1
|
Kosińska K, Skóra B, Holota S, Shepeta Y, Tabęcka-Łonczyńska A, Lesyk R, Szychowski KA. Role of 4-Thiazolidinone-Pyrazoline/Indoline Hybrids Les-4369 and Les-3467 in BJ and A549 Cell Lines. Cells 2024; 13:1007. [PMID: 38920636 PMCID: PMC11202306 DOI: 10.3390/cells13121007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 05/28/2024] [Accepted: 06/06/2024] [Indexed: 06/27/2024] Open
Abstract
Cancer is one of the most important problems of modern societies. Recently, studies have reported the anticancer properties of rosiglitazone related to its ability to bind peroxisome proliferator receptor γ (PPARγ), which has various effects on cancer and can inhibit cell proliferation. In this study, we investigated the effect of new 4-thiazolidinone (4-TZD) hybrids Les-4369 and Les-3467 and their effect on reactive oxygen species (ROS) production, metabolic activity, lactate dehydrogenase (LDH) release, caspase-3 activity, and gene and protein expression in human foreskin fibroblast (BJ) cells and lung adenocarcinoma (A549) cells. The ROS production and caspase-3 activity were mainly increased in the micromolar concentrations of the studied compounds in both cell lines. Les-3467 and Les-4369 increased the mRNA expression of PPARG, P53 (tumor protein P53), and ATM (ATM serine/threonine kinase) in the BJ cells, while the mRNA expression of these genes (except PPARG) was mainly decreased in the A549 cells treated with both of the tested compounds. Our results indicate a decrease in the protein expression of AhR, PPARγ, and PARP-1 in the BJ cells exposed to 1 µM Les-3467 and Les-4369. In the A549 cells, the protein expression of AhR, PPARγ, and PARP-1 increased in the treatment with 1 µM Les-3467 and Les-4369. We have also shown the PPARγ modulatory properties of Les-3467 and Les-4369. However, both compounds prove weak anticancer properties evidenced by their action at high concentrations and non-selective effects against BJ and A549 cells.
Collapse
Affiliation(s)
- Karolina Kosińska
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland; (B.S.); (A.T.-Ł.); (R.L.); (K.A.S.)
| | - Bartosz Skóra
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland; (B.S.); (A.T.-Ł.); (R.L.); (K.A.S.)
| | - Serhii Holota
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine;
- Department of Organic Chemistry and Pharmacy, Lesya Ukrainka Volyn National University, Volya Avenue 13, 43025 Lutsk, Ukraine
| | - Yulia Shepeta
- Department of Pharmaceutical Chemistry, National Pirogov Memorial Medical University, Pirogov 56, 21018 Vinnytsia, Ukraine;
| | - Anna Tabęcka-Łonczyńska
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland; (B.S.); (A.T.-Ł.); (R.L.); (K.A.S.)
| | - Roman Lesyk
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland; (B.S.); (A.T.-Ł.); (R.L.); (K.A.S.)
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine;
| | - Konrad A. Szychowski
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland; (B.S.); (A.T.-Ł.); (R.L.); (K.A.S.)
| |
Collapse
|
2
|
Ma S, Ming Y, Wu J, Cui G. Cellular metabolism regulates the differentiation and function of T-cell subsets. Cell Mol Immunol 2024; 21:419-435. [PMID: 38565887 PMCID: PMC11061161 DOI: 10.1038/s41423-024-01148-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 02/23/2024] [Indexed: 04/04/2024] Open
Abstract
T cells are an important component of adaptive immunity and protect the host from infectious diseases and cancers. However, uncontrolled T cell immunity may cause autoimmune disorders. In both situations, antigen-specific T cells undergo clonal expansion upon the engagement and activation of antigens. Cellular metabolism is reprogrammed to meet the increase in bioenergetic and biosynthetic demands associated with effector T cell expansion. Metabolites not only serve as building blocks or energy sources to fuel cell growth and expansion but also regulate a broad spectrum of cellular signals that instruct the differentiation of multiple T cell subsets. The realm of immunometabolism research is undergoing swift advancements. Encapsulating all the recent progress within this concise review in not possible. Instead, our objective is to provide a succinct introduction to this swiftly progressing research, concentrating on the metabolic intricacies of three pivotal nutrient classes-lipids, glucose, and amino acids-in T cells. We shed light on recent investigations elucidating the roles of these three groups of metabolites in mediating the metabolic and immune functions of T cells. Moreover, we delve into the prospect of "editing" metabolic pathways within T cells using pharmacological or genetic approaches, with the aim of synergizing this approach with existing immunotherapies and enhancing the efficacy of antitumor and antiinfection immune responses.
Collapse
Affiliation(s)
- Sicong Ma
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230601, China
| | - Yanan Ming
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230601, China
| | - Jingxia Wu
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230601, China.
| | - Guoliang Cui
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230601, China.
| |
Collapse
|
3
|
Wątroba M, Szewczyk G, Szukiewicz D. The Role of Sirtuin-1 (SIRT1) in the Physiology and Pathophysiology of the Human Placenta. Int J Mol Sci 2023; 24:16210. [PMID: 38003402 PMCID: PMC10671790 DOI: 10.3390/ijms242216210] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/04/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Sirtuins, especially SIRT1, play a significant role in regulating inflammatory response, autophagy, and cell response to oxidative stress. Since their discovery, sirtuins have been regarded as anti-ageing and longevity-promoting enzymes. Sirtuin-regulated processes seem to participate in the most prevalent placental pathologies, such as pre-eclampsia. Furthermore, more and more research studies indicate that SIRT1 may prevent pre-eclampsia development or at least alleviate its manifestations. Having considered this, we reviewed recent studies on the role of sirtuins, especially SIRT1, in processes determining normal or abnormal development and functioning of the placenta.
Collapse
Affiliation(s)
| | | | - Dariusz Szukiewicz
- Department of Biophysics, Physiology & Pathophysiology, Medical University of Warsaw, Chałubinskiego 5, 02-004 Warsaw, Poland; (M.W.); (G.S.)
| |
Collapse
|
4
|
Kim DJ, Yi YW, Seong YS. Beta-Transducin Repeats-Containing Proteins as an Anticancer Target. Cancers (Basel) 2023; 15:4248. [PMID: 37686524 PMCID: PMC10487276 DOI: 10.3390/cancers15174248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
Beta-transducin repeat-containing proteins (β-TrCPs) are E3-ubiquitin-ligase-recognizing substrates and regulate proteasomal degradation. The degradation of β-TrCPs' substrates is tightly controlled by various external and internal signaling and confers diverse cellular processes, including cell cycle progression, apoptosis, and DNA damage response. In addition, β-TrCPs function to regulate transcriptional activity and stabilize a set of substrates by distinct mechanisms. Despite the association of β-TrCPs with tumorigenesis and tumor progression, studies on the mechanisms of the regulation of β-TrCPs' activity have been limited. In this review, we studied publications on the regulation of β-TrCPs themselves and analyzed the knowledge gaps to understand and modulate β-TrCPs' activity in the future.
Collapse
Affiliation(s)
- Dong Joon Kim
- Department of Microbiology, College of Medicine, Dankook University, Cheonan-si 31116, Chungcheongnam-do, Republic of Korea;
- Multidrug-Resistant Refractory Cancer Convergence Research Center (MRCRC), Dankook University, Cheonan-si 31116, Chungcheongnam-do, Republic of Korea
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Science, College of Medicine, Zhengzhou University, Zhengzhou 450008, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou 450008, China
| | - Yong Weon Yi
- Multidrug-Resistant Refractory Cancer Convergence Research Center (MRCRC), Dankook University, Cheonan-si 31116, Chungcheongnam-do, Republic of Korea
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan-si 31116, Chungcheongnam-do, Republic of Korea
| | - Yeon-Sun Seong
- Multidrug-Resistant Refractory Cancer Convergence Research Center (MRCRC), Dankook University, Cheonan-si 31116, Chungcheongnam-do, Republic of Korea
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan-si 31116, Chungcheongnam-do, Republic of Korea
| |
Collapse
|
5
|
Wagner N, Wagner KD. Peroxisome Proliferator-Activated Receptors and the Hallmarks of Cancer. Cells 2022; 11:cells11152432. [PMID: 35954274 PMCID: PMC9368267 DOI: 10.3390/cells11152432] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 12/11/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) function as nuclear transcription factors upon the binding of physiological or pharmacological ligands and heterodimerization with retinoic X receptors. Physiological ligands include fatty acids and fatty-acid-derived compounds with low specificity for the different PPAR subtypes (alpha, beta/delta, and gamma). For each of the PPAR subtypes, specific pharmacological agonists and antagonists, as well as pan-agonists, are available. In agreement with their natural ligands, PPARs are mainly focused on as targets for the treatment of metabolic syndrome and its associated complications. Nevertheless, many publications are available that implicate PPARs in malignancies. In several instances, they are controversial for very similar models. Thus, to better predict the potential use of PPAR modulators for personalized medicine in therapies against malignancies, it seems necessary and timely to review the three PPARs in relation to the didactic concept of cancer hallmark capabilities. We previously described the functions of PPAR beta/delta with respect to the cancer hallmarks and reviewed the implications of all PPARs in angiogenesis. Thus, the current review updates our knowledge on PPAR beta and the hallmarks of cancer and extends the concept to PPAR alpha and PPAR gamma.
Collapse
Affiliation(s)
- Nicole Wagner
- Correspondence: (N.W.); (K.-D.W.); Tel.: +33-489-153-713 (K.-D.W.)
| | | |
Collapse
|
6
|
Tilekar K, Upadhyay N, Iancu CV, Pokrovsky V, Choe JY, Ramaa CS. Power of two: combination of therapeutic approaches involving glucose transporter (GLUT) inhibitors to combat cancer. Biochim Biophys Acta Rev Cancer 2020; 1874:188457. [PMID: 33096154 PMCID: PMC7704680 DOI: 10.1016/j.bbcan.2020.188457] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/16/2020] [Accepted: 10/16/2020] [Indexed: 12/20/2022]
Abstract
Cancer research of the Warburg effect, a hallmark metabolic alteration in tumors, focused attention on glucose metabolism whose targeting uncovered several agents with promising anticancer effects at the preclinical level. These agents' monotherapy points to their potential as adjuvant combination therapy to existing standard chemotherapy in human trials. Accordingly, several studies on combining glucose transporter (GLUT) inhibitors with chemotherapeutic agents, such as doxorubicin, paclitaxel, and cytarabine, showed synergistic or additive anticancer effects, reduced chemo-, radio-, and immuno-resistance, and reduced toxicity due to lowering the therapeutic doses required for desired chemotherapeutic effects, as compared with monotherapy. The combinations have been specifically effective in treating cancer glycolytic phenotypes, such as pancreatic and breast cancers. Even combining GLUT inhibitors with other glycolytic inhibitors and energy restriction mimetics seems worthwhile. Though combination clinical trials are in the early phase, initial results are intriguing. The various types of GLUTs, their role in cancer progression, GLUT inhibitors, and their anticancer mechanism of action have been reviewed several times. However, utilizing GLUT inhibitors as combination therapeutics has received little attention. We consider GLUT inhibitors agents that directly affect glucose transporters by binding to them or indirectly alter glucose transport by changing the transporters' expression level. This review mainly focuses on summarizing the effects of various combinations of GLUT inhibitors with other anticancer agents and providing a perspective on the current status.
Collapse
Affiliation(s)
- Kalpana Tilekar
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth’s College of Pharmacy, Navi Mumbai, Maharashtra, India
| | - Neha Upadhyay
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth’s College of Pharmacy, Navi Mumbai, Maharashtra, India
| | - Cristina V. Iancu
- East Carolina Diabetes and Obesity Institute, Department of Chemistry, East Carolina University, Greenville, North Carolina, USA
| | - Vadim Pokrovsky
- Laboratory of Combined Therapy, N.N. Blokhin Cancer Research Center, Moscow, Russia
- Department of Biochemistry, People’s Friendship University, Moscow, Russia
| | - Jun-yong Choe
- East Carolina Diabetes and Obesity Institute, Department of Chemistry, East Carolina University, Greenville, North Carolina, USA
| | - C. S. Ramaa
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth’s College of Pharmacy, Navi Mumbai, Maharashtra, India
| |
Collapse
|
7
|
Tilekar K, Upadhyay N, Hess JD, Macias LH, Mrowka P, Aguilera RJ, Meyer-Almes FJ, Iancu CV, Choe JY, Ramaa CS. Structure guided design and synthesis of furyl thiazolidinedione derivatives as inhibitors of GLUT 1 and GLUT 4, and evaluation of their anti-leukemic potential. Eur J Med Chem 2020; 202:112603. [PMID: 32634629 PMCID: PMC7451030 DOI: 10.1016/j.ejmech.2020.112603] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/14/2020] [Accepted: 06/18/2020] [Indexed: 12/24/2022]
Abstract
Cancer cells increase their glucose uptake and glycolytic activity to meet the high energy requirements of proliferation. Glucose transporters (GLUTs), which facilitate the transport of glucose and related hexoses across the cell membrane, play a vital role in tumor cell survival and are overexpressed in various cancers. GLUT1, the most overexpressed GLUT in many cancers, is emerging as a promising anti-cancer target. To develop GLUT1 inhibitors, we rationally designed, synthesized, structurally characterized, and biologically evaluated in-vitro and in-vivo a novel series of furyl-2-methylene thiazolidinediones (TZDs). Among 25 TZDs tested, F18 and F19 inhibited GLUT1 most potently (IC50 11.4 and 14.7 μM, respectively). F18 was equally selective for GLUT4 (IC50 6.8 μM), while F19 was specific for GLUT1 (IC50 152 μM in GLUT4). In-silico ligand docking studies showed that F18 interacted with conserved residues in GLUT1 and GLUT4, while F19 had slightly different interactions with the transporters. In in-vitro antiproliferative screening of leukemic/lymphoid cells, F18 was most lethal to CEM cells (CC50 of 1.7 μM). Flow cytometry analysis indicated that F18 arrested cell cycle growth in the subG0-G1 phase and lead to cell death due to necrosis and apoptosis. Western blot analysis exhibited alterations in cell signaling proteins, consistent with cell growth arrest and death. In-vivo xenograft study in a CEM model showed that F18 impaired tumor growth significantly.
Collapse
Affiliation(s)
- Kalpana Tilekar
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth's College of Pharmacy, Navi Mumbai, Maharashtra, India
| | - Neha Upadhyay
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth's College of Pharmacy, Navi Mumbai, Maharashtra, India
| | - Jessica D Hess
- The Cytometry, Screening and Imaging Core Facility & Border Biomedical Research Center & Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, USA
| | - Lucasantiago Henze Macias
- The Cytometry, Screening and Imaging Core Facility & Border Biomedical Research Center & Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, USA
| | - Piotr Mrowka
- Department of Biophysics and Human Physiology, Medical University of Warsaw, Chalubinskiego, Warsaw, Poland
| | - Renato J Aguilera
- The Cytometry, Screening and Imaging Core Facility & Border Biomedical Research Center & Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, USA
| | - Franz-Josef Meyer-Almes
- Department of Chemical Engineering and Biotechnology, University of Applied Sciences, Darmstadt, Germany
| | - Cristina V Iancu
- East Carolina Diabetes and Obesity Institute, Department of Chemistry, East Carolina University, Greenville, NC, USA
| | - Jun-Yong Choe
- East Carolina Diabetes and Obesity Institute, Department of Chemistry, East Carolina University, Greenville, NC, USA; Department of Biochemistry and Molecular Biology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA.
| | - C S Ramaa
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth's College of Pharmacy, Navi Mumbai, Maharashtra, India.
| |
Collapse
|
8
|
Permuted 2,4-thiazolidinedione (TZD) analogs as GLUT inhibitors and their in-vitro evaluation in leukemic cells. Eur J Pharm Sci 2020; 154:105512. [PMID: 32801003 DOI: 10.1016/j.ejps.2020.105512] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/24/2020] [Accepted: 08/10/2020] [Indexed: 01/04/2023]
Abstract
Cancer is a heterogeneous disease, and its treatment requires the identification of new ways to thwart tumor cells. Amongst such emerging targets are glucose transporters (GLUTs, SLC2 family), which are overexpressed by almost all types of cancer cells; their inhibition provides a strategy to disrupt tumor metabolism selectively, leading to antitumor effects. Here, novel thiazolidinedione (TZD) derivatives were designed, synthesized, characterized, and evaluated for their GLUT1, GLUT4, and GLUT5 inhibitory potential, followed by in-vitro cytotoxicity determination in leukemic cell lines. Compounds G5, G16, and G17 inhibited GLUT1, with IC50 values of 5.4 ± 1.3, 26.6 ± 1.8, and 12.6 ± 1.2 μM, respectively. G17 was specific for GLUT1, G16 inhibited GLUT4 (IC50 = 21.6 ± 4.5 μM) comparably but did not affect GLUT5. The most active compound, G5, inhibited all three GLUT types, with GLUT4 IC50 = 9.5 ± 2.8 μM, and GLUT5 IC50 = 34.5 ± 2.4 μM. Docking G5, G16, and G17 to the inward- and outward-facing structural models of GLUT1 predicted ligand binding affinities consistent with the kinetic inhibition data and implicated E380 and W388 of GLUT1 vs. their substitutions in GLUT5 (A388 and A396, respectively) in inhibitor preference for GLUT1. G5 inhibited the proliferation of leukemia CEM cells at low micromolar range (IC50 = 13.4 μM) while being safer for normal blood cells. Investigation of CEM cell cycle progression after treatment with G5 showed that cells accumulated in the G2/M phase. Flow cytometric apoptosis studies revealed that compound G5 induced both early and late-stage apoptosis in CEM cells.
Collapse
|
9
|
Upadhyay N, Tilekar K, Jänsch N, Schweipert M, Hess JD, Henze Macias L, Mrowka P, Aguilera RJ, Choe JY, Meyer-Almes FJ, Ramaa CS. Discovery of novel N-substituted thiazolidinediones (TZDs) as HDAC8 inhibitors: in-silico studies, synthesis, and biological evaluation. Bioorg Chem 2020; 100:103934. [PMID: 32446120 PMCID: PMC7302971 DOI: 10.1016/j.bioorg.2020.103934] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/11/2020] [Accepted: 05/11/2020] [Indexed: 01/01/2023]
Abstract
Epigenetics plays a fundamental role in cancer progression, and developing agents that regulate epigenetics is crucial for cancer management. Among Class I and Class II HDACs, HDAC8 is one of the essential epigenetic players in cancer progression. Therefore, we designed, synthesized, purified, and structurally characterized novel compounds containing N-substituted TZD (P1-P25). Cell viability assay of all compounds on leukemic cell lines (CEM, K562, and KCL22) showed the cytotoxic potential of P8, P9, P10, P12, P19, and P25. In-vitro screening of different HDACs isoforms revealed that P19 was the most potent and selective inhibitor for HDAC8 (IC50 - 9.3 μM). Thermal shift analysis (TSA) confirmed the binding of P19 to HDAC8. In-vitro screening of all compounds on the transport activity of GLUT1, GLUT4, and GLUT5 indicated that P19 inhibited GLUT1 (IC50 - 28.2 μM). P10 and P19 induced apoptotic cell death in CEM cells (55.19% and 60.97% respectively) and P19 was less cytotoxic on normal WBCs (CC50 - 104.2 μM) and human fibroblasts (HS27) (CC50 - 105.0 μM). Thus, among this novel series of TZD derivatives, compound P19 was most promising HDAC8 inhibitor and cytotoxic on leukemic cells. Thus, P19 could serve as a lead for further development of optimized molecules with enhanced selectivity and potency.
Collapse
Affiliation(s)
- Neha Upadhyay
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth's College of Pharmacy, Navi Mumbai, India
| | - Kalpana Tilekar
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth's College of Pharmacy, Navi Mumbai, India
| | - Niklas Jänsch
- Department of Chemical Engineering and Biotechnology, University of Applied Science, Darmstadt, Germany
| | - Markus Schweipert
- Department of Chemical Engineering and Biotechnology, University of Applied Science, Darmstadt, Germany
| | - Jessica D Hess
- The Cellular Characterization and Biorepository Core Facility & Border Biomedical Research Centre & Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, USA
| | - Luca Henze Macias
- The Cellular Characterization and Biorepository Core Facility & Border Biomedical Research Centre & Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, USA
| | - Piotr Mrowka
- Department of Biophysics and Human Physiology, Medical University of Warsaw, Chalubinskiego, Warsaw, Poland; Institute of Hematology and Blood Transfusion, Indira Gandhi St., Warsaw, Poland
| | - Renato J Aguilera
- The Cellular Characterization and Biorepository Core Facility & Border Biomedical Research Centre & Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, USA
| | - Jun-Yong Choe
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834, USA; Department of Biochemistry and Molecular Biology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Franz-Josef Meyer-Almes
- Department of Chemical Engineering and Biotechnology, University of Applied Science, Darmstadt, Germany.
| | - C S Ramaa
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth's College of Pharmacy, Navi Mumbai, India.
| |
Collapse
|
10
|
Hersi F, Omar HA, Al-Qawasmeh RA, Ahmad Z, Jaber AM, Zaher DM, Al-Tel TH. Design and synthesis of new energy restriction mimetic agents: Potent anti-tumor activities of hybrid motifs of aminothiazoles and coumarins. Sci Rep 2020; 10:2893. [PMID: 32076009 PMCID: PMC7031302 DOI: 10.1038/s41598-020-59685-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 02/03/2020] [Indexed: 11/09/2022] Open
Abstract
The incidence of obesity-related diseases like diabetes, cardiovascular diseases, and different types of cancers shed light on the importance of dietary control as preventive and treatment measures. However, long-term dietary control is challenging to achieve in most individuals. The use of energy restriction mimetic agents (ERMAs) as an alternative approach to affect the energy machinery of cancer cells has emerged as a promising approach for cancer therapy. ERMAs limit the high need for energy in rapidly growing tumor cells, with their survival rate strongly dependent on the robust availability of energy. In this context, initial phenotypic screening of an in-house pilot compound library identified a new class of aminothiazole anchored on coumarin scaffold as potent anticancer lead drug candidates with potential activity as ERMA. The identified chemotypes were able to inhibit glucose uptake and increase ROS content in cancer cells. Compounds 9b, 9c, 9i, 11b, and 11c were highly active against colorectal cancer cell lines, HCT116 and HT-29, with half-maximal inhibitory concertation (IC50) range from 0.25 to 0.38 µM. Further biological evaluations of 9b and 9f using Western blotting, caspase activity, glucose uptake, ROS production, and NADPH/NADP levels revealed the ability of these lead drug candidates to induce cancer cell death via, at least in part, energy restriction. Moreover, the assessment of 9b and 9f synergistic activity with cisplatin showed promising outcomes. The current work highlights the significant potential of the lead compounds, 9b, and 9f as potential anticancer agents via targeting the cellular energy machinery in cancer cells.
