Network


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

Hotspot


Dive into the research topics where Manikandan Paramasivam is active.

Publication


Featured researches published by Manikandan Paramasivam.


Molecular Cell | 2012

A Ubiquitin-Binding Protein, FAAP20, Links RNF8-Mediated Ubiquitination to the Fanconi Anemia DNA Repair Network

Zhijiang Yan; Rong Guo; Manikandan Paramasivam; Weiping Shen; Chen Ling; David Fox; Yucai Wang; Anneke B. Oostra; Julia Kuehl; Duck Yeon Lee; Minoru Takata; Maureen E. Hoatlin; Detlev Schindler; Hans Joenje; Johan P. de Winter; Lei Li; Michael M. Seidman; Weidong Wang

The Fanconi anemia (FA) protein network is necessary for repair of DNA interstrand crosslinks (ICLs), but its control mechanism remains unclear. Here we show that the network is regulated by a ubiquitin signaling cascade initiated by RNF8 and its partner, UBC13, and mediated by FAAP20, a component of the FA core complex. FAAP20 preferentially binds the ubiquitin product of RNF8-UBC13, and this ubiquitin-binding activity and RNF8-UBC13 are both required for recruitment of FAAP20 to ICLs. Both RNF8 and FAAP20 are required for recruitment of FA core complex and FANCD2 to ICLs, whereas RNF168 can modulate efficiency of the recruitment. RNF8 and FAAP20 are needed for efficient FANCD2 monoubiquitination, a key step of the FA network; RNF8 and the FA core complex work in the same pathway to promote cellular resistance to ICLs. Thus, the RNF8-FAAP20 ubiquitin cascade is critical for recruiting FA core complex to ICLs and for normal function of the FA network.


Cancer Research | 2013

ATR-Dependent Phosphorylation of FANCM at Serine 1045 Is Essential for FANCM Functions

Thiyam Ramsing Singh; Abdullah Mahmood Ali; Manikandan Paramasivam; Arun Pradhan; Kebola Wahengbam; Michael M. Seidman; Amom Ruhikanta Meetei

Fanconi anemia (FA) is a genome instability syndrome that has been associated with both cancer predisposition and bone marrow failure. FA proteins are involved in cellular response to replication stress in which they coordinate DNA repair with DNA replication and cell-cycle progression. One regulator of the replication stress response is the ATP-dependent DNA translocase FANCM, which we have shown to be hyperphosphorylated in response to various genotoxic agents. However, the significance of this phosphorylation remained unclear. Here, we show that genotoxic stress-induced FANCM phosphorylation is ATR-dependent and that this modification is highly significant for the cellular response to replication stress. We identified serine (S1045) residue of FANCM that is phosphorylated in response to genotoxic stress and this effect is ATR-dependent. We show that S1045 is required for FANCM functions including its role in FA pathway integrity, recruiting FANCM to the site of interstrand cross links, preventing the cells from entering mitosis prematurely, and efficient activation of the CHK1 and G2-M checkpoints. Overall, our data suggest that an ATR-FANCM feedback loop is present in the FA and replication stress response pathways and that it is required for both efficient ATR/CHK1 checkpoint activation and FANCM function.


Genes & Development | 2015

MERIT40 cooperates with BRCA2 to resolve DNA interstrand cross-links

Qinqin Jiang; Manikandan Paramasivam; Bernadette Aressy; Junmin Wu; Marina Bellani; Wei Tong; Michael M. Seidman; Roger A. Greenberg

MERIT40 is an essential component of the RAP80 ubiquitin recognition complex that targets BRCA1 to DNA damage sites. Although this complex is required for BRCA1 foci formation, its physiologic role in DNA repair has remained enigmatic, as has its relationship to canonical DNA repair mechanisms. Surprisingly, we found that Merit40(-/-) mice displayed marked hypersensitivity to DNA interstrand cross-links (ICLs) but not whole-body irradiation. MERIT40 was rapidly recruited to ICL lesions prior to FANCD2, and Merit40-null cells exhibited delayed ICL unhooking coupled with reduced end resection and homologous recombination at ICL damage. Interestingly, Merit40 mutation exacerbated ICL-induced chromosome instability in the context of concomitant Brca2 deficiency but not in conjunction with Fancd2 mutation. These findings implicate MERIT40 in the earliest stages of ICL repair and define specific functional interactions between RAP80 complex-dependent ubiquitin recognition and the Fanconi anemia (FA)-BRCA ICL repair network.


Journal of the American Chemical Society | 2014

Arsenite binds to the RING finger domains of RNF20-RNF40 histone E3 ubiquitin ligase and inhibits DNA double-strand break repair.

Fan Zhang; Manikandan Paramasivam; Qian Cai; Xiaoxia Dai; Pengcheng Wang; Krystal Lin; Jikui Song; Michael M. Seidman; Yinsheng Wang

Arsenic is a widespread environmental contaminant. However, the exact molecular mechanisms underlying the carcinogenic effects of arsenic remain incompletely understood. Core histones can be ubiquitinated by RING finger E3 ubiquitin ligases, among which the RNF20-RNF40 heterodimer catalyzes the ubiquitination of histone H2B at lysine 120. This ubiquitination event is important for the formation of open and biochemically accessible chromatin fiber that is conducive for DNA repair. Herein, we found that arsenite could bind directly to the RING finger domains of RNF20 and RNF40 in vitro and in cells, and treatment with arsenite resulted in substantially impaired H2B ubiquitination in multiple cell lines. Exposure to arsenite also diminished the recruitment of BRCA1 and RAD51 to laser-induced DNA double-strand break (DSB) sites, compromised DNA DSB repair in human cells, and rendered cells sensitive toward a radiomimetic agent, neocarzinostatin. Together, the results from the present study revealed, for the first time, that arsenite may exert its carcinogenic effect by targeting cysteine residues in the RING finger domains of histone E3 ubiquitin ligase, thereby altering histone epigenetic mark and compromising DNA DSB repair. Our results also suggest arsenite as a general inhibitor for RING finger E3 ubiquitin ligases.


Journal of Biological Chemistry | 2013

NEIL1 responds and binds to psoralen-induced DNA interstrand crosslinks

Daniel R. McNeill; Manikandan Paramasivam; Jakita Baldwin; Jing Huang; Vaddadi N. Vyjayanti; Michael M. Seidman; David M. Wilson

Background: Components of base excision repair participate in the processing of psoralen-induced DNA adducts. Results: NEIL1 glycosylase responds and binds to psoralen interstrand crosslinks, and interferes with efficient recognition and removal of these lesions. Conclusion: NEIL1 exhibits affinity for DNA interstrand crosslinks and regulates crosslink processing. Significance: Besides the efficiency of the canonical pathways, the abundance and composition of NEIL1 may influence responsiveness to environmental and therapeutic DNA crosslinking agents. Recent evidence suggests a role for base excision repair (BER) proteins in the response to DNA interstrand crosslinks, which block replication and transcription, and lead to cell death and genetic instability. Employing fluorescently tagged fusion proteins and laser microirradiation coupled with confocal microscopy, we observed that the endonuclease VIII-like DNA glycosylase, NEIL1, accumulates at sites of oxidative DNA damage, as well as trioxsalen (psoralen)-induced DNA interstrand crosslinks, but not to angelicin monoadducts. While recruitment to the oxidative DNA lesions was abrogated by the anti-oxidant N-acetylcysteine, this treatment did not alter the accumulation of NEIL1 at sites of interstrand crosslinks, suggesting distinct recognition mechanisms. Consistent with this conclusion, recruitment of the NEIL1 population variants, G83D, C136R, and E181K, to oxidative DNA damage and psoralen-induced interstrand crosslinks was differentially affected by the mutation. NEIL1 recruitment to psoralen crosslinks was independent of the nucleotide excision repair recognition factor, XPC. Knockdown of NEIL1 in LN428 glioblastoma cells resulted in enhanced recruitment of XPC, a more rapid removal of digoxigenin-tagged psoralen adducts, and decreased cellular sensitivity to trioxsalen plus UVA, implying that NEIL1 and BER may interfere with normal cellular processing of interstrand crosslinks. While exhibiting no enzymatic activity, purified NEIL1 protein bound stably to psoralen interstrand crosslink-containing synthetic oligonucleotide substrates in vitro. Our results indicate that NEIL1 recognizes specifically and distinctly interstrand crosslinks in DNA, and can obstruct the efficient removal of lethal crosslink adducts.


Molecular and Cellular Biology | 2015

Nuclear GIT2 Is an ATM Substrate and Promotes DNA Repair

Daoyuan Lu; Huan Cai; Sung-Soo Park; Sana Siddiqui; Richard T. Premont; Robert Schmalzigaug; Manikandan Paramasivam; Michael M. Seidman; Ionoa Bodogai; Arya Biragyn; Caitlin M. Daimon; Bronwen Martin; Stuart Maudsley

ABSTRACT Insults to nuclear DNA induce multiple response pathways to mitigate the deleterious effects of damage and mediate effective DNA repair. G-protein-coupled receptor kinase-interacting protein 2 (GIT2) regulates receptor internalization, focal adhesion dynamics, cell migration, and responses to oxidative stress. Here we demonstrate that GIT2 coordinates the levels of proteins in the DNA damage response (DDR). Cellular sensitivity to irradiation-induced DNA damage was highly associated with GIT2 expression levels. GIT2 is phosphorylated by ATM kinase and forms complexes with multiple DDR-associated factors in response to DNA damage. The targeting of GIT2 to DNA double-strand breaks was rapid and, in part, dependent upon the presence of H2AX, ATM, and MRE11 but was independent of MDC1 and RNF8. GIT2 likely promotes DNA repair through multiple mechanisms, including stabilization of BRCA1 in repair complexes; upregulation of repair proteins, including HMGN1 and RFC1; and regulation of poly(ADP-ribose) polymerase activity. Furthermore, GIT2-knockout mice demonstrated a greater susceptibility to DNA damage than their wild-type littermates. These results suggest that GIT2 plays an important role in MRE11/ATM/H2AX-mediated DNA damage responses.


Frontiers in Genetics | 2016

Single Molecule Analysis of Laser Localized Interstrand Crosslinks

Jing Huang; Himabindu Gali; Manikandan Paramasivam; Parameswary A. Muniandy; Julia Gichimu; Marina Bellani; Michael M. Seidman

DNA interstrand crosslinks (ICLs) block unwinding of the double helix, and have always been regarded as major challenges to replication and transcription. Compounds that form these lesions are very toxic and are frequently used in cancer chemotherapy. We have developed two strategies, both based on immunofluorescence (IF), for studying cellular responses to ICLs. The basis of each is psoralen, a photoactive (by long wave ultraviolet light, UVA) DNA crosslinking agent, to which we have linked an antigen tag. In the one approach, we have taken advantage of DNA fiber and immuno-quantum dot technologies for visualizing the encounter of replication forks with ICLs induced by exposure to UVA lamps. In the other, psoralen ICLs are introduced into nuclei in live cells in regions of interest defined by a UVA laser. The antigen tag can be displayed by conventional IF, as can the recruitment and accumulation of DNA damage response proteins to the laser localized ICLs. However, substantial difference between the technologies creates considerable uncertainty as to whether conclusions from one approach are applicable to those of the other. In this report, we have employed the fiber/quantum dot methodology to determine lesion density and spacing on individual DNA molecules carrying laser localized ICLs. We have performed the same measurements on DNA fibers with ICLs induced by exposure of psoralen to UVA lamps. Remarkably, we find little difference in the adduct distribution on fibers prepared from cells exposed to the different treatment protocols. Furthermore, there is considerable similarity in patterns of replication in the vicinity of the ICLs introduced by the two techniques.


DNA Repair | 2018

Imaging cellular responses to antigen tagged DNA damage

Marina Bellani; Jing Huang; Manikandan Paramasivam; Durga Pokharel; Julia Gichimu; Jing Zhang; Michael M. Seidman

Repair pathways of covalent DNA damage are understood in considerable detail due to decades of brilliant biochemical studies by many investigators. An important feature of these experiments is the defined adduct location on oligonucleotide or plasmid substrates that are incubated with purified proteins or cell free extracts. With some exceptions, this certainty is lost when the inquiry shifts to the response of living mammalian cells to the same adducts in genomic DNA. This reflects the limitation of assays, such as those based on immunofluorescence, that are widely used to follow responding proteins in cells exposed to a DNA reactive compound. The lack of effective reagents for adduct detection means that the proximity between responding proteins and an adduct must be assumed. Since these assumptions can be incorrect, models based on in vitro systems may fail to account for observations made in vivo. Here we discuss the use of a detection tag to address the problem of lesion location, as illustrated by our recent work on replication dependent and independent responses to interstrand crosslinks.


Journal of Visualized Experiments | 2017

Single Molecule Analysis of Laser Localized Psoralen Adducts

Jing Huang; Himabindu Gali; Julia Gichimu; Marina Bellani; Durga Pokharel; Manikandan Paramasivam; Michael M. Seidman

The DNA Damage Response (DDR) has been extensively characterized in studies of double strand breaks (DSBs) induced by laser micro beam irradiation in live cells. The DDR to helix distorting covalent DNA modifications, including interstrand DNA crosslinks (ICLs), is not as well defined. We have studied the DDR stimulated by ICLs, localized by laser photoactivation of immunotagged psoralens, in the nuclei of live cells. In order to address fundamental questions about adduct distribution and replication fork encounters, we combined laser localization with two other technologies. DNA fibers are often used to display the progress of replication forks by immunofluorescence of nucleoside analogues incorporated during short pulses. Immunoquantum dots have been widely employed for single molecule imaging. In the new approach, DNA fibers from cells carrying laser localized ICLs are spread onto microscope slides. The tagged ICLs are displayed with immunoquantum dots and the inter-lesion distances determined. Replication fork collisions with ICLs can be visualized and different encounter patterns identified and quantitated.


Cell Reports | 2015

UHRF1 Contributes to DNA Damage Repair as a Lesion Recognition Factor and Nuclease Scaffold

Yanyan Tian; Manikandan Paramasivam; Gargi Ghosal; Ding Chen; Xi Shen; Yaling Huang; Shamima Akhter; Randy J. Legerski; Junjie Chen; Michael M. Seidman; Jun Qin; Lei Li

Collaboration


Dive into the Manikandan Paramasivam's collaboration.

Top Co-Authors

Avatar

Michael M. Seidman

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Marina Bellani

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Jing Huang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Julia Gichimu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Arya Biragyn

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Bronwen Martin

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Caitlin M. Daimon

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Daoyuan Lu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Durga Pokharel

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Himabindu Gali

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge