1
|
Hartley T, Abdelmagid H, Abdulsalam Z, Mansion A, Howe E, Ramirez D, White K, Tadjuidje E. Embryotoxicity of statins and other prescribed drugs with reported off-target effects on cholesterol biosynthesis. Reprod Toxicol 2025; 132:108820. [PMID: 39667684 PMCID: PMC11890968 DOI: 10.1016/j.reprotox.2024.108820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 12/08/2024] [Accepted: 12/08/2024] [Indexed: 12/14/2024]
Abstract
Cholesterol plays pivotal cellular functions ranging from maintaining membrane fluidity to regulating cell-cell signaling. High cholesterol causes cardiovascular diseases, low cholesterol is linked to neuropsychiatric disorders, and inborn errors of cholesterol synthesis cause multisystem malformation syndromes. Statins lower cholesterol levels by inhibiting the first, rate-limiting reaction of the cholesterol biosynthesis pathway catalyzed by hydroxymethyl-glutaryl-Coenzyme A reductase (HMGCR). However, they have also been shown to interfere with cellular pathways that are unrelated to cholesterol synthesis. One of the last enzymes of cholesterol biosynthesis, 7-dehydrocholesterol reductase (DHCR7), is often mutated in the Smith-Lemli-Opitz syndrome (SLOS), a multisystem malformation syndrome. Strikingly, recent studies have shown that some prescribed psychotropic pharmaceuticals inhibit its activity. In this study, we used Xenopus laevis as a model organism to test the effects of 8 FDA-approved statins and selected prescribed psychotropic drugs on the developing vertebrate embryo. Drugs were tested at concentrations ranging from 0.1 µM to 50 µM. Embryos were exposed to the drugs from the blastula stage through the swimming tadpole stage with daily medium change. Our data show that statins are heterogenous with respect to their ability to cause embryonic lethality, with simvastatin, pitavastatin, lovastatin, cerivastatin, and fluvastatin being the most toxic ones. Observed phenotypes included delayed development, shortened body axis and pericardiac edema. On the other hand, psychotropic drugs were less embryonic lethal than statins but caused similar phenotypes as well as microcephaly and holoprosencephaly. Our findings suggest that the proximal and distal inhibition of cholesterol biosynthesis have different but overlapping effects on embryonic development.
Collapse
Affiliation(s)
- Taryn Hartley
- Department of Biological Sciences, Alabama State University, Montgomery, AL, United States; Center For NanoBiotechnology Research, Alabama State University, Montgomery, AL, United States
| | - Hagir Abdelmagid
- Department of Biological Sciences, Alabama State University, Montgomery, AL, United States; Center For NanoBiotechnology Research, Alabama State University, Montgomery, AL, United States
| | - Zeenat Abdulsalam
- Department of Biological Sciences, Alabama State University, Montgomery, AL, United States
| | | | - Emily Howe
- Department of Chemistry, Gettysburg College, Gettysburg, PA, United States
| | - Daniel Ramirez
- Department of Biology, Savannah State University, United States
| | - Kaylei White
- A & M College, Southern University, Baton Rouge, LA, United States
| | - Emmanuel Tadjuidje
- Department of Biological Sciences, Alabama State University, Montgomery, AL, United States; Center For NanoBiotechnology Research, Alabama State University, Montgomery, AL, United States.
| |
Collapse
|
2
|
Kobayashi K, Murakami K, Baba K. Effects of Lipophilic Statins on Cell Viability and Tissue Factor Expression in Canine Haemangiosarcoma Cells. Vet Comp Oncol 2024; 22:581-591. [PMID: 39319370 DOI: 10.1111/vco.13012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/18/2024] [Accepted: 09/06/2024] [Indexed: 09/26/2024]
Abstract
Canine haemangiosarcoma (HSA) is a highly aggressive cancer often associated with coagulation abnormalities. Statins, inhibitors of 3-hydroxy-3-methyl-glutaryl-CoA reductase (HMGCR) clinically prescribed for hypercholesterolemia, are also believed to possess antitumour and anticoagulant properties by inhibiting downstream Akt activation. Akt phosphorylation is involved in the mechanism of the antitumour and tissue factor (TF)-lowering effects of statins. In the present study, we aimed to investigate whether statins could inhibit cell viability while concurrently inducing anticoagulant properties by regulating the expression of TFs in canine HSA cells. Using reverse transcription-quantitative polymerase chain reaction (RT-qPCR), we initially exclusively detected HMGCR mRNA expression in canine HSA tissues and cell lines but not in normal cephalic vein and spleen tissues. Moreover, treatment with lipophilic statins, including atorvastatin, fluvastatin, and simvastatin, inhibited cell viability in a concentration-dependent manner and decreased TF expression both at the mRNA and protein levels, as evidenced by cell viability assays, RT-qPCR, and immunoblotting, respectively. Further investigation using cell viability assays and flow cytometry revealed that simvastatin decreased Akt phosphorylation, and MK-2206, a specific Akt inhibitor, mirrored the effect of simvastatin on cell viability and cell cycle arrest. However, MK-2206 exhibited different effects on TF expression depending on the cell type, indicating that Akt phosphorylation may not consistently regulate TF expression. Overall, this study provides insights into the potential therapeutic use of statins in targeting tumour growth and coagulation abnormalities in canine HSA. Further research is warranted to fully elucidate the underlying mechanisms and clinical applications of statins in canine HSA treatment.
Collapse
Affiliation(s)
- Kosuke Kobayashi
- Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Japan
| | - Kohei Murakami
- Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Japan
| | - Kenji Baba
- Laboratory of Veterinary Internal Medicine, The United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi, Japan
| |
Collapse
|
3
|
Gbelcová H, Rimpelová S, Jariabková A, Macášek P, Priščáková P, Ruml T, Šáchová J, Kubovčiak J, Kolář M, Vítek L. Highly variable biological effects of statins on cancer, non-cancer, and stem cells in vitro. Sci Rep 2024; 14:11830. [PMID: 38782983 PMCID: PMC11116523 DOI: 10.1038/s41598-024-62615-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 05/20/2024] [Indexed: 05/25/2024] Open
Abstract
Statins, the drugs used for the treatment of hypercholesterolemia, have come into the spotlight not only as chemoadjuvants, but also as potential stem cell modulators in the context of regenerative therapy. In our study, we compared the in vitro effects of all clinically used statins on the viability of human pancreatic cancer (MiaPaCa-2) cells, non-cancerous human embryonic kidney (HEK 293) cells and adipose-derived mesenchymal stem cells (ADMSC). Additionally, the effect of statins on viability of MiaPaCa-2 and ADMSC cells spheroids was tested. Furthermore, we performed a microarray analysis on ADMSCs treated with individual statins (12 μM) and compared the importance of the effects of statins on gene expression between stem cells and pancreatic cancer cells. Concentrations of statins that significantly affected cancer cells viability (< 40 μM) did not affect stem cells viability after 24 h. Moreover, statins that didn´t affect viability of cancer cells grown in a monolayer, induce the disintegration of cancer cell spheroids. The effect of statins on gene expression was significantly less pronounced in stem cells compared to pancreatic cancer cells. In conclusion, the low efficacy of statins on non-tumor and stem cells at concentrations sufficient for cancer cells growth inhibition, support their applicability in chemoadjuvant tumor therapy.
Collapse
Affiliation(s)
- Helena Gbelcová
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Bratislava, 813 72, Slovak Republic.
| | - Silvie Rimpelová
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, 166 28, Czech Republic
| | - Adriana Jariabková
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Bratislava, 813 72, Slovak Republic
| | - Patrik Macášek
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Bratislava, 813 72, Slovak Republic
| | - Petra Priščáková
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Bratislava, 813 72, Slovak Republic
| | - Tomáš Ruml
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, 166 28, Czech Republic
| | - Jana Šáchová
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, 142 20, Czech Republic
| | - Jan Kubovčiak
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, 142 20, Czech Republic
| | - Michal Kolář
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, 142 20, Czech Republic
- Department of Informatics and Chemistry, University of Chemistry and Technology, Prague, 166 28, Czech Republic
| | - Libor Vítek
- Institute of Medical Biochemistry and Laboratory Diagnostics, and 4Th Department of Internal Medicine, 1St Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, 121 08, Czech Republic
| |
Collapse
|
4
|
McKechnie T, Brown Z, Lovrics O, Yang S, Kazi T, Eskicioglu C, Parvez E. Concurrent Use of Statins in Patients Undergoing Curative Intent Treatment for Triple Negative Breast Cancer: A Systematic Review and Meta-Analysis. Clin Breast Cancer 2024; 24:e103-e115. [PMID: 38296737 DOI: 10.1016/j.clbc.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 02/02/2024]
Abstract
Pre-clinical studies in triple negative breast cancer (TNBC) suggest that statins may inhibit cell proliferation, promote cell-cycle arrest, induce apoptosis, change the tumor microenvironment, and improve effectiveness of other therapies. Observational studies have demonstrated variable effects from statin therapy on oncologic outcomes in these patients. As such, we aimed to pool previous data via a systematic review and meta-analysis to elucidate the impact of concurrent statin use on oncologic outcomes for patients with TNBC. Medline, EMBASE, CENTRAL, and PubMed were systematically searched from inception through to June 2022. Studies were included if they compared patients with TNBC receiving and not receiving statin therapy concurrently with oncologic treatment for curative intent in terms of recurrence and survival in a non-metastatic setting. The primary outcomes were 5-year disease-free survival (DFS) and 5-year overall survival (OS). A pairwise meta-analyses was performed using inverse variance random effects. Risk of bias was assessed with the ROBINS-I and the GRADE approach was conducted to assess quality of evidence. From 4014 citations, 5 studies with 625 patients on statin therapy and 2707 patients not on statin therapy were included. There was a significant increase in 5-year DFS for patients on statin therapy compared to patients not on statin therapy (OR 1.44, 95% CI 1.04-1.98, P = .03). No significant difference was noted in 5-year OS between the 2 groups (OR 1.12, 95% CI 0.86-1.47, P = .40). Included studies were at moderate-to-high risk of bias. The GRADE quality of evidence was very low. This review presents very low-quality evidence that concurrent use of statins with oncologic treatment may potentially improve long-term DFS for patients with TNBC undergoing curative intent therapy. Future research by way of large, prospective study is required to further clarify the clinical utility of statins on patients undergoing treatment for TNBC.
Collapse
Affiliation(s)
- Tyler McKechnie
- Division of General Surgery, Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Zachary Brown
- Division of General Surgery, Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Olivia Lovrics
- Division of General Surgery, Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Shuling Yang
- Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Tania Kazi
- Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Cagla Eskicioglu
- Division of General Surgery, Department of Surgery, McMaster University, Hamilton, Ontario, Canada; Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Elena Parvez
- Division of General Surgery, Department of Surgery, McMaster University, Hamilton, Ontario, Canada; Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
5
|
Ishikawa T, Irie N, Tashiro J, Osaki T, Warita T, Warita K, Naito M. Comparison of the anticancer effects of various statins on canine oral melanoma cells. Vet Comp Oncol 2024; 22:156-161. [PMID: 38044042 DOI: 10.1111/vco.12946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 10/29/2023] [Accepted: 11/17/2023] [Indexed: 12/05/2023]
Abstract
Canine oral melanoma is a highly malignant cancer with a poor prognosis. Statins, commonly used drugs for treating dyslipidemia, exhibit pleiotropic anticancer effects and marked anti-proliferative effects against melanoma cells. The anticancer effects among statins vary; in human cancers, lipophilic statins have shown stronger anticancer effects compared with hydrophilic statins. However, data on the differences in the effects of various statins on canine cancer cells are lacking, hence the optimal statins for treating canine melanoma remain unknown. Therefore, this study aimed to clarify the most effective statin by comparing the anticancer effects of hydrophilic rosuvastatin and lipophilic atorvastatin, simvastatin, fluvastatin and pitavastatin on three canine oral melanoma cell lines. Time-dependent measurement of cell confluence showed that lipophilic statins had a stronger anti-proliferative effect on all cell lines than hydrophilic rosuvastatin. Quantification of lactate dehydrogenase release, an indicator of cytotoxicity, showed that lipophilic statins more effectively induced cell death than hydrophilic rosuvastatin. Lipophilic statins affected both inhibition of cell proliferation and induction of cell death. The anticancer effects of statins on canine oral melanoma cells differed in the following ascending order of IC50 values: pitavastatin < fluvastatin = simvastatin < atorvastatin < rosuvastatin. The required concentration of pitavastatin was approximately 1/20th that of rosuvastatin. Among the statins used in this study, pitavastatin had the highest anticancer effect. Our results suggest lipophilic pitavastatin as the optimal statin for treating canine oral melanoma.
Collapse
Affiliation(s)
- Takuro Ishikawa
- Department of Anatomy, School of Medicine, Aichi Medical University, Nagakute, Aichi, Japan
| | - Nanami Irie
- Graduate School of Science and Technology, Kwansei Gakuin University, Sanda, Hyogo, Japan
| | - Jiro Tashiro
- Department of Veterinary Anatomy, School of Veterinary Medicine, Tottori University, Tottori, Japan
| | - Tomohiro Osaki
- Department of Veterinary Clinical Medicine, School of Veterinary Medicine, Tottori University, Tottori, Japan
| | - Tomoko Warita
- Graduate School of Science and Technology, Kwansei Gakuin University, Sanda, Hyogo, Japan
| | - Katsuhiko Warita
- Department of Veterinary Anatomy, School of Veterinary Medicine, Tottori University, Tottori, Japan
| | - Munekazu Naito
- Department of Anatomy, School of Medicine, Aichi Medical University, Nagakute, Aichi, Japan
| |
Collapse
|
6
|
Rebelo A, Kleeff J, Sunami Y. Cholesterol Metabolism in Pancreatic Cancer. Cancers (Basel) 2023; 15:5177. [PMID: 37958351 PMCID: PMC10650553 DOI: 10.3390/cancers15215177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/23/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
Pancreatic cancer's substantial impact on cancer-related mortality, responsible for 8% of cancer deaths and ranking fourth in the US, persists despite advancements, with a five-year relative survival rate of only 11%. Forecasts predict a 70% surge in new cases and a 72% increase in global pancreatic cancer-related deaths by 2040. This review explores the intrinsic metabolic reprogramming of pancreatic cancer, focusing on the mevalonate pathway, including cholesterol biosynthesis, transportation, targeting strategies, and clinical studies. The mevalonate pathway, central to cellular metabolism, significantly shapes pancreatic cancer progression. Acetyl coenzyme A (Acetyl-CoA) serves a dual role in fatty acid and cholesterol biosynthesis, fueling acinar-to-ductal metaplasia (ADM) and pancreatic intraepithelial neoplasia (PanIN) development. Enzymes, including acetoacetyl-CoA thiolase, 3-hydroxy-3methylglutaryl-CoA (HMG-CoA) synthase, and HMG-CoA reductase, are key enzymes in pancreatic cancer. Inhibiting HMG-CoA reductase, e.g., by using statins, shows promise in delaying PanIN progression and impeding pancreatic cancer. Dysregulation of cholesterol modification, uptake, and transport significantly impacts tumor progression, with Sterol O-acyltransferase 1 (SOAT1) driving cholesterol ester (CE) accumulation and disrupted low-density lipoprotein receptor (LDLR) expression contributing to cancer recurrence. Apolipoprotein E (ApoE) expression in tumor stroma influences immune suppression. Clinical trials targeting cholesterol metabolism, including statins and SOAT1 inhibitors, exhibit potential anti-tumor effects, and combination therapies enhance efficacy. This review provides insights into cholesterol metabolism's convergence with pancreatic cancer, shedding light on therapeutic avenues and ongoing clinical investigations.
Collapse
Affiliation(s)
| | | | - Yoshiaki Sunami
- Department of Visceral, Vascular and Endocrine Surgery, University Medical Center Halle, Martin-Luther-University Halle-Wittenberg, 06120 Halle, Germany; (A.R.); (J.K.)
| |
Collapse
|
7
|
Ivermectin Augments the Anti-Cancer Activity of Pitavastatin in Ovarian Cancer Cells. Diseases 2023; 11:diseases11010049. [PMID: 36975598 PMCID: PMC10047003 DOI: 10.3390/diseases11010049] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/10/2023] [Accepted: 03/10/2023] [Indexed: 03/16/2023] Open
Abstract
We have previously shown that pitavastatin has the potential to be used to treat ovarian cancer, although relatively high doses are likely to be necessary. One solution to this problem is to identify drugs that are synergistic with pitavastatin, thereby reducing the dose that is necessary to have a therapeutic effect. Here, we tested combinations of pitavastatin with the anti-parasitic drug ivermectin in six ovarian cancer cell lines. When tested on its own, ivermectin inhibited the growth of the cells but only with modest potency (IC50 = 10–20 µM). When the drugs were combined and assessed in cell growth assays, ivermectin showed synergy with pitavastatin in 3 cell lines and this was most evident in COV-318 cells (combination index ~ 0.6). Ivermectin potentiated the reduction in COV-318 cell viability caused by pitavastatin by 20–25% as well as potentiating apoptosis induced by pitavastatin, assessed by activation of caspase-3/7 (2–4 fold) and annexin-labelling (3–5 fold). These data suggest that ivermectin may be useful in the treatment of ovarian cancer when combined with pitavastatin, but methods to achieve an adequate ivermectin concentration in tumour tissue will be necessary.
Collapse
|
8
|
Cerivastatin Synergizes with Trametinib and Enhances Its Efficacy in the Therapy of Uveal Melanoma. Cancers (Basel) 2023; 15:cancers15030886. [PMID: 36765842 PMCID: PMC9913575 DOI: 10.3390/cancers15030886] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/27/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Metastatic uveal melanoma (MUM) is a highly aggressive, therapy-resistant disease. Driver mutations in Gα-proteins GNAQ and GNA11 activate MAP-kinase and YAP/TAZ pathways of oncogenic signalling. MAP-kinase and MEK-inhibitors do not significantly block MUM progression, likely due to persisting YAP/TAZ signalling. Statins inhibit YAP/TAZ activation by blocking the mevalonate pathway, geranyl-geranylation, and subcellular localisation of the Rho-GTPase. We investigated drugs that affect the YAP/TAZ pathway, valproic acid, verteporfin and statins, in combination with MEK-inhibitor trametinib. METHODS We established IC50 values of the individual drugs and monitored the effects of their combinations in terms of proliferation. We selected trametinib and cerivastatin for evaluation of cell cycle and apoptosis. Synergism was detected using isobologram and Chou-Talalay analyses. The most synergistic combination was tested in vivo. RESULTS Synergistic concentrations of trametinib and cerivastatin induced a massive arrest of proliferation and cell cycle and enhanced apoptosis, particularly in the monosomic, BAP1-mutated UPMM3 cell line. The combined treatment reduced ERK and AKT phosphorylation, increased the inactive, cytoplasmatic form of YAP and significantly impaired the growth of UM cells with monosomy of chromosome 3 in NSG mice. CONCLUSION Statins can potentiate the efficacy of MEK inhibitors in the therapy of UM.