Collapse
Affiliation(s)
- Fatema Hersi
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, 27272, United Arab Emirates.,College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Hany A Omar
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, 27272, United Arab Emirates. .,College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates. .,Department of Pharmacology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt.
| | - Raed A Al-Qawasmeh
- Department of Chemistry, Faculty of Science, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Zainab Ahmad
- Department of Chemistry, Faculty of Science, The University of Jordan, Amman, 11942, Jordan
| | - Areej M Jaber
- Department of Chemistry, Faculty of Science, The University of Jordan, Amman, 11942, Jordan
| | - Dana M Zaher
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, 27272, United Arab Emirates.,College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Taleb H Al-Tel
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, 27272, United Arab Emirates. .,College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates.
| |
Collapse
|
11
|
Mirza AZ, Althagafi II, Shamshad H. Role of PPAR receptor in different diseases and their ligands: Physiological importance and clinical implications. Eur J Med Chem 2019; 166:502-513. [DOI: 10.1016/j.ejmech.2019.01.067] [Citation(s) in RCA: 187] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 01/27/2019] [Accepted: 01/28/2019] [Indexed: 12/15/2022]
|
12
|
A Rise in ATP, ROS, and Mitochondrial Content upon Glucose Withdrawal Correlates with a Dysregulated Mitochondria Turnover Mediated by the Activation of the Protein Deacetylase SIRT1. Cells 2018; 8:cells8010011. [PMID: 30591661 PMCID: PMC6356350 DOI: 10.3390/cells8010011] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/17/2018] [Accepted: 12/20/2018] [Indexed: 11/17/2022] Open
Abstract
Glucose withdrawal has been used as a model for the study of homeostatic defense mechanisms, especially for how cells cope with a shortage of nutrient supply by enhancing catabolism. However, detailed cellular responses to glucose withdrawal have been poorly studied, and are controversial. In this study, we determined how glucose withdrawal affects mitochondrial activity, and the quantity and the role of SIRT1 in these changes. The results of our study indicate a substantial increase in ATP production from mitochondria, through an elevation of mitochondrial biogenesis, mediated by SIRT1 activation that is driven by increased NAD⁺/NADH ratio. Moreover, mitochondria persisted in the cells as elongated forms, and apparently evaded mitophagic removal. This led to a steady increase in mitochondria content and the reactive oxygen species (ROS) generated from them, indicating failure in ATP and ROS homeostasis, due to a misbalance in SIRT1-mediated mitochondria turnover in conditions of glucose withdrawal. Our results suggest that SIRT1 activation alone cannot properly manage energy homeostasis under certain metabolic crisis conditions.
Collapse
|
13
|
Protein N-glycosylation alteration and glycolysis inhibition both contribute to the antiproliferative action of 2-deoxyglucose in breast cancer cells. Breast Cancer Res Treat 2018; 171:581-591. [DOI: 10.1007/s10549-018-4874-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 07/01/2018] [Indexed: 12/12/2022]
|
14
|
Pham J, Arul Nambi Rajan K, Li P, Parast MM. The role of Sirtuin1-PPARγ axis in placental development and function. J Mol Endocrinol 2018; 60:R201-R212. [PMID: 29467141 PMCID: PMC8584848 DOI: 10.1530/jme-17-0315] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 02/21/2018] [Indexed: 12/11/2022]
Abstract
Placental development is important for proper in utero growth and development of the fetus, as well as maternal well-being during pregnancy. Abnormal differentiation of placental epithelial cells, called trophoblast, is at the root of multiple pregnancy complications, including miscarriage, the maternal hypertensive disorder preeclampsia and intrauterine growth restriction. The ligand-activated nuclear receptor, PPARγ, and nutrient sensor, Sirtuin-1, both play a role in numerous pathways important to cell survival and differentiation, metabolism and inflammation. However, each has also been identified as a key player in trophoblast differentiation and placental development. This review details these studies, and also describes how various stressors, including hypoxia and inflammation, alter the expression or activity of PPARγ and Sirtuin-1, thereby contributing to placenta-based pregnancy complications.
Collapse
Affiliation(s)
- Jonathan Pham
- Department of PathologyUniversity of California San Diego, La Jolla, California, USA
- Sanford Consortium for Regenerative MedicineUniversity of California San Diego, La Jolla, California, USA
| | - Kanaga Arul Nambi Rajan
- Department of PathologyUniversity of California San Diego, La Jolla, California, USA
- Sanford Consortium for Regenerative MedicineUniversity of California San Diego, La Jolla, California, USA
| | - Ping Li
- Department of PathologyMedical School of Jinan University, Guangzhou, China
| | - Mana M Parast
- Department of PathologyUniversity of California San Diego, La Jolla, California, USA
- Sanford Consortium for Regenerative MedicineUniversity of California San Diego, La Jolla, California, USA
| |
Collapse
|
15
|
Shinde DN, Trivedi R, Vamsi Krishna N, Lingamallu G, Sridhar B, Khursade PS, Reddy Shetty P. 2,4-Thiazolidinedione as a Bioactive Linker for Ferrocenyl Sugar-Triazole Conjugates: Synthesis, Characterization and Biological Properties. Eur J Inorg Chem 2018. [DOI: 10.1002/ejic.201800006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Dilip N. Shinde
- Inorganic and Physical Chemistry Division; CSIR-Indian Institute of Chemical Technology; 500007 Hyderabad Telangana India
- Academy of Scientific and Innovative Research (AcSIR); CSIR-IICT Campus; 500007 Hyderabad Telangana India
| | - Rajiv Trivedi
- Inorganic and Physical Chemistry Division; CSIR-Indian Institute of Chemical Technology; 500007 Hyderabad Telangana India
- Academy of Scientific and Innovative Research (AcSIR); CSIR-IICT Campus; 500007 Hyderabad Telangana India
| | - Narra Vamsi Krishna
- Inorganic and Physical Chemistry Division; CSIR-Indian Institute of Chemical Technology; 500007 Hyderabad Telangana India
| | - Giribabu Lingamallu
- Inorganic and Physical Chemistry Division; CSIR-Indian Institute of Chemical Technology; 500007 Hyderabad Telangana India
- Academy of Scientific and Innovative Research (AcSIR); CSIR-IICT Campus; 500007 Hyderabad Telangana India
| | - Balasubramanian Sridhar
- Center for X-ray Crystallography; CSIR-Indian Institute of Chemical Technology; 500007 Hyderabad Telangana India
| | - Parag S. Khursade
- Medicinal Chemistry and Biotechnology Division; CSIR-Indian Institute of Chemical Technology; 500007 Hyderabad Telangana India
| | - Prakasham Reddy Shetty
- Medicinal Chemistry and Biotechnology Division; CSIR-Indian Institute of Chemical Technology; 500007 Hyderabad Telangana India
| |
Collapse
|
16
|
Shinde DN, Trivedi R, Krishna JVS, Giribabu L, Sridhar B, Khursade PS, Prakasham RS. N-Arylation of ferrocenyl 2,4-thiazolidinedione conjugatesviaa copper-catalysed Chan–Lam cross coupling reaction with aryl boronic acids and their optoelectronic properties. NEW J CHEM 2018. [DOI: 10.1039/c8nj01598h] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Copper catalysed Chan–LamN-arylation of ferrocenyl 2,4-thiazolidinedione conjugates is described.
Collapse
Affiliation(s)
- Dilip Nivrutti Shinde
- Catalysis and Fine Chemicals Division
- CSIR-Indian Institute of Chemical Technology
- Hyderabad
- India
- Academy of Scientific and Innovative Research (AcSIR)
| | - Rajiv Trivedi
- Catalysis and Fine Chemicals Division
- CSIR-Indian Institute of Chemical Technology
- Hyderabad
- India
- Academy of Scientific and Innovative Research (AcSIR)
| | - Jonnadula V. S. Krishna
- Catalysis and Fine Chemicals Division
- CSIR-Indian Institute of Chemical Technology
- Hyderabad
- India
| | - L. Giribabu
- Catalysis and Fine Chemicals Division
- CSIR-Indian Institute of Chemical Technology
- Hyderabad
- India
- Academy of Scientific and Innovative Research (AcSIR)
| | - B. Sridhar
- Centre for X-ray Crystallography
- CSIR-Indian Institute of Chemical Technology
- Hyderabad 500007
- India
| | - Parag S. Khursade
- Organic Synthesis and Process Chemistry Division
- CSIR-Indian Institute of Chemical Technology
- Hyderabad 500007
- India
| | - R. S. Prakasham
- Organic Synthesis and Process Chemistry Division
- CSIR-Indian Institute of Chemical Technology
- Hyderabad 500007
- India
| |
Collapse
|
17
|
Szychowski KA, Leja ML, Kaminskyy DV, Kryshchyshyn AP, Binduga UE, Pinyazhko OR, Lesyk RB, Tobiasz J, Gmiński J. Anticancer properties of 4-thiazolidinone derivatives depend on peroxisome proliferator-activated receptor gamma (PPARγ). Eur J Med Chem 2017; 141:162-168. [DOI: 10.1016/j.ejmech.2017.09.071] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 09/24/2017] [Accepted: 09/29/2017] [Indexed: 10/18/2022]
|
18
|
Ndombera FT. Anti-cancer agents and reactive oxygen species modulators that target cancer cell metabolism. PURE APPL CHEM 2017. [DOI: 10.1515/pac-2016-1219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
AbstractTraditionally the perspective on reactive oxygen species (ROS) has centered on the role they play as carcinogenic or cancer-causing radicals. Over the years, characterization and functional studies have revealed the complexity of ROS as signaling molecules that regulate various physiological cellular responses or whose levels are altered in various diseases. Cancer cells often maintain high basal level of ROS and are vulnerable to any further increase in ROS levels beyond a certain protective threshold. Consequently, ROS-modulation has emerged as an anticancer strategy with synthesis of various ROS-inducing or responsive agents that target cancer cells. Of note, an increased carbohydrate uptake and/or induction of death receptors of cancer cells was exploited to develop glycoconjugates that potentially induce cellular stress, ROS and apoptosis. This mini review highlights the development of compounds that target cancer cells by taking advantage of redox or metabolic alteration in cancer cells.
Collapse
|
19
|
Yang J, Nishihara R, Zhang X, Ogino S, Qian ZR. Energy sensing pathways: Bridging type 2 diabetes and colorectal cancer? J Diabetes Complications 2017; 31:1228-1236. [PMID: 28465145 PMCID: PMC5501176 DOI: 10.1016/j.jdiacomp.2017.04.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 04/04/2017] [Accepted: 04/10/2017] [Indexed: 12/14/2022]
Abstract
The recently rapid increase of obesity and type 2 diabetes mellitus has caused great burden to our society. A positive association between type 2 diabetes and risk of colorectal cancer has been reported by increasing epidemiological studies. The molecular mechanism of this connection remains elusive. However, type 2 diabetes may result in abnormal carbohydrate and lipid metabolism, high levels of circulating insulin, insulin growth factor-1, and adipocytokines, as well as chronic inflammation. All these factors could lead to the alteration of energy sensing pathways such as the AMP activated kinase (PRKA), mechanistic (mammalian) target of rapamycin (mTOR), SIRT1, and autophagy signaling pathways. The resulted impaired SIRT1 and autophagy signaling pathway could increase the risk of gene mutation and cancer genesis by decreasing genetic stability and DNA mismatch repair. The dysregulated mTOR and PRKA pathway could remodel cell metabolism during the growth and metastasis of cancer in order for the cancer cell to survive the unfavorable microenvironment such as hypoxia and low blood supply. Moreover, these pathways may be coupling metabolic and epigenetic alterations that are central to oncogenic transformation. Further researches including molecular pathologic epidemiologic studies are warranted to better address the precise links between these two important diseases.
Collapse
Affiliation(s)
- Juhong Yang
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Ave., Boston, MA 02215; 211 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Key Laboratory of Hormone and Development (Ministry of Health), Metabolic Disease Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300070, China.
| | - Reiko Nishihara
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Ave., Boston, MA 02215; Division of MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School, 75 Francis Street, Boston, MA 02115; Department of Epidemiology, Harvard School of Public Health, 677 Huntington Ave., Boston, MA 02115
| | - Xuehong Zhang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, 75 Francis Street, Boston, MA 02115
| | - Shuji Ogino
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Ave., Boston, MA 02215; Division of MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School, 75 Francis Street, Boston, MA 02115; Department of Epidemiology, Harvard School of Public Health, 677 Huntington Ave., Boston, MA 02115
| | - Zhi Rong Qian
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Ave., Boston, MA 02215.
| |
Collapse
|
20
|
Moon SH, Lee SJ, Jung KH, Quach CHT, Park JW, Lee JH, Cho YS, Lee KH. Troglitazone Stimulates Cancer Cell Uptake of 18F-FDG by Suppressing Mitochondrial Respiration and Augments Sensitivity to Glucose Restriction. J Nucl Med 2015; 57:129-35. [DOI: 10.2967/jnumed.115.162016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 09/23/2015] [Indexed: 12/23/2022] Open
|
21
|
Omar HA, Tolba MF, Saber-Ayad MM. Potential targets of energy restriction mimetic agents in cancer cells. Future Oncol 2014; 10:2547-50. [DOI: 10.2217/fon.14.191] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Hany A Omar
- Department of Pharmacology, Faculty of Pharmacy, Beni-Suef University, Egypt
- Sharjah Institute for Medical Research, College of Pharmacy, University of Sharjah, Sharjah, UAE
| | - Mai F Tolba
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
- Biology Department, School of Science & Engineering, American University in Cairo, New Cairo, Egypt
| | - Maha M Saber-Ayad
- Sharjah Institute for Medical Research, College of Pharmacy, University of Sharjah, Sharjah, UAE
- Department of Pharmacology, Faculty of Medicine, Cairo University, Kasr Al Ainy, Egypt
| |
Collapse
|
22
|
Thiazolidine-2,4-diones as multi-targeted scaffold in medicinal chemistry: Potential anticancer agents. Eur J Med Chem 2014; 87:814-33. [DOI: 10.1016/j.ejmech.2014.10.025] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 08/22/2014] [Accepted: 10/10/2014] [Indexed: 12/17/2022]
|
23
|
Shin SJ, Kim JY, Kwon SY, Mun KC, Cho CH, Ha E. Ciglitazone enhances ovarian cancer cell death via inhibition of glucose transporter-1. Eur J Pharmacol 2014; 743:17-23. [DOI: 10.1016/j.ejphar.2014.09.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 08/28/2014] [Accepted: 09/07/2014] [Indexed: 02/08/2023]
|
24
|
Lai IL, Chou CC, Lai PT, Fang CS, Shirley LA, Yan R, Mo X, Bloomston M, Kulp SK, Bekaii-Saab T, Chen CS. Targeting the Warburg effect with a novel glucose transporter inhibitor to overcome gemcitabine resistance in pancreatic cancer cells. Carcinogenesis 2014; 35:2203-13. [PMID: 24879635 PMCID: PMC4178465 DOI: 10.1093/carcin/bgu124] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 04/23/2014] [Accepted: 05/16/2014] [Indexed: 01/05/2023] Open
Abstract
Gemcitabine resistance remains a significant clinical challenge. Here, we used a novel glucose transporter (Glut) inhibitor, CG-5, as a proof-of-concept compound to investigate the therapeutic utility of targeting the Warburg effect to overcome gemcitabine resistance in pancreatic cancer. The effects of gemcitabine and/or CG-5 on viability, survival, glucose uptake and DNA damage were evaluated in gemcitabine-sensitive and gemcitabine-resistant pancreatic cancer cell lines. Mechanistic studies were conducted to determine the molecular basis of gemcitabine resistance and the mechanism of CG-5-induced sensitization to gemcitabine. The effects of CG-5 on gemcitabine sensitivity were investigated in a xenograft tumor model of gemcitabine-resistant pancreatic cancer. In contrast to gemcitabine-sensitive pancreatic cancer cells, the resistant Panc-1 and Panc-1(GemR) cells responded to gemcitabine by increasing the expression of ribonucleotide reductase M2 catalytic subunit (RRM2) through E2F1-mediated transcriptional activation. Acting as a pan-Glut inhibitor, CG-5 abrogated this gemcitabine-induced upregulation of RRM2 through decreased E2F1 expression, thereby enhancing gemcitabine-induced DNA damage and inhibition of cell survival. This CG-5-induced inhibition of E2F1 expression was mediated by the induction of a previously unreported E2F1-targeted microRNA, miR-520f. The addition of oral CG-5 to gemcitabine therapy caused greater suppression of Panc-1(GemR) xenograft tumor growth in vivo than either drug alone. Glut inhibition may be an effective strategy to enhance gemcitabine activity for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- I-Lu Lai
- Division of Medicinal Chemistry, College of Pharmacy
| | | | - Po-Ting Lai
- Division of Medicinal Chemistry, College of Pharmacy
| | | | | | - Ribai Yan
- Division of Medicinal Chemistry, College of Pharmacy
| | | | | | | | - Tanios Bekaii-Saab
- Division of Medical Oncology, Department of Internal Medicine, College of Medicine, Ohio State University, Columbus, OH 43210, USA and
| | - Ching-Shih Chen
- Division of Medicinal Chemistry, College of Pharmacy
- Institute of Basic Medical Sciences, National Cheng-Kung University, Tainan 704, Taiwan
| |
Collapse
|
25
|
Chou CC, Salunke SB, Kulp SK, Chen CS. Prospects on strategies for therapeutically targeting oncogenic regulatory factors by small-molecule agents. J Cell Biochem 2014; 115:611-24. [PMID: 24166934 DOI: 10.1002/jcb.24704] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2013] [Accepted: 10/22/2013] [Indexed: 12/12/2022]
Abstract
Although the Human Genome Project has raised much hope for the identification of druggable genetic targets for cancer and other diseases, this genetic target-based approach has not improved productivity in drug discovery over the traditional approach. Analyses of known human target proteins of currently marketed drugs reveal that these drugs target only a limited number of proteins as compared to the whole proteome. In contrast to genome-based targets, mechanistic targets are derived from empirical research, at cellular or molecular levels, in disease models and/or in patients, thereby enabling the exploration of a greater number of druggable targets beyond the genome and epigenome. The paradigm shift has made a tremendous headway in developing new therapeutic agents targeting different clinically relevant mechanisms/pathways in cancer cells. In this Prospects article, we provide an overview of potential drug targets related to the following four emerging areas: (1) tumor metabolism (the Warburg effect), (2) dysregulated protein turnover (E3 ubiquitin ligases), (3) protein-protein interactions, and (4) unique DNA high-order structures and protein-DNA interactions. Nonetheless, considering the genetic and phenotypic heterogeneities that characterize cancer cells, the development of drug resistance in cancer cells by adapting signaling circuitry to take advantage of redundant pathways or feedback/crosstalk systems is possible. This "phenotypic adaptation" underlies the rationale of using therapeutic combinations of these targeted agents with cytotoxic drugs.
Collapse
Affiliation(s)
- Chih-Chien Chou
- Division of Medicinal Chemistry, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | | | | | | |
Collapse
|
26
|
Kuntz S, Mazerbourg S, Boisbrun M, Cerella C, Diederich M, Grillier-Vuissoz I, Flament S. Energy restriction mimetic agents to target cancer cells: comparison between 2-deoxyglucose and thiazolidinediones. Biochem Pharmacol 2014; 92:102-11. [PMID: 25083915 DOI: 10.1016/j.bcp.2014.07.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 07/18/2014] [Accepted: 07/21/2014] [Indexed: 01/02/2023]
Abstract
The use of energy restriction mimetic agents (ERMAs) to selectively target cancer cells addicted to glycolysis could be a promising therapeutic approach. Thiazolidinediones (TZDs) are synthetic agonists of the nuclear receptor peroxisome proliferator-activated receptor (PPAR)γ that were developed to treat type II diabetes. These compounds also display anticancer effects which appear mainly to be independent of their PPARγ agonist activity but the molecular mechanisms involved in the anticancer action are not yet well understood. Results obtained on ciglitazone derivatives, mainly in prostate cancer cell models, suggest that these compounds could act as ERMAs. In the present paper, we introduce how compounds like 2-deoxyglucose target the Warburg effect and then we discuss the possibility that the PPARγ-independent effects of various TZD could result from their action as ERMAs.
Collapse
Affiliation(s)
- Sandra Kuntz
- Université de Lorraine, CRAN, UMR 7039, Vandœuvre-lès-Nancy, F-54506, France; CNRS, CRAN, UMR 7039, Vandœuvre-lès-Nancy, F-54506, France
| | - Sabine Mazerbourg
- Université de Lorraine, CRAN, UMR 7039, Vandœuvre-lès-Nancy, F-54506, France; CNRS, CRAN, UMR 7039, Vandœuvre-lès-Nancy, F-54506, France
| | - Michel Boisbrun
- Université de Lorraine, SRSMC, UMR 7565, Vandœuvre-lès-Nancy, F-54506, France; CNRS, SRSMC, UMR 7565, Vandœuvre-lès-Nancy, F-54506, France
| | - Claudia Cerella
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer. Hôpital Kirchberg, L-2540, Luxembourg
| | - Marc Diederich
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer. Hôpital Kirchberg, L-2540, Luxembourg; Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 151-742, Korea
| | - Isabelle Grillier-Vuissoz
- Université de Lorraine, CRAN, UMR 7039, Vandœuvre-lès-Nancy, F-54506, France; CNRS, CRAN, UMR 7039, Vandœuvre-lès-Nancy, F-54506, France
| | - Stephane Flament
- Université de Lorraine, CRAN, UMR 7039, Vandœuvre-lès-Nancy, F-54506, France; CNRS, CRAN, UMR 7039, Vandœuvre-lès-Nancy, F-54506, France.
| |
Collapse
|
27
|
Bordessa A, Colin-Cassin C, Grillier-Vuissoz I, Kuntz S, Mazerbourg S, Husson G, Vo M, Flament S, Martin H, Chapleur Y, Boisbrun M. Optimization of troglitazone derivatives as potent anti-proliferative agents: towards more active and less toxic compounds. Eur J Med Chem 2014; 83:129-40. [PMID: 24953030 DOI: 10.1016/j.ejmech.2014.06.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 05/16/2014] [Accepted: 06/09/2014] [Indexed: 11/25/2022]
Abstract
Δ2-Troglitazone derivatives were shown to exhibit anti-proliferative activity in a PPARγ-independent manner. We prepared various compounds in order to increase their potency and decrease their toxicity towards non-malignant primary cultured hepatocytes. Many compounds induced viabilities less than 20% at 10 μM on various cancer cell lines. Furthermore, five of them showed hepatocyte viability of 80% or more at 200 μM. In addition, compounds 17 and 18 exhibited promising maximum tolerated doses on a murine model, enabling future investigations.
Collapse
Affiliation(s)
- Andrea Bordessa
- Université de Lorraine, SRSMC, UMR 7565, BP 70239, F-54506 Vandœuvre-lès-Nancy, France; CNRS, SRSMC, UMR 7565, BP 70239, F-54506 Vandœuvre-lès-Nancy, France
| | - Christelle Colin-Cassin
- Université de Lorraine, CRAN, UMR 7039, BP 70239, F-54506 Vandœuvre-lès-Nancy Cedex, France; CNRS, CRAN, UMR 7039, BP 70239, F-54506 Vandœuvre-lès-Nancy Cedex, France
| | - Isabelle Grillier-Vuissoz
- Université de Lorraine, CRAN, UMR 7039, BP 70239, F-54506 Vandœuvre-lès-Nancy Cedex, France; CNRS, CRAN, UMR 7039, BP 70239, F-54506 Vandœuvre-lès-Nancy Cedex, France
| | - Sandra Kuntz
- Université de Lorraine, CRAN, UMR 7039, BP 70239, F-54506 Vandœuvre-lès-Nancy Cedex, France; CNRS, CRAN, UMR 7039, BP 70239, F-54506 Vandœuvre-lès-Nancy Cedex, France
| | - Sabine Mazerbourg
- Université de Lorraine, CRAN, UMR 7039, BP 70239, F-54506 Vandœuvre-lès-Nancy Cedex, France; CNRS, CRAN, UMR 7039, BP 70239, F-54506 Vandœuvre-lès-Nancy Cedex, France
| | - Gauthier Husson
- Université de Lorraine, CRAN, UMR 7039, BP 70239, F-54506 Vandœuvre-lès-Nancy Cedex, France; CNRS, CRAN, UMR 7039, BP 70239, F-54506 Vandœuvre-lès-Nancy Cedex, France
| | - Myriam Vo
- Université de Lorraine, SRSMC, UMR 7565, BP 70239, F-54506 Vandœuvre-lès-Nancy, France; CNRS, SRSMC, UMR 7565, BP 70239, F-54506 Vandœuvre-lès-Nancy, France
| | - Stéphane Flament
- Université de Lorraine, CRAN, UMR 7039, BP 70239, F-54506 Vandœuvre-lès-Nancy Cedex, France; CNRS, CRAN, UMR 7039, BP 70239, F-54506 Vandœuvre-lès-Nancy Cedex, France
| | - Hélène Martin
- Université de Franche-Comté, Laboratoire de Toxicologie Cellulaire, EA 4267, 25030 Besançon Cedex, France
| | - Yves Chapleur
- Université de Lorraine, SRSMC, UMR 7565, BP 70239, F-54506 Vandœuvre-lès-Nancy, France; CNRS, SRSMC, UMR 7565, BP 70239, F-54506 Vandœuvre-lès-Nancy, France
| | - Michel Boisbrun
- Université de Lorraine, SRSMC, UMR 7565, BP 70239, F-54506 Vandœuvre-lès-Nancy, France; CNRS, SRSMC, UMR 7565, BP 70239, F-54506 Vandœuvre-lès-Nancy, France.
| |
Collapse
|
28
|
Abstract
Pyruvate is an obligatory intermediate in the oxidative disposal of glucose and a major precursor for the synthesis of glucose, glycerol, fatty acids, and non-essential amino acids. Stringent control of the fate of pyruvate is critically important for cellular homeostasis. The regulatory mechanisms for its metabolism are therefore of great interest. Recent advances include the findings that (a) the mitochondrial pyruvate carrier is sensitive to inhibition by thiazolidinediones; (b) pyruvate dehydrogenase kinases induce the Warburg effect in many disease states; and (c) pyruvate carboxylase is an important determinate of the rates of gluconeogenesis in humans with type 2 diabetes. These enzymes are potential therapeutic targets for several diseases.
Collapse
Affiliation(s)
- Nam Ho Jeoung
- Department of Fundamental Medical and Pharmaceutical Sciences, Catholic University of Daegu, Gyeongsan, Korea
| | | | | |
Collapse
|
29
|
OSU-CG5, a novel energy restriction mimetic agent, targets human colorectal cancer cells in vitro. Acta Pharmacol Sin 2014; 35:394-400. [PMID: 24464048 DOI: 10.1038/aps.2013.183] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Accepted: 11/27/2013] [Indexed: 12/22/2022]
Abstract
AIM Energy-restriction mimetic agents (ERMAs) are small-molecule agents that target various aspects of energy metabolism, which has emerged as a promising approach in cancer therapy. In the current study, we tested the ability of OSU-CG5, a novel ERMA, to target human colorectal cancer (CRC) in vitro. METHODS Two human CRC cell lines (HCT-116 and Caco-2) were tested. Cell viability was assessed using MTT assay. Caspase-3/7 activities were measured using Caspase-Glo 3/7 assay kit. Western blot analysis was used to measure the expression of relevant proteins in the cells. Glucose consumption of the cells was detected using glucose uptake cell-based assay kit. RESULTS OSU-CG5 dose-dependently inhibited HCT-116 and Caco-2 cell proliferation with the IC₅₀ values of 3.9 and 4.6 μmol/L, respectively, which were 20-25-fold lower than those of resveratrol, a reference ERMA. Both OSU-CG5 (5, 10, and 20 μmol/L) and resveratrol (50, 100, and 200 μmol/L) dose-dependently increased caspase-3/7 activity and PARP level in the cells. Furthermore, both OSU-CG5 and resveratrol induced dose-dependent energy restriction in the cells: they suppressed glucose uptake and Akt phosphorylation, decreased the levels of p-mTOR and p-p70S6K, increased the levels of ER stress response proteins GRP78 and GADD153, and increased the level of β-TrCP, which led to the downregulation of cyclin D1 and Sp1. CONCLUSION OSU-CG5 exhibits promising anti-cancer activity against human CRC cells in vitro, which was, at least in part, due to energy restriction and the consequent induction of ER stress and apoptosis.
Collapse
|
30
|
Colin-Cassin C, Yao X, Cerella C, Chbicheb S, Kuntz S, Mazerbourg S, Boisbrun M, Chapleur Y, Diederich M, Flament S, Grillier-Vuissoz I. PPARγ-inactive Δ2-troglitazone independently triggers ER stress and apoptosis in breast cancer cells. Mol Carcinog 2013; 54:393-404. [PMID: 24293218 DOI: 10.1002/mc.22109] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 10/08/2013] [Accepted: 10/23/2013] [Indexed: 01/02/2023]
Abstract
Our aim was to better understand peroxisome proliferator-activated receptor gamma (PPARγ)-independent pathways involved in anti-cancer effects of thiazolidinediones (TZDs). We focused on Δ2-troglitazone (Δ2-TGZ), a PPARγ inactive TZD that affects breast cancer cell viability. Appearance of TUNEL positive cells, changes in mitochondrial membrane potential, cleavage of poly(ADP-ribose) polymerase (PARP)-1 and caspase-7 revealed that apoptosis occurred in both hormone-dependent MCF7 and hormone-independent MDA-MB-231 breast cancer cells after 24 and 48 h of treatment. A microarray study identified endoplasmic reticulum (ER) stress as an essential cellular function since many genes involved in ER stress were upregulated in MCF7 cells following Δ2-TGZ treatment. Δ2-TGZ-induced ER stress was further confirmed in MCF7 cells by phosphorylation of pancreatic endoplasmic reticulum kinase-like endoplasmic reticulum kinase (PERK) and its target eIF2α after 1.5 h, rapid increase in activating transcription factor (ATF) 3 mRNA levels, splicing of X-box binding protein 1 (XBP1) after 3 h, accumulation of binding immunogloblulin protein (BiP) and CCAAT-enhancer-binding protein homologous protein (CHOP) after 6 h. Immunofluorescence microscopy indicated that CHOP was relocalized to the nucleus of treated cells. Similarly, in MDA-MB-231 cells, overexpression of ATF3, splicing of XBP1, and accumulation of BiP and CHOP were observed following Δ2-TGZ treatment. In MCF7 cells, knock-down of CHOP or the inhibition of c-Jun N-terminal kinase (JNK) did not impair cleavage of PARP-1 and caspase-7. Altogether, our results show that ER stress is an early response of major types of breast cancer cells to Δ2-TGZ, prior to, but not causative of apoptosis.
Collapse
Affiliation(s)
- Christelle Colin-Cassin
- Université de Lorraine, CRAN, UMR 7039, Vandœuvre-lès-Nancy, France; CNRS, CRAN, UMR 7039, Vandœuvre-lès-Nancy, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Venuti V, Cannavà C, Cristiano MC, Fresta M, Majolino D, Paolino D, Stancanelli R, Tommasini S, Ventura CA. A characterization study of resveratrol/sulfobutyl ether-β-cyclodextrin inclusion complex and in vitro anticancer activity. Colloids Surf B Biointerfaces 2013; 115:22-8. [PMID: 24321846 DOI: 10.1016/j.colsurfb.2013.11.025] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 10/18/2013] [Accepted: 11/13/2013] [Indexed: 12/27/2022]
Abstract
A resveratrol/sulfobutylether-β-cyclodextrin inclusion complex was prepared using the freeze-drying method and characterized in solution through UV-vis spectroscopy, solubility phase studies and Job's plot methods. At the solid state it was characterized using the FTIR-ATR technique. Sulfobutylether-β-cyclodextrin has a high affinity for the drug, and forms an inclusion complex with a 1:1 molar ratio both in solution and as a solid sample. It also has a high stability constant (Kc, 10,114 M(-1)). Complexation strongly increases the water solubility of resveratrol (from 0.03 mg/ml to 1.1 mg/ml, at 25 °C) and positively influences its in vitro anticancer activity which was observed on a human breast cancer cell line (MCF-7). In solid phase, FTIR-ATR revealed itself as being a useful technique in elucidating the complexation mechanism, which it did by emphasizing the functional groups involved in the activation of non-covalent "host-guest" interactions.
Collapse
Affiliation(s)
- Valentina Venuti
- Dipartimento di Fisica e di Scienze della Terra, Università degli Studi di Messina, Viale Ferdinando Stagno D'Alcontres 31, I-98166 Messina, Italy
| | - Carmela Cannavà
- Dipartimento di Scienze del Farmaco e Prodotti per la Salute, Università degli Studi di Messina, V.le Annunziata, I-98168 Messina, Italy
| | - Maria Chiara Cristiano
- Dipartimento di Scienze della Salute, Università Magna Graecia di Catanzaro, Viale Europa, I-88100 Catanzaro, Italy
| | - Massimo Fresta
- Dipartimento di Scienze della Salute, Università Magna Graecia di Catanzaro, Viale Europa, I-88100 Catanzaro, Italy
| | - Domenico Majolino
- Dipartimento di Fisica e di Scienze della Terra, Università degli Studi di Messina, Viale Ferdinando Stagno D'Alcontres 31, I-98166 Messina, Italy
| | - Donatella Paolino
- Dipartimento di Scienze della Salute, Università Magna Graecia di Catanzaro, Viale Europa, I-88100 Catanzaro, Italy
| | - Rosanna Stancanelli
- Dipartimento di Scienze del Farmaco e Prodotti per la Salute, Università degli Studi di Messina, V.le Annunziata, I-98168 Messina, Italy
| | - Silvana Tommasini
- Dipartimento di Scienze del Farmaco e Prodotti per la Salute, Università degli Studi di Messina, V.le Annunziata, I-98168 Messina, Italy
| | - Cinzia Anna Ventura
- Dipartimento di Scienze del Farmaco e Prodotti per la Salute, Università degli Studi di Messina, V.le Annunziata, I-98168 Messina, Italy.
| |
Collapse
|
32
|
Woo SR, Byun JG, Kim YH, Park ER, Joo HY, Yun M, Shin HJ, Kim SH, Shen YN, Park JE, Park GH, Lee KH. SIRT1 suppresses cellular accumulation of β-TrCP E3 ligase via protein degradation. Biochem Biophys Res Commun 2013; 441:831-7. [PMID: 24211209 DOI: 10.1016/j.bbrc.2013.10.146] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 10/28/2013] [Indexed: 10/26/2022]
Abstract
β-Transducin repeat-containing protein (β-TrCP), an E3 ligase, promotes the degradation of substrate proteins in response to various stimuli. Even though several β-TrCP substrates have been identified to date, limited information of its upstream regulators is available. Here, we showed that SIRT1 suppresses β-TrCP protein synthesis via post-translational degradation. SIRT1 depletion led to a significant increase in the β-TrCP accumulation without affecting the mRNA level. Consistently, β-TrCP protein accumulation induced by resveratrol was further enhanced upon SIRT1 depletion. Rescue of SIRT1 reversed the effect of resveratrol, leading to reduced β-TrCP protein levels. Proteasomal inhibition led to recovery of β-TrCP in cells with SIRT1 overexpression. Notably, the recovered β-TrCP colocalized mostly with SIRT1. Thus, SIRT1 acts as a negative regulator of β-TrCP synthesis via promoting protein degradation.
Collapse
Affiliation(s)
- Seon Rang Woo
- Division of Radiation Cancer Research, Korea Institute of Radiological & Medical Sciences, Seoul 139-706, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Tuccinardi T, Granchi C, Iegre J, Paterni I, Bertini S, Macchia M, Martinelli A, Qian Y, Chen X, Minutolo F. Oxime-based inhibitors of glucose transporter 1 displaying antiproliferative effects in cancer cells. Bioorg Med Chem Lett 2013; 23:6923-7. [PMID: 24200808 DOI: 10.1016/j.bmcl.2013.09.037] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 09/09/2013] [Accepted: 09/11/2013] [Indexed: 01/04/2023]
Abstract
An analysis of the main pharmacophoric features present in the still limited number of inhibitors of glucose transporter GLUT1 led to the identification of new oxime-based inhibitors, which proved to be able to efficiently hinder glucose uptake and cell growth in H1299 lung cancer cells. The most important interactions of a representative inhibitor were indicated by a novel computational model of GLUT1, which was purposely developed to explain these results and to provide useful indications for the design and the development of new and more efficient GLUT1 inhibitors.
Collapse
Affiliation(s)
- Tiziano Tuccinardi
- Dipartimento di Farmacia, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Cheng G, Zielonka J, McAllister DM, Mackinnon AC, Joseph J, Dwinell MB, Kalyanaraman B. Mitochondria-targeted vitamin E analogs inhibit breast cancer cell energy metabolism and promote cell death. BMC Cancer 2013; 13:285. [PMID: 23764021 PMCID: PMC3686663 DOI: 10.1186/1471-2407-13-285] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 06/07/2013] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Recent research has revealed that targeting mitochondrial bioenergetic metabolism is a promising chemotherapeutic strategy. Key to successful implementation of this chemotherapeutic strategy is the use of new and improved mitochondria-targeted cationic agents that selectively inhibit energy metabolism in breast cancer cells, while exerting little or no long-term cytotoxic effect in normal cells. METHODS In this study, we investigated the cytotoxicity and alterations in bioenergetic metabolism induced by mitochondria-targeted vitamin E analog (Mito-chromanol, Mito-ChM) and its acetylated ester analog (Mito-ChMAc). Assays of cell death, colony formation, mitochondrial bioenergetic function, intracellular ATP levels, intracellular and tissue concentrations of tested compounds, and in vivo tumor growth were performed. RESULTS Both Mito-ChM and Mito-ChMAc selectively depleted intracellular ATP and caused prolonged inhibition of ATP-linked oxygen consumption rate in breast cancer cells, but not in non-cancerous cells. These effects were significantly augmented by inhibition of glycolysis. Mito-ChM and Mito-ChMAc exhibited anti-proliferative effects and cytotoxicity in several breast cancer cells with different genetic background. Furthermore, Mito-ChM selectively accumulated in tumor tissue and inhibited tumor growth in a xenograft model of human breast cancer. CONCLUSIONS We conclude that mitochondria-targeted small molecular weight chromanols exhibit selective anti-proliferative effects and cytotoxicity in multiple breast cancer cells, and that esterification of the hydroxyl group in mito-chromanols is not a critical requirement for its anti-proliferative and cytotoxic effect.
Collapse
Affiliation(s)
- Gang Cheng
- Free Radical Research Center and Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
Lamontagne J, Jalbert-Arsenault É, Pepin É, Peyot ML, Ruderman NB, Nolan CJ, Joly E, Madiraju SM, Poitout V, Prentki M. Pioglitazone acutely reduces energy metabolism and insulin secretion in rats. Diabetes 2013; 62:2122-9. [PMID: 23378607 PMCID: PMC3661607 DOI: 10.2337/db12-0428] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Our objective was to determine if the insulin-sensitizing drug pioglitazone acutely reduces insulin secretion and causes metabolic deceleration in vivo independently of change in insulin sensitivity. We assessed glucose homeostasis by hyperinsulinemic-euglycemic and hyperglycemic clamp studies and energy expenditure by indirect calorimetry and biotelemetry in male Wistar and obese hyperinsulinemic Zucker diabetic fatty (ZDF) rats 45 min after a single oral dose of pioglitazone (30 mg/kg). In vivo insulin secretion during clamped hyperglycemia was reduced in both Wistar and ZDF rats after pioglitazone administration. Insulin clearance was slightly increased in Wistar but not in ZDF rats. Insulin sensitivity in Wistar rats assessed by the hyperinsulinemic-euglycemic clamp was minimally affected by pioglitazone at this early time point. Pioglitazone also reduced energy expenditure in Wistar rats without altering respiratory exchange ratio or core body temperature. Glucose-induced insulin secretion (GIIS) and oxygen consumption were reduced by pioglitazone in isolated islets and INS832/13 cells. In conclusion, pioglitazone acutely induces whole-body metabolic slowing down and reduces GIIS, the latter being largely independent of the insulin-sensitizing action of the drug. The results suggest that pioglitazone has direct metabolic deceleration effects on the β-cell that may contribute to its capacity to lower insulinemia and antidiabetic action.
Collapse
Affiliation(s)
- Julien Lamontagne
- Molecular Nutrition Unit and Montreal Diabetes Research Center at the Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, Quebec, Canada
- Department of Biochemistry, Université de Montréal, Montreal, Quebec, Canada
| | - Élise Jalbert-Arsenault
- Molecular Nutrition Unit and Montreal Diabetes Research Center at the Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, Quebec, Canada
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada
| | - Émilie Pepin
- Molecular Nutrition Unit and Montreal Diabetes Research Center at the Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, Quebec, Canada
- Program of Molecular Biology, Université de Montréal, Montreal, Quebec, Canada
| | - Marie-Line Peyot
- Molecular Nutrition Unit and Montreal Diabetes Research Center at the Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, Quebec, Canada
| | - Neil B. Ruderman
- Departments of Medicine and Physiology and Biophysics, Boston University School of Medicine and Diabetes Unit, Section of Endocrinology, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts
| | | | - Erik Joly
- Molecular Nutrition Unit and Montreal Diabetes Research Center at the Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, Quebec, Canada
| | - S.R. Murthy Madiraju
- Molecular Nutrition Unit and Montreal Diabetes Research Center at the Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, Quebec, Canada
| | - Vincent Poitout
- Montreal Diabetes Research Center at the CRCHUM and Departments of Medicine, Biochemistry and Nutrition, Université de Montréal, Montreal, Quebec, Canada
| | - Marc Prentki
- Molecular Nutrition Unit and Montreal Diabetes Research Center at the Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, Quebec, Canada
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada
- Department of Biochemistry, Université de Montréal, Montreal, Quebec, Canada
- Corresponding author: Marc Prentki,
| |
Collapse
|
36
|
Omar HA, Berman-Booty L, Weng JR. Energy restriction: stepping stones towards cancer therapy. Future Oncol 2013; 8:1503-6. [PMID: 23231510 DOI: 10.2217/fon.12.142] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
37
|
Thiazolidinediones are acute, specific inhibitors of the mitochondrial pyruvate carrier. Proc Natl Acad Sci U S A 2013; 110:5422-7. [PMID: 23513224 DOI: 10.1073/pnas.1303360110] [Citation(s) in RCA: 239] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Facilitated pyruvate transport across the mitochondrial inner membrane is a critical step in carbohydrate, amino acid, and lipid metabolism. We report that clinically relevant concentrations of thiazolidinediones (TZDs), a widely used class of insulin sensitizers, acutely and specifically inhibit mitochondrial pyruvate carrier (MPC) activity in a variety of cell types. Respiratory inhibition was overcome with methyl pyruvate, localizing the effect to facilitated pyruvate transport, and knockdown of either paralog, MPC1 or MPC2, decreased the EC50 for respiratory inhibition by TZDs. Acute MPC inhibition significantly enhanced glucose uptake in human skeletal muscle myocytes after 2 h. These data (i) report that clinically used TZDs inhibit the MPC, (ii) validate that MPC1 and MPC2 are obligatory components of facilitated pyruvate transport in mammalian cells, (iii) indicate that the acute effect of TZDs may be related to insulin sensitization, and (iv) establish mitochondrial pyruvate uptake as a potential therapeutic target for diseases rooted in metabolic dysfunction.
Collapse
|
38
|
Thiazolidine-2,4-diones: progress towards multifarious applications. Bioorg Med Chem 2013; 21:1599-620. [PMID: 23419324 DOI: 10.1016/j.bmc.2013.01.029] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 01/08/2013] [Accepted: 01/10/2013] [Indexed: 11/24/2022]
Abstract
The promising activity shown by compounds containing thiazolidine-2,4-dione nucleus in numerous categories such as anti-hyperglycaemics, aldose reductase inhibitors, anti-cancer, anti-inflammatory, anti-arthritics, anti-microbials, etc. has made it an indispensable anchor for development of new therapeutic agents. Varied substituents on the thiazolidine-2,4-dione nucleus have provided a wide spectrum of biological activities. Importance of this nucleus in some activities like, peroxisome proliferator activated receptor γ (PPARγ) agonism and PPARγ-dependent and -independent anti-cancer activities are reviewed separately in literature. Short reviews on biological importance of this nucleus are also known in literature. However, owing to fast development of new drugs possessing thiazolidine-2,4-dione nucleus many research reports are generated in short span of time. So, there is a need to couple the latest information with the earlier information to understand the current status of thiazolidine-2,4-dione nucleus in medicinal chemistry research. In the present review, various derivatives of thiazolidine-2,4-diones with different pharmacological activities are described on the basis of substitution pattern around the nucleus combined with the docking studies performed in the active site of the corresponding receptors with an aim to help medicinal chemists for developing an SAR on thiazolidine-2,4-dione derived compounds for each activity. This discussion will further help in the development of novel thiazolidine-2,4-dione compounds.
Collapse
|
39
|
Barar J, Omidi Y. Dysregulated pH in Tumor Microenvironment Checkmates Cancer Therapy. BIOIMPACTS : BI 2013; 3:149-62. [PMID: 24455478 PMCID: PMC3892734 DOI: 10.5681/bi.2013.036] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 12/05/2013] [Accepted: 12/07/2013] [Indexed: 04/13/2023]
Abstract
INTRODUCTION The dysregulation of pH by cancerous cells of solid tumors is able to create a unique milieu that is in favor of progression, invasion and metastasis as well as chemo-/immuno-resistance traits of solid tumors. Bioelements involved in pH dysregulation provide new set of oncotargets, inhibition of which may result in better clinical outcome. METHODS To study the impacts of pH dysregulation, we investigated the tumor development and progression in relation with Warburg effect, glycolysis and formation of aberrant tumor microenvironment. RESULTS The upregulation of glucose transporter GLUT-1 and several enzymes involve in glycolysis exacerbates this phenomenon. The accumulation of lactic acids in cancer cells provokes upregulation of several transport machineries (MCT-1, NHE-1, CA IX and H(+) pump V-ATPase) resulting in reinforced efflux of proton into extracellular fluid. This deviant event makes pH to be settled at 7.4 and 6.6 respectively in cancer cells cytoplasm and extracellular fluid within the tumor microenvironment, which in return triggers secretion of lysosomal components (various enzymes in acidic milieu with pH 5) into cytoplasm. All these anomalous phenomena make tumor microenvironment (TME) to be exposed to cocktail of various enzymes with acidic pH, upon which extracellular matrix (ECM) can be remodeled and even deformed, resulting in emergence of a complex viscose TME with high interstitial fluid pressure. CONCLUSION It seems that pH dysregulation is able to remodel various physiologic functions and make solid tumors to become much more invasive and metastatic. It also can cause undesired resistance to chemotherapy and immunotherapy. Hence, cancer therapy needs to be reinforced using specific inhibitors of bioelements involved in pH dysregulation of TME in solid tumors.
Collapse
Affiliation(s)
- Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
40
|
Berman-Booty LD, Chu PC, Thomas-Ahner JM, Bolon B, Wang D, Yang T, Clinton SK, Kulp SK, Chen CS. Suppression of prostate epithelial proliferation and intraprostatic progrowth signaling in transgenic mice by a new energy restriction-mimetic agent. Cancer Prev Res (Phila) 2012; 6:232-41. [PMID: 23275006 DOI: 10.1158/1940-6207.capr-12-0057] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Cells undergoing malignant transformation often exhibit a shift in cellular metabolism from oxidative phosphorylation to glycolysis. This glycolytic shift, called the Warburg effect, provides a mechanistic basis for targeting glycolysis to suppress carcinogenesis through the use of dietary caloric restriction and energy restriction-mimetic agents (ERMA). We recently reported the development of a novel class of ERMAs that exhibits high potency in eliciting starvation-associated cellular responses and epigenetic changes in cancer cells though glucose uptake inhibition. The lead ERMA in this class, OSU-CG5, decreases the production of ATP and NADH in LNCaP prostate cancer cells. In this study, we examined the effect of OSU-CG5 on the severity of preneoplastic lesions in male transgenic adenocarcinoma of the mouse prostate (TRAMP) mice. Daily oral treatment with OSU-CG5 at 100 mg/kg from 6 to 10 weeks of age resulted in a statistically significant decrease in the weight of urogenital tract and microdissected dorsal, lateral, and anterior prostatic lobes relative to vehicle controls. The suppressive effect of OSU-CG5 was evidenced by marked decreases in Ki67 immunostaining and proliferating cell nuclear antigen (PCNA) expression in the prostate. OSU-CG5 treatment was not associated with evidence of systemic toxicity. Microarray analysis indicated a central role for Akt, and Western blot analysis showed reduced phosphorylation and/or expression levels of Akt, Src, androgen receptor, and insulin-like growth factor-1 receptor in prostate lobes. These findings support further investigation of OSU-CG5 as a potential chemopreventive agent.
Collapse
Affiliation(s)
- Lisa D Berman-Booty
- College of Pharmacy, 336 Parks Hall, The Ohio State University, 500 West 12th Avenue, Columbus, OH 43210, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Lin HY, Kuo YC, Weng YI, Lai IL, Huang THM, Lin SP, Niu DM, Chen CS. Activation of silenced tumor suppressor genes in prostate cancer cells by a novel energy restriction-mimetic agent. Prostate 2012; 72:1767-78. [PMID: 22539223 PMCID: PMC3867924 DOI: 10.1002/pros.22530] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 03/30/2012] [Indexed: 01/25/2023]
Abstract
BACKGROUND Targeting tumor metabolism by energy restriction-mimetic agents (ERMAs) has emerged as a strategy for cancer therapy/prevention. Evidence suggests a mechanistic link between ERMA-mediated antitumor effects and epigenetic gene regulation. METHODS Microarray analysis showed that a novel thiazolidinedione-derived ERMA, CG-12, and glucose deprivation could suppress DNA methyltransferase (DNMT)1 expression and reactivate DNA methylation-silenced tumor suppressor genes in LNCaP prostate cancer cells. Thus, we investigated the effects of a potent CG-12 derivative, CG-5, vis-à-vis 2-deoxyglucose, glucose deprivation and/or 5-aza-deoxycytidine, on DNMT isoform expression (Western blotting, RT-PCR), DNMT1 transcriptional activation (luciferase reporter assay), and expression of genes frequently hypermethylated in prostate cancer (quantitative real-time PCR). Promoter methylation was assessed by pyrosequencing analysis. SiRNA-mediated knockdown and ectopic expression of DNMT1 were used to validate DNMT1 as a target of CG-5. RESULTS CG-5 and glucose deprivation upregulated the expression of DNA methylation-silenced tumor suppressor genes, including GADD45a, GADD45b, IGFBP3, LAMB3, BASP1, GPX3, and GSTP1, but also downregulated methylated tumor/invasion-promoting genes, including CD44, S100A4, and TACSTD2. In contrast, 5-aza-deoxycytidine induced global reactivation of these genes. CG-5 mediated these epigenetic effects by transcriptional repression of DNMT1, which was associated with reduced expression of Sp1 and E2F1. SiRNA-mediated knockdown and ectopic expression of DNMT1 corroborated DNMT1's role in the modulation of gene expression by CG-5. Pyrosequencing revealed differential effects of CG-5 versus 5-aza-deoxycytidine on promoter methylation in these genes. CONCLUSIONS These findings reveal a previously uncharacterized epigenetic effect of ERMAs on DNA methylation-silenced tumor suppressor genes, which may foster novel strategies for prostate cancer therapy.
Collapse
Affiliation(s)
- Hsiang-Yu Lin
- Division of Medicinal Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, U.S.A
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
- Department of Pediatrics, Mackay Memorial Hospital and Mackay Medicine, Nursing and Management College, Taipei, Taiwan
| | - Yi-Chiu Kuo
- Division of Medicinal Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, U.S.A
| | - Yu-I Weng
- Human Cancer Genetics Program, The Ohio State University, Columbus, OH 43210, U.S.A
| | - I-Lu Lai
- Division of Medicinal Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, U.S.A
| | - Tim H.-M. Huang
- Human Cancer Genetics Program, The Ohio State University, Columbus, OH 43210, U.S.A
| | - Shuan-Pei Lin
- Department of Pediatrics, Mackay Memorial Hospital and Mackay Medicine, Nursing and Management College, Taipei, Taiwan
| | - Dau-Ming Niu
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
- Corresponding authors: Ching-Shih Chen, College of Pharmacy, The Ohio State University, 500 West 12 Avenue, Columbus, OH 43210. Phone: 614-688-4008; Fax: 614-688-8556; . Dau-Ming Niu, Department of Pediatrics, Taipei Veterans General Hospital, No. 201, Section 2, Shih-Pai Road, Taipei 112, Taiwan. Tel & Fax: 886-2-28767181;
| | - Ching-Shih Chen
- Division of Medicinal Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, U.S.A
- Corresponding authors: Ching-Shih Chen, College of Pharmacy, The Ohio State University, 500 West 12 Avenue, Columbus, OH 43210. Phone: 614-688-4008; Fax: 614-688-8556; . Dau-Ming Niu, Department of Pediatrics, Taipei Veterans General Hospital, No. 201, Section 2, Shih-Pai Road, Taipei 112, Taiwan. Tel & Fax: 886-2-28767181;
| |
Collapse
|
42
|
Xu Y, Xia X, Pan H. Active autophagy in the tumor microenvironment: A novel mechanism for cancer metastasis. Oncol Lett 2012; 5:411-416. [PMID: 23420500 PMCID: PMC3573143 DOI: 10.3892/ol.2012.1015] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 10/09/2012] [Indexed: 12/19/2022] Open
Abstract
Autophagy is a lysosomal degradation process which is key for the regulation of the turnover of long-lived or damaged proteins and organelles and which promotes cell survival during nutrient deprivation or other microenvironmental stresses. Current evidence supports the hypothesis that autophagy suppresses tumorigenesis, particularly during the early stages of tumor initiation. However, in established tumors, autophagy promotes survival under stressful conditions during cancer progression and in response to chemotherapy; however, the mechanism by which autophagy influences cancer metastasis remains unknown. In this review, we discuss the capacity of an abnormal tumor environment to induce autophagy and consider how this relates to tumor metastasis and the attractive prospect of manipulating autophagic signaling pathways as potential targets for the treatment of cancer metastasis.
Collapse
Affiliation(s)
- Yinghua Xu
- Departments of Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | | | | |
Collapse
|
43
|
Chu PC, Chuang HC, Kulp SK, Chen CS. The mRNA-stabilizing factor HuR protein is targeted by β-TrCP protein for degradation in response to glycolysis inhibition. J Biol Chem 2012; 287:43639-50. [PMID: 23115237 DOI: 10.1074/jbc.m112.393678] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The mRNA-stabilizing protein HuR acts a stress response protein whose function and/or protein stability are modulated by diverse stress stimuli through posttranslational modifications. Here, we report a novel mechanism by which metabolic stress facilitates proteasomal degradation of HuR in cancer cells. In response to the glucose transporter inhibitor CG-5, HuR translocates to the cytoplasm, where it is targeted by the ubiquitin E3 ligase β-TrCP1 for degradation. The cytoplasmic localization of HuR is facilitated by PKCα-mediated phosphorylation at Ser-318 as the Ser-318 → alanine substitution abolishes the ability of the resulting HuR to bind PKCα and to undergo nuclear export. The mechanistic link between β-TrCP1 and HuR degradation was supported by the ability of ectopically expressed β-TrCP1 to mimic CG-5 to promote HuR degradation and by the protective effect of dominant negative inhibition of β-TrCP1 on HuR ubiquitination and degradation. Substrate targeting of HuR by β-TrCP1 was further verified by coimmunoprecipitation and in vitro GST pull-down assays and by the identification of a β-TrCP1 recognition site. Although HuR does not contain a DSG destruction motif, we obtained evidence that β-TrCP1 recognizes an unconventional motif, (296)EEAMAIAS(304), in the RNA recognition motif 3. Furthermore, mutational analysis indicates that IKKα-dependent phosphorylation at Ser-304 is crucial to the binding of HuR to β-TrCP1. Mechanistically, this HuR degradation pathway differs from that reported for heat shock and hypoxia, which underlies the complexity in the regulation of HuR turnover under different stress stimuli. The ability of glycolysis inhibitors to target the expression of oncogenic proteins through HuR degradation might foster novel strategies for cancer therapy.
Collapse
Affiliation(s)
- Po-Chen Chu
- Division of Medicinal Chemistry, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43221, USA
| | | | | | | |
Collapse
|
44
|
Wei S, Chu PC, Chuang HC, Hung WC, Kulp SK, Chen CS. Targeting the oncogenic E3 ligase Skp2 in prostate and breast cancer cells with a novel energy restriction-mimetic agent. PLoS One 2012; 7:e47298. [PMID: 23071779 PMCID: PMC3470570 DOI: 10.1371/journal.pone.0047298] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 09/12/2012] [Indexed: 11/18/2022] Open
Abstract
Substantial evidence supports the oncogenic role of the E3 ubiquitin ligase S-phase kinase-associated protein 2 (Skp2) in many types of cancers through its ability to target a broad range of signaling effectors for ubiquitination. Thus, this oncogenic E3 ligase represents an important target for cancer drug discovery. In this study, we report a novel mechanism by which CG-12, a novel energy restriction-mimetic agent (ERMA), down-regulates the expression of Skp2 in prostate cancer cells. Pursuant to our previous finding that upregulation of β-transducin repeat-containing protein (β-TrCP) expression represents a cellular response in cancer cells to ERMAs, including CG-12 and 2-deoxyglucose, we demonstrated that this β-TrCP accumulation resulted from decreased Skp2 expression. Evidence indicates that Skp2 targets β-TrCP for degradation via the cyclin-dependent kinase 2-facilitated recognition of the proline-directed phosphorylation motif 412SP. This Skp2 downregulation was attributable to Sirt1-dependent suppression of COP9 signalosome (Csn)5 expression in response to CG-12, leading to increased cullin 1 neddylation in the Skp1-cullin1-F-box protein complex and consequent Skp2 destabilization. Moreover, we determined that Skp2 and β-TrCP are mutually regulated, providing a feedback mechanism that amplifies the suppressive effect of ERMAs on Skp2. Specifically, cellular accumulation of β-TrCP reduced the expression of Sp1, a β-TrCP substrate, which, in turn, reduced Skp2 gene expression. This Skp2-β-TrCP-Sp1 feedback loop represents a novel crosstalk mechanism between these two important F-box proteins in cancer cells with aberrant Skp2 expression under energy restriction, which provides a proof-of-concept that the oncogenic Csn5/Skp2 signaling axis represents a “druggable” target for this novel ERMA.
Collapse
Affiliation(s)
- Shuo Wei
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio, United States of America
| | - Po-Chen Chu
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio, United States of America
| | - Hsiao-Ching Chuang
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio, United States of America
| | - Wen-Chun Hung
- National Institute of Cancer Research, National Health Research Institute, Zhunan, Miaoli County, Taiwan
| | - Samuel K. Kulp
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio, United States of America
| | - Ching-Shih Chen
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio, United States of America
- Institute of Basic Medical Sciences, National Cheng-Kung University, Tainan, Taiwan
- * E-mail:
| |
Collapse
|
45
|
Min KW, Zhang X, Imchen T, Baek SJ. A peroxisome proliferator-activated receptor ligand MCC-555 imparts anti-proliferative response in pancreatic cancer cells by PPARgamma-independent up-regulation of KLF4. Toxicol Appl Pharmacol 2012; 263:225-32. [PMID: 22750490 DOI: 10.1016/j.taap.2012.06.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Revised: 06/19/2012] [Accepted: 06/22/2012] [Indexed: 12/20/2022]
Abstract
MCC-555 is a novel PPARα/γ dual ligand of the thiazolidinedione class and was recently developed as an anti-diabetic drug with unique properties. MCC-555 also has anti-proliferative activity through growth inhibition and apoptosis induction in several cancer cell types. Our group has shown that MCC-555 targets several proteins in colorectal tumorigenesis including nonsteroidal anti-inflammatory drug (NSAID)-activated gene (NAG-1) which plays an important role in chemoprevention responsible for chemopreventive compounds. NAG-1 is a member of the TGF-β superfamily and is involved in tumor progression and development; however, NAG-1's roles in pancreatic cancer have not been studied. In this report, we found that MCC-555 alters not only NAG-1 expression, but also p21 and cyclin D1 expression. NAG-1 and p21 expression was not blocked by PPARγ-specific antagonist GW9662, suggesting that MCC-555-induced NAG-1 and p21 expression is independent of PPARγ activation. However, decreasing cyclin D1 by MCC-555 seems to be affected by PPARγ activation. Further, we found that the GC box located in the NAG-1 promoter play an important role in NAG-1 transactivation by MCC-555. Subsequently, we screened several transcription factors that may bind to the GC box region in the NAG-1 promoter and found that KLF4 potentially binds to this region. Expression of KLF4 precedes NAG-1 and p21 expression in the presence of MCC-555, whereas blocking KLF4 expression using specific KLF4 siRNA showed that both NAG-1 and p21 expression by MCC-555 was blocked. In conclusion, MCC-555's actions on anti-proliferation involve both PPARγ-dependent and -independent pathways, thereby enhancing anti-tumorigenesis in pancreatic cancer cells.
Collapse
Affiliation(s)
- Kyung-Won Min
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA
| | | | | | | |
Collapse
|
46
|
Ho FM, Liao YH, Yang AJ, Lee Chao PD, Hou YC, Huang CT, Lin SR, Lee KR, Huang KC, Lin WW. Anti-atherosclerotic action of Ger-Gen-Chyn-Lian-Tang and AMPK-dependent lipid lowering effect in hepatocytes. JOURNAL OF ETHNOPHARMACOLOGY 2012; 142:175-187. [PMID: 22543166 DOI: 10.1016/j.jep.2012.04.034] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 03/28/2012] [Accepted: 04/10/2012] [Indexed: 05/31/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Ger-Gen-Chyn-Lian-Tang (GGCLT), an officially standardized mixture of Chinese herbal medicines, consists of Puerariae Radix, Scutellariae Radix, Coptidis Rhizoma and Glycyrrhizae Radix in a ratio of 8:3:3:2. In this study, we evaluated the benefits of GGCLT in atherosclerotic progression. METHODS The major constituents of GGCLT were analyzed by HPLC. ApoE-/- mice taken 0.15% cholesterol diet were orally given vehicle or GGCLT (2 g/kg/day) for 12 weeks. Serum levels of lipid and glucose were analyzed, and atherosclerosis was examined by histological analyses. Cultures of vascular smooth muscle cells, hepatocytes and bone marrow-derived macrophages were used to investigate the action mechanisms of GGCLT. RESULTS Our quantitation results indicated that GGCLT contains puerarin, daidzin, daidzein, baicalin, baicalein, wogonin, palmatine, coptisine, berberine and glycyrrhizin. GGCLT decreased serum levels of total cholesterol and LDL, but not TG and HDL in ApoE-/- mice. In parallel, GGCLT treatment reduced atherosclerotic lesions and collagen expression in atheroma plaques. In vascular smooth muscle cells, GGCLT could reduce cell migration, but failed to affect cell viability and proliferation. In hepatocytes, GGCLT can reduce lipid accumulation, and this action was accompanied by the activation of AMPK, upregulation of PPARs, and downregulation of FAS. Pharmacological approach indicated that the latter two events contributing to the anti-lipogenesis is resulting from AMPK pathway, and the lipid lowering effect of GGCLT in hepatocytes is mediated by AMPK and PPARα pathways. Meanwhile, two of the major components of GGCLT, berberine and puerarin, also activated AMPK and decreased lipid accumulation in hepatocytes with berberine of higher efficacy. Besides in hepatocytes, AMPK signaling was also activated by GGCLT in vascular smooth muscle cells and macrophages. CONCLUSIONS These results demonstrate the anti-atherosclerotic action of Chinese medicine mixture GGCLT in ApoE-/- atherosclerotic mouse model. Mechanistic study suggests that activation of AMPK and PPARα in hepatocytes leading to a decrease of lipid formation contributes to the beneficial action of GGCLT in atherosclerosis treatment.
Collapse
Affiliation(s)
- Feng-Ming Ho
- Department of Internal Medicine, Tao-Yuan General Hospital Department of Health the Executive Yuan, Taoyuan, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Wang D, Chu PC, Yang CN, Yan R, Chuang YC, Kulp SK, Chen CS. Development of a novel class of glucose transporter inhibitors. J Med Chem 2012; 55:3827-36. [PMID: 22468970 DOI: 10.1021/jm300015m] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
On the basis of our finding that the antitumor effect of 5-{4-[(1-methylcyclohexyl)methoxy]benzyl}thiazolidine-2,4-dione, a thiazolidinedione peroxisome proliferator-activated receptor (PPAR)γ agonist, was, in part, attributable to its ability to block glucose uptake independently of PPARγ, we used its PPARγ-inactive analogue to develop a novel class of glucose transporter (GLUT) inhibitors. This lead optimization led to compound 30 {5-(4-hydroxy-3-trifluoromethylbenzylidene)-3-[4,4,4-trifluoro-2-methyl-2-(2,2,2-trifluoroethyl)butyl]thiazolidine-2,4-dione} as the optimal agent, which exhibited high antitumor potency through the suppression of glucose uptake (IC(50), 2.5 μM), while not cytotoxic to prostate and mammary epithelial cells. This glucose uptake inhibition was associated with the inhibition of GLUT1 (IC(50), 2 μM). Moreover, the mechanism of antitumor action of compound 30 was validated by its effect on a series of energy restriction-associated cellular responses. Homology modeling analysis suggests that the inhibitory effect of compound 30 on glucose entry was attributable to its ability to bind to the GLUT1 channel at a site distinct from that of glucose.
Collapse
Affiliation(s)
- Dasheng Wang
- Division of Medicinal Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Sikka A, Kaur M, Agarwal C, Deep G, Agarwal R. Metformin suppresses growth of human head and neck squamous cell carcinoma via global inhibition of protein translation. Cell Cycle 2012; 11:1374-82. [PMID: 22421144 DOI: 10.4161/cc.19798] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth leading cancer in the world; the main risk factors are alcohol and tobacco use. Advancements in therapies have yet to improve the prognosis of HNSCC. The connection between diabetes and cancer is being recognized, and metformin has been shown to decrease cancer incidence in diabetic patients. Accordingly, here, for the first time, we investigated metformin's efficacy on the growth and viability of human HNSCC FaDU and Detroit cells. Our results show that metformin treatment (5-20 mM) dose-dependently inhibits the growth of both cell lines. In FaDU cells, metformin caused 18-57% and 35-81% growth inhibition after 48 and 72 h treatments, respectively. Similarly, in Detroit 562 cells, 48 and 72 h metformin treatment resulted in 20-57% and 33-82% inhibition, respectively. Mechanistically, metformin caused G 1 arrest, which coincided with a decrease in the protein levels of CDKs (2, 4 and 6), cyclins (D1 and E) and CDK inhibitors (p15, p16, p18 and p27), but no change in p19 and p21. Metformin also decreased the levels of oncogenic proteins Skp2 and β-Trcp. In other studies, metformin decreased the phosphorylation of 4E-BP1 at Ser65, Thr37/46 and Thr70 sites, but drastically increased the phosphorylation of EF2 at Thr56 and AMPK at Thr172, which results in global translational inhibition. In summary, the observed wide spectrum of mechanistic effects of metformin on HNSCC cells provides support for the anticancer capability of the drug and its potential use in future therapies.
Collapse
Affiliation(s)
- Arron Sikka
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Denver, CO, USA
| | | | | | | | | |
Collapse
|
49
|
Salamone S, Colin C, Grillier-Vuissoz I, Kuntz S, Mazerbourg S, Flament S, Martin H, Richert L, Chapleur Y, Boisbrun M. Synthesis of new troglitazone derivatives: anti-proliferative activity in breast cancer cell lines and preliminary toxicological study. Eur J Med Chem 2012; 51:206-15. [PMID: 22409968 DOI: 10.1016/j.ejmech.2012.02.044] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 02/20/2012] [Accepted: 02/21/2012] [Indexed: 02/08/2023]
Abstract
Breast cancer is the most prevalent cancer in women. The development of resistances to therapeutic agents and the absence of targeted therapy for triple negative breast cancer motivate the search for alternative treatments. With this aim in mind, we synthesised new derivatives of troglitazone, a compound which was formerly used as an anti-diabetic agent and which exhibits anti-proliferative activity on various cancer cell lines. Among the compounds prepared, some displayed micromolar activity against hormone-dependent and hormone-independent breast cancer cells. Furthermore, the influence of the compounds on the viability of primary cultures of human hepatocytes was evaluated. This enabled us to obtain for the first time interesting structure-toxicity relationships in this family of compounds, resulting in 6b and 8b, which show good anti-proliferative activities and poor toxicity towards hepatocytes, compared to troglitazone.
Collapse
Affiliation(s)
- Stéphane Salamone
- Groupe SUCRES, UMR 7565, Nancy-Université-CNRS, BP 70239, F-54506 Vandoeuvre-lès-Nancy, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Kondratyuk TP, Park EJ, Marler LE, Ahn S, Yuan Y, Choi Y, Yu R, van Breemen RB, Sun B, Hoshino J, Cushman M, Jermihov KC, Mesecar AD, Grubbs CJ, Pezzuto JM. Resveratrol derivatives as promising chemopreventive agents with improved potency and selectivity. Mol Nutr Food Res 2011; 55:1249-65. [PMID: 21714126 PMCID: PMC4135049 DOI: 10.1002/mnfr.201100122] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Revised: 05/02/2011] [Accepted: 05/16/2011] [Indexed: 12/14/2022]
Abstract
SCOPE Despite scores of investigations, the actual impact of resveratrol (3,5,4'-trihydroxy-trans-stilbene) on human health, as a dietary component or supplement, remains moot. This is due to many factors, such as relatively low potency, pleiotropic mechanisms, and rapid metabolism. Nonetheless, as a promiscuous molecule that interacts with numerous targets, resveratrol can be viewed as a scaffold for designing structural relatives potentially capable of mediating more intense responses with greater mechanistic stringency. METHODS AND RESULTS We currently report the synthesis and biological evaluation of 92 stilbene analogs. The compounds were tested with in vitro assays for activation of quinone reductase 1, inhibition of quinone reductase 2, nitric oxide production, aromatase, NFκB, 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced ornithine decarboxylase, or cyclooxygenase-1 and -2, quenching of 2,2-diphenyl-1-picrylhydrazyl free radical, interaction with estrogen receptors, and as antiproliferative agents. Several compounds were found to mediate responses with much greater potency than resveratrol; some mediated pleiotropic responses, as is the case with the parent molecule, but others were highly specific or totally inactive. When administered to rats, higher serum concentrations and greater stability was demonstrated with prototype lead molecules. CONCLUSION Owing to structural simplicity, facile syntheses are available for large-scale production. These data support the promise of more advanced development of novel resveratrol derivatives as drug entities.
Collapse
Affiliation(s)
| | - Eun-Jung Park
- College of Pharmacy, University of Hawaii at Hilo, Hilo, Hawaii 96720, USA
| | - Laura E. Marler
- College of Pharmacy, University of Hawaii at Hilo, Hilo, Hawaii 96720, USA
| | - Soyoun Ahn
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois College of Pharmacy, Chicago, Illinois 60612, USA
| | - Yang Yuan
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois College of Pharmacy, Chicago, Illinois 60612, USA
| | - Yongsoo Choi
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois College of Pharmacy, Chicago, Illinois 60612, USA
| | - Rui Yu
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois College of Pharmacy, Chicago, Illinois 60612, USA
| | - Richard B. van Breemen
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois College of Pharmacy, Chicago, Illinois 60612, USA
| | - Bin Sun
- Department of Medicinal Chemistry and Molecular Pharmacology, School of Pharmacy and Pharmaceutical Sciences, and the Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907, USA
| | - Juma Hoshino
- Department of Medicinal Chemistry and Molecular Pharmacology, School of Pharmacy and Pharmaceutical Sciences, and the Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907, USA
| | - Mark Cushman
- Department of Medicinal Chemistry and Molecular Pharmacology, School of Pharmacy and Pharmaceutical Sciences, and the Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907, USA
| | - Katherine C. Jermihov
- Department of Biological Sciences, College of Science, and the Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907, USA
| | - Andrew D. Mesecar
- Department of Biological Sciences, College of Science, and the Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907, USA
| | - Clinton J. Grubbs
- Department of Surgery, Chemoprevention Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - John M. Pezzuto
- College of Pharmacy, University of Hawaii at Hilo, Hilo, Hawaii 96720, USA
| |
Collapse
|