1
|
Zha Z, Ge F, Li N, Zhang S, Wang C, Gong F, Miao J, Chen W. Effects of Na V1.5 and Rac1 on the Epithelial-Mesenchymal Transition in Breast Cancer. Cell Biochem Biophys 2025; 83:1483-1494. [PMID: 39673684 DOI: 10.1007/s12013-024-01625-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2024] [Indexed: 12/16/2024]
Abstract
Breast cancer is a disease that seriously endangers the health of women. However, it is difficult to treat due to the emergence of metastasis and drug resistance. Exploring the metastasis mechanism of breast cancer is helpful to aim for the appropriate target. The epithelial-mesenchymal transition (EMT) is an important mechanism of breast cancer metastasis. Sodium channel 1.5(NaV1.5) and the GTPase Rac1 are factors related to the degree of malignancy of breast tumors. The expression of NaV1.5 and the activation of Rac1 are both involved in EMT. In addition, NaV1.5 can change the plasma membrane potential (Vm) by promoting the inflow of Na+ to depolarize the cell membrane, induce the activation of Rac1 and produce a cascade of reactions that lead to EMT in breast cancer cells; this sequence of events further induces the movement, migration and invasion of tumor cells and affects the prognosis of breast cancer patients. In this paper, the roles of NaV1.5 and Rac1 in EMT-mediated breast cancer progression were reviewed.
Collapse
Affiliation(s)
- Zhuocen Zha
- First-Class Discipline Team of Kunming Medical University, Third Department of Breast Surgery, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
- Oncology department, Guizhou Hospital of the First Affiliated Hospital, Sun Yat-sen University, Guiyang, Guizhou, 550000, China
| | - Fei Ge
- Department of Breast Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Na Li
- First-Class Discipline Team of Kunming Medical University, Third Department of Breast Surgery, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| | - Shijun Zhang
- First-Class Discipline Team of Kunming Medical University, Third Department of Breast Surgery, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| | - Chenxi Wang
- First-Class Discipline Team of Kunming Medical University, Third Department of Breast Surgery, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| | - Fuhong Gong
- First-Class Discipline Team of Kunming Medical University, Third Department of Breast Surgery, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| | - Jingge Miao
- First-Class Discipline Team of Kunming Medical University, Third Department of Breast Surgery, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| | - Wenlin Chen
- First-Class Discipline Team of Kunming Medical University, Third Department of Breast Surgery, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China.
| |
Collapse
|
2
|
Li D, Huang Y, Lei H, Huang X. DOCK1/ELMO1/Rac1 Signaling is Essential for Vitreous-Induced Migration and Contraction of ARPE19 Cells. J Ocul Pharmacol Ther 2025; 41:217-225. [PMID: 39911025 DOI: 10.1089/jop.2024.0173] [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] [Indexed: 02/07/2025] Open
Abstract
Purpose: To test the effects of dedicator of cytokinesis protein 1 (DOCK1) with its binding partner engulfment and cell motility protein 1 (ELMO1)-Rac1 axis on the vitreous-induced biological functions of retinal pigment epithelial (RPE) cells. Methods: Rac1 activity in RPE cells after vitreous stimulation was detected via a pull-down assay. The related protein expression levels were examined via western blot analysis. DOCK1 and ELMO1 knockdown cells were generated via CRISPR-Cas9 technology. Cytoskeletal reorganization was detected by immunofluorescent localization of F-actin. Cell proliferation, migration, invasion, and contraction ability were measured via the CCK8 assay, wound healing assay, transwell invasion assay, and collagen contraction assay. Results: Rac1 activity was significantly elevated in ARPE-19 cells stimulated with vitreous fluid for 30 min to 3 h. Depletion of either DOCK1 or ELMO1 with CRISPR/Cas9 attenuated vitreous-stimulated Rac1 activity, thus reversing the vitreous-induced cytoskeletal rearrangements. The functional cell biology results revealed that deficiencies of DOCK1 and ELMO1 significantly impeded the migration, invasion, and contraction abilities of vitreous-stimulated human RPE cells. Conclusion: This study demonstrated that the DOCK1/ELMO1-Rac1 axis plays an essential role in the pathogenesis of proliferative vitreoretinopathy (PVR), thus suggesting that interruption of this axis has potential for PVR therapy.
Collapse
Affiliation(s)
- Duo Li
- Department of Ophthalmology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Department of Emergency Surgery, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Yikeng Huang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hetian Lei
- Department of Ophthalmology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Xionggao Huang
- Department of Ophthalmology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Department of Emergency Surgery, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital, Hainan Medical University, Haikou, China
| |
Collapse
|
3
|
Chang CC, Peng M, Keppeke GD, Tsai LK, Zhang Z, Pai LM, Sung LY, Liu JL. Y12C mutation disrupts IMPDH cytoophidia and alters cancer metabolism. FEBS J 2025. [PMID: 40186514 DOI: 10.1111/febs.70086] [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: 03/06/2024] [Revised: 12/17/2024] [Accepted: 03/25/2025] [Indexed: 04/07/2025]
Abstract
Guanosine triphosphate (GTP) is a building block for DNA and RNA, and plays a pivotal role in various cellular functions, serving as an energy source, enzyme cofactor and a key component of signal transduction. The activity of the rate-limiting enzyme in de novo GTP synthesis, inosine monophosphate dehydrogenase (IMPDH), is regulated by nucleotide binding. Recent studies have illuminated that IMPDH octamers can assemble into linear polymers, adding another dimension to its enzymatic regulation. This polymerisation reduces IMPDH's sensitivity to the inhibitory effects of GTP binding, thereby augmenting its activity under conditions with elevated GTP levels. Within cells, IMPDH polymers may cluster to form the distinctive structure known as the cytoophidium, which is postulated to reflect the cellular demand for increased GTP concentrations. Nevertheless, the functional significance of IMPDH polymerisation in in vivo metabolic regulation remains unclear. In this study, we report the widespread presence of IMPDH cytoophidia in various human cancer tissues. Utilising the ABEmax base editor, we introduced a Y12C point mutation into IMPDH2 across multiple cancer cell lines. This mutation disrupts the polymerisation interface of IMPDH and prevents cytoophidium assembly. In some cancer xenografts, the absence of IMPDH polymers led to a downregulation of IMPDH, as well as the glycolytic and pentose phosphate pathways. Furthermore, mutant HeLa-cell-derived xenografts were notably smaller than their wild-type counterparts. Our data suggest that IMPDH polymerisation and cytoophidium assembly could be instrumental in modulating metabolic homeostasis in certain cancers, offering insights into the clinical relevance of IMPDH cytoophidium.
Collapse
Affiliation(s)
- Chia-Chun Chang
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Min Peng
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Gerson Dierley Keppeke
- School of Life Science and Technology, ShanghaiTech University, China
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| | - Li-Kuang Tsai
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Ziheng Zhang
- School of Life Science and Technology, ShanghaiTech University, China
| | - Li-Mei Pai
- Department of Biochemistry and Molecular Biology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Li-Ying Sung
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
- Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
- Center for Biotechnology, National Taiwan University, Taipei, Taiwan
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| | - Ji-Long Liu
- School of Life Science and Technology, ShanghaiTech University, China
- Department of Physiology, Anatomy and Genetics, University of Oxford, UK
| |
Collapse
|
4
|
Li D, Linghu M, Tang J, Yang G, Li C, Yao H, Lei H, Huang Y, Huang X. Leveraging AAV1-Rac1T17N to prevent experimental proliferative vitreoretinopathy. J Transl Med 2025; 23:374. [PMID: 40140939 PMCID: PMC11948691 DOI: 10.1186/s12967-025-06391-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 03/17/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Platelet-derived growth factor receptor β (PDGFRβ) is the principal PDGFR isoform in retinal pigment epithelial (RPE) cells from the epiretinal membranes of patients with proliferative vitreoretinopathy (PVR). Ras-related C3 Botulinum toxin substrate 1 (Rac1), a member of the Rho family, is a crucial factor in the cell migration and contraction processes that are inherent to the pathogenesis of PVR. The mutants Rac1T17N and Rac1Q61L can block and promote Rac1 activation, respectively. The major objective of this research was to ascertain whether PDGFRβ mediates vitreous-induced Rac1 activation and whether Rac1T17N could be leveraged for the prevention of PVR pathogenesis in a rabbit model. METHODS A pull-down assay was used to examine GTP Rac1 levels, which are indicative of Rac1 activation, and western blotting was used to assess cellular protein expression. A CCK8 assay, a wound healing assay, a transwell invasion assay and a collagen contraction assay were employed to analyze cell proliferation, migration, invasion and contraction capacity, respectively. A PVR model was created by injecting platelet-rich plasma and human retinal pigment epithelial cells (ARPE-19) into the vitreous cavities of rabbits, and this model was used to evaluate the severity of PVR impacted by intravitreally injected ARPE-19 cells transduced with adeno-associated virus (AAV)1-Rac1T17N or Rac1Q61L. PVR grade was evaluated by a double-blinded investigator according to the Fastenberg classification; in addition, ultrasound and histological analyses were performed to assess PVR severity. RESULTS Vitreous-induced GTP Rac1 is mediated by PDGFRβ. There was a significant decrease in vitreous-induced GTP Rac1 in ARPE-19 cells transduced with AAV1-Rac1T17N compared with those transduced with AAV1-GFP. In addition, the suppression of GTP Rac1 production in human RPE cells by transduction with AAV1-Rac1T17N inhibited vitreous-induced proliferation, migration, invasion, and contractility. Importantly, the results of the animal experiments indicated that although there was a significant increase in PVR potential in rabbits intravitreally injected with ARPE-19 cells infected with AAV1-Rac1Q61L, there was a significant decrease in PVR potential in rabbits intravitreally injected with ARPE-19 cells infected with AAV1-Rac1T17N (P < 0.01). CONCLUSIONS AAV1-Rac1T17N has great potential for PVR therapy.
Collapse
Affiliation(s)
- Duo Li
- Department of Ophthalmology, The First Affiliated Hospital of Hainan Medical University, Haikou, 570102, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Department of Emergency Surgery, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital, Hainan Medical University, Haikou, 570102, China
| | - Minli Linghu
- Department of Ophthalmology, The First Affiliated Hospital of Hainan Medical University, Haikou, 570102, China
| | - Jisen Tang
- Department of Ophthalmology, The First Affiliated Hospital of Hainan Medical University, Haikou, 570102, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Department of Emergency Surgery, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital, Hainan Medical University, Haikou, 570102, China
| | - Gukun Yang
- Department of Ophthalmology, The First Affiliated Hospital of Hainan Medical University, Haikou, 570102, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Department of Emergency Surgery, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital, Hainan Medical University, Haikou, 570102, China
| | - Chuanwu Li
- Department of Ophthalmology, The First Affiliated Hospital of Hainan Medical University, Haikou, 570102, China
| | - Hang Yao
- Department of Ophthalmology, The First Affiliated Hospital of Hainan Medical University, Haikou, 570102, China
| | - Hetian Lei
- Department of Ophthalmology, The First Affiliated Hospital of Hainan Medical University, Haikou, 570102, China.
| | - Yikeng Huang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| | - Xionggao Huang
- Department of Ophthalmology, The First Affiliated Hospital of Hainan Medical University, Haikou, 570102, China.
- Key Laboratory of Emergency and Trauma of Ministry of Education, Department of Emergency Surgery, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital, Hainan Medical University, Haikou, 570102, China.
| |
Collapse
|
5
|
da Silva Fernandes T, Gillard BM, Dai T, Martin JC, Chaudhry KA, Dugas SM, Fisher AA, Sharma P, Wu R, Attwood KM, Dasgupta S, Takabe K, Rosario SR, Bianchi-Smiraglia A. Inosine monophosphate dehydrogenase 2 (IMPDH2) modulates response to therapy and chemo-resistance in triple negative breast cancer. Sci Rep 2025; 15:1061. [PMID: 39774345 PMCID: PMC11707137 DOI: 10.1038/s41598-024-85094-5] [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: 10/27/2024] [Accepted: 12/31/2024] [Indexed: 01/11/2025] Open
Abstract
Triple negative breast cancer (TNBC) is one of the deadliest subtypes of breast cancer, whose high frequency of relapse is often due to resistance to chemotherapy. Here, we identify inosine monophosphate dehydrogenase 2 (IMPDH2) as a contributor to doxorubicin resistance, in multiple TNBC models. Analysis of publicly available datasets reveals elevated IMPDH2 expression to associate with worse overall TNBC prognosis in the clinic, including lower recurrence-free survival post adjuvant/neoadjuvant therapy. Importantly, both genetic depletion and pharmacological inhibition of IMPDH2 leads to reduction of pro-tumorigenic phenotypes in multiple doxorubicin-resistant TNBC models, both in vitro and in vivo. Overall, we propose IMPDH2 as a novel vulnerability that could be leveraged therapeutically to suppress and/or prevent the growth of chemo-resistant lesions.
Collapse
Affiliation(s)
- Tatiane da Silva Fernandes
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, CGP L3-317, Buffalo, NY, 14263, USA
| | - Bryan M Gillard
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Tao Dai
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, CGP L3-317, Buffalo, NY, 14263, USA
| | - Jeffrey C Martin
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, CGP L3-317, Buffalo, NY, 14263, USA
| | - Kanita A Chaudhry
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, CGP L3-317, Buffalo, NY, 14263, USA
| | - Scott M Dugas
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, CGP L3-317, Buffalo, NY, 14263, USA
| | - Alyssa A Fisher
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, CGP L3-317, Buffalo, NY, 14263, USA
| | - Pia Sharma
- Department of Breast Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - RongRong Wu
- Department of Breast Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Kristopher M Attwood
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, RSC R-410, Buffalo, NY, 14263, USA
| | - Subhamoy Dasgupta
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, CGP L3-317, Buffalo, NY, 14263, USA
| | - Kazuaki Takabe
- Department of Breast Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Spencer R Rosario
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, RSC R-410, Buffalo, NY, 14263, USA.
| | - Anna Bianchi-Smiraglia
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, CGP L3-317, Buffalo, NY, 14263, USA.
| |
Collapse
|
6
|
Nie Y, Lu X, Zhu Y, Shi Y, Ren K, Li Z, Chen P, Han D, Li X. Circular Adhesion Substrates Inhibiting Cell Polarization and Proliferation via Graded Texture of Geometric Micropatterns. SMALL METHODS 2024:e2401471. [PMID: 39564718 DOI: 10.1002/smtd.202401471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/06/2024] [Indexed: 11/21/2024]
Abstract
Most melanomas that occur on the skin surface originate from a newly formed nevus and grow outward in a circular pattern and metastasize from the nevus center. Herein, a circular microfabricated substrate is constructed to explore the growth behavior of melanoma cells. Modeling software is used to calculate appropriate parameters, including shape and size, and then the substrates are processed with microfabrication technologies. The results show that the melanoma cells on the circular adhesion substrate are oval and are significant changes in cell spread length, nuclei, area, aspect ratio, Young's modulus, and orientation angles, indicating inhibition of cell polarization. Moreover, three different layers from circular adhesion substrates are selected to construct new substrates, which indicates that the polarization degree of cells is closely related to the number of micropillar arrays on the circular geometric substrate. In addition, flow cytometry demonstrates that the circular substrate reduced the transition from resting/gap 1 phase (G0/G1) to synthesis phase (S phase), thereby decreasing DNA synthesis and proliferation, reminding a potential method for treatment strategy. More importantly, the circular adhesion substrate influences the integrin signaling pathway, which has a potential application and research prospect in the treatment of melanoma.
Collapse
Affiliation(s)
- Yifeng Nie
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
| | - Xi Lu
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, P. R. China
| | - Yuting Zhu
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, P. R. China
| | - Yahong Shi
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100193, P. R. China
| | - Keli Ren
- The Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Zhongxian Li
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Peipei Chen
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
| | - Dong Han
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xiang Li
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
| |
Collapse
|
7
|
Zhao P, Zhang J, Song W, Qi D, Huang Y, Su Y, Wu R, Zhang L, Zhang S. Incarvine C and its analogues inhibit the formation of cell cytoskeleton by targeting Rac1. Bioorg Chem 2024; 149:107512. [PMID: 38833990 DOI: 10.1016/j.bioorg.2024.107512] [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: 03/01/2024] [Revised: 05/17/2024] [Accepted: 05/30/2024] [Indexed: 06/06/2024]
Abstract
Ras-related C3 botulinum toxin substrate 1 (Rac1) has emerged as a key regulator in the treatment of cancer metastasis because of its involvement in the formation of cell plate pseudopods and effects on cell migration. In this study, we found that incarvine C, a natural product isolated from Incarvillea sinensis, and its seven analogues exhibited antitumour activity by inhibiting cell cytoskeleton formation, with moderate cytotoxicity. Accordingly, these compounds inhibited the cytoskeleton-mediated migration and invasion of MDA-MB-231 cells, with inhibition rates ranging from 37.30 % to 69.72 % and 51.27 % to 70.90 % in vitro, respectively. Moreover, they induced G2/M phase cell cycle arrest in MDA-MB-231 cells. A pull-down assay revealed that the interaction between Rac1 and its downstream effector protein PAK1 was inhibited by these compounds and that the compound Ano-6 exhibited substantial activity, with an inhibition rate of more than 90 %. Molecular docking showed that incarvine C and its analogues could bind to the nucleotide-binding pocket of Rac1, maintaining high levels of inactivated Rac1. As Ano-6 exhibited significant activity in vitro, its anti-cancer activity was tested in vivo. Four weeks of oral treatment with Ano-6 was well-tolerated in mice, and it induced a potential anti-tumour response in xenografts of MDA-MB-231 cells. Further studies demonstrated that Ano-6 was enriched in tumour tissues after 2 h of administration and induced an increase in the number of dead tumour cells. In summary, these findings not only reveal the mechanism of incarvine C but also provide a new molecular template for Rac1 inhibitors and identify a promising candidate for breast cancer treatment.
Collapse
Affiliation(s)
- Pengxiang Zhao
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, 251 Ningda Road, Xining 810016, Qinghai, China; Department of Pharmacy, Medical College of Qinghai University, 16 Kunlun Road, Xining 810016, Qinghai, China
| | - Jie Zhang
- Qinghai University Affiliated Hospital, 29 Tongren Road, Xining 810016, Qinghai, China
| | - Weirong Song
- Department of Pharmacy, Medical College of Qinghai University, 16 Kunlun Road, Xining 810016, Qinghai, China
| | - Danshi Qi
- Department of Pharmacy, Medical College of Qinghai University, 16 Kunlun Road, Xining 810016, Qinghai, China
| | - Yongchun Huang
- Department of Pharmacy, Medical College of Qinghai University, 16 Kunlun Road, Xining 810016, Qinghai, China
| | - Yudong Su
- Department of Pharmacy, Medical College of Qinghai University, 16 Kunlun Road, Xining 810016, Qinghai, China
| | - Rumeng Wu
- Department of Pharmacy, Medical College of Qinghai University, 16 Kunlun Road, Xining 810016, Qinghai, China
| | - Lirong Zhang
- Department of Pharmacy, Medical College of Qinghai University, 16 Kunlun Road, Xining 810016, Qinghai, China
| | - Shoude Zhang
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, 251 Ningda Road, Xining 810016, Qinghai, China; Department of Pharmacy, Medical College of Qinghai University, 16 Kunlun Road, Xining 810016, Qinghai, China.
| |
Collapse
|
8
|
Zhou J, Zhang M, Gao A, Herman JG, Guo M. Epigenetic silencing of KCTD8 promotes hepatocellular carcinoma growth by activating PI3K/AKT signaling. Epigenomics 2024; 16:929-944. [PMID: 39023358 PMCID: PMC11370965 DOI: 10.1080/17501911.2024.2370590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 06/14/2024] [Indexed: 07/20/2024] Open
Abstract
Aim: The aim of current study is to explore the epigenetic changes and function of KCTD8 in human hepatocellular carcinoma (HCC). Materials & methods: HCC cell lines and tissue samples were employed. Methylation specific PCR, flow cytometry, immunoprecipitation and xenograft mouse models were used.Results: KCTD8 was methylated in 44.83% (104/232) of HCC and its methylation may act as an independent poor prognostic marker. KCTD8 expression was regulated by DNA methylation. KCTD8 suppressed HCC cell growth both in vitro and in vivo via inhibiting PI3K/AKT pathway.Conclusion: Methylation of KCTD8 is an independent poor prognostic marker, and epigenetic silencing of KCTD8 increases the malignant tendency in HCC.
Collapse
Affiliation(s)
- Jing Zhou
- School of Medicine, NanKai University, Tianjin, 300071, China
- Department of Gastroenterology & Hepatology, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Meiying Zhang
- Department of Gastroenterology & Hepatology, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Aiai Gao
- Department of Gastroenterology & Hepatology, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - James G Herman
- The Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA15213, USA
| | - Mingzhou Guo
- School of Medicine, NanKai University, Tianjin, 300071, China
- Department of Gastroenterology & Hepatology, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
- National Key Laboratory of Kidney Diseases, Beijing, 100853, China
| |
Collapse
|
9
|
Ruan X, Xiong Y, Li X, Yang E, Wang J. Lower ratio of IMPDH1 to IMPDH2 sensitizes gliomas to chemotherapy. Cancer Gene Ther 2024; 31:1081-1089. [PMID: 38871858 DOI: 10.1038/s41417-024-00793-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 05/24/2024] [Accepted: 05/29/2024] [Indexed: 06/15/2024]
Abstract
Gliomas are the most common primary tumors of the central nervous system, with approximately half of patients presenting with the most aggressive form of glioblastoma. Although several molecular markers for glioma have been identified, they are not sufficient to predict the prognosis due to the extensive genetic heterogeneity within glioma. Our study reveals that the ratio of IMPDH1 to IMPDH2 expression levels serves as a molecular indicator for glioma treatment prognosis. Patients with a higher IMPDH1/IMPDH2 ratio exhibit a worse prognosis, while those with a lower ratio display a more favorable prognosis. We further demonstrate that IMPDH1 plays a crucial role in maintaining cellular GTP/GDP levels following DNA damage compared to IMPDH2. In the absence of IMPDH1, cells experience an imbalance in the GTP/GDP ratio, impairing DNA damage repair capabilities and rendering them more sensitive to TMZ. This study not only introduces a novel prognostic indicator for glioma clinical diagnosis but also offers innovative insights for precise and stratified glioma treatment.
Collapse
Affiliation(s)
- Xiaoyu Ruan
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University International Cancer Institute, Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, 100191, Beijing, China
| | - Yundong Xiong
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University International Cancer Institute, Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, 100191, Beijing, China
| | - Xiaoman Li
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University International Cancer Institute, Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, 100191, Beijing, China.
| | - Ence Yang
- Department of Medical Bioinformatics, Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China.
| | - Jiadong Wang
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University International Cancer Institute, Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, 100191, Beijing, China.
- Department of Gastrointestinal Translational Research, Peking University Cancer Hospital, 100142, Beijing, China.
| |
Collapse
|
10
|
Scott KL, Halfmann CT, Hoefakker AD, Purkayastha P, Wang TC, Lele TP, Roux KJ. Nucleocytoplasmic transport rates are regulated by cellular processes that modulate GTP availability. J Cell Biol 2024; 223:e202308152. [PMID: 38683248 PMCID: PMC11059771 DOI: 10.1083/jcb.202308152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 03/08/2024] [Accepted: 04/01/2024] [Indexed: 05/01/2024] Open
Abstract
Nucleocytoplasmic transport (NCT), the facilitated diffusion of cargo molecules between the nucleus and cytoplasm through nuclear pore complexes (NPCs), enables numerous fundamental eukaryotic cellular processes. Ran GTPase uses cellular energy in the direct form of GTP to create a gradient across the nuclear envelope (NE) that drives the majority of NCT. We report here that changes in GTP availability resulting from altered cellular physiology modulate the rate of NCT, as monitored using synthetic and natural cargo, and the dynamics of Ran itself. Cell migration, cell spreading, and/or modulation of the cytoskeleton or its connection to the nucleus alter GTP availability and thus rates of NCT, regulating RNA export and protein synthesis. These findings support a model in which changes in cellular physiology that alter GTP availability can regulate the rate of NCT, impacting fundamental cellular processes that extensively utilize NCT.
Collapse
Affiliation(s)
- Kelsey L. Scott
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD, USA
| | | | - Allison D. Hoefakker
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD, USA
- Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
| | - Purboja Purkayastha
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Ting Ching Wang
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Tanmay P. Lele
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
- Department of Translational Medical Sciences, Texas A&M University, Houston, TX, USA
| | - Kyle J. Roux
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD, USA
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, USA
| |
Collapse
|
11
|
Wang L, Yang R, Kong Y, Zhou J, Chen Y, Li R, Chen C, Tang X, Chen X, Xia J, Chen X, Cheng B, Ren X. Integrative single-cell and bulk transcriptomes analyses reveals heterogeneity of serine-glycine-one-carbon metabolism with distinct prognoses and therapeutic vulnerabilities in HNSCC. Int J Oral Sci 2024; 16:44. [PMID: 38886346 PMCID: PMC11183126 DOI: 10.1038/s41368-024-00310-2] [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: 10/25/2023] [Revised: 04/03/2024] [Accepted: 04/28/2024] [Indexed: 06/20/2024] Open
Abstract
Metabolic heterogeneity plays a central role in sustaining uncontrolled cancer cell proliferation and shaping the tumor microenvironment (TME), which significantly compromises the clinical outcomes and responses to therapy in head and neck squamous cell carcinoma (HNSCC) patients. This highlights the urgent need to delineate the intrinsic heterogeneity and biological roles of metabolic vulnerabilities to advance precision oncology. The metabolic heterogeneity of malignant cells was identified using single-cell RNA sequencing (scRNA-seq) profiles and validated through bulk transcriptomes. Serine-glycine-one-carbon (SGOC) metabolism was screened out to be responsible for the aggressive malignant properties and poor prognosis in HNSCC patients. A 4-SGOC gene prognostic signature, constructed by LASSO-COX regression analysis, demonstrated good predictive performance for overall survival and therapeutic responses. Patients in the low-risk group exhibited greater infiltration of exhausted CD8+ T cells, and demonstrated better clinical outcomes after receiving immunotherapy and chemotherapy. Conversely, high-risk patients exhibited characteristics of cold tumors, with enhanced IMPDH1-mediated purine biosynthesis, resulting in poor responses to current therapies. IMPDH1 emerged as a potential therapeutic metabolic target. Treatment with IMPDH inhibitors effectively suppressed HNSCC cell proliferation and metastasis and induced apoptosis in vitro and in vivo by triggering GTP-exhaustion nucleolar stress. Our findings underscore the metabolic vulnerabilities of HNSCC in facilitating accurate patient stratification and individualized precise metabolic-targeted treatment.
Collapse
Affiliation(s)
- Lixuan Wang
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Rongchun Yang
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Yue Kong
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Jing Zhou
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Yingyao Chen
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Rui Li
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chuwen Chen
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Xinran Tang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaobing Chen
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Juan Xia
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Xijuan Chen
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Bin Cheng
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China.
| | - Xianyue Ren
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
12
|
Wu Y, Liu W, Li J, Shi H, Ma S, Wang D, Pan B, Xiao R, Jiang H, Liu X. Decreased Tiam1-mediated Rac1 activation is responsible for impaired directional persistence of chondrocyte migration in microtia. J Cell Mol Med 2024; 28:e18443. [PMID: 38837873 PMCID: PMC11149491 DOI: 10.1111/jcmm.18443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/06/2024] [Accepted: 05/11/2024] [Indexed: 06/07/2024] Open
Abstract
The human auricle has a complex structure, and microtia is a congenital malformation characterized by decreased size and loss of elaborate structure in the affected ear with a high incidence. Our previous studies suggest that inadequate cell migration is the primary cytological basis for the pathogenesis of microtia, however, the underlying mechanism is unclear. Here, we further demonstrate that microtia chondrocytes show a decreased directional persistence during cell migration. Directional persistence can define a leading edge associated with oriented movement, and any mistakes would affect cell function and tissue morphology. By the screening of motility-related genes and subsequent confirmations, active Rac1 (Rac1-GTP) is identified to be critical for the impaired directional persistence of microtia chondrocytes migration. Moreover, Rho guanine nucleotide exchange factors (GEFs) and Rho GTPase-activating proteins (GAPs) are detected, and overexpression of Tiam1 significantly upregulates the level of Rac1-GTP and improves directional migration in microtia chondrocytes. Consistently, decreased expression patterns of Tiam1 and active Rac1 are found in microtia mouse models, Bmp5se/J and Prkralear-3J/GrsrJ. Collectively, our results provide new insights into microtia development and therapeutic strategies of tissue engineering for microtia patients.
Collapse
Affiliation(s)
- Yi Wu
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Wei Liu
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Jia Li
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Hang Shi
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Shize Ma
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Di Wang
- Department of Auricular Reconstruction, Plastic Surgery Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Bo Pan
- Department of Auricular Reconstruction, Plastic Surgery Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Ran Xiao
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
- Key Laboratory of External Tissue and Organ Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Haiyue Jiang
- Department of Auricular Reconstruction, Plastic Surgery Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Xia Liu
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
- Key Laboratory of External Tissue and Organ Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
13
|
Toyoda S, Handa T, Yong H, Takahashi H, Shiwaku H. IMPDH2 forms spots at branching sites and distal ends of astrocyte stem processes. Genes Cells 2024; 29:150-158. [PMID: 38009721 DOI: 10.1111/gtc.13088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/12/2023] [Accepted: 11/15/2023] [Indexed: 11/29/2023]
Abstract
Inosine monophosphate dehydrogenase (IMPDH) is a rate-limiting enzyme in the de novo GTP biosynthesis pathway. Recent studies suggest that IMPDH2, an isoform of IMPDH, can localize to specific subcellular compartments under certain conditions and regulate site-specific GTP availability and small GTPase activity in invasive cancer cells. However, it is unclear whether IMPDH2 plays a site-specific regulatory role in subcellular functions in healthy cells. In this study, we focused on brain cells and examined the localization pattern of IMPDH2. We discovered that IMPDH2 forms localized spots in the astrocytes of the adult mouse hippocampus. Further analysis of spot distribution in primary astrocyte cultures revealed that IMPDH2 spots are predominantly localized on branching sites and distal ends of astrocyte stem processes. Our findings suggest a potential unidentified role for IMPDH2 and GTP synthesis specifically at specialized nodes of astrocyte branches.
Collapse
Affiliation(s)
- Saori Toyoda
- Department of Psychiatry and Behavioral Sciences, Tokyo Medical and Dental University Graduate School, Tokyo, Japan
| | - Takehisa Handa
- Department of Psychiatry and Behavioral Sciences, Tokyo Medical and Dental University Graduate School, Tokyo, Japan
| | - Huang Yong
- Department of Neuropathology, Medical Research Institute and Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hidehiko Takahashi
- Department of Psychiatry and Behavioral Sciences, Tokyo Medical and Dental University Graduate School, Tokyo, Japan
| | - Hiroki Shiwaku
- Department of Psychiatry and Behavioral Sciences, Tokyo Medical and Dental University Graduate School, Tokyo, Japan
| |
Collapse
|
14
|
Zhou W, Zhao Z, Lin A, Yang JZ, Xu J, Wilder-Romans K, Yang A, Li J, Solanki S, Speth JM, Walker N, Scott AJ, Wang L, Wen B, Andren A, Zhang L, Kothari AU, Yao Y, Peterson ER, Korimerla N, Werner CK, Ullrich A, Liang J, Jacobson J, Palavalasa S, O’Brien AM, Elaimy AL, Ferris SP, Zhao SG, Sarkaria JN, Győrffy B, Zhang S, Al-Holou WN, Umemura Y, Morgan MA, Lawrence TS, Lyssiotis CA, Peters-Golden M, Shah YM, Wahl DR. GTP Signaling Links Metabolism, DNA Repair, and Responses to Genotoxic Stress. Cancer Discov 2024; 14:158-175. [PMID: 37902550 PMCID: PMC10872631 DOI: 10.1158/2159-8290.cd-23-0437] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 09/07/2023] [Accepted: 10/19/2023] [Indexed: 10/31/2023]
Abstract
How cell metabolism regulates DNA repair is incompletely understood. Here, we define a GTP-mediated signaling cascade that links metabolism to DNA repair and has significant therapeutic implications. GTP, but not other nucleotides, regulates the activity of Rac1, a guanine nucleotide-binding protein, which promotes the dephosphorylation of serine 323 on Abl-interactor 1 (Abi-1) by protein phosphatase 5 (PP5). Dephosphorylated Abi-1, a protein previously not known to activate DNA repair, promotes nonhomologous end joining. In patients and mouse models of glioblastoma, Rac1 and dephosphorylated Abi-1 mediate DNA repair and resistance to standard-of-care genotoxic treatments. The GTP-Rac1-PP5-Abi-1 signaling axis is not limited to brain cancer, as GTP supplementation promotes DNA repair and Abi-1-S323 dephosphorylation in nonmalignant cells and protects mouse tissues from genotoxic insult. This unexpected ability of GTP to regulate DNA repair independently of deoxynucleotide pools has important implications for normal physiology and cancer treatment. SIGNIFICANCE A newly described GTP-dependent signaling axis is an unexpected link between nucleotide metabolism and DNA repair. Disrupting this pathway can overcome cancer resistance to genotoxic therapy while augmenting it can mitigate genotoxic injury of normal tissues. This article is featured in Selected Articles from This Issue, p. 5.
Collapse
Affiliation(s)
- Weihua Zhou
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Zitong Zhao
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Department of Oncology, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shan Xi, PR China
| | - Angelica Lin
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - John Z Yang
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Jie Xu
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Kari Wilder-Romans
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Annabel Yang
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Jing Li
- Cell Signaling Technology, Inc., Danvers, MA, USA
| | - Sumeet Solanki
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Jennifer M Speth
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Natalie Walker
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Andrew J Scott
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Lu Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA
| | - Bo Wen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA
| | - Anthony Andren
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Li Zhang
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Ayesha U Kothari
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Yangyang Yao
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Department of Oncology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, PR China
| | - Erik R Peterson
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Navyateja Korimerla
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Christian K Werner
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Alexander Ullrich
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Jessica Liang
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Janna Jacobson
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Sravya Palavalasa
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Alexandra M O’Brien
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Ameer L Elaimy
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Sean P Ferris
- Department of Pathology, Division of Neuropathology, University of Michigan, Ann Arbor, MI, USA
| | - Shuang G Zhao
- Department of Human Oncology, University of Wisconsin Madison, WI, USA
| | | | - Balázs Győrffy
- Department of Bioinformatics, Semmelweis University, Budapest, Hungary; and TTK Cancer Biomarker Research Group, Institute of Enzymology, Budapest, Hungary
| | - Shuqun Zhang
- Department of Oncology, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shan Xi, PR China
| | - Wajd N Al-Holou
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, United States
| | - Yoshie Umemura
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Meredith A Morgan
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Theodore S Lawrence
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Costas A Lyssiotis
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology and Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Marc Peters-Golden
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Yatrik M Shah
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Daniel R Wahl
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
- Lead contact
| |
Collapse
|
15
|
Scott KL, Halfmann CT, Hoefakker AD, Purkayastha P, Wang TC, Lele TP, Roux KJ. Nucleocytoplasmic transport rates are regulated by cellular processes that modulate GTP availability. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.29.573651. [PMID: 38234722 PMCID: PMC10793428 DOI: 10.1101/2023.12.29.573651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Nucleocytoplasmic transport (NCT), the facilitated diffusion of cargo molecules between the nucleus and cytoplasm through nuclear pore complexes (NPCs), enables numerous fundamental eukaryotic cellular processes. Ran GTPase uses cellular energy in the direct form of GTP to create a gradient across the nuclear envelope (NE) that drives the majority of NCT. We report here that changes in GTP availability resulting from altered cellular physiology modulate the rate of NCT, as monitored using synthetic and natural cargo, and the dynamics of Ran itself. Cell migration, cell spreading and/or modulation of the cytoskeleton or its connection to the nucleus alter GTP availability and thus rates of NCT, regulating RNA export and protein synthesis. These findings support a model in which changes in cellular physiology that alter GTP availability can regulate the rate of NCT, impacting fundamental cellular processes that extensively utilize NCT.
Collapse
Affiliation(s)
- Kelsey L. Scott
- Enabling Technologies Group, Sanford Research, Sioux Falls SD
| | | | - Allison D. Hoefakker
- Enabling Technologies Group, Sanford Research, Sioux Falls SD
- Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD
| | - Purboja Purkayastha
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas
| | - Ting Ching Wang
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas
| | - Tanmay P. Lele
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas
- Department of Translational Medical Sciences, Texas A&M University, Houston, Texas
| | - Kyle J. Roux
- Enabling Technologies Group, Sanford Research, Sioux Falls SD
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD
| |
Collapse
|
16
|
Fongang B, Wadop YN, Zhu Y, Wagner EJ, Kudlicki A, Rowicka M. Coevolution combined with molecular dynamics simulations provides structural and mechanistic insights into the interactions between the integrator complex subunits. Comput Struct Biotechnol J 2023; 21:5686-5697. [PMID: 38074468 PMCID: PMC10700540 DOI: 10.1016/j.csbj.2023.11.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 11/10/2023] [Accepted: 11/10/2023] [Indexed: 01/18/2024] Open
Abstract
Finding the 3D structure of large, multi-subunit complexes is difficult, despite recent advances in cryo-EM technology, due to remaining challenges to expressing and purifying subunits. Computational approaches that predict protein-protein interactions, including Direct Coupling Analysis (DCA), represent an attractive alternative for dissecting interactions within protein complexes. However, they are readily applicable only to small proteins due to high computational complexity and a high number of false positives. To solve this problem, we proposed a modified DCA approach, a powerful tool to predict the most likely interfaces of protein complexes. Since our modified approach cannot provide structural and mechanistic details of interacting peptides, we combine it with Molecular Dynamics (MD) simulations. To illustrate this novel approach, we predict interacting domains and structural details of interactions of two Integrator complex subunits, INTS9 and INTS11. Our predictions of interacting residues of INTS9/INTS11 are highly consistent with crystallographic structure. We then expand our procedure to two complexes whose structures are not well-studied: 1) The heterodimer formed by the Cleavage and Polyadenylation Specificity Factor 100-kD (CPSF100) and 73-kD (CPSF73); 2) The heterotrimer formed by INTS4/INTS9/INTS11. Experimental data supports our predictions of interactions within these two complexes, demonstrating that combining DCA and MD simulations is a powerful approach to revealing structural insights of large protein complexes.
Collapse
Affiliation(s)
- Bernard Fongang
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- Department of Biochemistry and Structural Biology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- Department of Population Health Sciences, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- Institute for Translational Sciences, The University of Texas Medical Branch, Galveston, TX, United States
| | - Yannick N. Wadop
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- Institute for Translational Sciences, The University of Texas Medical Branch, Galveston, TX, United States
| | - Yingjie Zhu
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, United States
- Institute for Translational Sciences, The University of Texas Medical Branch, Galveston, TX, United States
| | - Eric J. Wagner
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, United States
- Department of Biochemistry and Biophysics, The University of Rochester Medical Center, Rochester, NY, United States
- Institute for Translational Sciences, The University of Texas Medical Branch, Galveston, TX, United States
| | - Andrzej Kudlicki
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, United States
- Institute for Translational Sciences, The University of Texas Medical Branch, Galveston, TX, United States
- Informatics Service Center, The University of Texas Medical Branch, Galveston, TX, United States
| | - Maga Rowicka
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, United States
- Institute for Translational Sciences, The University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
17
|
Chaudhry KA, Jacobi JJ, Gillard BM, Karasik E, Martin JC, da Silva Fernandes T, Hurley E, Feltri ML, Attwood KM, Twist CJ, Smiraglia DJ, Long MD, Bianchi-Smiraglia A. Aryl hydrocarbon receptor is a tumor promoter in MYCN-amplified neuroblastoma cells through suppression of differentiation. iScience 2023; 26:108303. [PMID: 38026169 PMCID: PMC10654598 DOI: 10.1016/j.isci.2023.108303] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/25/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Neuroblastoma is the most common extracranial solid tumor in children. MYCN amplification is detected in almost half of high-risk cases and is associated with poorly differentiated tumors, poor patient prognosis and poor response to therapy, including retinoids. We identify the aryl hydrocarbon receptor (AhR) as a transcription factor promoting the growth and suppressing the differentiation of MYCN-amplified neuroblastoma. A neuroblastoma specific AhR transcriptional signature reveals an inverse correlation of AhR activity with patients' outcome, suggesting AhR activity is critical for disease progression. AhR modulates chromatin structures, reducing accessibility to regions responsive to retinoic acid. Genetic and pharmacological inhibition of AhR results in induction of differentiation. Importantly, AhR antagonism with clofazimine synergizes with retinoic acid in inducing differentiation both in vitro and in vivo. Thus, we propose AhR as a target for MYCN-amplified neuroblastoma and that its antagonism, combined with current standard-of-care, may result in a more durable response in patients.
Collapse
Affiliation(s)
- Kanita A. Chaudhry
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Justine J. Jacobi
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Bryan M. Gillard
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Ellen Karasik
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Jeffrey C. Martin
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | | | - Edward Hurley
- Department of Biochemistry and Neurology, Institute for Myelin and Glia Exploration, State University of New York at Buffalo, Buffalo, NY, USA
| | - Maria Laura Feltri
- Department of Biochemistry and Neurology, Institute for Myelin and Glia Exploration, State University of New York at Buffalo, Buffalo, NY, USA
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Foundation I.R.C.C.S. Carlo Besta Neurological Institute Milan, Italy
| | - Kristopher M. Attwood
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Clare J. Twist
- Department of Pediatric Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Dominic J. Smiraglia
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Mark D. Long
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Anna Bianchi-Smiraglia
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
18
|
Kichina JV, Maslov A, Kandel ES. PAK1 and Therapy Resistance in Melanoma. Cells 2023; 12:2373. [PMID: 37830586 PMCID: PMC10572217 DOI: 10.3390/cells12192373] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/23/2023] [Accepted: 09/27/2023] [Indexed: 10/14/2023] Open
Abstract
Malignant melanoma claims more lives than any other skin malignancy. While primary melanomas are usually cured via surgical excision, the metastatic form of the disease portents a poor prognosis. Decades of intense research has yielded an extensive armamentarium of anti-melanoma therapies, ranging from genotoxic chemo- and radiotherapies to targeted interventions in specific signaling pathways and immune functions. Unfortunately, even the most up-to-date embodiments of these therapies are not curative for the majority of metastatic melanoma patients, and the need to improve their efficacy is widely recognized. Here, we review the reports that implicate p21-regulated kinase 1 (PAK1) and PAK1-related pathways in the response of melanoma to various therapeutic modalities. Ample data suggest that PAK1 may decrease cell sensitivity to programmed cell death, provide additional stimulation to growth-promoting molecular pathways, and contribute to the creation of an immunosuppressive tumor microenvironment. Accordingly, there is mounting evidence that the concomitant inhibition of PAK1 enhances the potency of various anti-melanoma regimens. Overall, the available information suggests that a safe and effective inhibition of PAK1-dependent molecular processes would enhance the potency of the currently available anti-melanoma treatments, although considerable challenges in implementing such strategies still exist.
Collapse
Affiliation(s)
- Julia V. Kichina
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm & Carlton St., Buffalo, NY 14263, USA
| | - Alexei Maslov
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Elm & Carlton St., Buffalo, NY 14263, USA
| | - Eugene S. Kandel
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Elm & Carlton St., Buffalo, NY 14263, USA
| |
Collapse
|
19
|
Yuan H, Zhao Z, Xu J, Zhang R, Ma L, Han J, Zhao W, Guo M, Song Y. Hypoxia-induced TMTC3 expression in esophageal squamous cell carcinoma potentiates tumor angiogenesis through Rho GTPase/STAT3/VEGFA pathway. J Exp Clin Cancer Res 2023; 42:249. [PMID: 37752569 PMCID: PMC10521530 DOI: 10.1186/s13046-023-02821-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/02/2023] [Indexed: 09/28/2023] Open
Abstract
BACKGROUND Hypoxia is one of most typical features in the tumor microenvironment of solid tumor and an inducer of endoplasmic reticulum (ER) stress, and HIF-1α functions as a key transcription factor regulator to promote tumor angiogenesis in the adaptive response to hypoxia. Increasing evidence has suggested that hypoxia plays an important regulatory role of ER homeostasis. We previously identified TMTC3 as an ER stress mediator under nutrient-deficiency condition in esophageal squamous cell carcinoma (ESCC), but the molecular mechanism in hypoxia is still unclear. METHODS RNA sequencing data of TMTC3 knockdown cells and TCGA database were analyzed to determine the association of TMTC3 and hypoxia. Moreover, ChIP assay and dual-luciferase reporter assay were performed to detect the interaction of HIF-1α and TMTC3 promoter. In vitro and in vivo assays were used to investigate the function of TMTC3 in tumor angiogenesis. The molecular mechanism was determined using co-immunoprecipitation assays, immunofluorescence assays and western blot. The TMTC3 inhibitor was identified by high-throughput screening of FDA-approved drugs. The combination of TMTC3 inhibitor and cisplatin was conducted to confirm the efficiency in vitro and in vivo. RESULTS The expression of TMTC3 was remarkably increased under hypoxia and regulated by HIF-1α. Knockdown of TMTC3 inhibited the capability of tumor angiogenesis and ROS production in ESCC. Mechanistically, TMTC3 promoted the production of GTP through interacting with IMPDH2 Bateman domain. The activity of Rho GTPase/STAT3, regulated by cellular GTP levels, decreased in TMTC3 knockdown cells, whereas reversed by IMPDH2 overexpression. Additionally, TMTC3 regulated the expression of VEGFA through Rho GTPase/STAT3 pathway. Allopurinol inhibited the expression of TMTC3 and further reduced the phosphorylation and activation of STAT3 signaling pathway in a dose-dependent manner in ESCC. Additionally, the combination of allopurinol and cisplatin significantly inhibited the cell viability in vitro and tumor growth in vivo, comparing with single drug treatment, respectively. CONCLUSIONS Collectively, our study clarified the molecular mechanism of TMTC3 in regulating tumor angiogenesis and highlighted the potential therapeutic combination of TMTC3 inhibitor and cisplatin, which proposed a promising strategy for the treatment of ESCC.
Collapse
Affiliation(s)
- Hongyu Yuan
- Department of Gastroenterology & Hepatology, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Zitong Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jing Xu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Ruiping Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Liying Ma
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jing Han
- Department of Medical Oncology, Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Shijiazhuang, 050000, Hebei, China
| | - Weihong Zhao
- Medical Department, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Mingzhou Guo
- Department of Gastroenterology & Hepatology, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China.
| | - Yongmei Song
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
20
|
Zhou W, Zhao Z, Lin A, Yang J, Xu J, Kari WR, Yang A, Li J, Solanki S, Speth J, Walker N, Scott AJ, Kothari AU, Yao Y, Peterson ER, Korimerla N, Werner CK, Liang J, Jacobson J, Palavalasa S, Obrien AM, Elaimy AL, Ferris SP, Zhao SG, Sarkaria JN, Győrffy B, Zhang S, Al-Holou WN, Umemura Y, Morgan MA, Lawrence TS, Lyssiotis CA, Peters-Golden M, Shah YM, Wahl DR. GTP signaling links metabolism, DNA repair, and responses to genotoxic stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.12.536297. [PMID: 37090571 PMCID: PMC10120670 DOI: 10.1101/2023.04.12.536297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
How cell metabolism regulates DNA repair is incompletely understood. Here, we define a GTP-mediated signaling cascade that links metabolism to DNA repair and has significant therapeutic implications. GTP, but not other nucleotides, regulates the activity of Rac1, a G protein, that promotes the dephosphorylation of serine 323 on Abl-interactor 1 (Abi-1) by protein phosphatase 5 (PP5). Dephosphorylated Abi-1, a protein previously not known to activate DNA repair, promotes non-homologous end joining. In patients and mouse models of glioblastoma, Rac1 and dephosphorylated Abi-1 mediate DNA repair and resistance to standard of care genotoxic treatments. The GTP-Rac1-PP5-Abi-1 signaling axis is not limited to brain cancer, as GTP supplementation promotes DNA repair and Abi-1-S323 dephosphorylation in non-malignant cells and protects mouse tissues from genotoxic insult. This unexpected ability of GTP to regulate DNA repair independently of deoxynucleotide pools has important implications for normal physiology and cancer treatment.
Collapse
|
21
|
Martínez RAS, Pinky PD, Harlan BA, Brewer GJ. GTP energy dependence of endocytosis and autophagy in the aging brain and Alzheimer's disease. GeroScience 2023; 45:757-780. [PMID: 36622562 PMCID: PMC9886713 DOI: 10.1007/s11357-022-00717-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/15/2022] [Indexed: 01/10/2023] Open
Abstract
Increased interest in the aging and Alzheimer's disease (AD)-related impairments in autophagy in the brain raise important questions about regulation and treatment. Since many steps in endocytosis and autophagy depend on GTPases, new measures of cellular GTP levels are needed to evaluate energy regulation in aging and AD. The recent development of ratiometric GTP sensors (GEVALS) and findings that GTP levels are not homogenous inside cells raise new issues of regulation of GTPases by the local availability of GTP. In this review, we highlight the metabolism of GTP in relation to the Rab GTPases involved in formation of early endosomes, late endosomes, and lysosomal transport to execute the autophagic degradation of damaged cargo. Specific GTPases control macroautophagy (mitophagy), microautophagy, and chaperone-mediated autophagy (CMA). By inference, local GTP levels would control autophagy, if not in excess. Additional levels of control are imposed by the redox state of the cell, including thioredoxin involvement. Throughout this review, we emphasize the age-related changes that could contribute to deficits in GTP and AD. We conclude with prospects for boosting GTP levels and reversing age-related oxidative redox shift to restore autophagy. Therefore, GTP levels could regulate the numerous GTPases involved in endocytosis, autophagy, and vesicular trafficking. In aging, metabolic adaptation to a sedentary lifestyle could impair mitochondrial function generating less GTP and redox energy for healthy management of amyloid and tau proteostasis, synaptic function, and inflammation.
Collapse
Affiliation(s)
| | - Priyanka D. Pinky
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697 USA
| | - Benjamin A. Harlan
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697 USA
| | - Gregory J. Brewer
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697 USA
- Center for Neurobiology of Learning and Memory, University of California Irvine, Irvine, CA 92697 USA
- MIND Institute, University of California Irvine, Irvine, CA 92697 USA
| |
Collapse
|
22
|
Ma N, Xu E, Luo Q, Song G. Rac1: A Regulator of Cell Migration and A Potential Target for Cancer Therapy. Molecules 2023; 28:molecules28072976. [PMID: 37049739 PMCID: PMC10096471 DOI: 10.3390/molecules28072976] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/24/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Cell migration is crucial for physiological and pathological processes such as morphogenesis, wound repair, immune response and cancer invasion/metastasis. There are many factors affecting cell migration, and the regulatory mechanisms are complex. Rac1 is a GTP-binding protein with small molecular weight belonging to the Rac subfamily of the Rho GTPase family. As a key molecule in regulating cell migration, Rac1 participates in signal transduction from the external cell to the actin cytoskeleton and promotes the establishment of cell polarity which plays an important role in cancer cell invasion/metastasis. In this review, we firstly introduce the molecular structure and activity regulation of Rac1, and then summarize the role of Rac1 in cancer invasion/metastasis and other physiological processes. We also discuss the regulatory mechanisms of Rac1 in cell migration and highlight it as a potential target in cancer therapy. Finally, the current state as well as the future challenges in this area are considered. Understanding the role and the regulatory mechanism of Rac1 in cell migration can provide fundamental insights into Rac1-related cancer progression and further help us to develop novel intervention strategies for cancer therapy in clinic.
Collapse
|
23
|
Nchourupouo KWT, Nde J, Ngouongo YJW, Zekeng SS, Fongang B. Evolutionary Couplings and Molecular Dynamic Simulations Highlight Details of GPCRs Heterodimers' Interfaces. Molecules 2023; 28:1838. [PMID: 36838825 PMCID: PMC9966702 DOI: 10.3390/molecules28041838] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/03/2023] [Accepted: 02/08/2023] [Indexed: 02/18/2023] Open
Abstract
A growing body of evidence suggests that only a few amino acids ("hot-spots") at the interface contribute most of the binding energy in transient protein-protein interactions. However, experimental protocols to identify these hot-spots are highly labor-intensive and expensive. Computational methods, including evolutionary couplings, have been proposed to predict the hot-spots, but they generally fail to provide details of the interacting amino acids. Here we showed that unbiased evolutionary methods followed by biased molecular dynamic simulations could achieve this goal and reveal critical elements of protein complexes. We applied the methodology to selected G-protein coupled receptors (GPCRs), known for their therapeutic properties. We used the structure-prior-assisted direct coupling analysis (SP-DCA) to predict the binding interfaces of A2aR/D2R, CB1R/D2R, A2aR/CB1R, 5HT2AR/D2R, and 5-HT2AR/mGluR2 receptor heterodimers, which all agreed with published data. In order to highlight details of the interactions, we performed molecular dynamic (MD) simulations using the newly developed AWSEM energy model. We found that these receptors interact primarily through critical residues at the C and N terminal domains and the third intracellular loop (ICL3). The MD simulations showed that these residues are energetically necessary for dimerization and revealed their native conformational state. We subsequently applied the methodology to the 5-HT2AR/5-HTR4R heterodimer, given its implication in drug addiction and neurodegenerative pathologies such as Alzheimer's disease (AD). Further, the SP-DCA analysis showed that 5-HT2AR and 5-HTR4R heterodimerize through the C-terminal domain of 5-HT2AR and ICL3 of 5-HT4R. However, elucidating the details of GPCR interactions would accelerate the discovery of druggable sites and improve our knowledge of the etiology of common diseases, including AD.
Collapse
Affiliation(s)
- Karim Widad Temgbet Nchourupouo
- Laboratory of Mechanics, Materials, and Structures, Department of Physics, Faculty of Science, University of Yaoundé I, Yaoundé P.O. Box 812, Cameroon
| | - Jules Nde
- Department of Physics, University of Washington Seattle, Seattle, WA 98105, USA
| | - Yannick Joel Wadop Ngouongo
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Serge Sylvain Zekeng
- Laboratory of Mechanics, Materials, and Structures, Department of Physics, Faculty of Science, University of Yaoundé I, Yaoundé P.O. Box 812, Cameroon
| | - Bernard Fongang
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
- Department of Population Health Sciences, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
24
|
Xiao J, Zhao T, Fang W, Chen Y, Wu H, Li P, Chen X, Yan R, Jiang Y, Li S, Yang H, Wu C, Qin X, Liao X, Cai L, Li T, Liu Y. Caveolin-1 signaling-driven mitochondrial fission and cytoskeleton remodeling promotes breast cancer migration. Int J Biochem Cell Biol 2022; 152:106307. [PMID: 36162640 DOI: 10.1016/j.biocel.2022.106307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 09/15/2022] [Accepted: 09/21/2022] [Indexed: 10/31/2022]
Abstract
Mitochondria are highly dynamic organelles that constantly divide and fuse to maintain their proper structure and function. Cancer cells are often accompanied by an imbalance of mitochondrial fusion and fission, cancer progression is greatly affected by this imbalance. Here, we found that high-metastatic breast cancer MDA-MB-231 cells possess higher caveolin-1 (Cav-1) expression compared with low-metastatic breast cancer MCF-7 cells or normal breast epithelial MCF-10A cells. Downregulation of Cav-1 decreases the migratory and invasive abilities of MDA-MB-231 cells. Our results further demonstrated that downregulation of Cav-1 facilitated DRP1 and MFN2 to translocate to mitochondria, increasing the inhibitory phosphorylation level of DRP1 at Ser637 by protein kinase A (PKA), resulting in mitochondria elongation. We also showed that downregulation of Cav-1 significantly reduced the Rac1 activity by affecting intracellular reactive oxygen species (ROS) generation, which then inhibited F-actin formation. Based on these findings, we proposed that Cav-1 mediated mitochondrial fission-affected intracellular ROS generation and activated Rho GTPases, leading to F-actin-dependent formation of lamellipodia and promotion of breast cancer motility.
Collapse
Affiliation(s)
- Jinman Xiao
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Tian Zhao
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Wanli Fang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Yu Chen
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Hao Wu
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan, PR China
| | - Ping Li
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Xiangyan Chen
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Ran Yan
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, PR China; TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan, PR China
| | - Ying Jiang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Shun Li
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Hong Yang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Chunhui Wu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Xiang Qin
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Xiaoling Liao
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, Chongqing University of Science and Technology, Chongqing 401331, PR China
| | - Lulu Cai
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Tingting Li
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, PR China.
| | - Yiyao Liu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, PR China; TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan, PR China; Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, Chongqing University of Science and Technology, Chongqing 401331, PR China.
| |
Collapse
|
25
|
Wolff DW, Bianchi-Smiraglia A, Nikiforov MA. Compartmentalization and regulation of GTP in control of cellular phenotypes. Trends Mol Med 2022; 28:758-769. [PMID: 35718686 PMCID: PMC9420775 DOI: 10.1016/j.molmed.2022.05.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 10/18/2022]
Abstract
Genetic or pharmacological inhibition of enzymes involved in GTP biosynthesis has substantial biological effects, underlining the need to better understand the function of GTP levels in regulation of cellular processes and the significance of targeting GTP biosynthesis enzymes for therapeutic intervention. Our current understanding of spatiotemporal regulation of GTP metabolism and its role in physiological and pathological cellular processes is far from complete. Novel methodologies such as genetically encoded sensors of free GTP offered insights into intracellular distribution and function of GTP molecules. In the current Review, we provide analysis of recent discoveries in the field of GTP metabolism and evaluate the key enzymes as molecular targets.
Collapse
Affiliation(s)
- David W Wolff
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA.
| | - Anna Bianchi-Smiraglia
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
| | - Mikhail A Nikiforov
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA; Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
26
|
Chang CC, Peng M, Zhong J, Zhang Z, Keppeke GD, Sung LY, Liu JL. Molecular crowding facilitates bundling of IMPDH polymers and cytoophidium formation. Cell Mol Life Sci 2022; 79:420. [PMID: 35833994 PMCID: PMC11072341 DOI: 10.1007/s00018-022-04448-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/14/2022] [Accepted: 06/21/2022] [Indexed: 11/29/2022]
Abstract
The cytoophidium is a unique type of membraneless compartment comprising of filamentous protein polymers. Inosine monophosphate dehydrogenase (IMPDH) catalyzes the rate-limiting step of de novo GTP biosynthesis and plays critical roles in active cell metabolism. However, the molecular regulation of cytoophidium formation is poorly understood. Here we show that human IMPDH2 polymers bundle up to form cytoophidium-like aggregates in vitro when macromolecular crowders are present. The self-association of IMPDH polymers is suggested to rely on electrostatic interactions. In cells, the increase of molecular crowding with hyperosmotic medium induces cytoophidia, while the decrease of that by the inhibition of RNA synthesis perturbs cytoophidium assembly. In addition to IMPDH, CTPS and PRPS cytoophidium could be also induced by hyperosmolality, suggesting a universal phenomenon of cytoophidium-forming proteins. Finally, our results indicate that the cytoophidium can prolong the half-life of IMPDH, which is proposed to be one of conserved functions of this subcellular compartment.
Collapse
Affiliation(s)
- Chia-Chun Chang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
- Institute of Biotechnology, National Taiwan University, Taipei, 106, Taiwan
| | - Min Peng
- Institute of Biotechnology, National Taiwan University, Taipei, 106, Taiwan
| | - Jiale Zhong
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Ziheng Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Gerson Dierley Keppeke
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
- Rheumatology Division, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, Sao Paulo, SP, 04023-062, Brazil
| | - Li-Ying Sung
- Institute of Biotechnology, National Taiwan University, Taipei, 106, Taiwan
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, 115, Taiwan
| | - Ji-Long Liu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK.
| |
Collapse
|
27
|
Wolff DW, Deng Z, Bianchi-Smiraglia A, Foley CE, Han Z, Wang X, Shen S, Rosenberg MM, Moparthy S, Yun DH, Chen J, Baker BK, Roll MV, Magiera AJ, Li J, Hurley E, Feltri ML, Cox AO, Lee J, Furdui CM, Liu L, Bshara W, LaConte LE, Kandel ES, Pasquale EB, Qu J, Hedstrom L, Nikiforov MA. Phosphorylation of guanosine monophosphate reductase triggers a GTP-dependent switch from pro- to anti-oncogenic function of EPHA4. Cell Chem Biol 2022; 29:970-984.e6. [PMID: 35148834 PMCID: PMC9620470 DOI: 10.1016/j.chembiol.2022.01.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 11/19/2021] [Accepted: 01/11/2022] [Indexed: 12/11/2022]
Abstract
Signal transduction pathways post-translationally regulating nucleotide metabolism remain largely unknown. Guanosine monophosphate reductase (GMPR) is a nucleotide metabolism enzyme that decreases GTP pools by converting GMP to IMP. We observed that phosphorylation of GMPR at Tyr267 is critical for its activity and found that this phosphorylation by ephrin receptor tyrosine kinase EPHA4 decreases GTP pools in cell protrusions and levels of GTP-bound RAC1. EPHs possess oncogenic and tumor-suppressor activities, although the mechanisms underlying switches between these two modes are poorly understood. We demonstrated that GMPR plays a key role in EPHA4-mediated RAC1 suppression. This supersedes GMPR-independent activation of RAC1 by EPHA4, resulting in a negative overall effect on melanoma cell invasion and tumorigenicity. Accordingly, EPHA4 levels increase during melanoma progression and inversely correlate with GMPR levels in individual melanoma tumors. Therefore, phosphorylation of GMPR at Tyr267 is a metabolic signal transduction switch controlling GTP biosynthesis and transformed phenotypes.
Collapse
Affiliation(s)
- David W. Wolff
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA,Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | - Zhiyong Deng
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | - Anna Bianchi-Smiraglia
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
| | - Colleen E. Foley
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
| | - Zhannan Han
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA,Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | - Xingyou Wang
- Department of Chemistry, Brandeis University, Waltham, MA 02453, USA
| | - Shichen Shen
- Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, NY 14214, USA
| | | | - Sudha Moparthy
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | - Dong Hyun Yun
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | - Jialin Chen
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA,Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | - Brian K. Baker
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | - Matthew V. Roll
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA,Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | - Andrew J. Magiera
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | - Jun Li
- Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, NY 14214, USA
| | - Edward Hurley
- Department of Biochemistry and Neurology, Hunter James Kelly Research Institute, University at Buffalo, Buffalo NY, USA
| | - Maria Laura Feltri
- Department of Biochemistry and Neurology, Hunter James Kelly Research Institute, University at Buffalo, Buffalo NY, USA
| | - Anderson O. Cox
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem NC, USA
| | - Jingyun Lee
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem NC, USA
| | - Cristina M. Furdui
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem NC, USA
| | - Liang Liu
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | - Wiam Bshara
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo NY 14203, USA
| | - Leslie E.W. LaConte
- Fralin Biomedical Research Institute at Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
| | - Eugene S. Kandel
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
| | - Elena B. Pasquale
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Jun Qu
- Department of Chemistry, Brandeis University, Waltham, MA 02453, USA
| | - Lizbeth Hedstrom
- Department of Chemistry, Brandeis University, Waltham, MA 02453, USA,Department of Biology, Brandeis University, Waltham, MA 02453, USA
| | - Mikhail A. Nikiforov
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA,Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA,Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA,Corresponding author and lead contact: Mikhail A. Nikiforov,
| |
Collapse
|
28
|
Rac1 as a Target to Treat Dysfunctions and Cancer of the Bladder. Biomedicines 2022; 10:biomedicines10061357. [PMID: 35740379 PMCID: PMC9219850 DOI: 10.3390/biomedicines10061357] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 12/28/2022] Open
Abstract
Bladder pathologies, very common in the aged population, have a considerable negative impact on quality of life. Novel targets are needed to design drugs and combinations to treat diseases such as overactive bladder and bladder cancers. A promising new target is the ubiquitous Rho GTPase Rac1, frequently dysregulated and overexpressed in bladder pathologies. We have analyzed the roles of Rac1 in different bladder pathologies, including bacterial infections, diabetes-induced bladder dysfunctions and bladder cancers. The contribution of the Rac1 protein to tumorigenesis, tumor progression, epithelial-mesenchymal transition of bladder cancer cells and their metastasis has been analyzed. Small molecules selectively targeting Rac1 have been discovered or designed, and two of them—NSC23766 and EHT 1864—have revealed activities against bladder cancer. Their mode of interaction with Rac1, at the GTP binding site or the guanine nucleotide exchange factors (GEF) interaction site, is discussed. Our analysis underlines the possibility of targeting Rac1 with small molecules with the objective to combat bladder dysfunctions and to reduce lower urinary tract symptoms. Finally, the interest of a Rac1 inhibitor to treat advanced chemoresistance prostate cancer, while reducing the risk of associated bladder dysfunction, is discussed. There is hope for a better management of bladder pathologies via Rac1-targeted approaches.
Collapse
|
29
|
Hu J, Gong X, Strömblad S. Local temporal Rac1-GTP nadirs and peaks restrict cell protrusions and retractions. SCIENCE ADVANCES 2022; 8:eabl3667. [PMID: 35319996 PMCID: PMC8942371 DOI: 10.1126/sciadv.abl3667] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 01/28/2022] [Indexed: 06/14/2023]
Abstract
Cells probe their microenvironment using membrane protrusion-retraction cycles. Spatiotemporal coordination of Rac1 and RhoA GTP-binding activities initiates and reinforces protrusions and retractions, but the control of their finite lifetime remains unclear. We examined the relations of Rac1 and RhoA GTP-binding levels to key protrusion and retraction events, as well as to cell-ECM traction forces at physiologically relevant ECM stiffness. High RhoA-GTP preceded retractions and Rac1-GTP elevation before protrusions. Notable temporal Rac1-GTP nadirs and peaks occurred at the maximal edge velocity of local membrane protrusions and retractions, respectively, followed by declined edge velocity. Moreover, altered local Rac1-GTP consistently preceded similarly altered traction force. Local optogenetic Rac1-GTP perturbations defined a function of Rac1 in restricting protrusions and retractions and in promoting local traction force. Together, we show that Rac1 plays a fundamental role in restricting the size and durability of protrusions and retractions, plausibly in part through controlling traction forces.
Collapse
|