Network


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

Hotspot


Dive into the research topics where Muneer G. Hasham is active.

Publication


Featured researches published by Muneer G. Hasham.


Nature Immunology | 2010

Widespread genomic breaks generated by activation-induced cytidine deaminase are prevented by homologous recombination

Muneer G. Hasham; Nina M Donghia; Eliot Coffey; Jane Maynard; Kathy J. Snow; Jacquelyn Ames; Robert Y Wilpan; Yishu He; Benjamin L King; Kevin D. Mills

Activation-induced cytidine deaminase (AID) is required for somatic hypermutation and immunoglobulin class switching in activated B cells. Because AID has no known target-site specificity, there have been efforts to identify non-immunoglobulin AID targets. We show here that AID acts promiscuously, generating widespread DNA double-strand breaks (DSBs), genomic instability and cytotoxicity in B cells with less homologous recombination ability. We demonstrate that the homologous-recombination factor XRCC2 suppressed AID-induced off-target DSBs, promoting B cell survival. Finally, we suggest that aberrations that affect human chromosome 7q36, including XRCC2, correlate with genomic instability in B cell cancers. Our findings demonstrate that AID has promiscuous genomic DSB-inducing activity, identify homologous recombination as a safeguard against off-target AID action, and have implications for genomic instability in B cell cancers.


Proceedings of the National Academy of Sciences of the United States of America | 2007

A mouse model of conditional lipodystrophy

Suyeon Kim; Li-Wen Huang; Kathy J. Snow; Vitaly Ablamunits; Muneer G. Hasham; Timothy H. Young; Angelique C. Paulk; Joel E. Richardson; Jason P. Affourtit; Tali Shalom-Barak; Yaacov Barak

Lipodystrophies are syndromes of adipose tissue degeneration associated with severe defects in lipid and glucose homeostasis. We report here the generation and analysis of Ppargldi, a targeted allele that confers conditional dominant lipodystrophy in mice. The Ppargldi allele was generated by insertion of the Tet activator (tTA) and a tTA-regulated Flag-Pparg1 transgene into the Pparg gene. Unexpectedly, tTA elicits mild lipodystrophy, insulin resistance, and dyslipidemia, and the Flag-PPARγ1 transgene surprisingly exacerbates these traits. Doxycycline can both completely prevent and reverse these phenotypes, providing a mouse model of inducible lipodystrophy. Embryonic fibroblasts from either Ppargldi/+ or the phenotypically similar aP2-nSrebp1c (Sr) transgenic mice undergo robust adipogenesis, suggesting that neither strain develops lipodystrophy because of defective adipocyte differentiation. In addition, Ppargldi/+ adipose tissue shares extensive gene expression aberrations with that of Sr mice, authenticating the phenotype at the molecular level and revealing a common expression signature of lipodystrophic fat. Thus, the Ppargldi/+ mouse provides a conditional animal model for studying lipodystrophy and its associated physiology and gene expression.


Journal of Immunology | 2012

Activation-Induced Cytidine Deaminase-Initiated Off-Target DNA Breaks Are Detected and Resolved during S Phase

Muneer G. Hasham; Kathy J. Snow; Nina M Donghia; Jane Branca; Mark D. Lessard; Janet Stavnezer; Lindsay S. Shopland; Kevin D. Mills

Activation-induced cytidine deaminase (AID) initiates DNA double-strand breaks (DSBs) in the IgH gene (Igh) to stimulate isotype class switch recombination (CSR), and widespread breaks in non-Igh (off-target) loci throughout the genome. Because the DSBs that initiate class switching occur during the G1 phase of the cell cycle, and are repaired via end joining, CSR is considered a predominantly G1 reaction. By contrast, AID-induced non-Igh DSBs are repaired by homologous recombination. Although little is known about the connection between the cell cycle and either induction or resolution of AID-mediated non-Igh DSBs, their repair by homologous recombination implicates post-G1 phases. Coordination of DNA breakage and repair during the cell cycle is critical to promote normal class switching and prevent genomic instability. To understand how AID-mediated events are regulated through the cell cycle, we have investigated G1-to-S control in AID-dependent genome-wide DSBs. We find that AID-mediated off-target DSBs, like those induced in the Igh locus, are generated during G1. These data suggest that AID-mediated DSBs can evade G1/S checkpoint activation and persist beyond G1, becoming resolved during S phase. Interestingly, DSB resolution during S phase can promote not only non-Igh break repair, but also Ig CSR. Our results reveal novel cell cycle dynamics in response to AID-initiated DSBs, and suggest that the regulation of the repair of these DSBs through the cell cycle may ensure proper class switching while preventing AID-induced genomic instability.


Journal of Experimental Medicine | 2013

Attenuating homologous recombination stimulates an AID-induced antileukemic effect

Kristin R. Lamont; Muneer G. Hasham; Nina M Donghia; Jane Branca; Margaret Chavaree; Betsy Chase; Anne Breggia; Jacquelyn Hedlund; Ivette F. Emery; Francesca Cavallo; Maria Jasin; Jens Rüter; Kevin D. Mills

Inhibition of the RAD51 homologous recombination factor prevents the repair of AID-initiated DNA breaks and induces apoptosis preferentially in AID-expressing human CLL.


Molecular and Cellular Biology | 2008

Homologous recombination is necessary for normal lymphocyte development.

Lura Brianna Caddle; Muneer G. Hasham; William H. Schott; Bobbi-Jo Shirley; Kevin D. Mills

ABSTRACT Primary immunodeficiencies are rare but serious diseases with diverse genetic causes. Accumulating evidence suggests that defects in DNA double-strand break (DSB) repair can underlie many of these syndromes. In this context, the nonhomologous end joining pathway of DSB repair is absolutely required for lymphoid development, but possible roles for the homologous recombination (HR) pathway have remained more controversial. While recent evidence suggests that HR may indeed be important to suppress lymphoid transformation, the specific relationship of HR to normal lymphocyte development remains unclear. We have investigated roles of the X-ray cross-complementing 2 (Xrcc2) HR gene in lymphocyte development. We show that HR is critical for normal B-cell development, with Xrcc2 nullizygosity leading to p53-dependent early S-phase arrest. In the absence of p53 (encoded by Trp53), Xrcc2-null B cells can fully develop but show high rates of chromosome and chromatid fragmentation. We present a molecular model wherein Xrcc2 is important to preserve or restore replication forks during rapid clonal expansion of developing lymphocytes. Our findings demonstrate a key role for HR in lymphoid development and suggest that Xrcc2 defects could underlie some human primary immunodeficiencies.


Journal of Immunology | 2017

Genetic and Small Molecule Disruption of the AID/RAD51 Axis Similarly Protects Nonobese Diabetic Mice from Type 1 Diabetes through Expansion of Regulatory B Lymphocytes

Jeremy Ratiu; Jeremy J. Racine; Muneer G. Hasham; Qiming Wang; Jane Branca; Harold D. Chapman; Jing Zhu; Nina M Donghia; Vivek Philip; William H. Schott; Clive Wasserfall; Mark A. Atkinson; Kevin D. Mills; Caroline M. Leeth; David V. Serreze

B lymphocytes play a key role in type 1 diabetes (T1D) development by serving as a subset of APCs preferentially supporting the expansion of autoreactive pathogenic T cells. As a result of their pathogenic importance, B lymphocyte–targeted therapies have received considerable interest as potential T1D interventions. Unfortunately, the B lymphocyte–directed T1D interventions tested to date failed to halt β cell demise. IgG autoantibodies marking humans at future risk for T1D indicate that B lymphocytes producing them have undergone the affinity-maturation processes of class switch recombination and, possibly, somatic hypermutation. This study found that CRISPR/Cas9-mediated ablation of the activation-induced cytidine deaminase gene required for class switch recombination/somatic hypermutation induction inhibits T1D development in the NOD mouse model. The activation-induced cytidine deaminase protein induces genome-wide DNA breaks that, if not repaired through RAD51-mediated homologous recombination, result in B lymphocyte death. Treatment with the RAD51 inhibitor 4,4′-diisothiocyanatostilbene-2, 2′-disulfonic acid also strongly inhibited T1D development in NOD mice. The genetic and small molecule–targeting approaches expanded CD73+ B lymphocytes that exert regulatory activity suppressing diabetogenic T cell responses. Hence, an initial CRISPR/Cas9-mediated genetic modification approach has identified the AID/RAD51 axis as a target for a potentially clinically translatable pharmacological approach that can block T1D development by converting B lymphocytes to a disease-inhibitory CD73+ regulatory state.


Disease Models & Mechanisms | 2017

Systemic autoimmunity induced by the TLR7/8 agonist Resiquimod causes myocarditis and dilated cardiomyopathy in a new mouse model of autoimmune heart disease.

Muneer G. Hasham; Nicoleta Baxan; Daniel J. Stuckey; Jane Branca; Bryant Perkins; Oliver Dent; Ted Duffy; Tolani S. Hameed; Sarah E. Stella; Mohammed Bellahcene; Michael D. Schneider; Sian E. Harding; Nadia Rosenthal; Susanne Sattler

ABSTRACT Systemic autoimmune diseases such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA) show significant heart involvement and cardiovascular morbidity, which can be due to systemically increased levels of inflammation or direct autoreactivity targeting cardiac tissue. Despite high clinical relevance, cardiac damage secondary to systemic autoimmunity lacks inducible rodent models. Here, we characterise immune-mediated cardiac tissue damage in a new model of SLE induced by topical application of the Toll-like receptor 7/8 (TLR7/8) agonist Resiquimod. We observe a cardiac phenotype reminiscent of autoimmune-mediated dilated cardiomyopathy, and identify auto-antibodies as major contributors to cardiac tissue damage. Resiquimod-induced heart disease is a highly relevant mouse model for mechanistic and therapeutic studies aiming to protect the heart during autoimmunity. Summary: A novel mouse model of autoimmune-mediated heart damage to study the underlying mechanisms and test therapeutic options for systemic autoimmunity.


Cardiovascular Research | 2018

Immunomodulatory interventions in myocardial infarction and heart failure: a systematic review of clinical trials and meta-analysis of IL-1 inhibition

Mona Panahi; Angelos Papanikolaou; Azam Torabi; Ji-Gang Zhang; Habib Khan; Ali Vazir; Muneer G. Hasham; John G.F. Cleland; Nadia Rosenthal; Sian E. Harding; Susanne Sattler

Abstract Following a myocardial infarction (MI), the immune system helps to repair ischaemic damage and restore tissue integrity, but excessive inflammation has been implicated in adverse cardiac remodelling and development towards heart failure (HF). Pre-clinical studies suggest that timely resolution of inflammation may help prevent HF development and progression. Therapeutic attempts to prevent excessive post-MI inflammation in patients have included pharmacological interventions ranging from broad immunosuppression to immunomodulatory approaches targeting specific cell types or factors with the aim to maintain beneficial aspects of the early post-MI immune response. These include the blockade of early initiators of inflammation including reactive oxygen species and complement, inhibition of mast cell degranulation and leucocyte infiltration, blockade of inflammatory cytokines, and inhibition of adaptive B and T-lymphocytes. Herein, we provide a systematic review on post-MI immunomodulation trials and a meta-analysis of studies targeting the inflammatory cytokine Interleukin-1. Despite an enormous effort into a significant number of clinical trials on a variety of targets, a striking heterogeneity in study population, timing and type of treatment, and highly variable endpoints limits the possibility for meaningful meta-analyses. To conclude, we highlight critical considerations for future studies including (i) the therapeutic window of opportunity, (ii) immunological effects of routine post-MI medication, (iii) stratification of the highly diverse post-MI patient population, (iv) the potential benefits of combining immunomodulatory with regenerative therapies, and at last (v) the potential side effects of immunotherapies.


Cancer Research | 2018

Abstract 347: Targeting the AICDA/RAD51 axis: A novel gain-of-function synthetic lethal therapy for the treatment of AICDA-expressing cancers

Muneer G. Hasham; Kin-hoe Chow; Tyler Maclay; Amber Cyr; Darryl Patrick; Melinda Day; Kevin D. Mills

A key characteristic of many cancers is genomic instability, and is often associated with poor prognosis. While genomic instability can promote tumorigenesis, it also provides a therapeutic opportunity for synthetic lethality. The induction of DNA damage creates stress that necessitates highly active DNA repair as a critical survival system for many transformed cells. Recently, therapeutics that target PARP1 have been approved by the FDA as synthetic lethal therapeutics for cancers with deficiencies in BRCA1/2 and associated pathways. Here we present the early development of a new synthetic lethal therapy that leverages gain-of-function abnormalities to selectively target cancer cells. Activation Induced Cytidine Deaminase (AICDA or AID) is a DNA-directed cytidine deaminase that is normally expressed exclusively and transiently in activated B-lymphocytes, where it plays critical roles in somatic hypermutation and immunoglobulin class switching. AICDA is a DNA damaging enzyme, producing DNA base pair mismatches which can subsequently be converted into mutations, DNA single strand breaks (SSB), or DNA double strand breaks (DSB). Numerous cancers show constitutive overexpression of AICDA, leading to hypermutation, genomic instability, and tumor evolution. We have previously demonstrated that cells expressing AICDA are critically dependent upon the DNA repair factor RAD51. Here we present data illustrating the effectiveness of a novel small molecule, CYT02A, in targeting the AICDA/RAD51 axis. Cell culture assays showed CYT02A to be a potent DNA repair inhibitor that targets RAD51 subcellular localization and filament formation. Although CYT02A was stable when incubated with liver microsomes. Although CYT-02A showed limited oral bioavailability, it was stable in liver microsome assyas assays and the pharmacokinetic profile following intravenous injection revealed dose proportionality and acceptable in vivo exposures. CYT02A was efficacious in a human-to-mouse xenograft model of chronic lymphocytic leukemia (CLL). CYT02A was administered intravenously at a concentration of 50 mg/kg to AID+ CLL xenograft mice daily for up to 9 days. Treated mice showed a significant reduction in CLL burden in the bone marrow compared to vehicle control animals. CYT02A was well tolerated by the animals, with no observable change in behavior or complete blood counts (CBC). Taken together, these data validate a novel gain of function synthetic lethal approach targeting the RAD51/AICDA axis. Preclinical efficacy data support this strategy for the treatment of AICDA-expressing cancers. We continue to build upon this foundation to produce a new therapeutic paradigm that may be effective in a wide range of both hematologic malignancies and solid tumors. Citation Format: Muneer Hasham, Kin-hoe Chow, Tyler Maclay, Amber Cyr, Darryl Patrick, Melinda Day, Kevin D. Mills. Targeting the AICDA/RAD51 axis: A novel gain-of-function synthetic lethal therapy for the treatment of AICDA-expressing cancers [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 347.


Cancer Biology & Therapy | 2018

Enhancing the efficacy of glycolytic blockade in cancer cells via RAD51 inhibition

John Wilson; Kin-hoe Chow; Nathan LaBrie; Jane Branca; Thomas J. Sproule; Bryant Perkins; Elise E. Wolf; Mauro Costa; Grace Stafford; Christine Rosales; Kevin D. Mills; Derry C. Roopenian; Muneer G. Hasham

ABSTRACT Targeting the early steps of the glycolysis pathway in cancers is a well-established therapeutic strategy; however, the doses required to elicit a therapeutic effect on the cancer can be toxic to the patient. Consequently, numerous preclinical and clinical studies have combined glycolytic blockade with other therapies. However, most of these other therapies do not specifically target cancer cells, and thus adversely affect normal tissue. Here we first show that a diverse number of cancer models – spontaneous, patient-derived xenografted tumor samples, and xenografted human cancer cells – can be efficiently targeted by 2-deoxy-D-Glucose (2DG), a well-known glycolytic inhibitor. Next, we tested the cancer-cell specificity of a therapeutic compound using the MEC1 cell line, a chronic lymphocytic leukemia (CLL) cell line that expresses activation induced cytidine deaminase (AID). We show that MEC1 cells, are susceptible to 4,4ʹ-Diisothiocyano-2,2ʹ-stilbenedisulfonic acid (DIDS), a specific RAD51 inhibitor. We then combine 2DG and DIDS, each at a lower dose and demonstrate that this combination is more efficacious than fludarabine, the current standard- of- care treatment for CLL. This suggests that the therapeutic blockade of glycolysis together with the therapeutic inhibition of RAD51-dependent homologous recombination can be a potentially beneficial combination for targeting AID positive cancer cells with minimal adverse effects on normal tissue. Implications: Combination therapy targeting glycolysis and specific RAD51 function shows increased efficacy as compared to standard of care treatments in leukemias.

Collaboration


Dive into the Muneer G. Hasham's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Francesca Cavallo

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nadia Rosenthal

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Sian E. Harding

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Susanne Sattler

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ali Vazir

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge