1
|
Yan J, Xie Y, Si J, Gan L, Li H, Sun C, Di C, Zhang J, Huang G, Zhang X, Zhang H. Crosstalk of the Caspase Family and Mammalian Target of Rapamycin Signaling. Int J Mol Sci 2021; 22:E817. [PMID: 33467535 PMCID: PMC7830632 DOI: 10.3390/ijms22020817] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 12/20/2022] Open
Abstract
Cell can integrate the caspase family and mammalian target of rapamycin (mTOR) signaling in response to cellular stress triggered by environment. It is necessary here to elucidate the direct response and interaction mechanism between the two signaling pathways in regulating cell survival and determining cell fate under cellular stress. Members of the caspase family are crucial regulators of inflammation, endoplasmic reticulum stress response and apoptosis. mTOR signaling is known to mediate cell growth, nutrition and metabolism. For instance, over-nutrition can cause the hyperactivation of mTOR signaling, which is associated with diabetes. Nutrition deprivation can inhibit mTOR signaling via SH3 domain-binding protein 4. It is striking that Ras GTPase-activating protein 1 is found to mediate cell survival in a caspase-dependent manner against increasing cellular stress, which describes a new model of apoptosis. The components of mTOR signaling-raptor can be cleaved by caspases to control cell growth. In addition, mTOR is identified to coordinate the defense process of the immune system by suppressing the vitality of caspase-1 or regulating other interferon regulatory factors. The present review discusses the roles of the caspase family or mTOR pathway against cellular stress and generalizes their interplay mechanism in cell fate determination.
Collapse
Affiliation(s)
- Junfang Yan
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Yi Xie
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
| | - Jing Si
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
| | - Lu Gan
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
| | - Hongyan Li
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
| | - Chao Sun
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
| | - Cuixia Di
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
| | - Jinhua Zhang
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Guomin Huang
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Xuetian Zhang
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Hong Zhang
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
| |
Collapse
|
2
|
Román CL, Maiztegui B, Del Zotto H, Gagliardino JJ, Flores LE. INGAP-PP effects on β-cell mass and function are related to its positive effect on islet angiogenesis and VEGFA production. Mol Cell Endocrinol 2018; 470:269-280. [PMID: 29146554 DOI: 10.1016/j.mce.2017.11.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 11/10/2017] [Accepted: 11/10/2017] [Indexed: 01/09/2023]
Abstract
Our aim was to determine whether islet angiogenesis and VEGFA production/release participate in the mechanism by which INGAP-PP enhances β-cell function and mass. We used two models: a) in vivo (normal rats injected with INGAP-PP for 10 days) and b) in vitro (normal islets cultured for 4 days with INGAP-PP, VEGFA, Rapamycin, and the specific VEGF-Receptor inhibitor, SU5416). INGAP-PP administration enhanced insulin secretion, β-cell mass, islet vascularization, and angiogenesis without affecting glucose homeostasis. Normal islets cultured with INGAP-PP and VEGFA increased insulin and VEGFA secretion while apoptosis decreased. INGAP-PP-induced effects were prevented by both Rapamycin and SU5416. INGAP-PP effects on β-cell mass and function were significantly associated with a positive effect on islet angiogenesis and VEGFA production/release. VEGF-A possibly potentiates INGAP-PP effect through mTORC pathway.
Collapse
Affiliation(s)
- Carolina Lisi Román
- CENEXA, Centro de Endocrinología Experimental y Aplicada (UNLP-CONICET La Plata), Facultad de Ciencias Médicas UNLP, 60 y 120 (s/n) 4to piso, 1900 La Plata, Argentina
| | - Bárbara Maiztegui
- CENEXA, Centro de Endocrinología Experimental y Aplicada (UNLP-CONICET La Plata), Facultad de Ciencias Médicas UNLP, 60 y 120 (s/n) 4to piso, 1900 La Plata, Argentina
| | - Héctor Del Zotto
- CENEXA, Centro de Endocrinología Experimental y Aplicada (UNLP-CONICET La Plata), Facultad de Ciencias Médicas UNLP, 60 y 120 (s/n) 4to piso, 1900 La Plata, Argentina
| | - Juan José Gagliardino
- CENEXA, Centro de Endocrinología Experimental y Aplicada (UNLP-CONICET La Plata), Facultad de Ciencias Médicas UNLP, 60 y 120 (s/n) 4to piso, 1900 La Plata, Argentina
| | - Luis Emilio Flores
- CENEXA, Centro de Endocrinología Experimental y Aplicada (UNLP-CONICET La Plata), Facultad de Ciencias Médicas UNLP, 60 y 120 (s/n) 4to piso, 1900 La Plata, Argentina.
| |
Collapse
|
3
|
Afzal M, Bhardwaj DP, Khan R, Kazmi I, Saleem S, Al-Abbasi FA, Anwar F. Antineoplastic influence of nimesulide in chemically induced hepatocellular carcinoma by inhibition of DNA synthesis. Inflammopharmacology 2018; 27:89-98. [DOI: 10.1007/s10787-018-0481-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Accepted: 04/08/2018] [Indexed: 12/20/2022]
|
4
|
Bowyer C, Lewis AL, Lloyd AW, Phillips GJ, Macfarlane WM. Hypoxia as a target for drug combination therapy of liver cancer. Anticancer Drugs 2017; 28:771-780. [PMID: 28542038 PMCID: PMC5515631 DOI: 10.1097/cad.0000000000000516] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 04/19/2017] [Indexed: 01/06/2023]
Abstract
Hepatocellular carcinoma (HCC) is the third most frequent cause of cancer deaths worldwide. The standard of care for intermediate HCC is transarterial chemoembolization, which combines tumour embolization with locoregional delivery of the chemotherapeutic doxorubicin. Embolization therapies induce hypoxia, leading to the escape and proliferation of hypoxia-adapted cancer cells. The transcription factor that orchestrates responses to hypoxia is hypoxia-inducible factor 1 (HIF-1). The aim of this work is to show that targeting HIF-1 with combined drug therapy presents an opportunity for improving outcomes for HCC treatment. HepG2 cells were cultured under normoxic and hypoxic conditions exposed to doxorubicin, rapamycin and combinations thereof, and analyzed for viability and the expression of hypoxia-induced HIF-1α in response to these treatments. A pilot study was carried out to evaluate the antitumour effects of these drug combinations delivered from drug-eluting beads in vivo using an ectopic xenograft murine model of HCC. A therapeutic doxorubicin concentration that inhibits the viability of normoxic and hypoxic HepG2 cells and above which hypoxic cells are chemoresistant was identified, together with the lowest effective dose of rapamycin against normoxic and hypoxic HepG2 cells. It was shown that combinations of rapamycin and doxorubicin are more effective than doxorubicin alone. Western Blotting indicated that both doxorubicin and rapamycin inhibit hypoxia-induced accumulation of HIF-1α. Combination treatments were more effective in vivo than either treatment alone. mTOR inhibition can improve outcomes of doxorubicin treatment in HCC.
Collapse
Affiliation(s)
- Cressida Bowyer
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Lewes Road, Brighton
| | | | - Andrew W. Lloyd
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Lewes Road, Brighton
| | - Gary J. Phillips
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Lewes Road, Brighton
| | - Wendy M. Macfarlane
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Lewes Road, Brighton
| |
Collapse
|
5
|
Molecular signaling cascades involved in nonmelanoma skin carcinogenesis. Biochem J 2017; 473:2973-94. [PMID: 27679857 DOI: 10.1042/bcj20160471] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 06/10/2016] [Indexed: 12/17/2022]
Abstract
Nonmelanoma skin cancer (NMSC) is the most common cancer worldwide and the incidence continues to rise, in part due to increasing numbers in high-risk groups such as organ transplant recipients and those taking photosensitizing medications. The most significant risk factor for NMSC is ultraviolet radiation (UVR) from sunlight, specifically UVB, which is the leading cause of DNA damage, photoaging, and malignant transformation in the skin. Activation of apoptosis following UVR exposure allows the elimination of irreversibly damaged cells that may harbor oncogenic mutations. However, UVR also activates signaling cascades that promote the survival of these potentially cancerous cells, resulting in tumor initiation. Thus, the UVR-induced stress response in the skin is multifaceted and requires coordinated activation of numerous pathways controlling DNA damage repair, inflammation, and kinase-mediated signal transduction that lead to either cell survival or cell death. This review focuses on the central signaling mechanisms that respond to UVR and the subsequent cellular changes. Given the prevalence of NMSC and the resulting health care burden, many of these pathways provide promising targets for continued study aimed at both chemoprevention and chemotherapy.
Collapse
|
6
|
Xu SL, Zhang YC, Wang GY, Yang Q, Liu B, Zhang J, Li H, Wang GS, Yang Y, Chen GH. Survival analysis of sirolimus-based immunosuppression in liver transplantation in patients with hepatocellular carcinoma. Clin Res Hepatol Gastroenterol 2016; 40:674-681. [PMID: 27825633 DOI: 10.1016/j.clinre.2016.03.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 02/29/2016] [Accepted: 03/04/2016] [Indexed: 02/04/2023]
Abstract
AIM To investigate the effect of a sirolimus-based immunosuppressive protocol on tumor recurrence and survival after liver transplantation (LT) in hepatocellular carcinoma (HCC) patients. METHODS We retrospectively analyzed 142 HCC patients who underwent LT in our hospital from January 2006 to January 2012. The patients were divided into the sirolimus (SRL) group (62 cases) and non-sirolimus (control) group (80 cases). Disease-free survival (DFS) and tumor-bearing survival after tumor recurrence were compared using the Kaplan-Meier method. RESULTS No significant difference in DFS was observed between the two groups. Furthermore, DFS showed no significant differences in the subgroups of patients who met the Milan criteria, exceeded the Milan criteria but met the University of California, San Francisco (UCSF) criteria, or exceeded the UCSF criteria between the two groups. In the control group, 21 patients who were administered SRL after tumor recurrence had a median tumor-bearing survival time of 12months (3-34months), while 14 patients who did not experience a change in their immunosuppressive protocol after tumor recurrence had a median tumor-bearing survival time of 8months (6-22months). There was a significant difference in the tumor-bearing survival time between these patients (P=0.039). CONCLUSIONS Not all HCC patients benefited from the sirolimus-based immunosuppressive protocol after LT. However, sirolimus may prolong the survival time of patients after tumor recurrence.
Collapse
Affiliation(s)
- Shi-Lei Xu
- Liver Transplant Center, Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Institute of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangzhou 510630, PR China
| | - Ying-Cai Zhang
- Liver Transplant Center, Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Institute of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangzhou 510630, PR China
| | - Guo-Ying Wang
- Liver Transplant Center, Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Institute of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangzhou 510630, PR China
| | - Qing Yang
- Liver Transplant Center, Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Institute of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangzhou 510630, PR China
| | - Bo Liu
- Liver Transplant Center, Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Institute of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangzhou 510630, PR China
| | - Jian Zhang
- Liver Transplant Center, Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Institute of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangzhou 510630, PR China
| | - Hua Li
- Liver Transplant Center, Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Institute of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangzhou 510630, PR China
| | - Gen-Shu Wang
- Liver Transplant Center, Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Institute of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangzhou 510630, PR China
| | - Yang Yang
- Liver Transplant Center, Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Institute of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangzhou 510630, PR China.
| | - Gui-Hua Chen
- Liver Transplant Center, Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Institute of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangzhou 510630, PR China
| |
Collapse
|
7
|
da Silva EFG, Krause GC, Lima KG, Haute GV, Pedrazza L, Mesquita FC, Basso BS, Velasquez AC, Nunes FB, de Oliveira JR. Rapamycin and fructose-1,6-bisphosphate reduce the HEPG2 cell proliferation via increase of free radicals and apoptosis. Oncol Rep 2016; 36:2647-2652. [PMID: 27665945 DOI: 10.3892/or.2016.5111] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 06/21/2016] [Indexed: 11/06/2022] Open
Abstract
Hepatocellular carcinoma is the most prevalent type of tumor among primary tumors affecting the liver. Rapamycin is currently used as a basis for chemotherapy in the treatment of cancers, including the liver. Because it shows several adverse effects, minimizing these effects without compromising efficacy is important. In this sense other drugs may be used concomitantly. One of these drugs is fructose-1,6-bisphosphate (FBP), which has shown therapeutic effect in various pathological situations, having antioxidant and anti-inflammatory proprieties. The objective of the present study was to evaluate the activity of rapamycin in combination with the FBP in HepG2 cell proliferation and the mechanisms involved. HepG2 cells were analyzed after 72 h of treatment with both drugs. Cell proliferation, cytotoxicity, cytokines, apoptosis, senescence, autophagy and oxidative stress were accessed. Ιt was demonstrated that the combination is more efficient than the single use of substances, because subtherapeutic doses of rapamycin, when associated to FBP become effective, reducing cell proliferation, through a significant increase in the production of tiobarbituric acid reactive substances (TBARS), suggesting that this might be the cause of death by apoptosis. According to these results, we believe that the association of both drugs may be a promising choice for the treatment of hepatocarcinoma.
Collapse
Affiliation(s)
- Elisa Feller Gonçalves da Silva
- Laboratory of Cellular Biophysics and Inflammation, Department of Cellular and Molecular Biology, School of Biosciences, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, CEP 90619‑900, Brazil
| | - Gabriele Catyana Krause
- Laboratory of Cellular Biophysics and Inflammation, Department of Cellular and Molecular Biology, School of Biosciences, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, CEP 90619‑900, Brazil
| | - Kelly Goulart Lima
- Laboratory of Cellular Biophysics and Inflammation, Department of Cellular and Molecular Biology, School of Biosciences, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, CEP 90619‑900, Brazil
| | - Gabriela Viegas Haute
- Laboratory of Cellular Biophysics and Inflammation, Department of Cellular and Molecular Biology, School of Biosciences, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, CEP 90619‑900, Brazil
| | - Leonardo Pedrazza
- Laboratory of Cellular Biophysics and Inflammation, Department of Cellular and Molecular Biology, School of Biosciences, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, CEP 90619‑900, Brazil
| | - Fernanda Cristina Mesquita
- Laboratory of Cellular Biophysics and Inflammation, Department of Cellular and Molecular Biology, School of Biosciences, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, CEP 90619‑900, Brazil
| | - Bruno Souza Basso
- Laboratory of Cellular Biophysics and Inflammation, Department of Cellular and Molecular Biology, School of Biosciences, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, CEP 90619‑900, Brazil
| | - Anderson Catarina Velasquez
- Laboratory of Cellular Biophysics and Inflammation, Department of Cellular and Molecular Biology, School of Biosciences, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, CEP 90619‑900, Brazil
| | - Fernanda Bordignon Nunes
- Laboratory of Cellular Biophysics and Inflammation, Department of Cellular and Molecular Biology, School of Biosciences, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, CEP 90619‑900, Brazil
| | - Jarbas Rodrigues de Oliveira
- Laboratory of Cellular Biophysics and Inflammation, Department of Cellular and Molecular Biology, School of Biosciences, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, CEP 90619‑900, Brazil
| |
Collapse
|
8
|
Wu T, Leboeuf C, Durand S, Su B, Deschamps M, Zhang X, Ferrand C, Pessaux P, Robinet E. Suicide gene-modified killer cells as an allogeneic alternative to autologous cytokine-induced killer cell immunotherapy of hepatocellular carcinoma. Mol Med Rep 2016; 13:2645-54. [PMID: 26820174 DOI: 10.3892/mmr.2016.4811] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 12/15/2015] [Indexed: 11/06/2022] Open
Abstract
Adoptive immunotherapy using autologous cytokine-induced killer (CIK) cells reduces the recurrence rate of hepatocellular carcinoma (HCC) in association with transarterial chemoembolization or radiofrequency. However, a large‑scale development of this immunotherapy remains difficult to consider in an autologous setting, considering the logistical hurdles associated with the production of this cell therapy product. A previous study has provided the in vitro and in vivo proof‑of‑concept that allogeneic suicide gene‑modified killer cells (aSGMKCs) from healthy blood donors (a cell therapy product previously demonstrated to provide anti‑leukemic effects to patients receiving allogeneic hematopoietic transplantation) may exert a potent anti‑tumor effect towards HCC. Therefore, the development of a bank of 'ready‑for‑use' aSGMKCs was proposed as an approach allowing for the development of immunotherapies that are more convenient and on a broader scale than that of autologous therapies. In the present study, aSGMKCs were compared with CIK cells generated according to three different protocols. Similar to CIK cells, the cytotoxic activity of aSGMKCs toward the Huh‑7 HCC cell line was mediated by tumor necrosis factor‑related apoptosis‑inducing ligand, tumor necrosis factor‑α and interferon‑γ. Furthermore, the frequency of natural killer (NK), NK‑like T and T cells, and their in vitro and in vivo cytotoxicity activities were similar between aSGMKCs and CIK cells. Thus, the present study demonstrated that aSGMKCs are similar to CIK cells, further suggesting the possibility for future use of aSGMKCs in the treatment of solid tumors, including HCC.
