Skip to main content

Alexander C. Drohat, PhD

Academic Title:

Professor

Primary Appointment:

Biochemistry and Molecular Biology

Location:

108 N. Greene St., 419

Education and Training

University of Maryland, BS, Aerospace Engineering

University of Maryland School of Medicine, PhD, Biochemistry and Molecular Biology

Johns Hopkins University School of Medicine, NIST and Center for Advanced Research in Biotechnology, Postdoctoral Fellow

Biosketch

Our research centers on two broad areas, DNA repair and epigenetic regulation. The nucleobases of DNA are amenable to a broad range of chemical alterations, a feature that enables enzyme-mediated modifications but also allows for threatening DNA damage. We study enzymes that find and repair DNA lesions, thereby maintaining genomic integrity and protecting against cancer and other diseases. We also investigate enzymes that perform essential functions in epigenetic regulation, by acting on modified DNA bases. We use a broad range of biochemical, biophysical, structural, and molecular approaches, and collaborate with many other research groups. Some of our current research interests are summarized below.

Research/Clinical Keywords

DNA Repair, Epigenetics and DNA methylation, Enzymology, NMR, Structural Biology, SUMO modification

Highlighted Publications

Servius HW, Pidugu LS, Sherman ME, Drohat AC (2022) Rapid Excision of Oxidized Adenine by Thymine DNA Glycosylase, J Biol Chem2022 Nov 30;299(1):102756.

Pidugu LS, Bright H, Lin WJ, Majumdar C, Van Ostrand RP, David SS, Pozharski E, Drohat AC (2021) Structural Insights into the Mechanism of Base Excision by MBD4, J Mol Biol 433, 167097.

Pidugu LS, Dai Q, Malik SS, Pozharski E, Drohat AC (2019) Excision of 5-carboxylcytosine by Thymine DNA Glycosylase, J Am Chem Soc 141, 18851-61.

Dow BJ, Malik SS, Drohat AC (2019) Defining the Role of Nucleotide Flipping in Enzyme Specificity Using 19F NMR, J Am Chem Soc 141, 4952-62.

Coey CT, Drohat AC (2018) Defining the impact of sumoylation on substrate binding and catalysis by thymine DNA glycosylase, Nucleic Acids Res 46, 5159-70.

Pidugu LS, Flowers JW, Coey CT, Pozharski E, Greenberg MM, Drohat AC (2016) Structural basis for excision of 5-formylcytosine by Thymine DNA glycosylase, Biochemistry 55, 6205-8.

Drohat AC and Coey CT (2016) Role of Base Excision "Repair" Enzymes in Erasing Epigenetic Marks from DNA, Chem Rev 116, 12711-29.

Maiti A, Michelson AZ, Armwood CJ, Lee JK, Drohat AC (2013) Divergent Mechanisms for Enzymatic Excision of 5-formylcytosine and 5-carboxylcytosine from DNA, J Am Chem Soc 135, 15813-22.

Maiti A, Noon MS, MacKerell AD Jr, Pozhaski E, Drohat AC (2012) Lesion Processing by a Repair Enzyme is Severely Curtailed by Residues Needed to Prevent Aberrant Activity on Undamaged DNA, Proc Natl Acad Sci USA 109, 891-6.

Maiti A, Drohat AC (2011) Thymine DNA glycosylase can rapidly excise 5-formylcytosine and 5-carboxylcytosine: Potential implications for active demethylation of CpG sites, J Biol Chem 286, 35334-8.

Complete list of publications: NCBI and Google Scholar 

 

Research Interests

DNA Repair – Avoiding mutations caused by mC deamination

Often termed the “5th base” in DNA, 5-methylcytosine (mC) is a key epigenetic mark in eukaryotes, and it functions in restriction modification systems of archaea and bacteria. However, mC also threatens genetic and epigenetic integrity. Deamination of mC to T generates G/T mispairs, and, upon replication, CàT transitions. Through this process, mC deamination causes many point mutations in cancer and genetic disease. Two human glycosylases remove T from G/T mispairs, TDG (thymine DNA glycosylase) and MBD4 (methyl binding domain IV). Like other glycosylases, they flip a damaged base into their active site and cleave the base-sugar bond; follow-on base excision repair (BER) enzymes complete the repair process. While most glycosylases excise bases that are foreign to DNA (e.g., uracil), these mismatch enzymes remove a canonical base, thymine, from rare G/T mispairs but not from the vast background of A:T pairs. Because aberrant action on A:T pairs can be mutagenic and cytotoxic, specificity is critical for these enzymes, and we are studying how it is attained. We also study how TDG and MBD4 recognize other damaged bases, including uracil and 5-fluorouracil (5FU), among others. TDG excision of 5FU contributes to the antitumor effects of 5FU, which is used to treat cancer.

Pidugu et al. (2021) J Mol Biol

Dow et al (2019) J Am Chem Soc

Coey et al (2016) Nucleic Acids Res

Maiti et al. (2012) Proc Natl Acad Sci

Epigenetic Regulation - Base Excision Repair in active DNA Demethylation

Methyltransferases are known to convert C to mC, but the process for reversing this epigenetic modification had remained unclear. One mechanism involves DNA replication without subsequent remethylation. Recent studies establish a pathway for active DNA demethylation, involving TET (ten-eleven translocation) enzymes and TDG-initiated BER (Fig. 1). TET enzymes catalyze stepwise oxidation of mC, to give 5-hydroxymethyl-C (hmC), then 5-formyl-C (fC), and 5-carboxyl-C (caC). TDG excises fC and caC, and subsequent BER steps restore cytosine. This central function in epigenetic regulation likely explains findings that TDG is essential for embryonic development. We study the role of TDG and other BER enzymes in active DNA demethylation.

Pidugu et al (2019) J Am Chem Soc

Pidugu et al (2016) Biochemistry

Maiti et al (2013) J Am Chem Soc

Maiti and Drohat (2011) J Biol Chem

Protein Regulation by SUMO binding and SUMO conjugation

TDG can be covalently modified by SUMO (small ubiquitin-like modifier) proteins, and it also features a SUMO interacting motif (SIM) that binds non-covalentlyto SUMO domains. The SIM can bind free SUMO, a SUMO domain that is tethered to another protein, and intramolecularSUMO (tethered to TDG). We are investigating the effect of SUMO binding and SUMO conjugation on TDG function(s), and testing the current paradigm that SUMO modification of product-bound TDG is needed to relieve product inhibition and allow for efficient catalytic turnover. We are also studying how SUMO modification of TDG is regulated in cells.

Coey and Drohat (2018) Nucleic Acids Res

McLaughlin et al (2016) J Biol Chem

Coey at al. (2014) J Biol Chem

 

Awards and Affiliations

2022 - Elected Fellow, American Association for the Advancement of Science

2020-2025 - R35 MIRA Award, National Institute General Medical Sciences

2018-2022 - Member, NIH Molecular Genetics A (MGA) study section

2011-2015 - Member, DNA Mechanism in Cancer Peer Review Committee, American Cancer Society

2011-2014 - Editorial Advisory Board, ASBMB Today

2001 - Travel Award, American Chemical Society National Meeting

1998-2000 - National Research Council Postdoctoral Research Fellowship

1997 - Graduate Student Research Day Award, University of Maryland Baltimore

1996-97 - Graduate Merit Award, University of Maryland Baltimore

Grants and Contracts

Our work has been supported since 2005 by grants from the National Institutes of Health

 

R35GM136225          05/01/2020–04/30/2025

Mechanisms of BER in Genomic Integrity and Epigenetic Regulation

 

R01GM072711         02/01/2005 – 04/30/2022

Mechanism of Glycosylase Enzymes in DNA Repair and Demethylation