46 results on '"Ande Satyanarayana"'
Search Results
2. Supplemental material and methods from Nrf2 Drives Hepatocellular Carcinoma Progression through Acetyl-CoA–Mediated Metabolic and Epigenetic Regulatory Networks
- Author
-
Xi, Caixia, primary, Pang, Junfeng, primary, Barrett, Amanda, primary, Horuzsko, Anatolij, primary, Ande, Satyanarayana, primary, Mivechi, Nahid F., primary, and Zhu, Xingguo, primary
- Published
- 2023
- Full Text
- View/download PDF
3. Supplementary Figure 7 from Nrf2 Drives Hepatocellular Carcinoma Progression through Acetyl-CoA–Mediated Metabolic and Epigenetic Regulatory Networks
- Author
-
Xi, Caixia, primary, Pang, Junfeng, primary, Barrett, Amanda, primary, Horuzsko, Anatolij, primary, Ande, Satyanarayana, primary, Mivechi, Nahid F., primary, and Zhu, Xingguo, primary
- Published
- 2023
- Full Text
- View/download PDF
4. Data from Nrf2 Drives Hepatocellular Carcinoma Progression through Acetyl-CoA–Mediated Metabolic and Epigenetic Regulatory Networks
- Author
-
Xi, Caixia, primary, Pang, Junfeng, primary, Barrett, Amanda, primary, Horuzsko, Anatolij, primary, Ande, Satyanarayana, primary, Mivechi, Nahid F., primary, and Zhu, Xingguo, primary
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Figure 8 from Nrf2 Drives Hepatocellular Carcinoma Progression through Acetyl-CoA–Mediated Metabolic and Epigenetic Regulatory Networks
- Author
-
Xi, Caixia, primary, Pang, Junfeng, primary, Barrett, Amanda, primary, Horuzsko, Anatolij, primary, Ande, Satyanarayana, primary, Mivechi, Nahid F., primary, and Zhu, Xingguo, primary
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Figure 1 from Nrf2 Drives Hepatocellular Carcinoma Progression through Acetyl-CoA–Mediated Metabolic and Epigenetic Regulatory Networks
- Author
-
Xi, Caixia, primary, Pang, Junfeng, primary, Barrett, Amanda, primary, Horuzsko, Anatolij, primary, Ande, Satyanarayana, primary, Mivechi, Nahid F., primary, and Zhu, Xingguo, primary
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Figure 3 from Nrf2 Drives Hepatocellular Carcinoma Progression through Acetyl-CoA–Mediated Metabolic and Epigenetic Regulatory Networks
- Author
-
Xi, Caixia, primary, Pang, Junfeng, primary, Barrett, Amanda, primary, Horuzsko, Anatolij, primary, Ande, Satyanarayana, primary, Mivechi, Nahid F., primary, and Zhu, Xingguo, primary
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Figure 4 from Nrf2 Drives Hepatocellular Carcinoma Progression through Acetyl-CoA–Mediated Metabolic and Epigenetic Regulatory Networks
- Author
-
Xi, Caixia, primary, Pang, Junfeng, primary, Barrett, Amanda, primary, Horuzsko, Anatolij, primary, Ande, Satyanarayana, primary, Mivechi, Nahid F., primary, and Zhu, Xingguo, primary
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Figure 2 from Nrf2 Drives Hepatocellular Carcinoma Progression through Acetyl-CoA–Mediated Metabolic and Epigenetic Regulatory Networks
- Author
-
Xi, Caixia, primary, Pang, Junfeng, primary, Barrett, Amanda, primary, Horuzsko, Anatolij, primary, Ande, Satyanarayana, primary, Mivechi, Nahid F., primary, and Zhu, Xingguo, primary
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Figure 5 from Nrf2 Drives Hepatocellular Carcinoma Progression through Acetyl-CoA–Mediated Metabolic and Epigenetic Regulatory Networks
- Author
-
Xi, Caixia, primary, Pang, Junfeng, primary, Barrett, Amanda, primary, Horuzsko, Anatolij, primary, Ande, Satyanarayana, primary, Mivechi, Nahid F., primary, and Zhu, Xingguo, primary
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Figure 6 from Nrf2 Drives Hepatocellular Carcinoma Progression through Acetyl-CoA–Mediated Metabolic and Epigenetic Regulatory Networks
- Author
-
Xi, Caixia, primary, Pang, Junfeng, primary, Barrett, Amanda, primary, Horuzsko, Anatolij, primary, Ande, Satyanarayana, primary, Mivechi, Nahid F., primary, and Zhu, Xingguo, primary
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Figure 9 from Nrf2 Drives Hepatocellular Carcinoma Progression through Acetyl-CoA–Mediated Metabolic and Epigenetic Regulatory Networks
- Author
-
Xi, Caixia, primary, Pang, Junfeng, primary, Barrett, Amanda, primary, Horuzsko, Anatolij, primary, Ande, Satyanarayana, primary, Mivechi, Nahid F., primary, and Zhu, Xingguo, primary
- Published
- 2023
- Full Text
- View/download PDF
13. Supplemental Table 1 from Nrf2 Drives Hepatocellular Carcinoma Progression through Acetyl-CoA–Mediated Metabolic and Epigenetic Regulatory Networks
- Author
-
Xi, Caixia, primary, Pang, Junfeng, primary, Barrett, Amanda, primary, Horuzsko, Anatolij, primary, Ande, Satyanarayana, primary, Mivechi, Nahid F., primary, and Zhu, Xingguo, primary
- Published
- 2023
- Full Text
- View/download PDF
14. The Transcription Factor RXRα in CD11c+ APCs Regulates Intestinal Immune Homeostasis and Inflammation
- Author
-
Manoharan, Indumathi, primary, Shanmugam, Arulkumaran, additional, Ramalingam, Malarvizhi, additional, Patel, Nikhil, additional, Thangaraju, Muthusamy, additional, Ande, Satyanarayana, additional, Pacholczyk, Rafal, additional, Prasad, Puttur D., additional, and Manicassamy, Santhakumar, additional
- Published
- 2023
- Full Text
- View/download PDF
15. Nrf2 Drives Hepatocellular Carcinoma Progression through Acetyl-CoA–Mediated Metabolic and Epigenetic Regulatory Networks
- Author
-
Xi, Caixia, primary, Pang, Junfeng, additional, Barrett, Amanda, additional, Horuzsko, Anatolij, additional, Ande, Satyanarayana, additional, Mivechi, Nahid F., additional, and Zhu, Xingguo, additional
- Published
- 2023
- Full Text
- View/download PDF
16. The Promoter of Human Telomerase Reverse Transcriptase Is Activated During Liver Regeneration and Hepatocyte Proliferation
- Author
-
Sirma, Hüseyin, Kumar, Mukesh, Meena, Jitendra K., Witt, Britta, Weise, Julia M., Lechel, Andre, Ande, Satyanarayana, Sakk, Vadim, Guguen–Guillouzo, Christiane, Zender, Lars, Rudolph, Karl–Lenhard, and Günes, Cagatay
- Published
- 2011
- Full Text
- View/download PDF
17. Molecular Signaling Pathways and Therapeutic Targets in Hepatocellular Carcinoma
- Author
-
Ande Satyanarayana and Manali Dimri
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,signaling pathway ,Cancer Research ,Review ,lcsh:RC254-282 ,Receptor tyrosine kinase ,03 medical and health sciences ,0302 clinical medicine ,Protein kinase A ,Protein kinase B ,PI3K/AKT/mTOR pathway ,biology ,Wnt signaling pathway ,hepatocellular carcinoma ,targeted therapy ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Hedgehog signaling pathway ,digestive system diseases ,3. Good health ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Signal transduction - Abstract
Hepatocellular carcinoma (HCC) is a complex biological process and is often diagnosed at advanced stages with no effective treatment options. With advances in tumor biology and molecular genetic profiling, several different signaling pathways and molecular mechanisms have been identified as responsible for initiating and promoting HCC. Targeting these critical pathways, which include the receptor tyrosine kinase pathways, the Ras mitogen-activated protein kinase (Ras/Raf/MAPK), the phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR), the Wnt/β-catenin signaling pathway, the ubiquitin/proteasome degradation and the hedgehog signaling pathway has led to the identification of novel therapeutics for HCC treatment. In this review, we elaborated on our current understanding of the signaling pathways involved in the development and initiation of HCC and anticipate the potential targets for therapeutic drug development.