Collapse
|
9
|
Chen CY, Yang YF, Wang PC, Shan L, Lin S, Chen PJ, Chen YJ, Chiang HS, Lin JT, Hung CF, Liang YJ. Simvastatin Attenuated Tumor Growth in Different Pancreatic Tumor Animal Models. Pharmaceuticals (Basel) 2022; 15:1408. [PMID: 36422538 PMCID: PMC9692350 DOI: 10.3390/ph15111408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/01/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
Newly diagnosed pancreatic cancer increases year by year, while the prognosis of pancreatic cancer has not been very good. Statin drugs were found to have protective effects against a variety of cancers, but their association with pancreatic cancer remains to be clarified. This study used different pancreatic cancer cell lines and in different animal models to confirm the relationship between simvastatin and pancreatic cancer. Flow cytometry and luciferase-based bioluminescent images were used to investigate the cell cycle and tumor growth changes under simvastatin treatment. Simvastatin decreased the MIA PaCa-2 cells, PANC-1 cells, and BxPC-3 cell viability significantly and may arrest the cell cycle in the G0 phase. During in vivo study, subcutaneously implanted simvastatin pre-treated pancreatic cancer cells and intraperitoneally treated simvastatin continuously demonstrated a slower tumor growth rate and decreased the tumor/body weight ratio significantly. In intravenous implant models, implanted simvastatin-pre-treated BxPC-3 cells and cells treated along with simvastatin significantly decreased the tumor growth curve. Implanting the simvastatin-pre-treated pancreatic cells in the subcutaneous model showed better growth inhibition than the intravenous model. These results suggest simvastatin treatment may relate to different signaling pathways in local growth and metastasis. Pancreatic cancer cells presented different growth patterns in different animal-induced models, which could be important for clinical reference when it comes to the relationship of long-term statin use and pancreatic cancer.
Collapse
Affiliation(s)
- Chao-Yi Chen
- Department and Institute of Life Science, Fu-Jen Catholic University, New Taipei City 242062, Taiwan
- Graduate Institute of Applied Science and Engineering, Fu-Jen Catholic University, New Taipei City 242062, Taiwan
| | - Yi-Feng Yang
- Department and Institute of Life Science, Fu-Jen Catholic University, New Taipei City 242062, Taiwan
- Graduate Institute of Applied Science and Engineering, Fu-Jen Catholic University, New Taipei City 242062, Taiwan
| | - Paul C. Wang
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC 20060, USA
- College of Science and Engineering, Fu Jen Catholic University, New Taipei City 242062, Taiwan
| | - Liang Shan
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC 20060, USA
| | - Stephen Lin
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC 20060, USA
| | - Po-Jung Chen
- Department and Institute of Life Science, Fu-Jen Catholic University, New Taipei City 242062, Taiwan
- Graduate Institute of Applied Science and Engineering, Fu-Jen Catholic University, New Taipei City 242062, Taiwan
| | - Yi-Jung Chen
- Department and Institute of Life Science, Fu-Jen Catholic University, New Taipei City 242062, Taiwan
| | - Han-Sun Chiang
- School of Medicine, Fu-Jen Catholic University, New Taipei City 242062, Taiwan
| | - Jaw-Town Lin
- School of Medicine, Fu-Jen Catholic University, New Taipei City 242062, Taiwan
- Digestive Medicine Center, China Medical University Hospital, Taichung 404327, Taiwan
| | - Chi-Feng Hung
- School of Medicine, Fu-Jen Catholic University, New Taipei City 242062, Taiwan
| | - Yao-Jen Liang
- Department and Institute of Life Science, Fu-Jen Catholic University, New Taipei City 242062, Taiwan
- Graduate Institute of Applied Science and Engineering, Fu-Jen Catholic University, New Taipei City 242062, Taiwan
| |
Collapse
|
10
|
Wu Y, Meng W, Guan M, Zhao X, Zhang C, Fang Q, Zhang Y, Sun Z, Cai M, Huang D, Yang X, Yu Y, Cui Y, He S, Chai R. Pitavastatin protects against neomycin-induced ototoxicity through inhibition of endoplasmic reticulum stress. Front Mol Neurosci 2022; 15:963083. [PMID: 35992197 PMCID: PMC9381809 DOI: 10.3389/fnmol.2022.963083] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
Irreversible injury to inner ear hair cells induced by aminoglycoside antibiotics contributes to the formation of sensorineural hearing loss. Pitavastatin (PTV), a 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor, has been reported to exert neuroprotective effects. However, its role in aminoglycoside-induced hearing loss remains unknown. The objectives of this study were to investigate the beneficial effects, as well as the mechanism of action of PTV against neomycin-induced ototoxicity. We found that PTV remarkably reduced hair cell loss in mouse cochlear explants and promoted auditory HEI-OC1 cells survival after neomycin stimulation. We also observed that the auditory brainstem response threshold that was increased by neomycin was significantly reduced by pretreatment with PTV in mice. Furthermore, neomycin-induced endoplasmic reticulum stress in hair cells was attenuated by PTV treatment through inhibition of PERK/eIF2α/ATF4 signaling. Additionally, we found that PTV suppressed the RhoA/ROCK/JNK signal pathway, which was activated by neomycin stimulation in HEI-OC1 cells. Collectively, our results showed that PTV might serve as a promising therapeutic agent against aminoglycoside-induced ototoxicity.
Collapse
Affiliation(s)
- Yunhao Wu
- State Key Laboratory of Bioelectronics, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Department of Otolaryngology Head and Neck Surgery, School of Life Sciences and Technology, Zhongda Hospital, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Wei Meng
- Department of Otorhinolaryngology Head and Neck Surgery, School of Medicine, Nanjing Tongren Hospital, Southeast University, Nanjing, China
| | - Ming Guan
- Department of Otolaryngology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaolong Zhao
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Chen Zhang
- Beijing Key Laboratory of Neural Regeneration and Repair, Department of Neurobiology, Advanced Innovation Center for Human Brain Protection, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Qiaojun Fang
- State Key Laboratory of Bioelectronics, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Department of Otolaryngology Head and Neck Surgery, School of Life Sciences and Technology, Zhongda Hospital, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Yuhua Zhang
- State Key Laboratory of Bioelectronics, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Department of Otolaryngology Head and Neck Surgery, School of Life Sciences and Technology, Zhongda Hospital, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Zihui Sun
- Department of Otorhinolaryngology Head and Neck Surgery, School of Medicine, Nanjing Tongren Hospital, Southeast University, Nanjing, China
| | - Mingjing Cai
- Department of Otorhinolaryngology Head and Neck Surgery, School of Medicine, Nanjing Tongren Hospital, Southeast University, Nanjing, China
| | - Dongdong Huang
- Department of Otorhinolaryngology Head and Neck Surgery, School of Medicine, Nanjing Tongren Hospital, Southeast University, Nanjing, China
| | - Xuechun Yang
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yafeng Yu
- Department of Otolaryngology, First Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Yafeng Yu,
| | - Yong Cui
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- The Second School of Clinical Medicine, South Medical University, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
- Yong Cui,
| | - Shuangba He
- Department of Otorhinolaryngology Head and Neck Surgery, School of Medicine, Nanjing Tongren Hospital, Southeast University, Nanjing, China
- Shuangba He,
| | - Renjie Chai
- State Key Laboratory of Bioelectronics, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Department of Otolaryngology Head and Neck Surgery, School of Life Sciences and Technology, Zhongda Hospital, Advanced Institute for Life and Health, Southeast University, Nanjing, China
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
- Renjie Chai,
| |
Collapse
|
11
|
Mutant p53, the Mevalonate Pathway and the Tumor Microenvironment Regulate Tumor Response to Statin Therapy. Cancers (Basel) 2022; 14:cancers14143500. [PMID: 35884561 PMCID: PMC9323637 DOI: 10.3390/cancers14143500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/15/2022] [Accepted: 07/18/2022] [Indexed: 11/16/2022] Open
Abstract
Tumor cells have the ability to co-opt multiple metabolic pathways, enhance glucose uptake and utilize aerobic glycolysis to promote tumorigenesis, which are characteristics constituting an emerging hallmark of cancer. Mutated tumor suppressor and proto-oncogenes are frequently responsible for enhanced metabolic pathway signaling. The link between mutant p53 and the mevalonate (MVA) pathway has been implicated in the advancement of various malignancies, with tumor cells relying heavily on increased MVA signaling to fuel their rapid growth, metastatic spread and development of therapy resistance. Statin drugs inhibit HMG-CoA reductase, the pathway’s rate-limiting enzyme, and as such, have long been studied as a potential anti-cancer therapy. However, whether statins provide additional anti-cancer properties is worthy of debate. Here, we examine retrospective, prospective and pre-clinical studies involving the use of statins in various cancer types, as well as potential issues with statins’ lack of efficacy observed in clinical trials and future considerations for upcoming clinical trials.
Collapse
|
12
|
Comprehensive Analysis of Differential Gene Expression Profile via RNA Sequencing in the Human Ovarian Cancer SKOV3 Cells Treated with Simvastatin. Chem Res Chin Univ 2022. [DOI: 10.1007/s40242-022-1325-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
13
|
Dorsch M, Kowalczyk M, Planque M, Heilmann G, Urban S, Dujardin P, Forster J, Ueffing K, Nothdurft S, Oeck S, Paul A, Liffers ST, Kaschani F, Kaiser M, Schramm A, Siveke JT, Winslow MM, Fendt SM, Nalbant P, Grüner BM. Statins affect cancer cell plasticity with distinct consequences for tumor progression and metastasis. Cell Rep 2021; 37:110056. [PMID: 34818551 PMCID: PMC8640221 DOI: 10.1016/j.celrep.2021.110056] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 09/21/2021] [Accepted: 11/03/2021] [Indexed: 12/12/2022] Open
Abstract
Statins are among the most commonly prescribed drugs, and around every fourth person above the age of 40 is on statin medication. Therefore, it is of utmost clinical importance to understand the effect of statins on cancer cell plasticity and its consequences to not only patients with cancer but also patients who are on statins. Here, we find that statins induce a partial epithelial-to-mesenchymal transition (EMT) phenotype in cancer cells of solid tumors. Using a comprehensive STRING network analysis of transcriptome, proteome, and phosphoproteome data combined with multiple mechanistic in vitro and functional in vivo analyses, we demonstrate that statins reduce cellular plasticity by enforcing a mesenchymal-like cell state that increases metastatic seeding ability on one side but reduces the formation of (secondary) tumors on the other due to heterogeneous treatment responses. Taken together, we provide a thorough mechanistic overview of the consequences of statin use for each step of cancer development, progression, and metastasis.
Collapse
Affiliation(s)
- Madeleine Dorsch
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen at the University Duisburg-Essen, Duisburg, Germany
| | - Manuela Kowalczyk
- Department of Molecular Cell Biology, Center for Medical Biotechnology, University of Duisburg-Essen, Duisburg, Germany
| | - Mélanie Planque
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Geronimo Heilmann
- Department of Chemical Biology, Center for Medical Biotechnology, University of Duisburg-Essen, Duisburg, Germany
| | - Sebastian Urban
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen at the University Duisburg-Essen, Duisburg, Germany
| | - Philip Dujardin
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen at the University Duisburg-Essen, Duisburg, Germany
| | - Jan Forster
- Department of Genome Informatics, Institute for Human Genetics, University of Duisburg-Essen, Duisburg, Germany; German Cancer Consortium (DKTK) partner site Essen, Essen, Germany
| | - Kristina Ueffing
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen at the University Duisburg-Essen, Duisburg, Germany
| | - Silke Nothdurft
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen at the University Duisburg-Essen, Duisburg, Germany
| | - Sebastian Oeck
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen at the University Duisburg-Essen, Duisburg, Germany
| | - Annika Paul
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen at the University Duisburg-Essen, Duisburg, Germany
| | - Sven T Liffers
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Medicine Essen, Essen, Germany
| | - Farnusch Kaschani
- Department of Chemical Biology, Center for Medical Biotechnology, University of Duisburg-Essen, Duisburg, Germany
| | - Markus Kaiser
- Department of Chemical Biology, Center for Medical Biotechnology, University of Duisburg-Essen, Duisburg, Germany
| | - Alexander Schramm
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen at the University Duisburg-Essen, Duisburg, Germany
| | - Jens T Siveke
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Medicine Essen, Essen, Germany; Division of Solid Tumor Translational Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), partner site Essen, Heidelberg, Germany
| | - Monte M Winslow
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA; Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Perihan Nalbant
- Department of Molecular Cell Biology, Center for Medical Biotechnology, University of Duisburg-Essen, Duisburg, Germany
| | - Barbara M Grüner
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen at the University Duisburg-Essen, Duisburg, Germany; German Cancer Consortium (DKTK) partner site Essen, Essen, Germany.
| |
Collapse
|
14
|
Duarte RRR, Copertino DC, Iñiguez LP, Marston JL, Bram Y, Han Y, Schwartz RE, Chen S, Nixon DF, Powell TR. Identifying FDA-approved drugs with multimodal properties against COVID-19 using a data-driven approach and a lung organoid model of SARS-CoV-2 entry. Mol Med 2021; 27:105. [PMID: 34503440 PMCID: PMC8426591 DOI: 10.1186/s10020-021-00356-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 08/16/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Vaccination programs have been launched worldwide to halt the spread of COVID-19. However, the identification of existing, safe compounds with combined treatment and prophylactic properties would be beneficial to individuals who are waiting to be vaccinated, particularly in less economically developed countries, where vaccine availability may be initially limited. METHODS We used a data-driven approach, combining results from the screening of a large transcriptomic database (L1000) and molecular docking analyses, with in vitro tests using a lung organoid model of SARS-CoV-2 entry, to identify drugs with putative multimodal properties against COVID-19. RESULTS Out of thousands of FDA-approved drugs considered, we observed that atorvastatin was the most promising candidate, as its effects negatively correlated with the transcriptional changes associated with infection. Atorvastatin was further predicted to bind to SARS-CoV-2's main protease and RNA-dependent RNA polymerase, and was shown to inhibit viral entry in our lung organoid model. CONCLUSIONS Small clinical studies reported that general statin use, and specifically, atorvastatin use, are associated with protective effects against COVID-19. Our study corroborrates these findings and supports the investigation of atorvastatin in larger clinical studies. Ultimately, our framework demonstrates one promising way to fast-track the identification of compounds for COVID-19, which could similarly be applied when tackling future pandemics.
Collapse
Affiliation(s)
- Rodrigo R R Duarte
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, Cornell University, Belfer Research Building, 5th floor, 413 E. 69th St., New York, NY, 10021, USA.
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
| | - Dennis C Copertino
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, Cornell University, Belfer Research Building, 5th floor, 413 E. 69th St., New York, NY, 10021, USA
| | - Luis P Iñiguez
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, Cornell University, Belfer Research Building, 5th floor, 413 E. 69th St., New York, NY, 10021, USA
| | - Jez L Marston
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, Cornell University, Belfer Research Building, 5th floor, 413 E. 69th St., New York, NY, 10021, USA
| | - Yaron Bram
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Yuling Han
- Department of Surgery, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Robert E Schwartz
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Douglas F Nixon
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, Cornell University, Belfer Research Building, 5th floor, 413 E. 69th St., New York, NY, 10021, USA
| | - Timothy R Powell
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, Cornell University, Belfer Research Building, 5th floor, 413 E. 69th St., New York, NY, 10021, USA
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| |
Collapse
|
15
|
Støer NC, Bouche G, Pantziarka P, Sloan EK, Andreassen BK, Botteri E. Use of non-cancer drugs and survival among patients with pancreatic adenocarcinoma: a nationwide registry-based study in Norway. Acta Oncol 2021; 60:1146-1153. [PMID: 34338111 DOI: 10.1080/0284186x.2021.1953136] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND The prognosis of pancreatic cancer is poor and new treatment strategies are urgently needed. To identify non-cancer drugs that could be re-purposed for cancer, we investigated the association between the use of selected drugs and cancer-specific mortality in a nationwide cohort of pancreatic cancer patients. MATERIAL AND METHODS The study is based on linkage between the Cancer Registry of Norway and the Norwegian Prescription Database, comprising 2614 pancreatic cancer patients diagnosed between 2007 and 2014. We evaluated the association between use at diagnosis of a pre-defined list of non-cancer drugs, including metformin, antihypertensives, and statins, and pancreatic cancer-specific mortality, using Cox regression. Patients were defined as users of a particular drug if it was prescribed before diagnosis, and the prescription covered the date of diagnosis. RESULTS In total, 2096 (80.2%) patients died from pancreatic cancer; median survival was 6 months. Statin users (n = 621) had lower mortality (hazard ratio (HR): 0.86; 95% confidence interval (CI) 0.76-0.97) compared to non-users (n = 1993). This association was more pronounced (P-heterogeneity 0.062) in users of hydrophilic (n = 37, HR: 0.61; 95% CI 0.42-0.90) than lipophilic (n = 587, HR: 0.87; 95% CI 0.78-0.98) statins. An indication for lower mortality (HR: 0.85; 95% CI 0.69-1.05) was observed in users of non-selective beta-blockers (n = 113) compared to non-users (n = 2501). Notably, when compared to users of other antihypertensives (n = 643), users of non-selective beta-blockers (n = 40) had lower mortality (HR 0.67; 95% CI 0.47-0.96). The use of other drugs, including selective beta-blockers and metformin, was not associated with mortality. CONCLUSION The findings suggest an association between the use of statins and non-selective beta-blockers and reduced pancreatic cancer mortality, and add to the literature supporting the design of randomised clinical trials to evaluate those drugs in the management of pancreatic cancer.
Collapse
Affiliation(s)
| | | | | | - Erica K. Sloan
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Division of Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | | | - Edoardo Botteri
- Department of Research, Cancer Registry of Norway, Oslo, Norway
- Section for Colorectal Cancer Screening, Cancer Registry of Norway, Oslo, Norway
| |
Collapse
|
16
|
Comparison of Transcriptomic Profiles of MiaPaCa-2 Pancreatic Cancer Cells Treated with Different Statins. Molecules 2021; 26:molecules26123528. [PMID: 34207840 PMCID: PMC8226792 DOI: 10.3390/molecules26123528] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/21/2021] [Accepted: 05/26/2021] [Indexed: 01/24/2023] Open
Abstract
Statins have been widely used for the treatment of hypercholesterolemia due to their ability to inhibit HMG-CoA reductase, the rate-limiting enzyme of de novo cholesterol synthesis, via the so-called mevalonate pathway. However, their inhibitory action also causes depletion of downstream intermediates of the pathway, resulting in the pleiotropic effects of statins, including the beneficial impact in the treatment of cancer. In our study, we compared the effect of all eight existing statins on the expression of genes, the products of which are implicated in cancer inhibition and suggested the molecular mechanisms of their action in epigenetic and posttranslational regulation, and in cell-cycle arrest, death, migration, or invasion of the cancer cells.
Collapse
|
17
|
Wujak M, Kozakiewicz A, Ciarkowska A, Loch JI, Barwiolek M, Sokolowska Z, Budny M, Wojtczak A. Assessing the Interactions of Statins with Human Adenylate Kinase Isoenzyme 1: Fluorescence and Enzyme Kinetic Studies. Int J Mol Sci 2021; 22:ijms22115541. [PMID: 34073952 PMCID: PMC8197361 DOI: 10.3390/ijms22115541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 05/16/2021] [Accepted: 05/21/2021] [Indexed: 11/16/2022] Open
Abstract
Statins are the most effective cholesterol-lowering drugs. They also exert many pleiotropic effects, including anti-cancer and cardio- and neuro-protective. Numerous nano-sized drug delivery systems were developed to enhance the therapeutic potential of statins. Studies on possible interactions between statins and human proteins could provide a deeper insight into the pleiotropic and adverse effects of these drugs. Adenylate kinase (AK) was found to regulate HDL endocytosis, cellular metabolism, cardiovascular function and neurodegeneration. In this work, we investigated interactions between human adenylate kinase isoenzyme 1 (hAK1) and atorvastatin (AVS), fluvastatin (FVS), pravastatin (PVS), rosuvastatin (RVS) and simvastatin (SVS) with fluorescence spectroscopy. The tested statins quenched the intrinsic fluorescence of hAK1 by creating stable hAK1-statin complexes with the binding constants of the order of 104 M−1. The enzyme kinetic studies revealed that statins inhibited hAK1 with significantly different efficiencies, in a noncompetitive manner. Simvastatin inhibited hAK1 with the highest yield comparable to that reported for diadenosine pentaphosphate, the only known hAK1 inhibitor. The determined AK sensitivity to statins differed markedly between short and long type AKs, suggesting an essential role of the LID domain in the AK inhibition. Our studies might open new horizons for the development of new modulators of short type AKs.
Collapse
Affiliation(s)
- Magdalena Wujak
- Faculty of Pharmacy, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, Jurasza 2, 85-089 Bydgoszcz, Poland;
- Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Lwowska 1, 87-100 Toruń, Poland;
| | - Anna Kozakiewicz
- Faculty of Chemistry, Nicolaus Copernicus University in Toruń, Gagarina 7, 87-100 Toruń, Poland; (M.B.); (Z.S.); (A.W.)
- Correspondence: ; Tel.: +48-56-611-4511
| | - Anna Ciarkowska
- Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Lwowska 1, 87-100 Toruń, Poland;
| | - Joanna I. Loch
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Kraków, Poland;
| | - Magdalena Barwiolek
- Faculty of Chemistry, Nicolaus Copernicus University in Toruń, Gagarina 7, 87-100 Toruń, Poland; (M.B.); (Z.S.); (A.W.)
| | - Zuzanna Sokolowska
- Faculty of Chemistry, Nicolaus Copernicus University in Toruń, Gagarina 7, 87-100 Toruń, Poland; (M.B.); (Z.S.); (A.W.)
| | - Marcin Budny
- Synthex Technologies Sp. z o.o., Gagarina 7/134B, 87-100 Toruń, Poland;
| | - Andrzej Wojtczak
- Faculty of Chemistry, Nicolaus Copernicus University in Toruń, Gagarina 7, 87-100 Toruń, Poland; (M.B.); (Z.S.); (A.W.)
| |
Collapse
|
18
|
Suazo KF, Jeong A, Ahmadi M, Brown C, Qu W, Li L, Distefano MD. Metabolic labeling with an alkyne probe reveals similarities and differences in the prenylomes of several brain-derived cell lines and primary cells. Sci Rep 2021; 11:4367. [PMID: 33623102 PMCID: PMC7902609 DOI: 10.1038/s41598-021-83666-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 02/03/2021] [Indexed: 01/31/2023] Open
Abstract
Protein prenylation involves the attachment of one or two isoprenoid group(s) onto cysteine residues positioned near the C-terminus. This modification is essential for many signal transduction processes. In this work, the use of the probe C15AlkOPP for metabolic labeling and identification of prenylated proteins in a variety of cell lines and primary cells is explored. Using a single isoprenoid analogue, 78 prenylated protein groups from the three classes of prenylation substrates were identified including three novel prenylation substrates in a single experiment. Applying this method to three brain-related cell lines including neurons, microglia, and astrocytes showed substantial overlap (25%) in the prenylated proteins identified. In addition, some unique prenylated proteins were identified in each type. Eight proteins were observed exclusively in neurons, five were observed exclusively in astrocytes and three were observed exclusively in microglia, suggesting their unique roles in these cells. Furthermore, inhibition of farnesylation in primary astrocytes revealed the differential responses of farnesylated proteins to an FTI. Importantly, these results provide a list of 19 prenylated proteins common to all the cell lines studied here that can be monitored using the C15AlkOPP probe as well as a number of proteins that were observed in only certain cell lines. Taken together, these results suggest that this chemical proteomic approach should be useful in monitoring the levels and exploring the underlying role(s) of prenylated proteins in various diseases.
Collapse
Affiliation(s)
- Kiall F Suazo
- Department of Chemistry, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Angela Jeong
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Mina Ahmadi
- Department of Chemistry, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Caroline Brown
- Department of Chemistry, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Wenhui Qu
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Ling Li
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Mark D Distefano
- Department of Chemistry, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
19
|
Juarez D, Fruman DA. Targeting the Mevalonate Pathway in Cancer. Trends Cancer 2021; 7:525-540. [PMID: 33358111 DOI: 10.1016/j.trecan.2020.11.008] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/21/2020] [Accepted: 11/23/2020] [Indexed: 02/06/2023]
Abstract
The mevalonate synthesis inhibitors, statins, are mainstay therapeutics for cholesterol management and cardiovascular health. Thirty years of research have uncovered supportive roles for the mevalonate pathway in numerous cellular processes that support oncogenesis, most recently macropinocytosis. Central to the diverse mechanisms of statin sensitivity is an acquired dependence on one mevalonate pathway output, protein geranylgeranylation. New chemical prenylation probes and the discovery of a novel geranylgeranyl transferase hold promise to deepen our understanding of statin mechanisms of action. Further, insights into statin selection and the counterproductive role of dietary geranylgeraniol highlight how we should assess statins in the clinic. Lastly, rational combination strategies preview how statins will enter the oncology toolbox.
Collapse
Affiliation(s)
- Dennis Juarez
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, USA
| | - David A Fruman
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
20
|
Anti-inflammatory Effects of Statins in Lung Vascular Pathology: From Basic Science to Clinical Trials. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1303:33-56. [PMID: 33788186 DOI: 10.1007/978-3-030-63046-1_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
HMG-CoA reductase inhibitors (or statins) are cholesterol-lowering drugs and are among the most widely prescribed medications in the United States. Statins exhibit pleiotropic effects that extend beyond cholesterol reduction including anti-atherosclerotic, antiproliferative, anti-inflammatory, and antithrombotic effects. Over the last 20 years, statins have been studied and examined in pulmonary vascular disorders, including both chronic pulmonary vascular disease such as pulmonary hypertension, and acute pulmonary vascular endothelial injury such as acute lung injury. In both research and clinical settings, statins have demonstrated promising vascular protection through modulation of the endothelium, attenuation of vascular leak, and promotion of endothelial repair following lung inflammation. This chapter provides a summary of the rapidly changing literature, summarizes the anti-inflammatory mechanism of statins on pulmonary vascular disorders, and explores clinical evidence for statins as a potential therapeutic approach to modulation of the endothelium as well as a means to broaden our understanding of pulmonary vasculopathy pathophysiology.
Collapse
|
21
|
MOHAMMADIAN-HAFSHEJANI ABDOLLAH, SHERWIN CATHERINEM, HEIDARI-SOURESHJANI SAEID. Do statins play any role in reducing the incidence and mortality of ovarian cancer? A systematic review and meta-analysis. JOURNAL OF PREVENTIVE MEDICINE AND HYGIENE 2020; 61:E331-E339. [PMID: 33150223 PMCID: PMC7595068 DOI: 10.15167/2421-4248/jpmh2020.61.3.1497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 05/18/2020] [Indexed: 01/15/2023]
Abstract
Introduction This systematic review and meta-analysis aimed to investigate the relationship between statin consumption and risk of incidence of ovarian cancer (OC) and associated mortality. Methods Computerized searches were conducted in three electronic databases (PubMed, Web of Science, and Scopus). Two calibrated authors performed the publications selection, data extraction, and quality assessment of the selected publications. The quality of the included articles was evaluated using the Newcastle-Ottawa Scale (NOS) for observational studies, and Jadad criteria for randomized clinical trials (RCTs). The electronic searches retrieved 2272 titles/abstracts. After the deletion of duplicate publications, 2030 titles/abstracts were assessed. Eighteen articles were included. Results Meta-analysis demonstrated that risk ratio (RR) of the association between statin consumption and OC incidence was 0.88 (95% CI = 0.75-1.03, P = 0.109). Patients receiving statin were less likely to die than those who did not receive statin, with a statistically significant association [RR = 0.76 (95% CI 0.67-0.86, P = 0.0001)]. There was no evidence of publication bias in examining the association between statin consumption and the risk of incidence and mortality from OC. Conclusions This study determined that statin use reduced the incidence risk of OC and significantly increased the survival in OC patients.
Collapse
Affiliation(s)
- ABDOLLAH MOHAMMADIAN-HAFSHEJANI
- Department of Epidemiology and Biostatistics, School of Public Health, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Pediatric Clinical Pharmacology, Department of Pediatrics, Wright State University Boonshoft School of Medicine, Dayton Children’s Hospital, One Children’s Plaza, Dayton, Ohio, USA
| | - CATHERINE M.T. SHERWIN
- Pediatric Clinical Pharmacology, Department of Pediatrics, Wright State University Boonshoft School of Medicine, Dayton Children’s Hospital, One Children’s Plaza, Dayton, Ohio, USA
| | - SAEID HEIDARI-SOURESHJANI
- Shahrekord University of Medical Sciences, Shahrekord, Iran
- Correspondence: Saeid Heidari-Soureshjani, Circuit of Research and Technology, Shahrekord University of Medical Sciences, Shahrekord, Iran - Tel. +98 9131833509 - Fax: +98 383351031 - E-mail:
| |
Collapse
|
22
|
Feltrin S, Ravera F, Traversone N, Ferrando L, Bedognetti D, Ballestrero A, Zoppoli G. Sterol synthesis pathway inhibition as a target for cancer treatment. Cancer Lett 2020; 493:19-30. [DOI: 10.1016/j.canlet.2020.07.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 07/05/2020] [Accepted: 07/09/2020] [Indexed: 12/21/2022]
|
23
|
Hao F, Xu Q, Wang J, Yu S, Chang HH, Sinnett-Smith J, Eibl G, Rozengurt E. Lipophilic statins inhibit YAP nuclear localization, co-activator activity and colony formation in pancreatic cancer cells and prevent the initial stages of pancreatic ductal adenocarcinoma in KrasG12D mice. PLoS One 2019; 14:e0216603. [PMID: 31100067 PMCID: PMC6524808 DOI: 10.1371/journal.pone.0216603] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 04/24/2019] [Indexed: 01/06/2023] Open
Abstract
We examined the impact of statins on Yes-associated Protein (YAP) localization, phosphorylation and transcriptional activity in human and mouse pancreatic ductal adenocarcinoma (PDAC) cells. Exposure of sparse cultures of PANC-1 and MiaPaCa-2 cells to cerivastatin or simvastatin induced a striking re-localization of YAP from the nucleus to the cytoplasm and inhibited the expression of the YAP/TEAD-regulated genes Connective Tissue Growth Factor (CTGF) and Cysteine-rich angiogenic inducer 61 (CYR61). Statins also prevented YAP nuclear import and expression of CTGF and CYR61 stimulated by the mitogenic combination of insulin and neurotensin in dense culture of these PDAC cells. Cerivastatin, simvastatin, atorvastatin and fluvastatin also inhibited colony formation by PANC-1 and MiaPaCa-2 cells in a dose-dependent manner. In contrast, the hydrophilic statin pravastatin did not exert any inhibitory effect even at a high concentration (10 μM). Mechanistically, cerivastatin did not alter the phosphorylation of YAP at Ser127 in either PANC-1 or MiaPaCa-2 cells incubated without or with neurotensin and insulin but blunted the assembly of actin stress fiber in these cells. We extended these findings with human PDAC cells using primary KC and KPC cells, (expressing KrasG12D or both KrasG12D and mutant p53, respectively) isolated from KC or KPC mice. Using cultures of these murine cells, we show that lipophilic statins induced striking YAP translocation from the nucleus to the cytoplasm, inhibited the expression of Ctgf, Cyr61 and Birc5 and profoundly inhibited colony formation of these cells. Administration of simvastatin to KC mice subjected to diet-induced obesity prevented early pancreatic acini depletion and PanIN formation. Collectively, our results show that lipophilic statins restrain YAP activity and proliferation in pancreatic cancer cell models in vitro and attenuates early lesions leading to PDAC in vivo.
Collapse
Affiliation(s)
- Fang Hao
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- Tianjin Medical University, Tianjin, China
| | - Qinhong Xu
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- Xi'an Jiaotong University, Xi'an, China
| | - Jing Wang
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- Xi'an Jiaotong University, Xi'an, China
| | - Shuo Yu
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- Xi'an Jiaotong University, Xi'an, China
| | - Hui-Hua Chang
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- CURE: Digestive Diseases Research Center, Los Angeles, California, United States of America
| | - James Sinnett-Smith
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- CURE: Digestive Diseases Research Center, Los Angeles, California, United States of America
- VA Greater Los Angeles Health Care System, Los Angeles, California, United States of America
| | - Guido Eibl
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- CURE: Digestive Diseases Research Center, Los Angeles, California, United States of America
| | - Enrique Rozengurt
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- CURE: Digestive Diseases Research Center, Los Angeles, California, United States of America
- VA Greater Los Angeles Health Care System, Los Angeles, California, United States of America
| |
Collapse
|
24
|
Heijnen NFL, Bergmans DCJJ, Schnabel RM, Bos LDJ. Targeted treatment of acute respiratory distress syndrome with statins-a commentary on two phenotype stratified re-analysis of randomized controlled trials. J Thorac Dis 2019; 11:S296-S299. [PMID: 30997202 DOI: 10.21037/jtd.2019.01.23] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Nanon F L Heijnen
- Department of Intensive Care, Maastricht UMC+, Maastricht, The Netherlands
| | | | - Ronny M Schnabel
- Department of Intensive Care, Maastricht UMC+, Maastricht, The Netherlands
| | - Lieuwe D J Bos
- Department of Intensive Care, UMC Amsterdam, Location AMC, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Abstract
Epidemiologic studies have, variably, shown the concomitant use of statin drugs to be beneficial to cancer outcomes. Statin drugs have been FDA approved for three decades for the treatment of high cholesterol and atherosclerotic coronary artery disease and are widely used. This has engendered studies as to their influence on concomitant diseases, including cancers. In this context, statin use has been correlated, variably, with a decrease in deaths from breast cancer. However, there is no extant model for this effect, and the extent of efficacy is open to question.The overarching goal of this article is to communicate to the reader of the potential of statins to reduce breast cancer progression and mortality. This is the use as a secondary prevention measure, and not as a therapy to directly counter active cancer. First, salient aspects of statin pharmacology, as relates to cardiovascular disease, will be discussed. Second, the basic and clinical research studies that investigate statin usage in breast cancer will be presented. Additionally, statin effects in other cancer types will be included for context. Finally, proposals for future basic and clinical research studies to determine the role of statins in breast cancer management will be presented.
Collapse
Affiliation(s)
- Colin H. Beckwitt
- Department of Pathology, University of Pittsburgh, Pittsburgh, 15231 PA USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15231 USA
- Pittsburgh VA Health System, Pittsburgh, 15240 PA USA
| | - Adam Brufsky
- Magee-Women’s Hospital of Pittsburgh, 300 Halket St., Pittsburgh, 15213 PA USA
| | - Zoltán N. Oltvai
- Department of Pathology, University of Pittsburgh, Pittsburgh, 15231 PA USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, 15231 PA USA
| | - Alan Wells
- Department of Pathology, University of Pittsburgh, Pittsburgh, 15231 PA USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, 15231 PA USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, 15231 PA USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15231 USA
- Pittsburgh VA Health System, Pittsburgh, 15240 PA USA
- Magee-Women’s Hospital of Pittsburgh, 300 Halket St., Pittsburgh, 15213 PA USA
| |
Collapse
|
26
|
Camacho L, Zabala-Letona A, Carracedo A. Re-evaluating statin activity in cancer. Aging (Albany NY) 2018; 10:1538-1539. [PMID: 30036187 PMCID: PMC6075430 DOI: 10.18632/aging.101504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 07/21/2018] [Indexed: 11/25/2022]
Affiliation(s)
- Laura Camacho
- CIC bioGUNE, Bizkaia Technology Park, Derio, Spain.,Biochemistry and Molecular Biology Department, University of the Basque Country, Bilbao, Spain
| | | | - Arkaitz Carracedo
- CIC bioGUNE, Bizkaia Technology Park, Derio, Spain.,Biochemistry and Molecular Biology Department, University of the Basque Country, Bilbao, Spain.,CIBERONC, Madrid, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
27
|
Abdullah MI, de Wolf E, Jawad MJ, Richardson A. The poor design of clinical trials of statins in oncology may explain their failure - Lessons for drug repurposing. Cancer Treat Rev 2018; 69:84-89. [PMID: 29936313 DOI: 10.1016/j.ctrv.2018.06.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 06/14/2018] [Accepted: 06/15/2018] [Indexed: 01/27/2023]
Abstract
Statins are widely used to treat hypercholesterolaemia. However, by inhibiting the production of mevalonate, they also reduce the production of several isoprenoids that are necessary for the function of small GTPase oncogenes such as Ras. As such, statins offer an attractive way to inhibit an "undruggable" target, suggesting that they may be usefully repurposed to treat cancer. However, despite numerous studies, there is still no consensus whether statins are useful in the oncology arena. Numerous preclinical studies have provided evidence justifying the evaluation of statins in cancer patients. Some retrospective studies of patients taking statins to control cholesterol have identified a reduced risk of cancer mortality. However, prospective clinical studies have mostly not been successful. We believe that this has occurred because many of the prospective clinical trials have been poorly designed. Many of these trials have failed to take into account some or all of the factors identified in preclinical studies that are likely to be necessary for statins to be efficacious. We suggest an improved trial design which takes these factors into account. Importantly, we suggest that the design of clinical trials of drugs which are being considered for repurposing should not assume it is appropriate to use them in the same way as they are used in their original indication. Rather, such trials deserve to be informed by preclinical studies that are comparable to those for any novel drug.
Collapse
Affiliation(s)
- Marwan I Abdullah
- Institute for Science and Technology in Medicine, Guy Hilton Research Centre, Keele University, Thornburrow Drive, Stoke-on-Trent ST4 7QB, United Kingdom
| | - Elizabeth de Wolf
- Institute for Science and Technology in Medicine, Guy Hilton Research Centre, Keele University, Thornburrow Drive, Stoke-on-Trent ST4 7QB, United Kingdom
| | - Mohammed J Jawad
- Institute for Science and Technology in Medicine, Guy Hilton Research Centre, Keele University, Thornburrow Drive, Stoke-on-Trent ST4 7QB, United Kingdom
| | - Alan Richardson
- Institute for Science and Technology in Medicine, Guy Hilton Research Centre, Keele University, Thornburrow Drive, Stoke-on-Trent ST4 7QB, United Kingdom; School of Pharmacy, Guy Hilton Research Centre, Keele University, Thornburrow Drive, Stoke-on-Trent ST4 7QB, United Kingdom.
| |
Collapse
|
28
|
A transcriptomic analysis of turmeric: Curcumin represses the expression of cholesterol biosynthetic genes and synergizes with simvastatin. Pharmacol Res 2018; 132:176-187. [DOI: 10.1016/j.phrs.2018.01.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 01/24/2018] [Accepted: 01/29/2018] [Indexed: 02/08/2023]
|
29
|
Ye X, Zhang G, Righolt C, Johnston JB, Banerji V, Gibson SB, Mahmud SM. Associations between statin use and risk of non-Hodgkin lymphomas by subtype. Int J Cancer 2018. [PMID: 29524215 DOI: 10.1002/ijc.31373] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Non-Hodgkin lymphomas (NHL) are a group of cancers with highly heterogeneous biology and clinical features. Statins are increasingly prescribed to prevent cardiovascular diseases. Early evidence shows a preventive effect of statins for some cancers, but their effect on NHL risk is unclear. We conducted a population-based nested case-control study involving 5,541 NHL cases and 27,315 controls matched for gender, age, place of residence and length of period of available prescription drug data. We assessed the use of statins prior to diagnosis (excluding the 12 months prior to the index date). We used conditional logistic regression models to estimate odds ratio (OR) and 95% confidence interval (CI) for use of any statin, adjusting for medical conditions, number of family physician visits for 5 years prior to index date, healthcare utilization, income and use of other medications. Over one-quarter of cases and controls were prescribed statins. Ever-use of any statin was associated with lower risk of Total NHL (OR = 0.82, 95% CI 0.76-0.89) and of certain subtypes including diffuse large B-cell lymphomas (DLBCL, OR = 0.77, 95% CI 0.65-0.92), plasma cell neoplasms (PCN, OR = 0.76, 95% CI 0.63-0.91) and other B-cell NHL (0.75, 0.59-0.95). Analysis by statin type suggested that the association was limited to high potency statin and lipophilic statin users. No clear duration or dose-response relationships were observed. Our findings provide evidence that statin use can reduce the risk of DLBCL and plasma cell lymphomas, but not other NHL types. Further studies are warranted to verify these associations and to examine the biological mechanisms.
Collapse
Affiliation(s)
- Xibiao Ye
- Department of Community Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Vaccine and Drug Evaluation Centre, University of Manitoba, Winnipeg, MB, Canada
| | - Geng Zhang
- Department of Community Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Vaccine and Drug Evaluation Centre, University of Manitoba, Winnipeg, MB, Canada
| | - Christiaan Righolt
- Department of Community Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Vaccine and Drug Evaluation Centre, University of Manitoba, Winnipeg, MB, Canada
| | - James B Johnston
- Research Institute of Oncology and Hematology, Cancer Care Manitoba, Winnipeg, MB, Canada
- Department of Internal Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Versha Banerji
- Research Institute of Oncology and Hematology, Cancer Care Manitoba, Winnipeg, MB, Canada
- Department of Internal Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Spencer B Gibson
- Research Institute of Oncology and Hematology, Cancer Care Manitoba, Winnipeg, MB, Canada
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada
| | - Salaheddin M Mahmud
- Department of Community Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Vaccine and Drug Evaluation Centre, University of Manitoba, Winnipeg, MB, Canada
- College of Pharmacy, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
30
|
Mira E, Carmona-Rodríguez L, Pérez-Villamil B, Casas J, Fernández-Aceñero MJ, Martínez-Rey D, Martín-González P, Heras-Murillo I, Paz-Cabezas M, Tardáguila M, Oury TD, Martín-Puig S, Lacalle RA, Fabriás G, Díaz-Rubio E, Mañes S. SOD3 improves the tumor response to chemotherapy by stabilizing endothelial HIF-2α. Nat Commun 2018; 9:575. [PMID: 29422508 PMCID: PMC5805714 DOI: 10.1038/s41467-018-03079-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 01/18/2018] [Indexed: 02/08/2023] Open
Abstract
One drawback of chemotherapy is poor drug delivery to tumor cells, due in part to hyperpermeability of the tumor vasculature. Extracellular superoxide dismutase (SOD3) is an antioxidant enzyme usually repressed in the tumor milieu. Here we show that specific SOD3 re-expression in tumor-associated endothelial cells (ECs) increases doxorubicin (Doxo) delivery into and chemotherapeutic effect on tumors. Enhanced SOD3 activity fostered perivascular nitric oxide accumulation and reduced vessel leakage by inducing vascular endothelial cadherin (VEC) transcription. SOD3 reduced HIF prolyl hydroxylase domain protein activity, which increased hypoxia-inducible factor-2α (HIF-2α) stability and enhanced its binding to a specific VEC promoter region. EC-specific HIF-2α ablation prevented both the SOD3-mediated increase in VEC transcription and the enhanced Doxo effect. SOD3, VEC, and HIF-2α levels correlated positively in primary colorectal cancers, which suggests a similar interconnection of these proteins in human malignancy.
Collapse
Affiliation(s)
- Emilia Mira
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, Madrid, 28049, Spain
| | - Lorena Carmona-Rodríguez
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, Madrid, 28049, Spain
| | - Beatriz Pérez-Villamil
- Genomics and Microarray Laboratory, Medical Oncology & Surgical Pathology Departments, Instituto de Investigación Sanitaria San Carlos Hospital Clínico San Carlos, Univ. Complutense de Madrid, CIBERONC, Profesor Martín Lagos, S/N, Madrid, 28040, Spain
| | - Josefina Casas
- Department of Biomedicinal Chemistry, Institute of Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18-26, Barcelona, 08034, Spain
| | - María Jesús Fernández-Aceñero
- Genomics and Microarray Laboratory, Medical Oncology & Surgical Pathology Departments, Instituto de Investigación Sanitaria San Carlos Hospital Clínico San Carlos, Univ. Complutense de Madrid, CIBERONC, Profesor Martín Lagos, S/N, Madrid, 28040, Spain
| | - Diego Martínez-Rey
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, Madrid, 28049, Spain
| | - Paula Martín-González
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, Madrid, 28049, Spain
| | - Ignacio Heras-Murillo
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, Madrid, 28049, Spain
| | - Mateo Paz-Cabezas
- Genomics and Microarray Laboratory, Medical Oncology & Surgical Pathology Departments, Instituto de Investigación Sanitaria San Carlos Hospital Clínico San Carlos, Univ. Complutense de Madrid, CIBERONC, Profesor Martín Lagos, S/N, Madrid, 28040, Spain
| | - Manuel Tardáguila
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, Madrid, 28049, Spain
- Genetics Institute, University of Florida, 2033 Mowry Road, Gainesville, FL, 32610, USA
| | - Tim D Oury
- Department of Pathology, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA, 15261, USA
| | - Silvia Martín-Puig
- Myocardial Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares, Calle de Melchor Fernández Almagro, 3, Madrid, 28029, Spain
| | - Rosa Ana Lacalle
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, Madrid, 28049, Spain
| | - Gemma Fabriás
- Department of Biomedicinal Chemistry, Institute of Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18-26, Barcelona, 08034, Spain
| | - Eduardo Díaz-Rubio
- Genomics and Microarray Laboratory, Medical Oncology & Surgical Pathology Departments, Instituto de Investigación Sanitaria San Carlos Hospital Clínico San Carlos, Univ. Complutense de Madrid, CIBERONC, Profesor Martín Lagos, S/N, Madrid, 28040, Spain
| | - Santos Mañes
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, Madrid, 28049, Spain.
| |
Collapse
|
31
|
Tanaka M, Yasuoka A, Yoshinuma H, Saito Y, Asakura T, Tanabe S. Seasoning ingredients in a medium-fat diet regulate lipid metabolism in peripheral tissues via the hypothalamic-pituitary axis in growing rats. Biosci Biotechnol Biochem 2018; 82:497-506. [PMID: 29370734 DOI: 10.1080/09168451.2018.1427551] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
We fed rats noodle (N) -diet containing 30 wt.% instant noodle with a 26% fat-to-energy ratio for 30 days (N-group). Compared with rats that were fed the same amount of nutrients (C-group), the N-group showed lower liver triacylglycerol levels and higher fecal cholesterol levels. We then analyzed transcriptome of the hypothalamic-pituitary (HP), the liver and the white adipose tissue (WAT). Thyroid stimulating hormone (Tshb), and its partner, glycoprotein hormone genes were up-regulated in the HP of N-group. Sterol regulatory element binding transcription factors were activated in the liver of N-group, while an up-regulation of the angiogenic signal occurred in the WAT of N-group. N-group showed higher urine noradrenaline (NA) level suggesting that these tissue signals are regulated by NA and Tshb. The N-diet contains 0.326 wt.% glutamate, 0.00236 wt.% 6-shogaol and Maillard reaction products. Our results suggest that these ingredients may affect lipid homeostasis via the HP axis.
Collapse
Key Words
- AM: adrenal medulla
- ANGPT 2: angiopoietin 2
- CNS: central nervous system
- Cga: glycoprotein hormones alpha polypeptide
- DEGs: differentially expressed genes
- DHBA: 3,4-dihydroxybenzylamine
- FDR: false discovery rate
- HP: hypothalamic-pituitary
- HPLC: High Performance Liquid Chromatography
- IPA: Ingenuity Pathway Analysis
- NA: noradrenaline
- NCBI: National Center of Biotechnology Information
- SN: sympathetic nerve
- SREBF: sterol regulatory element binding transcription factor
- TG: triacylglycerol
- TH: thyroid hormone
- TRPs: transient receptor potential channels
- The designation of following abbreviations, Acsm5, Avp, Ch25h, CREB, Cyp51a1, Dhcr7, ERBB4, F2R, Gpd1, Hdc, Hmgcs, Maob, NEDD9, NFkB, SMARCB1, SPDEF, SPI1, STAT4, TGFBR1, Vip, WNT3A, and XBP1, are shown in Table 5 and 6.
- Trh: Thyrotropin releasing hormone
- Tshb: thyroid stimulating hormone B
- WAT: white adipose tissue
- hypothalamus
- instant noodle
- lipid metabolism
- noradrenaline
- seasoning ingredients
Collapse
Affiliation(s)
- Mitsuru Tanaka
- a Nissin Global Innovation Center , Nissin Foods Holdings , Hachioji , Japan
| | - Akihito Yasuoka
- b Project on Health and Anti-Aging, Kanagawa Academy of Science and Technology , Kawasaki , Japan
| | - Haruka Yoshinuma
- a Nissin Global Innovation Center , Nissin Foods Holdings , Hachioji , Japan
| | - Yoshikazu Saito
- c Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences , The University of Tokyo , Tokyo , Japan
| | - Tomiko Asakura
- c Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences , The University of Tokyo , Tokyo , Japan
| | - Soichi Tanabe
- a Nissin Global Innovation Center , Nissin Foods Holdings , Hachioji , Japan
| |
Collapse
|
32
|
Targeting of stress response pathways in the prevention and treatment of cancer. Biotechnol Adv 2018; 36:583-602. [PMID: 29339119 DOI: 10.1016/j.biotechadv.2018.01.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 01/08/2018] [Accepted: 01/10/2018] [Indexed: 12/12/2022]
Abstract
The hallmarks of tumor tissue are not only genetic aberrations but also the presence of metabolic and oxidative stress as a result of hypoxia and lactic acidosis. The stress activates several prosurvival pathways including metabolic remodeling, autophagy, antioxidant response, mitohormesis, and glutaminolysis, whose upregulation in tumors is associated with a poor survival of patients, while their activation in healthy tissue with statins, metformin, physical activity, and natural compounds prevents carcinogenesis. This review emphasizes the dual role of stress response pathways in cancer and suggests the integrative understanding as a basis for the development of rational therapy targeting the stress response.
Collapse
|
33
|
Gbelcová H, Rimpelová S, Knejzlík Z, Šáchová J, Kolář M, Strnad H, Repiská V, D'Acunto WC, Ruml T, Vítek L. Isoprenoids responsible for protein prenylation modulate the biological effects of statins on pancreatic cancer cells. Lipids Health Dis 2017; 16:250. [PMID: 29262834 PMCID: PMC5738693 DOI: 10.1186/s12944-017-0641-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 12/05/2017] [Indexed: 01/08/2023] Open
Abstract
Background Statin treatment of hypercholesterolemia is accompanied also with depletion of the mevalonate intermediates, including farnesyl pyrophosphate (FPP) and geranylgeranyl pyrophosphate (GGPP) necessary for proper function of small GTPases. These include Ras proteins, prevalently mutated in pancreatic cancer. In our study, we evaluated the effect of three key intermediates of the mevalonate pathway on GFP-K-Ras protein localization and the gene expression profile in pancreatic cancer cells after exposure to individual statins. Methods These effects were tested on MiaPaCa-2 human pancreatic cancer cells carrying a K-Ras activating mutation (G12C) after exposure to individual statins (20 μM). The effect of statins (atorvastatin, lovastatin, simvastatin, fluvastatin, cerivastatin, rosuvastatin, and pitavastatin) and mevalonate intermediates on GFP-K-Ras protein translocation was analyzed using fluorescence microscopy. The changes in gene expression induced in MiaPaCa-2 cells treated with simvastatin, FPP, GGPP, and their combinations with simvastatin were examined by whole genome DNA microarray analysis. Results All tested statins efficiently inhibited K-Ras protein trafficking from cytoplasm to the cell membrane of the MiaPaCa-2 cells. The inhibitory effect of statins on GFP-K-Ras protein trafficking was partially prevented by addition of any of the mevalonate pathway’s intermediates tested. Expressions of genes involved in metabolic and signaling pathways modulated by simvastatin treatment was normalized by the concurrent addition of FPP or GGPP. K-Ras protein trafficking within the pancreatic cancer cells is effectively inhibited by the majority of statins; the inhibition is eliminated by isoprenoid intermediates of the mevalonate pathway. Conclusions Our data indicate that the anticancer effects of statins observed in numerous studies to a large extent are mediated through isoprenoid intermediates of the mevalonate pathway, as they influence expression of genes involved in multiple intracellular pathways. Electronic supplementary material The online version of this article (10.1186/s12944-017-0641-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Helena Gbelcová
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Bratislava, Slovakia.,Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, Czech Republic
| | - Silvie Rimpelová
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, Czech Republic
| | - Zdeněk Knejzlík
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, Czech Republic
| | - Jana Šáchová
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Michal Kolář
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Hynek Strnad
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Vanda Repiská
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Walter Cosimo D'Acunto
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, Czech Republic
| | - Tomáš Ruml
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, Czech Republic.
| | - Libor Vítek
- Institute of Medical Biochemistry and Laboratory Diagnostics, and 4th Department of Internal Medicine, 1st Faculty of Medicine, Charles University, Prague, Czech Republic.
| |
Collapse
|
34
|
Dietary geranylgeraniol can limit the activity of pitavastatin as a potential treatment for drug-resistant ovarian cancer. Sci Rep 2017; 7:5410. [PMID: 28710496 PMCID: PMC5511264 DOI: 10.1038/s41598-017-05595-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 06/05/2017] [Indexed: 12/14/2022] Open
Abstract
Pre-clinical and retrospective studies of patients using statins to reduce plasma cholesterol have suggested that statins may be useful to treat cancer. However, prospective clinical trials have yet to demonstrate significant efficacy. We have previously shown that this is in part because a hydrophobic statin with a long half-life is necessary. Pitavastatin, the only statin with this profile, has not undergone clinical evaluation in oncology. The target of pitavastatin, hydroxymethylglutarate coenzyme-A reductase (HMGCR), was found to be over-expressed in all ovarian cancer cell lines examined and upregulated by mutated TP53, a gene commonly altered in ovarian cancer. Pitavastatin-induced apoptosis was blocked by geranylgeraniol and mevalonate, products of the HMGCR pathway, confirming that pitavastatin causes cell death through inhibition of HMGCR. Solvent extracts of human and mouse food were also able to block pitavastatin-induced apoptosis, suggesting diet might influence the outcome of clinical trials. When nude mice were maintained on a diet lacking geranylgeraniol, oral pitavastatin caused regression of Ovcar-4 tumour xenografts. However, when the animal diet was supplemented with geranylgeraniol, pitavastatin failed to prevent tumour growth. This suggests that a diet containing geranylgeraniol can limit the anti-tumour activity of pitavastatin and diet should be controlled in clinical trials of statins.
Collapse
|