1
|
Li Y, Zhang H, Yang F, Zhu D, Chen S, Wang Z, Wei Z, Yang Z, Jia J, Zhang Y, Wang D, Ma M, Kang X. Mechanisms and therapeutic potential of disulphidptosis in cancer. Cell Prolif 2025; 58:e13752. [PMID: 39354653 DOI: 10.1111/cpr.13752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/30/2024] [Accepted: 09/14/2024] [Indexed: 10/04/2024] Open
Abstract
SLC7A11 plays a pivotal role in tumour development by facilitating cystine import to enhance glutathione synthesis and counteract oxidative stress. Disulphidptosis, an emerging form of cell death observed in cells with high expression of SLC7A11 under glucose deprivation, is regulated through reduction-oxidation reactions and disulphide bond formation. This process leads to contraction and collapse of the F-actin cytoskeleton from the plasma membrane, ultimately resulting in cellular demise. Compared to other forms of cell death, disulphidptosis exhibits distinctive characteristics and regulatory mechanisms. This mechanism provides novel insights and innovative strategies for cancer treatment while also inspiring potential therapeutic approaches for other diseases. Our review focuses on elucidating the molecular mechanism underlying disulphidptosis and its connection with the actin cytoskeleton, identifying alternative metabolic forms of cell death, as well as offering insights into disulphidptosis-based cancer therapy. A comprehensive understanding of disulphidptosis will contribute to our knowledge about fundamental cellular homeostasis and facilitate the development of groundbreaking therapies for disease treatment.
Collapse
Affiliation(s)
- Yanhu Li
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Haijun Zhang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
- The Second People's Hospital of Gansu Province, Lanzhou, PR China
| | - Fengguang Yang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Daxue Zhu
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Shijie Chen
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Zhaoheng Wang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Ziyan Wei
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Zhili Yang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Jingwen Jia
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Yizhi Zhang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Dongxin Wang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Mingdong Ma
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Xuewen Kang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| |
Collapse
|
2
|
Dong Y, Zhang Z, Huang H, Yu Y, Rao B, Kuang X, Zeng J, Zhao E, Chen Y, Lu J, Qiu F. ZFHX2-AS1 interacts with DKC1 to regulate ARHGAP5 pseudouridylation and suppress ovarian cancer progression. Cell Signal 2024; 124:111441. [PMID: 39368791 DOI: 10.1016/j.cellsig.2024.111441] [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: 06/17/2024] [Revised: 09/11/2024] [Accepted: 09/29/2024] [Indexed: 10/07/2024]
Abstract
Ovarian cancer (OCa) remains a highly lethal disease, largely due to late-stage diagnosis and limited treatment options for recurrent metastatic tumors. Long non-coding RNAs (lncRNAs) have been recognized as key regulators of cancer hallmarks, yet their specific roles in driving OCa progression are not fully understood. In this study, we employed an integrated approach combining clinical correlation, functional assays, and mechanistic investigations to reveal that lncRNA ZFHX2-AS1 is significantly downregulated in OCa tissues and cells, with its reduced expression associated with poor clinical outcomes. Using in vitro and in vivo models, we demonstrated that overexpression of ZFHX2-AS1 suppresses OCa cell proliferation, migration and invasion, whereas ZFHX2-AS1 knockdown enhances these malignant phenotypes. Mechanistically, we defined that ZFHX2-AS1 interacts with and attenuates the enzymatic activity of the pseudouridine synthase DKC1, thereby reducing pseudouridylation and stabilizing the oncogenic ARHGAP5 mRNA. Re-expression of ARHGAP5 could partially reverse the tumor-suppressive effects of ZFHX2-AS1. Further, we found that ARHGAP5 promotes epithelial-mesenchymal transition (EMT) by regulating Rho GTPases activities, and that ZFHX2-AS1 inhibits EMT in OCa by downregulating ARHGAP5 expression and suppressing the Rho GTPase signaling pathway. Taken together, our findings identify ZFHX2-AS1 as a potent tumor suppressor in OCa, acting through the modulation of DKC1-mediated pseudouridylation of ARHGAP5 and the inhibition of the Rho GTPase pathway, thus offering a potential therapeutic target for combating OCa progression.
Collapse
Affiliation(s)
- Yongshun Dong
- The Key Laboratory of Advanced Interdisciplinary Studies, Institute for Chemical Carcinogenesis, School of Public Health, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, China; Shenzhen Luohu Hospital of Traditional Chinese Medicine, Shenzhen, Guangdong, China
| | - Zili Zhang
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Hongmei Huang
- The Key Laboratory of Advanced Interdisciplinary Studies, Institute for Chemical Carcinogenesis, School of Public Health, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, China
| | - Yonghui Yu
- The Key Laboratory of Advanced Interdisciplinary Studies, Institute for Chemical Carcinogenesis, School of Public Health, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, China
| | - Boqi Rao
- The Key Laboratory of Advanced Interdisciplinary Studies, Institute for Chemical Carcinogenesis, School of Public Health, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, China
| | - Xinjie Kuang
- The Key Laboratory of Advanced Interdisciplinary Studies, Institute for Chemical Carcinogenesis, School of Public Health, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, China
| | - Jie Zeng
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China
| | - Eryong Zhao
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou 510000, China
| | - Yongxiu Chen
- Department of Gynaecology & Obstetrics, Guangdong Women and Children Hospital, Guangzhou 511400, China
| | - Jiachun Lu
- The Key Laboratory of Advanced Interdisciplinary Studies, Institute for Chemical Carcinogenesis, School of Public Health, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, China
| | - Fuman Qiu
- The Key Laboratory of Advanced Interdisciplinary Studies, Institute for Chemical Carcinogenesis, School of Public Health, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, China.
| |
Collapse
|
3
|
Karthikeyan S, Casey PJ, Wang M. RAB4A is a master regulator of cancer cell stemness upstream of NUMB-NOTCH signaling. Cell Death Dis 2024; 15:778. [PMID: 39463384 PMCID: PMC11514220 DOI: 10.1038/s41419-024-07172-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: 05/10/2024] [Revised: 10/03/2024] [Accepted: 10/18/2024] [Indexed: 10/29/2024]
Abstract
Cancer stem cells (CSCs) are a group of specially programmed tumor cells that possess the characteristics of perpetual cell renewal, increased invasiveness, and often, drug resistance. Hence, eliminating CSCs is a major challenge for cancer treatment. Understanding the cellular programs that maintain CSCs, and identifying the critical regulators for such programs, are major undertakings in both basic and translational cancer research. Recently, we have reported that RAB4A is a major regulator of epithelial-to-mesenchymal transition (EMT) and it does so mainly through regulating the activation of RAC1 GTPase. In the current study, we have delineated a new signaling circuitry through which RAB4A transmits its control of cancer stemness. Using in vitro and in vivo studies, we show that RAB4A, as the upstream regulator, relays signal stepwise to NUMB, NOTCH1, RAC1, and then SOX2 to control the self-renewal property of multiple cancer cells of diverse tissue origins. Knockdown of NUMB, or overexpression of NICD (the active fragment NOTCH1) or SOX2, rescued the in vitro sphere-forming and in vivo tumor-forming abilities that were lost upon RAB4A knockdown. Furthermore, we discovered that the chain of control is mostly through transcriptional regulation at every step of the pathway. The discovery of the novel signaling axis of RAB4A-NUMB-NOTCH-SOX2 opens the path for further expansion of the signaling chain and for the identification of new regulators and interacting proteins important for CSC functions, which can be explored to develop new and effective therapies.
Collapse
Affiliation(s)
| | - Patrick J Casey
- Program in Cancer Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Mei Wang
- Program in Cancer Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore.
- Department of Biochemistry, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
4
|
Li Y, Gu A, Yang L, Wang Q. RAC1 serves as a prognostic factor and correlated with immune infiltration in liver hepatocellular carcinoma. J Cancer Res Clin Oncol 2024; 150:418. [PMID: 39264423 PMCID: PMC11393158 DOI: 10.1007/s00432-024-05933-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: 06/20/2024] [Accepted: 08/28/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Hepatocellular carcinoma (LIHC) has severe consequences due to late diagnosis and the lack of effective therapies. Currently, potential biomarkers for the diagnosis and prognosis of LIHC have not been systematically evaluated. Previous studies have reported that RAC1 is associated with the B cell receptor signaling pathway in various tumor microenvironments, but its relationship with LIHC remains unclear. We investigated the relationship between RAC1 and the prognosis and immune infiltration microenvironment of LIHC, exploring its potential as a prognostic biomarker for this type of cancer. METHODS In this study, we analyzed data from The Cancer Genome Atlas (TCGA) using the Wilcoxon signed-rank test and logistic regression to assess the association between RAC1 expression and clinical characteristics in LIHC patients. Additionally, Kaplan-Meier and Cox regression methods were employed to confirm the impact of RAC1 expression levels on overall survival. Immunohistochemistry was used to validate RAC1 protein expression in LIHC. We constructed RAC1 knockdown LIHC cells and studied the effects of RAC1 protein on cell proliferation and migration at both cellular and animal levels. RESULTS RAC1 expression levels were significantly elevated in LIHC tissues compared to normal tissues. High RAC1 expression was strongly associated with advanced pathological stages and was identified as an independent factor negatively affecting overall survival. At both cellular and animal levels, RAC1 knockdown significantly inhibited the proliferation and migration of LIHC cells. Furthermore, RAC1 expression was positively correlated with the infiltration of Th2 cells and macrophages in the tumor microenvironment, suggesting that RAC1 may contribute to the deterioration of the tumor immunosuppressive microenvironment and potentially lead to reduced patient survival. CONCLUSION These findings indicate that RAC1 expression promotes LIHC proliferation and migration and influences the landscape of immune cell infiltration in the tumor microenvironment. Based on these results, RAC1 is proposed as a potential prognostic biomarker for LIHC, associated with both cancer progression and tumor immune cell infiltration.
Collapse
Affiliation(s)
- Yuan Li
- Department of Chemotherapy, The Second Hospital of Nanjing,Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Aidong Gu
- Department of Hepatobiliary Surgery, The Second Hospital of Nanjing,Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Lili Yang
- Department of Chemotherapy, The Second Hospital of Nanjing,Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Qingbo Wang
- Department of Chemotherapy, The Second Hospital of Nanjing,Affiliated to Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
5
|
Wang W, Xia H, Feng P, Dai B. Identification and characterization of RAC1-related immune and prognostic subtypes of hepatocellular carcinoma. J Gene Med 2024; 26:e3719. [PMID: 38979878 DOI: 10.1002/jgm.3719] [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: 01/28/2024] [Revised: 06/18/2024] [Accepted: 06/25/2024] [Indexed: 07/10/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a malignant tumor with significant variability in prognosis among patients. Ras-related C3 botulinum toxin substrate 1 (RAC1) is a key focus in the area of cancer research. However, the molecular mechanisms of RAC1 in HCC remain incompletely elucidated. MATERIALS AND METHODS In this study, bioinformatics analysis was used, and public databases were used to obtain information about HCC cases. The samples were categorized into two groups of high and low expression based on the expression level of RAC1 gene. The limma package was used to calculate the differentially expressed genes between the two groups, and univariate Cox regression analysis was used to screen the prognostic related factors. Consensus clustering analysis was performed using the ConsensusClusterPlus package to identify molecular subtypes of HCC patients. Immune cell infiltration and ESTIMATE scores were assessed using the single sample gene set enrichment analysis and ESTIMATE algorithms. The sensitivity of different isoforms to chemotherapeutic agents was predicted by the oncoPredict package. Finally, we also performed cell function experiments to validate the biological role of RAC1 in vitro. Initially, we classified patients into high and low expression groups based on RAC1 gene expression levels and identified 195 up-regulated genes and 107 down-regulated genes. Through univariate Cox regression analysis, we screened out 169 prognosis-related factors. Furthermore, HCC patients were categorized into two subtypes. Subsequently, Kaplan-Meier survival curves showed that there was a significant difference in prognosis between the two molecular subtypes. Further analysis indicated substantial differences in gene expression levels and TIDE scores between two molecular subtypes. Moreover, these two subtypes exhibited varying sensitivity to chemotherapy drugs, as evidenced by differences in IC50 values. In addition, we found that the silence of RAC1 could effectively inhibit the migration and invasion of HCC cells in vitro. CONCLUSION This study sheds light on the molecular intricacies of RAC1 in HCC and identifies patient populations that may benefit from immunotherapeutic interventions, with potential implications for tailored treatment strategies.
Collapse
Affiliation(s)
- Wei Wang
- Department of Hepatobiliary Surgery, Wuhan No.1 Hospital, Wuhan, China
| | - Hui Xia
- Department of Hepatobiliary Surgery, Wuhan No.1 Hospital, Wuhan, China
| | - Pei Feng
- Department of Rehabilitation Medicine, Wuhan No.1 Hospital, Wuhan, China
| | - Bin Dai
- Department of Hepatobiliary Surgery, Wuhan No.1 Hospital, Wuhan, China
| |
Collapse
|
6
|
Luyckx M, Verougstraete C, Jouret M, Sawadogo K, Waterkeyn M, Grandjean F, Van Gossum JP, Dubois N, Malvaux V, Verreth L, Grandjean P, Jadoul P, Maillard C, Gerday A, Dieu A, Forget P, Baurain JF, Squifflet JL. Intraoperative Ketorolac and Outcomes after Ovarian Cancer Surgery. J Clin Med 2024; 13:1546. [PMID: 38541772 PMCID: PMC10971204 DOI: 10.3390/jcm13061546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/08/2024] [Accepted: 02/20/2024] [Indexed: 11/11/2024] Open
Abstract
INTRODUCTION Surgery is the cornerstone of ovarian cancer treatment. However, surgery and perioperative inflammation have been described as potentially pro-metastagenic. In various animal models and other human cancers, intraoperative administration of non-steroidal anti-inflammatory drugs (NSAIDs) appears to have a positive impact on patient outcomes. MATERIALS AND METHODS In this unicentric retrospective study, we provide an exploratory analysis of the safety and potential benefit of intraoperative administration of ketorolac on the outcome of patients undergoing surgery for ovarian cancer. The study population included all patients who were given a diagnosis of ovarian, fallopian tube or peritoneal cancer by the multidisciplinary oncology committee (MOC) of the Cliniques universitaires Saint-Luc between 2015 and 2020. RESULTS We included 166 patients in our analyses, with a median follow-up of 21.8 months. Both progression-free survival and overall survival were superior in patients who received an intraoperative injection of ketorolac (34.4 months of progression-free survival in the ketorolac group versus 21.5 months in the non-ketorolac group (p = 0.002), and median overall survival was not reached in either group but there was significantly higher survival in the ketorolac group (p = 0.004)). We also performed subgroup analyses to minimise bias due to imbalance between groups on factors that could influence patient survival, and the group of patients receiving ketorolac systematically showed a better outcome. Uni- and multivariate analyses confirmed that administration of ketorolac intraoperatively was associated with better progression-free survival (HR = 0.47 on univariate analysis and 0.43 on multivariate analysis, p = 0.003 and 0.023, respectively). In terms of complications, there were no differences between the two groups, either intraoperatively or postoperatively. CONCLUSION Our study has shown a favourable association between the use of ketorolac during surgery and the postoperative progression of ovarian cancer in a group of 166 patients, without any rise in intra- or postoperative complications. These encouraging results point to the need for a prospective study to confirm the benefit of intraoperative administration of ketorolac in ovarian cancer surgery.
Collapse
Affiliation(s)
- Mathieu Luyckx
- UNGO (UCLouvain Network of Gynaecological Oncology), 1200 Brussel, Belgium; (M.W.); (F.G.); (J.-P.V.G.); (N.D.); (V.M.); (L.V.); (P.G.); (J.-F.B.); (J.-L.S.)
- Gynaecological Oncology Board, Institut Roi Albert II, Cliniques Universitaires Saint-Luc, UCLouvain, 1200 Brussels, Belgium; (C.V.); (P.J.); (C.M.); (A.G.)
- TILS Group, De Duve Institute, UCLouvain, 1200 Brussels, Belgium
| | - Céline Verougstraete
- Gynaecological Oncology Board, Institut Roi Albert II, Cliniques Universitaires Saint-Luc, UCLouvain, 1200 Brussels, Belgium; (C.V.); (P.J.); (C.M.); (A.G.)
| | - Mathieu Jouret
- Obstetrics and Gynecology Department, Centre Hospitaliser de Wallonie Picard (CHWaPi), 7500 Tournai, Belgium;
| | - Kiswendsida Sawadogo
- Statistical Support Unit, Cliniques Universitaire Saint-Luc, UCLouvain, 1200 Brussels, Belgium;
| | - Marc Waterkeyn
- UNGO (UCLouvain Network of Gynaecological Oncology), 1200 Brussel, Belgium; (M.W.); (F.G.); (J.-P.V.G.); (N.D.); (V.M.); (L.V.); (P.G.); (J.-F.B.); (J.-L.S.)
- Obstetrics and Gynecology Department, Cliniques de l’Europe, St. Elisabeth Branch, 1180 Brussel, Belgium
| | - Frédéric Grandjean
- UNGO (UCLouvain Network of Gynaecological Oncology), 1200 Brussel, Belgium; (M.W.); (F.G.); (J.-P.V.G.); (N.D.); (V.M.); (L.V.); (P.G.); (J.-F.B.); (J.-L.S.)
- Obstetrics and Gynecology Department, Cliniques de l’Europe, St. Michel Branch, 1040 Brussel, Belgium
| | - Jean-Paul Van Gossum
- UNGO (UCLouvain Network of Gynaecological Oncology), 1200 Brussel, Belgium; (M.W.); (F.G.); (J.-P.V.G.); (N.D.); (V.M.); (L.V.); (P.G.); (J.-F.B.); (J.-L.S.)
- Obstetrics and Gynecology Department, Clinique St-Jean, 1000 Brussel, Belgium
| | - Nathanael Dubois
- UNGO (UCLouvain Network of Gynaecological Oncology), 1200 Brussel, Belgium; (M.W.); (F.G.); (J.-P.V.G.); (N.D.); (V.M.); (L.V.); (P.G.); (J.-F.B.); (J.-L.S.)
- Obstetrics and Gynecology Department, Clinique St-Jean, 1000 Brussel, Belgium
| | - Vincent Malvaux
- UNGO (UCLouvain Network of Gynaecological Oncology), 1200 Brussel, Belgium; (M.W.); (F.G.); (J.-P.V.G.); (N.D.); (V.M.); (L.V.); (P.G.); (J.-F.B.); (J.-L.S.)
- Obstetrics and Gynecology Department, Clinique St-Pierre, 1340 Ottignies, Belgium
| | - Lucie Verreth
- UNGO (UCLouvain Network of Gynaecological Oncology), 1200 Brussel, Belgium; (M.W.); (F.G.); (J.-P.V.G.); (N.D.); (V.M.); (L.V.); (P.G.); (J.-F.B.); (J.-L.S.)
- Obstetrics and Gynecology Department, Clinique St-Pierre, 1340 Ottignies, Belgium
| | - Pascale Grandjean
- UNGO (UCLouvain Network of Gynaecological Oncology), 1200 Brussel, Belgium; (M.W.); (F.G.); (J.-P.V.G.); (N.D.); (V.M.); (L.V.); (P.G.); (J.-F.B.); (J.-L.S.)
- Obstetrics and Gynecology Department, Centre Hospitalier Régional, 7000 Mons, Belgium
| | - Pascale Jadoul
- Gynaecological Oncology Board, Institut Roi Albert II, Cliniques Universitaires Saint-Luc, UCLouvain, 1200 Brussels, Belgium; (C.V.); (P.J.); (C.M.); (A.G.)
| | - Charlotte Maillard
- Gynaecological Oncology Board, Institut Roi Albert II, Cliniques Universitaires Saint-Luc, UCLouvain, 1200 Brussels, Belgium; (C.V.); (P.J.); (C.M.); (A.G.)
| | - Amandine Gerday
- Gynaecological Oncology Board, Institut Roi Albert II, Cliniques Universitaires Saint-Luc, UCLouvain, 1200 Brussels, Belgium; (C.V.); (P.J.); (C.M.); (A.G.)
| | - Audrey Dieu
- Anesthesiology Department, Cliniques Universitaire Saint-Luc, UCLouvain, 1200 Brussels, Belgium;
| | - Patrice Forget
- Epidemiology Group, Department of Anaesthesia, School of Medicine, Medical Sciences and Nutrition, Institute of Applied Health Sciences, University of Aberdeen, NHS Grampian, Aberdeen AB24 3UE, UK
| | - Jean-François Baurain
- UNGO (UCLouvain Network of Gynaecological Oncology), 1200 Brussel, Belgium; (M.W.); (F.G.); (J.-P.V.G.); (N.D.); (V.M.); (L.V.); (P.G.); (J.-F.B.); (J.-L.S.)
- Gynaecological Oncology Board, Institut Roi Albert II, Cliniques Universitaires Saint-Luc, UCLouvain, 1200 Brussels, Belgium; (C.V.); (P.J.); (C.M.); (A.G.)
- Medical Oncology Department, Institut Roi Albert II, Cliniques Universitaires Saint-Luc, UCLouvain, 1200 Brussels, Belgium
| | - Jean-Luc Squifflet
- UNGO (UCLouvain Network of Gynaecological Oncology), 1200 Brussel, Belgium; (M.W.); (F.G.); (J.-P.V.G.); (N.D.); (V.M.); (L.V.); (P.G.); (J.-F.B.); (J.-L.S.)
- Gynaecological Oncology Board, Institut Roi Albert II, Cliniques Universitaires Saint-Luc, UCLouvain, 1200 Brussels, Belgium; (C.V.); (P.J.); (C.M.); (A.G.)
| |
Collapse
|
7
|
Yan F, Ha JH, Yan Y, Ton SB, Wang C, Mutembei B, Alhajeri ZA, McNiel AF, Keddissi AJ, Zhang Q, Jayaraman M, Dhanasekaran DN, Tang Q. Optical Coherence Tomography of Tumor Spheroids Identifies Candidates for Drug Repurposing in Ovarian Cancer. IEEE Trans Biomed Eng 2023; 70:1891-1901. [PMID: 37015385 PMCID: PMC10482500 DOI: 10.1109/tbme.2022.3231835] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Multicellular tumor spheroids (MCTs) are indispensable models for evaluating drug efficacy for precision cancer therapeutic strategies as well as for repurposing FDA-approved drugs for ovarian cancer. However, current imaging techniques cannot provide effective monitoring of pathological responses due to shallow penetration and experimentally operative destruction. We plan to utilize a noninvasive optical imaging tool to achieve in vivo longitudinal monitoring of the growth of MCTs and therapeutic responses to repurpose three FDA-approved drugs for ovarian cancer therapy. METHODS A swept-source optical coherence tomography (SS-OCT) system was used to monitor the volume growth of MCTs over 11 days. Three inhibitors of 2-Methoxyestradiol (2-ME), AZD1208, and R-Ketorolac (R-keto) with concentrations of 1, 10, and 25 µM were employed to treat ovarian MCTs on day 5. Three-dimensional (3D), intrinsic optical attenuation contrast, and degree of uniformity were applied to analyze the therapeutic effect of these inhibitors on ovarian MCTs. RESULTS We found that 2-ME, AZD1208, and R-keto with concentration of 10 and 25 µM significantly inhibited the volume growth of ovarian MCTs. There was no effect to necrotic tissues from all concentrations of 2-ME, AZD1208, and R-keto inhibitors from our OCT results. 2-ME and AZD1208 inhibited the growth of high uniformity tissues within MCTs and higher concentrations provided more significant inhibitory effects. CONCLUSION Our results indicated that OCT was capable and reliable to monitor the therapeutic effect of inhibitors to ovarian MCTs and it can be used for the rapid characterization of novel therapeutics for ovarian cancers in the future.
Collapse
Affiliation(s)
- Feng Yan
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA
| | - Ji-Hee Ha
- Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Yuyang Yan
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA
| | - Sam B. Ton
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA
| | - Chen Wang
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA
| | - Bornface Mutembei
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA
| | - Zaid A. Alhajeri
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA
| | - Aubrey F. McNiel
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA
| | - Andrew J. Keddissi
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA
| | - Qinghao Zhang
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA
| | - Muralidharan Jayaraman
- Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Danny N. Dhanasekaran
- Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Qinggong Tang
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA
- Institute for Biomedical Engineering, Science, and Technology (IBEST), University of Oklahoma, Norman, OK 73019, USA
| |
Collapse
|
8
|
Rivera M, Toledo-Jacobo L, Romero E, Oprea TI, Moses ME, Hudson LG, Wandinger-Ness A, Grimes MM. Agent-based modeling predicts RAC1 is critical for ovarian cancer metastasis. Mol Biol Cell 2022; 33:ar138. [PMID: 36200848 PMCID: PMC9727804 DOI: 10.1091/mbc.e21-11-0540] [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] [Indexed: 02/04/2023] Open
Abstract
Experimental and computational studies pinpoint rate-limiting step(s) in metastasis governed by Rac1. Using ovarian cancer cell and animal models, Rac1 expression was manipulated, and quantitative measurements of cell-cell and cell-substrate adhesion, cell invasion, mesothelial clearance, and peritoneal tumor growth discriminated the tumor behaviors most highly influenced by Rac1. The experimental data were used to parameterize an agent-based computational model simulating peritoneal niche colonization, intravasation, and hematogenous metastasis to distant organs. Increased ovarian cancer cell survival afforded by the more rapid adhesion and intravasation upon Rac1 overexpression is predicted to increase the numbers of and the rates at which tumor cells are disseminated to distant sites. Surprisingly, crowding of cancer cells along the blood vessel was found to decrease the numbers of cells reaching a distant niche irrespective of Rac1 overexpression or knockdown, suggesting that sites for tumor cell intravasation are rate limiting and become accessible if cells intravasate rapidly or are displaced due to diminished viability. Modeling predictions were confirmed through animal studies of Rac1-dependent metastasis to the lung. Collectively, the experimental and modeling approaches identify cell adhesion, rapid intravasation, and survival in the blood as parameters in the ovarian metastatic cascade that are most critically dependent on Rac1.
Collapse
Affiliation(s)
- Melanie Rivera
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
| | - Leslie Toledo-Jacobo
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
| | - Elsa Romero
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
| | - Tudor I. Oprea
- Division of Translational Informatics, Department of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131,Translational Informatics, Roivant Discovery, Boston, MA 02210
| | - Melanie E. Moses
- Department of Computer Science, University of New Mexico, Albuquerque, NM 87131
| | - Laurie G. Hudson
- Cancer Research Facility, Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM 87131,Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
| | - Angela Wandinger-Ness
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131,Cancer Research Facility, Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM 87131,*Address correspondence to: Angela Wandinger-Ness ()
| | - Martha M. Grimes
- Cancer Research Facility, Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM 87131,Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
| |
Collapse
|
9
|
Sauzeau V, Beignet J, Vergoten G, Bailly C. Overexpressed or hyperactivated Rac1 as a target to treat hepatocellular carcinoma. Pharmacol Res 2022; 179:106220. [PMID: 35405309 DOI: 10.1016/j.phrs.2022.106220] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/31/2022] [Accepted: 04/05/2022] [Indexed: 12/12/2022]
Abstract
Despite novel targeted and immunotherapies, the prognosis remains bleak for patients with hepatocellular carcinoma (HCC), especially for advanced and/or metastatic forms. The rapid emergence of drug resistance is a major obstacle in the success of chemo-, targeted-, immuno-therapies of HCC. Novel targets are needed. The prominent roles of the small GTPase Rac1 in the development and progression of HCC are discussed here, together with its multiple protein partners, and the targeting of Rac1 with RNA-based regulators and small molecules. We discuss the oncogenic functions of Rac1 in HCC, including the contribution of Rac1 mutants and isoform Rac1b. Rac1 is a ubiquitous target, but the protein is frequently overexpressed and hyperactivated in HCC. It contributes to the aggressivity of the disease, with key roles in cancer cell proliferation, tumor metastasis and resistance to treatment. Small molecule targeting Rac1, indirectly or directly, have shown anticancer effects in HCC experimental models. Rac1-binding agents such as EHT 1864 and analogues offer novel opportunities to combat HCC. We discuss the different modalities to repress Rac1 overactivation in HCC with small molecules and the combination with reference drugs to promote cancer cell death and to repress cell invasion. We highlight the necessity to combine Rac1-targeted approach with appropriate biomarkers to select Rac1 activated tumors. Our analysis underlines the prominent oncogenic functions of Rac1 in HCC and discuss the modalities to target this small GTPase. Rac1 shall be considered as a valid target to limit the acquired and intrinsic resistance of HCC tumors and their metastatic potential.
Collapse
Affiliation(s)
- Vincent Sauzeau
- Université de Nantes, CHU Nantes, CNRS, INSERM, Institut du Thorax, Nantes, France.
| | - Julien Beignet
- SATT Ouest Valorisation, 30 boulevard Vincent Gâche, CS 70211, 44202 Nantes Cedex, France
| | - Gérard Vergoten
- University of Lille, Inserm, INFINITE - U1286, Institut de Chimie Pharmaceutique Albert Lespagnol (ICPAL), Faculté de Pharmacie, 3 rue du Professeur Laguesse, BP-83, 59006, Lille, France
| | - Christian Bailly
- OncoWitan, Scientific Consulting Office, Lille, Wasquehal 59290, France.
| |
Collapse
|
10
|
Wang LL, Luo J, He ZH, Liu YQ, Li HG, Xie D, Cai MY. STEAP3 promotes cancer cell proliferation by facilitating nuclear trafficking of EGFR to enhance RAC1-ERK-STAT3 signaling in hepatocellular carcinoma. Cell Death Dis 2021; 12:1052. [PMID: 34741044 PMCID: PMC8571373 DOI: 10.1038/s41419-021-04329-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 09/30/2021] [Accepted: 10/13/2021] [Indexed: 12/24/2022]
Abstract
STEAP3 (Six-transmembrane epithelial antigen of the prostate 3, TSAP6, dudulin-2) has been reported to be involved in tumor progression in human malignancies. Nevertheless, how it participates in the progression of human cancers, especially HCC, is still unknown. In the present study, we found that STEAP3 was aberrantly overexpressed in the nuclei of HCC cells. In a large cohort of clinical HCC tissues, high expression level of nuclear STEAP3 was positively associated with tumor differentiation and poor prognosis (p < 0.001), and it was an independent prognostic factor for HCC patients. In HCC cell lines, nuclear expression of STEAP3 significantly promoted HCC cells proliferation by promoting stemness phenotype and cell cycle progression via RAC1-ERK-STAT3 and RAC1-JNK-STAT6 signaling axes. Through upregulating the expression and nuclear trafficking of EGFR, STEAP3 participated in regulating EGFR-mediated STAT3 transactivity in a manner of positive feedback. In summary, our findings support that nuclear expression of STEAP3 plays a critical oncogenic role in the progression of HCC via modulation on EGFR and intracellular signaling, and it could be a candidate for prognostic marker and therapeutic target in HCC.
Collapse
MESH Headings
- Animals
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Cycle/genetics
- Cell Cycle Proteins/metabolism
- Cell Line, Tumor
- Cell Nucleus/metabolism
- Cell Proliferation
- Disease Progression
- ErbB Receptors/metabolism
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- MAP Kinase Signaling System
- Male
- Mice, Inbred BALB C
- Mice, Nude
- Middle Aged
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Oxidoreductases/metabolism
- Phosphorylation
- Prognosis
- Protein Transport
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- STAT3 Transcription Factor/metabolism
- Signal Transduction
- Spheroids, Cellular/metabolism
- Spheroids, Cellular/pathology
- Treatment Outcome
- rac1 GTP-Binding Protein/metabolism
- Mice
Collapse
Affiliation(s)
- Li-Li Wang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Jie Luo
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Zhang-Hai He
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Ye-Qing Liu
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Hai-Gang Li
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Dan Xie
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China.
- Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China.
| | - Mu-Yan Cai
- Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China.
| |
Collapse
|
11
|
Mulens-Arias V, Rojas JM, Barber DF. The Use of Iron Oxide Nanoparticles to Reprogram Macrophage Responses and the Immunological Tumor Microenvironment. Front Immunol 2021; 12:693709. [PMID: 34177955 PMCID: PMC8221395 DOI: 10.3389/fimmu.2021.693709] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 05/24/2021] [Indexed: 12/12/2022] Open
Abstract
The synthesis and functionalization of iron oxide nanoparticles (IONPs) is versatile, which has enhanced the interest in studying them as theranostic agents over recent years. As IONPs begin to be used for different biomedical applications, it is important to know how they affect the immune system and its different cell types, especially their interaction with the macrophages that are involved in their clearance. How immune cells respond to therapeutic interventions can condition the systemic and local tissue response, and hence, the final therapeutic outcome. Thus, it is fundamental to understand the effects that IONPs have on the immune response, especially in cancer immunotherapy. The biological effects of IONPs may be the result of intrinsic features of their iron oxide core, inducing reactive oxygen species (ROS) and modulating intracellular redox and iron metabolism. Alternatively, their effects are driven by the nanoparticle coating, for example, through cell membrane receptor engagement. Indeed, exploiting these properties of IONPs could lead to the development of innovative therapies. In this review, after a presentation of the elements that make up the tumor immunological microenvironment, we will review and discuss what is currently known about the immunomodulatory mechanisms triggered by IONPs, mainly focusing on macrophage polarization and reprogramming. Consequently, we will discuss the implications of these findings in the context of plausible therapeutic scenarios for cancer immunotherapy.
Collapse
Affiliation(s)
- Vladimir Mulens-Arias
- Department of Immunology and Oncology, and NanoBiomedicine Initiative, Centro Nacional de Biotecnología (CNB)-CSIC, Madrid, Spain
| | - José Manuel Rojas
- Centro de Investigación en Sanidad Animal, Centro Nacional Instituto de Investigación y Tecnología Agraria y Alimentaria (CISA-INIA)-CSIC, Valdeolmos, Madrid, Spain
| | - Domingo F Barber
- Department of Immunology and Oncology, and NanoBiomedicine Initiative, Centro Nacional de Biotecnología (CNB)-CSIC, Madrid, Spain
| |
Collapse
|
12
|
Grimes MM, Kenney SR, Dominguez DR, Brayer KJ, Guo Y, Wandinger-Ness A, Hudson LG. The R-enantiomer of ketorolac reduces ovarian cancer tumor burden in vivo. BMC Cancer 2021; 21:40. [PMID: 33413202 PMCID: PMC7791840 DOI: 10.1186/s12885-020-07716-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 12/08/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Rho-family GTPases, including Ras-related C3 botulinum toxin substrate 1 (Rac1) and cell division control protein 42 (Cdc42), are important modulators of cancer-relevant cell functions and are viewed as promising therapeutic targets. Based on high-throughput screening and cheminformatics we identified the R-enantiomer of an FDA-approved drug (ketorolac) as an inhibitor of Rac1 and Cdc42. The corresponding S-enantiomer is a non-steroidal anti-inflammatory drug (NSAID) with selective activity against cyclooxygenases. We reported previously that R-ketorolac, but not the S-enantiomer, inhibited Rac1 and Cdc42-dependent downstream signaling, growth factor stimulated actin cytoskeleton rearrangements, cell adhesion, migration and invasion in ovarian cancer cell lines and patient-derived tumor cells. METHODS In this study we treated mice with R-ketorolac and measured engraftment of tumor cells to the omentum, tumor burden, and target GTPase activity. In order to gain insights into the actions of R-ketorolac, we also performed global RNA-sequencing (RNA-seq) analysis on tumor samples. RESULTS Treatment of mice with R-ketorolac decreased omental engraftment of ovarian tumor cells at 18 h post tumor cell injection and tumor burden after 2 weeks of tumor growth. R-ketorolac treatment inhibited tumor Rac1 and Cdc42 activity with little impact on mRNA or protein expression of these GTPase targets. RNA-seq analysis revealed that R-ketorolac decreased expression of genes in the HIF-1 signaling pathway. R-ketorolac treatment also reduced expression of additional genes associated with poor prognosis in ovarian cancer. CONCLUSION These findings suggest that R-ketorolac may represent a novel therapeutic approach for ovarian cancer based on its pharmacologic activity as a Rac1 and Cdc42 inhibitor. R-ketorolac modulates relevant pathways and genes associated with disease progression and worse outcome.
Collapse
Affiliation(s)
- Martha M. Grimes
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, New Mexico USA
| | - S. Ray Kenney
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, New Mexico USA
- Division of Molecular Medicine, School of Medicine, University of New Mexico, Albuquerque, New Mexico USA
| | - Dayna R. Dominguez
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, New Mexico USA
| | - Kathryn J. Brayer
- Analytical and Translational Genomics Shared Resource, Comprehensive Cancer Center, University of New Mexico, Albuquerque, New Mexico USA
- Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, New Mexico USA
| | - Yuna Guo
- Department of Pathology, School of Medicine, University of New Mexico, Albuquerque, New Mexico USA
| | - Angela Wandinger-Ness
- Department of Pathology, School of Medicine, University of New Mexico, Albuquerque, New Mexico USA
| | - Laurie G. Hudson
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, New Mexico USA
| |
Collapse
|
13
|
Ahn HS, Yeom J, Yu J, Kwon YI, Kim JH, Kim K. Convergence of Plasma Metabolomics and Proteomics Analysis to Discover Signatures of High-Grade Serous Ovarian Cancer. Cancers (Basel) 2020; 12:cancers12113447. [PMID: 33228226 PMCID: PMC7709037 DOI: 10.3390/cancers12113447] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/10/2020] [Accepted: 11/17/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary In-time diagnosing ovarian cancer, intractable cancer that has no symptoms can increase the survival of women. The aim of this study was to discover biomarkers from liquid biopsy samples using multi-omics approach, metabolomics and proteomics for the diagnosis of ovarian cancer. To verify our biomarker candidates, we conducted comparative analysis with other previous published studies. Despite the limitations of non-invasive samples, our findings are able to discover emerging properties through the interplay between metabolites and proteins and mechanism-based biomarkers through integrated protein and metabolite analysis. Abstract The 5-year survival rate in the early and late stages of ovarian cancer differs by 63%. In addition, a liquid biopsy is necessary because there are no symptoms in the early stage and tissue collection is difficult without using invasive methods. Therefore, there is a need for biomarkers to achieve this goal. In this study, we found blood-based metabolite or protein biomarker candidates for the diagnosis of ovarian cancer in the 20 clinical samples (10 ovarian cancer patients and 10 healthy control subjects). Plasma metabolites and proteins were measured and quantified using mass spectrometry in ovarian cancer patients and control groups. We identified the differential abundant biomolecules (34 metabolites and 197 proteins) and statistically integrated molecules of different dimensions to better understand ovarian cancer signal transduction and to identify novel biological mechanisms. In addition, the biomarker reliability was verified through comparison with existing research results. Integrated analysis of metabolome and proteome identified emerging properties difficult to grasp with the single omics approach, more reliably interpreted the cancer signaling pathway, and explored new drug targets. Especially, through this analysis, proteins (PPCS, PMP2, and TUBB) and metabolites (L-carnitine and PC-O (30:0)) related to the carnitine system involved in cancer plasticity were identified.
Collapse
Affiliation(s)
- Hee-Sung Ahn
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea; (H.-S.A.); (J.Y.)
| | - Jeonghun Yeom
- Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea;
| | - Jiyoung Yu
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea; (H.-S.A.); (J.Y.)
| | | | - Jae-Hoon Kim
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06237, Korea
- Correspondence: (J.-H.K.); (K.K.); Tel.: +82-2-2019-3436 (J.-H.K.); +82-2-1688-7575 (K.K.)
| | - Kyunggon Kim
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea; (H.-S.A.); (J.Y.)
- Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea;
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul 05505, Korea
- Clinical Proteomics Core Laboratory, Convergence Medicine Research Center, Asan Medical Center, Seoul 05505, Korea
- Bio-Medical Institute of Technology, Asan Medical Center, Seoul 05505, Korea
- Correspondence: (J.-H.K.); (K.K.); Tel.: +82-2-2019-3436 (J.-H.K.); +82-2-1688-7575 (K.K.)
| |
Collapse
|
14
|
Hufnagel DH, Cozzi GD, Crispens MA, Beeghly-Fadiel A. Platelets, Thrombocytosis, and Ovarian Cancer Prognosis: Surveying the Landscape of the Literature. Int J Mol Sci 2020; 21:ijms21218169. [PMID: 33142915 PMCID: PMC7663176 DOI: 10.3390/ijms21218169] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 10/26/2020] [Accepted: 10/27/2020] [Indexed: 12/16/2022] Open
Abstract
Platelets are critical components of a number of physiologic processes, including tissue remodeling after injury, wound healing, and maintenance of vascular integrity. Increasing evidence suggests that platelets may also play important roles in cancer. In ovarian cancer, thrombocytosis, both at the time of initial diagnosis and at recurrence, has been associated with poorer prognosis. This review describes current evidence for associations between thrombocytosis and ovarian cancer prognosis and discusses the clinical relevance of platelet count thresholds and timing of assessment. In addition, we discuss several mechanisms from in vitro, in vivo, and clinical studies that may underlie these associations and recommend potential approaches for novel therapeutic targets for this lethal disease.
Collapse
Affiliation(s)
- Demetra H. Hufnagel
- Vanderbilt University School of Medicine, 2209 Garland Avenue, Nashville, TN 37240, USA; (D.H.H.); (G.D.C.)
| | - Gabriella D. Cozzi
- Vanderbilt University School of Medicine, 2209 Garland Avenue, Nashville, TN 37240, USA; (D.H.H.); (G.D.C.)
| | - Marta A. Crispens
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, USA;
- Vanderbilt-Ingram Cancer Center, 1301 Medical Center Drive, Nashville, TN 37232, USA
| | - Alicia Beeghly-Fadiel
- Vanderbilt-Ingram Cancer Center, 1301 Medical Center Drive, Nashville, TN 37232, USA
- Department of Medicine, Division of Epidemiology, Vanderbilt University Medical Center, 2525 West End Avenue, Nashville, TN 37203, USA
- Correspondence:
| |
Collapse
|
15
|
Wang CX, Xiong HF, Wang S, Wang J, Nie X, Guo Q, Li X, Qi Y, Liu JJ, Lin B. Overexpression of TEM8 promotes ovarian cancer progression via Rac1/Cdc42/JNK and MEK/ERK/STAT3 signaling pathways. Am J Transl Res 2020; 12:3557-3576. [PMID: 32774719 PMCID: PMC7407733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 06/04/2020] [Indexed: 06/11/2023]
Abstract
Tumor endothelial cell marker 8 (TEM8) is a type I transmembrane protein, that has been widely studied in the areas of anthrax toxin infection and tumor angiogenesis. However, the role of TEM8 in the progression of epithelial ovarian cancer (EOC) remains unclear. In this study, we determined that TEM8 was highly expressed in ovarian cancer and associated with poor prognosis in EOC patients. In vitro experiments showed that TEM8 overexpression significantly promoted ovarian cancer proliferation. TEM8 overexpression also promoted the G0/G1 phase transition, migration, and invasion of ovarian cancer cells but suppressed apoptosis. Moreover, experimental verification confirmed that TEM8 overexpression increased the expression of Ki-67, cyclin D1, Bcl2/Bax, MMP2, MMP9, and VEGFA and the phosphorylation of Rac1/Cdc42, JNK, MEK, ERK, and STAT3 (Ser727). Subsequently, the addition of RAC1 (EHop-016) and MEK (PD98059) pathway inhibitors suppressed malignant behaviors in the TEM8 overexpression group, which robustly indicated that TEM8 activated Rac1/Cdc42/JNK and MEK/ERK/STAT3 signaling pathways. In addition, we also revealed that the transcription factor GATA2 bound to the TATTAGTTATCTTT site of the TEM8 promoter region and regulated its expression. In conclusion, our study may provide a new theoretical basis for TEM8 application as a clinical biomarker and potential target in EOC patients.
Collapse
Affiliation(s)
- Cai-Xia Wang
- Department of Obstetrics and Gynaecology, Shengjing Hospital Affiliated to China Medical UniversityLiaoning, People’s Republic of China
- Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning ProvinceLiaoning, People’s Republic of China
| | - Hui-Fang Xiong
- Department of Obstetrics and Gynaecology, The Affiliated Longyan First Hospital of Fujian Medical UniversityFujian, People’s Republic of China
| | - Shuang Wang
- Department of Obstetrics and Gynaecology, Shengjing Hospital Affiliated to China Medical UniversityLiaoning, People’s Republic of China
- Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning ProvinceLiaoning, People’s Republic of China
| | - Jing Wang
- Department of Obstetrics and Gynaecology, Shengjing Hospital Affiliated to China Medical UniversityLiaoning, People’s Republic of China
- Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning ProvinceLiaoning, People’s Republic of China
| | - Xin Nie
- Department of Obstetrics and Gynaecology, Shengjing Hospital Affiliated to China Medical UniversityLiaoning, People’s Republic of China
- Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning ProvinceLiaoning, People’s Republic of China
| | - Qian Guo
- Department of Obstetrics and Gynaecology, Shengjing Hospital Affiliated to China Medical UniversityLiaoning, People’s Republic of China
- Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning ProvinceLiaoning, People’s Republic of China
| | - Xiao Li
- Department of Obstetrics and Gynaecology, Shengjing Hospital Affiliated to China Medical UniversityLiaoning, People’s Republic of China
- Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning ProvinceLiaoning, People’s Republic of China
| | - Yue Qi
- Department of Obstetrics and Gynaecology, Shengjing Hospital Affiliated to China Medical UniversityLiaoning, People’s Republic of China
- Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning ProvinceLiaoning, People’s Republic of China
| | - Juan-Juan Liu
- Department of Obstetrics and Gynaecology, Shengjing Hospital Affiliated to China Medical UniversityLiaoning, People’s Republic of China
- Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning ProvinceLiaoning, People’s Republic of China
| | - Bei Lin
- Department of Obstetrics and Gynaecology, Shengjing Hospital Affiliated to China Medical UniversityLiaoning, People’s Republic of China
- Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning ProvinceLiaoning, People’s Republic of China
| |
Collapse
|
16
|
Active RAC1 Promotes Tumorigenic Phenotypes and Therapy Resistance in Solid Tumors. Cancers (Basel) 2020; 12:cancers12061541. [PMID: 32545340 PMCID: PMC7352592 DOI: 10.3390/cancers12061541] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/04/2020] [Accepted: 06/10/2020] [Indexed: 12/16/2022] Open
Abstract
Acting as molecular switches, all three members of the Guanosine triphosphate (GTP)-ase-family, Ras-related C3 botulinum toxin substrate (RAC), Rho, and Cdc42 contribute to various processes of oncogenic transformations in several solid tumors. We have reviewed the distribution of patterns regarding the frequency of Ras-related C3 botulinum toxin substrate 1 (RAC1)-alteration(s) and their modes of actions in various cancers. The RAC1 hyperactivation/copy-number gain is one of the frequently observed features in various solid tumors. We argued that RAC1 plays a critical role in the progression of tumors and the development of resistance to various therapeutic modalities applied in the clinic. With this perspective, here we interrogated multiple functions of RAC1 in solid tumors pertaining to the progression of tumors and the development of resistance with a special emphasis on different tumor cell phenotypes, including the inhibition of apoptosis and increase in the proliferation, epithelial-to-mesenchymal transition (EMT), stemness, pro-angiogenic, and metastatic phenotypes. Our review focuses on the role of RAC1 in adult solid-tumors and summarizes the contextual mechanisms of RAC1 involvement in the development of resistance to cancer therapies.
Collapse
|
17
|
Rho GTPases in Gynecologic Cancers: In-Depth Analysis toward the Paradigm Change from Reactive to Predictive, Preventive, and Personalized Medical Approach Benefiting the Patient and Healthcare. Cancers (Basel) 2020; 12:cancers12051292. [PMID: 32443784 PMCID: PMC7281750 DOI: 10.3390/cancers12051292] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 12/24/2022] Open
Abstract
Rho guanosine triphospatases (GTPases) resemble a conserved family of GTP-binding proteins regulating actin cytoskeleton dynamics and several signaling pathways central for the cell. Rho GTPases create a so-called Ras-superfamily of GTPases subdivided into subgroups comprising at least 20 members. Rho GTPases play a key regulatory role in gene expression, cell cycle control and proliferation, epithelial cell polarity, cell migration, survival, and apoptosis, among others. They also have tissue-related functions including angiogenesis being involved in inflammatory and wound healing processes. Contextually, any abnormality in the Rho GTPase function may result in severe consequences at molecular, cellular, and tissue levels. Rho GTPases also play a key role in tumorigenesis and metastatic disease. Corresponding mechanisms include a number of targets such as kinases and scaffold/adaptor-like proteins initiating GTPases-related signaling cascades. The accumulated evidence demonstrates the oncogenic relevance of Rho GTPases for several solid malignancies including breast, liver, bladder, melanoma, testicular, lung, central nervous system (CNS), head and neck, cervical, and ovarian cancers. Furthermore, Rho GTPases play a crucial role in the development of radio- and chemoresistance e.g. under cisplatin-based cancer treatment. This article provides an in-depth overview on the role of Rho GTPases in gynecological cancers, highlights relevant signaling pathways and pathomechanisms, and sheds light on their involvement in tumor progression, metastatic spread, and radio/chemo resistance. In addition, insights into a spectrum of novel biomarkers and innovative approaches based on the paradigm shift from reactive to predictive, preventive, and personalized medicine are provided.
Collapse
|
18
|
Liu M, Banerjee R, Rossa C, D'Silva NJ. RAP1-RAC1 Signaling Has an Important Role in Adhesion and Migration in HNSCC. J Dent Res 2020; 99:959-968. [PMID: 32401565 DOI: 10.1177/0022034520917058] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cell-cell adhesion is a key mechanism to control tissue integrity and migration. In head and neck squamous cell carcinoma (HNSCC), cell migration facilitates distant metastases and is correlated with poor prognosis. RAP1, a ras-like protein, has an important role in the progression of HNSCC. RAC1 is an integrin-linked, ras-like protein that promotes cell migration. Here we show that loss of cell-cell adhesion is correlated with inactivation of RAP1 confirmed by 2 different biochemical approaches. RAP1 activation is required for cell-matrix adhesion confirmed by adhesion to fibronectin-coated plates with cells that have biochemically activated RAP1. This effect is reversed when RAP1 is inactivated. In addition, RAP1GTP-mediated adhesion is only facilitated through α5β1 integrin complex and is not a function of either α5 or β1 integrin alone. Moreover, the inside-out signaling of RAP1 activation is coordinated with RAC1 activation. These findings show that RAP1 has a prominent role in cell-matrix adhesion via extracellular matrix molecule fibronectin-induced α5β1 integrin and supports a critical role for the RAP1/RAC1 signaling axis in HNSCC cell migration.
Collapse
Affiliation(s)
- M Liu
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - R Banerjee
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - C Rossa
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,Department of Diagnosis and Surgery, School of Dentistry at Araraquara, UNESP-Univ Estadual Paulista, Araraquara, SP, Brazil
| | - N J D'Silva
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,Department of Pathology, Medical School; University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
19
|
Pérez‐Venegas M, Rodríguez‐Treviño AM, Juaristi E. Dual Mechanoenzymatic Kinetic Resolution of (±)‐Ketorolac. ChemCatChem 2020. [DOI: 10.1002/cctc.201902292] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Mario Pérez‐Venegas
- Department of ChemistryCentro de Investigación y de Estudios Avanzados Av. IPN 2508 Ciudad de México 07360 Mexico
| | | | - Eusebio Juaristi
- Department of ChemistryCentro de Investigación y de Estudios Avanzados Av. IPN 2508 Ciudad de México 07360 Mexico
- El Colegio Nacional Donceles 104 Ciudad de México 06020 Mexico
| |
Collapse
|
20
|
Strategies for Delivery of siRNAs to Ovarian Cancer Cells. Pharmaceutics 2019; 11:pharmaceutics11100547. [PMID: 31652539 PMCID: PMC6835428 DOI: 10.3390/pharmaceutics11100547] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/15/2019] [Accepted: 10/18/2019] [Indexed: 12/13/2022] Open
Abstract
The unmet need for novel therapeutic options for ovarian cancer (OC) deserves further investigation. Among the different novel drugs, small interfering RNAs (siRNAs) are particularly attractive because of their specificity of action and efficacy, as documented in many experimental setups. However, the fragility of these molecules in the biological environment necessitates the use of delivery materials able to protect them and possibly target them to the cancer cells. Among the different delivery materials, those based on polymers and lipids are considered very interesting because of their biocompatibility and ability to carry/deliver siRNAs. Despite these features, polymers and lipids need to be engineered to optimize their delivery properties for OC. In this review, we concentrated on the description of the therapeutic potential of siRNAs and polymer-/lipid-based delivery systems for OC. After a brief description of OC and siRNA features, we summarized the strategies employed to minimize siRNA delivery problems, the targeting strategies to OC, and the preclinical models available. Finally, we discussed the most interesting works published in the last three years about polymer-/lipid-based materials for siRNA delivery.
Collapse
|
21
|
Bearer EL, Wu C. Herpes Simplex Virus, Alzheimer's Disease and a Possible Role for Rab GTPases. Front Cell Dev Biol 2019; 7:134. [PMID: 31448273 PMCID: PMC6692634 DOI: 10.3389/fcell.2019.00134] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 07/04/2019] [Indexed: 12/17/2022] Open
Abstract
Herpes simplex virus (HSV) is a common pathogen, infecting 85% of adults in the United States. After reaching the nucleus of the long-lived neuron, HSV may enter latency to persist throughout the life span. Re-activation of latent herpesviruses is associated with progressive cognitive impairment and Alzheimer's disease (AD). As an enveloped DNA virus, HSV exploits cellular membrane systems for its life cycle, and thereby comes in contact with the Rab family of GTPases, master regulators of intracellular membrane dynamics. Knock-down and overexpression of specific Rabs reduce HSV production. Disheveled membrane compartments could lead to AD because membrane sorting and trafficking are crucial for synaptic vesicle formation, neuronal survival signaling and Abeta production. Amyloid precursor protein (APP), a transmembrane glycoprotein, is the parent of Abeta, the major component of senile plaques in AD. Up-regulation of APP expression due to HSV is significant since excess APP interferes with Rab5 endocytic trafficking in neurons. Here, we show that purified PC12-cell endosomes transport both anterograde and retrograde when injected into the squid giant axon at rates similar to isolated HSV. Intracellular HSV co-fractionates with these endosomes, contains APP, Rab5 and TrkA, and displays a second membrane. HSV infected PC12 cells up-regulate APP expression. Whether interference with Rabs has a specific effect on HSV or indirectly affects membrane compartment dynamics co-opted by virus needs further study. Ultimately Rabs, their effectors or their membrane-binding partners may serve as handles to reduce the impact of viral re-activation on cognitive function, or even as more general-purpose anti-microbial therapies.
Collapse
Affiliation(s)
- Elaine L. Bearer
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Chengbiao Wu
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
22
|
Dual Actions of Ketorolac in Metastatic Ovarian Cancer. Cancers (Basel) 2019; 11:cancers11081049. [PMID: 31344967 PMCID: PMC6721416 DOI: 10.3390/cancers11081049] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/11/2019] [Accepted: 07/17/2019] [Indexed: 12/21/2022] Open
Abstract
Cytoreductive surgery and chemotherapy are cornerstones of ovarian cancer treatment, yet disease recurrence remains a significant clinical issue. Surgery can release cancer cells into the circulation, suppress anti-tumor immunity, and induce inflammatory responses that support the growth of residual disease. Intervention within the peri-operative window is an under-explored opportunity to mitigate these consequences of surgery and influence the course of metastatic disease to improve patient outcomes. One drug associated with improved survival in cancer patients is ketorolac. Ketorolac is a chiral molecule administered as a 1:1 racemic mixture of the S- and R-enantiomers. The S-enantiomer is considered the active component for its FDA indication in pain management with selective activity against cyclooxygenase (COX) enzymes. The R-enantiomer has a previously unrecognized activity as an inhibitor of Rac1 (Ras-related C3 botulinum toxin substrate) and Cdc42 (cell division control protein 42) GTPases. Therefore, ketorolac differs from other non-steroidal anti-inflammatory drugs (NSAIDs) by functioning as two distinct pharmacologic entities due to the independent actions of each enantiomer. In this review, we summarize evidence supporting the benefits of ketorolac administration for ovarian cancer patients. We also discuss how simultaneous inhibition of these two distinct classes of targets, COX enzymes and Rac1/Cdc42, by S-ketorolac and R-ketorolac respectively, could each contribute to anti-cancer activity.
Collapse
|
23
|
RAC1 Takes the Lead in Solid Tumors. Cells 2019; 8:cells8050382. [PMID: 31027363 PMCID: PMC6562738 DOI: 10.3390/cells8050382] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/18/2019] [Accepted: 04/24/2019] [Indexed: 12/20/2022] Open
Abstract
Three GTPases, RAC, RHO, and Cdc42, play essential roles in coordinating many cellular functions during embryonic development, both in healthy cells and in disease conditions like cancers. We have presented patterns of distribution of the frequency of RAC1-alteration(s) in cancers as obtained from cBioPortal. With this background data, we have interrogated the various functions of RAC1 in tumors, including proliferation, metastasis-associated phenotypes, and drug-resistance with a special emphasis on solid tumors in adults. We have reviewed the activation and regulation of RAC1 functions on the basis of its sub-cellular localization in tumor cells. Our review focuses on the role of RAC1 in cancers and summarizes the regulatory mechanisms, inhibitory efficacy, and the anticancer potential of RAC1-PAK targeting agents.
Collapse
|
24
|
Hühn J, Musielak M, Schmitz HP, Heinisch JJ. Fungal homologues of human Rac1 as emerging players in signal transduction and morphogenesis. Int Microbiol 2019; 23:43-53. [PMID: 31020478 DOI: 10.1007/s10123-019-00077-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/28/2019] [Accepted: 04/02/2019] [Indexed: 02/06/2023]
Abstract
A wealth of data is accumulating on the physiological functions of human Rac1, a member of the Rho GTPase family of molecular switches and substrate of botulinum toxin, which was first identified as a regulator of cell motility through its effect on the actin cytoskeleton. Later on, it was found to be involved in different diseases like cancers, cardiac function, neuronal disorders, and apoptotic cell death. Despite the presence of Rac1 homologues in most fungi investigated so far, including Rho5 in the genetically tractable model yeast Saccharomyces cerevisiae, knowledge on their physiological functions is still scarce, let alone the details of the molecular mechanisms of their actions and interactions. Nevertheless, all functions proposed for human Rac1 seem to be conserved in one or the other fungus. This includes the regulation of MAPK cascades, polarized growth, and actin dynamics. Moreover, both the production and response to reactive oxygen species, as well as the reaction to nutrient availability, can be affected. We here summarize the studies performed on fungal Rac1 homologues, with a special focus on S. cerevisiae Rho5, which may be of use in drug development in medicine and agriculture.
Collapse
Affiliation(s)
- Julia Hühn
- Department of Biology and Chemistry, Genetics group, University of Osnabrück, Barbarastrasse 11, 49076, Osnabrück, Germany
| | - Marius Musielak
- Department of Biology and Chemistry, Genetics group, University of Osnabrück, Barbarastrasse 11, 49076, Osnabrück, Germany
| | - Hans-Peter Schmitz
- Department of Biology and Chemistry, Genetics group, University of Osnabrück, Barbarastrasse 11, 49076, Osnabrück, Germany
| | - Jürgen J Heinisch
- Department of Biology and Chemistry, Genetics group, University of Osnabrück, Barbarastrasse 11, 49076, Osnabrück, Germany.
| |
Collapse
|
25
|
The Tumor Microenvironment of High Grade Serous Ovarian Cancer. Cancers (Basel) 2018; 11:cancers11010021. [PMID: 30587822 PMCID: PMC6357134 DOI: 10.3390/cancers11010021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 12/25/2018] [Indexed: 12/26/2022] Open
|