair by NHEJ and HR. Our results suggest that silencing of A-type lamins by DNA methylation in some cancers could contribute to the genomic instability that drives malignancy. In addition, lamins-deficient tumor cells could represent a good target for radiation therapy.


Journal of Cell Biology | 2013

BRCA1 loss activates cathepsin L–mediated degradation of 53BP1 in breast cancer cells

David A. Grotsky; Ignacio Gonzalez-Suarez; Anna Novell; Martin Neumann; Sree C. Yaddanapudi; Monica Croke; Montserrat Martínez-Alonso; Abena B. Redwood; Sylvia Ortega-Martinez; Zhihui Feng; Enrique Lerma; Teresa Ramón y Cajal; Junran Zhang; Xavier Matias-Guiu; Adriana Dusso; Susana Gonzalo

Cathepsin L degrades 53BP1 to overcome genomic instability and growth arrest in BRCA1-deficient and triple-negative breast cancers.


Cell Cycle | 2009

Loss of A-type lamins and genomic instability

Ignacio Gonzalez-Suarez; Abena B. Redwood; Susana Gonzalo

Research performed in the last few years has revealed important roles for the spatial and temporal organization of the genome on genome function and integrity. A challenge in the field is to determine the molecular mechanisms involved in the organization of genome function. A-type lamins, key structural components of the nucleus, have been implicated in the maintenance of nuclear architecture and chromatin structure. Interestingly, alterations of A-type lamins lead to defects in DNA replication and repair as well as gene transcription and silencing. Elucidating the functions of these proteins is a topical subject since alterations of A-type lamins are associated with a variety of human diseases, ranging from muscular dystrophies and premature aging syndromes to cancer. Here, we discuss novels roles for A-type lamins in the maintenance of telomere structure, length and function as well as in the stabilization of a key DNA damage response factor. These studies support the notion that increased genomic instability due to defects in telomere biology and DNA repair contribute to the pathogenesis of lamin-related diseases.


Cell Cycle | 2011

Regulating the levels of key factors in cell cycle and DNA repair: New pathways revealed by lamins

Abena B. Redwood; Ignacio Gonzalez-Suarez; Susana Gonzalo

Spatial and temporal organization of the genome represents an additional step in the regulation of nuclear functions. The nuclear lamina, a polymeric meshwork formed by lamins (A/C and B type) and lamin-associated proteins, plays a key role in the maintenance of genome localization, structure and function. Specifically, mutations in the LMNA gene encoding lamins A/C or changes in its expression, either upregulation or silencing, are associated with defects in DNA replication, transcription and repair, as well as alterations in epigenetic modifications of chromatin. These data, together with the fact that defects in A-type lamins are associated with a whole variety of degenerative disorders, premature aging syndromes and cancer, support the notion that these proteins operate as caretakers of the genome. However, our understanding of their functions is limited due to the lack of well-defined mechanisms behind the genomic instability observed in lamin-related diseases. Here, we summarize our recent discovery of new pathways that are affected by the loss of A-type lamins. In particular, we found that A-type lamins control transcription and degradation of proteins with key roles in cell cycle regulation and DNA double-strand breaks (DSBs) repair by nonhomologous end-joining (NHEJ) and homologous-recombination (HR). Importantly, the proteins regulated by A-type lamins—Rb family members, 53BP1, BRCA1 and RAD51— exert tumor suppressor functions, with their loss being associated with cancer susceptibility. Moreover, our studies revealed novel pathways that contribute to genomic instability and that can be activated in disease states independent of the status of A-type lamins.


Nucleus | 2013

Lamin A Δexon9 mutation leads to telomere and chromatin defects but not genomic instability

Arindam Das; David A. Grotsky; Martin Neumann; Ray Kreienkamp; Ignacio Gonzalez-Suarez; Abena B. Redwood; Brian K. Kennedy; Colin L. Stewart; Susana Gonzalo

Over 300 mutations in the LMNA gene, encoding A-type lamins, are associated with 15 human degenerative disorders and premature aging syndromes. Although genomic instability seems to contribute to the pathophysiology of some laminopathies, there is limited information about what mutations cause genomic instability and by which molecular mechanisms. Mouse embryonic fibroblasts depleted of A-type lamins or expressing mutants lacking exons 8–11 (LmnaΔ8–11/Δ8–11) exhibit alterations in telomere biology and DNA repair caused by cathepsin L-mediated degradation of 53BP1 and reduced expression of BRCA1 and RAD51. Thus, a region encompassing exons 8–11 seems essential for genome integrity. Given that deletion of lamin A exon 9 in the mouse (LmnaΔ9/Δ9) results in a progeria phenotype, we tested if this domain is important for genome integrity. LmnaΔ9/Δ9 MEFs exhibit telomere shortening and heterochromatin alterations but do not activate cathepsin L-mediated degradation of 53BP1 and maintain expression of BRCA1 and RAD51. Accordingly, LmnaΔ9/Δ9 MEFs do not present genomic instability, and expression of mutant lamin A Δexon9 in lamin-depleted cells restores DNA repair factors levels and partially rescues nuclear abnormalities. These data reveal that the domain encoded by exon 9 is important to maintain telomere homeostasis and heterochromatin structure but does not play a role in DNA repair, thus pointing to other exons in the lamin A tail as responsible for the genomic instability phenotype in LmnaΔ8–11/Δ8–11 mice. Our study also suggests that the levels of DNA repair factors 53BP1, BRCA1 and RAD51 could potentially serve as biomarkers to identify laminopathies that present with genomic instability.


