1
|
Ji Y, Li R, Tang G, Wang W, Chen C, Yang Q. The interrelated roles of RAB family proteins in the advancement of neoplastic growth. Front Oncol 2025; 15:1513360. [PMID: 40196733 PMCID: PMC11974252 DOI: 10.3389/fonc.2025.1513360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/26/2025] [Indexed: 04/09/2025] Open
Abstract
Rab Proteins, A Subfamily Of The Ras Superfamily Of Small Gtpases, Are Critical Regulators Of Intracellular Vesicular Trafficking, Which Is Intricately Linked To Various Cellular Processes. These Proteins Play Essential Roles Not Only In Maintaining Cellular Homeostasis But Also In Mediating The Complex Interplay Between Cancer Cells and Their Microenvironment. Rab Proteins Can Act As Either Oncogenic Factors Or Tumor Suppressors, With Their Functions Highly Dependent On The Cellular Context. Mechanistic Studies Have Revealed That Rab Proteins Are Involved In A Variety Of Processes, Including Vesicular Transport, Tumor Microenvironment Regulation, Autophagy, Drug Resistance, and Metabolic Regulation, and Play Either A Promotional Or Inhibitory Role In Cancer Development. Consequently, Targeting Rab Gtpases To Restore Dysregulated Vesicular Transport Systems May Offer A Promising Therapeutic Strategy To Inhibit Cancer Progression. However, It Is Equally Important To Consider The Potential Risks Of Disrupting Rab Functions, As Their Roles Are Highly Context-Dependent and May Have Opposing Effects In Different Malignancies. This Review Focuses On The Multifaceted Involvement Of Rab Family Proteins In Cancer Progression Underscores Their Importance As Potential Therapeutic Targets and Underscores The Need For A Deeper Understanding Of Their Complex Roles In Tumorigenesis.
Collapse
Affiliation(s)
- Yuxin Ji
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu, Anhui, China
- Clinical Testing and Diagnose Experimental Center, Bengbu Medical University, Bengbu, Anhui, China
| | - Ruonan Li
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu, Anhui, China
- Clinical Testing and Diagnose Experimental Center, Bengbu Medical University, Bengbu, Anhui, China
| | - Guohui Tang
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu, Anhui, China
- Department of Biotechnology, Bengbu Medical College, Bengbu, Anhui, China
| | - Wenrui Wang
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu, Anhui, China
- Department of Biotechnology, Bengbu Medical College, Bengbu, Anhui, China
| | - Changjie Chen
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu, Anhui, China
- Department of Biotechnology, Bengbu Medical College, Bengbu, Anhui, China
| | - Qingling Yang
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu, Anhui, China
- Clinical Testing and Diagnose Experimental Center, Bengbu Medical University, Bengbu, Anhui, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
| |
Collapse
|
2
|
Chastney MR, Kaivola J, Leppänen VM, Ivaska J. The role and regulation of integrins in cell migration and invasion. Nat Rev Mol Cell Biol 2025; 26:147-167. [PMID: 39349749 DOI: 10.1038/s41580-024-00777-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2024] [Indexed: 01/29/2025]
Abstract
Integrin receptors are the main molecular link between cells and the extracellular matrix (ECM) as well as mediating cell-cell interactions. Integrin-ECM binding triggers the formation of heterogeneous multi-protein assemblies termed integrin adhesion complexes (IACs) that enable integrins to transform extracellular cues into intracellular signals that affect many cellular processes, especially cell motility. Cell migration is essential for diverse physiological and pathological processes and is dysregulated in cancer to favour cell invasion and metastasis. Here, we discuss recent findings on the role of integrins in cell migration with a focus on cancer cell dissemination. We review how integrins regulate the spatial distribution and dynamics of different IACs, covering classical focal adhesions, emerging adhesion types and adhesion regulation. We discuss the diverse roles integrins have during cancer progression from cell migration across varied ECM landscapes to breaching barriers such as the basement membrane, and eventual colonization of distant organs.
Collapse
Affiliation(s)
- Megan R Chastney
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Jasmin Kaivola
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Veli-Matti Leppänen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Johanna Ivaska
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland.
- Department of Life Technologies, University of Turku, Turku, Finland.
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland.
- Western Finnish Cancer Center (FICAN West), University of Turku, Turku, Finland.
- Foundation for the Finnish Cancer Institute, Helsinki, Finland.
| |
Collapse
|
3
|
Luan C, Gao Y, Zhao J, Zhang X, Wang C, Sun W, Li Y, Yang X, Chen J, Liu W, Gong W, Ma X. Chloride intracellular channel CLIC3 mediates fibroblast cellular senescence by interacting with ERK7. Commun Biol 2025; 8:51. [PMID: 39809890 PMCID: PMC11732983 DOI: 10.1038/s42003-025-07482-5] [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: 04/22/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025] Open
Abstract
Cellular senescence (CS) is recognized as a critical driver of aging and age-related disorders. Recent studies have emphasized the roles of ion channels as key mediators of CS. Nonetheless, the roles and regulatory mechanisms of chloride intracellular channels (CLICs) during CS remain largely unexplored. In this study, we conducted RNA sequencing on bleomycin-induced senescent lung tissues from mice and identified Clic3 as the most significantly upregulated CLIC member. Furthermore, our findings revealed that the knockdown of CLIC3 mitigated intracellular chloride ion lose, mitochondrial dysfunction, nuclear enlargement, DNA damage, CS progression, and expression of senescence-associated secretory phenotype (SASP) triggered by bleomycin. Mechanistically, CLIC3 controls CS by translocating to the membrane where it interacts with extracellular signal-regulated kinase 7 (ERK7). Overall, our work demonstrates that the chloride intracellular channel CLIC3 modulates CS by repressing ERK7 activity and provides novel insights into the role of chloride channels.
Collapse
Affiliation(s)
- Changjiao Luan
- Laboratory of Intensive Care, Laboratory for Prevention and Translation of Geriatric Diseases, The Affiliated Hospital of Yangzhou University, Yangzhou, China
- Department of Lung, The Third People's Hospital of Yangzhou, Yangzhou, China
| | - Yue Gao
- Laboratory of Intensive Care, Laboratory for Prevention and Translation of Geriatric Diseases, The Affiliated Hospital of Yangzhou University, Yangzhou, China
- Department of Pathology, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Jun Zhao
- Department of Lung, The Third People's Hospital of Yangzhou, Yangzhou, China
| | - Xiaohui Zhang
- Department of Thoracic Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Chaofan Wang
- Laboratory of Intensive Care, Laboratory for Prevention and Translation of Geriatric Diseases, The Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Wentao Sun
- Laboratory of Intensive Care, Laboratory for Prevention and Translation of Geriatric Diseases, The Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Yucheng Li
- Laboratory of Intensive Care, Laboratory for Prevention and Translation of Geriatric Diseases, The Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Xinxing Yang
- Laboratory of Intensive Care, Laboratory for Prevention and Translation of Geriatric Diseases, The Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Jiaxiao Chen
- Laboratory of Intensive Care, Laboratory for Prevention and Translation of Geriatric Diseases, The Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Weili Liu
- Laboratory of Intensive Care, Laboratory for Prevention and Translation of Geriatric Diseases, The Affiliated Hospital of Yangzhou University, Yangzhou, China.
| | - Weijuan Gong
- Laboratory of Intensive Care, Laboratory for Prevention and Translation of Geriatric Diseases, The Affiliated Hospital of Yangzhou University, Yangzhou, China.
| | - Xingjie Ma
- Laboratory of Intensive Care, Laboratory for Prevention and Translation of Geriatric Diseases, The Affiliated Hospital of Yangzhou University, Yangzhou, China.
| |
Collapse
|
4
|
Yu K, Wang GM, Guo SS, Bassermann F, Fässler R. The USP12/46 deubiquitinases protect integrins from ESCRT-mediated lysosomal degradation. EMBO Rep 2024; 25:5687-5718. [PMID: 39506038 PMCID: PMC11624278 DOI: 10.1038/s44319-024-00300-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 10/02/2024] [Accepted: 10/16/2024] [Indexed: 11/08/2024] Open
Abstract
The functions of integrins are tightly regulated via multiple mechanisms including trafficking and degradation. Integrins are repeatedly internalized, routed into the endosomal system and either degraded by the lysosome or recycled back to the plasma membrane. The ubiquitin system dictates whether internalized proteins are degraded or recycled. Here, we use a genetic screen and proximity-dependent biotin identification to identify deubiquitinase(s) that control integrin surface levels. We find that a ternary deubiquitinating complex, comprised of USP12 (or the homologous USP46), WDR48 and WDR20, stabilizes β1 integrin (Itgb1) by preventing ESCRT-mediated lysosomal degradation. Mechanistically, the USP12/46-WDR48-WDR20 complex removes ubiquitin from the cytoplasmic tail of internalized Itgb1 in early endosomes, which in turn prevents ESCRT-mediated sorting and Itgb1 degradation.
Collapse
Affiliation(s)
- Kaikai Yu
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Guan M Wang
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Shiny Shengzhen Guo
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Florian Bassermann
- Department of Medicine III, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Heidelberg, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Reinhard Fässler
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, Martinsried, Germany.
| |
Collapse
|
5
|
Zeger M, Stanisławczyk LS, Bulić M, Binder AM, Huber A. tsCRISPR based identification of Rab proteins required for the recycling of Drosophila TRPL ion channel. Front Cell Dev Biol 2024; 12:1444953. [PMID: 39372952 PMCID: PMC11450138 DOI: 10.3389/fcell.2024.1444953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 09/05/2024] [Indexed: 10/08/2024] Open
Abstract
In polarized cells, the precise regulation of protein transport to and from the plasma membrane is crucial to maintain cellular function. Dysregulation of intracellular protein transport in neurons can lead to neurodegenerative diseases such as Retinitis Pigmentosa, Alzheimer's and Parkinson's disease. Here we used the light-dependent transport of the TRPL (transient receptor potential-like) ion channel in Drosophila photoreceptor cells to study the role of Rab proteins in TRPL recycling. TRPL is located in the rhabdomeric membrane of dark-adapted flies, but it is transported out of the rhabdomere upon light exposure and localizes at the Endoplasmatic Reticulum within 12 h. Upon subsequent dark adaptation, TRPL is recycled back to the rhabdomeric membrane within 90 min. To screen for Rab proteins involved in TRPL recycling, we established a tissue specific (ts) CRISPR/Cas9-mediated knock-out of individual Rab genes in Drosophila photoreceptors and assessed TRPL localization using an eGFP tagged TRPL protein in the intact eyes of these mutants. We observed severe TRPL recycling defects in the knockouts of Rab3, Rab4, Rab7, Rab32, and RabX2. Using immunohistochemistry, we further showed that Rab3 and RabX2 each play a significant role in TRPL recycling and also influence TRPL transport. We localized Rab3 to the late endosome in Drosophila photoreceptors and observed disruption of TRPL transport to the ER in Rab3 knock-out mutants. TRPL transport from the ER to the rhabdomere ensues from the trans-Golgi where RabX2 is located. We observed accumulated TRPL at the trans-Golgi in RabX2 knock-out mutants. In summary, our study reveals the requirement of specific Rab proteins for different steps of TRPL transport in photoreceptor cells and provides evidence for a unique retrograde recycling pathway of TRPL from the ER via the trans-Golgi.
Collapse
Affiliation(s)
| | | | | | | | - Armin Huber
- Department of Biochemistry, Institute of Biology, University of Hohenheim, Stuttgart, Germany
| |
Collapse
|
6
|
Jin Y, Zhao J, Qi T, Tian D, Liao Y, Yang Q, Li M, Zhu Q, Chen J, Shen Y, Liu Y, Lu H. Reduced B cell frequencies in cord blood of HIV-exposed uninfected infants: an immunological and transcriptomic analysis. Front Immunol 2024; 15:1445239. [PMID: 39295873 PMCID: PMC11408218 DOI: 10.3389/fimmu.2024.1445239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/15/2024] [Indexed: 09/21/2024] Open
Abstract
Introduction In the course of immune development, HIV-exposed uninfected (HEU) infants exhibit abnormal immune function and increased infectious morbidity compared to HIV-unexposed uninfected (HUU) infants. Yet the specific functional phenotypes and regulatory mechanisms associated with in-utero HIV and/or ART exposure remain largely obscure. Methods We utilized flow cytometry and RNA-seq technologies to conduct the immunological and transcriptomic profiling in cord blood from 9 HEU mother-infant pairs and 24 HUU pairs. On top of that, we compared the cord blood dataset with the maternal venous blood dataset to characterize unique effects induced by in-utero HIV and/or ART exposure. Results Flow cytometry immunophenotyping revealed that the level of B lymphocyte subsets was significantly decreased in HEU cord blood as compared to HUU (P < 0.001). Expression profiling-based cell abundance assessment, includes CIBERSORT and ssGSEA algorithm, showed a significantly reduced abundance of naive B cells in HEU cord blood (both P < 0.05), supporting the altered composition of B lymphocyte subsets in HEU. Functional enrichment analysis demonstrated suppressed innate immune responses and impaired immune regulatory function of B cells in HEU cord blood. Furthermore, through differential expression analysis, co-expression network analysis using WGCNA, and feature selection analysis using LASSO, we identified a 4-gene signature associated with HEU status. This signature effectively assesses B cell levels in cord blood, enabling discrimination between HEU and HUU infants. Discussion Our study provides the first comprehensive immunological and transcriptomic characterization of HEU cord blood. Additionally, we establish a 4-gene-based classifier that holds potential for predict immunological abnormalities in HEU infants.
Collapse
Affiliation(s)
- Ye Jin
- Department of Infection Immunology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- Children's Health Department, Shanghai Center for Women and Children's Health, Shanghai, China
| | - Jun Zhao
- Department of Pediatrics, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Tangkai Qi
- Department of Infection Immunology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Di Tian
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Yixin Liao
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Qing Yang
- Child Healthcare Department, Songjiang Maternity and Child Health Hospital, Shanghai, China
| | - Minming Li
- Department of Infection Immunology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Qingqing Zhu
- Child Healthcare Department, Songjiang Maternity and Child Health Hospital, Shanghai, China
| | - Jun Chen
- Department of Infection Immunology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Yinzhong Shen
- Department of Infection Immunology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Yabin Liu
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Hongzhou Lu
- Department of Infection Immunology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- National Clinical Research Center for Infectious Diseases, Third People's Hospital of Shenzhen, Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
7
|
Lei T, Lin Y, Lai X, Zhang Y, Ma Y, Wang X, Liu W, Tang Q, Yang T, Feng W, Song W. ITGB5 facilitates gastric cancer metastasis by promoting TGFBR2 endosomal recycling. Cancer Lett 2024; 592:216953. [PMID: 38729557 DOI: 10.1016/j.canlet.2024.216953] [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: 01/09/2024] [Revised: 04/30/2024] [Accepted: 05/07/2024] [Indexed: 05/12/2024]
Abstract
TGFBR2, a key regulator of the TGFβ signaling pathway, plays a crucial role in gastric cancer (GC) metastasis through its endosomal recycling process. Despite its importance, the mechanisms governing this process remain unclear. Here, we identify integrin β5 (ITGB5) as a critical mediator that promotes TGFBR2 endosomal recycling. Our study reveals elevated expression of ITGB5 in GC, particularly in metastatic cases, correlating with poor patient outcomes. Knockdown of ITGB5 impairs GC cell metastasis both in vitro and in vivo. Mechanistically, ITGB5 facilitates epithelial-mesenchymal transition mediated by TGFβ signaling, thereby enhancing GC metastasis. Acting as a scaffold, ITGB5 interacts with TGFBR2 and SNX17, facilitating SNX17-mediated endosomal recycling of TGFBR2 and preventing lysosomal degradation, thereby maintaining its surface distribution on tumor cells. Notably, TGFβ signaling directly upregulates ITGB5 expression, establishing a positive feedback loop that exacerbates GC metastasis. Our findings shed light on the role of ITGB5 in promoting GC metastasis through SNX17-mediated endosomal recycling of TGFBR2, providing insights for the development of targeted cancer therapies.
Collapse
Affiliation(s)
- Tianxiang Lei
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Yingying Lin
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Xiaofan Lai
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Yongxin Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Yuanchen Ma
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Xiaofeng Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Wenwei Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Qiao Tang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Ting Yang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Wei Feng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China; Department of Oncology-Pathology, Karolinska Institutet, Karolinska University Hospital, Bioclinicum, Solna, 17177, Sweden
| | - Wu Song
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
| |
Collapse
|
8
|
Zhang J, Fu L, Wang H, Yonemura A, Semba T, Yasuda-Yoshihara N, Nishimura A, Tajiri T, Tong Y, Yasuda T, Uchihara T, Yamazaki M, Okamoto Y, Yamasaki J, Nagano O, Baba H, Ishimoto T. RAC1-mediated integrin alpha-6 expression in E-cadherin-deficient gastric cancer cells promotes interactions with the stroma and peritoneal dissemination. Cancer Lett 2024; 591:216901. [PMID: 38641311 DOI: 10.1016/j.canlet.2024.216901] [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: 12/18/2023] [Revised: 04/09/2024] [Accepted: 04/15/2024] [Indexed: 04/21/2024]
Abstract
Diffuse-type gastric cancer (DGC) is a subtype of gastric cancer that is prone to peritoneal dissemination, with poor patient prognosis. Although intercellular adhesion loss between cancer cells is a major characteristic of DGCs, the mechanism underlying the alteration in cell-to-extracellular matrix (ECM) adhesion is unclear. We investigated how DGCs progress and cause peritoneal dissemination through interactions between DGC cells and the tumour microenvironment (TME). P53 knockout and KRASG12V-expressing (GAN-KP) cells and Cdh1-deleted GAN-KP (GAN-KPC) cells were orthotopically transplanted into the gastric wall to mimic peritoneal dissemination. The GAN-KPC tumour morphology was similar to that of human DGCs containing abundant stroma. RNA sequencing revealed that pathways related to Rho GTPases and integrin-ECM interactions were specifically increased in GAN-KPC cells compared with GAN-KP cells. Notably, we found that Rac Family Small GTPase 1 (RAC1) induces Integrin Subunit Alpha 6 (ITGA6) trafficking, leading to its enrichment on the GC cell membrane. Fibroblasts activate the FAK/AKT pathway in GC cells by mediating extracellular matrix (ECM)-Itga6 interactions, exacerbating the malignant phenotype. In turn, GC cells induce abnormal expression of fibroblast collagen and its transformation into cancer-associated fibroblasts (CAFs), resulting in DGC-like subtypes. These findings indicate that Cdh1 gene loss leads to abnormal expression and changes in the subcellular localization of ITGA6 through RAC1 signalling. The latter, through interactions with CAFs, allows for peritoneal dissemination.
Collapse
Affiliation(s)
- Jun Zhang
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan; Gastrointestinal Cancer Biology, International Research Center of Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan; Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Lingfeng Fu
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan; Gastrointestinal Cancer Biology, International Research Center of Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan; Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Huaitao Wang
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan; Gastrointestinal Cancer Biology, International Research Center of Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan; Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Atsuko Yonemura
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan; Gastrointestinal Cancer Biology, International Research Center of Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan; Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takashi Semba
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan; Gastrointestinal Cancer Biology, International Research Center of Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Noriko Yasuda-Yoshihara
- Gastrointestinal Cancer Biology, International Research Center of Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan; Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Akiho Nishimura
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan; Gastrointestinal Cancer Biology, International Research Center of Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Takuya Tajiri
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan; Gastrointestinal Cancer Biology, International Research Center of Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan; Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yilin Tong
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan; Gastrointestinal Cancer Biology, International Research Center of Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan; Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tadahito Yasuda
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan; Gastrointestinal Cancer Biology, International Research Center of Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan; Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tomoyuki Uchihara
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan; Gastrointestinal Cancer Biology, International Research Center of Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan; Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Masaya Yamazaki
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yuya Okamoto
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Juntaro Yamasaki
- Division of Gene Regulation, Cancer Center, Fujita Health University, Toyoake, Japan
| | - Osamu Nagano
- Division of Gene Regulation, Cancer Center, Fujita Health University, Toyoake, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan; Center for Metabolic Regulation of Healthy Ageing, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takatsugu Ishimoto
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan; Gastrointestinal Cancer Biology, International Research Center of Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan; Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
9
|
Dong W, Song CY, Liu MQ, Gao YH, Zhao ZW, Zhang XB, Moussian B, Zhang JZ. Osiris17 is essential for stable integrin localization and function during insect wing epithelia remodeling. Int J Biol Macromol 2024; 263:130245. [PMID: 38367779 DOI: 10.1016/j.ijbiomac.2024.130245] [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: 12/07/2023] [Revised: 02/13/2024] [Accepted: 02/14/2024] [Indexed: 02/19/2024]
Abstract
The dynamic adhesion between cells and their extracellular matrix is essential for the development and function of organs. During insect wing development, two epithelial sheets contact each other at their basal sites through the interaction of βPS integrins with the extracellular matrix. We report that Osiris17 contributes to the maintenance of βPS integrins localization and function in developing wing of Drosophila and locust. In flies with reduced Osiris17 expression the epithelia sheets fail to maintain the integrity of basal cytoplasmic junctional bridges and basal adhesion. In contrast to the continuous basal integrin localization in control wings, this localization is disrupted during late stages of wing development in Osiris17 depleted flies. In addition, the subcellular localization revealed that Osiris17 co-localizes with the endosomal markers Rab5 and Rab11. This observation suggests an involvement of Osiris17 in endosomal recycling of integrins. Indeed, Osiris17 depletion reduced the numbers of Rab5 and Rab11 positive endosomes. Moreover, overexpression of Osiris17 increased co-localization of Rab5 and βPS integrins and partially rescued the detachment phenotype in flies with reduced βPS integrins. Taken together, our data suggest that Osiris17 is an endosome related protein that contributes to epithelial remodeling and morphogenesis by assisting basal integrins localization in insects.
Collapse
Affiliation(s)
- Wei Dong
- Research Institute of Applied Biology, Shanxi Key Laboratory of Nucleic Acid Biopesticides, Shanxi University, Taiyuan, Shanxi, China.
| | - Chen-Yang Song
- Research Institute of Applied Biology, Shanxi Key Laboratory of Nucleic Acid Biopesticides, Shanxi University, Taiyuan, Shanxi, China
| | - Meng-Qi Liu
- Research Institute of Applied Biology, Shanxi Key Laboratory of Nucleic Acid Biopesticides, Shanxi University, Taiyuan, Shanxi, China
| | - Ying-Hao Gao
- Research Institute of Applied Biology, Shanxi Key Laboratory of Nucleic Acid Biopesticides, Shanxi University, Taiyuan, Shanxi, China
| | - Zhang-Wu Zhao
- Research Institute of Applied Biology, Shanxi Key Laboratory of Nucleic Acid Biopesticides, Shanxi University, Taiyuan, Shanxi, China
| | - Xu-Bo Zhang
- Research Institute of Applied Biology, Shanxi Key Laboratory of Nucleic Acid Biopesticides, Shanxi University, Taiyuan, Shanxi, China.
| | - Bernard Moussian
- INRAE, CNRS, Institut Sophia Agrobiotech, Sophia Antipolis, Université Côte d(')Azur, 06108 Nice, France.
| | - Jian-Zhen Zhang
- Research Institute of Applied Biology, Shanxi Key Laboratory of Nucleic Acid Biopesticides, Shanxi University, Taiyuan, Shanxi, China.
| |
Collapse
|
10
|
Guo H, Zhou C, Zheng M, Zhang J, Wu H, He Q, Ding L, Yang B. Insights into the role of derailed endocytic trafficking pathway in cancer: From the perspective of cancer hallmarks. Pharmacol Res 2024; 201:107084. [PMID: 38295915 DOI: 10.1016/j.phrs.2024.107084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/12/2024] [Accepted: 01/25/2024] [Indexed: 02/06/2024]
Abstract
The endocytic trafficking pathway is a highly organized cellular program responsible for the regulation of membrane components and uptake of extracellular substances. Molecules internalized into the cell through endocytosis will be sorted for degradation or recycled back to membrane, which is determined by a series of sorting events. Many receptors, enzymes, and transporters on the membrane are strictly regulated by endocytic trafficking process, and thus the endocytic pathway has a profound effect on cellular homeostasis. However, the endocytic trafficking process is typically dysregulated in cancers, which leads to the aberrant retention of receptor tyrosine kinases and immunosuppressive molecules on cell membrane, the loss of adhesion protein, as well as excessive uptake of nutrients. Therefore, hijacking endocytic trafficking pathway is an important approach for tumor cells to obtain advantages of proliferation and invasion, and to evade immune attack. Here, we summarize how dysregulated endocytic trafficking process triggers tumorigenesis and progression from the perspective of several typical cancer hallmarks. The impact of endocytic trafficking pathway to cancer therapy efficacy is also discussed.
Collapse
Affiliation(s)
- Hongjie Guo
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chen Zhou
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Mingming Zheng
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jie Zhang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Honghai Wu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; The Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou 310018, China; Cancer Center of Zhejiang University, Hangzhou 310058, China
| | - Ling Ding
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Nanhu Brain-computer Interface Institute, Hangzhou 311100, China.
