1
|
Villegas-Vázquez EY, Quintas-Granados LI, Cortés H, González-Del Carmen M, Leyva-Gómez G, Rodríguez-Morales M, Bustamante-Montes LP, Silva-Adaya D, Pérez-Plasencia C, Jacobo-Herrera N, Reyes-Hernández OD, Figueroa-González G. Lithium: A Promising Anticancer Agent. Life (Basel) 2023; 13:537. [PMID: 36836894 PMCID: PMC9966411 DOI: 10.3390/life13020537] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/08/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
Lithium is a therapeutic cation used to treat bipolar disorders but also has some important features as an anti-cancer agent. In this review, we provide a general overview of lithium, from its transport into cells, to its innovative administration forms, and based on genomic, transcriptomic, and proteomic data. Lithium formulations such as lithium acetoacetate (LiAcAc), lithium chloride (LiCl), lithium citrate (Li3C6H5O7), and lithium carbonate (Li2CO3) induce apoptosis, autophagy, and inhibition of tumor growth and also participate in the regulation of tumor proliferation, tumor invasion, and metastasis and cell cycle arrest. Moreover, lithium is synergistic with standard cancer therapies, enhancing their anti-tumor effects. In addition, lithium has a neuroprotective role in cancer patients, by improving their quality of life. Interestingly, nano-sized lithium enhances its anti-tumor activities and protects vital organs from the damage caused by lipid peroxidation during tumor development. However, these potential therapeutic activities of lithium depend on various factors, such as the nature and aggressiveness of the tumor, the type of lithium salt, and its form of administration and dosage. Since lithium has been used to treat bipolar disorder, the current study provides an overview of its role in medicine and how this has changed. This review also highlights the importance of this repurposed drug, which appears to have therapeutic cancer potential, and underlines its molecular mechanisms.
Collapse
Affiliation(s)
- Edgar Yebrán Villegas-Vázquez
- Unidad Multidisciplinaria de Investigación Experimental Zaragoza, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México 09230, Mexico
| | | | - Hernán Cortés
- Laboratorio de Medicina Genómica, Departamento de Genómica, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Ciudad de México 14389, Mexico
| | | | - Gerardo Leyva-Gómez
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Miguel Rodríguez-Morales
- Licenciatura en Médico Cirujano, Facultad de Ciencias de la Salud Universidad Anáhuac Norte, Academia de Genética Médica, Naucalpan de Juárez 52786, Mexico
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico
| | | | - Daniela Silva-Adaya
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Instituto Nacional de Neurología y Neurocirugía, Ciudad de México 14269, Mexico
| | - Carlos Pérez-Plasencia
- Laboratorio de Genómica, Instituto Nacional de Cancerología (INCan), Ciudad de México 14080, Mexico
- Laboratorio de Genómica, Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico
| | - Nadia Jacobo-Herrera
- Unidad de Bioquímica, Instituto Nacional de Ciencias Medicas y Nutrición Salvador Zubirán (INCMNSZ), Ciudad de México 14080, Mexico
| | - Octavio Daniel Reyes-Hernández
- Laboratorio de Biología Molecular del Cáncer, Unidad Multidisciplinaria de Investigación Experimental Zaragoza, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México 09230, Mexico
| | - Gabriela Figueroa-González
- Laboratorio de Farmacogenética, Unidad Multidisciplinaria de Investigación Experimental Zaragoza, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México 09230, Mexico
| |
Collapse
|
2
|
Yang C, Zhu B, Zhan M, Hua ZC. Lithium in Cancer Therapy: Friend or Foe? Cancers (Basel) 2023; 15:cancers15041095. [PMID: 36831437 PMCID: PMC9954674 DOI: 10.3390/cancers15041095] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/29/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023] Open
Abstract
Lithium, a trace element important for fetal health and development, is considered a metal drug with a well-established clinical regime, economical production process, and a mature storage system. Several studies have shown that lithium affects tumor development by regulating inositol monophosphate (IMPase) and glycogen synthase kinase-3 (GSK-3). Lithium can also promote proliferation and programmed cell death (PCD) in tumor cells through a number of new targets, such as the nuclear receptor NR4A1 and Hedgehog-Gli. Lithium may increase cancer treatment efficacy while reducing side effects, suggesting that it can be used as an adjunctive therapy. In this review, we summarize the effects of lithium on tumor progression and discuss the underlying mechanisms. Additionally, we discuss lithium's limitations in antitumor clinical applications, including its narrow therapeutic window and potential pro-cancer effects on the tumor immune system.
Collapse
Affiliation(s)
- Chunhao Yang
- School of Biopharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Bo Zhu
- School of Biopharmacy, China Pharmaceutical University, Nanjing 211198, China
- Correspondence: (B.Z.); (Z.-C.H.)
| | - Mingjie Zhan
- School of Biopharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Zi-Chun Hua
- School of Biopharmacy, China Pharmaceutical University, Nanjing 211198, China
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
- Correspondence: (B.Z.); (Z.-C.H.)
| |
Collapse
|
3
|
Bahmad HF, Daher D, Aljamal AA, Elajami MK, Oh KS, Alvarez Moreno JC, Delgado R, Suarez R, Zaldivar A, Azimi R, Castellano A, Sackstein R, Poppiti RJ. Repurposing of Anticancer Stem Cell Drugs in Brain Tumors. J Histochem Cytochem 2021; 69:749-773. [PMID: 34165342 PMCID: PMC8647630 DOI: 10.1369/00221554211025482] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 05/03/2021] [Indexed: 11/22/2022] Open
Abstract
Brain tumors in adults may be infrequent when compared with other cancer etiologies, but they remain one of the deadliest with bleak survival rates. Current treatment modalities encompass surgical resection, chemotherapy, and radiotherapy. However, increasing resistance rates are being witnessed, and this has been attributed, in part, to cancer stem cells (CSCs). CSCs are a subpopulation of cancer cells that reside within the tumor bulk and have the capacity for self-renewal and can differentiate and proliferate into multiple cell lineages. Studying those CSCs enables an increasing understanding of carcinogenesis, and targeting CSCs may overcome existing treatment resistance. One approach to weaponize new drugs is to target these CSCs through drug repurposing which entails using drugs, which are Food and Drug Administration-approved and safe for one defined disease, for a new indication. This approach serves to save both time and money that would otherwise be spent in designing a totally new therapy. In this review, we will illustrate drug repurposing strategies that have been used in brain tumors and then further elaborate on how these approaches, specifically those that target the resident CSCs, can help take the field of drug repurposing to a new level.
Collapse
Affiliation(s)
- Hisham F. Bahmad
- Arkadi M. Rywlin M.D. Department of Pathology
and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach,
Florida
| | - Darine Daher
- Faculty of Medicine, American University of
Beirut, Beirut, Lebanon
| | - Abed A. Aljamal
- Department of Internal Medicine, Mount Sinai
Medical Center, Miami Beach, Florida
| | - Mohamad K. Elajami
- Department of Internal Medicine, Mount Sinai
Medical Center, Miami Beach, Florida
| | - Kei Shing Oh
- Arkadi M. Rywlin M.D. Department of Pathology
and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach,
Florida
| | - Juan Carlos Alvarez Moreno
- Arkadi M. Rywlin M.D. Department of Pathology
and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach,
Florida
| | - Ruben Delgado
- Arkadi M. Rywlin M.D. Department of Pathology
and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach,
Florida
| | - Richard Suarez
- Department of Pathology, Herbert Wertheim
College of Medicine, Florida International University, Miami, Florida
| | - Ana Zaldivar
- Arkadi M. Rywlin M.D. Department of Pathology
and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach,
Florida
| | - Roshanak Azimi
- Arkadi M. Rywlin M.D. Department of Pathology
and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach,
Florida
| | - Amilcar Castellano
- Arkadi M. Rywlin M.D. Department of Pathology
and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach,
Florida
- Department of Pathology, Herbert Wertheim
College of Medicine, Florida International University, Miami, Florida
| | - Robert Sackstein
- Department of Translational Medicine,
Translational Glycobiology Institute, Herbert Wertheim College of Medicine,
Florida International University, Miami, Florida
| | - Robert J. Poppiti
- Arkadi M. Rywlin M.D. Department of Pathology
and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach,
Florida
- Department of Pathology, Herbert Wertheim
College of Medicine, Florida International University, Miami, Florida
| |
Collapse
|
4
|
In Vitro and In Vivo Antitumor Activity of Vitamin D3 in Malignant Gliomas: A Systematic Review. INTERNATIONAL JOURNAL OF CANCER MANAGEMENT 2020. [DOI: 10.5812/ijcm.94542] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
5
|
Emerging therapeutic potential of anti-psychotic drugs in the management of human glioma: A comprehensive review. Oncotarget 2019; 10:3952-3977. [PMID: 31231472 PMCID: PMC6570463 DOI: 10.18632/oncotarget.26994] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 05/13/2019] [Indexed: 12/12/2022] Open
Abstract
Despite numerous advancements in the last decade, human gliomas such as astrocytoma and glioblastoma multiforme have the worst prognoses among all cancers. Anti-psychotic drugs are commonly prescribed to treat mental disorders among cancer patients, and growing empirical evidence has revealed their antitumor, anti-metastatic, anti-angiogenic, anti-proliferative, chemo-preventive, and neo-adjuvant efficacies in various in vitro, in vivo, and clinical glioma models. Anti-psychotic drugs have drawn the attention of physicians and researchers owing to their beneficial effects in the prevention and treatment of gliomas. This review highlights data on the therapeutic potential of various anti-psychotic drugs as anti-proliferative, chemopreventive, and anti-angiogenic agents in various glioma models via the modulation of upstream and downstream molecular targets involved in apoptosis, autophagy, oxidative stress, inflammation, and the cell cycle in in vitro and in vivo preclinical and clinical stages among glioma patients. The ability of anti-psychotic drugs to modulate various signaling pathways and multidrug resistance-conferring proteins that enhance the efficacy of chemotherapeutic drugs with low side-effects exemplifies their great potential as neo-adjuvants and potential chemotherapeutics in single or multimodal treatment approach. Moreover, anti-psychotic drugs confer the ability to induce glioma into oligodendrocyte-like cells and neuronal-like phenotype cells with reversal of epigenetic alterations through inhibition of histone deacetylase further rationalize their use in glioma treatment. The improved understanding of anti-psychotic drugs as potential chemotherapeutic drugs or as neo-adjuvants will provide better information for their use globally as affordable, well-tolerated, and effective anticancer agents for human glioma.
Collapse
|
6
|
Li XX, Guo H, Zhou JD, Wu DH, Ma JC, Wen XM, Zhang W, Xu ZJ, Lin J, Jun Q. Overexpression of CTNNB1: Clinical implication in Chinese de novo acute myeloid leukemia. Pathol Res Pract 2018; 214:361-367. [PMID: 29496308 DOI: 10.1016/j.prp.2018.01.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 12/25/2017] [Accepted: 01/05/2018] [Indexed: 12/28/2022]
Abstract
Activation of Wnt/β-catenin signaling played a crucial role in tumorigenesis, and β-catenin (CTNNB1) overexpression has been identified in numerous solid tumors. The present study was designed to determine CTNNB1 expression and its clinical significance in Chinese de novo acute myeloid leukemia (AML) patients. Real-time quantitative PCR was carried out to detect the pattern of CTNNB1 expression in 140 AML patients and 46 controls. The level of CTNNB1 transcript in AML patients was significantly up-regulated compared with controls (P < 0.001). CTNNB1high patients showed significantly older age than CTNNB1low patients (P < 0.05). The frequency of high CTNNB1 expression was significantly observed in patients with intermediate/poor karyotypes. CTNNB1high patients had a significantly lower complete remission (CR) rate than CTNNB1low patients (P = 0.004). Among cytogenetically normal AML (CN-AML), CTNNB1high patients presented significantly shorter overall survival (OS, P = 0.004) and leukemia-free survival (LFS, P = 0.038) than CTNNB1low patients. Multivariate analysis confirmed that CTNNB1 expression was an independent prognostic factor for OS among CN-AML. Moreover, CTNNB1 expression level significantly decreased after CR stage (P = 0.032) and increased in relapsed stage (P = 0.015). Our findings suggest that CTNNB1 is overexpressed and confers a poor prognosis in AML, and could be used as a biomarker in monitoring disease recurrence.
Collapse
Affiliation(s)
- Xi-Xi Li
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China; The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China; School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Hong Guo
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China; The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China
| | - Jing-Dong Zhou
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China; The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China; School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - De-Hong Wu
- Department of Hematology, The Third People's Hospital of KunShan City, Suzhou, Jiangsu, People's Republic of China
| | - Ji-Chun Ma
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China; The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China
| | - Xiang-Mei Wen
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China; The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China
| | - Wei Zhang
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China; The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China
| | - Zi-Jun Xu
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China; The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China; School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Jiang Lin
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China; The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China
| | - Qian Jun
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China; The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China.
| |
Collapse
|
7
|
Dong Y, Furuta T, Sabit H, Kitabayashi T, Jiapaer S, Kobayashi M, Ino Y, Todo T, Teng L, Hirao A, Zhao SG, Nakada M. Identification of antipsychotic drug fluspirilene as a potential anti-glioma stem cell drug. Oncotarget 2017; 8:111728-111741. [PMID: 29340087 PMCID: PMC5762355 DOI: 10.18632/oncotarget.22904] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 11/15/2017] [Indexed: 12/14/2022] Open
Abstract
Glioma stem cell (GSC)-targeted therapy is expected to be one of the most innovative approaches to treat patients with glioblastoma (GBM). A number of the drugs that restrain the signaling pathway essential for GSC maintenance have been under clinical trials. Here, we identified fluspirilene, a traditional antipsychotic drug, as a GSC-targeting agent, selected from thousands of existing drugs, and investigated its therapeutic effects against GBM with the purpose of drug repositioning. To develop novel therapeutics targeting GSCs, we initially screened drug libraries for small-molecule compounds showing a greater efficacy, compared to that of controls, in inhibiting the proliferation and survival of different GSC lines using cell proliferation assay. Drugs already reported to show therapeutic effects against GBM or those under clinical trials were excluded from subsequent screening. Finally, we found three drugs showing remarkable antiproliferative effects on GSCs at low concentrations and investigated their therapeutic effects on GSCs, glioma cell lines, and in a GBM mouse model. Of the three compounds, fluspirilene demonstrated a significant inhibitory effect on the proliferation and invasion of glioma cells as well as in the model mice treated with the drug. These effects were associated with the inactivation of the signal transducer and activator of transcription 3 (STAT3). Redeveloping of fluspirilene is a promising approach for the treatment of GBM.
Collapse
Affiliation(s)
- Yu Dong
- Department of Neurosurgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Takuya Furuta
- Department of Neurosurgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
| | - Hemragul Sabit
- Department of Neurosurgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Tomohiro Kitabayashi
- Department of Neurosurgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Shabierjiang Jiapaer
- Department of Neurosurgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Masahiko Kobayashi
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Yasushi Ino
- Laboratory of Innovative Cancer Therapy, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Tomoki Todo
- Laboratory of Innovative Cancer Therapy, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Lei Teng
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Atsushi Hirao
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Shi-Guang Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Mitsutoshi Nakada
- Department of Neurosurgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
8
|
Han S, Meng L, Jiang Y, Cheng W, Tie X, Xia J, Wu A. Lithium enhances the antitumour effect of temozolomide against TP53 wild-type glioblastoma cells via NFAT1/FasL signalling. Br J Cancer 2017; 116:1302-1311. [PMID: 28359080 PMCID: PMC5482734 DOI: 10.1038/bjc.2017.89] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 03/10/2017] [Indexed: 12/30/2022] Open
Abstract
Background: We previously showed that activation of the nuclear factor of activated T cells (NFAT)1/Fas ligand (FasL) pathway induces glioma cell death. Lithium (Li) is an inhibitor of glycogen synthase kinase (GSK)-3 that activates NFAT1/FasL signalling. Temozolomide (TMZ) inhibits GSK-3 and activates Fas in tumour protein (TP)53 wild-type (TP53wt) glioma cells. The present study investigated the combinational effects of TMZ and low-dose Li on TP53wt glioma cells. Methods: The combined effect of TMZ and Li was examined in TP53wt U87 and primary glioma cells and a mouse xenograft model. Results: Combination with 1.2 mM Li potentiated TMZ-induced cell death in TP53wt glioma cells, as determined by neurosphere formation and apoptosis assays. Temozolomide combined with Li treatment inhibited GSK-3 activation, promoted NFAT1 nuclear translocation and upregulated Fas/FasL expression. Targeted knockdown of NFAT1 expression blocked the induction of cell death by TMZ and Li via FasL inhibition. In vivo, combined treatment with TMZ and Li suppressed tumour growth and prolonged the survival of tumour-bearing mice. However, the combination of TMZ and Li did not produce a statistically significant effect in TP53mut glioma cells. Conclusions: Temozolomide combined with low-dose Li induces TP53wt glioma cell death via NFAT1/FasL signalling. This represents a potential therapeutic strategy for TP53wt glioma treatment.
Collapse
Affiliation(s)
- Sheng Han
- Department of Neurosurgery, The First Hospital of China Medical University, Nanjing Street 155, Heping District, Shenyang 110001, China
| | - Lingxuan Meng
- Department of Neurosurgery, The First Hospital of China Medical University, Nanjing Street 155, Heping District, Shenyang 110001, China
| | - Yang Jiang
- Department of Neurosurgery, The First Hospital of China Medical University, Nanjing Street 155, Heping District, Shenyang 110001, China
| | - Wen Cheng
- Department of Neurosurgery, The First Hospital of China Medical University, Nanjing Street 155, Heping District, Shenyang 110001, China
| | - Xinxin Tie
- Department of Neurosurgery, The First Hospital of China Medical University, Nanjing Street 155, Heping District, Shenyang 110001, China
| | - Junzhe Xia
- Department of Neurosurgery, The First Hospital of China Medical University, Nanjing Street 155, Heping District, Shenyang 110001, China
| | - Anhua Wu
- Department of Neurosurgery, The First Hospital of China Medical University, Nanjing Street 155, Heping District, Shenyang 110001, China
| |
Collapse
|
9
|
de Araujo WM, Robbs BK, Bastos LG, de Souza WF, Vidal FCB, Viola JPB, Morgado-Diaz JA. PTEN Overexpression Cooperates With Lithium to Reduce the Malignancy and to Increase Cell Death by Apoptosis via PI3K/Akt Suppression in Colorectal Cancer Cells. J Cell Biochem 2016. [PMID: 26224641 DOI: 10.1002/jcb.25294] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Lithium is a well-established non-competitive inhibitor of glycogen synthase kinase-3β (GSK-3β), a kinase that is involved in several cellular processes related to cancer progression. GSK-3β is regulated upstream by PI3K/Akt, which is negatively modulated by PTEN. The role that lithium plays in cancer is controversial because lithium can activate or inhibit survival signaling pathways depending on the cell type. In this study, we analyzed the mechanisms by which lithium can modulate events related to colorectal cancer (CRC) progression and evaluated the role that survival signaling pathways such as PI3K/Akt and PTEN play in this context. We show that the administration of lithium decreased the proliferative potential of CRC cells in a GSK-3β-independent manner but induced the accumulation of cells in G2/M phase. Furthermore, high doses of lithium increased apoptosis, which was accompanied by decreased proteins levels of Akt and PTEN. Then, cells that were induced to overexpress PTEN were treated with lithium; we observed that low doses of lithium strongly increased apoptosis. Additionally, PTEN overexpression reduced proliferation, but this effect was minor compared with that in cells treated with lithium alone. Furthermore, we demonstrated that PTEN overexpression and lithium treatment separately reduced cell migration, colony formation, and invasion, and these effects were enhanced when lithium treatment and PTEN overexpression were combined. In conclusion, our findings indicate that PTEN overexpression and lithium treatment cooperate to reduce the malignancy of CRC cells and highlight lithium and PTEN as potential candidates for studies to identify new therapeutic approaches for CRC treatment.
Collapse
Affiliation(s)
- Wallace Martins de Araujo
- Grupo de Biologia Estrutural, Divisão de Biologia Celular, Centro de Pesquisas, Instituto Nacional de Câncer, Rua André Cavalcanti, 37, 5andar, Rio de Janeiro, Brasil
| | - Bruno Kaufmann Robbs
- Departamento de Ciências Básicas, Campus Universitário de Nova Friburgo, Universidade Federal Fluminense, UFF, Nova Friburgo, Rio de Janeiro, Brasil
| | - Lilian G Bastos
- Grupo de Biologia Estrutural, Divisão de Biologia Celular, Centro de Pesquisas, Instituto Nacional de Câncer, Rua André Cavalcanti, 37, 5andar, Rio de Janeiro, Brasil
| | - Waldemir F de Souza
- Grupo de Biologia Estrutural, Divisão de Biologia Celular, Centro de Pesquisas, Instituto Nacional de Câncer, Rua André Cavalcanti, 37, 5andar, Rio de Janeiro, Brasil
| | - Flávia C B Vidal
- Banco de Tumores e DNA do Maranhão, Universidade Federal do Maranhão, Rua Coelho Neto, 311, São Luís, MA, Brasil
| | - João P B Viola
- Grupo de Regulação Gênica, Programa de Biologia Celular, Centro de Pesquisas, Instituto Nacional de Câncer, Rua André Cavalcanti, 37, 5andar, Rio de Janeiro, Brasil
| | - Jose A Morgado-Diaz
- Grupo de Biologia Estrutural, Divisão de Biologia Celular, Centro de Pesquisas, Instituto Nacional de Câncer, Rua André Cavalcanti, 37, 5andar, Rio de Janeiro, Brasil
| |
Collapse
|
10
|
Rattanawarawipa P, Pavasant P, Osathanon T, Sukarawan W. Effect of lithium chloride on cell proliferation and osteogenic differentiation in stem cells from human exfoliated deciduous teeth. Tissue Cell 2016; 48:425-31. [PMID: 27590780 DOI: 10.1016/j.tice.2016.08.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 08/11/2016] [Accepted: 08/19/2016] [Indexed: 12/11/2022]
Abstract
Lithium Chloride (LiCl) has been used as a canonical Wnt pathway activator due to its ability to inhibit a glycogen synthase kinase-3. The aim of the present study was to investigate the effect of LiCl on cell proliferation and osteogenic differentiation in stem cells isolated from human exfoliated deciduous teeth (SHEDs). SHEDs were isolated and cultured in media supplemented with LiCl at 5, 10, or 20mM. The results demonstrated that LiCl significantly decreased SHEDs colony forming unit ability in a dose dependent manner. LiCl significantly enhanced the percentage of cells in the sub G0 phase, accompanied by a reduction of the percentage of cells in the G1 phase at day 3 and 7 after treatment. Further, LiCl markedly decreased OSX and DMP1 mRNA expression after treating SHEDs in an osteogenic induction medium for 7 days. In addition, no significant difference in alkaline phosphatase enzymatic activity or mineral deposition was found. Together, these results imply that LiCl influences SHEDs behavior.
Collapse
Affiliation(s)
- Panarat Rattanawarawipa
- Department of Pediatric Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330 Thailand; Mineralized Tissue Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330 Thailand
| | - Prasit Pavasant
- Mineralized Tissue Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330 Thailand; Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330 Thailand
| | - Thanaphum Osathanon
- Mineralized Tissue Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330 Thailand; Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330 Thailand
| | - Waleerat Sukarawan
- Department of Pediatric Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330 Thailand; Mineralized Tissue Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330 Thailand.
| |
Collapse
|
11
|
Elmaci İ, Altinoz MA. A Metabolic Inhibitory Cocktail for Grave Cancers: Metformin, Pioglitazone and Lithium Combination in Treatment of Pancreatic Cancer and Glioblastoma Multiforme. Biochem Genet 2016; 54:573-618. [PMID: 27377891 DOI: 10.1007/s10528-016-9754-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 06/23/2016] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer (PC) and glioblastoma multiforme (GBM) are among the human cancers with worst prognosis which require an urgent need for efficient therapies. Here, we propose to apply to treat both malignancies with a triple combination of drugs, which are already in use for different indications. Recent studies demonstrated a considerable link between risk of PC and diabetes. In experimental models, anti-diabetogenic agents suppress growth of PC, including metformin (M), pioglitazone (P) and lithium (L). L is used in psychiatric practice, yet also bears anti-diabetic potential and selectively inhibits glycogen synthase kinase-3 beta (GSK-3β). M, a biguanide class anti-diabetic agent shows anticancer activity via activating AMP-activated protein kinase (AMPK). Glitazones bind to PPAR-γ and inhibit NF-κB, triggering cell proliferation, apoptosis resistance and synthesis of inflammatory cytokines in cancer cells. Inhibition of inflammatory cytokines could simultaneously decrease tumor growth and alleviate cancer cachexia, having a major role in PC mortality. Furthermore, mutual synergistic interactions exist between PPAR-γ and GSK-3β, between AMPK and GSK-3β and between AMPK and PPAR-γ. In GBM, M blocks angiogenesis and migration in experimental models. Very noteworthy, among GBM patients with type 2 diabetes, usage of M significantly correlates with better survival while reverse is true for sulfonylureas. In experimental models, P synergies with ligands of RAR, RXR and statins in reducing growth of GBM. Further, usage of P was found to be lesser in anaplastic astrocytoma and GBM patients, indicating a protective effect of P against high-grade gliomas. L is accumulated in GBM cells faster and higher than in neuroblastoma cells, and its levels further increase with chronic exposure. Recent studies revealed anti-invasive potential of L in GBM cell lines. Here, we propose that a triple-agent regime including drugs already in clinical usage may provide a metabolic adjuvant therapy for PC and GBM.
Collapse
Affiliation(s)
- İlhan Elmaci
- Department of Neurosurgery, Memorial Hospital, Istanbul, Turkey
- Neuroacademy Group, Istanbul, Turkey
| | - Meric A Altinoz
- Department of Immunology, Experimental Medicine Research Center, Istanbul, Turkey.
| |
Collapse
|
12
|
Castillo-Quan JI, Kinghorn KJ, Bjedov I. Genetics and pharmacology of longevity: the road to therapeutics for healthy aging. ADVANCES IN GENETICS 2015; 90:1-101. [PMID: 26296933 DOI: 10.1016/bs.adgen.2015.06.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Aging can be defined as the progressive decline in tissue and organismal function and the ability to respond to stress that occurs in association with homeostatic failure and the accumulation of molecular damage. Aging is the biggest risk factor for human disease and results in a wide range of aging pathologies. Although we do not completely understand the underlying molecular basis that drives the aging process, we have gained exceptional insights into the plasticity of life span and healthspan from the use of model organisms such as the worm Caenorhabditis elegans and the fruit fly Drosophila melanogaster. Single-gene mutations in key cellular pathways that regulate environmental sensing, and the response to stress, have been identified that prolong life span across evolution from yeast to mammals. These genetic manipulations also correlate with a delay in the onset of tissue and organismal dysfunction. While the molecular genetics of aging will remain a prosperous and attractive area of research in biogerontology, we are moving towards an era defined by the search for therapeutic drugs that promote healthy aging. Translational biogerontology will require incorporation of both therapeutic and pharmacological concepts. The use of model organisms will remain central to the quest for drug discovery, but as we uncover molecular processes regulated by repurposed drugs and polypharmacy, studies of pharmacodynamics and pharmacokinetics, drug-drug interactions, drug toxicity, and therapeutic index will slowly become more prevalent in aging research. As we move from genetics to pharmacology and therapeutics, studies will not only require demonstration of life span extension and an underlying molecular mechanism, but also the translational relevance for human health and disease prevention.
Collapse
Affiliation(s)
- Jorge Iván Castillo-Quan
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK; Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Kerri J Kinghorn
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK; Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Ivana Bjedov
- Cancer Institute, University College London, London, UK
| |
Collapse
|
13
|
Fu Y, Jiao Y, Zheng S, Liang A, Hu F. Combination of lithium chloride and pEGFP-N1-BmK CT effectively decreases proliferation and migration of C6 glioma cells. Cytotechnology 2014; 68:197-202. [PMID: 25286828 DOI: 10.1007/s10616-014-9768-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 07/01/2014] [Indexed: 12/20/2022] Open
Abstract
Deleterious invasiveness of glioma cells into the normal brain tissue is endorsed by its inherent ability to regulate the receptor-mediated adhesive properties, extracellular matrix degradation and remodeling and elevated secretory ability of metalloproteinase (MMPs) such as MMP-2. By doing so, it will create an intercellular space for the invasion of glioma cells. Here, we reported that combination of gene therapy Buthus martensii Karsch (BmK) CT, a type of scorpion toxin peptide, with lithium chloride (LiCl), clinically used as mood stabilizer, could inhibit the migration and invasion of C6 glioma cells. The results showed that concomitant administration of LiCl and pEGFP-N1-BmK CT on glioma cells would hamper pro-MMP2 secretion and in the meantime, inhibited its proliferation in a synergistic manner. These results try to extrapolate the potential interplay between the combined treatment of LiCl and BmK CT with signaling pathways β-catenin, MMP, GSK-3 in C6 glioma cells. This strategy can stand for a novel approach designated for the development of a new method for glioma therapy.
Collapse
Affiliation(s)
- Yuejun Fu
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan, 030006, People's Republic of China.
| | - Yanmei Jiao
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan, 030006, People's Republic of China
| | - Shuhua Zheng
- University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Aihua Liang
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan, 030006, People's Republic of China
| | - Fengyun Hu
- Department of Neurology, Shanxi Provincial People's Hospital, Taiyuan, 030012, People's Republic of China.
| |
Collapse
|