Cancer Research | 2017

CHK1 inhibition in small cell lung cancer produces single-agent activity in biomarker-defined disease subsets and combination activity with cisplatin or olaparib

Triparna Sen; Pan Tong; C. Allison Stewart; Sandra Cristea; Aly Valliani; David S. Shames; Abena B. Redwood; You Hong Fan; Lerong Li; Bonnie S. Glisson; John D. Minna; Julien Sage; Don L. Gibbons; Helen Piwnica-Worms; John V. Heymach; Jing Wang; Lauren Averett Byers

Effective targeted therapies for small-cell lung cancer (SCLC), the most aggressive form of lung cancer, remain urgently needed. Here we report evidence of preclinical efficacy evoked by targeting the overexpressed cell-cycle checkpoint kinase CHK1 in SCLC. Our studies employed RNAi-mediated attenuation or pharmacologic blockade with the novel second-generation CHK1 inhibitor prexasertib (LY2606368), currently in clinical trials. In SCLC models in vitro and in vivo, LY2606368 exhibited strong single-agent efficacy, augmented the effects of cisplatin or the PARP inhibitor olaparib, and improved the response of platinum-resistant models. Proteomic analysis identified CHK1 and MYC as top predictive biomarkers of LY2606368 sensitivity, suggesting that CHK1 inhibition may be especially effective in SCLC with MYC amplification or MYC protein overexpression. Our findings provide a preclinical proof of concept supporting the initiation of a clinical efficacy trial in patients with platinum-sensitive or platinum-resistant relapsed SCLC. Cancer Res; 77(14); 3870-84. ©2017 AACR.


Cancer Research | 2017

CDKN2A/p16 deletion in head and neck cancer cells is associated with cdk2 activation, replication stress, and vulnerability to CHK1 inhibition

Mayur A. Gadhikar; Jiexin Zhang; Li Shen; Xiayu Rao; Jing Wang; Mei Zhao; Nene N. Kalu; Faye M. Johnson; Lauren Averett Byers; John V. Heymach; Walter N. Hittelman; Durga Udayakumar; Raj K. Pandita; Tej K. Pandita; Curtis R. Pickering; Abena B. Redwood; Helen Piwnica-Worms; Katharina Schlacher; Mitchell J. Frederick; Jeffrey N. Myers

Checkpoint kinase inhibitors (CHKi) exhibit striking single-agent activity in certain tumors, but the mechanisms accounting for hypersensitivity are poorly understood. We screened a panel of 49 established human head and neck squamous cell carcinoma (HNSCC) cell lines and report that nearly 20% are hypersensitive to CHKi monotherapy. Hypersensitive cells underwent early S-phase arrest at drug doses sufficient to inhibit greater than 90% of CHK1 activity. Reduced rate of DNA replication fork progression and chromosomal shattering were also observed, suggesting replication stress as a root causative factor in CHKi hypersensitivity. To explore genomic underpinnings of CHKi hypersensitivity, comparative genomic analysis was performed between hypersensitive cells and cells categorized as least sensitive because they showed drug IC50 value greater than the cell panel median and lacked early S-phase arrest. Novel association between CDKN2A/p16 copy number loss, CDK2 activation, replication stress, and hypersensitivity of HNSCC cells to CHKi monotherapy was found. Restoring p16 in cell lines harboring CDKN2A/p16 genomic deletions alleviated CDK2 activation and replication stress, attenuating CHKi hypersensitivity. Taken together, our results suggest a biomarker-driven strategy for selecting HNSCC patients who may benefit the most from CHKi therapy.Significance: These results suggest a biomarker-driven strategy for selecting HNSCC patients who may benefit the most from therapy with CHK inhibitors. Cancer Res; 78(3); 781-97. ©2017 AACR.

Collaboration


Dive into the Abena B. Redwood's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ignacio Gonzalez-Suarez

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Helen Piwnica-Worms

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Colin L. Stewart

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

David A. Grotsky

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Adriana Dusso

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Jing Wang

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

John V. Heymach

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge