Network


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

Hotspot


Dive into the research topics where Shakur Mohibi is active.

Publication


Featured researches published by Shakur Mohibi.


Journal of Carcinogenesis | 2011

Mouse models of estrogen receptor-positive breast cancer

Shakur Mohibi; Sameer Mirza; Hamid Band; Vimla Band

Breast cancer is the most frequent malignancy and second leading cause of cancer-related deaths among women. Despite advances in genetic and biochemical analyses, the incidence of breast cancer and its associated mortality remain very high. About 60 – 70% of breast cancers are Estrogen Receptor alpha (ER-α) positive and are dependent on estrogen for growth. Selective estrogen receptor modulators (SERMs) have therefore provided an effective targeted therapy to treat ER-α positive breast cancer patients. Unfortunately, development of resistance to endocrine therapy is frequent and leads to cancer recurrence. Our understanding of molecular mechanisms involved in the development of ER-α positive tumors and their resistance to ER antagonists is currently limited due to lack of experimental models of ER-α positive breast cancer. In most mouse models of breast cancer, the tumors that form are typically ER-negative and independent of estrogen for their growth. However, in recent years more attention has been given to develop mouse models that develop different subtypes of breast cancers, including ER-positive tumors. In this review, we discuss the currently available mouse models that develop ER-α positive mammary tumors and their potential use to elucidate the molecular mechanisms of ER-α positive breast cancer development and endocrine resistance.


Journal of Biological Chemistry | 2012

Mammalian Alteration/Deficiency in Activation 3 (Ada3) Is Essential for Embryonic Development and Cell Cycle Progression

Shakur Mohibi; Channabasavaiah B. Gurumurthy; Alo Nag; Jun Wang; Sameer Mirza; Yousaf Mian; Meghan Quinn; Bryan J. Katafiasz; James D. Eudy; Sanjit Pandey; Chittibabu Guda; Mayumi Naramura; Hamid Band; Vimla Band

Background: Ada3 is a core component of HAT containing coactivator complexes. Results: Germline deletion of Ada3 is embryonic lethal, and cell deletion leads to abnormal cell cycle progression. Conclusion: Ada3 is a critical protein at organismic and cellular level. Significance: This study describes a novel role of Ada3, a component of HAT complexes, as a critical regulator of cell survival. Ada3 protein is an essential component of histone acetyl transferase containing coactivator complexes conserved from yeast to human. We show here that germline deletion of Ada3 in mouse is embryonic lethal, and adenovirus-Cre mediated conditional deletion of Ada3 in Ada3FL/FL mouse embryonic fibroblasts leads to a severe proliferation defect which was rescued by ectopic expression of human Ada3. A delay in G1 to S phase of cell cycle was also seen that was due to accumulation of Cdk inhibitor p27 which was an indirect effect of c-myc gene transcription control by Ada3. We further showed that this defect could be partially reverted by knocking down p27. Additionally, drastic changes in global histone acetylation and changes in global gene expression were observed in microarray analyses upon loss of Ada3. Lastly, formation of abnormal nuclei, mitotic defects and delay in G2/M to G1 transition was seen in Ada3 deleted cells. Taken together, we provide evidence for a critical role of Ada3 in embryogenesis and cell cycle progression as an essential component of HAT complex.


Cell Cycle | 2012

Alteration/deficiency in activation-3 (Ada3) plays a critical role in maintaining genomic stability.

Sameer Mirza; Bryan J. Katafiasz; Rakesh Kumar; Jun Wang; Shakur Mohibi; Smrati Jain; Channabasavaiah B. Gurumurthy; Tej K. Pandita; Bhavana J. Dave; Hamid Band; Vimla Band

Cell cycle regulation and DNA repair following damage are essential for maintaining genome integrity. DNA damage activates checkpoints in order to repair damaged DNA prior to exit to the next phase of cell cycle. Recently, we have shown the role of Ada3, a component of various histone acetyltransferase complexes, in cell cycle regulation, and loss of Ada3 results in mouse embryonic lethality. Here, we used adenovirus-Cre-mediated Ada3 deletion in Ada3fl/fl mouse embryonic fibroblasts (MEFs) to assess the role of Ada3 in DNA damage response following exposure to ionizing radiation (IR). We report that Ada3 depletion was associated with increased levels of phospho-ATM (pATM), γH2AX, phospho-53BP1 (p53BP1) and phospho-RAD51 (pRAD51) in untreated cells; however, radiation response was intact in Ada3−/− cells. Notably, Ada3−/− cells exhibited a significant delay in disappearance of DNA damage foci for several critical proteins involved in the DNA repair process. Significantly, loss of Ada3 led to enhanced chromosomal aberrations, such as chromosome breaks, fragments, deletions and translocations, which further increased upon DNA damage. Notably, the total numbers of aberrations were more clearly observed in S-phase, as compared with G₁ or G₂ phases of cell cycle with IR. Lastly, comparison of DNA damage in Ada3fl/fl and Ada3−/− cells confirmed higher residual DNA damage in Ada3−/− cells, underscoring a critical role of Ada3 in the DNA repair process. Taken together, these findings provide evidence for a novel role for Ada3 in maintenance of the DNA repair process and genomic stability.


Journal of Biological Chemistry | 2015

Alteration/Deficiency in Activation 3 (ADA3) Protein, a Cell Cycle Regulator, Associates with the Centromere through CENP-B and Regulates Chromosome Segregation

Shakur Mohibi; Shashank Srivastava; Jun Wang-France; Sameer Mirza; Xiangshan Zhao; Hamid Band; Vimla Band

Background: ADA3, a conserved component of several HAT complexes, regulates mitosis. Results: ADA3 associates with the centromere through CENP-B and regulates chromosome segregation by directing centromeric loading of CENP-B. Conclusion: ADA3 regulates mitosis by its association with the CENP-B/centromere and regulates segregation of chromosomes. Significance: This study provides the mechanism of how ADA3 regulates mitosis to maintain genomic stability. ADA3 (alteration/deficiency in activation 3) is a conserved component of several transcriptional co-activator and histone acetyltransferase (HAT) complexes. Recently, we generated Ada3 knock-out mice and demonstrated that deletion of Ada3 leads to early embryonic lethality. The use of Ada3FL/FL mouse embryonic fibroblasts with deletion of Ada3 using adenovirus Cre showed a critical role of ADA3 in cell cycle progression through mitosis. Here, we demonstrate an association of ADA3 with the higher order repeat region of the α-satellite region on human X chromosome centromeres that is consistent with its role in mitosis. Given the role of centromere proteins (CENPs) in mitosis, we next analyzed whether ADA3 associates with the centromere through CENPs. Both an in vivo proximity ligation assay and immunofluorescence studies confirmed the association of ADA3 with CENP-B protein, a highly conserved centromeric protein that binds to the 17-bp DNA sequences on α-satellite DNA. Deletional analysis showed that ADA3 directly associates with CENP-B through its N terminus, and a CENP-B binding-deficient mutant of ADA3 was incompetent in cell proliferation rescue. Notably, knockdown of ADA3 decreased binding of CENP-B onto the centromeres, suggesting that ADA3 is required for the loading of CENP-B onto the centromeres. Finally, we show that deletion of Ada3 from Ada3FL/FL mouse embryonic fibroblasts exhibited various chromosome segregation defects. Taken together, we demonstrate a novel ADA3 interaction with CENP-B-centromere that may account for its previously known function in mitosis. This study, together with its known function in maintaining genomic stability and its mislocalization in cancers, suggests an important role of ADA3 in mitosis.


Molecular and Cellular Biology | 2016

Acetylation of Mammalian ADA3 Is Required for Its Functional Roles in Histone Acetylation and Cell Proliferation

Shakur Mohibi; Shashank Srivastava; Aditya Bele; Sameer Mirza; Hamid Band; Vimla Band

ABSTRACT Alteration/deficiency in activation 3 (ADA3) is an essential component of specific histone acetyltransferase (HAT) complexes. We have previously shown that ADA3 is required for establishing global histone acetylation patterns and for normal cell cycle progression (S. Mohibi et al., J Biol Chem 287:29442–29456, 2012, http://dx.doi.org/10.1074/jbc.M112.378901). Here, we report that these functional roles of ADA3 require its acetylation. We show that ADA3 acetylation, which is dynamically regulated in a cell cycle-dependent manner, reflects a balance of coordinated actions of its associated HATs, GCN5, PCAF, and p300, and a new partner that we define, the deacetylase SIRT1. We use mass spectrometry and site-directed mutagenesis to identify major sites of ADA3 acetylated by GCN5 and p300. Acetylation-defective mutants are capable of interacting with HATs and other components of HAT complexes but are deficient in their ability to restore ADA3-dependent global or locus-specific histone acetylation marks and cell proliferation in Ada3-deleted murine embryonic fibroblasts (MEFs). Given the key importance of ADA3-containing HAT complexes in the regulation of various biological processes, including the cell cycle, our study presents a novel mechanism to regulate the function of these complexes through dynamic ADA3 acetylation.


Breast Cancer Research | 2016

ADA3 regulates normal and tumor mammary epithelial cell proliferation through c-MYC

Nicolas I. Griffin; Gayatri Sharma; Xiangshan Zhao; Sameer Mirza; Shashank Srivastava; Bhavana J. Dave; Mohammed A. Aleskandarany; Emad A. Rakha; Shakur Mohibi; Hamid Band; Vimla Band

BackgroundWe have established the critical role of ADA3 as a coactivator of estrogen receptor (ER), as well as its role in cell cycle progression. Furthermore, we showed that ADA3 is predominantly nuclear in mammary epithelium, and in ER+, but is cytoplasmic in ER- breast cancers, the latter correlating with poor survival. However, the role of nuclear ADA3 in human mammary epithelial cells (hMECs), and in ER+ breast cancer cells, as well as the importance of ADA3 expression in relation to patient prognosis and survival in ER+ breast cancer have remained uncharacterized.MethodsWe overexpressed ADA3 in hMECs or in ER+ breast cancer cells and assessed the effect on cell proliferation. The expression of ADA3 was analyzed then correlated with the expression of various prognostic markers, as well as survival of breast cancer patients.ResultsOverexpression of ADA3 in ER- hMECs as well as in ER+ breast cancer cell lines enhanced cell proliferation. These cells showed increased cyclin B and c-MYC, decreased p27 and increased SKP2 levels. This was accompanied by increased mRNA levels of early response genes c-FOS, EGR1, and c-MYC. Analysis of breast cancer tissue specimens showed a significant correlation of ADA3 nuclear expression with c-MYC expression. Furthermore, nuclear ADA3 and c-MYC expression together showed significant correlation with tumor grade, mitosis, pleomorphism, NPI, ER/PR status, Ki67 and p27 expression. Importantly, within ER+ cases, expression of nuclear ADA3 and c-MYC also significantly correlated with Ki67 and p27 expression. Univariate Kaplan Meier analysis of four groups in the whole, as well as the ER+ patients showed that c-MYC and ADA3 combinatorial phenotypes showed significantly different breast cancer specific survival with c-MYC-high and ADA3-Low subgroup had the worst outcome. Using multivariate analyses within the whole cohort and the ER+ subgroups, the significant association of ADA3 and c-MYC expression with patients’ outcome was independent of tumor grade, stage and size, and ER status.ConclusionADA3 overexpression enhances cell proliferation that is associated with increased expression of c-MYC. Expression patterns with respect to ADA3/c-MYC can divide patients into four significantly different subgroups, with c-MYC High and ADA3 Low status independently predicting poor survival in patients.


Cell Cycle | 2017

Epidermal Growth Factor Receptor activation promotes ADA3 acetylation through the AKT-p300 pathway

Shashank Srivastava; Shakur Mohibi; Sameer Mirza; Hamid Band; Vimla Band

ABSTRACT The ADA3 (Alteration/Deficiency in Activation 3) protein is an essential adaptor component of several Lysine Acetyltransferase (KAT) complexes involved in chromatin modifications. Previously, we and others have demonstrated a crucial role of ADA3 in cell cycle progression and in maintenance of genomic stability. Recently, we have shown that acetylation of ADA3 is key to its role in cell cycle progression. Here, we demonstrate that AKT activation downstream of Epidermal Growth Factor Receptor (EGFR) family proteins stimulation leads to phosphorylation of p300, which in turn promotes the acetylation of ADA3. Inhibition of upstream receptor tyrosine kinases (RTKs), HER1 (EGFR)/HER2 by lapatinib and the accompanying reduction of phospho-AKT levels led to a decrease in p300 phosphorylation and ADA3 protein levels. The p300/PCAF inhibitor garcinol also destabilized the ADA3 protein in a proteasome-dependent manner and an ADA3 mutant with K→R mutations exhibited a marked increase in half-life, consistent with opposite role of acetylation and ubiquitination of ADA3 on shared lysine residues. ADA3 knockdown led to cell cycle inhibitory effects, as well as apoptosis similar to those induced by lapatinib treatment of HER2+ breast cancer cells, as seen by accumulation of CDK inhibitor p27, reduction in mitotic marker pH3(S10), and a decrease in the S-phase marker PCNA, as well as the appearance of cleaved PARP. Taken together our results reveal a novel RTK-AKT-p300-ADA3 signaling pathway involved in growth factor-induced cell cycle progression.


Archive | 2014

Role of Alteration/Deficiency in Activation (ADA) Complex in Cell Cycle, Genomic Instability and Cancer

Shakur Mohibi; Shashank Srivastava; Hamid Band; Vimla Band

In eukaryotes, DNA wraps around histone proteins to form highly condensed chromatin structures that usually remain inert and inaccessible to proteins involved in DNA-related processes. Thus, multitudes of important DNA-related biological processes, including transcription, replication, DNA repair, apoptosis, chromosome condensation, and segregation, are dependent upon alteration of this chromatin structure so that proteins involved in these processes can access the DNA. This required change in chromatin structure is brought about by binding of various chromatin modifying proteins that loosen the chromatin by distinct mechanisms, one of which is covalent histone modification. Various histone post-translational modifications, specifically acetylation, play a major role in opening up of this highly condensed chromatin allowing access to proteins involved in the several important processes. Histone acetyl transferases (HATs) and histone deacetylases (HDACs) are important for maintaining a steady-state level of this particular post-translational modification in cells and are present in multi-subunit complexes. One such multi-subunit HAT complex is the alteration/deficiency in activation (ADA) complex, which was originally discovered in yeast and is now known to be also present in mammalian cells as part of much larger HAT complexes. In this chapter, we discuss various components of the ADA complex with a special focus on the adaptor proteins Ada3 and Ada2 (Ada2a and Ada2b) for their role in important physiological processes, such as the cell cycle, genomic integrity, DNA repair response, and in pathology such as cancer. Further, we discuss recent developments using various inhibitors to target the HAT enzymes and disrupt HAT complex function as an anti-cancer strategy.


Advances in Experimental Medicine and Biology | 2011

Breast Cancer Subtypes: Two Decades of Journey from Cell Culture to Patients

Xiangshan Zhao; Channabasavaiah B. Gurumurthy; Gautam K. Malhotra; Sameer Mirza; Shakur Mohibi; Aditya Bele; Meghan Quinn; Hamid Band; Vimla Band

Recent molecular profiling has identified six major subtypes of breast cancers that exhibit different survival outcomes for patients. To address the origin of different subtypes of breast cancers, we have now identified, isolated, and immortalized (using hTERT) mammary stem/progenitor cells which maintain their stem/progenitor properties even after immortalization. Our decade long research has shown that these stem/progenitor cells are highly susceptible to oncogenesis. Given the emerging evidence that stem/progenitor cells are precursors of cancers and that distinct subtypes of breast cancer have different survival outcome, these cellular models provide novel tools to understand the oncogenic process leading to various subtypes of breast cancers and for future development of novel therapeutic strategies to treat different subtypes of breast cancers.


Cancer Research | 2016

Abstract 2719: ADA3, a cell cycle regulator, regulates chromosome segregation

Shashank Srivastava; Shakur Mohibi; June Wang-France; Sameer Mirza; Xiangshan Zhao; Hamid Band; Vimla Bnad

The RNA polymerase II mediated transcription requires the higher degree structure of chromatin to be relieved so that general transcription machinery can access the DNA. The acetylation of DNA bound histones at specific loci is the major epigenetic modification by which opening of chromatin is achieved. HATs (Histone Acetyl Transferase) are the enzymes that catalyze the acetylation of histones and require association with mediator proteins for their function. One such mediator protein is ADA3 (Alteration/Deficiency in Activation 3), which was initially discovered as a component of multi-protein complex that contains either GCN5 (General Control Non-repressed 5) or PCAF (p300/CBP Associated Factor) as HAT and subsequent studies showed that ADA3 associates with another HAT, p300. As a HAT interacting protein, ADA3 enhances the acetylation of histones as well as non-histone proteins, such as p53. We have recently shown that ADA3 is a cell cycle regulatory protein and is important for both G1 to S phase transition as well as mitosis. Conditional deletion of Ada3 from Ada3FL/FL MEFs (Mouse Embryonic Fibroblasts) causes cell cycle arrest and severe mitotic defects. To further explore the mechanism of ADA3 mediated mitosis, we performed ChIP-seq analyses and found that ADA3 bound to higher order repeat region of the centromere across most of the chromosomes. Further studies showed ADA3 interaction with centromere was mediated by a centromeric protein CENP-B. More importantly, siRNA mediated knockdown of ADA3 decreases the occupancy of CENP-B at centromere and causes chromosomal segregation defects during mitosis. These studies demonstrate a novel function of ADA3 in cell cycle regulation. Our current studies are focused on defining the exact mechanism of recruitment of ADA3 complex to CENP-B to regulate mitosis and its effect on genomic stability. Citation Format: Shashank Srivastava, Shakur Mohibi, June Wang-France, Sameer Mirza, Xiangshan Zhao, Hamid Band, Vimla Bnad. ADA3, a cell cycle regulator, regulates chromosome segregation. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2719.

Collaboration


Dive into the Shakur Mohibi's collaboration.

Top Co-Authors

Avatar

Hamid Band

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Vimla Band

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Sameer Mirza

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Aditya Bele

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Channabasavaiah B. Gurumurthy

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Xiangshan Zhao

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Shashank Srivastava

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Bryan J. Katafiasz

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jun Wang

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Alaa Alshareeda

Nottingham University Hospitals NHS Trust

View shared research outputs
Researchain Logo
Decentralizing Knowledge