- Published
- 2020
18. Id1 Promotes Obesity by Suppressing Brown Adipose Thermogenesis and White Adipose Browning
- Author
-
Ande Satyanarayana, Judith Chang, Shweta Kapil, Sawsan Elattar, Bal Krishan Sharma, Jinling Yuan, and Mallikarjun Patil
- Subjects
Inhibitor of Differentiation Protein 1 ,0301 basic medicine ,medicine.medical_specialty ,Adipose Tissue, White ,Endocrinology, Diabetes and Metabolism ,Adipose tissue ,Mice, Transgenic ,White adipose tissue ,Biology ,Diet, High-Fat ,Real-Time Polymerase Chain Reaction ,Mice ,03 medical and health sciences ,Adipose Tissue, Brown ,Internal medicine ,Coactivator ,Brown adipose tissue ,Basic Helix-Loop-Helix Transcription Factors ,Internal Medicine ,medicine ,Animals ,Adipocytes, Beige ,Obesity ,Uncoupling Protein 1 ,PRDM16 ,Thermogenesis ,Stromal vascular fraction ,Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha ,Thermogenin ,Cold Temperature ,DNA-Binding Proteins ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Gene Expression Regulation ,Obesity Studies ,Transcription Factors - Abstract
Obesity results from increased energy intake or defects in energy expenditure. Brown adipose tissue (BAT) is specialized for energy expenditure, a process called adaptive thermogenesis. Peroxisome proliferator–activated receptor γ coactivator 1α (PGC1α) controls BAT-mediated thermogenesis by regulating the expression of Ucp1. Inhibitor of differentiation 1 (Id1) is a helix-loop-helix transcription factor that plays an important role in cell proliferation and differentiation. We demonstrate a novel function of Id1 in BAT thermogenesis and programming of beige adipocytes in white adipose tissue (WAT). We found that adipose tissue–specific overexpression of Id1 causes age-associated and high-fat diet–induced obesity in mice. Id1 suppresses BAT thermogenesis by binding to and suppressing PGC1α transcriptional activity. In WAT, Id1 is mainly localized in the stromal vascular fraction, where the adipose progenitor/precursors reside. Lack of Id1 increases beige gene and Ucp1 expression in the WAT in response to cold exposure. Furthermore, brown-like differentiation is increased in Id1-deficient mouse embryonic fibroblasts. At the molecular level, Id1 directly interacts with and suppresses Ebf2 transcriptional activity, leading to reduced expression of Prdm16, which determines beige/brown adipocyte cell fate. Overall, the study highlights the existence of novel regulatory mechanisms between Id1/PGC1α and Id1/Ebf2 in controlling brown fat metabolism, which has significant implications in the treatment of obesity and its associated diseases, such as diabetes.
- Published
- 2017
- Full Text
- View/download PDF
19. NAD(P)H Quinone Dehydrogenase 1 Ablation Inhibits Activation of the Phosphoinositide 3-Kinase/Akt Serine/Threonine Kinase and Mitogen-Activated Protein Kinase/Extracellular Signal-Regulated Kinase Pathways and Blocks Metabolic Adaptation in Hepatocellular Carcinoma
- Author
-
Ashley Humphries, Archana Laknaur, Ashok Sharma, Ravindra Kolhe, Manali Dimri, Ande Satyanarayana, Sawsan Elattar, and Tae Jin Lee
- Subjects
Serine/threonine-specific protein kinase ,MAPK/ERK pathway ,Male ,Carcinoma, Hepatocellular ,Hepatology ,biology ,Chemistry ,Kinase ,Liver Neoplasms ,Protein phosphatase 2 ,Article ,Cell biology ,Mice ,Phosphatidylinositol 3-Kinases ,Mitogen-activated protein kinase ,biology.protein ,NAD(P)H Dehydrogenase (Quinone) ,Animals ,Humans ,Protein kinase A ,Extracellular Signal-Regulated MAP Kinases ,Protein kinase B ,Proto-Oncogene Proteins c-akt ,PI3K/AKT/mTOR pathway ,Signal Transduction - Abstract
Cancer cells undergo metabolic adaptation to sustain uncontrolled proliferation. Aerobic glycolysis and glutaminolysis are two of the most essential characteristics of cancer metabolic reprogramming. Hyperactivated phosphoinositide 3-kinase (PI3K)/Akt serine/threonine kinase (Akt) and mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signaling pathways play central roles in cancer cell metabolic adaptation given that their downstream effectors, such as Akt and c-Myc, control most of the glycolytic and glutaminolysis genes. Here, we report that the cytosolic flavoprotein, NAD(P)H quinone dehydrogenase 1 (Nqo1), is strongly overexpressed in mouse and human hepatocellular carcinoma (HCC). Knockdown of Nqo1 enhanced activity of the serine/threonine phosphatase, protein phosphatase 2A, which operates at the intersection of the PI3K/Akt and MAPK/ERK pathways and dephosphorylates and inactivates pyruvate dehydrogenase kinase 1, Akt, Raf, mitogen-activated protein kinase kinase, and ERK1/2. Nqo1 ablation also induced the expression of phosphatase and tensin homolog, a dual protein/lipid phosphatase that blocks PI3K/Akt signaling, through the ERK/cAMP-responsive element-binding protein/c-Jun pathway. Together, Nqo1 ablation triggered simultaneous inhibition of the PI3K/Akt and MAPK/ERK pathways, suppressed the expression of glycolysis and glutaminolysis genes and blocked metabolic adaptation in liver cancer cells. Conversely, Nqo1 overexpression caused hyperactivation of the PI3K/Akt and MAPK/ERK pathways and promoted metabolic adaptation. Conclusion: In conclusion, Nqo1 functions as an upstream activator of both the PI3K/Akt and MAPK/ERK pathways in liver cancer cells, and Nqo1 ablation blocked metabolic adaptation and inhibited liver cancer cell proliferation and HCC growth in mice. Therefore, our results suggest that Nqo1 may function as a therapeutic target to inhibit liver cancer cell proliferation and inhibit HCC.
- Published
- 2018
20. The tumor secretory factor ZAG promotes white adipose tissue browning and energy wasting
- Author
-
Manali Dimri, Ande Satyanarayana, and Sawsan Elattar
- Subjects
0301 basic medicine ,Cachexia ,Adipose Tissue, White ,Adipose tissue ,White adipose tissue ,Zn-Alpha-2-Glycoprotein ,Biochemistry ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Adipose Tissue, Brown ,Neoplasms ,Brown adipose tissue ,Genetics ,medicine ,Lipolysis ,Animals ,Molecular Biology ,Transcription factor ,Uncoupling Protein 1 ,PRDM16 ,Chemistry ,Research ,Seminal Plasma Proteins ,Biological Transport ,Thermogenesis ,Thermogenin ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Energy Metabolism ,Biotechnology ,Transcription Factors - Abstract
Cachexia is a complex tissue-wasting syndrome characterized by inflammation, hypermetabolism, increased energy expenditure, and anorexia. Browning of white adipose tissue (WAT) is one of the significant factors that contribute to energy wasting in cachexia. By utilizing a cell implantation model, we demonstrate here that the lipid mobilizing factor zinc-α(2)-glycoprotein (ZAG) induces WAT browning in mice. Increased circulating levels of ZAG not only induced lipolysis in adipose tissues but also caused robust browning in WAT. Stimulating WAT progenitors with ZAG recombinant protein or expression of ZAG in mouse embryonic fibroblasts (MEFs) strongly enhanced brown-like differentiation. At the molecular level, ZAG stimulated peroxisome proliferator–activated receptor γ (PPARγ) and early B cell factor 2 expression and promoted their recruitment to the PR/SET domain 16 (Prdm16) promoter, leading to enhanced expression of Prdm16, which determines brown cell fate. In brown adipose tissue, ZAG stimulated the expression of PPARγ and PPARγ coactivator 1α and promoted recruitment of PPARγ to the uncoupling protein 1 (Ucp1) promoter, leading to increased expression of Ucp1. Overall, our results reveal a novel function of ZAG in WAT browning and highlight the targeting of ZAG as a potential therapeutic application in humans with cachexia.—Elattar, S., Dimri, M., Satyanarayana, A. The tumor secretory factor ZAG promotes white adipose tissue browning and energy wasting.
- Published
- 2018
21. Inhibitor of differentiation 1 transcription factor promotes metabolic reprogramming in hepatocellular carcinoma cells
- Author
-
Stephen M. Black, Ande Satyanarayana, Ravindra Kolhe, Bal Krishan Sharma, Nahid F. Mivechi, and Jonathan R. Keller
- Subjects
Inhibitor of Differentiation Protein 1 ,Male ,0301 basic medicine ,Carcinoma, Hepatocellular ,Cell ,Biology ,Biochemistry ,Proto-Oncogene Proteins c-myc ,Mice ,Research Communication ,03 medical and health sciences ,Basic Helix-Loop-Helix Transcription Factors ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,Transcription factor ,Feedback, Physiological ,Gene knockdown ,Glutaminolysis ,Oncogene ,Tumor Suppressor Proteins ,Liver Neoplasms ,Cancer ,Hep G2 Cells ,Hypoxia-Inducible Factor 1, alpha Subunit ,medicine.disease ,Cell Hypoxia ,Mice, Inbred C57BL ,Oxygen ,030104 developmental biology ,medicine.anatomical_structure ,Anaerobic glycolysis ,Cancer cell ,Cancer research ,Glycolysis ,Biotechnology - Abstract
Reprograming of metabolism is one of the central hallmarks of cancer. The majority of cancer cells depend on high rates of glycolysis and glutaminolysis for their growth and survival. A number of oncogenes and tumor suppressors have been connected to the regulation of altered glucose and glutamine metabolism in cancer cells. For example, the oncogene c-Myc plays vital roles in cancer cell metabolic adaptation by directly regulating various genes that participate in aerobic glycolysis and glutaminolysis. Inhibitor of differentiation 1 (Id1) is a helix-loop-helix transcription factor that plays important roles in cell proliferation, differentiation, and cell fate determination. Overexpression of Id1 causes intestinal adenomas and thymic lymphomas in mice, suggesting that Id1 could function as an oncogene. Despite it being an oncogene, whether Id1 plays any prominent role in cancer cell metabolic reprograming is unknown. Here, we demonstrate that Id1 is strongly expressed in human and mouse liver tumors and in hepatocellular carcinoma (HCC) cell lines, whereas its expression is very low or undetectable in normal liver tissues. In HCC cells, Id1 expression is regulated by the MAPK/ERK pathway at the transcriptional level. Knockdown of Id1 suppressed aerobic glycolysis and glutaminolysis, suggesting that Id1 promotes a metabolic shift toward aerobic glycolysis. At the molecular level, Id1 mediates its metabolic effects by regulating the expression levels of c-Myc. Knockdown of Id1 resulted in down-regulation (∼75%) of c-Myc, whereas overexpression of Id1 strongly induced (3-fold) c-Myc levels. Interestingly, knockdown of c-Myc resulted in down-regulation (∼60%) of Id1, suggesting a positive feedback-loop regulatory mechanism between Id1 and c-Myc. Under anaerobic conditions, both Id1 and c-Myc are down-regulated (50–70%), and overexpression of oxygen-insensitive hypoxia-inducible factor 1α (Hif1α) or its downstream target Mxi1 resulted in a significant reduction of c-Myc and Id1 (∼70%), suggesting that Hif1α suppresses Id1 and c-Myc under anaerobic conditions via Mxi1. Together, our findings indicate a prominent novel role for Id1 in liver cancer cell metabolic adaptation.—Sharma, B. K., Kolhe, R., Black, S. M., Keller, J. R., Mivechi, N. F., Satyanarayana, A. Inhibitor of differentiation 1 transcription factor promotes metabolic reprogramming in hepatocellular carcinoma cells.
- Published
- 2015
- Full Text
- View/download PDF
22. Can Brown Fat Win the Battle Against White Fat?
- Author
-
Sawsan Elattar and Ande Satyanarayana
- Subjects
education.field_of_study ,medicine.medical_specialty ,Physiology ,Clinical Biochemistry ,Population ,Energy balance ,Adipose tissue ,Cell Biology ,White adipose tissue ,Biology ,Overweight ,medicine.disease ,Obesity ,medicine.anatomical_structure ,Endocrinology ,Internal medicine ,Brown adipose tissue ,medicine ,medicine.symptom ,education ,Thermogenesis - Abstract
A rapid growth in the overweight and obese population in the last few decades suggest that the current diet, exercise, awareness or drug strategies are still not effectively restraining the obesity epidemic. Obesity results from increased energy intake, and the body's energy balance shifts towards energy abundance. Therefore, current research is focused on developing new strategies aimed at increasing energy expenditure. As a result, brown adipose tissue (BAT) is receiving tremendous attention since the major function of BAT is to dissipate energy as heat. For example, mouse models that have increased BAT activity or increased numbers of brown-like adipocytes within the white adipose tissue (WAT) are lean and protected from obesity. Alternatively, mouse models that lack BAT activity are more susceptible to age and diet-induced obesity. However, a significant loss of BAT mass during the natural growth process in humans has created enormous challenges in effectively utilizing this tissue to increase energy expenditure. New strategies are primarily focused on expanding the BAT mass and/or activating the existing BAT. In this regard, recent finding that expression of early B cell factor-2 (Ebf2) reprograms the white pre-adipocytes into brown adipocytes is a significant break-through in developing BAT-mediated strategies to treat obesity. Here we review the major biological functions of WAT and BAT, which play critical but opposing roles in the energy spectrum, energy storage versus energy expenditure, and we evaluate whether activation and/or expansion of BAT is practically achievable to treat obesity in humans.
- Published
- 2015
- Full Text
- View/download PDF
23. The cell polarity protein Scrib functions as a tumor suppressor in liver cancer
- Author
-
Mallikarjun Patil, Shweta Kapil, Bal Krishan Sharma, Steven X. Hou, Sawsan Elattar, Jinling Yuan, Ande Satyanarayana, and Ravindra Kolhe
- Subjects
0301 basic medicine ,Pathology ,Gene Expression ,Molecular oncology ,Mice ,0302 clinical medicine ,Medicine ,hippo signaling ,Cyclin D1 ,HCC ,nuclear localization ,YAP1 ,Cell Cycle ,Liver Neoplasms ,3. Good health ,Protein Transport ,ERK ,Cell Transformation, Neoplastic ,Oncology ,Hippo signaling ,030220 oncology & carcinogenesis ,Heterografts ,Stem cell ,Liver cancer ,Protein Binding ,Signal Transduction ,Research Paper ,SCRIB ,medicine.medical_specialty ,MAP Kinase Signaling System ,Active Transport, Cell Nucleus ,Yap1 ,Protein Serine-Threonine Kinases ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,Cell Line, Tumor ,Animals ,Humans ,Hippo Signaling Pathway ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,business.industry ,Tumor Suppressor Proteins ,Cancer ,Membrane Proteins ,YAP-Signaling Proteins ,medicine.disease ,Phosphoproteins ,Disease Models, Animal ,030104 developmental biology ,Cancer research ,business ,Transcription Factors - Abstract
// Shweta Kapil 1, * , Bal Krishan Sharma 1, * , Mallikarjun Patil 1, * , Sawsan Elattar 1 , Jinling Yuan 1 , Steven X. Hou 2 , Ravindra Kolhe 3 , Ande Satyanarayana 1 1 Department of Biochemistry and Molecular Biology, Molecular Oncology & Biomarkers Program, Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA 2 Stem Cell Regulation and Animal Aging Section, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA 3 Department of Pathology, Augusta University, Augusta, GA 30912, USA * These authors contributed equally to this work Correspondence to: Ande Satyanarayana, email: sande@augusta.edu Keywords: HCC, nuclear localization, ERK, hippo signaling, Yap1 Received: January 13, 2017 Accepted: February 15, 2017 Published: February 24, 2017 ABSTRACT Scrib is a membrane protein that is involved in the maintenance of apical-basal cell polarity of the epithelial tissues. However, Scrib has also been shown to be mislocalized to the cytoplasm in breast and prostate cancer. Here, for the first time, we report that Scrib not only translocates to the cytoplasm but also to the nucleus in hepatocellular carcinoma (HCC) cells, and in mouse and human liver tumor samples. We demonstrate that Scrib overexpression suppresses the growth of HCC cells in vitro, and Scrib deficiency enhances liver tumor growth in vivo. At the molecular level, we have identified the existence of a positive feed-back loop between Yap1 and c-Myc in HCC cells, which Scrib disrupts by simultaneously regulating the MAPK/ERK and Hippo signaling pathways. Overall, Scrib inhibits liver cancer cell proliferation by suppressing the expression of three oncogenes, Yap1, c-Myc and cyclin D1, thereby functioning as a tumor suppressor in liver cancer.
- Published
- 2017
24. Negative Regulators of Brown Adipose Tissue (BAT)-Mediated Thermogenesis
- Author
-
Bal Krishan Sharma, Ande Satyanarayana, and Mallikarjun Patil
- Subjects
medicine.medical_specialty ,animal structures ,Physiology ,Clinical Biochemistry ,Repressor ,Adipose tissue ,Cell Biology ,Biology ,Thermogenin ,Active Bat ,medicine.anatomical_structure ,Endocrinology ,Mechanism of action ,Energy expenditure ,Internal medicine ,Brown adipose tissue ,medicine ,medicine.symptom ,Thermogenesis - Abstract
Brown adipose tissue (BAT) is specialized for energy expenditure, a process called adaptive thermogenesis. PET-CT scans recently demonstrated the existence of metabolically active BAT in adult humans, which revitalized our interest in BAT. Increasing the amount and/or activity of BAT holds tremendous promise for the treatment of obesity and its associated diseases. PGC1α is the master regulator of UCP1-mediated thermogenesis in BAT. A number of proteins have been identified to influence thermogenesis either positively or negatively through regulating the expression or transcriptional activity of PGC1α. Therefore, BAT activation can be achieved by either inducing the expression of positive regulators of PGC1α or by inhibiting the repressors of the PGC1α/UCP1 pathway. Here, we review the most important negative regulators of PGC1α/UCP1 signaling and their mechanism of action in BAT-mediated thermogenesis.
- Published
- 2014
- Full Text
- View/download PDF
25. Abstract 1835: Nqo1 ablation inhibits activation of the PI3K/Akt and MAPK/ERK pathways and blocks metabolic adaptation in hepatocellular carcinoma
- Author
-
Manali Dimri, Ashley Humphries, Archana Laknaur, Sawsan Elattar, Ashok Sharma, Ravindra Kolhe, and Ande Satyanarayana
- Subjects
Cancer Research ,Oncology - Abstract
According to the World Health Organization, hepatocellular carcinoma (HCC) is the 2nd leading cause of cancer related deaths worldwide. HCC is highly resistant to conventional chemotherapies; therefore, identification of specific molecular targets for the development of targeted therapies is indispensable. Cancer cells undergo metabolic adaptation to sustain uncontrolled proliferation and depends on the high rate of glycolysis and glutaminolysis. Hence, identifying and targeting proteins that assist cancer cells in metabolic reprogramming is an effective strategy to block their proliferation. We detected a strong upregulation of NAD(P)H Quinone Dehydrogenase 1 (Nqo1), a cytosolic flavoprotein, in vivo in mouse and human liver tumors as well as in liver cancer cell lines in vitro. Knockdown of Nqo1 in liver cancer cells by shRNA caused a significant downregulation of both glycolytic and glutaminolysis genes, impaired cell proliferation, colony formation and in vivo xenograft tumor growth. Nqo1-/- mice with HCC displayed significant reduction in tumor number, size and growth. In Nqo1+/+ mice, histological analysis of liver tumors revealed large tumor cells with hyperchromatic nuclei in a compact growth pattern, whereas Nqo1-/- livers appeared normal. Hyper-activated PI3K/Akt and MAPK/ERK signaling pathways play central role in cancer cell metabolic adaptation since their downstream effectors such as Akt and c-Myc control most of the glycolytic and glutaminolysis genes. Interestingly, at the molecular level, Nqo1 knockdown caused strong upregulation of the tumor suppressor PTEN, and enhanced activation of the serine/threonine phosphatase PP2A, leading to impaired activation of the PI3K/Akt and MAPK/ERK/c-Myc pathways. Our findings indicate that, by targeting PP2A and PTEN, Nqo1 enhances the expression and/or activity of PDK1, Akt, ERK1/2 and c-Myc and promotes cancer cell metabolic reprogramming and hence may function as a key therapeutic target for HCC inhibition. Citation Format: Manali Dimri, Ashley Humphries, Archana Laknaur, Sawsan Elattar, Ashok Sharma, Ravindra Kolhe, Ande Satyanarayana. Nqo1 ablation inhibits activation of the PI3K/Akt and MAPK/ERK pathways and blocks metabolic adaptation in hepatocellular carcinoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 1835.
- Published
- 2019
- Full Text
- View/download PDF
26. Speedy A-Cdk2 binding mediates initial telomere-nuclear envelope attachment during meiotic prophase I independent of Cdk2 activation
- Author
-
Hongbin Liu, Vincenzo Coppola, Kui Liu, Zi-Jiang Chen, Lino Tessarollo, Zhaowei Tu, Philipp Kaldis, Chunsheng Han, Meenakshi Singh, Ande Satyanarayana, Chunwei Zheng, Mustafa Bilal Bayazit, Kiran Busayavalasa, Jingchen Shao, Jan-Åke Gustafsson, Jingjing Zhang, and Sanjiv Risal
- Subjects
0301 basic medicine ,Chromosome movement ,Male ,Telomere localization ,Nuclear Envelope ,Cell Cycle Proteins ,Biology ,03 medical and health sciences ,Meiotic Prophase I ,Mice ,0302 clinical medicine ,Meiosis ,Spermatocytes ,Homologous chromosome ,Animals ,Protein Interaction Domains and Motifs ,Genetics ,Mice, Knockout ,Multidisciplinary ,Cyclin-Dependent Kinase 2 ,Synapsis ,Telomere ,Biological Sciences ,Cell biology ,Enzyme Activation ,Mice, Inbred C57BL ,030104 developmental biology ,Oocytes ,Female ,biological phenomena, cell phenomena, and immunity ,Homologous recombination ,030217 neurology & neurosurgery - Abstract
Telomere attachment to the nuclear envelope (NE) is a prerequisite for chromosome movement during meiotic prophase I that is required for pairing of homologous chromosomes, synapsis, and homologous recombination. Here we show that Speedy A, a noncanonical activator of cyclin-dependent kinases (Cdks), is specifically localized to telomeres in prophase I male and female germ cells in mice, and plays an essential role in the telomere-NE attachment. Deletion of Spdya in mice disrupts telomere-NE attachment, and this impairs homologous pairing and synapsis and leads to zygotene arrest in male and female germ cells. In addition, we have identified a telomere localization domain on Speedy A covering the distal N terminus and the Cdk2-binding Ringo domain, and this domain is essential for the localization of Speedy A to telomeres. Furthermore, we found that the binding of Cdk2 to Speedy A is indispensable for Cdk2's localization on telomeres, suggesting that Speedy A and Cdk2 might be the initial components that are recruited to the NE for forming the meiotic telomere complex. However, Speedy A-Cdk2-mediated telomere-NE attachment is independent of Cdk2 activation. Our results thus indicate that Speedy A and Cdk2 might mediate the initial telomere-NE attachment for the efficient assembly of the telomere complex that is essential for meiotic prophase I progression.
- Published
- 2016
27. Ablation of the transcriptional regulator Id1 enhances energy expenditure, increases insulin sensitivity, and protects against age and diet induced insulin resistance, and hepatosteatosis
- Author
-
Ande Satyanarayana, Jonathan R. Keller, Oksana Gavrilova, and Kimberly D. Klarmann
- Subjects
Inhibitor of Differentiation Protein 1 ,Male ,Aging ,medicine.medical_specialty ,Mice, 129 Strain ,Adipose Tissue, White ,Adipose tissue ,White adipose tissue ,Biology ,Biochemistry ,Research Communications ,Mice ,chemistry.chemical_compound ,Insulin resistance ,Adipose Tissue, Brown ,Pregnancy ,Internal medicine ,Brown adipose tissue ,Adipocytes ,Genetics ,medicine ,Animals ,Molecular Biology ,Cells, Cultured ,Mice, Knockout ,Adipogenesis ,Triglyceride ,Leptin ,Fatty Acids ,Cell Differentiation ,Thermogenesis ,Fibroblasts ,medicine.disease ,Fatty Liver ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Endocrinology ,chemistry ,Female ,Insulin Resistance ,Energy Metabolism ,Oxidation-Reduction ,Biotechnology - Abstract
Obesity is a major health concern that contributes to the development of diabetes, hyperlipidemia, coronary artery disease, and cancer. Id proteins are helix-loop-helix transcription factors that regulate the proliferation and differentiation of cells from multiple tissues, including adipocytes. We screened mouse tissues for the expression of Id1 and found that Id1 protein is highly expressed in brown adipose tissue (BAT) and white adipose tissue (WAT), suggesting a role for Id1 in adipogenesis and cell metabolism. Id1−/− mice are viable but show a significant reduction in fat mass (P
- Published
- 2011
- Full Text
- View/download PDF
28. Abstract 447: ZAG promotes cachexia-associated white adipose tissue browning and energy wastage
- Author
-
Elattar, Sawsan, primary and Ande, Satyanarayana, additional
- Published
- 2017
- Full Text
- View/download PDF
29. The cellular level of telomere dysfunction determines induction of senescence or apoptosis in vivo
- Author
-
Ande Satyanarayana, Nisar P. Malek, Ludwig Wilkens, Sonja Schaetzlein, K. Lenhard Rudolph, Michael P. Manns, Zhenyu Ju, Stephanie U Wiemann, Ruben R. Plentz, Gabriele Saretzki, Jan Buer, Cornelia Rudolph, and André Lechel
- Subjects
Senescence ,Liver cytology ,Scientific Report ,Apoptosis ,Biology ,medicine.disease_cause ,Biochemistry ,Mice ,In vivo ,Genetics ,medicine ,Animals ,Telomeric Repeat Binding Protein 2 ,Molecular Biology ,Gene ,Cellular Senescence ,Mice, Knockout ,Mutation ,Telomere ,Cell biology ,Mice, Inbred C57BL ,Liver ,Female ,Tumor Suppressor Protein p53 ,Cell aging - Abstract
Telomere dysfunction induces two types of cellular response: cellular senescence and apoptosis. We analysed the extent to which the cellular level of telomere dysfunction and p53 gene status affect these cellular responses in mouse liver using the experimental system of TRF2 inhibition by a dominant-negative version of the protein (TRF2delta B delta M). We show that the level of telomere dysfunction correlates with the level of TRF2delta B delta M protein expression resulting in chromosomal fusions, aberrant mitotic figures and aneuploidy of liver cells. These alterations provoked p53-independent apoptosis, but a strictly p53-dependent senescence response in distinct populations of mouse liver cells depending on the cellular level of TRF2delta B delta M expression. Apoptosis was associated with higher expression of TRF2delta B delta M, whereas cellular senescence was associated with low levels of TRF2delta B delta M) expression. Our data provide experimental evidence that induction of senescence or apoptosis in vivo depends on the cellular level of telomere dysfunction and differentially on p53 gene function.
- Published
- 2005
- Full Text
- View/download PDF
30. Gene Expression Profile at the G1/S Transition of Liver Regeneration after Partial Hepatectomy in Mice
- Author
-
Robert Geffers, K. Lenhard Rudolph, Michael P. Manns, Ande Satyanarayana, and Jan Buer
- Subjects
Time Factors ,Transcription, Genetic ,Cell division ,Liver cytology ,Biology ,S Phase ,Mice ,Cytochrome P-450 Enzyme System ,Transcriptional regulation ,Animals ,Hepatectomy ,RNA, Messenger ,Molecular Biology ,Oligonucleotide Array Sequence Analysis ,Regulation of gene expression ,Gene Expression Profiling ,G1 Phase ,Membrane Transport Proteins ,G1/S transition ,Cell Biology ,Cell cycle ,Molecular biology ,Liver regeneration ,Liver Regeneration ,Mice, Inbred C57BL ,Gene Expression Regulation ,Liver ,Signal transduction ,Acute-Phase Proteins ,Developmental Biology - Abstract
Liver is a quiescent organ with >90% of the cells present in the G(0) stage of the cell cycle. However, adult hepatocytes have enormous ability to proliferate in response to liver injury. After 70% liver resection hepatocytes enter the cell cycle in a highly synchronized manner and undergo 1 to 2 rounds of cell division to restore the lost organ mass, thus, representing one of the most reliable model systems to study cell cycle progression in vivo. Using high density oligonucleotide micro-array we analyzed the expression patterns of genes at the G(1)/S transition of liver regeneration in comparison to quiescent livers. The G(1)/S boundary was identified by in vivo BrdU pulse labeling and we observed 199 genes/ESTs which were either up/down regulated at this time point. These differentially regulated genes have a wide range of functions including transcriptional regulation, signal transduction, cell cycle regulation, chromatin reorganization, protein targeting, metabolism, transport, surface receptors, circadian rhythms, xenobiotic metabolism, inflammation and acute phase response. The functions of most of the genes identified in this screen are not known in the process of liver regeneration and cell cycle control at G(1)/S transition.
- Published
- 2004
- Full Text
- View/download PDF
31. Telomere shortening impairs organ regeneration by inhibiting cell cycle re-entry of a subpopulation of cells
- Author
-
Michael P. Manns, Torsten Wüstefeld, Ande Satyanarayana, Stefanie U. Wiemann, Maria A. Blasco, Jörg Lauber, Jan Buer, K.L. Rudolph, and Kurt E.J. Dittmar
- Subjects
Senescence ,Telomerase ,Cell division ,Liver cytology ,Biology ,Polymerase Chain Reaction ,General Biochemistry, Genetics and Molecular Biology ,Mice ,Animals ,Regeneration ,Molecular Biology ,Oligonucleotide Array Sequence Analysis ,Mice, Knockout ,Genetics ,General Immunology and Microbiology ,General Neuroscience ,Regeneration (biology) ,Cell Cycle ,Articles ,Telomere ,Cell cycle ,Immunohistochemistry ,Liver regeneration ,Cell biology ,Liver ,RNA ,Cell Division - Abstract
Telomere shortening limits the regenerative capacity of primary cells in vitro by inducing cellular senescence characterized by a permanent growth arrest of cells with critically short telomeres. To test whether this in vitro model of cellular senescence applies to impaired organ regeneration induced by telomere shortening in vivo, we monitored liver regeneration after partial hepatectomy in telomerase-deficient mice. Our study shows that telomere shortening is heterogeneous at the cellular level and inhibits a subpopulation of cells with critically short telomeres from entering the cell cycle. This subpopulation of cells with impaired proliferative capacity shows senescence-associated beta-galactosidase activity, while organ regeneration is accomplished by cells with sufficient telomere reserves that are capable of additional rounds of cell division. This study provides experimental evidence for the existence of an in vivo process of cellular senescence induced by critical telomere shortening that has functional impact on organ regeneration.
- Published
- 2003
- Full Text
- View/download PDF
32. Id transcriptional regulators in adipogenesis and adipose tissue metabolism
- Author
-
Ande Satyanarayana, Bal Krishan Sharma, and Mallikarjun Patil
- Subjects
Regulation of gene expression ,Adipogenesis ,Models, Genetic ,Cellular differentiation ,Adipose tissue ,DNA-binding domain ,Biology ,Cell fate determination ,Article ,chemistry.chemical_compound ,Biochemistry ,chemistry ,Adipose Tissue ,Gene Expression Regulation ,Adipocyte ,Basic Helix-Loop-Helix Transcription Factors ,Animals ,Humans ,Protein Isoforms ,Inhibitor of Differentiation Proteins ,Transcription factor ,Signal Transduction - Abstract
Id proteins (Id1-Id4) are helix-loop-helix (HLH) transcriptional regulators that lack a basic DNA binding domain. They act as negative regulators of basic helix-loop-helix (bHLH) transcription factors by forming heterodimers and inhibit their DNA binding and transcriptional activity. Id proteins are implicated in the regulation of various cellular mechanisms such as cell proliferation, cellular differentiation, cell fate determination, angiogenesis and tumorigenesis. A handful of recent studies also disclosed that Id proteins have critical functions in adipocyte differentiation and adipose tissue metabolism. Here, we reviewed the progress made thus far in understanding the specific functions of Id proteins in adipose tissue differentiation and metabolism. In addition to reviewing the known mechanisms of action, we also discuss possible additional mechanisms in which Id proteins might participate in regulating adipogenic and metabolic pathways.
- Published
- 2014
33. Novel Targets in Myelogenous Leukemia: The Id Family of Proteins
- Author
-
Emily Bowers, Huajie Li, Bjorg Gudmundsdottir, Ande Satyanarayana, Wonil Kim, Jonathan R. Keller, Kimberly D. Klarmann, and Ming Ji
- Subjects
education.field_of_study ,Kinase ,business.industry ,Population ,Imatinib ,Drug resistance ,medicine.disease ,Leukemia ,Myelogenous ,hemic and lymphatic diseases ,medicine ,Cancer research ,education ,business ,neoplasms ,Tyrosine kinase ,Chronic myelogenous leukemia ,medicine.drug - Abstract
1.1 Myelogenous leukemia and the need for novel drug targets The successful treatment of myelogenous leukemia depends upon the continuing coordinate efforts of the medical and research communities and patients to understand the complex nature of leukemia development and progression, and to battle drug resistant cells. Novel drug therapies, like Imatinib, have been developed which target the mutant BCR-ABL (Breakpoint Cluster Region-Abelson Kinase) protein, and have significantly changed the treatment regimens for chronic myelogenous leukemia (CML). However, while Imatinib is frequently effective against the majority of the leukemic cells, and patients often undergo remission after treatment, these patients also relapse due to a leukemic drug-resistant population (Schemionek et al., 2010). In comparison to CML, the treatment for acute myelogenous leukemia (AML) is predominantly induction therapy using strong anti-mitotic agents, followed by consolidation therapy to kill residual diseased cells (Venditti et al., 2000). These therapies are not specifically targeted to mutated proteins in the cells, and therefore can be more toxic to the patient's normal cells. Researchers have attempted to target mutated proteins in AML. However, unlike BCR-ABL, targeting FLT3-ITD (FMS-like tyrosine kinase 3-Internal Tandem Duplication) and FLT3-TKD (FMS-like tyrosine kinase 3Tyrosine Kinase Domain) mutations have not been as successful as CML-Imatinib therapy (Kindler et al., 2010). Therefore, challenges facing patients with AML remain significant, and drug-resistant relapses remain a threat.
- Published
- 2011
34. Mammalian cell-cycle regulation: several Cdks, numerous cyclins and diverse compensatory mechanisms
- Author
-
Philipp Kaldis and Ande Satyanarayana
- Subjects
Cancer Research ,Cell Cycle Proteins ,Models, Biological ,Mice ,Mitotic cell cycle ,Cyclin-dependent kinase ,Cyclins ,CDC2 Protein Kinase ,Genetics ,Animals ,Humans ,Molecular Biology ,Gene knockout ,Cyclin ,Mice, Knockout ,Cyclin-dependent kinase 1 ,biology ,Cyclin-dependent kinase 2 ,Cell Cycle ,Cyclin-Dependent Kinase 2 ,Cyclin-Dependent Kinase 4 ,Cell cycle ,Cyclin-Dependent Kinases ,Cell biology ,enzymes and coenzymes (carbohydrates) ,Meiosis ,Knockout mouse ,biology.protein ,biological phenomena, cell phenomena, and immunity ,DNA Damage - Abstract
After a decade of extensive work on gene knockout mouse models of cell-cycle regulators, the classical model of cell-cycle regulation was seriously challenged. Several unexpected compensatory mechanisms were uncovered among cyclins and Cdks in these studies. The most astonishing observation is that Cdk2 is dispensable for the regulation of the mitotic cell cycle with both Cdk4 and Cdk1 covering for Cdk2's functions. Similar to yeast, it was recently discovered that Cdk1 alone can drive the mammalian cell cycle, indicating that the regulation of the mammalian cell cycle is highly conserved. Nevertheless, cell-cycle-independent functions of Cdks and cyclins such as in DNA damage repair are still under investigation. Here we review the compensatory mechanisms among major cyclins and Cdks in mammalian cell-cycle regulation.
- Published
- 2009
35. Genetic substitution of Cdk1 by Cdk2 leads to embryonic lethality and loss of meiotic function of Cdk2
- Author
-
Ande Satyanarayana, Philipp Kaldis, Javier Lopez-Molina, Vincenzo Coppola, Cyril Berthet, and Lino Tessarollo
- Subjects
Male ,Locus (genetics) ,Biology ,environment and public health ,Article ,Mice ,Cyclin-dependent kinase ,Complementary DNA ,CDC2 Protein Kinase ,Animals ,Molecular Biology ,Gene ,Cells, Cultured ,Cyclin-dependent kinase 1 ,Cyclin-dependent kinase 2 ,Cyclin-Dependent Kinase 2 ,Cell cycle ,Fibroblasts ,Embryo, Mammalian ,Molecular biology ,Immunohistochemistry ,Cell biology ,enzymes and coenzymes (carbohydrates) ,Meiosis ,biology.protein ,Female ,Genes, Lethal ,biological phenomena, cell phenomena, and immunity ,Developmental Biology - Abstract
It was believed that Cdk2-cyclin E complexes are essential to drive cells through the G1-S phase transition. However, it was discovered recently that the mitotic kinase Cdk1 (Cdc2a) compensates for the loss of Cdk2. In the present study, we tested whether Cdk2 can compensate for the loss of Cdk1. We generated a knockin mouse in which the Cdk2 cDNA was knocked into the Cdk1 locus (Cdk1Cdk2KI). Substitution of both copies of Cdk1 by Cdk2 led to early embryonic lethality,even though Cdk2 was expressed from the Cdk1 locus. In addition, we generated Cdk2-/-Cdk1+/Cdk2KI mice in which one copy of Cdk2 and one copy of Cdk1 were expressed from the Cdk1 locus and the Cdk2 gene was deleted from the endogenous Cdk2 locus. We found that both male and female Cdk2-/-Cdk1+/Cdk2KI mice were sterile, similar to Cdk2-/- mice, even though they expressed the Cdk2 protein from the Cdk1 locus in testes. The translocational and cell cycle properties of knockin Cdk2 in Cdk2-/-Cdk1+/Cdk2KI cells were comparable to those of endogenous Cdk2, but we detected premature transcriptional activation of Cdk1during liver regeneration in the absence of Cdk2. This study provides evidence of the molecular differences between Cdk2 and Cdk1 and highlights that the timing of transcriptional activation and the genetic locus play important roles in determining the function of Cdk proteins in vivo.
- Published
- 2008
36. p21 Inhibits Cdk1 in the absence of Cdk2 to maintain the G1/S phase DNA damage checkpoint
- Author
-
Ande Satyanarayana, Philipp Kaldis, and Mary Beth Hilton
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,DNA Repair ,DNA damage ,DNA repair ,Chromosomal translocation ,Radiation Tolerance ,S Phase ,Mice ,CDC2 Protein Kinase ,medicine ,Animals ,CHEK1 ,Molecular Biology ,Cell Nucleus ,biology ,Cyclin-dependent kinase 2 ,Cyclin-Dependent Kinase 2 ,G1 Phase ,Cell Biology ,Articles ,G2-M DNA damage checkpoint ,Fibroblasts ,Molecular biology ,Cell biology ,Protein Transport ,medicine.anatomical_structure ,Cytoplasm ,Gamma Rays ,biology.protein ,biological phenomena, cell phenomena, and immunity ,Tumor Suppressor Protein p53 ,Nucleus ,DNA Damage - Abstract
Cdk1 was proposed to compensate for the loss of Cdk2. Here we present evidence that this is possible due to premature translocation of Cdk1 from the cytoplasm to the nucleus in the absence of Cdk2. We also investigated the consequence of loss of Cdk2 on the maintenance of the G1/S DNA damage checkpoint. Cdk2−/−mouse embryonic fibroblasts in vitro as well as regenerating liver cells after partial hepatectomy (PH) in Cdk2−/−mice, arrest promptly at the G1/S checkpoint in response to γ-irradiation due to activation of p53 and p21 inhibiting Cdk1. Furthermore re-entry into S phase after irradiation was delayed in Cdk2−/−cells due to prolonged and impaired DNA repair activity. In addition, Cdk2−/−mice were more sensitive to lethal irradiation compared to wild-type and displayed delayed resumption of DNA replication in regenerating liver cells. Our results suggest that the G1/S DNA damage checkpoint is intact in the absence of Cdk2, but Cdk2 is important for proper repair of the damaged DNA.
- Published
- 2007
37. Telomeres and Telomerase: Distinctive Roles in Liver Regeneration, Cirrhosis and Carcinogenesis
- Author
-
Ande Satyanarayana and K. Lenhard Rudolph
- Subjects
Pathology ,medicine.medical_specialty ,Telomerase ,Cell division ,biology ,DNA polymerase ,Reverse transcriptase ,Cell biology ,Telomere ,Telomerase RNA component ,chemistry.chemical_compound ,chemistry ,biology.protein ,medicine ,Telomerase reverse transcriptase ,DNA - Abstract
Telomeres are specialized nucleo-protein structures at the end of linear eukaryotic chromosomes [6, 8], consisting of small tandem nucleotide repeats of several kilo base pairs, which provide a platform for a variety of proteins that bind to telomeres [12, 35, 41, 42, 59, 64, 81] and are possibly involved in telomere-mediated responses [7, 27, 34]. Some of these proteins are necessary for the stabilization of telomeres and formation of higher-order telomere structures, such as T loops, D loops [24, 25] or G quartets [5, 80]. The main function of telomeres is to cap and protect chromosomal ends, thus to prevent chromosomal fusions [7, 15, 27, 33, 65]. The inability of DNA polymerase to replicate at the extreme ends of linear DNA molecules leads to telomere shortening during each round of cell division [43], which is overcome by the reverse transcriptase telomerase, which synthesizes telomere repeats de novo [23]. Telomerase holoenzyme consists of two essential components: (a) the telomerase RNA component (TERC), which serves as a template for the synthesis of telomere sequence [22], and (b) the telomerase reverse transcriptase (TERT), which is the catalytic component of the holoenzyme [9, 46]. TERC is ubiquitously expressed in humans, whereas TERT expression is suppressed and acts as a rate-limiting factor for telomerase activity in most human cells and tissues, including the liver. This leads to inevitable loss of telomere repeats at a rate of 50–100 bp after each round of replication [26]. When the telomeres reach a critically short length they lose capping function and the uncapped, dysfunctional telomeres trigger responses similar to DNA-damage signaling involving the p53-dependent DNA-damage pathway, leading to permanent cell cycle arrest, termed cellular senescence and/or apoptosis [7, 15, 33, 65, 72]. The fact that telomere shortening is prevalent in most human tissues during aging [2, 13, 18, 32, 44, 45] and in a variety of chronic diseases [18, 52, 53, 71, 76] has fueled the debate that it might limit the regenerative capacity of organs and tissues. Contrary to the adverse effects of telomere shortening on regeneration, the suppression of telomerase activity and the hindrance of cell division by telomere shortening might be beneficial for the organism since it acts as a tumor suppressor mechanism [77].
- Published
- 2005
- Full Text
- View/download PDF
38. Contrasting effects of telomere shortening on organ homeostasis, tumor suppression, and survival during chronic liver damage
- Author
-
Ande Satyanarayana, Karl Lenhard Rudolph, Stefanie U. Wiemann, Kenji Kamino, Jan Buer, and Michael P. Manns
- Subjects
Cancer Research ,Telomerase ,Liver tumor ,Apoptosis ,Mice, Transgenic ,Gene delivery ,Biology ,medicine.disease_cause ,Mice ,Genetics ,medicine ,Animals ,Homeostasis ,Molecular Biology ,beta Catenin ,Cell Nucleus ,Mice, Knockout ,Carbon Tetrachloride Poisoning ,Liver cell ,Liver Neoplasms ,medicine.disease ,Telomere ,DNA-Binding Proteins ,Cytoskeletal Proteins ,Liver ,Tumor progression ,Immunology ,Cancer research ,Trans-Activators ,Carcinogenesis ,Cell Division - Abstract
Telomere shortening limits the regenerative capacity of cells during aging and chronic disease but at the same time inhibits tumor progression, and it has yet to be determined which of these mechanisms is dominantly affecting organismal survival. Here we show that telomere shortening in telomerase knockout (mTERC-/-) mice in combination with chronic liver damage significantly reduced organismal survival even though telomere shortening strongly inhibited liver tumor formation. Decreased survival induced by telomere shortening correlated with an imbalance between liver cell proliferation and liver cell apoptosis. Specific changes in gene expression were associated with telomere shortening and chronic liver damage and these gene expression changes were partially reversed by adenovirus mediated telomerase gene delivery. This study gives experimental evidence that the negative impact of telomere shortening on organ homeostasis and organismal survival can surpass the beneficial effects of telomere shortening on suppression of tumor growth in the setting of chronic organ damage.
- Published
- 2004
39. p16 and ARF: activation of teenage proteins in old age
- Author
-
Ande Satyanarayana and K. Lenhard Rudolph
- Subjects
Senescence ,Aging ,Cellular senescence ,Biology ,Models, Biological ,Retinoblastoma Protein ,Article ,Downregulation and upregulation ,Animals ,Humans ,neoplasms ,Cellular Senescence ,Cyclin-Dependent Kinase Inhibitor p16 ,Regulation of gene expression ,Cell growth ,Retinoblastoma protein ,General Medicine ,Cell cycle ,Cell biology ,Telomere ,Up-Regulation ,Gene Expression Regulation ,Cancer research ,biology.protein ,ADP-Ribosylation Factor 1 ,Tumor Suppressor Protein p53 ,Biomarkers - Abstract
The Ink4a/Arf locus encodes 2 tumor suppressor molecules, p16INK4a and Arf, which are principal mediators of cellular senescence. To study the links between senescence and aging in vivo, we examined Ink4a/Arf expression in rodent models of aging. We show that expression of p16INK4a and Arf markedly increases in almost all rodent tissues with advancing age, while there is little or no change in the expression of other related cell cycle inhibitors. The increase in expression is restricted to well-defined compartments within each organ studied and occurs in both epithelial and stromal cells of diverse lineages. The age-associated increase in expression of p16INK4a and Arf is attenuated in the kidney, ovary, and heart by caloric restriction, and this decrease correlates with diminished expression of an in vivo marker of senescence, as well as decreased pathology of those organs. Last, the age-related increase in Ink4a/Arf expression can be independently attributed to the expression of Ets-1, a known p16INK4a transcriptional activator, as well as unknown Ink4a/Arf coregulatory molecules. These data suggest that expression of the Ink4a/Arf tumor suppressor locus is a robust biomarker, and possible effector, of mammalian aging.
- Published
- 2004
40. Telomeres, telomerase and cancer: an endless search to target the ends
- Author
-
Ande, Satyanarayana, Michael P, Manns, and K Lenhard, Rudolph
- Subjects
DNA-Binding Proteins ,Cell Transformation, Neoplastic ,Neoplasms ,Animals ,Humans ,RNA ,Antineoplastic Agents ,Telomere ,Telomerase ,Cell Division ,Cell Proliferation - Abstract
Maintenance of functional telomeres, the highly complex nucleo-protein structures, at the end of linear eukaryotic chromosomes appears to be essential for growth and survival of the cells. The compelling correlation between telomerase reactivation and cellular immortalization led to the idea that inhibition of telomerase may provide a way for effective hindrance of cancer cell growth by interfering with telomere maintenance. In addition to targeting the components of telomerase enzyme directly to prevent telomere synthesis, several approaches including disruption of telomeres, interference with telomerase component assembly, translocation of the catalytic component of telomerase have also been under extensive investigation due to the advances in understanding the biology of telomeres and telomerase in recent years. This review will focus on the so far identified approaches to prevent cancer cell growth by targeting telomerase and telomeres with a brief introduction about structure and function of telomeres and telomerase.
- Published
- 2004
41. Telomeres and telomerase: a dual role in hepatocarcinogenesis
- Author
-
Michael P. Manns, Ande Satyanarayana, and K. Lenhard Rudolph
- Subjects
Pathology ,medicine.medical_specialty ,Telomerase ,Carcinoma, Hepatocellular ,Biology ,medicine.disease_cause ,Enzyme activator ,Mice ,Liver Neoplasms, Experimental ,Chromosome instability ,medicine ,Animals ,Humans ,Telomerase reverse transcriptase ,Mice, Knockout ,Hepatology ,Liver Neoplasms ,Cancer ,Telomere ,medicine.disease ,DNA-Binding Proteins ,Enzyme Activation ,Tumor progression ,Cancer research ,Disease Progression ,RNA ,Carcinogenesis - Abstract
Telomere shortening limits the proliferative capacity of primary human cells and restrains the regenerative capacity of organ systems during chronic diseases and aging. Telomere shortening apparently has a dual role in tumor development and progression. On the one hand, it induces chromosomal instability and the initiation of cancer; on the other hand, tumor progression requires stabilization of telomeres. The predominant mechanism of telomere stabilization in tumor cells is the activation of the telomere-synthesizing enzyme telomerase. The potential use of telomerase activators for the treatment of regenerative disorders will ultimately depend on their effects on tumorigenesis. This review focuses on the role of telomere shortening and telomerase in carcinogenesis with a special focus on hepatocellular carcinoma.
- Published
- 2004
42. Mitogen Stimulation Cooperates with Telomere Shortening To Activate DNA Damage Responses and Senescence Signaling
- Author
-
William C. Hahn, Uwe M. Martens, Ande Satyanarayana, Kenkichi Masutomi, Stefan Zimmermann, Sonja Schaetzlein, Karl Lenhard Rudolph, Jan Buer, Roger A. Greenberg, and Michael P. Manns
- Subjects
Senescence ,Male ,Telomerase ,DNA, Complementary ,Cell division ,DNA damage ,Biology ,Mice ,Animals ,Humans ,Molecular Biology ,Cell Growth and Development ,Cells, Cultured ,Cellular Senescence ,Mice, Knockout ,Base Sequence ,Cell Cycle ,Cell Biology ,Cell cycle ,Telomere ,Cell biology ,Mice, Inbred C57BL ,Phenotype ,RNA ,Signal transduction ,Mitogens ,Cell aging ,Cell Division ,DNA Damage ,Signal Transduction - Abstract
Replicative senescence is induced by critical telomere shortening and limits the proliferation of primary cells to a finite number of divisions. To characterize the activity status of the replicative senescence program in the context of cell cycle activity, we analyzed the senescence phenotypes and signaling pathways in quiescent and growth-stimulated primary human fibroblasts in vitro and liver cells in vivo. This study shows that replicative senescence signaling operates at a low level in cells with shortened telomeres but becomes fully activated when cells are stimulated to enter the cell cycle. This study also shows that the dysfunctional telomeres and nontelomeric DNA lesions in senescent cells do not elicit a DNA damage signal unless the cells are induced to enter the cell cycle by mitogen stimulation. The amplification of senescence signaling and DNA damage responses by mitogen stimulation in cells with shortened telomeres is mediated in part through the MEK/mitogen-activated protein kinase pathway. These findings have implications for the further understanding of replicative senescence and analysis of its role in vivo.
- Published
- 2004
43. 883 The impact of telomere shortening on liver regeneration, DNA-damage-response and hepatocarcinogenesis
- Author
-
A.C. Kabbasch, M.P. Manns, Stefanie U. Wiemann, Ande Satyanarayana, and Karl Lenhard Rudolph
- Subjects
Hepatology ,DNA damage ,Biology ,Liver regeneration ,Telomere ,Cell biology - Published
- 2003
- Full Text
- View/download PDF
44. A dual role of Cdk2 in DNA damage response
- Author
-
Ande Satyanarayana and Philipp Kaldis
- Subjects
Genetics ,Cyclin-dependent kinase 1 ,biology ,lcsh:Cytology ,DNA damage ,Cyclin-dependent kinase 2 ,Cell Biology ,G2-M DNA damage checkpoint ,Cell cycle ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,Biochemistry ,Cell biology ,Dual role ,Cyclin-dependent kinase ,biology.protein ,Commentary ,lcsh:QH573-671 ,biological phenomena, cell phenomena, and immunity ,Molecular Biology ,Gene knockout - Abstract
Once it was believed that Cdk2 was the master regulator of S phase entry. Gene knockout mouse studies of cell cycle regulators revealed that Cdk2 is dispensable for S phase initiation and progression whereby Cdk1 can compensate for the loss of Cdk2. Nevertheless, recent evidence indicates that Cdk2 is involved in cell cycle independent functions such as DNA damage repair. Whether these properties are unique to Cdk2 or also being compensated by other Cdks in the absence of Cdk2 is under extensive investigation. Here we review the emerging new role of Cdk2 in DNA damage repair and also discuss how the loss of Cdk2 impacts the G1/S phase DNA damage checkpoint.
- Published
- 2009
45. Hepatocyte telomere shortening and senescence are general markers of human liver cirrhosis
- Author
-
Ande, Satyanarayana, primary, Wiemann, Stefanie, additional, Tsahuridu, Martina, additional, Tillmann, Hans, additional, Manns, Michael P., additional, and Rudolph, K.Lenhard, additional
- Published
- 2002
- Full Text
- View/download PDF
46. Telomeres, telomerase and cancer: an endless search to target the ends
- Author
-
Ande Satyanarayana, Michael P. Manns, and K. Lenhard Rudolph
- Subjects
Genetics ,Telomerase ,Cell Biology ,Biology ,Cell cycle ,G-quadruplex ,Telomere ,Cell biology ,Telomerase RNA component ,Eukaryotic chromosome fine structure ,Cancer cell ,Telomerase reverse transcriptase ,Molecular Biology ,Developmental Biology - Abstract
Maintenance of functional telomeres, the highly complex nucleo-protein structures, at the end of linear eukaryotic chromosomes appears to be essential for growth and survival of the cells. The compelling correlation between telomerase re-activation and cellular immortalization led to the idea that inhibition of telomerase may provide a way for effective hindrance of cancer cell growth by interfering with telomere maintenance. In addition to targeting the components of telomerase enzyme directly to prevent telomere synthesis, several approaches including disruption of telomeres, interference with telomerase component assembly, translocation of the catalytic component of telomerase etc., have also been under extensive investigation due to the advances in understanding the biology of telomeres and telomerase in recent years. This review will focus on the so far identified approaches to prevent cancer cell growth by targeting telomerase and telomeres with a brief introduction about structure and function of t...
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.