1
|
Chang YC, Yu MH, Huang HP, Chen DH, Yang MY, Wang CJ. Mulberry leaf extract inhibits obesity and protects against diethylnitrosamine-induced hepatocellular carcinoma in rats. J Tradit Complement Med 2024; 14:266-275. [PMID: 38707917 PMCID: PMC11068992 DOI: 10.1016/j.jtcme.2024.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 05/07/2024] Open
Abstract
Mulberry leaf has been recognized as a traditional Chinese medicinal plant, which was distributed throughout the Asia. The aqueous extract of mulberry leaf extract (MLE) has various biologically active components such as polyphenols and flavonoids. However, the inhibitory effect of MLE in hepatocarcinogenesis is poorly understood. In this study, we determined the role of MLE supplementation in preventing hepatocarcinogenesis in a carcinogen-initiated high-fat diet (HFD)-promoted Sprague-Dawley (SD) rat model. The rats were fed an HFD to induce obesity and spontaneous hepatomas by administering 0.01% diethylnitrosamine (DEN) in their drinking water for 12 weeks (HD group), and also to fed MLE through oral ingestion at daily doses of 0.5%, 1%, or 2%. At the end of the 12-week experimental period, the liver tumors were analyzed to identify markers of oxidative stress and antioxidant enzyme activities, and their serum was analyzed to determine their nutritional status and liver function. Histopathological analysis revealed that MLE supplementation significantly suppressed the severity and incidence of hepatic tumors. Furthermore, compared with the HFD + DEN groups, the expression of protein kinase C (PKC)-α and Rac family small GTPase 1 (Rac1) was lower in the MLE groups. These findings suggest that MLE prevents obesity-enhanced, carcinogen-induced hepatocellular carcinoma development, potentially through the protein kinase C (PKC)α/Rac1 signaling pathway. MLE might be an effective chemoprevention modality for nonalcoholic fatty liver disease (NAFLD)/nonalcoholic steatohepatitis (NASH)-related hepatocarcinogenesis.
Collapse
Affiliation(s)
- Yun-Ching Chang
- Department of Health Diet and Industry Management, Chung Shan Medical University, Taichung, 402, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, 402, Taiwan
| | - Meng-Hsun Yu
- Department of Nutrition, Chung Shan Medical University, Taichung, 402, Taiwan
- Department of Nutrition, Chung Shan Medical University Hospital, Taichung, 402, Taiwan
| | - Hui-Pei Huang
- Department of Biochemistry, School of Medicine, Medical College, Chung Shan Medical University, Taichung, 402, Taiwan
| | - Dong-Hui Chen
- Institute of Medicine, Chung Shan Medical University, Taichung, 402, Taiwan
| | - Mon-Yuan Yang
- Department of Health Diet and Industry Management, Chung Shan Medical University, Taichung, 402, Taiwan
| | - Chau-Jong Wang
- Department of Health Diet and Industry Management, Chung Shan Medical University, Taichung, 402, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, 402, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, 402, Taiwan
| |
Collapse
|
2
|
Desert R, Ge X, Song Z, Han H, Lantvit D, Chen W, Das S, Athavale D, Abraham-Enachescu I, Blajszczak C, Chen Y, Musso O, Guzman G, Hoshida Y, Nieto N. Role of Hepatocyte-Derived Osteopontin in Liver Carcinogenesis. Hepatol Commun 2021; 6:692-709. [PMID: 34730871 PMCID: PMC8948552 DOI: 10.1002/hep4.1845] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 09/13/2021] [Accepted: 10/02/2021] [Indexed: 12/24/2022] Open
Abstract
Osteopontin (OPN) expression correlates with tumor progression in many cancers, including hepatocellular carcinoma (HCC); however, its role in the onset of HCC remains unclear. We hypothesized that increased hepatocyte‐derived OPN is a driver of hepatocarcinogenesis. Analysis of a tissue microarray of 366 human samples revealed a continuous increase in OPN expression during hepatocarcinogenesis. In patients with cirrhosis, a transcriptome‐based OPN correlation network was associated with HCC incidence along 10 years of follow‐up, together with messenger RNA (mRNA) signatures of carcinogenesis. After diethylnitrosamine (DEN) injection, mice with conditional overexpression of Opn in hepatocytes (OpnHep transgenic [Tg]) showed increased tumor burden. Surprisingly, mice with conditional ablation of Opn in hepatocytes (OpnΔHep) expressed a similar phenotype. The acute response to DEN was reduced in OpnΔHep, which also showed more cancer stem/progenitor cells (CSCs, CD44+AFP+) at 5 months. CSCs from OpnHep Tg mice expressed several mRNA signatures known to promote carcinogenesis, and mRNA signatures from OpnHep Tg mice were associated with poor outcome in human HCC patients. Treatment with rOPN had little effect on CSCs, and their progression to HCC was similar in Opn−/− compared with wild‐type mice. Finally, ablation of Cd44, an OPN receptor, did not reduce tumor burden in Cd44−/−OpnHep Tg mice. Conclusions: Hepatocyte‐derived OPN acts as a tumor suppressor at physiological levels by controlling the acute response to DEN and the presence of CSCs, while induction of OPN is pro‐tumorigenic. This is primarily due to intracellular events rather that by the secretion of the protein and receptor activation.
Collapse
Affiliation(s)
- Romain Desert
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA
| | - Xiaodong Ge
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA.,Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zhuolun Song
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA
| | - Hui Han
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA
| | - Daniel Lantvit
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA
| | - Wei Chen
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA
| | - Sukanta Das
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA
| | - Dipti Athavale
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA
| | - Ioana Abraham-Enachescu
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chuck Blajszczak
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA
| | - Yu Chen
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA
| | - Orlando Musso
- INSERM, University of Rennes, INRA, Institut NuMeCAN (Nutrition Metabolisms and Cancer), Rennes, France
| | - Grace Guzman
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA
| | - Yujin Hoshida
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Liver Tumor Translational Research Program, Harold C. Simmons Comprehensive Cancer Center, Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Natalia Nieto
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA.,Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
3
|
Liu L, Liu B, Yu J, Zhang D, Shi J, Liang P. Development of a Toll-Like Receptor-Based Gene Signature That Can Predict Prognosis, Tumor Microenvironment, and Chemotherapy Response for Hepatocellular Carcinoma. Front Mol Biosci 2021; 8:729789. [PMID: 34621787 PMCID: PMC8490642 DOI: 10.3389/fmolb.2021.729789] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/16/2021] [Indexed: 11/19/2022] Open
Abstract
Objective: Emerging evidence highlights the implications of the toll-like receptor (TLR) signaling pathway in the pathogenesis and therapeutic regimens of hepatocellular carcinoma (HCC). Herein, a prognostic TLR-based gene signature was conducted for HCC. Methods: HCC-specific TLRs were screened in the TCGA cohort. A LASSO model was constructed based on prognosis-related HCC-specific TLRs. The predictive efficacy, sensitivity, and independency of this signature was then evaluated and externally verified in the ICGC, GSE14520, and GSE76427 cohorts. The associations between this signature and tumor microenvironment (stromal/immune score, immune checkpoint expression, and immune cell infiltrations) and chemotherapy response were assessed in HCC specimens. The expression of TLRs in this signature was verified in HCC and normal liver tissues by Western blot. Following si-MAP2K2 transfection, colony formation and apoptosis of Huh7 and HepG2 cells were examined. Results: Herein, we identified 60 HCC-specific TLRs. A TLR-based gene signature (MAP2K2, IRAK1, RAC1, TRAF3, MAP3K7, and SPP1) was conducted for HCC prognosis. High-risk patients exhibited undesirable outcomes. ROC curves confirmed the well prediction performance of this signature. Multivariate Cox regression analysis demonstrated that the signature was an independent prognostic indicator. Also, high-risk HCC was characterized by an increased immune score, immune checkpoint expression, and immune cell infiltration. Meanwhile, high-risk patients displayed higher sensitivity to gemcitabine and cisplatin. The dysregulation of TLRs in the signature was confirmed in HCC. MAP2K2 knockdown weakened colony formation and elevated apoptosis of Huh7 and HepG2 cells. Conclusion: Collectively, this TLR-based gene signature might assist clinicians to select personalized therapy programs for HCC patients.
Collapse
Affiliation(s)
- Lixia Liu
- Department of Interventional Ultrasound, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China.,Department of Ultrasound, Affiliated Hospital of Hebei University, Baoding, China
| | - Bin Liu
- Central Laboratory, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Affiliated Hospital of Hebei University, Institute of Life Science and Green Development, Hebei University, Baoding, China
| | - Jie Yu
- Department of Interventional Ultrasound, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Dongyun Zhang
- Department of Interventional Ultrasound, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Jianhong Shi
- Central Laboratory, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Affiliated Hospital of Hebei University, Institute of Life Science and Green Development, Hebei University, Baoding, China
| | - Ping Liang
- Department of Interventional Ultrasound, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| |
Collapse
|
4
|
Singh D, Yadav E, Kumar V, Verma A. Madhuca longifolia Embedded Silver Nanoparticles Attenuate Diethylnitrosamine (DEN)-Induced Renal Cancer via Regulating Oxidative Stress. Curr Drug Deliv 2021; 18:634-644. [PMID: 32914714 DOI: 10.2174/1567201817666200910154301] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 07/21/2020] [Accepted: 07/28/2020] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Madhuca longifolia has been used for the treatment of renal cancer. Therefore, the current study describes the protective effects of biofabricated silver nanoparticles (MLAg- NPs) using Madhuca longifolia aqueous leaves extract against diethylnitrosamine (DEN) induced Renal Cell Carcinoma (RCC) in rats. METHODS Animals were categorized into five groups and treated with doses of silver nanoparticles for 16 weeks. Antineoplastic effect in renal cancer was dose dependent to control the macroscopical variations when compared to DEN induced group. Significant changes were observed in biochemical parameters and dose graded improvement in the level of antioxidants parameters were accountable for its protective nature. RESULTS Silver nanoparticles in dose dependent manner was effective to modify the raised levels of pro-inflammatory cytokines and inflammatory mediators during renal cancer. Alteration in renal histopathology were also detected in the silver nanoparticles treated group, which show its safety concern. Biofabricated silver nanoparticles (MLAgNPs) using Madhuca longifolia can convey significant chemo-protective effect against renal cancer by suppressing the IL-6, TNF-α and IL-1β by nuclear factor-kappa B (NF-κB) pathway. CONCLUSION Our outcomes implicates that biofabricated MLAgNPs exhibited a chemoprotective potential in the prevention and intervention of RCC.
Collapse
Affiliation(s)
- Deepika Singh
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, Sam Higginbottom University of Agriculture, Technology & Sciences, Allahabad-211007, Uttar Pradesh, India
| | - Ekta Yadav
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, Sam Higginbottom University of Agriculture, Technology & Sciences, Allahabad-211007, Uttar Pradesh, India
| | - Vikas Kumar
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, Sam Higginbottom University of Agriculture, Technology & Sciences, Allahabad-211007, Uttar Pradesh, India
| | - Amita Verma
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, Sam Higginbottom University of Agriculture, Technology & Sciences, Allahabad-211007, Uttar Pradesh, India
| |
Collapse
|
5
|
Krupenko NI, Sharma J, Fogle HM, Pediaditakis P, Strickland KC, Du X, Helke KL, Sumner S, Krupenko SA. Knockout of Putative Tumor Suppressor Aldh1l1 in Mice Reprograms Metabolism to Accelerate Growth of Tumors in a Diethylnitrosamine (DEN) Model of Liver Carcinogenesis. Cancers (Basel) 2021; 13:cancers13133219. [PMID: 34203215 PMCID: PMC8268287 DOI: 10.3390/cancers13133219] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/15/2021] [Accepted: 06/22/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Cancers often loose the enzyme of folate metabolism ALDH1L1. We proposed that such loss is advantageous for the malignant tumor growth and tested this hypothesis in mice proficient or deficient (gene knockout) in ALDH1L1 expression. Liver cancer in both groups was induced by injection of chemical carcinogen diethylnitrosamine. While the number of tumors observed in ALDH1L1 proficient and deficient mice was similar, tumors grew faster and to a larger size in the knockout mice. We conclude that the ALDH1L1 loss promotes liver tumor growth without affecting tumor initiation or multiplicity. Accelerated growth of tumors lacking the enzyme was linked to several metabolic pathways, which are beneficial for rapid proliferation. Abstract Cytosolic 10-formyltetrahydrofolate dehydrogenase (ALDH1L1) is commonly downregulated in human cancers through promoter methylation. We proposed that ALDH1L1 loss promotes malignant tumor growth. Here, we investigated the effect of the Aldh1l1 mouse knockout (Aldh1l1−/−) on hepatocellular carcinoma using a chemical carcinogenesis model. Fifteen-day-old male Aldh1l1 knockout mice and their wild-type littermate controls (Aldh1l1+/+) were injected intraperitoneally with 20 μg/g body weight of DEN (diethylnitrosamine). Mice were sacrificed 10, 20, 28, and 36 weeks post-DEN injection, and livers were examined for tumor multiplicity and size. We observed that while tumor multiplicity did not differ between Aldh1l1−/− and Aldh1l1+/+ animals, larger tumors grew in Aldh1l1−/− compared to Aldh1l1+/+ mice at 28 and 36 weeks. Profound differences between Aldh1l1−/− and Aldh1l1+/+ mice in the expression of inflammation-related genes were seen at 10 and 20 weeks. Of note, large tumors from wild-type mice showed a strong decrease of ALDH1L1 protein at 36 weeks. Metabolomic analysis of liver tissues at 20 weeks showed stronger differences in Aldh1l1+/+ versus Aldh1l1−/− metabotypes than at 10 weeks, which underscores metabolic pathways that respond to DEN in an ALDH1L1-dependent manner. Our study indicates that Aldh1l1 knockout promoted liver tumor growth without affecting tumor initiation or multiplicity.
Collapse
Affiliation(s)
- Natalia I. Krupenko
- Department of Nutrition, University of North Carolina, Chapel Hill, NC 27599, USA; (N.I.K.); (S.S.)
- Nutrition Research Institute, University of North Carolina, Kannapolis, NC 28081, USA; (J.S.); (H.M.F.); (P.P.)
| | - Jaspreet Sharma
- Nutrition Research Institute, University of North Carolina, Kannapolis, NC 28081, USA; (J.S.); (H.M.F.); (P.P.)
| | - Halle M. Fogle
- Nutrition Research Institute, University of North Carolina, Kannapolis, NC 28081, USA; (J.S.); (H.M.F.); (P.P.)
| | - Peter Pediaditakis
- Nutrition Research Institute, University of North Carolina, Kannapolis, NC 28081, USA; (J.S.); (H.M.F.); (P.P.)
| | | | - Xiuxia Du
- Department of Bioinformatics & Genomics, UNC Charlotte, Charlotte, NC 28223, USA;
| | - Kristi L. Helke
- Department of Comparative Medicine, Medical University of South Carolina, Charleston, SC 29425, USA;
| | - Susan Sumner
- Department of Nutrition, University of North Carolina, Chapel Hill, NC 27599, USA; (N.I.K.); (S.S.)
- Nutrition Research Institute, University of North Carolina, Kannapolis, NC 28081, USA; (J.S.); (H.M.F.); (P.P.)
| | - Sergey A. Krupenko
- Department of Nutrition, University of North Carolina, Chapel Hill, NC 27599, USA; (N.I.K.); (S.S.)
- Nutrition Research Institute, University of North Carolina, Kannapolis, NC 28081, USA; (J.S.); (H.M.F.); (P.P.)
- Correspondence:
| |
Collapse
|
6
|
Ganapathy-Kanniappan S. Rac1 repression reverses chemoresistance by targeting tumor metabolism. Cancer Biol Ther 2020; 21:888-890. [PMID: 32866423 DOI: 10.1080/15384047.2020.1809923] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Tumor metabolism is exemplified by the increased rate of glucose utilization, a biochemical signature of cancer cells. The enhanced glucose hydrolysis enabled by the augmentation of glycolytic flux and the pentose phosphate pathway (PPP) plays a pivotal role in the growth and survival of neoplastic cells. In a recent report, it has been shown that in human breast cancer the GTP binding protein, Rac1 enables resistance to therapy, particularly against the DNA-damaging therapeutics. Significantly, the findings demonstrate that Rac1-dependent chemoresistance involves the upregulation of glycolytic flux as well as PPP. Using multiple approaches, the study demonstrates that disruption of Rac1 activity sensitizes cancer cells to DNA-damaging agents. More importantly, the data uncover a previously unknown PPP regulatory role of Rac1 in breast cancer. Finally, the authors also show the effectiveness and the feasibility of in vivo targeting of Rac1 to enhance the chemosensitivity of breast cancer. This elegant report provokes scientific curiosity to expand our understanding of the intricacies of the role and regulation of Rac1 in cancer.
Collapse
Affiliation(s)
- Shanmugasundaram Ganapathy-Kanniappan
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Interventional Radiology, The Johns Hopkins University School of Medicine , Baltimore, MD, USA
| |
Collapse
|
7
|
Park MK, Lee JC, Lee JW, Kang S, Kim J, Park MH, Hwang SJ, Lee M. Effects of fermented rice bran on DEN-induced oxidative stress in mice: GSTP1, LINE-1 methylation, and telomere length ratio. J Food Biochem 2020; 44:e13274. [PMID: 32468620 DOI: 10.1111/jfbc.13274] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/01/2020] [Accepted: 04/18/2020] [Indexed: 12/12/2022]
Abstract
N-diethylnitrosamine (DEN), a well-known carcinogen, not only induces excessive reactive oxygen species but also suppresses DNA methylation. This study investigated the effect of fermented rice bran (FRB) treatment on DEN-induced oxidative stress through DNA methylation and telomere length analysis. To evaluate the potential protective role of FRB in oxidative stress, two different doses of FRB, DEN, and their combination were administered to mice that were preadapted or not to FRB. Glutathione-S-transferase P1 (GSTP1) methylation levels significantly decreased at 2 and 24 hr after FRB and DEN co-administration in mice with and without pre-adaptation. Moreover, GSTP1 mRNA was upregulated under DEN-induced oxidative stress. Furthermore, changes in long interspersed nuclear element-1 methylation were observed from the viewpoint of genomic instability. In addition, FRB preadapted mice displayed a lower telomere length ratio than the non-adapted mice, suggesting that FRB adaptation offers advantages over the non-adapted conditions in terms of inflammation suppression. PRACTICAL APPLICATIONS: DEN induces excessive ROS, which is associated with oxidative stress on DNA and other cellular components, resulting in inflammation. This study shows that FRB may alleviate DEN-triggered oxidative stress, based on changes in GSTP1, LINE-1 methylation, and telomere length ratios, thereby, revealing the potential of dietary intervention during inflammation. Furthermore, this study furthers the current understanding of DNA methylation mechanisms underlying the antioxidant and anti-inflammatory effects of functional food components. These results indicate that dietary inclusion of FRB may help decrease oxidative DNA damage and its associated inflammation at early stages of a disease.
Collapse
Affiliation(s)
- Min-Koo Park
- Nutrigenetics Institute, Bio-Innovation Park, Erom, Inc., Uiwang, Republic of Korea
| | - Jeong-Chan Lee
- Nutrigenetics Institute, Bio-Innovation Park, Erom, Inc., Uiwang, Republic of Korea
| | - Ji-Won Lee
- Nutrigenetics Institute, Bio-Innovation Park, Erom, Inc., Uiwang, Republic of Korea
| | - Sujin Kang
- Bio R&D Division, Bio-Innovation Park, Erom, Inc., Chuncheon, Republic of Korea
| | - JoongHark Kim
- Bio R&D Division, Bio-Innovation Park, Erom, Inc., Chuncheon, Republic of Korea
| | - Mi Houn Park
- Bio R&D Division, Bio-Innovation Park, Erom, Inc., Chuncheon, Republic of Korea
| | - Sung-Joo Hwang
- Integrated Medicine Institute, Loving Care Hospital, Sampyeong, Republic of Korea
| | - MinJae Lee
- College of Veterinary Medicine, Kangwon National University, Chuncheon, Republic of Korea
| |
Collapse
|
8
|
Coordination between Rac1 and Rab Proteins: Functional Implications in Health and Disease. Cells 2019; 8:cells8050396. [PMID: 31035701 PMCID: PMC6562727 DOI: 10.3390/cells8050396] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 04/25/2019] [Accepted: 04/26/2019] [Indexed: 02/07/2023] Open
Abstract
The small GTPases of the Rho family regulate many aspects of actin dynamics, but are functionally connected to many other cellular processes. Rac1, a member of this family, besides its known function in the regulation of actin cytoskeleton, plays a key role in the production of reactive oxygen species, in gene transcription, in DNA repair, and also has been proven to have specific roles in neurons. This review focuses on the cooperation between Rac1 and Rab proteins, analyzing how the coordination between these GTPases impact on cells and how alterations of their functions lead to disease.
Collapse
|
9
|
Henninger C, Pohlmann S, Ziegler V, Ohlig J, Schmitt J, Fritz G. Distinct contribution of Rac1 expression in cardiomyocytes to anthracycline-induced cardiac injury. Biochem Pharmacol 2019; 164:82-93. [PMID: 30936017 DOI: 10.1016/j.bcp.2019.03.038] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 03/28/2019] [Indexed: 12/16/2022]
Abstract
Cardiotoxicity is the dose limiting adverse effect of anthracycline-based anticancer therapy. Inhibitor studies point to Rac1 as therapeutic target to prevent anthracycline-induced cardiotoxicity. Yet, supporting genetic evidence is still missing and the pathophysiological relevance of different cardiac cell types is unclear. Here, we employed a tamoxifen-inducible cardiomyocyte-specific rac1 knock-out mouse model (Rac1flox/flox/MHC-MerCreMer) to investigate the impact of Rac1 expression in cardiomyocytes on cardiac injury following doxorubicin treatment. Distinctive stress responses resulting from doxorubicin treatment were observed, including upregulation of systemic markers of inflammation (IL-6, IL-1α, MCP-1), cardiac damage (ANP, BNP), DNA damage (i.e. DNA double-strand breaks (DSB)), DNA damage response (DDR) and cell death. Measuring the acute doxorubicin response, the serum level of MCP-1 was elevated, cardiac mRNA expression of Hsp70 was reduced and cardiac DDR was specifically enhanced in Rac1 deficient mice. The frequency of apoptotic heart cells remained unaffected by Rac1. Employing a subactue model, the number of doxorubicin-induced DSB was significantly reduced if Rac1 is absent. Yet, the doxorubicin-triggered increase in serum ANP and BNP levels remained unaffected by Rac1. Overall, knock-out of rac1 in cardiomyocytes confers partial protection against doxorubicin-induced cardiac injury. Hence, the data provide first genetic evidence supporting the view that pharmacological targeting of Rac1 is useful to widen the therapeutic window of anthracycline-based anticancer therapy by alleviating acute/subacute cardiomyocyte damage. Furthermore, considering published data obtained from the use of pharmacological Rac1 inhibitors, the results of our study indicate that Rac1-regulated functions of cardiac cell types others than cardiomyocytes additionally influence the adverse outcomes of anthracycline treatment on the heart.
Collapse
Affiliation(s)
- Christian Henninger
- Institute of Toxicology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstrasse 5, D-40225 Düsseldorf, Germany
| | - Stephanie Pohlmann
- Institute of Toxicology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstrasse 5, D-40225 Düsseldorf, Germany
| | - Verena Ziegler
- Institute of Toxicology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstrasse 5, D-40225 Düsseldorf, Germany
| | - Jan Ohlig
- Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstrasse 5, D-40225 Düsseldorf, Germany
| | - Joachim Schmitt
- Institute of Pharmacology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstrasse 5, D-40225 Düsseldorf, Germany
| | - Gerhard Fritz
- Institute of Toxicology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstrasse 5, D-40225 Düsseldorf, Germany.
| |
Collapse
|
10
|
Liu N, Liu Q, Yang X, Zhang F, Li X, Ma Y, Guan F, Zhao X, Li Z, Zhang L, Ye X. Hepatitis B Virus-Upregulated LNC-HUR1 Promotes Cell Proliferation and Tumorigenesis by Blocking p53 Activity. Hepatology 2018; 68:2130-2144. [PMID: 29790592 DOI: 10.1002/hep.30098] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 05/12/2018] [Indexed: 02/06/2023]
Abstract
Recent studies have indicated that a number of long noncoding RNAs (lncRNAs) are dysregulated in hepatocellular carcinoma, while their aberrant expressions are associated with tumorigenesis and poor prognosis. To identify hepatitis B virus (HBV)-related lncRNAs, we used RNA deep sequencing to quantify the abundances of lncRNAs in HepG2 cells and HBV transgenic HepG2-4D14 cells. Here, we demonstrate that lnc-HUR1 is significantly upregulated in HepG2-4D14 cells. We found that HBV-encoded hepatitis B x protein can enhance the transcription of lnc-HUR1. Overexpression of lnc-HUR1 promotes cell proliferation, whereas knockdown of lnc-HUR1 inhibits cell growth. We identified that lnc-HUR1 can interact with p53 and inhibit its transcriptional regulation on downstream genes, such as p21 and B cell lymphoma 2-associated X protein. We generated lnc-HUR1 transgenic mice and performed the partial hepatectomy (PHx) to examine liver regeneration. The data showed that the ratio of liver weight to body weight in lnc-HUR1 transgenic mice is higher than that in wild-type (WT) littermates at day 2 and day 3 following hepatectomy. Consistently, the results of bromodeoxyuridine staining on liver sections following hepatectomy indicate that the ratio of bromodeoxyuridine-positive cells in lnc-HUR1 transgenic mice is significantly higher than that in WT mice, suggesting that lnc-HUR1 promotes cell proliferation during liver regeneration. Next, we performed the experiment of diethylnitrosamine-induced tumorigenesis. The data demonstrate that tumor number in lnc-HUR1 transgenic mice is higher compared with control mice, indicating that lnc-HUR1 enhances diethylnitrosamine-induced tumorigenesis. Conclusion: We reveal that HBV-upregulated lnc-HUR1 promotes cell proliferation and tumorigenesis by interacting with p53 to block downstream gene transcription. Our findings suggest that lnc-HUR1 plays an important role in HBV-related hepatocellular carcinoma development and may serve as a therapeutic marker for hepatocellular carcinoma. (Hepatology 2018; 00:000-000).
Collapse
Affiliation(s)
- Ningning Liu
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Qi Liu
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiaohai Yang
- Institute of Health Sciences, Anhui University, Hefei, China
| | - Fang Zhang
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xinda Li
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yuanwu Ma
- Key Laboratory of Human Disease Comparative Medicine, NHFPC, Institute of Laboratory Animal Science, Peking Union Medicine College, Chinese Academy of Medical Sciences, Beijing, China
| | - Feifei Guan
- Key Laboratory of Human Disease Comparative Medicine, NHFPC, Institute of Laboratory Animal Science, Peking Union Medicine College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xin Zhao
- 302 Hospital of PLA, Beijing, China
| | | | - Lianfeng Zhang
- Key Laboratory of Human Disease Comparative Medicine, NHFPC, Institute of Laboratory Animal Science, Peking Union Medicine College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xin Ye
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
11
|
Hepatic Rac1 GTPase contributes to liver-mediated basal immune homeostasis and LPS-induced endotoxemia. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:1277-1292. [DOI: 10.1016/j.bbamcr.2018.06.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 05/30/2018] [Accepted: 06/17/2018] [Indexed: 12/16/2022]
|
12
|
Genetic inactivation of Nrf2 prevents clonal expansion of initiated cells in a nutritional model of rat hepatocarcinogenesis. J Hepatol 2018; 69:635-643. [PMID: 29758334 DOI: 10.1016/j.jhep.2018.05.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 04/10/2018] [Accepted: 05/03/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Dysregulation of the Keap1-Nrf2 pathway has been observed in experimental and human tumors, suggesting possible roles of the pathway in cancer development. Herein, we examined whether Nrf2 (Nfe2l2) activation occurs at early steps of rat hepatocarcinogenesis, to assess critical contributions of Nrf2 to the onset of hepatocellular carcinoma (HCC). METHODS We used wild-type (WT) and Nrf2 knockout (Nrf2KO) rats treated with a single injection of diethylnitrosamine (DENA) followed by choline-devoid methionine-deficient (CMD) diet. This experimental model causes massive fatty liver and steatohepatitis with fibrosis and enables identification of early stages of hepatocarcinogenesis. RESULTS We found that Nrf2 activation takes place in early preneoplastic lesions identified by the marker glutathione S-transferase placental form (GSTP). Nrf2 missense mutations, known to disrupt the Keap1-Nrf2 binding, were present in 65.7% of GSTP-positive foci. Nrf2KO rats were used to directly investigate whether Nrf2 is critical for initiation and/or clonal expansion of DENA-damaged hepatocytes. While Nrf2 genetic inactivation did not alter DENA-induced initiation, it led to increased liver injury and chronic compensatory hepatocyte regeneration when rats were fed a CMD diet. However, in spite of such a permissive environment, the livers of Nrf2KO rats did not display any preneoplastic lesion unlike those of WT rats. CONCLUSIONS These results demonstrate that, in a model of hepatocarcinogenesis resembling human non-alcoholic fatty liver disease: i) Nrf2 is activated at early steps of the tumorigenic process and ii) Nrf2 is mandatory for the clonal expansion of initiated cells, indicating that Nrf2 is critical in the onset of HCC. LAY SUMMARY Dysregulation of the Keap1-Nrf2 molecular pathway has been observed in human tumors. In a nutritional model of hepatocarcinogenesis, the protein Nrf2 is frequently mutated/activated at early steps of the tumorigenic process. Herein, we show that Nrf2 is mandatory for the development of preneoplastic lesions. These results suggest that Nrf2 has a critical role in the onset of hepatocellular carcinoma.
Collapse
|
13
|
Cheng H, Wang W, Wang G, Wang A, Du L, Lou W. Silencing Ras-Related C3 Botulinum Toxin Substrate 1 Inhibits Growth and Migration of Hypopharyngeal Squamous Cell Carcinoma via the P38 Mitogen-Activated Protein Kinase Signaling Pathway. Med Sci Monit 2018; 24:768-781. [PMID: 29410394 PMCID: PMC5812251 DOI: 10.12659/msm.907468] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Ras-related C3 botulinum toxin substrate 1 (Rac1) is implicated in a variety of cellular functions and is related to tumor growth and metastasis. This study aimed to explore the role of Rac1 in hypopharyngeal squamous cell carcinoma (HSCC). MATERIAL AND METHODS The Rac1 expression in HSCC tissues was determined by quantitative real-time polymerase chain reaction and Western blot analysis. The level of Rac1 in HSCC cells was downregulated by a Rac1-specific shRNA. Then, the growth and metastasis of HSCC cells were assessed in vitro by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide assay, flow cytometry, Hoechst staining, and Transwell assay. Moreover, cells transfected with Rac1 shRNA or negative control were injected subcutaneously into the right axilla of mice, and then the effects of Rac1 silencing on the growth of HSCC were also explored in vivo. Additionally, activation of the P38 mitogen-activated protein kinase (MAPK) signaling pathway was assessed by Western blot. RESULTS Rac1 was highly expressed in HSCC tissues. Silencing Rac1 inhibited the proliferation and cell cycle progress of HSCC cells, and induced their apoptosis. Rac1 silencing also suppressed the migration and invasion of HSCC cells. In vivo study showed that silencing Rac1 suppressed the growth of tumor bodies. Moreover, the P38 MAPK signaling pathway was implicated in the tumor-suppressing effect of Rac1 silencing in vitro and in vivo. CONCLUSIONS Silencing Rac1 suppressed the growth and migration of HSCC through the P38 MAPK signaling pathway. Due to its contribution in HSCC, Rac1 has the potential to become a promising antitumor therapeutic target for HSCC.
Collapse
Affiliation(s)
- Huijuan Cheng
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Weiwei Wang
- Department of Otolaryngology, Henan Provincial People's Hospital, Zhengzhou, Henan, China (mainland)
| | - Guangke Wang
- Department of Otolaryngology, Henan Provincial People's Hospital, Zhengzhou, Henan, China (mainland)
| | - Anran Wang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Linfang Du
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Weihua Lou
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| |
Collapse
|
14
|
Horng CT, Huang CW, Yang MY, Chen TH, Chang YC, Wang CJ. Nelumbo nucifera leaf extract treatment attenuated preneoplastic lesions and oxidative stress in the livers of diethylnitrosamine-treated rats. ENVIRONMENTAL TOXICOLOGY 2017; 32:2327-2340. [PMID: 28804948 DOI: 10.1002/tox.22434] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 04/26/2017] [Accepted: 04/26/2017] [Indexed: 06/07/2023]
Abstract
Lotus (Nelumbo nucifera Gaertn) possesses antioxidant, hepatoprotective, and anticancer potential. This study determined the protective role of aqueous extract from Nelumbo nucifera leaves (NLE) against N-diethylnitrosamine (DEN)-induced oxidative stress and hepatocellular carcinogenesis in a sample of Sprague-Dawley rats. NLE was fed orally to rats in which hepatic carcinoma was induced with DEN for 12 weeks. Five groups of 12 rats each were used for the study: Group I (control group) rats received distilled water; Group II rats were induced with DEN; Group III rats were induced with DEN and cotreated with 0.5% NLE; Group IV rats were induced with DEN and cotreated with 1.0% NLE; and Group V rats were induced with DEN and cotreated with 2.0% NLE. Clinical chemistry, organ weight, inflammatory marker, protein expression, enzyme, and antioxidant analyses were conducted. NLE administration to rats resulted in significantly decreased levels of serum alanine aminotransferase, aspartate aminotransferase, and albumin, which is indicative of hepatocellular damage, compared with the control group. DEN-induced oxidative stress was inhibited by NLE and this inhibition was paralleled by decreased lipid peroxides and increased glutathione transferase, superoxide dismutase, catalase, and glutathione peroxidase activity in liver tissues. The status of nonenzymatic antioxidants, such as reduced glutathione, was also found to be increased in NLE-administered rats. Furthermore, NLE decreased tumor size, hepatic Rac1, PKCα, and GSTπ expressions compared with the DEN-only group. Thus, supplementation of NLE reduced the adverse changes that occur because of liver cancer. These results prove that NLE protects against liver carcinogenesis induced because of treatment with DEN through blocking lipid peroxidation, hepatic cell damage, and enhancing the antioxidant defense system.
Collapse
Affiliation(s)
- Chi-Ting Horng
- Department of Ophthalmology, Kaohsiung Armed Forces General Hospital, Kaohsiung City, 802, Taiwan
- Institute of Biochemistry, Microbiology and Immunology, Medical College, Chung Shan Medical University, Taichung City, 402, Taiwan
| | - Chien-Wei Huang
- Division of Gastroenterology, Kaohsiung Armed Forces General Hospital, Kaohsiung City, 802, Taiwan
| | - Mon-Yuan Yang
- Institute of Biochemistry, Microbiology and Immunology, Medical College, Chung Shan Medical University, Taichung City, 402, Taiwan
| | - Tzu-Hsin Chen
- Institute of Biochemistry, Microbiology and Immunology, Medical College, Chung Shan Medical University, Taichung City, 402, Taiwan
| | - Yun-Ching Chang
- Institute of Biochemistry, Microbiology and Immunology, Medical College, Chung Shan Medical University, Taichung City, 402, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung City, 402, Taiwan
| | - Chau-Jong Wang
- Institute of Biochemistry, Microbiology and Immunology, Medical College, Chung Shan Medical University, Taichung City, 402, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung City, 402, Taiwan
| |
Collapse
|
15
|
Deshmukh J, Pofahl R, Haase I. Epidermal Rac1 regulates the DNA damage response and protects from UV-light-induced keratinocyte apoptosis and skin carcinogenesis. Cell Death Dis 2017; 8:e2664. [PMID: 28277539 PMCID: PMC5386559 DOI: 10.1038/cddis.2017.63] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 12/31/2016] [Accepted: 01/09/2017] [Indexed: 12/13/2022]
Abstract
Non-melanoma skin cancer (NMSC) is the most common type of cancer. Increased expression and activity of Rac1, a small Rho GTPase, has been shown previously in NMSC and other human cancers; suggesting that Rac1 may function as an oncogene in skin. DMBA/TPA skin carcinogenesis studies in mice have shown that Rac1 is required for chemically induced skin papilloma formation. However, UVB radiation by the sun, which causes DNA damage, is the most relevant cause for NMSC. A potential role of Rac1 in UV-light-induced skin carcinogenesis has not been investigated so far. To investigate this, we irradiated mice with epidermal Rac1 deficiency (Rac1-EKO) and their controls using a well-established protocol for long-term UV-irradiation. Most of the Rac1-EKO mice developed severe skin erosions upon long-term UV-irradiation, unlike their controls. These skin erosions in Rac1-EKO mice healed subsequently. Surprisingly, we observed development of squamous cell carcinomas (SCCs) within the UV-irradiation fields. This shows that the presence of Rac1 in the epidermis protects from UV-light-induced skin carcinogenesis. Short-term UV-irradiation experiments revealed increased UV-light-induced apoptosis of Rac1-deficient epidermal keratinocytes in vitro as well as in vivo. Further investigations using cyclobutane pyrimidine dimer photolyase transgenic mice revealed that the observed increase in UV-light-induced keratinocyte apoptosis in Rac1-EKO mice is DNA damage dependent and correlates with caspase-8 activation. Furthermore, Rac1-deficient keratinocytes showed reduced levels of p53, γ-H2AX and p-Chk1 suggesting an attenuated DNA damage response upon UV-irradiation. Taken together, our data provide direct evidence for a protective role of Rac1 in UV-light-induced skin carcinogenesis and keratinocyte apoptosis probably through regulating mechanisms of the DNA damage response and repair pathways.
Collapse
Affiliation(s)
- Jayesh Deshmukh
- Department of Dermatology, University of Cologne, Kerpener Strasse 62, Cologne 50937, Germany
| | - Ruth Pofahl
- Department of Dermatology, University of Cologne, Kerpener Strasse 62, Cologne 50937, Germany
| | - Ingo Haase
- Department of Dermatology, University of Cologne, Kerpener Strasse 62, Cologne 50937, Germany
| |
Collapse
|
16
|
Zhou J, Liu Y, Luo X, Shen R, Yang C, Yang T, Shi S. Identification and association of RAC1 gene polymorphisms with mRNA and protein expression levels of Rac1 in solid organ (kidney, liver, heart) transplant recipients. Mol Med Rep 2016; 14:1379-88. [PMID: 27279566 DOI: 10.3892/mmr.2016.5383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 05/11/2016] [Indexed: 11/06/2022] Open
Abstract
The activation of Ras-related C3 botulinum toxin substrate 1 (Rac1) is critical in the renal, hepatic and cardiac diseases that lead to the requirement for transplantation, however, no investigations have been performed in Chinese populations to determine the association between RAC1 genotypes and the activation of Rac1. In the present study, 304 solid organ transplant recipients (SOTRs), consisting of 164 renal transplantations, 85 hepatic transplantations and 55 cardiac transplantations, and 332 Chinese healthy control subjects were recruited to investigate whether differences existed in the mRNA and protein expression levels of Rac1 in the different groups. Furthermore, the present study identified and investigated associations of the RAC1 (rs702482, rs10951982, rs702483 and rs6954996) genotypes with the mRNA expression levels of RAC1, and the protein expression levels of total Rac1 and active Rac1‑guanosine triphosphatase (GTP). It was identified that the healthy population had significantly higher levels of Rac1 and Rac1‑GTP, compared with the kidney, liver and heart transplantation populations (P<0.001 for all comparisons). Significant associations (P<0.05) were observed between the RAC1 genotypes and the expression levels of mRNA, Rac1 and Rac1‑GTP. However, the changes in the mRNA expression levels of RAC1 with genotypes were different from those of the proteins. The results of the present study represent the first, to the best of our knowledge, to report that Rac1 and Rac1‑GTP proteins can be downregulated in SOTRs, and that RAC1 genetic polymorphisms can potentially affect the mRNA expression of RAC1, and the protein expression of Rac1 and Rac1‑GTP. These results provide a foundation for further functional investigations to determine the biological and molecular functions of the RAC1 gene in SOTRs.
Collapse
Affiliation(s)
- Jiali Zhou
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yani Liu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Xiaomei Luo
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Rufei Shen
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Chunxiao Yang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Tingyu Yang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Shaojun Shi
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
17
|
Rho GTPases: Novel Players in the Regulation of the DNA Damage Response? Biomolecules 2015; 5:2417-34. [PMID: 26437439 PMCID: PMC4693241 DOI: 10.3390/biom5042417] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 09/02/2015] [Accepted: 09/09/2015] [Indexed: 12/26/2022] Open
Abstract
The Ras-related C3 botulinum toxin substrate 1 (Rac1) belongs to the family of Ras-homologous small GTPases. It is well characterized as a membrane-bound signal transducing molecule that is involved in the regulation of cell motility and adhesion as well as cell cycle progression, mitosis, cell death and gene expression. Rac1 also adjusts cellular responses to genotoxic stress by regulating the activity of stress kinases, including c-Jun-N-terminal kinase/stress-activated protein kinase (JNK/SAPK) and p38 kinases as well as related transcription factors. Apart from being found on the inner side of the outer cell membrane and in the cytosol, Rac1 has also been detected inside the nucleus. Different lines of evidence indicate that genotoxin-induced DNA damage is able to activate nuclear Rac1. The exact mechanisms involved and the biological consequences, however, are unclear. The data available so far indicate that Rac1 might integrate DNA damage independent and DNA damage dependent cellular stress responses following genotoxin treatment, thereby coordinating mechanisms of the DNA damage response (DDR) that are related to DNA repair, survival and cell death.
Collapse
|