22 results on '"Sunil K. Malonia"'
Search Results
2. Protocol for monitoring phagocytosis of cancer cells by TAM-like macrophages using imaging cytometry
- Author
-
Alok K. Mishra, Shahid Banday, Ritesh P. Thakare, Sunil K. Malonia, and Michael R. Green
- Subjects
Cell Biology ,Cell culture ,Cancer ,Immunology ,Microscopy ,Biotechnology and bioengineering ,Science (General) ,Q1-390 - Abstract
Summary: Here, we present a protocol for monitoring phagocytosis by M2-type macrophages using automated counting of phagocytic events with an imaging cytometer. We describe steps for isolating and differentiating peripheral blood mononuclear cell (PBMC)-derived monocytes into M2-like macrophages, preparing cancer cells expressing a green fluorescence marker, labeling with a pH-sensitive dye, and co-culturing with macrophages. We then outline procedures for enumerating phagocytic events using an imaging cytometer.For complete details on the use and execution of this protocol, please refer to Mishra et al.1 : Publisher’s note: Undertaking any experimental protocol requires adherence to local institutional guidelines for laboratory safety and ethics.
- Published
- 2024
- Full Text
- View/download PDF
3. Identification of WNK1 as a therapeutic target to suppress IgH/MYC expression in multiple myeloma
- Author
-
Tianyi Ye, Alok K. Mishra, Shahid Banday, Rui Li, Kai Hu, Madison M. Coleman, Yi Shan, Shreya Roy Chowdhury, Lin Zhou, Magnolia L. Pak, Tessa M. Simone, Sunil K. Malonia, Lihua Julie Zhu, Michelle A. Kelliher, and Michael R. Green
- Subjects
CP: Cancer ,Biology (General) ,QH301-705.5 - Abstract
Summary: Multiple myeloma (MM) remains an incurable hematological malignancy demanding innovative therapeutic strategies. Targeting MYC, the notorious yet traditionally undruggable oncogene, presents an appealing avenue. Here, using a genome-scale CRISPR-Cas9 screen, we identify the WNK lysine-deficient protein kinase 1 (WNK1) as a regulator of MYC expression in MM cells. Genetic and pharmacological inhibition of WNK1 reduces MYC expression and, further, disrupts the MYC-dependent transcriptional program. Mechanistically, WNK1 inhibition attenuates the activity of the immunoglobulin heavy chain (IgH) enhancer, thus reducing MYC transcription when this locus is translocated near the MYC locus. WNK1 inhibition profoundly impacts MM cell behaviors, leading to growth inhibition, cell-cycle arrest, senescence, and apoptosis. Importantly, the WNK inhibitor WNK463 inhibits MM growth in primary patient samples as well as xenograft mouse models and exhibits synergistic effects with various anti-MM compounds. Collectively, our study uncovers WNK1 as a potential therapeutic target in MM.
- Published
- 2024
- Full Text
- View/download PDF
4. Targeting the GPI transamidase subunit GPAA1 abrogates the CD24 immune checkpoint in ovarian cancer
- Author
-
Alok K. Mishra, Tianyi Ye, Shahid Banday, Ritesh P. Thakare, Chinh Tran-To Su, Ngoc N.H. Pham, Amjad Ali, Ankur Kulshreshtha, Shreya Roy Chowdhury, Tessa M. Simone, Kai Hu, Lihua Julie Zhu, Birgit Eisenhaber, Sara K. Deibler, Karl Simin, Paul R. Thompson, Michelle A. Kelliher, Frank Eisenhaber, Sunil K. Malonia, and Michael R. Green
- Subjects
CP: Cancer ,Biology (General) ,QH301-705.5 - Abstract
Summary: CD24 is frequently overexpressed in ovarian cancer and promotes immune evasion by interacting with its receptor Siglec10, present on tumor-associated macrophages, providing a “don’t eat me” signal that prevents targeting and phagocytosis by macrophages. Factors promoting CD24 expression could represent novel immunotherapeutic targets for ovarian cancer. Here, using a genome-wide CRISPR knockout screen, we identify GPAA1 (glycosylphosphatidylinositol anchor attachment 1), a factor that catalyzes the attachment of a glycosylphosphatidylinositol (GPI) lipid anchor to substrate proteins, as a positive regulator of CD24 cell surface expression. Genetic ablation of GPAA1 abolishes CD24 cell surface expression, enhances macrophage-mediated phagocytosis, and inhibits ovarian tumor growth in mice. GPAA1 shares structural similarities with aminopeptidases. Consequently, we show that bestatin, a clinically advanced aminopeptidase inhibitor, binds to GPAA1 and blocks GPI attachment, resulting in reduced CD24 cell surface expression, increased macrophage-mediated phagocytosis, and suppressed growth of ovarian tumors. Our study highlights the potential of targeting GPAA1 as an immunotherapeutic approach for CD24+ ovarian cancers.
- Published
- 2024
- Full Text
- View/download PDF
5. Correction: Satam et al. Next-Generation Sequencing Technology: Current Trends and Advancements. Biology 2023, 12, 997
- Author
-
Heena Satam, Kandarp Joshi, Upasana Mangrolia, Sanober Waghoo, Gulnaz Zaidi, Shravani Rawool, Ritesh P. Thakare, Shahid Banday, Alok K. Mishra, Gautam Das, and Sunil K. Malonia
- Subjects
n/a ,Biology (General) ,QH301-705.5 - Abstract
We are very thankful to the commentator for pointing out the issues in the review article by Satam et al [...]
- Published
- 2024
- Full Text
- View/download PDF
6. Next-Generation Sequencing Technology: Current Trends and Advancements
- Author
-
Heena Satam, Kandarp Joshi, Upasana Mangrolia, Sanober Waghoo, Gulnaz Zaidi, Shravani Rawool, Ritesh P. Thakare, Shahid Banday, Alok K. Mishra, Gautam Das, and Sunil K. Malonia
- Subjects
next-generation sequencing ,genomics ,microbiome ,molecular diagnostics ,bioinformatics ,Nanopore ,Biology (General) ,QH301-705.5 - Abstract
The advent of next-generation sequencing (NGS) has brought about a paradigm shift in genomics research, offering unparalleled capabilities for analyzing DNA and RNA molecules in a high-throughput and cost-effective manner. This transformative technology has swiftly propelled genomics advancements across diverse domains. NGS allows for the rapid sequencing of millions of DNA fragments simultaneously, providing comprehensive insights into genome structure, genetic variations, gene expression profiles, and epigenetic modifications. The versatility of NGS platforms has expanded the scope of genomics research, facilitating studies on rare genetic diseases, cancer genomics, microbiome analysis, infectious diseases, and population genetics. Moreover, NGS has enabled the development of targeted therapies, precision medicine approaches, and improved diagnostic methods. This review provides an insightful overview of the current trends and recent advancements in NGS technology, highlighting its potential impact on diverse areas of genomic research. Moreover, the review delves into the challenges encountered and future directions of NGS technology, including endeavors to enhance the accuracy and sensitivity of sequencing data, the development of novel algorithms for data analysis, and the pursuit of more efficient, scalable, and cost-effective solutions that lie ahead.
- Published
- 2023
- Full Text
- View/download PDF
7. Macrophages as a Potential Immunotherapeutic Target in Solid Cancers
- Author
-
Alok K. Mishra, Shahid Banday, Ravi Bharadwaj, Amjad Ali, Romana Rashid, Ankur Kulshreshtha, and Sunil K. Malonia
- Subjects
TAMs ,immunotherapy ,inflammation ,prognosis ,metastasis ,phagocytosis ,Medicine - Abstract
The revolution in cancer immunotherapy over the last few decades has resulted in a paradigm shift in the clinical care of cancer. Most of the cancer immunotherapeutic regimens approved so far have relied on modulating the adaptive immune system. In recent years, strategies and approaches targeting the components of innate immunity have become widely recognized for their efficacy in targeting solid cancers. Macrophages are effector cells of the innate immune system, which can play a crucial role in the generation of anti-tumor immunity through their ability to phagocytose cancer cells and present tumor antigens to the cells of adaptive immunity. However, the macrophages that are recruited to the tumor microenvironment predominantly play pro-tumorigenic roles. Several strategies targeting pro-tumorigenic functions and harnessing the anti-tumorigenic properties of macrophages have shown promising results in preclinical studies, and a few of them have also advanced to clinical trials. In this review, we present a comprehensive overview of the pathobiology of TAMs and their role in the progression of solid malignancies. We discuss various mechanisms through which TAMs promote tumor progression, such as inflammation, genomic instability, tumor growth, cancer stem cell formation, angiogenesis, EMT and metastasis, tissue remodeling, and immunosuppression, etc. In addition, we also discuss potential therapeutic strategies for targeting TAMs and explore how macrophages can be used as a tool for next-generation immunotherapy for the treatment of solid malignancies.
- Published
- 2022
- Full Text
- View/download PDF
8. Emerging Trends in Immunotherapy for Cancer
- Author
-
Alok K. Mishra, Amjad Ali, Shubham Dutta, Shahid Banday, and Sunil K. Malonia
- Subjects
FDA ,checkpoint inhibitors ,monoclonal antibody ,bispecific antibody antibody drug-conjugate ,CAR-T ,CAR NK ,Medicine - Abstract
Recent advances in cancer immunology have enabled the discovery of promising immunotherapies for various malignancies that have shifted the cancer treatment paradigm. The innovative research and clinical advancements of immunotherapy approaches have prolonged the survival of patients with relapsed or refractory metastatic cancers. Since the U.S. FDA approved the first immune checkpoint inhibitor in 2011, the field of cancer immunotherapy has grown exponentially. Multiple therapeutic approaches or agents to manipulate different aspects of the immune system are currently in development. These include cancer vaccines, adoptive cell therapies (such as CAR-T or NK cell therapy), monoclonal antibodies, cytokine therapies, oncolytic viruses, and inhibitors targeting immune checkpoints that have demonstrated promising clinical efficacy. Multiple immunotherapeutic approaches have been approved for specific cancer treatments, while others are currently in preclinical and clinical trial stages. Given the success of immunotherapy, there has been a tremendous thrust to improve the clinical efficacy of various agents and strategies implemented so far. Here, we present a comprehensive overview of the development and clinical implementation of various immunotherapy approaches currently being used to treat cancer. We also highlight the latest developments, emerging trends, limitations, and future promises of cancer immunotherapy.
- Published
- 2022
- Full Text
- View/download PDF
9. FBXL20 promotes breast cancer malignancy by inhibiting apoptosis through degradation of PUMA and BAX
- Author
-
Sunil K. Malonia, Asha Patel, Manas Kumar Santra, Sarathkumar Edachery, Shahid Banday, Yashika Agrawal, Praveenkumar Shetty, Avinash Kumar, and Rajesh Kumar Manne
- Subjects
Programmed cell death ,SCF complex ,Apoptosis ,Breast Neoplasms ,immunoprecipitation ,DOXO, doxorubicin ,Biochemistry ,FBXO31 ,oligomerization ,Bcl-2-associated X protein ,comet assay ,Puma ,Proto-Oncogene Proteins ,Humans ,p53 upregulated modulator of apoptosis ,Protein kinase A ,Molecular Biology ,Protein kinase B ,Tissue homeostasis ,bcl-2-Associated X Protein ,PUMA, p53 upregulated modulator of apoptosis ,biology ,Chemistry ,AKT ,F-Box Proteins ,GSK3β ,Cell Biology ,biology.organism_classification ,BAX, BCL2-associated X protein ,HEK293 Cells ,Drug Resistance, Neoplasm ,Cancer cell ,Proteolysis ,biology.protein ,Cancer research ,MCF-7 Cells ,Female ,CPT, camptothecin ,PVDF, polyvinylidenedifluoride ,Apoptosis Regulatory Proteins ,IHC, immunohistochemistry ,Research Article - Abstract
Apoptosis is a programmed cell death that efficiently removes damaged cells to maintain tissue homeostasis. Defect in apoptotic machinery can lead to tumor development, progression, and resistance to chemotherapy. PUMA (p53 upregulated modulator of apoptosis) and BAX (BCL2-associated X protein) are among the most well-known inducers of apoptosis. It has been reported that expression levels of BAX and PUMA are controlled at the posttranslational level by phosphorylation. However, the posttranslational regulation of these proapoptotic proteins remains largely unexplored. In this study, using biochemical, molecular biology, flow cytometric, and immunohistochemistry techniques, we show that PUMA and BAX are the direct target of the F-box protein FBXL20, which restricts their cellular levels. FBXL20 directs the proteasomal degradation of PUMA and BAX in a protein kinase AKT1-dependent manner to promote cancer cell proliferation and tumor growth. Interestingly, inactivation of AKT1 results in activation of another protein kinase GSK3α/β, which facilitates the proteasomal degradation of FBXL20 by another F-box protein, FBXO31. Thus, a switch between two signaling kinases AKT1 and GSK3α/β modulates the functional activity of these proapoptotic regulators, thereby determining cell survival or death. RNAi-mediated ablation of FBXL20 results in increased levels of PUMA as well as BAX, which further enhances the sensitivity of cancer cells to chemotherapeutic drugs. We showed that high level expression of FBXL20 in cancer cells reduces therapeutic drug-induced apoptosis and promotes chemoresistance. Overall, this study highlights the importance of targeting FBXL20 in cancers in conjunction with chemotherapy and may represent a promising anticancer strategy to overcome chemoresistance.
- Published
- 2021
10. F‐box protein FBXO16 functions as a tumor suppressor by attenuating nuclear β‐catenin function
- Author
-
Praveen Kumar Shetty, Biswanath Maity, Srikanth Rapole, Ramanamurthy Boppana, Debasish Paul, Rajesh Kumar Manne, US Dinesh, Sehbanul Islam, Manas Kumar Santra, and Sunil K. Malonia
- Subjects
0301 basic medicine ,proteasomal degradation ,Epithelial-Mesenchymal Transition ,tumor suppressor ,F-box protein ,Pathology and Forensic Medicine ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Ubiquitin ,law ,Cell Line, Tumor ,medicine ,Humans ,Genes, Tumor Suppressor ,Wnt Signaling Pathway ,beta Catenin ,Cell Nucleus ,Original Paper ,biology ,Chemistry ,F-Box Proteins ,Wnt signaling pathway ,Nuclear Proteins ,Cancer ,medicine.disease ,Original Papers ,Wnt signaling ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Proteasome ,FBXO16 ,030220 oncology & carcinogenesis ,Catenin ,Cancer cell ,biology.protein ,Cancer research ,β‐catenin ,Suppressor - Abstract
Aberrant activation of β‐catenin has been implicated in a variety of human diseases, including cancer. In spite of significant progress, the regulation of active Wnt/β‐catenin‐signaling pathways is still poorly understood. In this study, we show that F‐box protein 16 (FBXO16) is a putative tumor suppressor. It is a component of the SCF (SKP1‐Cullin1‐F‐box protein) complex, which targets the nuclear β‐catenin protein to facilitate proteasomal degradation through the 26S proteasome. FBXO16 interacts physically with the C‐terminal domain of β‐catenin and promotes its lysine 48‐linked polyubiquitination. In addition, it inhibits epithelial‐to‐mesenchymal transition (EMT) by attenuating the level of β‐catenin. Therefore, depletion of FBXO16 leads to increased levels of β‐catenin, which then promotes cell invasion, tumor growth, and EMT of cancer cells. Furthermore, FBXO16 and β‐catenin share an inverse correlation of cellular expression in clinical breast cancer patient samples. In summary, we propose that FBXO16 functions as a putative tumor suppressor by forming an SCFFBXO16 complex that targets nuclear β‐catenin in a unique manner for ubiquitination and subsequent proteasomal degradation to prevent malignancy. This work suggests a novel therapeutic strategy against human cancers related to aberrant β‐catenin activation. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
- Published
- 2019
- Full Text
- View/download PDF
11. F-box protein FBXO31 directs degradation of MDM2 to facilitate p53-mediated growth arrest following genotoxic stress
- Author
-
Manas Kumar Santra, Sunil K. Malonia, Michael R. Green, and Parul Dutta
- Subjects
DNA damage ,Ataxia Telangiectasia Mutated Proteins ,Genotoxic Stress ,Biology ,Cell Line ,Ubiquitin ,Proto-Oncogene Proteins c-mdm2 ,Humans ,Phosphorylation ,Multidisciplinary ,F-Box Proteins ,Tumor Suppressor Proteins ,Biological Sciences ,Molecular biology ,Ubiquitin ligase ,enzymes and coenzymes (carbohydrates) ,Proteasome ,Proteolysis ,biology.protein ,Mdm2 ,Tumor Suppressor Protein p53 ,Protein Processing, Post-Translational ,Cell Division ,DNA Damage ,Mutagens - Abstract
The tumor suppressor p53 plays a critical role in maintaining genomic stability. In response to genotoxic stress, p53 levels increase and induce cell-cycle arrest, senescence, or apoptosis, thereby preventing replication of damaged DNA. In unstressed cells, p53 is maintained at a low level. The major negative regulator of p53 is MDM2, an E3 ubiquitin ligase that directly interacts with p53 and promotes its polyubiquitination, leading to the subsequent destruction of p53 by the 26S proteasome. Following DNA damage, MDM2 is degraded rapidly, resulting in increased p53 stability. Because of the important role of MDM2 in modulating p53 function, it is critical to understand how MDM2 levels are regulated. Here we show that the F-box protein FBXO31, a candidate tumor suppressor encoded in 16q24.3 for which there is loss of heterozygosity in various solid tumors, is responsible for promoting MDM2 degradation. Following genotoxic stress, FBXO31 is phosphorylated by the DNA damage serine/threonine kinase ATM, resulting in increased levels of FBXO31. FBXO31 then interacts with and directs the degradation of MDM2, which is dependent on phosphorylation of MDM2 by ATM. FBXO31-mediated loss of MDM2 leads to elevated levels of p53, resulting in growth arrest. In cells depleted of FBXO31, MDM2 is not degraded and p53 levels do not increase following genotoxic stress. Thus, FBXO31 is essential for the classic robust increase in p53 levels following DNA damage.
- Published
- 2015
- Full Text
- View/download PDF
12. Nuclear matrix binding protein SMAR1 regulates T-cell differentiation and allergic airway disease
- Author
-
Bhalchandra Mirlekar, Sunil K. Malonia, A J Badhwar, S V Chemmannur, Samit Chattopadhyay, Neerja Wadhwa, Subeer S. Majumdar, Mehak Gupta, R Bopanna, Balaram Ghosh, and Ulaganathan Mabalirajan
- Subjects
Chromatin Immunoprecipitation ,T cell ,Cellular differentiation ,Immunology ,Blotting, Western ,Fluorescent Antibody Technique ,Cell Cycle Proteins ,Electrophoretic Mobility Shift Assay ,Immunoglobulin E ,Real-Time Polymerase Chain Reaction ,Mice ,Immune system ,Interferon ,Conditional gene knockout ,medicine ,Hypersensitivity ,Immunology and Allergy ,Animals ,Nuclear protein ,Mice, Knockout ,biology ,Nuclear Proteins ,Cell Differentiation ,T-Lymphocytes, Helper-Inducer ,Articles ,Flow Cytometry ,Asthma ,DNA-Binding Proteins ,Mice, Inbred C57BL ,Ovalbumin ,Disease Models, Animal ,medicine.anatomical_structure ,biology.protein ,medicine.drug - Abstract
Asthma is a complex airway allergic disease involving the interplay of various cell types, cytokines, and transcriptional factors. Though many factors contribute to disease etiology, the molecular control of disease phenotype and responsiveness is not well understood. Here we report an essential role of the matrix attachment region (MAR)-binding protein SMAR1 in regulating immune response during allergic airway disease. Conditional knockout of SMAR1 in T cells rendered the mice resistant to eosinophilic airway inflammation against ovalbumin (OVA) allergen with low immunoglobulin E (IgE) and interleukin-5 (IL-5) levels. Moreover, a lower IgE/IgG2a ratio and higher interferon-γ (IFN-γ) response suggested aberrant skewing of T-cell differentiation toward type 1 helper T cell (Th1) response. We show that SMAR1 functions as a negative regulator of Th1 and Th17 differentiation by interacting with two potential and similar MAR regions present on the promoters of T-bet and IL-17. Thus, we present SMAR1 as a regulator of T-cell differentiation that favors the establishment of Th2 cells by modulating Th1 and Th17 responses.
- Published
- 2015
13. Degradation of FBXO31 by APC/C is regulated by AKT- and ATM-mediated phosphorylation
- Author
-
Ganga Sankaran, Srinadh Choppara, Michael R. Green, Manas Kumar Santra, and Sunil K. Malonia
- Subjects
0301 basic medicine ,Proteasome Endopeptidase Complex ,Cdc20 Proteins ,CDC20 ,Ataxia Telangiectasia Mutated Proteins ,Models, Biological ,Anaphase-Promoting Complex-Cyclosome ,03 medical and health sciences ,0302 clinical medicine ,Ubiquitin ,Antigens, CD ,Humans ,Phosphorylation ,RNA, Small Interfering ,Protein kinase B ,Multidisciplinary ,biology ,Chemistry ,Kinase ,F-Box Proteins ,Tumor Suppressor Proteins ,Ubiquitination ,Cell Cycle Checkpoints ,Cell cycle ,Biological Sciences ,Cadherins ,Cell biology ,030104 developmental biology ,HEK293 Cells ,030220 oncology & carcinogenesis ,Gene Knockdown Techniques ,Proteolysis ,biology.protein ,Mdm2 ,Proto-Oncogene Proteins c-akt ,Cullin ,DNA Damage - Abstract
The F-box protein FBXO31 is a tumor suppressor that is encoded in 16q24.3, for which there is loss of heterozygosity in various solid tumors. FBXO31 serves as the substrate-recognition component of the SKP/Cullin/F-box protein class of E3 ubiquitin ligases and has been shown to direct degradation of pivotal cell-cycle regulatory proteins including cyclin D1 and the p53 antagonist MDM2. FBXO31 levels are normally low but increase substantially following genotoxic stress through a mechanism that remains to be determined. Here we show that the low levels of FBXO31 are maintained through proteasomal degradation by anaphase-promoting complex/cyclosome (APC/C). We find that the APC/C coactivators CDH1 and CDC20 bind to a destruction-box (D-box) motif present in FBXO31 to promote its polyubiquitination and degradation in a cell-cycle-regulated manner, which requires phosphorylation of FBXO31 on serine-33 by the prosurvival kinase AKT. Following genotoxic stress, phosphorylation of FBXO31 on serine-278 by another kinase, the DNA damage kinase ATM, results in disruption of its interaction with CDH1 and CDC20, thereby preventing FBXO31 degradation. Collectively, our results reveal how alterations in FBXO31 phosphorylation, mediated by AKT and ATM, underlie physiological regulation of FBXO31 levels in unstressed and genotoxically stressed cells.
- Published
- 2018
14. Genetic disruption of oncogenic Kras sensitizes lung cancer cells to Fas receptor-mediated apoptosis
- Author
-
Yingxiang Li, Jill Moore, Soren Hough, Michael R. Green, Zhiping Weng, Deniz M. Ozata, Chun-Qing Song, Haiwei Mou, Jordan L. Smith, Andrew H. Fischer, Sunil K. Malonia, and Wen Xue
- Subjects
0301 basic medicine ,Lung Neoplasms ,endocrine system diseases ,medicine.drug_class ,Cell Survival ,Cell ,Biology ,medicine.disease_cause ,Antibodies ,Epigenesis, Genetic ,Proto-Oncogene Proteins p21(ras) ,03 medical and health sciences ,Gene Knockout Techniques ,Mice ,RNA interference ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Epigenetics ,fas Receptor ,Promoter Regions, Genetic ,neoplasms ,Multidisciplinary ,Gene Expression Profiling ,Histone deacetylase inhibitor ,Biological Sciences ,Fas receptor ,digestive system diseases ,respiratory tract diseases ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,medicine.anatomical_structure ,Apoptosis ,Cancer cell ,Mutation ,Cancer research ,KRAS ,Neoplasm Transplantation - Abstract
Genetic lesions that activate KRAS account for ∼30% of the 1.6 million annual cases of lung cancer. Despite clinical need, KRAS is still undruggable using traditional small-molecule drugs/inhibitors. When oncogenic Kras is suppressed by RNA interference, tumors initially regress but eventually recur and proliferate despite suppression of Kras Here, we show that tumor cells can survive knockout of oncogenic Kras, indicating the existence of Kras-independent survival pathways. Thus, even if clinical KRAS inhibitors were available, resistance would remain an obstacle to treatment. Kras-independent cancer cells exhibit decreased colony formation in vitro but retain the ability to form tumors in mice. Comparing the transcriptomes of oncogenic Kras cells and Kras knockout cells, we identified 603 genes that were specifically up-regulated in Kras knockout cells, including the Fas gene, which encodes a cell surface death receptor involved in physiological regulation of apoptosis. Antibodies recognizing Fas receptor efficiently induced apoptosis of Kras knockout cells but not oncogenic Kras-expressing cells. Increased Fas expression in Kras knockout cells was attributed to decreased association of repressive epigenetic marks at the Fas promoter. Concordant with this observation, treating oncogenic Kras cells with histone deacetylase inhibitor and Fas-activating antibody efficiently induced apoptosis, thus bypassing the need to inhibit Kras. Our results suggest that activation of Fas could be exploited as an Achilles' heel in tumors initiated by oncogenic Kras.
- Published
- 2017
15. Oncogenic RAS directs silencing of tumor suppressor genes through ordered recruitment of transcriptional repressors
- Author
-
Michael R. Green, Rajendra Kumar Palakurthy, Narendra Wajapeyee, and Sunil K. Malonia
- Subjects
Repressor ,Biology ,Models, Biological ,DNA methyltransferase ,Epigenesis, Genetic ,law.invention ,Mice ,Research Communication ,RNA interference ,law ,Genetics ,Animals ,Gene silencing ,Genes, Tumor Suppressor ,Gene Silencing ,fas Receptor ,DNA Methylation ,Gene Expression Regulation, Neoplastic ,DNA methylation ,NIH 3T3 Cells ,ras Proteins ,Cancer research ,DNMT1 ,Suppressor ,RNA Interference ,Signal transduction ,Protein Binding ,Signal Transduction ,Developmental Biology - Abstract
We previously identified 28 cofactors through which a RAS oncoprotein directs transcriptional silencing of Fas and other tumor suppressor genes (TSGs). Here we performed RNAi-based epistasis experiments and found that RAS-directed silencing occurs through a highly ordered pathway that is initiated by binding of ZFP354B, a sequence-specific DNA-binding protein, and culminates in recruitment of the DNA methyltransferase DNMT1. RNAi and pharmacological inhibition experiments reveal that silencing requires continuous function of RAS and its cofactors and can be rapidly reversed, which may have therapeutic implications for reactivation of silenced TSGs in RAS-positive cancers.
- Published
- 2013
- Full Text
- View/download PDF
16. Chromatin remodelling protein SMAR1 inhibits p53 dependent transactivation by regulating acetyl transferase p300
- Author
-
Samit Chattopadhyay, Jayanta K. Pal, Surajit Sinha, Jinumary Mathai, Smriti P.K. Mittal, and Sunil K. Malonia
- Subjects
Transcriptional Activation ,DNA damage ,Lactacystin ,Apoptosis ,Cell Cycle Proteins ,Biology ,Transfection ,Biochemistry ,chemistry.chemical_compound ,Transactivation ,MG132 ,Humans ,p300-CBP Transcription Factors ,RNA, Small Interfering ,Gene knockdown ,Ubiquitination ,Nuclear Proteins ,Acetylation ,Cell Biology ,Chromatin Assembly and Disassembly ,HCT116 Cells ,Chromatin ,DNA-Binding Proteins ,chemistry ,Cancer research ,Ectopic expression ,Tumor Suppressor Protein p53 ,DNA Damage ,Signal Transduction - Abstract
Acetylation of p53 is indispensable for its transcriptional activities and induction of apoptosis upon DNA damage. Here, we show that chromatin remodelling protein SMAR1 inhibits p53 acetylation and p53 dependent apoptosis by repressing p300 expression in response to DNA damage. The repression of p300 expression by SMAR1 is relieved upon treatment with proteosomal inhibitors MG132 and Lactacystin. We demonstrate that SMAR1 interacts with p53–p300 transcriptional complex and SMAR1 overexpression antagonizes p300 interaction with p53 and suppresses activation of p53 apoptotic targets and p53 regulated miRNA miR-34a. Conversely, knockdown of SMAR1 promotes p300 accumulation and p53 acetylation while ectopic expression of p300 rescues SMAR1 inhibition on p53. Collectively, these results indicate that SMAR1 is an important player in p300–p53 regulated DNA damage signalling pathway and can exert its effect on apoptosis in a transcription independent manner.
- Published
- 2012
- Full Text
- View/download PDF
17. Tumor Necrosis Factor alpha (TNFα) regulates CD40 expression through SMAR1 phosphorylation
- Author
-
Samit Chattopadhyay, Surajit Sinha, Sunil K. Malonia, and Kamini Singh
- Subjects
Cytoplasm ,Biophysics ,Repressor ,Cell Cycle Proteins ,Biology ,Biochemistry ,Cell Line ,Phosphorylation cascade ,Mice ,Cyclin D1 ,Serine ,Animals ,STAT1 ,CD40 Antigens ,Phosphorylation ,Promoter Regions, Genetic ,Molecular Biology ,Janus Kinases ,Regulation of gene expression ,General transcription factor ,Tumor Necrosis Factor-alpha ,Nuclear Proteins ,Promoter ,Cell Biology ,DNA-Binding Proteins ,Protein Transport ,STAT1 Transcription Factor ,Gene Expression Regulation ,Cancer research ,biology.protein - Abstract
CD40 plays an important role in mediating inflammatory response and is mainly induced by JAK/STAT phosphorylation cascade. TNFalpha is the key cytokine that activates CD40 during inflammation and tumorigenesis. We have earlier shown that SMAR1 can repress the transcription of Cyclin D1 promoter by forming a HDAC1 dependent repressor complex. In this study, we show that SMAR1 regulates the transcription of NF-kappaB target gene CD40. SMAR1 recruits HDAC1 and forms a repressor complex on CD40 promoter and keeps its basal transcription in check. Further, we show that TNFalpha stimulation induces SMAR1 phosphorylation at Ser-347 and promotes its cytoplasmic translocation, thus releasing its negative effect. Concomitantly, TNFalpha induced phosphorylation of STAT1 at Tyr-701 by JAK1 facilitates its nuclear translocation and activation of CD40 through p300 recruitment and core Histone-3 acetylation. Thus, TNFalpha mediated regulation of CD40 expression occurs by dual phosphorylation of SMAR1 and STAT1.
- Published
- 2010
- Full Text
- View/download PDF
18. Tumor Suppressor SMAR1 Represses IκBα Expression and Inhibits p65 Transactivation through Matrix Attachment Regions
- Author
-
Kamini Singh, Samit Chattopadhyay, Sunil K. Malonia, Pradeep Bist, Vinay Tergaonkar, and Surajit Sinha
- Subjects
Transcriptional Activation ,Transcription, Genetic ,Molecular Sequence Data ,Down-Regulation ,Cell Cycle Proteins ,Histone Deacetylase 1 ,Biology ,medicine.disease_cause ,Biochemistry ,Histone Deacetylases ,law.invention ,Mice ,Transactivation ,law ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Promoter Regions, Genetic ,Scaffold/matrix attachment region ,Molecular Biology ,Binding Sites ,Tumor Necrosis Factor-alpha ,Transcription Factor RelA ,Nuclear Proteins ,DNA ,Cell Biology ,Molecular biology ,I-kappa B Kinase ,Cell biology ,DNA-Binding Proteins ,IκBα ,Phosphorylation ,Suppressor ,Tumor necrosis factor alpha ,Protein Multimerization ,Carcinogenesis ,Corepressor ,Protein Binding - Abstract
Aberrant NF-kappaB activity promotes tumorigenesis. However, NF-kappaB also inhibits tumor growth where tumor suppressor pathways remain unaltered. Thus, its role in tumorigenesis depends upon the function of other cellular factors. Tumor suppressor SMAR1 down-modulated in high grade breast cancers is regulated by p53 and is reported to interact and stabilize p53. Because both SMAR1 and NF-kappaB are involved in tumorigenesis, we investigated the effect of SMAR1 upon NF-kappaB activity. We show that SMAR1 induction by doxorubicin or overexpression produces functional NF-kappaB complexes that are competent for binding to NF-kappaB consensus sequence. However, SMAR1 induced p65-p50 complex is phosphorylation- and transactivation-deficient. Induction of functional NF-kappaB complexes stems from down-regulation of IkappaBalpha transcription through direct binding of SMAR1 to the matrix attachment region site present in IkappaBalpha promoter and recruitment of corepressor complex. Real time PCR array for NF-kappaB target genes revealed that SMAR1 down-regulates a subset of NF-kappaB target genes that are involved in tumorigenesis. We also show that SMAR1 inhibits tumor necrosis factor alpha-induced induction of NF-kappaB suggesting that activation of NF-kappaB by SMAR1 is independent and different from classical pathway. Thus, for the first time we report that a tumor suppressor protein SMAR1 can modulate NF-kappaB transactivation and inhibit tumorigenesis by regulating NF-kappaB target genes.
- Published
- 2009
- Full Text
- View/download PDF
19. Abstract 1028: Genetic disruption of Kras sensitizes lung cancer cells to Fas-mediated apoptosis
- Author
-
Yingxiang Li, Jill Moore, Andrew T. Fisher, Michael R. Green, Deniz M. Ozata, Haiwei Mou, Jordan L. Smith, Shipping Weng, Chun-Qing Song, Sunil K. Malonia, Wen Xue, Daniel G. Anderson, Has Yin, and Soren Hough
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,medicine.disease_cause ,medicine.disease ,Fas mediated apoptosis ,Internal medicine ,Cancer research ,Medicine ,KRAS ,business ,Lung cancer - Abstract
Genetic lesions that activate KRAS account for ~30% of the 1.6 million annual cases of lung cancer. Despite clinical need, KRAS is still undruggable using traditional small molecule drugs/inhibitors. When oncogenic Kras is suppressed by RNA interference, tumors initially regress but eventually recur and proliferate despite suppression of Kras. Here we show that tumor cells can survive CRISPR-mediated knockout of oncogenic Kras, indicating the existence of Kras-independent survival pathways. Thus even if clinical KRAS inhibitors were available, resistance would remain an obstacle to treatment. Kras-independent cancer cells exhibit decreased colony formation in vitro but retain the ability to form tumors in mice. Comparing the transcriptomes of oncogenic Kras cells and Kras knockout cells, we identified 603 genes that were specifically upregulated in Kras knockout cells, including the Fas gene, which encodes a cell surface death receptor involved in physiological regulation of apoptosis. Antibodies recognizing Fas-receptor efficiently induced apoptosis of Kras knockout cells but not oncogenic Kras expressing cells. Increased Fas expression in Kras knockout cells was attributed to decreased association of repressive epigenetic marks at the Fas promoter. Concordant with this observation, treating oncogenic Kras cells with histone-deacetylase inhibitor and Fas-activating antibody efficiently induced apoptosis, thus bypassing the need to inhibit Kras. Our results suggest that activation of Fas could be exploited as an Achilles’ heel in tumors initiated by oncogenic Kras. Citation Format: Haiwei Mou, Jill Moore, Sunil Malonia, Yingxiang Li, Deniz Ozata, Soren Hough, Chunqing Song, Jordan Smith, Has Yin, Andrew Fisher, Daniel Anderson, Shipping Weng, Michael Green, Wen Xue. Genetic disruption of Kras sensitizes lung cancer cells to Fas-mediated apoptosis [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 1028. doi:10.1158/1538-7445.AM2017-1028
- Published
- 2017
- Full Text
- View/download PDF
20. Chromatin remodeling protein SMAR1 regulates NF-κB dependent Interleukin-8 transcription in breast cancer
- Author
-
Bhawna Yadav, Sunil K. Malonia, Surajit Sinha, Gwendel Lazennec, Samit Chattopadhyay, Cellules Souches, Plasticité Cellulaire, Médecine Régénératrice et Immunothérapies (IRMB), Université de Montpellier (UM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre Hospitalier Régional Universitaire [Montpellier] (CHRU Montpellier), Sys2Diag-Modélisation et Ingénierie des Systèmes Complexes Biologiques pour le Diagnostic (Sys2Diag), Centre National de la Recherche Scientifique (CNRS)-Alcediag, and Thomas Jefferson National Accelerator Facility (Jefferson Lab)
- Subjects
Transcription, Genetic ,MESH: NF-kappa B ,Cell Cycle Proteins ,Histone Deacetylase 1 ,Biochemistry ,NF-κB ,Transactivation ,chemistry.chemical_compound ,0302 clinical medicine ,MESH: Transcription Factor RelA ,MESH: Reverse Transcriptase Polymerase Chain Reaction ,Transcriptional regulation ,Promoter Regions, Genetic ,0303 health sciences ,MESH: Immunoblotting ,Reverse Transcriptase Polymerase Chain Reaction ,NF-kappa B ,Nuclear Proteins ,Acetylation ,MESH: Gene Expression Regulation, Neoplastic ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,MESH: MCF-7 Cells ,030220 oncology & carcinogenesis ,MCF-7 Cells ,RNA Interference ,Transcription ,SMAR1 ,MESH: Acetylation ,Protein Binding ,MESH: Cell Line, Tumor ,Immunoblotting ,MESH: RNA Interference ,Breast Neoplasms ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Biology ,Chromatin remodeling ,03 medical and health sciences ,MESH: Cell Cycle Proteins ,Cell Line, Tumor ,MESH: Promoter Regions, Genetic ,Humans ,MESH: Protein Binding ,Interleukin 8 ,Scaffold/matrix attachment region ,030304 developmental biology ,MESH: Humans ,MESH: Transcription, Genetic ,Interleukin-8 ,Transcription Factor RelA ,MESH: Chromatin Assembly and Disassembly ,Cell Biology ,Chromatin Assembly and Disassembly ,HDAC1 ,MESH: Interleukin-8 ,IκBα ,chemistry ,Cancer research ,Repressor ,MESH: Histone Deacetylase 1 ,Chromatin immunoprecipitation ,MESH: Nuclear Proteins ,MESH: Breast Neoplasms ,MESH: DNA-Binding Proteins - Abstract
International audience; Interleukin-8 (IL-8) is a pleiotropic chemokine involved in metastasis and angiogenesis of breast tumors. The expression of IL-8 is deregulated in metastatic breast carcinomas owing to aberrant NF-κB activity, which is known to positively regulate IL-8 transcription. Earlier, we have shown that tumor suppressor SMAR1 suppresses NF-κB transcriptional activity by modulating IκBα function. Here, we show that NF-κB target gene IL-8, is a direct transcriptional target of SMAR1. Using chromatin immunoprecipitation and reporter assays, we demonstrate that SMAR1 binds to IL-8 promoter MAR (matrix attachment region) and recruits HDAC1 dependent co-repressor complex. Further, we also show that SMAR1 antagonizes p300-mediated acetylation of RelA/p65, a post-translational modification indispensable for IL-8 transactivation. Thus, we decipher a new role of SMAR1 in NF-κB dependent transcriptional regulation of pro-angiogenic chemokine IL-8.
- Published
- 2014
- Full Text
- View/download PDF
21. Gene regulation by SMAR1: Role in cellular homeostasis and cancer
- Author
-
Samit Chattopadhyay, Sunil K. Malonia, Kamini Singh, Shravanti Rampalli, Pavithra Lakshminarasimhan, Ruchika Kaul-Ghanekar, Archana Jalota-Badhwar, and Surajit Sinha
- Subjects
Cancer Research ,Cellular homeostasis ,Apoptosis ,Cell Cycle Proteins ,Biology ,medicine.disease_cause ,Transforming Growth Factor beta ,Neoplasms ,Gene expression ,Genetics ,medicine ,Animals ,Homeostasis ,Humans ,Neoplasm Invasiveness ,Scaffold/matrix attachment region ,Regulation of gene expression ,Cell Cycle ,Nuclear Proteins ,Nuclear matrix ,Genes, bcl-1 ,Cell biology ,Chromatin ,DNA-Binding Proteins ,Oncology ,Gene Expression Regulation ,Signal transduction ,Carcinogenesis ,Signal Transduction - Abstract
Changes in the composition of nuclear matrix associated proteins contribute to alterations in nuclear structure, one of the major phenotypes of malignant cancer cells. The malignancy-induced changes in this structure lead to alterations in chromatin folding, the fidelity of genome replication and gene expression programs. The nuclear matrix forms a scaffold upon which the chromatin is organized into periodic loop domains called matrix attachment regions (MAR) by binding to various MAR binding proteins (MARBPs). Aberrant expression of MARBPs modulates the chromatin organization and disrupt transcriptional network that leads to oncogenesis. Dysregulation of nuclear matrix associated MARBPs has been reported in different types of cancers. Some of these proteins have tumor specific expression and are therefore considered as promising diagnostic or prognostic markers in few cancers. SMAR1 (scaffold/matrix attachment region binding protein 1), is one such nuclear matrix associated protein whose expression is drastically reduced in higher grades of breast cancer. SMAR1 gene is located on human chromosome 16q24.3 locus, the loss of heterozygosity (LOH) of which has been reported in several types of cancers. This review elaborates on the multiple roles of nuclear matrix associated protein SMAR1 in regulating various cellular target genes involved in cell growth, apoptosis and tumorigenesis.
- Published
- 2010
22. Comparative evaluation of Löwenstein-Jensen proportion method, BacT/ALERT 3D system, and enzymatic pyrazinamidase assay for pyrazinamide susceptibility testing of Mycobacterium tuberculosis
- Author
-
G.P. Jadaun, Abhay Kumar Mishra, PK Upadhyay, Unnati Gupta, Vishwas Sharma, Clement Wesley, V. M. Katoch, Pushpendra Singh, Kalpana Singh, Rathindra Nath Das, P.S.P. Gupta, Pushp Sharma, Sunil K. Malonia, Jaya Faujdar, Krishnamurthy Venkatesan, and Durg Singh Chauhan
- Subjects
Microbiology (medical) ,Susceptibility testing ,Antitubercular Agents ,Microbial Sensitivity Tests ,Sensitivity and Specificity ,Comparative evaluation ,Microbiology ,Amidohydrolases ,Mycobacterium tuberculosis ,Predictive Value of Tests ,Medicine ,Humans ,Antibacterial agent ,chemistry.chemical_classification ,biology ,business.industry ,Bact alert ,Reproducibility of Results ,Mycobacteriology and Aerobic Actinomycetes ,Gold standard (test) ,Pyrazinamide ,biology.organism_classification ,Culture Media ,Enzyme ,chemistry ,business ,medicine.drug - Abstract
Pyrazinamide (PZA) is an important first-line antituberculosis drug because of its sterilizing activity against semidormant tubercle bacilli. In spite of its very high in vivo activity, its in vitro activity is not apparent unless an acidic environment is available, which makes PZA susceptibility testing difficult by conventional methods. The present study was, therefore, planned to assess the performance of the colorimetric BacT/ALERT 3D system and compare the results with those from conventional tests, i.e., the Löwenstein-Jensen (LJ) proportion method (pH 4.85) and Wayne's pyrazinamidase (PZase) assay, using 107 clinical isolates. The concordance among all of these tests was 89.71% after the first round of testing and reached 92.52% after resolution of the discordant results by retesting. Prolonged incubation of the PZase tube for up to 10 days was found to increase the specificity of the PZase test. The concordances between LJ proportion and BacT/ALERT 3D, LJ proportion and the PZase assay, and BacT/ALERT 3D and the PZase assay were found to be 99.06%, 93.46%, and 92.52%, respectively. Using the LJ results as the gold standard, the sensitivities of BacT/ALERT 3D and the PZase assay were 100 and 82.85%, respectively, while the specificity was 98.61% for both of the tests. The difference between the sensitivities of BacT/ALERT 3D and the PZase assay was significant ( P = 0.025). The mean turnaround times for the detection of resistant and susceptible results by BacT/ALERT 3D were 8.04 and 11.32 days, respectively. While the major limitations associated with the PZase assay and the LJ proportion method are lower sensitivity in previously treated patients and a longer time requirement, respectively, the BacT/ALERT 3D system was found to be rapid, highly sensitive, and specific.
- Published
- 2006
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.