Network


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

Hotspot


Dive into the research topics where Abdul M. Mondal is active.

Publication


Featured researches published by Abdul M. Mondal.


Nature Cell Biology | 2009

p53 isoforms Δ133p53 and p53β are endogenous regulators of replicative cellular senescence

Kaori Fujita; Abdul M. Mondal; Izumi Horikawa; Giang Hong Nguyen; Kensuke Kumamoto; Jane J. Sohn; Elise D. Bowman; Ewy Mathe; Aaron J. Schetter; Sharon R. Pine; Helen Ji; Borivoj Vojtesek; Jean-Christophe Bourdon; David P. Lane; Curtis C. Harris

The finite proliferative potential of normal human cells leads to replicative cellular senescence, which is a critical barrier to tumour progression in vivo. We show that the human p53 isoforms Δ133p53 and p53β function in an endogenous regulatory mechanism for p53-mediated replicative senescence. Induced p53β and diminished Δ133p53 were associated with replicative senescence, but not oncogene-induced senescence, in normal human fibroblasts. The replicatively senescent fibroblasts also expressed increased levels of miR-34a, a p53-induced microRNA, the antisense inhibition of which delayed the onset of replicative senescence. The siRNA (short interfering RNA)-mediated knockdown of endogenous Δ133p53 induced cellular senescence, which was attributed to the regulation of p21WAF1 and other p53 transcriptional target genes. In overexpression experiments, whereas p53β cooperated with full-length p53 to accelerate cellular senescence, Δ133p53 repressed miR-34a expression and extended the cellular replicative lifespan, providing a functional connection of this microRNA to the p53 isoform-mediated regulation of senescence. The senescence-associated signature of p53 isoform expression (that is, elevated p53β and reduced Δ133p53) was observed in vivo in colon adenomas with senescent phenotypes. The increased Δ133p53 and decreased p53β isoform expression found in colon carcinoma may signal an escape from the senescence barrier during the progression from adenoma to carcinoma.


Nature Cell Biology | 2010

Positive feedback between p53 and TRF2 during telomere-damage signalling and cellular senescence

Kaori Fujita; Izumi Horikawa; Abdul M. Mondal; Lisa M. Miller Jenkins; Ettore Appella; Borivoj Vojtesek; Jean-Christophe Bourdon; David P. Lane; Curtis C. Harris

The telomere-capping complex shelterin protects functional telomeres and prevents the initiation of unwanted DNA-damage-response pathways. At the end of cellular replicative lifespan, uncapped telomeres lose this protective mechanism and DNA-damage signalling pathways are triggered that activate p53 and thereby induce replicative senescence. Here, we identify a signalling pathway involving p53, Siah1 (a p53-inducible E3 ubiquitin ligase) and TRF2 (telomere repeat binding factor 2; a component of the shelterin complex). Endogenous Siah1 and TRF2 were upregulated and downregulated, respectively, during replicative senescence with activated p53. Experimental manipulation of p53 expression demonstrated that p53 induces Siah1 and represses TRF2 protein levels. The p53-dependent ubiquitylation and proteasomal degradation of TRF2 are attributed to the E3 ligase activity of Siah1. Knockdown of Siah1 stabilized TRF2 and delayed the onset of cellular replicative senescence, suggesting a role for Siah1 and TRF2 in p53-regulated senescence. This study reveals that p53, a downstream effector of telomere-initiated damage signalling, also functions upstream of the shelterin complex.


Oncogene | 2013

Downregulation of splicing factor SRSF3 induces p53β, an alternatively spliced isoform of p53 that promotes cellular senescence.

Tang Y; Izumi Horikawa; Masahiko Ajiro; Ana I. Robles; Kaori Fujita; Abdul M. Mondal; Stauffer Jk; Zhi-Ming Zheng; Curtis C. Harris

Most human pre-mRNA transcripts are alternatively spliced, but the significance and fine-tuning of alternative splicing in different biological processes is only starting to be understood. SRSF3 (SRp20) is a member of a highly conserved family of splicing factors that have critical roles in key biological processes, including tumor progression. Here, we show that SRSF3 regulates cellular senescence, a p53-mediated process to suppress tumorigenesis, through TP53 alternative splicing. Downregulation of SRSF3 was observed in normal human fibroblasts undergoing replicative senescence, and was associated with the upregulation of p53β, an alternatively spliced isoform of p53 that promotes p53-mediated senescence. Knockdown of SRSF3 by short interfering RNA (siRNA) in early-passage fibroblasts induced senescence, which was associated with elevated expression of p53β at mRNA and protein levels. Knockdown of p53 partially rescued SRSF3-knockdown-induced senescence, suggesting that SRSF3 acts on p53-mediated cellular senescence. RNA pulldown assays demonstrated that SRSF3 binds to an alternatively spliced exon uniquely included in p53β mRNA through the consensus SRSF3-binding sequences. RNA crosslinking and immunoprecipitation assays (CLIP) also showed that SRSF3 in vivo binds to endogenous p53 pre-mRNA at the region containing the p53β-unique exon. Splicing assays using a transfected TP53 minigene in combination with siRNA knockdown of SRSF3 showed that SRSF3 functions to inhibit the inclusion of the p53β-unique exon in splicing of p53 pre-mRNA. These data suggest that downregulation of SRSF3 represents an endogenous mechanism for cellular senescence that directly regulates the TP53 alternative splicing to generate p53β. This study uncovers the role for general splicing machinery in tumorigenesis, and suggests that SRSF3 is a direct regulator of p53.


Journal of Clinical Investigation | 2013

p53 isoforms regulate aging- and tumor-associated replicative senescence in T lymphocytes

Abdul M. Mondal; Izumi Horikawa; Sharon R. Pine; Kaori Fujita; Katherine M. Morgan; Elsa Vera; Sharlyn J. Mazur; Ettore Appella; Borivoj Vojtesek; Maria A. Blasco; David P. Lane; Curtis C. Harris

Cellular senescence contributes to aging and decline in tissue function. p53 isoform switching regulates replicative senescence in cultured fibroblasts and is associated with tumor progression. Here, we found that the endogenous p53 isoforms Δ133p53 and p53β are physiological regulators of proliferation and senescence in human T lymphocytes in vivo. Peripheral blood CD8+ T lymphocytes collected from healthy donors displayed an age-dependent accumulation of senescent cells (CD28-CD57+) with decreased Δ133p53 and increased p53β expression. Human lung tumor-associated CD8+ T lymphocytes also harbored senescent cells. Cultured CD8+ blood T lymphocytes underwent replicative senescence that was associated with loss of CD28 and Δ133p53 protein. In poorly proliferative, Δ133p53-low CD8+CD28- cells, reconstituted expression of either Δ133p53 or CD28 upregulated endogenous expression of each other, which restored cell proliferation, extended replicative lifespan and rescued senescence phenotypes. Conversely, Δ133p53 knockdown or p53β overexpression in CD8+CD28+ cells inhibited cell proliferation and induced senescence. This study establishes a role for Δ133p53 and p53β in regulation of cellular proliferation and senescence in vivo. Furthermore, Δ133p53-induced restoration of cellular replicative potential may lead to a new therapeutic paradigm for treating immunosenescence disorders, including those associated with aging, cancer, autoimmune diseases, and HIV infection.


Nature Communications | 2014

Autophagic degradation of the inhibitory p53 isoform Δ133p53α as a regulatory mechanism for p53-mediated senescence

Izumi Horikawa; Kaori Fujita; Lisa M. Miller Jenkins; Yukiharu Hiyoshi; Abdul M. Mondal; Borivoj Vojtesek; David P. Lane; Ettore Appella; Curtis C. Harris

Δ133p53α, a p53 isoform that can inhibit full-length p53, is downregulated at replicative senescence in a manner independent of mRNA regulation and proteasome-mediated degradation. Here we demonstrate that, unlike full-length p53, Δ133p53α is degraded by autophagy during replicative senescence. Pharmacological inhibition of autophagy restores Δ133p53α expression levels in replicatively senescent fibroblasts, without affecting full-length p53. The siRNA-mediated knockdown of pro-autophagic proteins (ATG5, ATG7 and Beclin-1) also restores Δ133p53α expression. The chaperone-associated E3 ubiquitin ligase STUB1, which is known to regulate autophagy, interacts with Δ133p53α and is downregulated at replicative senescence. The siRNA knockdown of STUB1 in proliferating, early-passage fibroblasts induces the autophagic degradation of Δ133p53α and thereby induces senescence. Upon replicative senescence or STUB1 knockdown, Δ133p53α is recruited to autophagosomes, consistent with its autophagic degradation. This study reveals that STUB1 is an endogenous regulator of Δ133p53α degradation and senescence, and identifies a p53 isoform-specific protein turnover mechanism that orchestrates p53-mediated senescence.


Cell Death & Differentiation | 2017

|[Delta]|133p53 represses p53-inducible senescence genes and enhances the generation of human induced pluripotent stem cells

Izumi Horikawa; Kye-yoon Park; Kazunobu Isogaya; Yukiharu Hiyoshi; Han Li; Katsuhiro Anami; Ana I. Robles; Abdul M. Mondal; Kaori Fujita; Manuel Serrano; Curtis C. Harris

p53 functions to induce cellular senescence, which is incompatible with self-renewal of pluripotent stem cells such as induced pluripotent stem cells (iPSC) and embryonic stem cells (ESC). However, p53 also has essential roles in these cells through DNA damage repair for maintaining genomic integrity and high sensitivity to apoptosis for eliminating severely damaged cells. We hypothesized that Δ133p53, a physiological inhibitory p53 isoform, is involved in the balanced regulation of self-renewing capacity, DNA damage repair and apoptosis. We examined 12 lines of human iPSC and their original fibroblasts, as well as three ESC lines, for endogenous protein levels of Δ133p53 and full-length p53 (FL-p53), and mRNA levels of various p53 target genes. While FL-p53 levels in iPSC and ESC widely ranged from below to above those in the fibroblasts, all iPSC and ESC lines expressed elevated levels of Δ133p53. The p53-inducible genes that mediate cellular senescence (p21WAF1, miR-34a, PAI-1 and IGFBP7), but not those for apoptosis (BAX and PUMA) and DNA damage repair (p53R2), were downregulated in iPSC and ESC. Consistent with these endogenous expression profiles, overexpression of Δ133p53 in human fibroblasts preferentially repressed the p53-inducible senescence mediators and significantly enhanced their reprogramming to iPSC. The iPSC lines derived from Δ133p53-overexpressing fibroblasts formed well-differentiated, benign teratomas in immunodeficient mice and had fewer numbers of somatic mutations than an iPSC derived from p53-knocked-down fibroblasts, suggesting that Δ133p53 overexpression is non- or less oncogenic and mutagenic than total inhibition of p53 activities. Overexpressed Δ133p53 prevented FL-p53 from binding to the regulatory regions of p21WAF1 and miR-34a promoters, providing a mechanistic basis for its dominant-negative inhibition of a subset of p53 target genes. This study supports the hypothesis that upregulation of Δ133p53 is an endogenous mechanism that facilitates human somatic cells to become self-renewing pluripotent stem cells with maintained apoptotic and DNA repair activities.


Cancer Research | 2013

Abstract PR08: Autophagic degradation of Δ133p53 during replicative cellular senescence: An isoform-specific protein degradation mechanism for p53

Izumi Horikawa; Kaori Fujita; Lisa M. Miller Jenkins; Yukiharu Hiyoshi; Abdul M. Mondal; Borivoj Vojtesek; David P. Lane; Ettore Appella; Curtis C. Harris

Δ133p53, a p53 isoform that dominant-negatively inhibits full-length p53, is downregulated at replicative senescence in a manner independent of mRNA regulation and proteasome-mediated degradation. We here demonstrate that, in contrast to proteasomal degradation of full-length p53, Δ133p53 is degraded by autophagy. Pharmacological inhibition of autophagy restores the reduced expression of Δ133p53 in replicatively senescent fibroblasts. The siRNA-mediated knockdown of pro-autophagic proteins (ATG5, ATG7 and Beclin-1) also results in restored expression of Δ133p53. The degradation of Δ133p53 via selective autophagy is indicated by its ubiquitination at the C-terminal lysine residues, its colocalization in the cytoplasm with a ubiquitin-binding protein p62/SQSTM1 and an autophagosome component LC3-II upon autophagy induction, and its stabilization by siRNA knockdown of p62/SQSTM1. A chaperone-associated E3 ubiquitin ligase STUB1, rather than MDM2, physically interacts with Δ133p53 and, consistent with the downregulation of endogenous STUB1 at replicative senescence, siRNA knockdown of STUB1 induces autophagic degradation of Δ133p53 and thereby senescence. Our data reveal that Δ133p53 degradation is controlled specifically by STUB1, rather than overall activity of autophagy. This study has identified for the first time a p53 isoform-specific protein turnover mechanism that orchestrates p53-mediated senescence and supports the hypothesis that Δ133p53 functions at the crossroads of senescence and autophagy. This abstract is also presented as poster B19. Citation Format: Izumi Horikawa, Kaori Fujita, Lisa Miller Jenkins, Yukiharu Hiyoshi, Abdul M. Mondal, Borivoj Vojtesek, David P. Lane, Ettore Appella, Curtis C. Harris. Autophagic degradation of Δ133p53 during replicative cellular senescence: An isoform-specific protein degradation mechanism for p53. [abstract]. In: Proceedings of the Third AACR International Conference on Frontiers in Basic Cancer Research; Sep 18-22, 2013; National Harbor, MD. Philadelphia (PA): AACR; Cancer Res 2013;73(19 Suppl):Abstract nr PR08.


Cancer Research | 2010

Abstract 3199: p53 represses TRF2 through E3 ubiquitin ligase Siah-1: Feedback regulation in telomere-initiated damage signaling

Izumi Horikawa; Kaori Fujita; Abdul M. Mondal; Borivoj Vojtesek; Jean-Christophe Bourdon; David P. Lane; Curtis C. Harris

Proceedings: AACR 101st Annual Meeting 2010‐‐ Apr 17‐21, 2010; Washington, DC p53 plays critical roles in tumor suppression, stem cell functions and aging in vivo. The telomere-capping protein complex (shelterin) prevents functional telomeres from undergoing erosion or end-to-end fusion and from initiating unwanted DNA damage response. Uncapped, dysfunctional telomeres at the end of cellular replicative lifespan lose this protective mechanism and trigger telomere-initiated DNA damage signaling to activate p53 and thereby induce cellular senescence. Here we report that p53 in turn controls a component of the shelterin complex, TRF2, through Siah-1, a p53-inducible E3 ubiquitin ligase. Endogenous TRF2 and Siah-1 were repressed and induced, respectively, in normal human fibroblasts at replicative senescence, when p53 was physiologically activated. Spontaneous allelic loss, shRNA-mediated knockdown, dominant-negative inhibition, nutlin-3a activation and overexpression of p53 all showed that p53 induced Siah-1 and repressed TRF2. TRF2 was subject to proteasomal degradation in a p53-dependent manner. Anti-TRF2 antibody-immunoprecipitated proteins were found to undergo p53- and Siah-1-mediated ubiquitination. In vitro and in vivo ubiquitination experiments showed that the E3 ligase activity of Siah-1 was responsible for TRF2 ubiquitination. Biologically, Siah-1 knockdown or TRF2 overexpression delayed the onset of replicative senescence, suggesting that the proteolytic control of TRF2 cooperates with the transcriptional regulation of p53 target genes (e.g., p21WAF1 and microRNA-34a) to regulate p53-mediated replicative senescence. This study reveals that p53, which is a downstream effector of the DNA damage signaling from uncapped telomeres, also functions upstream to regulate the telomere-capping complex, and suggests that the p53-Siah-1-TRF2 pathway takes an integral part in orchestrating the DNA damage response at telomeres. Given that TRF2 inhibits a p53-activating kinase ATM at telomeres, a positive feedback loop involving TRF2, ATM and p53 may function to amplify DNA damage-induced and p53-mediated cellular responses. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 3199.


Cancer Research | 2010

Abstract 2915: p53 isoforms Δ133p53 and p53β are endogenous regulators of replicative cellular senescence

Kaori Fujita; Abdul M. Mondal; Izumi Horikawa; G Nguyen; Kensuke Kumamoto; Jane J. Sohn; Elise D. Bowman; Mathe A. Mathe; Aaron J. Schetter; Sharon R. Pine; Helen Ji; Borivoj Vojtesek; Jean-Christophe Bourdon; David P. Lane; Curtis C. Harris

Proceedings: AACR 101st Annual Meeting 2010‐‐ Apr 17‐21, 2010; Washington, DC The finite proliferative potential of normal human cells leads to replicative cellular senescence, which is a critical barrier to tumor progression in vivo. The p53 signaling pathway plays central roles in the regulation of cellular senescence. Humans, as well as Drosophila and zebrafish, have p53 isoforms; however, their regulation and function are poorly understood. We here examine the expression profiles of two human p53 isoforms, p53β (lacking the C-terminal oligomerization domain due to an alternative mRNA splicing) and Δ133p53 (lacking the N-terminal transactivation and proline-rich domains due to the transcription from an alternative promoter in intron 4), during in vitro and in vivo cellular senescence and their biological activities in regulating cellular senescence. Induced p53β and diminished Δ133p53 were associated with replicative senescence, but not oncogene-induced senescence, in normal human fibroblasts. The replicatively senescent fibroblasts also expressed increased levels of miR-34a, a p53-induced microRNA, the antisense inhibition of which delayed the onset of replicative senescence. The siRNA-mediated knockdown of endogenous Δ133p53 induced cellular senescence, which was attributed to the regulation of p21WAF1 and other p53 transcriptional target genes. In overexpression experiments, while p53β cooperated with full-length p53 to accelerate cellular senescence, Δ133p53 repressed miR-34a expression and extended cellular replicative lifespan, providing a functional connection of this microRNA to the p53 isoform-mediated regulation of senescence. The senescence-associated signature of p53 isoform expression (i.e., elevated p53β and reduced Δ133p53) was observed in vivo in colon adenomas with senescent phenotypes. The decreased p53β and increased Δ133p53 expression found in colon carcinomas might signal an escape from the senescence barrier during the progression from premalignant to malignant tumors in vivo. This study shows that natural p53 isoforms constitute an endogenous regulatory mechanism for p53-mediated replicative senescence and may open up a new p53-based, senescence-mediated strategy to manipulate carcinogenesis and aging. The molecular details of the senescence-associated Δ133p53 repression and p53β induction are currently under investigation. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 2915.


Cancer Research | 2017

Abstract 922: Delta133p53 represses p53-inducible senescence genes and enhances the generation of human induced pluripotent stem cells

Izumi Horikawa; Kye-yoon Park; Han Li; Kazunobu Isogaya; Yukiharu Hiyoshi; Katsuhiro Anami; Ana I. Robles; Abdul M. Mondal; Kaori Fujita; Manuel Serrano; Curtis C. Harris

Collaboration


Dive into the Abdul M. Mondal's collaboration.

Top Co-Authors

Avatar

Curtis C. Harris

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Izumi Horikawa

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kaori Fujita

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Borivoj Vojtesek

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ettore Appella

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Sharon R. Pine

New York Medical College

View shared research outputs
Top Co-Authors

Avatar

Ana I. Robles

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge