1
|
Pethe A, Hartmann TN. The cytoskeletal control of B cell receptor and integrin signaling in normal B cells and chronic lymphocytic leukemia. FEBS Lett 2025. [PMID: 40243025 DOI: 10.1002/1873-3468.70045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/20/2025] [Accepted: 03/25/2025] [Indexed: 04/18/2025]
Abstract
B cells migrate within lymphoid organs during maturation and activation, processes orchestrated by the interplay between B cell receptor (BCR) signaling and microenvironmental cues. Integrins act as mechanoreceptors, linking BCR activation to cytoskeletal remodeling, facilitating immune synapse formation, antigen recognition, and extraction. BCR activation models describe receptor clustering and mechanical changes within the antigen-BCR complex. Upon activation, immune synapses form, enabling antigen extraction and downstream signaling. Integrins stabilize these synapses, amplify BCR signaling, and modulate BCR positioning via actin reorganization. In chronic lymphocytic leukemia (CLL), aberrant BCR signaling and integrins are major players in leukemic cell homing, prognosis, and therapy resistance. In this review, we summarize the current understanding of the interplay of BCR mechanics and B cell localization, with a particular focus on communication between BCR signaling and integrin-mediated processes via actin dynamics. We give insights into normal B cell biology and then outline aspects typical to CLL.
Collapse
Affiliation(s)
- Abhishek Pethe
- Department of Medicine I, Medical Centre-University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Tanja Nicole Hartmann
- Department of Medicine I, Medical Centre-University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| |
Collapse
|
2
|
Matsumura I, Oda T, Kasamatsu T, Murakami Y, Ishihara R, Ohmori A, Matsumoto A, Gotoh N, Kobayashi N, Miyazawa Y, Ogawa Y, Yokohama A, Sasaki N, Saitoh T, Handa H. Role of Rac1 in p53-Related Proliferation and Drug Sensitivity in Multiple Myeloma. Cancers (Basel) 2025; 17:461. [PMID: 39941828 PMCID: PMC11815915 DOI: 10.3390/cancers17030461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/14/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
In this work, the study presented in [...].
Collapse
Affiliation(s)
- Ikuko Matsumura
- Hematology, Graduate School of Medicine, Gunma University, Maebashi 371-8511, Gunma, Japan
| | - Tsukasa Oda
- Mucosal Ecosystem Design, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi 371-8511, Gunma, Japan
| | - Tetsuhiro Kasamatsu
- Faculty of Medical Technology and Clinical Engineering, Gunma University of Health and Welfare, Maebashi 371-0823, Gunma, Japan
| | - Yuki Murakami
- Faculty of Medical Technology and Clinical Engineering, Gunma University of Health and Welfare, Maebashi 371-0823, Gunma, Japan
| | - Rei Ishihara
- Faculty of Medical Technology and Clinical Engineering, Gunma University of Health and Welfare, Maebashi 371-0823, Gunma, Japan
| | - Ayane Ohmori
- Department of Laboratory Sciences, Graduate School of Health Sciences, Gunma University, Maebashi 371-8511, Gunma, Japan
| | - Akira Matsumoto
- Hematology, Graduate School of Medicine, Gunma University, Maebashi 371-8511, Gunma, Japan
| | - Nanami Gotoh
- Department of Laboratory Sciences, Graduate School of Health Sciences, Gunma University, Maebashi 371-8511, Gunma, Japan
| | - Nobuhiko Kobayashi
- Hematology, Graduate School of Medicine, Gunma University, Maebashi 371-8511, Gunma, Japan
| | - Yuri Miyazawa
- Hematology, Graduate School of Medicine, Gunma University, Maebashi 371-8511, Gunma, Japan
| | - Yoshiyuki Ogawa
- Hematology, Graduate School of Medicine, Gunma University, Maebashi 371-8511, Gunma, Japan
| | - Akihiko Yokohama
- Division of Blood Transfusion, Gunma University Hospital, Maebashi 371-8511, Gunma, Japan
| | - Nobuo Sasaki
- Mucosal Ecosystem Design, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi 371-8511, Gunma, Japan
| | - Takayuki Saitoh
- Department of Laboratory Sciences, Graduate School of Health Sciences, Gunma University, Maebashi 371-8511, Gunma, Japan
| | - Hiroshi Handa
- Hematology, Graduate School of Medicine, Gunma University, Maebashi 371-8511, Gunma, Japan
| |
Collapse
|
3
|
Zhang Y, Zhang H, Zhao S, Qi Z, He Y, Zhang X, Wu W, Yan K, Hu L, Sun S, Tang X, Zhou Q, Chen F, Gu A, Wang L, Zhang Z, Yu B, Wang D, Han Y, Xie L, Ji Y. S-Nitrosylation of Septin2 Exacerbates Aortic Aneurysm and Dissection by Coupling the TIAM1-RAC1 Axis in Macrophages. Circulation 2024; 149:1903-1920. [PMID: 38357802 DOI: 10.1161/circulationaha.123.066404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 01/26/2024] [Indexed: 02/16/2024]
Abstract
BACKGROUND S-Nitrosylation (SNO), a prototypic redox-based posttranslational modification, is involved in cardiovascular disease. Aortic aneurysm and dissection are high-risk cardiovascular diseases without an effective cure. The aim of this study was to determine the role of SNO of Septin2 in macrophages in aortic aneurysm and dissection. METHODS Biotin-switch assay combined with liquid chromatography-tandem mass spectrometry was performed to identify the S-nitrosylated proteins in aortic tissue from both patients undergoing surgery for aortic dissection and Apoe-/- mice infused with angiotensin II. Angiotensin II-induced aortic aneurysm model and β-aminopropionitrile-induced aortic aneurysm and dissection model were used to determine the role of SNO of Septin2 (SNO-Septin2) in aortic aneurysm and dissection development. RNA-sequencing analysis was performed to recapitulate possible changes in the transcriptome profile of SNO-Septin2 in macrophages in aortic aneurysm and dissection. Liquid chromatography-tandem mass spectrometry and coimmunoprecipitation were used to uncover the TIAM1-RAC1 (Ras-related C3 botulinum toxin substrate 1) axis as the downstream target of SNO-Septin2. Both R-Ketorolac and NSC23766 treatments were used to inhibit the TIAM1-RAC1 axis. RESULTS Septin2 was identified S-nitrosylated at cysteine 111 (Cys111) in both aortic tissue from patients undergoing surgery for aortic dissection and Apoe-/- mice infused with Angiotensin II. SNO-Septin2 was demonstrated driving the development of aortic aneurysm and dissection. By RNA-sequencing, SNO-Septin2 in macrophages was demonstrated to exacerbate vascular inflammation and extracellular matrix degradation in aortic aneurysm. Next, TIAM1 (T lymphoma invasion and metastasis-inducing protein 1) was identified as a SNO-Septin2 target protein. Mechanistically, compared with unmodified Septin2, SNO-Septin2 reduced its interaction with TIAM1 and activated the TIAM1-RAC1 axis and consequent nuclear factor-κB signaling pathway, resulting in stronger inflammation and extracellular matrix degradation mediated by macrophages. Consistently, both R-Ketorolac and NSC23766 treatments protected against aortic aneurysm and dissection by inhibiting the TIAM1-RAC1 axis. CONCLUSIONS SNO-Septin2 drives aortic aneurysm and dissection through coupling the TIAM1-RAC1 axis in macrophages and activating the nuclear factor-κB signaling pathway-dependent inflammation and extracellular matrix degradation. Pharmacological blockade of RAC1 by R-Ketorolac or NSC23766 may therefore represent a potential treatment against aortic aneurysm and dissection.
Collapse
Affiliation(s)
- Yan Zhang
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Jiangsu, China (Y.Z., H.Z., S.Z., Z.Q., Y.H., X.Z., W.W., K.Y., L.H., S.S., F.C., L.X., Y.J.)
| | - Hao Zhang
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Jiangsu, China (Y.Z., H.Z., S.Z., Z.Q., Y.H., X.Z., W.W., K.Y., L.H., S.S., F.C., L.X., Y.J.)
| | - Shuang Zhao
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Jiangsu, China (Y.Z., H.Z., S.Z., Z.Q., Y.H., X.Z., W.W., K.Y., L.H., S.S., F.C., L.X., Y.J.)
| | - Zhenhua Qi
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Jiangsu, China (Y.Z., H.Z., S.Z., Z.Q., Y.H., X.Z., W.W., K.Y., L.H., S.S., F.C., L.X., Y.J.)
| | - Yiwei He
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Jiangsu, China (Y.Z., H.Z., S.Z., Z.Q., Y.H., X.Z., W.W., K.Y., L.H., S.S., F.C., L.X., Y.J.)
| | - Xuhong Zhang
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Jiangsu, China (Y.Z., H.Z., S.Z., Z.Q., Y.H., X.Z., W.W., K.Y., L.H., S.S., F.C., L.X., Y.J.)
| | - Wencheng Wu
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Jiangsu, China (Y.Z., H.Z., S.Z., Z.Q., Y.H., X.Z., W.W., K.Y., L.H., S.S., F.C., L.X., Y.J.)
| | - Ke Yan
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Jiangsu, China (Y.Z., H.Z., S.Z., Z.Q., Y.H., X.Z., W.W., K.Y., L.H., S.S., F.C., L.X., Y.J.)
| | - Lulu Hu
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Jiangsu, China (Y.Z., H.Z., S.Z., Z.Q., Y.H., X.Z., W.W., K.Y., L.H., S.S., F.C., L.X., Y.J.)
| | - Shixiu Sun
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Jiangsu, China (Y.Z., H.Z., S.Z., Z.Q., Y.H., X.Z., W.W., K.Y., L.H., S.S., F.C., L.X., Y.J.)
| | - Xinlong Tang
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Institute of Cardiothoracic Vascular Disease, Nanjing University, China (X.T., Q.Z., D.W.)
| | - Qing Zhou
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Institute of Cardiothoracic Vascular Disease, Nanjing University, China (X.T., Q.Z., D.W.)
| | - Feng Chen
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Jiangsu, China (Y.Z., H.Z., S.Z., Z.Q., Y.H., X.Z., W.W., K.Y., L.H., S.S., F.C., L.X., Y.J.)
- Department of Forensic Medicine (F.C.), Nanjing Medical University, China
| | - Aihua Gu
- School of Public Health (A.G.), Nanjing Medical University, China
| | - Liansheng Wang
- Departments of Cardiology, First Affiliated Hospital of Nanjing Medical University, China (L.W.)
| | - Zhiren Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Heilongjiang, PR China (Z.Z., Y.J.)
| | - Bo Yu
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Heilongjiang, China (B.Y.)
| | - Dongjin Wang
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Institute of Cardiothoracic Vascular Disease, Nanjing University, China (X.T., Q.Z., D.W.)
| | - Yi Han
- Department of Geriatrics, the First Affiliated Hospital of Nanjing Medical University, China (Y.H.)
| | - Liping Xie
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Jiangsu, China (Y.Z., H.Z., S.Z., Z.Q., Y.H., X.Z., W.W., K.Y., L.H., S.S., F.C., L.X., Y.J.)
- Gusu School, Nanjing Medical University, Suzhou, China (L.X., Y.J.)
| | - Yong Ji
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Jiangsu, China (Y.Z., H.Z., S.Z., Z.Q., Y.H., X.Z., W.W., K.Y., L.H., S.S., F.C., L.X., Y.J.)
- Gusu School, Nanjing Medical University, Suzhou, China (L.X., Y.J.)
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Heilongjiang, PR China (Z.Z., Y.J.)
| |
Collapse
|
4
|
Hornigold K, Baker MJ, Machin PA, Chetwynd SA, Johnsson AK, Pantarelli C, Islam P, Stammers M, Crossland L, Oxley D, Okkenhaug H, Walker S, Walker R, Segonds-Pichon A, Fukui Y, Malliri A, Welch HCE. The Rac-GEF Tiam1 controls integrin-dependent neutrophil responses. Front Immunol 2023; 14:1223653. [PMID: 38077328 PMCID: PMC10703174 DOI: 10.3389/fimmu.2023.1223653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 10/20/2023] [Indexed: 12/18/2023] Open
Abstract
Rac GTPases are required for neutrophil adhesion and migration, and for the neutrophil effector responses that kill pathogens. These Rac-dependent functions are impaired when neutrophils lack the activators of Rac, Rac-GEFs from the Prex, Vav, and Dock families. In this study, we demonstrate that Tiam1 is also expressed in neutrophils, governing focal complexes, actin cytoskeletal dynamics, polarisation, and migration, in a manner depending on the integrin ligand to which the cells adhere. Tiam1 is dispensable for the generation of reactive oxygen species but mediates degranulation and NETs release in adherent neutrophils, as well as the killing of bacteria. In vivo, Tiam1 is required for neutrophil recruitment during aseptic peritonitis and for the clearance of Streptococcus pneumoniae during pulmonary infection. However, Tiam1 functions differently to other Rac-GEFs. Instead of promoting neutrophil adhesion to ICAM1 and stimulating β2 integrin activity as could be expected, Tiam1 restricts these processes. In accordance with these paradoxical inhibitory roles, Tiam1 limits the fMLP-stimulated activation of Rac1 and Rac2 in adherent neutrophils, rather than activating Rac as expected. Tiam1 promotes the expression of several regulators of small GTPases and cytoskeletal dynamics, including αPix, Psd4, Rasa3, and Tiam2. It also controls the association of Rasa3, and potentially αPix, Git2, Psd4, and 14-3-3ζ/δ, with Rac. We propose these latter roles of Tiam1 underlie its effects on Rac and β2 integrin activity and on cell responses. Hence, Tiam1 is a novel regulator of Rac-dependent neutrophil responses that functions differently to other known neutrophil Rac-GEFs.
Collapse
Affiliation(s)
- Kirsti Hornigold
- Signalling Programme, Babraham Institute, Cambridge, United Kingdom
| | - Martin J. Baker
- Signalling Programme, Babraham Institute, Cambridge, United Kingdom
- Cell Signalling Group, Cancer Research UK Manchester Institute, University of Manchester, Macclesfield, United Kingdom
| | - Polly A. Machin
- Signalling Programme, Babraham Institute, Cambridge, United Kingdom
| | | | | | | | - Priota Islam
- Signalling Programme, Babraham Institute, Cambridge, United Kingdom
| | | | | | - David Oxley
- Mass Spectrometry Facility, Babraham Institute, Cambridge, United Kingdom
| | | | - Simon Walker
- Imaging Facility, Babraham Institute, Cambridge, United Kingdom
| | - Rachael Walker
- Flow Cytometry Facility, Babraham Institute, Cambridge, United Kingdom
| | | | - Yoshinori Fukui
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Angeliki Malliri
- Cell Signalling Group, Cancer Research UK Manchester Institute, University of Manchester, Macclesfield, United Kingdom
| | | |
Collapse
|
5
|
Powis G, Meuillet EJ, Indarte M, Booher G, Kirkpatrick L. Pleckstrin Homology [PH] domain, structure, mechanism, and contribution to human disease. Biomed Pharmacother 2023; 165:115024. [PMID: 37399719 DOI: 10.1016/j.biopha.2023.115024] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/14/2023] [Indexed: 07/05/2023] Open
Abstract
The pleckstrin homology [PH] domain is a structural fold found in more than 250 proteins making it the 11th most common domain in the human proteome. 25% of family members have more than one PH domain and some PH domains are split by one, or several other, protein domains although still folding to give functioning PH domains. We review mechanisms of PH domain activity, the role PH domain mutation plays in human disease including cancer, hyperproliferation, neurodegeneration, inflammation, and infection, and discuss pharmacotherapeutic approaches to regulate PH domain activity for the treatment of human disease. Almost half PH domain family members bind phosphatidylinositols [PIs] that attach the host protein to cell membranes where they interact with other membrane proteins to give signaling complexes or cytoskeleton scaffold platforms. A PH domain in its native state may fold over other protein domains thereby preventing substrate access to a catalytic site or binding with other proteins. The resulting autoinhibition can be released by PI binding to the PH domain, or by protein phosphorylation thus providing fine tuning of the cellular control of PH domain protein activity. For many years the PH domain was thought to be undruggable until high-resolution structures of human PH domains allowed structure-based design of novel inhibitors that selectively bind the PH domain. Allosteric inhibitors of the Akt1 PH domain have already been tested in cancer patients and for proteus syndrome, with several other PH domain inhibitors in preclinical development for treatment of other human diseases.
Collapse
Affiliation(s)
- Garth Powis
- PHusis Therapeutics Inc., 6019 Folsom Drive, La Jolla, CA 92037, USA.
| | | | - Martin Indarte
- PHusis Therapeutics Inc., 6019 Folsom Drive, La Jolla, CA 92037, USA
| | - Garrett Booher
- PHusis Therapeutics Inc., 6019 Folsom Drive, La Jolla, CA 92037, USA
| | - Lynn Kirkpatrick
- PHusis Therapeutics Inc., 6019 Folsom Drive, La Jolla, CA 92037, USA
| |
Collapse
|
6
|
Chen F, Gurler SB, Novo D, Selli C, Alferez DG, Eroglu S, Pavlou K, Zhang J, Sims AH, Humphreys NE, Adamson A, Campbell A, Sansom OJ, Tournier C, Clarke RB, Brennan K, Streuli CH, Ucar A. RAC1B function is essential for breast cancer stem cell maintenance and chemoresistance of breast tumor cells. Oncogene 2023; 42:679-692. [PMID: 36599922 PMCID: PMC9957727 DOI: 10.1038/s41388-022-02574-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 11/29/2022] [Accepted: 12/08/2022] [Indexed: 01/06/2023]
Abstract
Breast cancer stem cells (BCSC) are presumed to be responsible for treatment resistance, tumor recurrence and metastasis of breast tumors. However, development of BCSC-targeting therapies has been held back by their heterogeneity and the lack of BCSC-selective molecular targets. Here, we demonstrate that RAC1B, the only known alternatively spliced variant of the small GTPase RAC1, is expressed in a subset of BCSCs in vivo and its function is required for the maintenance of BCSCs and their chemoresistance to doxorubicin. In human breast cancer cell line MCF7, RAC1B is required for BCSC plasticity and chemoresistance to doxorubicin in vitro and for tumor-initiating abilities in vivo. Unlike Rac1, Rac1b function is dispensable for normal mammary gland development and mammary epithelial stem cell (MaSC) activity. In contrast, loss of Rac1b function in a mouse model of breast cancer hampers the BCSC activity and increases their chemosensitivity to doxorubicin treatment. Collectively, our data suggest that RAC1B is a clinically relevant molecular target for the development of BCSC-targeting therapies that may improve the effectiveness of doxorubicin-mediated chemotherapy.
Collapse
Affiliation(s)
- Fuhui Chen
- Manchester Breast Centre, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Sevim B Gurler
- Manchester Breast Centre, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - David Novo
- Wellcome Trust Centre for Cell Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Cigdem Selli
- Applied Bioinformatics of Cancer, Institute of Genetics and Cancer, University of Edinburgh Cancer Research Centre, Edinburgh, UK
| | - Denis G Alferez
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Secil Eroglu
- Manchester Breast Centre, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Kyriaki Pavlou
- Manchester Breast Centre, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jingwei Zhang
- Manchester Breast Centre, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Andrew H Sims
- Applied Bioinformatics of Cancer, Institute of Genetics and Cancer, University of Edinburgh Cancer Research Centre, Edinburgh, UK
| | - Neil E Humphreys
- Genome Editing Unit, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Antony Adamson
- Genome Editing Unit, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | | | - Owen J Sansom
- Cancer Research UK Beatson Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Cathy Tournier
- Manchester Breast Centre, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Robert B Clarke
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Keith Brennan
- Manchester Breast Centre, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Charles H Streuli
- Wellcome Trust Centre for Cell Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Ahmet Ucar
- Manchester Breast Centre, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
7
|
Liu C, Nguyen RY, Pizzurro GA, Zhang X, Gong X, Martinez AR, Mak M. Self-assembly of mesoscale collagen architectures and applications in 3D cell migration. Acta Biomater 2023; 155:167-181. [PMID: 36371004 PMCID: PMC9805527 DOI: 10.1016/j.actbio.2022.11.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 11/11/2022]
Abstract
3D in vitro tumor models have recently been investigated as they can recapitulate key features in the tumor microenvironment. Reconstruction of a biomimetic scaffold is critical in these models. However, most current methods focus on modulating local properties, e.g. micro- and nano-scaled topographies, without capturing the global millimeter or intermediate mesoscale features. Here we introduced a method for modulating the collagen I-based extracellular matrix structure by disruption of fibrillogenesis and the gelation process through mechanical agitation. With this method, we generated collagen scaffolds that are thickened and wavy at a larger scale while featuring global softness. Thickened collagen patches were interconnected with loose collagen networks, highly resembling collagen architecture in the tumor stroma. This thickened collagen network promoted tumor cell dissemination. In addition, this novel modified scaffold triggered differences in morphology and migratory behaviors of tumor cells. Altogether, our method for altered collagen architecture paves new ways for studying in detail cell behavior in physiologically relevant biological processes. STATEMENT OF SIGNIFICANCE: Tumor progression usually involves chronic tissue damage and repair processes. Hallmarks of tumors are highly overlapped with those of wound healing. To mimic the tumor milieu, collagen-based scaffolds are widely used. These scaffolds focus on modulating microscale topographies and mechanics, lacking global architecture similarity compared with in vivo architecture. Here we introduced one type of thick collagen bundles that mimics ECM architecture in human skin scars. These thickened collagen bundles are long and wavy while featuring global softness. This collagen architecture imposes fewer steric restraints and promotes tumor cell dissemination. Our findings demonstrate a distinct picture of cell behaviors and intercellular interactions, highlighting the importance of collagen architecture and spatial heterogeneity of the tumor microenvironment.
Collapse
Affiliation(s)
- Chang Liu
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, United States
| | - Ryan Y Nguyen
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, United States
| | - Gabriela A Pizzurro
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, United States
| | - Xingjian Zhang
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, United States
| | - Xiangyu Gong
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, United States
| | | | - Michael Mak
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, United States.
| |
Collapse
|
8
|
Yang Z, Chen S, Ying H, Yao W. Targeting syndecan-1: new opportunities in cancer therapy. Am J Physiol Cell Physiol 2022; 323:C29-C45. [PMID: 35584326 PMCID: PMC9236862 DOI: 10.1152/ajpcell.00024.2022] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/16/2022] [Accepted: 05/16/2022] [Indexed: 12/02/2022]
Abstract
Syndecan-1 (SDC1, CD138) is one of the heparan sulfate proteoglycans and is essential for maintaining normal cell morphology, interacting with the extracellular and intracellular protein repertoire, as well as mediating signaling transduction upon environmental stimuli. The critical role of SDC1 in promoting tumorigenesis and metastasis has been increasingly recognized in various cancer types, implying a promising potential of utilizing SDC1 as a novel target for cancer therapy. This review summarizes the current knowledge on SDC1 structure and functions, including its role in tumor biology. We also discuss the highlights and limitations of current SDC1-targeted therapies as well as the obstacles in developing new therapeutic methods, offering our perspective on the future directions to target SDC1 for cancer treatment.
Collapse
Affiliation(s)
- Zecheng Yang
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shuaitong Chen
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Haoqiang Ying
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wantong Yao
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
9
|
Vadakumchery A, Faraidun H, Ayoubi OE, Outaleb I, Schmid V, Abdelrasoul H, Amendt T, Khadour A, Setz C, Göhring K, Lodd K, Hitzing C, Alkhatib A, Bilal M, Benckendorff J, Al Shugri AK, Brakebusch CH, Engels N, Datta M, Hobeika E, Alsadeq A, Jumaa H. The Small GTPase RHOA Links SLP65 Activation to PTEN Function in Pre B Cells and Is Essential for the Generation and Survival of Normal and Malignant B Cells. Front Immunol 2022; 13:842340. [PMID: 35371049 PMCID: PMC8965026 DOI: 10.3389/fimmu.2022.842340] [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: 12/23/2021] [Accepted: 02/18/2022] [Indexed: 11/13/2022] Open
Abstract
The generation, differentiation, survival and activation of B cells are coordinated by signals emerging from the B cell antigen receptor (BCR) or its precursor, the pre-BCR. The adaptor protein SLP65 (also known as BLNK) is an important signaling factor that controls pre-B cell differentiation by down-regulation of PI3K signaling. Here, we investigated the mechanism by which SLP65 interferes with PI3K signaling. We found that SLP65 induces the activity of the small GTPase RHOA, which activates PTEN, a negative regulator of PI3K signaling, by enabling its translocation to the plasma membrane. The essential role of RHOA is confirmed by the complete block in early B cell development in conditional RhoA-deficient mice. The RhoA-deficient progenitor B cells showed defects in activation of immunoglobulin gene rearrangement and fail to survive both in vitro and in vivo. Reconstituting the RhoA-deficient cells with RhoA or Foxo1, a transcription factor repressed by PI3K signaling and activated by PTEN, completely restores the survival defect. However, the defect in differentiation can only be restored by RhoA suggesting a unique role for RHOA in B cell generation and selection. In full agreement, conditional RhoA-deficient mice develop increased amounts of autoreactive antibodies with age. RHOA function is also required at later stage, as inactivation of RhoA in peripheral B cells or in a transformed mature B cell line resulted in cell loss. Together, these data show that RHOA is the key signaling factor for B cell development and function by providing a crucial SLP65-activated link between BCR signaling and activation of PTEN. Moreover, the identified essential role of RHOA for the survival of transformed B cells offers the opportunity for targeting B cell malignancies by blocking RHOA function.
Collapse
Affiliation(s)
| | - Hemin Faraidun
- Department of Molecular Immunology, Faculty of Biology, Albert Ludwigs University of Freiburg, Freiburg, Germany
| | - Omar El Ayoubi
- Institute of Immunology, Ulm University Medical Center, Ulm, Germany
| | - Issame Outaleb
- Department of Molecular Immunology, Faculty of Biology, Albert Ludwigs University of Freiburg, Freiburg, Germany
| | - Vera Schmid
- Institute of Immunology, Ulm University Medical Center, Ulm, Germany
| | - Hend Abdelrasoul
- Institute of Immunology, Ulm University Medical Center, Ulm, Germany
| | - Timm Amendt
- Institute of Immunology, Ulm University Medical Center, Ulm, Germany
| | - Ahmad Khadour
- Institute of Immunology, Ulm University Medical Center, Ulm, Germany
| | - Corinna Setz
- Institute of Immunology, Ulm University Medical Center, Ulm, Germany
| | - Katharina Göhring
- Institute of Immunology, Ulm University Medical Center, Ulm, Germany
| | - Karoline Lodd
- Institute of Immunology, Ulm University Medical Center, Ulm, Germany
| | - Christoffer Hitzing
- Institute of Cellular & Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Alabbas Alkhatib
- Department of Molecular Immunology, Faculty of Biology, Albert Ludwigs University of Freiburg, Freiburg, Germany
| | - Mayas Bilal
- Institute of Immunology, Ulm University Medical Center, Ulm, Germany
| | | | | | - Cord Herbert Brakebusch
- Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Niklas Engels
- Institute of Cellular & Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Moumita Datta
- Institute of Immunology, Ulm University Medical Center, Ulm, Germany
| | - Elias Hobeika
- Institute of Immunology, Ulm University Medical Center, Ulm, Germany
| | - Ameera Alsadeq
- Institute of Immunology, Ulm University Medical Center, Ulm, Germany
| | - Hassan Jumaa
- Institute of Immunology, Ulm University Medical Center, Ulm, Germany.,Department of Molecular Immunology, Faculty of Biology, Albert Ludwigs University of Freiburg, Freiburg, Germany
| |
Collapse
|
10
|
Huang Z, Pu J, Luo Y, Fan J, Li K, Peng D, Zong K, Zhou B, Guan X, Zhou F. FAM49B, restrained by miR-22, relieved hepatic ischemia/reperfusion injury by inhibiting TRAF6/IKK signaling pathway in a Rac1-dependent manner. Mol Immunol 2022; 143:135-146. [PMID: 35131594 DOI: 10.1016/j.molimm.2022.01.015] [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: 05/03/2021] [Revised: 01/22/2022] [Accepted: 01/27/2022] [Indexed: 11/29/2022]
Abstract
Hepatic ischemia/reperfusion (I/R) injury plays a pivotal pathogenic role in trauma, hepatectomy, and liver transplantation. However, the whole mechanism remains undescribed. The objective of this study is to investigate the internal mechanism by which microRNA-22 (miR-22) targets family with sequence similarity 49 member B (FAM49B), thus aggravating hepatic I/R injury. Here, we found that miR-22 was upregulated while FAM49B was reduced in hepatic I/R injury. Inhibition of miR-22 in vitro was able to intensify expression of FAM49B, thus reducing phosphorylation of inhibitors of nuclear factor kappa-B kinase (IKK) and downstream pro-inflammatory proteins. A dual luciferase reporter assay indicated that miR-22 directly targeted FAM49B. Remission of hepatic pathologic alterations, apoptosis, and release of cytokines derived from constraints of miR-22 were abolished in vivo by repressing FAM49B. Further interference of Ras-related C3 botulinum toxin substrate 1 (Rac1) reversed the function of FAM49B inhibition, thus achieving anti-inflammatory consequences.
Collapse
Affiliation(s)
- Zuotian Huang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Junliang Pu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yunhai Luo
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Fan
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kaili Li
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dadi Peng
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kezhen Zong
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Baoyong Zhou
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiangdong Guan
- Department of Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Fachun Zhou
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
11
|
Gong FH, Chen XL, Zhang Q, Xiao XQ, Yang YS, Song BJ, Chao SP, Cheng WL. MicroRNA-183 as a Novel Regulator Protects Against Cardiomyocytes Hypertrophy via Targeting TIAM1. Am J Hypertens 2022; 35:87-95. [PMID: 32870256 DOI: 10.1093/ajh/hpaa144] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 07/10/2020] [Accepted: 08/29/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND MicroRNAs serve as important regulators of the pathogenesis of cardiac hypertrophy. Among them, miR-183 is well documented as a novel tumor suppressor in previous studies, whereas it exhibits a downregulated expression in cardiac hypertrophy recently. The present study was aimed to examine the effect of miR-183 on cardiomyocytes hypertrophy. METHODS Angiotensin II (Ang II) was used for establishment of cardiac hypertrophy model in vitro. Neonatal rat ventricular cardiomyocytes transfected with miR-183 mimic or negative control were further utilized for the phenotype analysis. Moreover, the bioinformatics analysis and luciferase reporter assays were used for exploring the potential target of miR-183 in cardiomyocytes. RESULTS We observed a significant decreased expression of miR-183 in hypertrophic cardiomyocytes. Overexpression of miR-183 significantly attenuated the cardiomyocytes size morphologically and prohypertrophic genes expression. Moreover, we demonstrated that TIAM1 was a direct target gene of miR-183 verified by bioinformatics analysis and luciferase reporter assays, which showed a decreased mRNA and protein expression in the cardiomyocytes transfected with miR-183 upon Ang II stimulation. Additionally, the downregulated TIAM1 expression was required for the attenuated effect of miR-183 on cardiomyocytes hypertrophy. CONCLUSIONS Taken together, these evidences indicated that miR-183 acted as a cardioprotective regulator for the development of cardiomyocytes hypertrophy via directly regulation of TIAM1.
Collapse
Affiliation(s)
- Fu-han Gong
- Department of Cardiology, Tongren Municipal People’s Hospital, Tongren, China
| | - Xi-Lu Chen
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Quan Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-qiang Xiao
- Department of Cardiology, Tongren Municipal People’s Hospital, Tongren, China
| | - Yong-sheng Yang
- Department of Cardiology, Tongren Municipal People’s Hospital, Tongren, China
| | - Bian-jing Song
- Department of Cardiology, Tongren Municipal People’s Hospital, Tongren, China
| | - Sheng-ping Chao
- Department of Cardiology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Wen-Lin Cheng
- Department of Cardiology, Zhongnan Hospital, Wuhan University, Wuhan, China
| |
Collapse
|
12
|
Fan G, Lou L, Song Z, Zhang X, Xiong XF. Targeting mutated GTPase KRAS in tumor therapies. Eur J Med Chem 2021; 226:113816. [PMID: 34520956 DOI: 10.1016/j.ejmech.2021.113816] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/24/2021] [Accepted: 08/29/2021] [Indexed: 12/13/2022]
Abstract
Kirsten rat sarcoma virus oncogene (KRAS) mutation accounts for approximately 85% of RAS-driven cancers, and participates in multiple signaling pathways and mediates cell proliferation, differentiation and metabolism. KRAS has been considered as an "undruggable" target due to the lack of effective direct inhibitors, although high frequency of KRAS mutations have been identified in multiple carcinomas in the past decades. Encouragingly, the KRASG12C inhibitor AMG510 (sotorasib), which has been approved for treating NSCLC and CRC recently, makes directly targeting KRAS the most promising strategy for cancer therapy. To better understand the current state of KRAS inhibitors, this review summarizes the biological functions of KRAS, the structure-activity relationship studies of the small-molecule inhibitors that directly target KRAS, and highlights the therapeutic agents with improved selectivity, bioavailability and physicochemical properties. Furthermore, the combined medication that can enhance efficacy and overcome drug resistance of KRAS covalent inhibitors is also reviewed.
Collapse
Affiliation(s)
- Guangjin Fan
- Guangdong Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Linlin Lou
- Guangdong Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Zhendong Song
- Guangdong Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China.
| | - Xiaolei Zhang
- Guangdong Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China.
| | - Xiao-Feng Xiong
- Guangdong Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China.
| |
Collapse
|
13
|
Scala M, Nishikawa M, Nagata KI, Striano P. Pathophysiological Mechanisms in Neurodevelopmental Disorders Caused by Rac GTPases Dysregulation: What's behind Neuro-RACopathies. Cells 2021; 10:3395. [PMID: 34943902 PMCID: PMC8699292 DOI: 10.3390/cells10123395] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/23/2021] [Accepted: 11/30/2021] [Indexed: 02/07/2023] Open
Abstract
Rho family guanosine triphosphatases (GTPases) regulate cellular signaling and cytoskeletal dynamics, playing a pivotal role in cell adhesion, migration, and cell cycle progression. The Rac subfamily of Rho GTPases consists of three highly homologous proteins, Rac 1-3. The proper function of Rac1 and Rac3, and their correct interaction with guanine nucleotide-exchange factors (GEFs) and GTPase-activating proteins (GAPs) are crucial for neural development. Pathogenic variants affecting these delicate biological processes are implicated in different medical conditions in humans, primarily neurodevelopmental disorders (NDDs). In addition to a direct deleterious effect produced by genetic variants in the RAC genes, a dysregulated GTPase activity resulting from an abnormal function of GEFs and GAPs has been involved in the pathogenesis of distinctive emerging conditions. In this study, we reviewed the current pertinent literature on Rac-related disorders with a primary neurological involvement, providing an overview of the current knowledge on the pathophysiological mechanisms involved in the neuro-RACopathies.
Collapse
Affiliation(s)
- Marcello Scala
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16132 Genoa, Italy;
- Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Masashi Nishikawa
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, 713-8 Kamiya, Kasugai 480-0392, Japan; (M.N.); (K.-i.N.)
| | - Koh-ichi Nagata
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, 713-8 Kamiya, Kasugai 480-0392, Japan; (M.N.); (K.-i.N.)
- Department of Neurochemistry, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Nagoya 466-8550, Japan
| | - Pasquale Striano
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16132 Genoa, Italy;
- Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| |
Collapse
|
14
|
Youssef A, Haskali MB, Gorringe KL. The Protein Landscape of Mucinous Ovarian Cancer: Towards a Theranostic. Cancers (Basel) 2021; 13:5596. [PMID: 34830751 PMCID: PMC8616050 DOI: 10.3390/cancers13225596] [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: 10/11/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 01/20/2023] Open
Abstract
MOC is a rare histotype of epithelial ovarian cancer, and current management options are inadequate for the treatment of late stage or recurrent disease. A shift towards personalised medicines in ovarian cancer is being observed, with trials targeting specific molecular pathways, however, MOC lags due to its rarity. Theranostics is a rapidly evolving category of personalised medicine, encompassing both a diagnostic and therapeutic approach by recognising targets that are expressed highly in tumour tissue in order to deliver a therapeutic payload. The present review evaluates the protein landscape of MOC in recent immunohistochemical- and proteomic-based research, aiming to identify potential candidates for theranostic application. Fourteen proteins were selected based on cell membrane localisation: HER2, EGFR, FOLR1, RAC1, GPR158, CEACAM6, MUC16, PD-L1, NHE1, CEACAM5, MUC1, ACE2, GP2, and PTPRH. Optimal proteins to target using theranostic agents must exhibit high membrane expression on cancerous tissue with low expression on healthy tissue to afford improved disease outcomes with minimal off-target effects and toxicities. We provide guidelines to consider in the selection of a theranostic target for MOC and suggest future directions in evaluating the results of this review.
Collapse
Affiliation(s)
- Arkan Youssef
- Department of Medicine, The University of Melbourne, Melbourne, VIC 3000, Australia;
| | - Mohammad B. Haskali
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3000, Australia;
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Kylie L. Gorringe
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3000, Australia;
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| |
Collapse
|
15
|
Crosas-Molist E, Samain R, Kohlhammer L, Orgaz J, George S, Maiques O, Barcelo J, Sanz-Moreno V. RhoGTPase Signalling in Cancer Progression and Dissemination. Physiol Rev 2021; 102:455-510. [PMID: 34541899 DOI: 10.1152/physrev.00045.2020] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Rho GTPases are a family of small G proteins that regulate a wide array of cellular processes related to their key roles controlling the cytoskeleton. On the other hand, cancer is a multi-step disease caused by the accumulation of genetic mutations and epigenetic alterations, from the initial stages of cancer development when cells in normal tissues undergo transformation, to the acquisition of invasive and metastatic traits, responsible for a large number of cancer related deaths. In this review, we discuss the role of Rho GTPase signalling in cancer in every step of disease progression. Rho GTPases contribute to tumour initiation and progression, by regulating proliferation and apoptosis, but also metabolism, senescence and cell stemness. Rho GTPases play a major role in cell migration, and in the metastatic process. They are also involved in interactions with the tumour microenvironment and regulate inflammation, contributing to cancer progression. After years of intensive research, we highlight the importance of relevant models in the Rho GTPase field, and we reflect on the therapeutic opportunities arising for cancer patients.
Collapse
Affiliation(s)
- Eva Crosas-Molist
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Remi Samain
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Leonie Kohlhammer
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Jose Orgaz
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom.,Instituto de Investigaciones Biomédicas 'Alberto Sols', CSIC-UAM, 28029, Madrid, Spain
| | - Samantha George
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Oscar Maiques
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Jaume Barcelo
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | | |
Collapse
|
16
|
Hosseini A, Hamblin MR, Mirzaei H, Mirzaei HR. Role of the bone marrow microenvironment in drug resistance of hematological malignances. Curr Med Chem 2021; 29:2290-2305. [PMID: 34514979 DOI: 10.2174/0929867328666210910124319] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 07/07/2021] [Accepted: 07/20/2021] [Indexed: 11/22/2022]
Abstract
The unique features of the tumor microenvironment (TME) govern the biological properties of many cancers, including hematological malignancies. TME factors can trigger invasion, and protect against drug cytotoxicity by inhibiting apoptosis and activating specific signaling pathways (e.g. NF-ΚB). TME remodeling is facilitated due to the high self-renewal ability of the bone marrow. Progressing tumor cells can alter some extracellular matrix (ECM) components which act as a barrier to drug penetration in the TME. The initial progression of the cell cycle is controlled by the MAPK pathway (Raf/MEK/ERK) and Hippo pathway, while the final phase is regulated by the PI3K/Akt /mTOR and WNT pathways. In this review we summarize the main signaling pathways involved in drug resistance (DR) and some mechanisms by which DR can occur in the bone marrow. The relationship between autophagy, endoplasmic reticulum stress, and cellular signaling pathways in DR and apoptosis are covered in relation to the TME.
Collapse
Affiliation(s)
- Alireza Hosseini
- Laboratory Hematology and Blood Banking, Tehran University of Medical Sciences, Tehran. Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028. South Africa
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan. Iran
| | - Hamid Reza Mirzaei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran. Iran
| |
Collapse
|
17
|
Liang J, Oyang L, Rao S, Han Y, Luo X, Yi P, Lin J, Xia L, Hu J, Tan S, Tang L, Pan Q, Tang Y, Zhou Y, Liao Q. Rac1, A Potential Target for Tumor Therapy. Front Oncol 2021; 11:674426. [PMID: 34079763 PMCID: PMC8165220 DOI: 10.3389/fonc.2021.674426] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/29/2021] [Indexed: 12/20/2022] Open
Abstract
RAS-related C3 botulinum toxin substrate 1 (Rac.1) is one of the important members of Rho GTPases. It is well known that Rac1 is a cytoskeleton regulation protein that regulates cell adhesion, morphology, and movement. Rac1 is highly expressed in different types of tumors, which is related to poor prognosis. Studies have shown that Rac1 not only participates in the tumor cell cycle, apoptosis, proliferation, invasion, migration and angiogenesis, but also participates in the regulation of tumor stem cell, thus promoting the occurrence of tumors. Rac1 also plays a key role in anti-tumor therapy and participates in immune escape mediated by the tumor microenvironment. In addition, the good prospects of Rac1 inhibitors in cancer prevention and treatment are exciting. Therefore, Rac1 is considered as a potential target for the prevention and treatment of cancer. The necessity and importance of Rac1 are obvious, but it still needs further study.
Collapse
Affiliation(s)
- Jiaxin Liang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Linda Oyang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Shan Rao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yaqian Han
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Xia Luo
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Pin Yi
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Jinguan Lin
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Longzheng Xia
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Jiaqi Hu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Shiming Tan
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Lu Tang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,University of South China, Hengyang, China
| | - Qing Pan
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,University of South China, Hengyang, China
| | - Yanyan Tang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Clinical Research Center for Wound Healing in Hunan Province, Changsha, China
| | - Yujuan Zhou
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Qianjin Liao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Clinical Research Center for Wound Healing in Hunan Province, Changsha, China
| |
Collapse
|
18
|
Fragliasso V, Tameni A, Inghirami G, Mularoni V, Ciarrocchi A. Cytoskeleton Dynamics in Peripheral T Cell Lymphomas: An Intricate Network Sustaining Lymphomagenesis. Front Oncol 2021; 11:643620. [PMID: 33928032 PMCID: PMC8076600 DOI: 10.3389/fonc.2021.643620] [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: 12/18/2020] [Accepted: 03/17/2021] [Indexed: 12/04/2022] Open
Abstract
Defects in cytoskeleton functions support tumorigenesis fostering an aberrant proliferation and promoting inappropriate migratory and invasive features. The link between cytoskeleton and tumor features has been extensively investigated in solid tumors. However, the emerging genetic and molecular landscape of peripheral T cell lymphomas (PTCL) has unveiled several alterations targeting structure and function of the cytoskeleton, highlighting its role in cell shape changes and the aberrant cell division of malignant T cells. In this review, we summarize the most recent evidence about the role of cytoskeleton in PTCLs development and progression. We also discuss how aberrant signaling pathways, like JAK/STAT3, NPM-ALK, RhoGTPase, and Aurora Kinase, can contribute to lymphomagenesis by modifying the structure and the signaling properties of cytoskeleton.
Collapse
Affiliation(s)
- Valentina Fragliasso
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Annalisa Tameni
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy.,Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Giorgio Inghirami
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Valentina Mularoni
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Alessia Ciarrocchi
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| |
Collapse
|
19
|
Lareau CA, Ludwig LS, Muus C, Gohil SH, Zhao T, Chiang Z, Pelka K, Verboon JM, Luo W, Christian E, Rosebrock D, Getz G, Boland GM, Chen F, Buenrostro JD, Hacohen N, Wu CJ, Aryee MJ, Regev A, Sankaran VG. Massively parallel single-cell mitochondrial DNA genotyping and chromatin profiling. Nat Biotechnol 2021; 39:451-461. [PMID: 32788668 PMCID: PMC7878580 DOI: 10.1038/s41587-020-0645-6] [Citation(s) in RCA: 178] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 07/17/2020] [Indexed: 12/24/2022]
Abstract
Natural mitochondrial DNA (mtDNA) mutations enable the inference of clonal relationships among cells. mtDNA can be profiled along with measures of cell state, but has not yet been combined with the massively parallel approaches needed to tackle the complexity of human tissue. Here, we introduce a high-throughput, droplet-based mitochondrial single-cell assay for transposase-accessible chromatin with sequencing (scATAC-seq), a method that combines high-confidence mtDNA mutation calling in thousands of single cells with their concomitant high-quality accessible chromatin profile. This enables the inference of mtDNA heteroplasmy, clonal relationships, cell state and accessible chromatin variation in individual cells. We reveal single-cell variation in heteroplasmy of a pathologic mtDNA variant, which we associate with intra-individual chromatin variability and clonal evolution. We clonally trace thousands of cells from cancers, linking epigenomic variability to subclonal evolution, and infer cellular dynamics of differentiating hematopoietic cells in vitro and in vivo. Taken together, our approach enables the study of cellular population dynamics and clonal properties in vivo.
Collapse
Affiliation(s)
- Caleb A Lareau
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Division of Medical Sciences, Harvard Medical School, Boston, MA, USA.
| | - Leif S Ludwig
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Christoph Muus
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Satyen H Gohil
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Academic Haematology, UCL Cancer Institute, London, UK
| | - Tongtong Zhao
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Zachary Chiang
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Medical Sciences, Harvard Medical School, Boston, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Karin Pelka
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jeffrey M Verboon
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Wendy Luo
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Elena Christian
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Medical Sciences, Harvard Medical School, Boston, MA, USA
| | - Daniel Rosebrock
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Gad Getz
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Genevieve M Boland
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Fei Chen
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jason D Buenrostro
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Nir Hacohen
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Catherine J Wu
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Martin J Aryee
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Pathology, Harvard Medical School, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
- Department of Biology and Koch Institute, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Vijay G Sankaran
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
20
|
Butera A, Cassandri M, Rugolo F, Agostini M, Melino G. The ZNF750-RAC1 axis as potential prognostic factor for breast cancer. Cell Death Discov 2020; 6:135. [PMID: 33298895 PMCID: PMC7701147 DOI: 10.1038/s41420-020-00371-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 12/14/2022] Open
Abstract
The human zinc finger (C2H2-type) protein ZNF750 is a transcription factor regulated by p63 that plays a critical role in epithelial tissues homoeostasis, as well as being involved in the pathogenesis of cancer. Indeed, missense mutations, truncation and genomic deletion have been found in oesophageal squamous cell carcinoma. In keeping, we showed that ZNF750 negatively regulates cell migration and invasion in breast cancer cells; in particular, ZNF750 binds and recruits KDM1A and HDAC1 on the LAMB3 and CTNNAL1 promoters. This interaction, in turn, represses the transcription of LAMB3 and CTNNAL1 genes, which are involved in cell migration and invasion. Given that ZNF750 is emerging as a crucial transcription factor that acts as tumour suppressor gene, here, we show that ZNF750 represses the expression of the small GTPase, Ras-related C3 botulinum toxin substrate 1 (RAC1) in breast cancer cell lines, by directly binding its promoter region. In keeping with ZNF750 controlling RAC1 expression, we found an inverse correlation between ZNF750 and RAC1 in human breast cancer datasets. More importantly, we found a significant upregulation of RAC1 in human breast cancer datasets and we identified a direct correlation between RAC1 expression and the survival rate of breast cancer patient. Overall, our findings provide a novel molecular mechanism by which ZNF750 acts as tumour suppressor gene. Hence, we report a potential clinical relevance of ZNF750/RAC1 axis in breast cancer.
Collapse
Affiliation(s)
- Alessio Butera
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - Matteo Cassandri
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", 00133, Rome, Italy.,Department of Oncohematology, Bambino Gesu' Children's Hospital, 00146, Rome, Italy
| | - Francesco Rugolo
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - Massimiliano Agostini
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", 00133, Rome, Italy.
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", 00133, Rome, Italy.
| |
Collapse
|
21
|
Chen X, Yin L, Qiao G, Li Y, Li B, Bai Y, Feng F. Inhibition of Rac1 reverses enzalutamide resistance in castration-resistant prostate cancer. Oncol Lett 2020; 20:2997-3005. [PMID: 32782617 PMCID: PMC7400968 DOI: 10.3892/ol.2020.11823] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 02/13/2020] [Indexed: 12/24/2022] Open
Abstract
Enzalutamide, an androgen receptor inhibitor, has been clinically approved for the treatment of metastatic castration-resistant prostate cancer (CRPC) in the United States. However, patients only benefit from enzalutamide for a short period of time as resistance may develop. Therefore, it is vital to develop a novel strategy to overcome enzalutamide resistance. Ras-related C3 botulinum toxin substrate 1 (Rac1), which is commonly upregulated in human cancer types, has been recognized as a key molecular component in tumorigenesis, invasion and metastasis. However, the role of Rac1 in enzalutamide-resistance in prostate cancer (PCa) remains unknown. In the present study, Rac1 was demonstrated to be upregulated in enzalutamide-resistant PCa cells, and Rac1 knockdown inhibited enzalutamide-resistant cell proliferation and colony formation. Western blotting results indicated that enzalutamide treatment downregulated the expression levels of JNK and activated transcription factor 2, as well as enhanced the Bax/Bcl-2 ratio and induced cleavage of poly-ADP ribose polymerase. Moreover, knockdown of Rac1 in MR49F cells significantly inhibited cell migration and invasion via the downregulation of Snail and the upregulation of E-cadherin. The results of a nude mouse xenograft tumor model using 22RV1 cells demonstrated that enzalutamide inhibited tumor growth after Rac1 knockdown dramatically, compared to vehicle and single treatment groups. Therefore, the present study provided novel evidence that Rac1 may serve a crucial role in enzalutamide resistance, and that targeting Rac1 may be a potential approach for the treatment of CRPC.
Collapse
Affiliation(s)
- Xiaoliang Chen
- School of Medicine, Shanxi Datong University, Datong, Shanxi 037009, P.R. China
| | - Lili Yin
- School of Life Sciences, Shanxi Datong University, Datong, Shanxi 037009, P.R. China
| | - Gan Qiao
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Yanhua Li
- School of Medicine, Shanxi Datong University, Datong, Shanxi 037009, P.R. China
| | - Baoyuan Li
- School of Life Sciences, Shanxi Datong University, Datong, Shanxi 037009, P.R. China
| | - Yunfeng Bai
- Department of Chemistry, School of Chemistry and Environmental Engineering, Shanxi Datong University, Datong, Shanxi 037009, P.R. China
| | - Feng Feng
- Department of Chemistry, School of Chemistry and Environmental Engineering, Shanxi Datong University, Datong, Shanxi 037009, P.R. China
| |
Collapse
|
22
|
Mastelaro de Rezende M, Zenker Justo G, Julian Paredes-Gamero E, Gosens R. Wnt-5A/B Signaling in Hematopoiesis throughout Life. Cells 2020; 9:cells9081801. [PMID: 32751131 PMCID: PMC7465103 DOI: 10.3390/cells9081801] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 02/08/2023] Open
Abstract
Wnt signaling is well-known to play major roles in the hematopoietic system, from embryogenesis to aging and disease. In addition to the main β-catenin-dependent pathway, it is now clear that Wnt5a and the structurally related Wnt5b are essential for hematopoiesis, bone marrow colonization and the final steps of hematopoietic stem cell (HSC) maturation via β-catenin-independent signaling. Wnt5a and Wnt5b ligands prevent hematopoietic exhaustion (by maintaining quiescent, long-term HSCs), induce the proliferation of progenitors, and guide myeloid development, in addition to being involved in the development of aging-related alterations. The aim of this review is to summarize the current knowledge on these roles of Wnt5a and Wn5b signaling in the hematopoietic field.
Collapse
Affiliation(s)
- Marina Mastelaro de Rezende
- Departamento de Bioquímica, Universidade Federal de São Paulo (UNIFESP), São Paulo 04044-020, Brazil; (M.M.d.R.); (G.Z.J.); (E.J.P.-G.)
- Department of Molecular Pharmacology, University of Groningen, Groningen 9713 AV, The Netherlands
| | - Giselle Zenker Justo
- Departamento de Bioquímica, Universidade Federal de São Paulo (UNIFESP), São Paulo 04044-020, Brazil; (M.M.d.R.); (G.Z.J.); (E.J.P.-G.)
- Departamento de Ciências Farmacêuticas, Universidade Federal de São Paulo (UNIFESP), Diadema 09913-030, Brazil
| | - Edgar Julian Paredes-Gamero
- Departamento de Bioquímica, Universidade Federal de São Paulo (UNIFESP), São Paulo 04044-020, Brazil; (M.M.d.R.); (G.Z.J.); (E.J.P.-G.)
- Faculdade de Ciências Farmacêuticas, Universidade Federal de Mato Grosso do Sul, Campo Grande 79070-900, Brazil
| | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen, Groningen 9713 AV, The Netherlands
- Correspondence: ; Tel.: +31-50363-8177
| |
Collapse
|
23
|
Chan YL, Lai WC, Chen JS, Tseng JTC, Chuang PC, Jou J, Lee CT, Sun HS. TIAM2S Mediates Serotonin Homeostasis and Provokes a Pro-Inflammatory Immune Microenvironment Permissive for Colorectal Tumorigenesis. Cancers (Basel) 2020; 12:cancers12071844. [PMID: 32650570 PMCID: PMC7408714 DOI: 10.3390/cancers12071844] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 07/05/2020] [Accepted: 07/07/2020] [Indexed: 12/19/2022] Open
Abstract
The short isoform of human TIAM2 has been shown to promote proliferation and invasion in various cancer cells. However, the roles of TIAM2S in immune cells in relation to tumor development have not been investigated. To characterize the effects of TIAM2S, we generated TIAM2S-overexpressing mouse lines and found that aged TIAM2S-transgenic (TIAM2S-TG) developed significantly higher occurrence of lymphocytic infiltration and tumorigenesis in various organs, including colon. In addition, TIAM2S-TG is more sensitized to AOM-induced colon tumor development, suggesting a priming effect toward tumorigenesis. In the light of our recent findings that TIAM2S functions as a novel regulator of cellular serotonin level, we found that serotonin, in addition to Cox2, is a unique inflammation marker presented in the colonic lesion sites in the aged TG animals. Furthermore, our results demonstrated that ectopic TIAM2S altered immunity via the expansion of T lymphocytes; this was especially pronounced in CD8+ T cells in combination with CXCL13/BCA-1 pro-inflammatory chemokine in the serum of TIAM2S-TG mice. Consequently, T lymphocytes and B cells were recruited to the lesion sites and stimulated IL-23/IL17A expression to form the tertiary lymphoid organs. Collectively, our research suggests that TIAM2S provokes a pro-inflammatory immune microenvironment permissive to colorectal tumorigenesis through the serotonin-induced immunomodulatory effects.
Collapse
Affiliation(s)
- Ya-Ling Chan
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan 701, Taiwan
| | - Wei-Chung Lai
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Jia-Shing Chen
- School of Medicine for International Students, I-Shou University, Kaohsiung 840, Taiwan
| | - Joseph Ta-Chien Tseng
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan 701, Taiwan
| | - Pei-Chin Chuang
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Jonathan Jou
- Department of Surgery, University of Illinois, College of Medicine at Peoria, Peoria, IL 61605, USA
| | - Chung-Ta Lee
- Department of Pathology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - H Sunny Sun
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| |
Collapse
|
24
|
Rac1 activation in human breast carcinoma as a prognostic factor associated with therapeutic resistance. Breast Cancer 2020; 27:919-928. [PMID: 32314182 DOI: 10.1007/s12282-020-01091-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 04/02/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND RAS-related C3 botulinus toxin substrate 1 (Rac1) is a molecular switch fluctuating between GDP-bound inactive form (Rac1-GDP) and GTP-bound active form (Rac1-GTP) and involved in diverse function in both normal and malignant cells such as breast carcinoma cells. Although several studies have demonstrated immunolocalization of Rac1 protein in human breast carcinoma tissues, activation status of Rac1 still remains to be elucidated. METHODS We immunolocalized active form of Rac1 (Rac1-GTP) as well as total Rac1 using antibody specific for them in 115 invasive breast carcinoma tissues and correlated with clinicopathological parameters and clinical outcomes. RESULTS Rac1-GTP was frequently immunolocalized in the cytoplasm or cell membrane of breast carcinoma cells and it was positively correlated with Ki-67 labeling index and total Rac1 while negatively correlated with progesterone receptor. On the other hand, immunohistochemical Rac1-GTP status was significantly correlated with increased risk of recurrence and breast cancer-specific mortality of breast cancer patients and multivariate analyses did demonstrate Rac1-GTP as an independent worse prognostic factor for both disease-free and breast cancer-specific survival. In addition, Rac1-GTP was still correlated with worse prognosis in the patients who had received adjuvant chemotherapy or endocrine therapy. CONCLUSION These findings suggested Rac1 activation played pivotal roles in the progression and therapeutic resistance of breast cancers and Rac1 might be an important therapeutic target for improvement of the therapy for breast cancer patients.
Collapse
|
25
|
Zheng CW, Zeng RJ, Xu LY, Li EM. Rho GTPases: Promising candidates for overcoming chemotherapeutic resistance. Cancer Lett 2020; 475:65-78. [PMID: 31981606 DOI: 10.1016/j.canlet.2020.01.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/17/2020] [Accepted: 01/17/2020] [Indexed: 02/06/2023]
Abstract
Despite therapeutic advances, resistance to chemotherapy remains a major challenge to patients with malignancies. Rho GTPases are essential for the development and progression of various diseases including cancer, and a vast number of studies have linked Rho GTPases to chemoresistance. Therefore, understanding the underlying mechanisms can expound the effects of Rho GTPases towards chemotherapeutic agents, and targeting Rho GTPases is a promising strategy to downregulate the chemo-protective pathways and overcome chemoresistance. Importantly, exceptions in certain biological conditions and interactions among the members of Rho GTPases should be noted. In this review, we focus on the role of Rho GTPases, particularly Rac1, in regulating chemoresistance and provide an overview of their related mechanisms and available inhibitors, which may offer novel options for future targeted cancer therapy.
Collapse
Affiliation(s)
- Chun-Wen Zheng
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041, China; The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, 515041, China
| | - Rui-Jie Zeng
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041, China; The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, 515041, China
| | - Li-Yan Xu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041, China; Institute of Oncologic Pathology, Shantou University Medical College, Shantou, 515041, China.
| | - En-Min Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041, China; The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, 515041, China.
| |
Collapse
|
26
|
Maldonado MDM, Medina JI, Velazquez L, Dharmawardhane S. Targeting Rac and Cdc42 GEFs in Metastatic Cancer. Front Cell Dev Biol 2020; 8:201. [PMID: 32322580 PMCID: PMC7156542 DOI: 10.3389/fcell.2020.00201] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 03/09/2020] [Indexed: 12/20/2022] Open
Abstract
The Rho family GTPases Rho, Rac, and Cdc42 have emerged as key players in cancer metastasis, due to their essential roles in regulating cell division and actin cytoskeletal rearrangements; and thus, cell growth, migration/invasion, polarity, and adhesion. This review will focus on the close homologs Rac and Cdc42, which have been established as drivers of metastasis and therapy resistance in multiple cancer types. Rac and Cdc42 are often dysregulated in cancer due to hyperactivation by guanine nucleotide exchange factors (GEFs), belonging to both the diffuse B-cell lymphoma (Dbl) and dedicator of cytokinesis (DOCK) families. Rac/Cdc42 GEFs are activated by a myriad of oncogenic cell surface receptors, such as growth factor receptors, G-protein coupled receptors, cytokine receptors, and integrins; consequently, a number of Rac/Cdc42 GEFs have been implicated in metastatic cancer. Hence, inhibiting GEF-mediated Rac/Cdc42 activation represents a promising strategy for targeted metastatic cancer therapy. Herein, we focus on the role of oncogenic Rac/Cdc42 GEFs and discuss the recent advancements in the development of Rac and Cdc42 GEF-interacting inhibitors as targeted therapy for metastatic cancer, as well as their potential for overcoming cancer therapy resistance.
Collapse
Affiliation(s)
- Maria Del Mar Maldonado
- Department of Biochemistry, School of Medicine, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
| | - Julia Isabel Medina
- Department of Biochemistry, School of Medicine, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
| | - Luis Velazquez
- Department of Biochemistry, School of Medicine, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
| | - Suranganie Dharmawardhane
- Department of Biochemistry, School of Medicine, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
| |
Collapse
|
27
|
Li Q, Qin T, Bi Z, Hong H, Ding L, Chen J, Wu W, Lin X, Fu W, Zheng F, Yao Y, Luo ML, Saw PE, Wulf GM, Xu X, Song E, Yao H, Hu H. Rac1 activates non-oxidative pentose phosphate pathway to induce chemoresistance of breast cancer. Nat Commun 2020; 11:1456. [PMID: 32193458 PMCID: PMC7081201 DOI: 10.1038/s41467-020-15308-7] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 02/27/2020] [Indexed: 02/08/2023] Open
Abstract
Resistance development to one chemotherapeutic reagent leads frequently to acquired tolerance to other compounds, limiting the therapeutic options for cancer treatment. Herein, we find that overexpression of Rac1 is associated with multi-drug resistance to the neoadjuvant chemotherapy (NAC). Mechanistically, Rac1 activates aldolase A and ERK signaling which up-regulates glycolysis and especially the non-oxidative pentose phosphate pathway (PPP). This leads to increased nucleotides metabolism which protects breast cancer cells from chemotherapeutic-induced DNA damage. To translate this finding, we develop endosomal pH-responsive nanoparticles (NPs) which deliver Rac1-targeting siRNA together with cisplatin and effectively reverses NAC-chemoresistance in PDXs from NAC-resistant breast cancer patients. Altogether, our findings demonstrate that targeting Rac1 is a potential strategy to overcome acquired chemoresistance in breast cancer.
Collapse
Affiliation(s)
- Qingjian Li
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 510120, Guangzhou, People's Republic of China
| | - Tao Qin
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 510120, Guangzhou, People's Republic of China
| | - Zhuofei Bi
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 510120, Guangzhou, People's Republic of China
| | - Huangming Hong
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 510120, Guangzhou, People's Republic of China
| | - Lin Ding
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 510120, Guangzhou, People's Republic of China
| | - Jiewen Chen
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 510120, Guangzhou, People's Republic of China
| | - Wei Wu
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 510120, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 510120, Guangzhou, China
| | - Xiaorong Lin
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 510120, Guangzhou, People's Republic of China
| | - Wenkui Fu
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 510120, Guangzhou, People's Republic of China
| | - Fang Zheng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 510120, Guangzhou, China
| | - Yandan Yao
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 510120, Guangzhou, People's Republic of China
| | - Man-Li Luo
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 510120, Guangzhou, China
| | - Phei Er Saw
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 510120, Guangzhou, China
| | - Gerburg M Wulf
- Division of Hematology and Oncology, Beth Israel Deaconess Medical Center and Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Xiaoding Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 510120, Guangzhou, China
| | - Erwei Song
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 510120, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 510120, Guangzhou, China
| | - Herui Yao
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 510120, Guangzhou, People's Republic of China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 510120, Guangzhou, China.
| | - Hai Hu
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 510120, Guangzhou, People's Republic of China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 510120, Guangzhou, China.
| |
Collapse
|
28
|
Yang C, Ma C, Li Y, Mo P, Yang Y. High Tiam1 expression predicts positive lymphatic metastasis and worse survival in patients with malignant solid tumors: a systematic review and meta-analysis. Onco Targets Ther 2019; 12:5925-5936. [PMID: 31413590 PMCID: PMC6663076 DOI: 10.2147/ott.s191571] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 05/10/2019] [Indexed: 12/16/2022] Open
Abstract
Background Many studies have explored the prognostic value of T-cell lymphoma invasion and metastasis inducing factor 1 (Tiam1) and its association with lymphatic metastasis in malignant solid tumors, but the conclusions remain controversial. Therefore, we performed a meta-analysis to systematically assess the prognostic value of Tiam1 expression and its association with lymphatic metastasis in malignant solid tumors. Methods We searched eligible studies in PubMed, Web of Science and EMBASE databases (from inception up to October 2018). The combined HR with 95% CI was used to estimate the prognostic value of Tiam1 expression. The correlation between Tiam1 expression and lymphatic metastasis was assessed using the combined odds ratio (OR) with 95% CI. Results A total of 17 studies with 2,228 patients with solid tumors were included in this meta-analysis. The overall estimated results showed that high Tiam1 expression was significantly associated with shorter overall survival (HR= 2.08, 95% CI: 1.62-2.68, P<0.01), and disease-free survival (HR = 1.86, 95% CI: 1.49-2.32, P<0.01). Besides, we also found that there was a close relationship between high Tiam1 expression and positive lymphatic metastasis (OR=2.63; 95% CI: 1.79-3.84, P<0.01). Conclusion High Tiam1 expression was significantly associated with shorter survival and positive lymphatic metastasis in patients with malignant solid tumors. Therefore, Tiam1 may be a promising prognostic biomarker and an effective therapeutic target for malignant solid tumors.
Collapse
Affiliation(s)
- Caixia Yang
- Department of Stomatology, Qinghai province people's Hospital, Qinghai, People's Republic of China
| | - Chenlin Ma
- Department of Stomatology, Qinghai province people's Hospital, Qinghai, People's Republic of China
| | - Yingchun Li
- Department of Stomatology, Qinghai province people's Hospital, Qinghai, People's Republic of China
| | - Peng Mo
- Department of Stomatology, Qinghai province people's Hospital, Qinghai, People's Republic of China
| | - Yusheng Yang
- Department of Pathology, Ninbo Yinzhou No. 2 Hospital, Ninbo, People's Republic of China
| |
Collapse
|
29
|
Liu W, Chen G, Sun L, Zhang Y, Han J, Dai Y, He J, Shi S, Chen B. TUFT1 Promotes Triple Negative Breast Cancer Metastasis, Stemness, and Chemoresistance by Up-Regulating the Rac1/β-Catenin Pathway. Front Oncol 2019; 9:617. [PMID: 31338333 PMCID: PMC6629836 DOI: 10.3389/fonc.2019.00617] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 06/24/2019] [Indexed: 12/26/2022] Open
Abstract
Objectives: Triple negative breast cancer (TNBC) is a subtype of breast cancer with stronger invasion and metastasis, but its specific mechanism of action is still unclear. Tuft1 plays an important regulatory role in the survival of breast cancer cells; however, its role in regulating TNBC metastatic potential has not been well-characterized. Our aim was therefore to systematically study the mechanism of TUFT1 in the metastasis, stemness, and chemoresistance of TNBC and provide new predictors and targets for BC treatment. Methods: We used western blotting and IHC to measure TUFT1and Rac1-GTP expression levels in both human BC samples and cell lines. A combination of shRNA, migration/invasion assays, sphere formation assay, apoptosis assays, nude mouse xenograft tumor model, and GTP activity assays was used for further mechanistic studies. Results: We demonstrated that silencing TUFT1 in TNBC cells significantly inhibited cell metastasis and stemness in vitro. A nude mouse xenograft tumor model revealed that TUFT1 knockdown greatly decreased spontaneous lung metastasis of TNBC tumors. Mechanism studies showed that TUFT1 promoted tumor cell metastasis and stemness by up-regulating the Rac1/β-catenin pathway. Moreover, mechanistic studies indicated that the lack of TUFT1 expression in TNBC cells conferred more sensitive to chemotherapy and increased cell apoptosis via down-regulating the Rac1/β-catenin signaling pathway. Further, TUFT1 expression positively correlated with Rac1-GTP in TNBC samples, and co-expression of TUFT1 and Rac1-GTP predicted poor prognosis in TNBC patients who treated with chemotherapy. Conclusion: Our findings suggest that TUFT1/Rac1/β-catenin pathway may provide a potential target for more effective treatment of TNBC.
Collapse
Affiliation(s)
- Weiguang Liu
- Department of Breast Surgery, Affiliated Hospital of Hebei University of Engineering, Handan, China
| | - Guanglei Chen
- Department of Breast Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Lisha Sun
- Department of Breast Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yue Zhang
- Department of Physiology, Dalian Medical University, Dalian, China
| | - Jianjun Han
- Department of Breast Surgery, Affiliated Hospital of Hebei University of Engineering, Handan, China
| | - Yuna Dai
- Department of Breast Surgery, Affiliated Hospital of Hebei University of Engineering, Handan, China
| | - Jianchao He
- Department of Breast Surgery, Affiliated Hospital of Hebei University of Engineering, Handan, China
| | - Sufang Shi
- Department of Breast Surgery, Affiliated Hospital of Hebei University of Engineering, Handan, China
| | - Bo Chen
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
30
|
Kawai H, Matsushita H, Suzuki R, Kitamura Y, Ogawa Y, Kawada H, Ando K. Overcoming Tyrosine Kinase Inhibitor Resistance in Transformed Cell Harboring SEPT9-ABL1 Chimeric Fusion Protein. Neoplasia 2019; 21:788-801. [PMID: 31276931 PMCID: PMC6611969 DOI: 10.1016/j.neo.2019.06.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 12/14/2022] Open
Abstract
Hematological malignancies harboring various ABL1 fusions are expected to be sensitive to tyrosine kinase inhibitors (TKIs), similar to those with BCR-ABL1. However, SEPT9-ABL1 exhibits TKI resistance both in vitro and in vivo. SEPT9-ABL1 has the same ABL1 region as seen in BCR-ABL1 but no point mutation in its kinase domain, which is one of the main mechanisms underlying TKI resistance in the leukemic cells harboring BCR-ABL1. The purpose of this study was to reveal the mechanism underlying TKI resistance induced by SEPT9-ABL1. We focused on the TP53 status because TKI-induced apoptosis in BCR-ABL1–positive cells is achieved through TP53. Mouse TP53 homologue TRP53 was downregulated and less phosphorylated in the cells expressing SEPT9-ABL1 than in those with BCR-ABL1, resulting in the prevention of apoptosis induced by TKIs. The CRM1 inhibitor KPT-330 accumulated nuclear TRP53 and NFKB1A (also known as IκBα), which is thought to capture TRP53 in the cytoplasm, and induced apoptosis in the hematopoietic cells expressing SEPT9-ABL1. In addition, the combination treatment of KPT-330 and imatinib, which induced the marked nuclear accumulation of PP2A and SET, reactivated PP2A through its dephosphorylation and inhibited SET expression, resulting in the effective induction of the apoptosis in the cells expressing SEPT9-ABL1. The combination treatment with KPT-330 and imatinib successfully reduced the subcutaneous masses expressing SEPT9-ABL1 and extended the survival of the mice intraperitoneally transplanted with SEPT9-ABL1–expressing cells. These results show that therapy with CRM1 inhibitors may be effective for overcoming TKI resistance induced by SEPT9-ABL1.
Collapse
Affiliation(s)
- Hidetsugu Kawai
- Research Center for Cancer Stem Cell, Tokai University School of Medicine, Isehara, Kanagawa, Japan; Department of Hematology/Oncology, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Hiromichi Matsushita
- Research Center for Cancer Stem Cell, Tokai University School of Medicine, Isehara, Kanagawa, Japan; Division of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, Japan.
| | - Rikio Suzuki
- Research Center for Cancer Stem Cell, Tokai University School of Medicine, Isehara, Kanagawa, Japan; Department of Hematology/Oncology, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Yuka Kitamura
- Research Center for Cancer Stem Cell, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Yoshiaki Ogawa
- Department of Hematology/Oncology, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Hiroshi Kawada
- Research Center for Cancer Stem Cell, Tokai University School of Medicine, Isehara, Kanagawa, Japan; Department of Hematology/Oncology, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Kiyoshi Ando
- Research Center for Cancer Stem Cell, Tokai University School of Medicine, Isehara, Kanagawa, Japan; Department of Hematology/Oncology, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| |
Collapse
|
31
|
Ibrutinib induces chromatin reorganisation of chronic lymphocytic leukaemia cells. Oncogenesis 2019; 8:32. [PMID: 31076570 PMCID: PMC6510766 DOI: 10.1038/s41389-019-0142-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 04/01/2019] [Indexed: 12/12/2022] Open
Abstract
Chronic lymphocytic leukaemia (CLL) is the most common leukaemia in Western countries. It has recently been shown that the homogeneity of the chromatin landscape between CLL cells contrasts with the important observed genetic heterogeneity of the disease. To gain further insight into the consequences of disease evolution on the epigenome's plasticity, we monitored changes in chromatin structure occurring in vivo in CLL cells from patients receiving continuous Ibrutinib treatment. Ibrutinib, an oral inhibitor of the Bruton's tyrosine kinase (BTK) has proved to be remarkably efficient against treatment naïve (TN), heavily pre-treated and high-risk chronic lymphocytic leukaemia (CLL), with limited adverse events. We established that the chromatin landscape is significantly and globally affected in response to Ibrutinib. However, we observed that prior to treatment, CLL cells show qualitative and quantitative variations in chromatin structure correlated with both EZH2 protein level and cellular response to external stimuli. Then, under prolonged exposure to Ibrutinib, a loss of the two marks associated with lysine 27 (acetylation and trimethylation) was observed. Altogether, these data indicate that the epigenome of CLL cells from the peripheral blood change dynamically in response to stimuli and suggest that these cells might adapt to the Ibrutinib "hit" in a process leading toward a possible reduced sensitivity to treatment.
Collapse
|
32
|
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
|
33
|
Izumi D, Toden S, Ureta E, Ishimoto T, Baba H, Goel A. TIAM1 promotes chemoresistance and tumor invasiveness in colorectal cancer. Cell Death Dis 2019; 10:267. [PMID: 30890693 PMCID: PMC6425043 DOI: 10.1038/s41419-019-1493-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 02/18/2019] [Accepted: 02/28/2019] [Indexed: 12/21/2022]
Abstract
Accumulating evidence suggests that cancer cells with stem cell-like features have higher resistance to chemotherapeutic agents. Herein, we identified T-lymphoma invasion and metastasis-inducing protein-1 (TIAM1) as one of the Wnt-signaling associated genes which drives self-renewal and its expression is upregulated by cancer associated fibroblasts (CAFs). TIAM1 expression was assessed in resected colorectal cancer (CRC) tissues from 300 patients who did or did not respond to chemotherapy. siRNA and CRISPR/Cas9 was used to examine whether the inhibition of TIAM1 affects chemosensitivity of CRC. We demonstrate that stemness through Wnt signaling regulates chemosensitivity and this phenomenon occurs exclusively in cancer stem cells. Subsequently, we established patient-derived CAFs and tested whether the drug sensitivity of CRC cell lines is altered with CAF-derived conditioned medium. High-TIAM1 expression correlated significantly with poor prognosis of CRC patients, and was overexpressed in patients who did not respond to chemotherapy. We demonstrated that the inhibition of TIAM1 enhanced sensitivity to chemotherapeutic drugs and reduced tumor invasiveness in a series of experiments in vitro. Moreover, CAF-derived conditioned media increased stemness and chemoresistance in CRC cell lines through TIAM1 overexpression. In addition, we validated TIAM1 associated drug sensitivity using a xenograft model. We have demonstrated that TIAM1 is overexpressed in CRC tumors from patients who did not respond to chemotherapeutic drugs and levels of TIAM1 expression served as an independent prognostic factor. Mechanistically, CAFs enhanced CRC chemoresistance through TIAM1 overexpression. Collectively, these results suggest that TIAM1 regulates chemosensitivity in tumors and stroma and thus may be an attractive therapeutic target.
Collapse
Affiliation(s)
- Daisuke Izumi
- Center for Gastrointestinal Research, Center for Translational Genomics and Oncology, Baylor Scott & White Research Institute and Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, TX, USA.,Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,Department of Surgery, Japan Community Health Care Organization Kumamoto General Hospital, Yatsushiro, Japan
| | - Shusuke Toden
- Center for Gastrointestinal Research, Center for Translational Genomics and Oncology, Baylor Scott & White Research Institute and Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, TX, USA
| | - Elsie Ureta
- Center for Gastrointestinal Research, Center for Translational Genomics and Oncology, Baylor Scott & White Research Institute and Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, TX, USA
| | - Takatsugu Ishimoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,The International Research Center for Medicine Sciences, Kumamoto University, Kumamoto, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Ajay Goel
- Center for Gastrointestinal Research, Center for Translational Genomics and Oncology, Baylor Scott & White Research Institute and Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, TX, USA.
| |
Collapse
|
34
|
Durand-Onaylı V, Haslauer T, Härzschel A, Hartmann TN. Rac GTPases in Hematological Malignancies. Int J Mol Sci 2018; 19:ijms19124041. [PMID: 30558116 PMCID: PMC6321480 DOI: 10.3390/ijms19124041] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 12/11/2018] [Accepted: 12/12/2018] [Indexed: 12/22/2022] Open
Abstract
Emerging evidence suggests that crosstalk between hematologic tumor cells and the tumor microenvironment contributes to leukemia and lymphoma cell migration, survival, and proliferation. The supportive tumor cell-microenvironment interactions and the resulting cellular processes require adaptations and modulations of the cytoskeleton. The Rac subfamily of the Rho family GTPases includes key regulators of the cytoskeleton, with essential functions in both normal and transformed leukocytes. Rac proteins function downstream of receptor tyrosine kinases, chemokine receptors, and integrins, orchestrating a multitude of signals arising from the microenvironment. As such, it is not surprising that deregulation of Rac expression and activation plays a role in the development and progression of hematological malignancies. In this review, we will give an overview of the specific contribution of the deregulation of Rac GTPases in hematologic malignancies.
Collapse
Affiliation(s)
- Valerie Durand-Onaylı
- Department of Internal Medicine III with Hematology, Medical Oncology, Hemostaseology, Infectious Disease, Rheumatology, Oncologic Center, Salzburg Cancer Research Institute-Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), Paracelsus Medical University, Cancer Cluster Salzburg, 5020 Salzburg, Austria.
| | - Theresa Haslauer
- Department of Internal Medicine III with Hematology, Medical Oncology, Hemostaseology, Infectious Disease, Rheumatology, Oncologic Center, Salzburg Cancer Research Institute-Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), Paracelsus Medical University, Cancer Cluster Salzburg, 5020 Salzburg, Austria.
| | - Andrea Härzschel
- Department of Internal Medicine III with Hematology, Medical Oncology, Hemostaseology, Infectious Disease, Rheumatology, Oncologic Center, Salzburg Cancer Research Institute-Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), Paracelsus Medical University, Cancer Cluster Salzburg, 5020 Salzburg, Austria.
| | - Tanja Nicole Hartmann
- Department of Internal Medicine III with Hematology, Medical Oncology, Hemostaseology, Infectious Disease, Rheumatology, Oncologic Center, Salzburg Cancer Research Institute-Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), Paracelsus Medical University, Cancer Cluster Salzburg, 5020 Salzburg, Austria.
- Department of Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine and Medical Center, University of Freiburg, 79106 Freiburg, Germany.
| |
Collapse
|
35
|
Sondka Z, Bamford S, Cole CG, Ward SA, Dunham I, Forbes SA. The COSMIC Cancer Gene Census: describing genetic dysfunction across all human cancers. Nat Rev Cancer 2018; 18:696-705. [PMID: 30293088 PMCID: PMC6450507 DOI: 10.1038/s41568-018-0060-1] [Citation(s) in RCA: 978] [Impact Index Per Article: 139.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The Catalogue of Somatic Mutations in Cancer (COSMIC) Cancer Gene Census (CGC) is an expert-curated description of the genes driving human cancer that is used as a standard in cancer genetics across basic research, medical reporting and pharmaceutical development. After a major expansion and complete re-evaluation, the 2018 CGC describes in detail the effect of 719 cancer-driving genes. The recent expansion includes functional and mechanistic descriptions of how each gene contributes to disease generation in terms of the key cancer hallmarks and the impact of mutations on gene and protein function. These functional characteristics depict the extraordinary complexity of cancer biology and suggest multiple cancer-related functions for many genes, which are often highly tissue-dependent or tumour stage-dependent. The 2018 CGC encompasses a second tier, describing an expanding list of genes (currently 145) from more recent cancer studies that show supportive but less detailed indications of a role in cancer.
Collapse
Affiliation(s)
- Zbyslaw Sondka
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK.
- Open Targets, Wellcome Genome Campus, Hinxton, Cambridge, UK.
| | - Sally Bamford
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Charlotte G Cole
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Sari A Ward
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Ian Dunham
- Open Targets, Wellcome Genome Campus, Hinxton, Cambridge, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Simon A Forbes
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| |
Collapse
|
36
|
Hudson LG, Gillette JM, Kang H, Rivera MR, Wandinger-Ness A. Ovarian Tumor Microenvironment Signaling: Convergence on the Rac1 GTPase. Cancers (Basel) 2018; 10:cancers10100358. [PMID: 30261690 PMCID: PMC6211091 DOI: 10.3390/cancers10100358] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 09/25/2018] [Accepted: 09/25/2018] [Indexed: 02/06/2023] Open
Abstract
The tumor microenvironment for epithelial ovarian cancer is complex and rich in bioactive molecules that modulate cell-cell interactions and stimulate numerous signal transduction cascades. These signals ultimately modulate all aspects of tumor behavior including progression, metastasis and therapeutic response. Many of the signaling pathways converge on the small GTPase Ras-related C3 botulinum toxin substrate (Rac)1. In addition to regulating actin cytoskeleton remodeling necessary for tumor cell adhesion, migration and invasion, Rac1 through its downstream effectors, regulates cancer cell survival, tumor angiogenesis, phenotypic plasticity, quiescence, and resistance to therapeutics. In this review we discuss evidence for Rac1 activation within the ovarian tumor microenvironment, mechanisms of Rac1 dysregulation as they apply to ovarian cancer, and the potential benefits of targeting aberrant Rac1 activity in this disease. The potential for Rac1 contribution to extraperitoneal dissemination of ovarian cancer is addressed.
Collapse
Affiliation(s)
- Laurie G Hudson
- Department of Pharmaceutical Sciences, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| | - Jennifer M Gillette
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| | - Huining Kang
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
- Department of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| | - Melanie R Rivera
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| | - Angela Wandinger-Ness
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| |
Collapse
|
37
|
Mele S, Devereux S, Pepper AG, Infante E, Ridley AJ. Calcium-RasGRP2-Rap1 signaling mediates CD38-induced migration of chronic lymphocytic leukemia cells. Blood Adv 2018; 2:1551-1561. [PMID: 29970392 PMCID: PMC6039665 DOI: 10.1182/bloodadvances.2017014506] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 05/24/2018] [Indexed: 01/08/2023] Open
Abstract
CD38 is a transmembrane exoenzyme that is associated with poor prognosis in chronic lymphocytic leukemia (CLL). High CD38 levels in CLL cells are linked to increased cell migration, but the molecular basis is unknown. CD38 produces nicotinic acid adenine dinucleotide phosphate and adenosine 5'-diphosphate-ribose, both of which can act to increase intracellular Ca2+ levels. Here we show that CD38 expression increases basal intracellular Ca2+ levels and stimulates CLL cell migration both with and without chemokine stimulation. We find that CD38 acts via intracellular Ca2+ to increase the activity of the Ras family GTPase Rap1, which is in turn regulated by the Ca2+-sensitive Rap1 guanine-nucleotide exchange factor RasGRP2. Both Rap1 and RasGRP2 are required for CLL cell migration, and RasGRP2 is polarized in primary CLL cells with high CD38 levels. These results indicate that CD38 promotes RasGRP2/Rap1-mediated CLL cell adhesion and migration by increasing intracellular Ca2+ levels.
Collapse
Affiliation(s)
- Silvia Mele
- Randall Centre for Cell and Molecular Biophysics, and
- School of Cancer Sciences, King's College London, London, United Kingdom
| | - Stephen Devereux
- School of Cancer Sciences, King's College London, London, United Kingdom
| | - Andrea G Pepper
- School of Cancer Sciences, King's College London, London, United Kingdom
- Brighton and Sussex Medical School, University of Sussex, Brighton, United Kingdom; and
| | | | - Anne J Ridley
- Randall Centre for Cell and Molecular Biophysics, and
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
38
|
Bustelo XR. RHO GTPases in cancer: known facts, open questions, and therapeutic challenges. Biochem Soc Trans 2018; 46:741-760. [PMID: 29871878 PMCID: PMC7615761 DOI: 10.1042/bst20170531] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/17/2018] [Accepted: 05/03/2018] [Indexed: 02/06/2023]
Abstract
RHO GTPases have been traditionally associated with protumorigenic functions. While this paradigm is still valid in many cases, recent data have unexpectedly revealed that RHO proteins can also play tumor suppressor roles. RHO signaling elements can also promote both pro- and antitumorigenic effects using GTPase-independent mechanisms, thus giving an extra layer of complexity to the role of these proteins in cancer. Consistent with these variegated roles, both gain- and loss-of-function mutations in RHO pathway genes have been found in cancer patients. Collectively, these observations challenge long-held functional archetypes for RHO proteins in both normal and cancer cells. In this review, I will summarize these data and discuss new questions arising from them such as the functional and clinical relevance of the mutations found in patients, the mechanistic orchestration of those antagonistic functions in tumors, and the pros and cons that these results represent for the development of RHO-based anticancer drugs.
Collapse
Affiliation(s)
- Xosé R Bustelo
- Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, and Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
39
|
Cirmtuzumab inhibits ibrutinib-resistant, Wnt5a-induced Rac1 activation and proliferation in mantle cell lymphoma. Oncotarget 2018; 9:24731-24736. [PMID: 29872501 PMCID: PMC5973864 DOI: 10.18632/oncotarget.25340] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 03/21/2018] [Indexed: 02/02/2023] Open
Abstract
Cirmtuzumab may enhance the therapeutic activity of ibrutinib by inhibiting ROR1-dependent signaling pathway in patients with chronic lymphocytic leukemia (CLL). Mantle cell lymphoma (MCL) is B-cell malignancy that also expresses ROR1. In this study, we found that the plasma of patients with MCL had high levels of Wnt5a, a ROR1 ligand, that were comparable to those found in patients with CLL; in contrast Wnt5a was virtually undetectable in the plasma of age-matched healthy adults. We also found that Wnt5a induced Rac1 activation in the primary MCL cells. Cirmtuzumab, but not ibrutinib, could inhibit the capacity of Wnt5a to induce primary MCL cells to activate Rac1. Addition of exogenous Wnt5a in vitro significantly enhanced the numbers of MCL cell divisions and the proportion of dividing MCL cells entering S/G2 in MCL cells over time in the presence of CD154 and IL-4/10. Treatment of the MCL cells with cirmtuzumab, but not ibrutinib, blocked Wnt5a-enhanced proliferation of MCL cells. This study indicates that cirmtuzumab and ibrutinib may have complementary activity in the treatment of patients with MCL.
Collapse
|
40
|
Cardama GA, Alonso DF, Gonzalez N, Maggio J, Gomez DE, Rolfo C, Menna PL. Relevance of small GTPase Rac1 pathway in drug and radio-resistance mechanisms: Opportunities in cancer therapeutics. Crit Rev Oncol Hematol 2018; 124:29-36. [PMID: 29548483 DOI: 10.1016/j.critrevonc.2018.01.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 11/21/2017] [Accepted: 01/31/2018] [Indexed: 10/18/2022] Open
Abstract
Rac1 GTPase signaling pathway has a critical role in the regulation of a plethora of cellular functions governing cancer cell behavior. Recently, it has been shown a critical role of Rac1 in the emergence of resistance mechanisms to cancer therapy. This review describes the current knowledge regarding Rac1 pathway deregulation and its association with chemoresistance, radioresistance, resistance to targeted therapies and immune evasion. This supports the idea that interfering Rac1 signaling pathway could be an interesting approach to tackle cancer resistance.
Collapse
Affiliation(s)
- G A Cardama
- Laboratory of Molecular Oncology, National University of Quilmes, Buenos Aires, Argentina
| | - D F Alonso
- Laboratory of Molecular Oncology, National University of Quilmes, Buenos Aires, Argentina; National Council of Scientific and Technical Research (CONICET), Buenos Aires, Argentina
| | - N Gonzalez
- Laboratory of Molecular Oncology, National University of Quilmes, Buenos Aires, Argentina
| | - J Maggio
- Laboratory of Molecular Oncology, National University of Quilmes, Buenos Aires, Argentina
| | - D E Gomez
- Laboratory of Molecular Oncology, National University of Quilmes, Buenos Aires, Argentina; National Council of Scientific and Technical Research (CONICET), Buenos Aires, Argentina
| | - C Rolfo
- Phase I-Early Clinical trials Unit, Oncology Department Antwerp University Hospital & Center for Oncological Research (CORE), Antwerp University, Belgium.
| | - P L Menna
- Laboratory of Molecular Oncology, National University of Quilmes, Buenos Aires, Argentina; National Council of Scientific and Technical Research (CONICET), Buenos Aires, Argentina
| |
Collapse
|
41
|
Kou W, Xu X, Ji S, Chen M, Liu D, Wang K, Zhuang J, Yu Q, Zhao Q, Xu Y, Zhang H, Peng W. The inhibition of the effect and mechanism of vascular intimal hyperplasia in Tiam1 knockout mice. Biochem Biophys Res Commun 2018; 497:248-255. [DOI: 10.1016/j.bbrc.2018.02.065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 02/07/2018] [Indexed: 12/21/2022]
|
42
|
Rac1 plays a role in CXCL12 but not CCL3-induced chemotaxis and Rac1 GEF inhibitor NSC23766 has off target effects on CXCR4. Cell Signal 2018; 42:88-96. [DOI: 10.1016/j.cellsig.2017.10.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 10/11/2017] [Accepted: 10/13/2017] [Indexed: 12/17/2022]
|
43
|
Ikram M, Lim Y, Baek SY, Jin S, Jeong YH, Kwak JY, Yoon S. Co-targeting of Tiam1/Rac1 and Notch ameliorates chemoresistance against doxorubicin in a biomimetic 3D lymphoma model. Oncotarget 2017; 9:2058-2075. [PMID: 29416753 PMCID: PMC5788621 DOI: 10.18632/oncotarget.23156] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 11/16/2017] [Indexed: 12/11/2022] Open
Abstract
Lymphoma is a heterogeneous disease with a highly variable clinical course and prognosis. Improving the prognosis for patients with relapsed and treatment-resistant lymphoma remains challenging. Current in vitro drug testing models based on 2D cell culture lack natural tissue-like structural organization and result in disappointing clinical outcomes. The development of efficient drug testing models using 3D cell culture that more accurately reflects in vivo behaviors is vital. Our aim was to establish an in vitro 3D lymphoma model that can imitate the in vivo 3D lymphoma microenvironment. Using this model, we explored strategies to enhance chemosensitivity to doxorubicin, an important chemotherapeutic drug widely used for the treatment of hematological malignancies. Lymphoma cells grown in this model exhibited excellent biomimetic properties compared to conventional 2D culture including (1) enhanced chemotherapy resistance, (2) suppressed rate of apoptosis, (3) upregulated expression of drug resistance genes (MDR1, MRP1, BCRP and HIF-1α), (4) elevated levels of tumor aggressiveness factors including Notch (Notch-1, -2, -3, and -4) and its downstream molecules (Hes-1 and Hey-1), VEGF and MMPs (MMP-2 and MMP-9), and (5) enrichment of a lymphoma stem cell population. Tiam1, a potential biomarker of tumor progression, metastasis, and chemoresistance, was activated in our 3D lymphoma model. Remarkably, we identified two synergistic therapeutic oncotargets, Tiam1 and Notch, as a strategy to combat resistance against doxorubicin in EL4 T and A20 B lymphoma. Therefore, our data suggest that our 3D lymphoma model is a promising in vitro research platform for studying lymphoma biology and therapeutic approaches.
Collapse
Affiliation(s)
- Muhammad Ikram
- Department of Anatomy, Pusan National University School of Medicine, Yangsan 50612, Korea
| | - Yeseon Lim
- Department of Anatomy, Pusan National University School of Medicine, Yangsan 50612, Korea
| | - Sun-Yong Baek
- Department of Anatomy, Pusan National University School of Medicine, Yangsan 50612, Korea
| | - Songwan Jin
- Department of Mechanical Engineering, Korea Polytechnic University, Siheung 15073, Korea
| | - Young Hun Jeong
- Department of Mechanical Engineering, Kyungpook National University, Daegu 41566, Korea
| | - Jong-Young Kwak
- Department of Pharmacology, Ajou University School of Medicine, Suwon 16499, Korea
| | - Sik Yoon
- Department of Anatomy, Pusan National University School of Medicine, Yangsan 50612, Korea
| |
Collapse
|
44
|
Janovská P, Bryja V. Wnt signalling pathways in chronic lymphocytic leukaemia and B-cell lymphomas. Br J Pharmacol 2017; 174:4701-4715. [PMID: 28703283 PMCID: PMC5727250 DOI: 10.1111/bph.13949] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 06/19/2017] [Accepted: 06/29/2017] [Indexed: 12/31/2022] Open
Abstract
In this review, we discuss the intricate roles of the Wnt signalling network in the development and progression of mature B-cell-derived haematological malignancies, with a focus on chronic lymphocytic leukaemia (CLL) and related B-cell lymphomas. We review the current literature and highlight the differences between the β-catenin-dependent and -independent branches of Wnt signalling. Special attention is paid to the role of the non-canonical Wnt/planar cell polarity (PCP) pathway, mediated by the Wnt-5-receptor tyrosine kinase-like orphan receptor (ROR1)-Dishevelled signalling axis in CLL. This is mainly because the Wnt/PCP co-receptor ROR1 was found to be overexpressed in CLL and the Wnt/PCP pathway contributes to numerous aspects of CLL pathogenesis. We also discuss the possibilities of therapeutically targeting the Wnt signalling pathways as an approach to disrupt the crucial interaction between malignant cells and their micro-environment. We also advocate the need for research in this direction for other lymphomas, namely, diffuse large B-cell lymphoma, Hodgkin lymphoma, mantle cell lymphoma, Burkitt lymphoma and follicular lymphoma where the Wnt signalling pathway probably plays a similar role. LINKED ARTICLES This article is part of a themed section on WNT Signalling: Mechanisms and Therapeutic Opportunities. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.24/issuetoc.
Collapse
Affiliation(s)
- Pavlína Janovská
- Institute of Experimental Biology, Faculty of ScienceMasaryk UniversityBrnoCzech Republic
| | - Vítězslav Bryja
- Institute of Experimental Biology, Faculty of ScienceMasaryk UniversityBrnoCzech Republic
| |
Collapse
|
45
|
Namasu CY, Katzerke C, Bräuer-Hartmann D, Wurm AA, Gerloff D, Hartmann JU, Schwind S, Müller-Tidow C, Hilger N, Fricke S, Christopeit M, Niederwieser D, Behre G. ABR, a novel inducer of transcription factor C/EBPα, contributes to myeloid differentiation and is a favorable prognostic factor in acute myeloid leukemia. Oncotarget 2017; 8:103626-103639. [PMID: 29262589 PMCID: PMC5732755 DOI: 10.18632/oncotarget.22093] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 09/22/2017] [Indexed: 11/29/2022] Open
Abstract
Active BCR related (ABR) gene deactivates ras-related C3 botulinum toxin substrate 1 (RAC1), which plays an essential role in regulating normal hematopoiesis and in leukemia. BCR gene, closely related to ABR, acts as a tumor suppressor in chronic myeloid leukemia and has overlapping functions with ABR. Evidence for a putative tumor suppressor role of ABR has been shown in several solid tumors, in which deletion of ABR is present. Our results show downregulation of ABR in AML. A block of ABR prevents myeloid differentiation and leads to repression of the myeloid transcription factor C/EBPα, a major regulator of myeloid differentiation and functionally impaired in leukemia. Conversely, stable overexpression of ABR enhances myeloid differentiation. Inactivation of the known ABR target RAC1 by treatment with the RAC1 inhibitor NSC23766 resulted in an increased expression of C/EBPα in primary AML samples and in AML cell lines U937 and MV4;11. Finally, AML patients with high ABR expression at diagnosis showed a significant longer overall survival and patients who respond to azacitidine therapy showed a significant higher ABR expression. This is the first report showing that ABR expression plays a critical role in both myelopoiesis and AML. Our data indicate the tumor suppressor potential of ABR and underline its potential role in leukemia therapeutic strategies.
Collapse
Affiliation(s)
| | - Christiane Katzerke
- Division of Hematology and Oncology, University Hospital Leipzig, Leipzig, Germany
| | | | | | - Dennis Gerloff
- Division of Dermatology and Venereology, University Hospital Halle, Halle, Germany
| | - Jens-Uwe Hartmann
- Division of Hematology and Oncology, University Hospital Leipzig, Leipzig, Germany
| | - Sebastian Schwind
- Division of Hematology and Oncology, University Hospital Leipzig, Leipzig, Germany
| | - Carsten Müller-Tidow
- Division of Hematology and Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Nadja Hilger
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Stephan Fricke
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Maximilian Christopeit
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dietger Niederwieser
- Division of Hematology and Oncology, University Hospital Leipzig, Leipzig, Germany
| | - Gerhard Behre
- Division of Hematology and Oncology, University Hospital Leipzig, Leipzig, Germany
| |
Collapse
|
46
|
Kazanietz MG, Caloca MJ. The Rac GTPase in Cancer: From Old Concepts to New Paradigms. Cancer Res 2017; 77:5445-5451. [PMID: 28807941 DOI: 10.1158/0008-5472.can-17-1456] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 06/29/2017] [Accepted: 08/01/2017] [Indexed: 01/24/2023]
Abstract
Rho family GTPases are critical regulators of cellular functions that play important roles in cancer progression. Aberrant activity of Rho small G-proteins, particularly Rac1 and their regulators, is a hallmark of cancer and contributes to the tumorigenic and metastatic phenotypes of cancer cells. This review examines the multiple mechanisms leading to Rac1 hyperactivation, particularly focusing on emerging paradigms that involve gain-of-function mutations in Rac and guanine nucleotide exchange factors, defects in Rac1 degradation, and mislocalization of Rac signaling components. The unexpected pro-oncogenic functions of Rac GTPase-activating proteins also challenged the dogma that these negative Rac regulators solely act as tumor suppressors. The potential contribution of Rac hyperactivation to resistance to anticancer agents, including targeted therapies, as well as to the suppression of antitumor immune response, highlights the critical need to develop therapeutic strategies to target the Rac pathway in a clinical setting. Cancer Res; 77(20); 5445-51. ©2017 AACR.
Collapse
Affiliation(s)
- Marcelo G Kazanietz
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Maria J Caloca
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas, Universidad de Valladolid, Valladolid, Spain.
| |
Collapse
|
47
|
Yu J, Chen L, Chen Y, Hasan MK, Ghia EM, Zhang L, Wu R, Rassenti LZ, Widhopf GF, Shen Z, Briggs SP, Kipps TJ. Wnt5a induces ROR1 to associate with 14-3-3ζ for enhanced chemotaxis and proliferation of chronic lymphocytic leukemia cells. Leukemia 2017; 31:2608-2614. [PMID: 28465528 PMCID: PMC5670032 DOI: 10.1038/leu.2017.132] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 03/03/2017] [Accepted: 03/21/2017] [Indexed: 11/09/2022]
Abstract
Wnt5a can activate Rho GTPases in chronic lymphocytic leukemia (CLL) cells by inducing the recruitment of ARHGEF2 to ROR1. Mass spectrometry on immune precipitates of Wnt5a-activated ROR1 identified 14-3-3ζ, which was confirmed by co-immunoprecipitation. The capacity of Wnt5a to induce ROR1 to complex with 14-3-3ζ could be blocked in CLL cells by treatment with cirmtuzumab, a humanized mAb targeting ROR1. Silencing 14-3-3ζ via small interfering RNA impaired the capacity of Wnt5a to: (1) induce recruitment of ARHGEF2 to ROR1, (2) enhance in vitro exchange activity of ARHGEF2 and (3) induce activation of RhoA and Rac1 in CLL cells. Furthermore, CRISPR/Cas9 deletion of 14-3-3ζ in ROR1-negative CLL cell-line MEC1, and in MEC1 cells transfected to express ROR1 (MEC1-ROR1), demonstrated that 14-3-3ζ was necessary for the growth/engraftment advantage of MEC1-ROR1 over MEC1 cells. We identified a binding motif (RSPS857SAS) in ROR1 for 14-3-3ζ. Site-directed mutagenesis of ROR1 demonstrated that serine-857 was required for the recruitment of 14-3-3ζ and ARHGEF2 to ROR1, and activation of RhoA and Rac1. Collectively, this study reveals that 14-3-3ζ plays a critical role in Wnt5a/ROR1 signaling, leading to enhanced CLL migration and proliferation.
Collapse
Affiliation(s)
- J Yu
- Moores Cancer Center, University of California-San Diego, La Jolla, CA, USA
| | - L Chen
- Moores Cancer Center, University of California-San Diego, La Jolla, CA, USA
| | - Y Chen
- Moores Cancer Center, University of California-San Diego, La Jolla, CA, USA
| | - M K Hasan
- Moores Cancer Center, University of California-San Diego, La Jolla, CA, USA
| | - E M Ghia
- Moores Cancer Center, University of California-San Diego, La Jolla, CA, USA
| | - L Zhang
- Moores Cancer Center, University of California-San Diego, La Jolla, CA, USA
| | - R Wu
- Moores Cancer Center, University of California-San Diego, La Jolla, CA, USA
| | - L Z Rassenti
- Moores Cancer Center, University of California-San Diego, La Jolla, CA, USA
| | - G F Widhopf
- Moores Cancer Center, University of California-San Diego, La Jolla, CA, USA
| | - Z Shen
- Section of Cell and Developmental Biology, University of California-San Diego, La Jolla, CA, USA
| | - S P Briggs
- Section of Cell and Developmental Biology, University of California-San Diego, La Jolla, CA, USA
| | - T J Kipps
- Moores Cancer Center, University of California-San Diego, La Jolla, CA, USA
| |
Collapse
|
48
|
Yu J, Chen L, Cui B, Wu C, Choi MY, Chen Y, Zhang L, Rassenti LZ, Widhopf Ii GF, Kipps TJ. Cirmtuzumab inhibits Wnt5a-induced Rac1 activation in chronic lymphocytic leukemia treated with ibrutinib. Leukemia 2016; 31:1333-1339. [PMID: 27904138 PMCID: PMC5462858 DOI: 10.1038/leu.2016.368] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 10/24/2016] [Accepted: 11/04/2016] [Indexed: 12/15/2022]
Abstract
Signaling via the B cell receptor (BCR) plays an important role in the pathogenesis and progression of chronic lymphocytic leukemia (CLL). This is underscored by the clinical effectiveness of ibrutinib, an inhibitor of Bruton's tyrosine kinase (BTK) that can block BCR-signaling. However, ibrutinib cannot induce complete responses (CR) or durable remissions without continued therapy, suggesting alternative pathways also contribute to CLL growth/survival that are independent of BCR-signaling. ROR1 is a receptor for Wnt5a, which can promote activation of Rac1 to enhance CLL-cell proliferation and survival. In this study, we found that CLL cells of patients treated with ibrutinib had activated Rac1. Moreover, Wnt5a could induce Rac1 activation and enhance proliferation of CLL cells treated with ibrutinib at concentrations that were effective in completely inhibiting BTK and BCR-signaling. Wnt5a-induced Rac1 activation could be blocked by cirmtuzumab (UC-961), an anti-ROR1 mAb. We found that treatment with cirmtuzumab and ibrutinib was significantly more effective than treatment with either agent alone in clearing leukemia cells in vivo. This study indicates that cirmtuzumab may enhance the activity of ibrutinib in the treatment of patients with CLL or other ROR1+ B-cell malignancies.
Collapse
Affiliation(s)
- J Yu
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - L Chen
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - B Cui
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Christina Wu
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - M Y Choi
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Y Chen
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - L Zhang
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - L Z Rassenti
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - G F Widhopf Ii
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - T J Kipps
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
49
|
Morath I, Hartmann T, Orian-Rousseau V. CD44: More than a mere stem cell marker. Int J Biochem Cell Biol 2016; 81:166-173. [DOI: 10.1016/j.biocel.2016.09.009] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 09/05/2016] [Accepted: 09/10/2016] [Indexed: 01/15/2023]
|
50
|
Kurdi AT, Bassil R, Olah M, Wu C, Xiao S, Taga M, Frangieh M, Buttrick T, Orent W, Bradshaw EM, Khoury SJ, Elyaman W. Tiam1/Rac1 complex controls Il17a transcription and autoimmunity. Nat Commun 2016; 7:13048. [PMID: 27725632 PMCID: PMC5062600 DOI: 10.1038/ncomms13048] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 08/30/2016] [Indexed: 12/14/2022] Open
Abstract
RORγt is a master transcription factor of Th17 cells and considered as a promising drug target for the treatment of autoimmune diseases. Here, we show the guanine nucleotide exchange factor, Tiam1, and its cognate Rho-family G protein, Rac1, regulate interleukin (IL)17A transcription and autoimmunity. Whereas Tiam1 genetic deficiency weakens IL-17A expression partially and inhibits the development of experimental autoimmune encephalomyelitis (EAE), deletion of Rac1 in T cells exhibits more robust effects on Th17 cells and EAE. We demonstrate Tiam1 and Rac1 form a complex with RORγt in the nuclear compartment of Th17 cells, and together bind and activate the Il17 promoter. The clinical relevance of these findings is emphasized by pharmacological targeting of Rac1 that suppresses both murine and human Th17 cells as well as EAE. Thus, our findings highlight a regulatory pathway of Tiam1/Rac1 in Th17 cells and suggest that it may be a therapeutic target in multiple sclerosis. Tiam1 is a guanine nucleotide exchange factor for the Rho-family GTPase Rac1. Here, the authors show that nuclear Tiam1 and Rac1 bind to RORγt on the IL-17 promoter, activating its transcription, and that inhibiting Tiam1/Rac1 is beneficial in a mouse model of autoimmunity.
Collapse
Affiliation(s)
- Ahmed T Kurdi
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Ribal Bassil
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Marta Olah
- Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Broad Institute at Harvard University and MIT, Boston, Massachusetts 02115, USA
| | - Chuan Wu
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Sheng Xiao
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Mariko Taga
- Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Broad Institute at Harvard University and MIT, Boston, Massachusetts 02115, USA
| | - Michael Frangieh
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Thomas Buttrick
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - William Orent
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Elizabeth M Bradshaw
- Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Broad Institute at Harvard University and MIT, Boston, Massachusetts 02115, USA
| | - Samia J Khoury
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA.,Abu Haidar Neuroscience Institute, American University of Beirut Medical Center, Riad El Solh, Beirut 1107 2020, Lebanon
| | - Wassim Elyaman
- Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Broad Institute at Harvard University and MIT, Boston, Massachusetts 02115, USA
| |
Collapse
|