Collapse
Affiliation(s)
- Tao Wu
- Department of Hepatobiliary and Pancreatic Surgery, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Céline Leboeuf
- French National Institute of Health and Medical Research, Research Unit 1110, F-67000 Strasbourg, France
| | - Sarah Durand
- French National Institute of Health and Medical Research, Research Unit 1110, F-67000 Strasbourg, France
| | - Bin Su
- French National Institute of Health and Medical Research, Research Unit 1110, F-67000 Strasbourg, France
| | - Marina Deschamps
- French National Blood Service (Bourgogne/Franche‑Comté), Research Unit 1098, F-25000 Besançon, France
| | - Xiaowen Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Christophe Ferrand
- French National Blood Service (Bourgogne/Franche‑Comté), Research Unit 1098, F-25000 Besançon, France
| | - Patrick Pessaux
- French National Institute of Health and Medical Research, Research Unit 1110, F-67000 Strasbourg, France
| | - Eric Robinet
- French National Institute of Health and Medical Research, Research Unit 1110, F-67000 Strasbourg, France
| |
Collapse
|
9
|
Dickinson SE, Janda J, Criswell J, Blohm-Mangone K, Olson ER, Liu Z, Barber C, Petricoin EF, Calvert VS, Einspahr J, Dickinson JE, Stratton SP, Curiel-Lewandrowski C, Saboda K, Hu C, Bode AM, Dong Z, Alberts DS, Timothy Bowden G. Inhibition of Akt Enhances the Chemopreventive Effects of Topical Rapamycin in Mouse Skin. Cancer Prev Res (Phila) 2016; 9:215-24. [PMID: 26801880 DOI: 10.1158/1940-6207.capr-15-0419] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 01/01/2016] [Indexed: 11/16/2022]
Abstract
The PI3Kinase/Akt/mTOR pathway has important roles in cancer development for multiple tumor types, including UV-induced nonmelanoma skin cancer. Immunosuppressed populations are at increased risk of aggressive cutaneous squamous cell carcinoma (SCC). Individuals who are treated with rapamycin (sirolimus, a classical mTOR inhibitor) have significantly decreased rates of developing new cutaneous SCCs compared with those that receive traditional immunosuppression. However, systemic rapamycin use can lead to significant adverse events. Here, we explored the use of topical rapamycin as a chemopreventive agent in the context of solar-simulated light (SSL)-induced skin carcinogenesis. In SKH-1 mice, topical rapamycin treatment decreased tumor yields when applied after completion of 15 weeks of SSL exposure compared with controls. However, applying rapamycin during SSL exposure for 15 weeks, and continuing for 10 weeks after UV treatment, increased tumor yields. We also examined whether a combinatorial approach might result in more significant tumor suppression by rapamycin. We validated that rapamycin causes increased Akt (S473) phosphorylation in the epidermis after SSL, and show for the first time that this dysregulation can be inhibited in vivo by a selective PDK1/Akt inhibitor, PHT-427. Combining rapamycin with PHT-427 on tumor prone skin additively caused a significant reduction of tumor multiplicity compared with vehicle controls. Our findings indicate that patients taking rapamycin should avoid sun exposure, and that combining topical mTOR inhibitors and Akt inhibitors may be a viable chemoprevention option for individuals at high risk for cutaneous SCC.
Collapse
Affiliation(s)
- Sally E Dickinson
- The University of Arizona Cancer Center, Tucson, Arizona. Department of Pharmacology, The University of Arizona, Tucson, Arizona.
| | - Jaroslav Janda
- The University of Arizona Cancer Center, Tucson, Arizona
| | - Jane Criswell
- The University of Arizona Cancer Center, Tucson, Arizona
| | | | - Erik R Olson
- The University of Arizona Cancer Center, Tucson, Arizona
| | - Zhonglin Liu
- Department of Medical Imaging, The University of Arizona, Tucson, Arizona
| | - Christy Barber
- Department of Medical Imaging, The University of Arizona, Tucson, Arizona
| | - Emanuel F Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia
| | - Valerie S Calvert
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia
| | - Janine Einspahr
- The University of Arizona Cancer Center, Tucson, Arizona. Department of Medicine, The University of Arizona, Tucson, Arizona
| | - Jesse E Dickinson
- Arizona Water Science Center, U.S. Geological Survey, Tucson, Arizona
| | - Steven P Stratton
- The University of Arizona Cancer Center, Tucson, Arizona. Department of Medicine, The University of Arizona, Tucson, Arizona
| | - Clara Curiel-Lewandrowski
- The University of Arizona Cancer Center, Tucson, Arizona. Department of Medicine, The University of Arizona, Tucson, Arizona
| | | | - Chengcheng Hu
- The University of Arizona Cancer Center, Tucson, Arizona
| | - Ann M Bode
- Department of Molecular Medicine and Biopharmaceutical Sciences, The Hormel Institute, The University of Minnesota, Austin, Minnesota
| | - Zigang Dong
- Department of Molecular Medicine and Biopharmaceutical Sciences, The Hormel Institute, The University of Minnesota, Austin, Minnesota
| | - David S Alberts
- The University of Arizona Cancer Center, Tucson, Arizona. Department of Medicine, The University of Arizona, Tucson, Arizona
| | - G Timothy Bowden
- The University of Arizona Cancer Center, Tucson, Arizona. Department of Medicine, The University of Arizona, Tucson, Arizona. Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona
| |
Collapse
|
10
|
Hispidulin Enhances the Anti-Tumor Effects of Temozolomide in Glioblastoma by Activating AMPK. Cell Biochem Biophys 2014; 71:701-6. [DOI: 10.1007/s12013-014-0252-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
11
|
Han X, Shang D, Han T, Xu X, Tian Y. Interferon-α enhances the susceptibility of renal cell carcinoma to rapamycin by suppressing mTOR activity. Exp Ther Med 2014; 8:267-273. [PMID: 24944633 PMCID: PMC4061221 DOI: 10.3892/etm.2014.1691] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 03/19/2014] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the antiproliferative effects of interferon (IFN)-α and rapamycin (RPM) on renal cell carcinoma (RCC) cells and examine the synergistic growth suppression conferred by IFN-α and RPM. The effects of IFN-α and/or RPM on RCC cells were determined using a WST-1 assay and the synergy of IFN-α and RPM against three RCC cell lines was analyzed with isobolographic analysis. The expression of mammalian target of rapamycin (mTOR) was downregulated by RNAi, and the expression and phosphorylation of proteins in the mTOR pathway following treatment with IFN-α and/or RPM was examined by western blot analysis. The observations indicated that IFN-α significantly increased the susceptibility of RCC cells to RPM and the synergistic effect of IFN-α and RPM against RCC cells was confirmed in all three RCC cell lines. The mTOR pathway was shown to be associated with the synergistic effect of IFN-α and RPM against RCC. IFN-α and RPM alone decreased the phosphorylation of mTOR, p70 S6 kinase, S6 and 4E binding protein 1, and IFN-α significantly enhanced the RPM-induced suppression of the mTOR pathway. However, in RCC cells with low mTOR activity, the synergy of IFN-α and RPM was eliminated. Therefore, the results of the present study indicate that the mTOR pathway plays an important role in the synergistic effect of IFN-α and RPM against RCC cells. Thus, mTOR may serve as an effective therapeutic target in the treatment of advanced RCC.
Collapse
Affiliation(s)
- Xiao Han
- Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Donghao Shang
- Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Tiandong Han
- Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Xiuhong Xu
- Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Ye Tian
- Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| |
Collapse
|
12
|
Kaneko M, Nozawa H, Hiyoshi M, Tada N, Murono K, Nirei T, Emoto S, Kishikawa J, Iida Y, Sunami E, Tsuno NH, Kitayama J, Takahashi K, Watanabe T. Temsirolimus and chloroquine cooperatively exhibit a potent antitumor effect against colorectal cancer cells. J Cancer Res Clin Oncol 2014; 140:769-81. [PMID: 24619662 DOI: 10.1007/s00432-014-1628-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Accepted: 02/22/2014] [Indexed: 01/08/2023]
Abstract
PURPOSE Temsirolimus (TEM) is a novel, water-soluble mammalian target of rapamycin (mTOR) inhibitor that has shown activity against a wide range of cancers in preclinical models, but its efficacy against colorectal cancer (CRC) has not been fully explored. METHODS We evaluated the antitumor effect of TEM in CRC cell lines (CaR-1, HT-29, Colon26) in vitro and in vivo. In vitro, cell growth inhibition was assessed using a MTS assay. Apoptosis induction and cell cycle effects were measured using flow cytometry. Modulation of mTOR signaling was measured using immunoblotting. Antitumor activity as a single agent was evaluated in a mouse subcutaneous tumor model of CRC. The effects of adding chloroquine, an autophagy inhibitor, to TEM were evaluated in vitro and in vivo. RESULTS In vitro, TEM was effective in inhibiting the growth of two CRC cell lines with highly activated AKT, possibly through the induction of G1 cell cycle arrest via a reduction in cyclin D1 expression, whereas TEM reduced HIF-1α and VEGF in all three cell lines. In a mouse subcutaneous tumor model, TEM inhibited the growth of tumors in all cell lines, not only through direct growth inhibition but also via an anti-angiogenic effect. We also explored the effects of adding chloroquine, an autophagy inhibitor, to TEM. Chloroquine significantly potentiated the antitumor activity of TEM in vitro and in vivo. Moreover, the combination therapy triggered enhanced apoptosis, which corresponded to an increased Bax/Bcl-2 ratio. CONCLUSIONS Based on these data, we propose TEM with or without chloroquine as a new treatment option for CRC.
Collapse
Affiliation(s)
- Manabu Kaneko
- Department of Surgical Oncology, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan,
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Jin M, Shen X, Zhao C, Qin X, Liu H, Huang L, Qiu Z, Liu Y. In vivo study of effects of artesunate nanoliposomes on human hepatocellular carcinoma xenografts in nude mice. Drug Deliv 2013; 20:127-33. [PMID: 23731485 DOI: 10.3109/10717544.2013.801047] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
To investigate the effect of artesunate nanoliposomes on cultured cells in vitro and hepatocellular carcinoma xenografts in BALB/c-nu mice. Fluorescence polarization was applied for measurement of mitochondrial membrane fluidities; inhibition test of tumor cell proliferation in vitro was performed and nude mice xenograft model from human hepatocellular carcinoma (HCC) was established. Cytotoxicity of these compounds was evaluated by MTT assay on hepatocellular carcinoma xenografts in nude mice. Anisotropy (r-value) of blank nanoliposomes didn't change, it had no statistically significance between the blank nanoliposomes group and the control group, it indicated that artesunate had no obvious effect on L-O2 human normal liver cells. IC₅₀ values of artesunate nanoliposomes and artesunate API (active pharmaceutical ingredient) against HepG-2 cells were 15.997 and 19.706 μg/ml; IC₅₀ values of the same drugs against L-O2 normal human liver cells were 100.23 and 105.54 μg/ml, respectively. Tumor growth inhibitory effect of artesunate nanoliposomes was 32.7%, and artesunate API was 20.5%, respectively. HepG-2 cells treated with artesunate nanoliposomes showed dose-dependent apoptosis. The antitumor effect of artesunate nanoliposomes on human hepatoma HepG2 cells were stronger than that of artesunate API at the same concentration.
Collapse
Affiliation(s)
- Meihua Jin
- College of Pharmacy, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
14
|
The expression of URGCP gene in prostate cancer cell lines: correlation with rapamycin. Mol Biol Rep 2012; 39:10173-7. [PMID: 23007575 DOI: 10.1007/s11033-012-1891-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Accepted: 09/18/2012] [Indexed: 01/02/2023]
Abstract
Molecular targets in prostate cancer are continually being explored, for which there are currently few therapeutic options. Rapamycin (RPM) is an antifungal macrolide antibiotic isolated from Streptomyces hygroscopicus which can inhibit the G1 to S transition. URGCP (upregulator of cell proliferation) is a novel gene located on chromosome 7p13. We aimed to investigate the role of URGCP gene expression changes in PC3, DU145, and LNCAP cell lines with/out RPM. Average cell viability and cytotoxic effect of rapamycin were investigated at 24 h intervals for three days by using Trypan blue dye exclusion test and XTT assay. Cytotoxic effects of rapamycin in DU145, PC3 and LNCAP cells were detected in time and dose dependent manner with the IC(50) doses within the range of 1-100 nM. As the results were evaluated, IC(50) doses in the DU145, PC3, and LNCaP cells were detected as 10, 25, and 50 nM, respectively. The mean relative ratios of URGCP gene expression in DU145, LNCAP and PC3 cells were found as -1.48, 6.59 and -13.00, respectively, when compared to rapamycin-free cells. The False Discovery Rate adjusted p value in DU145, LNCAP and PC3 were 1.25 × 10(-5), 2.20 × 10(-8) and 6.20 × 10(-9), respectively. When the URGCP gene expression level is compared between the dose and control group, we found that URGCP gene expression was significantly decreased in dose groups of DU145 and PC3 cells.
Collapse
|
15
|
Sun Y, Chen X, Zhao J, Zou X, Li G, Li X, Shen B, Sun S. Combined use of rapamycin and leflunomide in prevention of acute cardiac allografts rejection in rats. Transpl Immunol 2012; 27:19-24. [DOI: 10.1016/j.trim.2012.04.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Revised: 03/31/2012] [Accepted: 04/08/2012] [Indexed: 01/11/2023]
|
16
|
MIYAKE NAOMI, CHIKUMI HIROKI, TAKATA MIYAKO, NAKAMOTO MASAKI, IGISHI TADASHI, SHIMIZU EIJI. Rapamycin induces p53-independent apoptosis through the mitochondrial pathway in non-small cell lung cancer cells. Oncol Rep 2012; 28:848-54. [DOI: 10.3892/or.2012.1855] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Accepted: 05/02/2012] [Indexed: 11/06/2022] Open
|
17
|
Yuan Y, Xue X, Guo RB, Sun XL, Hu G. Resveratrol enhances the antitumor effects of temozolomide in glioblastoma via ROS-dependent AMPK-TSC-mTOR signaling pathway. CNS Neurosci Ther 2012; 18:536-46. [PMID: 22530672 DOI: 10.1111/j.1755-5949.2012.00319.x] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE Resveratrol has been regarded as a promising candidate for cancer prevention and treatment. The present study was to investigate the impact of resveratrol on the antitumor effects of temozolomide (TMZ), a standard treatment regiment of glioblastoma (GBM), in vitro and in vivo. METHODS AND RESULTS We found that the combination of resveratrol and TMZ significantly resulted in G(2)/M cell cycle arrest by flow cytometry, triggered a robust increase in expression of astrocyte differentiation marker glial fibrillary acid protein (GFAP), downregulated the expression of matrix metalloproteinase-9 (MMP-9) by immunohistochemistry and western blot analysis as well as inhibited cell migration by scratch wound assay. Further study revealed that TMZ in combination with resveratrol remarkably increased reactive oxygen species (ROS) production, which serves as an upstream signal for AMP-activated protein kinase (AMPK) activation. Subsequently, activated AMPK inhibited mTOR signaling and downregulated antiapoptosis protein Bcl-2, which was contributed to the additive antiproliferation effects of combination treatment. In an orthotopic xenograft model of GBM, TMZ plus resveratrol treatment significantly reduced the volume of tumor, which was confirmed by decreased expression of Ki-67, a marker of proliferation index. CONCLUSIONS Our findings demonstrate for the first time that resveratrol can enhance TMZ-mediated antitumor effects in GBM in vitro and in vivo, via ROS-dependent AMPK-TSC-mTOR signaling pathway.
Collapse
Affiliation(s)
- Yuan Yuan
- Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, China
| | | | | | | | | |
Collapse
|
18
|
Morikawa Y, Koike H, Sekine Y, Matsui H, Shibata Y, Ito K, Suzuki K. Rapamycin enhances docetaxel-induced cytotoxicity in a androgen-independent prostate cancer xenograft model by survivin downregulation. Biochem Biophys Res Commun 2012; 419:584-9. [DOI: 10.1016/j.bbrc.2012.02.089] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 02/15/2012] [Indexed: 10/28/2022]
|
19
|
Ponziani F, Ojetti V, Tortora A, Di Maurizio L, Purchiaroni F, Gasbarrini A. The metabolic and toxicological considerations for mTOR inhibitors in the treatment of hepatocarcinoma. Expert Opin Drug Metab Toxicol 2011; 7:1535-46. [PMID: 22032293 DOI: 10.1517/17425255.2011.631911] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Hepatocellular carcinoma (HCC) is a major health problem worldwide. Several molecular pathways involved in HCC growth and progression have recently been identified. Rapamycin analogs are able to inhibit one of the most active oncogenic molecular pathways in HCC cells: the mammalian target of rapamycin (mTOR) pathway. AREAS COVERED In this review, the authors analyze the principal molecular features of the mTOR pathway and the use of rapamycin analogs in the treatment of hepatocarcinoma. The article also looks at the reoccurrence of HCC following liver transplantation as well as after the treatment of de novo neoplasms. Finally, the authors discuss the advantage of using a combined HCC pharmacological therapy to obtain a synergistic effect on tumor mass. EXPERT OPINION Among the available options for the treatment of advanced-stage HCC, mTOR pathway inhibitors show great promise. Once these agents have their safety and efficacy confirmed, in the treatment of liver disease, their use should be considered in patients affected by HCC. This should especially be the case for those who have had liver transplants or suffered with de novo tumors. Moreover, the authors believe that mTOR inhibitors could be used in a combined pharmacological approach to improve HCC molecular-targeted therapy by producing a multiple-level block of tumor intracellular signaling.
Collapse
Affiliation(s)
- Francesca Ponziani
- Catholic University of Sacred Heart, Gemelli Hospital, Largo A. Gemelli, 8 Rome 00168, Italy
| | | | | | | | | | | |
Collapse
|
20
|
Chen MB, Wu XY, Gu JH, Guo QT, Shen WX, Lu PH. Activation of AMP-activated protein kinase contributes to doxorubicin-induced cell death and apoptosis in cultured myocardial H9c2 cells. Cell Biochem Biophys 2011; 60:311-22. [PMID: 21274754 DOI: 10.1007/s12013-011-9153-0] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Despite its potent antitumor effect, clinical use of Doxorubicin is limited because of serious side effects including myocardial toxicity. Understanding the cellular mechanism involved in this process in a better manner is beneficial for optimizing Doxorubicin treatment. In the current study, the authors focus on the AMP-activated protein kinase (AMPK) in the said process. In this study, the authors discovered for the first time that Doxorubicin induces AMPK activation in cultured rat embryonic ventricular myocardial H9c2 cells. Reactive oxygen species (ROS)-dependent LKB1 activation serves as the upstream signal for AMPK activation by Doxorubicin. Evidence in support of the activation of AMPK contributing to Doxorubicin-induced H9c2 cell death/apoptosis--probably by modulating multiple downstream signal targets, including regulating JNK, p53, and inhibiting mTORC1--is provided in this article.
Collapse
Affiliation(s)
- Min-Bin Chen
- Department of Medical Oncology, Kunshan First People's Hospital Affiliated to Jiangsu University, 91 Qianjin Road, Kunshan 215300, Jiangsu Province, China.
| | | | | | | | | | | |
Collapse
|
21
|
Pignataro G, Capone D, Polichetti G, Vinciguerra A, Gentile A, Di Renzo G, Annunziato L. Neuroprotective, immunosuppressant and antineoplastic properties of mTOR inhibitors: current and emerging therapeutic options. Curr Opin Pharmacol 2011; 11:378-394. [PMID: 21646048 DOI: 10.1016/j.coph.2011.05.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Revised: 05/12/2011] [Accepted: 05/13/2011] [Indexed: 02/05/2023]
Abstract
The acronym mTOR defines a family of serine-threonine protein kinase called mammalian target of rapamycin. The major role of these kinases in the cell is to merge extracellular instructions with information about cellular metabolic resources and to control the rate of anabolic and catabolic processes accordingly. In mammalian cells mTOR is present in two distinct heteromeric protein complexes commonly referred to as mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2), involved in the control of a wide variety of cellular processes. It has been recently reported that compounds acting modulating mTOR activity, beside mediating the well recognized processes exploited in the anticancer and immunosuppressant effects, are provided with neuroprotective properties. In fact, mTOR is involved in the mechanism of PI3K/Akt-induced upregulation of glutamate transporter 1, GLT1, that is linked to several neuronal disorders such as stroke, Alzheimer's disease, and amyotrophic lateral sclerosis. Furthermore, in adult brain mTOR is crucial for numerous physiological processes such as synaptic plasticity, learning, memory, and brain control of food uptake. Moreover, the activation of mTOR pathway is involved in neuronal development, dendrite development and spine morphogenesis.
Collapse
Affiliation(s)
- Giuseppe Pignataro
- Division of Pharmacology, Department of Neuroscience, School of Medicine, Federico II University of Naples, Via Pansini, 5, 80131 Naples, Italy
| | | | | | | | | | | | | |
Collapse
|
22
|
Chen MB, Shen WX, Yang Y, Wu XY, Gu JH, Lu PH. Activation of AMP-activated protein kinase is involved in vincristine-induced cell apoptosis in B16 melanoma cell. J Cell Physiol 2011; 226:1915-25. [PMID: 21506122 DOI: 10.1002/jcp.22522] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The molecular basis for induction of apoptosis in melanoma cells by vincristine remains unknown. Here we tested the potential involvement of AMP-activated protein kinase (AMPK) in this process. We found for the first time that vincristine induces AMPK activation (AMPKα, Thr 172) and Acetyl-CoA carboxylase (ACC, Ser 79) (a downstream molecular target of AMPK) phosphorylation in cultured melanoma cells in vitro. Reactive oxygen species (ROS) dependent LKB1 activation serves as the upstream signal for AMPK activation. AMPK inhibitor (compound C) or AMPKα siRNA knockdown inhibits vincristine induced B16 melanoma cell apoptosis, while AMPK activator 5-aminoimidazole-4-carboxamide-1-β-riboside (AICAR) enhances it. AMPK activation is involved in vincristine induced p53 phosphorylation and stabilization, the latter is known to mediate melanoma cell apoptosis. Further, activation of AMPK by vincristine inhibits mTOR Complex 1 (mTORC1) in B16 melanoma cells, which serves as another important mechanism to induce melanoma cell apoptosis. Our study provides new insights into understanding the cellular and molecular mechanisms of vincristine induced cancer cell death/apoptosis. We suggest that combining AMPK activator AICAR with vincristine may have potential to be used as a new therapeutic intervention against melanoma.
Collapse
Affiliation(s)
- Min-Bin Chen
- Department of Medical Oncology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu Province, China
| | | | | | | | | | | |
Collapse
|
23
|
Zhang WB, Wang Z, Shu F, Jin YH, Liu HY, Wang QJ, Yang Y. Activation of AMP-activated protein kinase by temozolomide contributes to apoptosis in glioblastoma cells via p53 activation and mTORC1 inhibition. J Biol Chem 2010; 285:40461-71. [PMID: 20880848 DOI: 10.1074/jbc.m110.164046] [Citation(s) in RCA: 157] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Methylating drugs such as temozolomide (TMZ) are widely used in the treatment of brain tumors including malignant glioblastoma. The mechanism of TMZ-induced glioblastoma cell death and apoptosis, however, is not fully understood. Here, we tested the potential involvement of AMP-activated protein kinase (AMPK) in this process. We found that methylating agents TMZ and N-methyl-N'-nitro-N-nitrosoguanidine induce AMPK activation in primary cultured human glioblastoma and glioblastoma cell lines. TMZ-induced O(6)-methylguanine production is involved in AMPK activation. O(6)-benzylguanine, an O(6)-methylguanine-DNA methyltransferase inhibitor, enhances TMZ-induced O(6)-methylguanine production, leading to enhanced reactive oxygen species production, which serves as an upstream signal for AMPK activation. Activation of AMPK is involved in TMZ-induced glioblastoma cell death and apoptosis. AMPK inhibitor (Compound C) or AMPKα siRNA knockdown inhibits TMZ-induced glioblastoma cell death and apoptosis, whereas AMPK activator 5-aminoimidazole-4-carboxamide-1-β-d-ribofuranoside enhances it. In further studies, we found that activation of AMPK is involved in TMZ-induced p53 activation and subsequent p21, Noxa, and Bax up-regulation. Activation of AMPK by TMZ also inhibits mTOR complex 1 (mTORC1) signaling and promotes anti-apoptosis protein Bcl-2 down-regulation, which together mediate TMZ-induced pro-cell apoptosis effects. Our study suggests that activation of AMPK by TMZ contributes to glioblastoma cell apoptosis, probably by promoting p53 activation and inhibiting mTORC1 signaling.
Collapse
Affiliation(s)
- Wen-bin Zhang
- Department of Physiology, China Pharmaceutical University, Nanjing 210009, China
| | | | | | | | | | | | | |
Collapse
|
24
|
Exogenous cell-permeable C6 ceramide sensitizes multiple cancer cell lines to Doxorubicin-induced apoptosis by promoting AMPK activation and mTORC1 inhibition. Oncogene 2010; 29:6557-68. [PMID: 20802518 DOI: 10.1038/onc.2010.379] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
New chemotherapy-enhancing strategies are needed for better cancer therapy. Previous studies suggest that exogenous cell-permeable C6 ceramide may be a useful adjunct to the anti-tumor effects of chemotherapeutic agents (such as Taxol) against multiple cancers. Here we demonstrate that exogenous cell-permeable C6 ceramide largely sensitizes multiple progressive cancer cell lines to Doxorubicin-induced cell death and apoptosis. We found for the first time that Doxorubicin induces AMP-activated protein kinase (AMPK) activation in a reactive oxygen species-dependent manner. Activation of AMPK contributes to Doxorubicin-induced cancer cell death and apoptosis. Inhibition of AMPK by small interfering RNA knockdown or a pharmacological inhibitor reduces Doxorubicin-induced cancer cell apoptosis, whereas AMPK activator AICAR enhances it. Importantly, we found that C6 ceramide largely enhances Doxorubicin-induced activation of AMPK, which leads to mTOR complex 1 inhibition and chemo-sensitization. Our data suggest that the combination of C6 ceramide with traditional chemotherapy drugs such as Doxorubicin may have the potential to be used as a new therapeutic intervention against multiple cancers.
Collapse
|
25
|
Chu YK, Liu Y, Yin JK, Wang N, Cai L, Lu JG. Effect of mycophenolate mofetil plus adriamycin on HepG-2 cells. World J Hepatol 2010; 2:311-7. [PMID: 21161014 PMCID: PMC2998975 DOI: 10.4254/wjh.v2.i8.311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Revised: 08/12/2010] [Accepted: 08/17/2010] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the influence of mycophenolate mofetil (MMF) plus adriamycin (ADM) on hepatocellular carcinoma (HCC) cells. METHODS HCC cells were treated with 100 μg/ml of MMF alone (MMF group), 1 μg/mL of adriamycin (ADM group) alone, or a combination of the drugs (MMF + ADM group). We performed an 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide (MTT) assay to measure the growth inhibition rate of HCC cells. Flow cytometry was used to determine the percentage of cells in different phases of the cell cycle and the number of apoptotic cells. Hoechst 33258 staining revealed the morphological changes associated with apoptosis in HCC cells. RESULTS The results of MTT assays revealed that monotherapy with ADM or MMF showed inhibition of cell growth, while MMF + ADM therapy afforded an inhibition rate of more than 90% with cell distribution in G1 and G2/M phase greater than that in S phase. MMF + ADM treatment also downregulated Bcl-2 expression markedly. The growth of HCC cells was markedly inhibited and apoptosis was enhanced in all the 3 groups. Compared with other 2 groups, the MMF + ADM group showed more obvious apoptosis of cells. CONCLUSION The MMF plus ADM combination exerts remarkable inhibitory effects on the growth of HCC cells.
Collapse
Affiliation(s)
- Yan-Kui Chu
- Yan-Kui Chu, Yi Liu, Ji-Kai Yin, Nan Wang, Liang Cai, Jian-Guo Lu, Department of General Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, Shaanxi Province, China
| | | | | | | | | | | |
Collapse
|
26
|
Kim KW, Moretti L, Mitchell LR, Jung DK, Lu B. Combined Bcl-2/mammalian target of rapamycin inhibition leads to enhanced radiosensitization via induction of apoptosis and autophagy in non-small cell lung tumor xenograft model. Clin Cancer Res 2009; 15:6096-105. [PMID: 19773376 DOI: 10.1158/1078-0432.ccr-09-0589] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE Radiotherapy has a central role in the treatment of non-small cell lung cancer. Effectiveness of this modality, however, is often limited as resistance results from defects in cell death. EXPERIMENTAL DESIGN We investigated whether simultaneous up-regulation of apoptosis, via Bcl-2 inhibitor ABT-737, and autophagy, via mammalian target of rapamycin inhibitor rapamycin, can be used to enhance radiosensitivity of H460 cells in vitro and growth delay in a xenograft model. RESULTS In vitro studies confirmed that ABT-737 and rapamycin induce apoptosis and autophagy, respectively. ABT-737 induced cleaved caspase-3, a marker of apoptosis, and rapamycin correlated with an increase in punctate localization of green fluorescent protein-LC3, characteristic of autophagy. The combination ABT-737/rapamycin markedly enhanced sensitivity of H460 cells to radiation (dose enhancement ratio = 2.47; P = 0.002) in clonogenic assay. In addition, the combination ABT-737/rapamycin/radiation showed a dramatic tumor growth delay in a mouse xenograft model. In vivo immunohistochemistry staining showed that combination therapy yielded over a 100% increase in caspase-3 activity (apoptosis) and a 6-fold decrease in p62 protein level (indicative of autophagic flux) compared with radiation alone control group. Moreover, cell proliferation (Ki-67 staining) was reduced by 77% (P = 0.001) and vascular density (von Willebrand factor staining) by 67.5% (P = 0.09) compared with radiation alone. Additional in vitro studies in human umbilical vein endothelial cells indicated that combined therapy also significantly decreases tubule formation. CONCLUSION These results suggest that concurrent induction of apoptosis and autophagy enhances radiation therapy both in vitro and in lung cancer xenograft models. Further investigations are warranted to assess the clinical potential of such strategy in lung cancer patients.
Collapse
Affiliation(s)
- Kwang Woon Kim
- Department of Radiation Oncology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-5671, USA
| | | | | | | | | |
Collapse
|
27
|
Both tacrolimus and sirolimus decrease Th1/Th2 ratio, and increase regulatory T lymphocytes in the liver after ischemia/reperfusion. J Transl Med 2009; 89:433-45. [PMID: 19188906 DOI: 10.1038/labinvest.2009.3] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The protective effects of immunosuppressants against ischemia/reperfusion (I/R) injury have been attributed to their non-specific anti-inflammatory effect. However, these effects may also depend on their effect on T lymphocytes, which are increasingly considered to be key players in I/R. Here, we studied the effects of tacrolimus and sirolimus on lymphocyte subpopulations in an I/R rat model. The animals were treated with tacrolimus, sirolimus or vehicle, before undergoing a 60-min ischemia event of the right hepatic lobe, followed by excision of the remaining liver. After 2 h, I/R rats showed increased mortality, plasma lactate dehydrogenase (LDH) levels, hepatocyte apoptosis, liver histological injury and parenchymal infiltration by neutrophils, macrophages, NK cells and T lymphocytes. Most of the changes were antagonized by both immunosuppressants. Tacrolimus augmented the proportion of cycling cells in I/R rats, whereas sirolimus showed the opposite effect. The increased Th1/Th2 ratio found in I/R livers after 2 h was reverted by immunosuppressants, which also amplified the proportion of CD4(+)CD25(+)Foxp3(+) regulatory T lymphocytes at 24 h. The protective effects of both tacrolimus and sirolimus correlated well with a decreased ratio of proinflammatory to anti-inflammatory T lymphocytes, and with an increase in the Treg proportion. This suggests a new mechanism to explain the known beneficial effect shown by immunosuppressants early after I/R.
Collapse
|
28
|
Kent MS, Collins CJ, Ye F. Activation of the AKT and mammalian target of rapamycin pathways and the inhibitory effects of rapamycin on those pathways in canine malignant melanoma cell lines. Am J Vet Res 2009; 70:263-9. [PMID: 19231960 DOI: 10.2460/ajvr.70.2.263] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVE To investigate the activation of the AKT and mammalian target of rapamycin (mTOR) pathways and assess the inhibitory effects of rapamycin on those pathways in canine malignant melanoma cells. SAMPLE POPULATION 3 established primary canine melanoma cell lines generated from naturally occurring tumors. PROCEDURES Expressions of total and phosphorylated AKT, mTOR, and p70 ribosomal S6 kinase 1 (p70S6K) in canine melanoma cells that were or were not exposed to 10nM rapamycin were assessed via western blot analysis. Clonogenic assays were performed to determine the surviving fraction of melanoma cells after exposure to 0.1, 1, 10, or 100nM rapamycin. RESULTS Expressions of total and phosphorylated AKT, mTOR, and p70S6K proteins were detected (ie, the AKT and mTOR pathways were activated) in all 3 cell lines. Rapamycin treatment resulted in decreases in phosphorylated mTOR expression and phosphorylated p70S6K expression but no change in phosphorylated AKT expression. Expression of total AKT, mTOR, and p70S6K persisted after rapamycin treatment. There was a significant dose-dependent decrease in surviving tumor cell fraction for each cell line following treatment with rapamycin. CONCLUSIONS AND CLINICAL RELEVANCE These data indicated that AKT and mTOR, as well as their downstream product p70S6K, are present and active in canine melanoma cells. Activation of the mTOR pathway can be inhibited by rapamycin; treatment of melanoma cells with rapamycin decreased the surviving tumor cell fraction. Use of mTOR inhibitors as antineoplastic treatments in dogs with melanoma warrants investigation. Furthermore, these data support the use of canine melanoma cells as a molecular model for melanoma in humans.
Collapse
Affiliation(s)
- Michael S Kent
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | | | | |
Collapse
|
29
|
Okuno T, Ueda M, Tsuruyama T, Haga H, Takada Y, Maetani Y, Tamaki K, Manabe T, Tanaka K, Uemoto S. Loss of heterozygosity on 10q23 is involved in metastatic recurrence of hepatocellular carcinoma. Cancer Sci 2009; 100:520-8. [PMID: 19077004 PMCID: PMC11158083 DOI: 10.1111/j.1349-7006.2008.01056.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
We performed loss of heterozygosity (LOH) analysis on five chromosomal arms (1p, 3p, 9p, 10q, 17p) in hepatocellular carcinoma (HCC). Univariate analyses of 80 patients who underwent liver transplantation demonstrated significant correlations between cancer recurrence and the following variables: LOH on 3p26, LOH on 10q23, LOH on 17p13, tumor diameter > or = 5 cm, number of tumors > or = 4, histologic Grade 3, alpha-fetoprotein (AFP) > or = 400 ng/mL, American Joint Committee on Cancer (AJCC) pT classification, and portal invasion. Patients with LOH on 10q23 exhibited a significantly higher 3-year recurrence rate (38.9%vs 11.9%, P = 0.0009). Multivariate analysis identified LOH on 10q23, histologic Grade 3, tumor nodules > or = 4, and AFP > or = 400 ng/mL as the risk factors of advanced HCC recurrence. These results suggest that LOH on 10q23 is associated with metastatic recurrence of HCC.
Collapse
Affiliation(s)
- Tomoko Okuno
- Laboratory of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Pinto-Leite R, Botelho P, Ribeiro E, Oliveira PA, Santos L. Effect of sirolimus on urinary bladder cancer T24 cell line. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2009; 28:3. [PMID: 19128503 PMCID: PMC2628648 DOI: 10.1186/1756-9966-28-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2008] [Accepted: 01/07/2009] [Indexed: 12/18/2022]
Abstract
Background Sirolimus is recently reported to have antitumour effects on a large variety of cancers. The present study was performed to investigate sirolimus's ability to inhibit growth in T24 bladder cancer cells. Methods T24 bladder cancer cells were treated with various concentrations of sirolimus. MTT assay was used to evaluate the proliferation inhibitory effect on T24 cell line. The viability of T24 cell line was determined by Trypan blue exclusion analysis. Results Sirolimus inhibits the growth of bladder carcinoma cells and decreases their viability. Significant correlations were found between cell proliferation and sirolimus concentration (r = 0.830; p < 0.01) as well as between cell viability and sirolimus concentration (r = -0.896; p < 0.01). Conclusion Sirolimus has an anti-proliferation effect on the T24 bladder carcinoma cell line. The information from our results is useful for a better understanding sirolimus's anti-proliferative activity in the T24 bladder cancer cell line.
Collapse
Affiliation(s)
- Rosario Pinto-Leite
- Genetic Service, Cytogenetic laboratory, Hospital center of Trás-os-Montes and Alto Douro, Vila Real, Portugal.
| | | | | | | | | |
Collapse
|
31
|
Decaens T, Hurtova M, Duvoux C. Transplantation hépatique pour carcinome hépatocellulaire. ACTA ACUST UNITED AC 2009; 33:61-9. [DOI: 10.1016/j.gcb.2008.11.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
32
|
Sun Q, Zheng Y, Liu Q, Cao X. Rapamycin reverses TLR4 signaling-triggered tumor apoptosis resistance by disrupting Akt-mediated Bcl-xL upregulation. Int Immunopharmacol 2008; 8:1854-8. [DOI: 10.1016/j.intimp.2008.08.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2008] [Revised: 08/09/2008] [Accepted: 08/20/2008] [Indexed: 01/15/2023]
|
33
|
Sirolimus inhibits the growth and metastatic progression of hepatocellular carcinoma. J Cancer Res Clin Oncol 2008; 135:715-22. [PMID: 19002496 DOI: 10.1007/s00432-008-0506-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2007] [Accepted: 10/20/2008] [Indexed: 12/23/2022]
Abstract
PURPOSE Immunosuppressive therapy after liver transplantation for hepatocellular carcinoma (HCC) is one of the major contributory factors for HCC recurrence and metastasis. Sirolimus, a potent immunosuppressant, has been reported to be an effective inhibitor in a variety of tumors. The present study is designed to explore whether sirolimus could block the growth and metastatic progression of HCC. METHODS MHCC97H cells were used as targets to explore the effect of sirolimus on cell cycle progression, apoptosis, proliferation, and its antiangiogenic mechanism. LCI-D20, a highly metastatic model of human HCC in nude mice, was also used as the model tumor to explore the effect of sirolimus on tumor growth and metastatic progression. RESULTS In vitro, sirolimus induced cell cycle arrest at the G1 checkpoint and blocked proliferation of MHCC97H cells but did not induce apoptosis. In vivo, sirolimus prevented tumor growth and metastatic progression in LCI-D20. Intratumoral microvessel density and circulating levels of VEGF in tumor-bearing mice were also significantly reduced in sirolimus treatment group. Quantitative RT-PCR showed that sirolimus down-regulated the mRNA expression of VEGF and HIF-1a, but not of bFGF, and TGF-b in MHCC97H cells. Furthermore, western blot analysis confirmed that sirolimus also decreased expression of HIF-1a at protein level, in parallel with the down-regulation of the levels of VEGF protein excretion in a time-dependent manner as compared to untreated control cells following anoxia. CONCLUSIONS The immunosuppressive macrolide sirolimus prevents the growth and metastatic progression of HCC, and suppresses VEGF synthesis and secretion by downregulating HIF-1a expression. Sirolimus may be useful for clinical application in patients who received a liver transplant for HCC.
Collapse
|
34
|
Rapamycin inhibits proliferation and migration of hepatoma cells in vitro. J Surg Res 2008; 159:705-13. [PMID: 19482307 DOI: 10.1016/j.jss.2008.07.035] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2008] [Revised: 07/11/2008] [Accepted: 07/21/2008] [Indexed: 12/17/2022]
Abstract
BACKGROUND Systemic immunosuppression represents the major factor for cancer recurrence after orthotopic liver transplantation for hepatocellular carcinoma (HCC). Rapamycin is an immunosuppressant with unique antitumoral properties. Although rapamycin has been successfully used in HCC patients after liver transplantation, the detailed mechanisms of rapamycin action on tumor cells are poorly understood. METHODS Two HCC cell lines (PLC5 and HuH7) were used to evaluate the effect of rapamycin. Tumor cell proliferation was analyzed using cell counting and BrdU incorporation assay. Expression of phosphorylated Akt was studied using enzyme linked immunosorbent assay. Digital time-lapse microscopy was utilized to measure tumor cell migration in vitro. RESULTS Rapamycin induced a strongly dose-dependent inhibition of tumor cell proliferation in both HCC cell lines. Additionally, rapamycin inhibited activation of Akt phosphorylation and tumor cell migration after prolonged treatment. CONCLUSIONS Rapamycin suppresses tumor progression due to inhibition of phosphorylated Akt, cell proliferation, and migration. The data of the present study strengthen clinical implications of rapamycin after liver transplantation with HCC.
Collapse
|
35
|
Wang Z, Zhou J, Fan J, Qiu SJ, Yu Y, Huang XW, Tang ZY. Effect of rapamycin alone and in combination with sorafenib in an orthotopic model of human hepatocellular carcinoma. Clin Cancer Res 2008; 14:5124-30. [PMID: 18698030 DOI: 10.1158/1078-0432.ccr-07-4774] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE Novel therapeutic strategies are needed to prevent the tumor recurrence or metastasis after liver transplantation for hepatocellular carcinoma (HCC). This study was to investigate the effect of rapamycin, alone and in combination with sorafenib, on HCC in vivo. EXPERIMENTAL DESIGN Xenograft of a highly metastatic human HCC tumor (LCI-D20) was used to evaluate primary tumor growth and lung metastasis after treatment with rapamycin alone or in combination with sorafenib. Tumor cell proliferation was determined by Ki-67 immunostaining. To detect tumor cell apoptosis, the terminal deoxynucleotidyl-transferase-mediated dUTP nick-end labeling assay was used. Tumor angiogenesis was investigated by using a monoclonal anti-CD31 antibody. A vascular endothelial growth factor ELISA kit was used to measure vascular endothelial growth factor protein levels in the mice serum. RESULTS Rapamycin, alone and in combination with sorafenib, strongly inhibited primary tumor growth and lung metastases in LCI-D20 model. Furthermore, the combination therapy significantly enhanced the effect of antitumor on primary tumor growth compared with single treatment with either rapamycin (P < 0.001) or sorafenib (P < 0.001). Rapamycin alone inhibited HCC cell proliferation, induced apoptosis, and decreased tumor angiogenesis. Nevertheless, the combination therapy showed a significant inhibition of tumor cell proliferation (P < 0.05). Additionally, the combination therapy also further enhanced suppression of tumor cell angiogenesis compared with rapamycin treatment (P < 0.01). However, the induction of apoptosis in combination therapy group was not significantly higher than in the rapamycin-treated group (P > 0.05). CONCLUSIONS The combination therapy of rapamycin and sorafenib could be a new and promising therapeutic approach to the treatment of HCC and prevention of HCC recurrence after liver transplantation.
Collapse
Affiliation(s)
- Zheng Wang
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, PR China
| | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
New advancements have emerged in the field of hepatocellular carcinoma (HCC) in recent years. There has been a switch in the type of presentation of HCC in developed countries, with a clear increase of tumors <2 cm in diameter as a result of the wide implementation of surveillance programs. Non-invasive radiological techniques have been developed and validated for the diagnosis of small and tiny HCCs. Simultaneously, diagnostic criteria based on molecular profiling of early tumors have been proposed. The current clinical classification of HCC divides patients into 5 stages with a specific treatment-oriented schedule. There is no established molecular classification of HCC, although preliminary proposals have already been published. Advancements in the treatment arena have come from well designed trials. Radiofrequency ablation is currently consolidated as providing better local control of the disease compared with percutaneous ethanol injection. New devices are available to improve the anti-tumoral efficacy of conventional chemoembolization. Sorafenib, a multikinase inhibitor, has shown survival benefits in patients at advanced stages of the disease. This advancement represents a breakthrough in the management of this complex disease, and proves that molecular targeted therapies can be effective in this otherwise chemo-resistant tumor. Consequently, sorafenib will become the standard of care in advanced cases, and the control arm for future trials. Now, the research effort faces other areas of unmet need, such as the adjuvant setting of resection/local ablation and combination therapies.
Collapse
Affiliation(s)
- Josep M Llovet
- Barcelona Clínic Liver Cancer (BCLC) Group, Liver Unit, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), CIBERehd, Hospital Clìnic, Villarroel 170, 08036 Barcelona, Catalonia, Spain.
| | | |
Collapse
|
37
|
Yeap SK, Alitheen NB, Ali AM, Omar AR, Raha AR, Suraini AA, Muhajir AH. Effect of Rhaphidophora korthalsii methanol extract on human peripheral blood mononuclear cell (PBMC) proliferation and cytolytic activity toward HepG2. JOURNAL OF ETHNOPHARMACOLOGY 2007; 114:406-11. [PMID: 17884317 DOI: 10.1016/j.jep.2007.08.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2007] [Revised: 08/10/2007] [Accepted: 08/14/2007] [Indexed: 05/17/2023]
Abstract
The study of bioactivity of natural product is one of the major researches for drug discovery. The aim of this finding was to study the proliferation effect of Rhaphidophora korthalsii methanol extract on human PBMC and subsequently the cytotoxic effect of activated PBMC toward HepG2 human hepatocellular carcinoma. In this present study, MTT assay, cell cycle study and Annexin 5 binding assay were used to study the immunomodulatory and cytotoxic effects. In vitro cytotoxic screening of Rhaphidophora korthalsii methanol extract showed that the extract was non-toxic against hepatocellular carcinoma (HepG2). In contrast, the extract was able to stimulate the proliferation of human PBMC at 48 h and 72 h in MTT assay and cell cycle progress study. The application of immunomodulator in tumor research was studied by using MTT microcytotoxicity assay and flow cytometric Annexin V. Results indicated that pre-treated PBMC with Rhaphidophora korthalsii methanol extract induced the highest cytotoxicity (44.87+/-6.06% for MTT microcytotoxicity assay and 51.51+/-3.85% for Annexin V) toward HepG2. This finding demonstrates that Rhaphidophora korthalsii methanol extract are potent to stimulate the cytotoxic effect of immune cells toward HepG2.
Collapse
Affiliation(s)
- S K Yeap
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | | | | | | | | | | | | |
Collapse
|
38
|
The role of sirolimus in liver transplantation. Curr Opin Organ Transplant 2007. [DOI: 10.1097/mot.0b013e3282f1fbc7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|