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; School of Medicine, Hangzhou City University, Hangzhou 310015, China; The Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou 310018, China.
| |
Collapse
|
11
|
Shuai Y, Zhang H, Liu C, Wang J, Jiang Y, Sun J, Gao X, Bo X, Xiao X, Liao X, Huang C, Chen H, Jiang G. CLIC3 interacts with NAT10 to inhibit N4-acetylcytidine modification of p21 mRNA and promote bladder cancer progression. Cell Death Dis 2024; 15:9. [PMID: 38182571 PMCID: PMC10770081 DOI: 10.1038/s41419-023-06373-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 11/27/2023] [Accepted: 12/05/2023] [Indexed: 01/07/2024]
Abstract
Chromatin accessibility plays important roles in revealing the regulatory networks of gene expression, while its application in bladder cancer is yet to be fully elucidated. Chloride intracellular channel 3 (CLIC3) protein has been reported to be associated with the progression of some tumors, whereas the specific mechanism of CLIC3 in tumor remains unclear. Here, we screened for key genes in bladder cancer through the identification of transcription factor binding site clustered region (TFCR) on the basis of chromatin accessibility and TF motif. CLIC3 was identified by joint profiling of chromatin accessibility data with TCGA database. Clinically, CLIC3 expression was significantly elevated in bladder cancer and was negatively correlated with patient survival. CLIC3 promoted the proliferation of bladder cancer cells by reducing p21 expression in vitro and in vivo. Mechanistically, CLIC3 interacted with NAT10 and inhibited the function of NAT10, resulting in the downregulation of ac4C modification and stability of p21 mRNA. Overall, these findings uncover an novel mechanism of mRNA ac4C modification and CLIC3 may act as a potential therapeutic target for bladder cancer.
Collapse
Affiliation(s)
- Yujun Shuai
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hui Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Changhao Liu
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Junting Wang
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Yangkai Jiang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiayin Sun
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xincheng Gao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaochen Bo
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Xingyuan Xiao
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xin Liao
- Department of General Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chao Huang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Hebing Chen
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, China.
| | - Guosong Jiang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
12
|
Johannes L, Shafaq-Zadah M, Dransart E, Wunder C, Leffler H. Endocytic Roles of Glycans on Proteins and Lipids. Cold Spring Harb Perspect Biol 2024; 16:a041398. [PMID: 37735065 PMCID: PMC10759989 DOI: 10.1101/cshperspect.a041398] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/23/2023]
Abstract
Most cell surface proteins are decorated by glycans, and the plasma membrane is rich in glycosylated lipids. The mechanisms by which the enormous complexity of these glycan structures on proteins and lipids is exploited to control glycoprotein activity by setting their cell surface residence time and the ways by which they are taken up into cells are still under active investigation. Here, two mechanisms are presented, termed galectin lattices and glycolipid-lectin (GL-Lect)-driven endocytosis, which are among the most prominent to establish a link between glycan information and endocytosis. Types of glycans on glycoproteins and glycolipids are reviewed from the angle of their interaction with glycan-binding proteins that are at the heart of galectin lattices and GL-Lect-driven endocytosis. Examples are given to show how these mechanisms affect cellular functions ranging from cell migration and signaling to vascularization and immune modulation. Finally, outstanding challenges on the link between glycosylation and endocytosis are discussed.
Collapse
Affiliation(s)
- Ludger Johannes
- Cellular and Chemical Biology Unit, Institut Curie, 75248 Paris Cedex 05, France
| | | | - Estelle Dransart
- Cellular and Chemical Biology Unit, Institut Curie, 75248 Paris Cedex 05, France
| | - Christian Wunder
- Cellular and Chemical Biology Unit, Institut Curie, 75248 Paris Cedex 05, France
| | - Hakon Leffler
- Section MIG (Microbiology, Immunology, Glycobiology), Department of Laboratory Medicine, Lund University, 22362 Lund, Sweden
| |
Collapse
|
13
|
Loyo-Celis V, Patel D, Sanghvi S, Kaur K, Ponnalagu D, Zheng Y, Bindra S, Bhachu HR, Deschenes I, Gururaja Rao S, Singh H. Biophysical characterization of chloride intracellular channel 6 (CLIC6). J Biol Chem 2023; 299:105349. [PMID: 37838179 PMCID: PMC10641671 DOI: 10.1016/j.jbc.2023.105349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 09/27/2023] [Accepted: 09/30/2023] [Indexed: 10/16/2023] Open
Abstract
Chloride intracellular channels (CLICs) are a family of proteins that exist in soluble and transmembrane forms. The newest discovered member of the family CLIC6 is implicated in breast, ovarian, lung gastric, and pancreatic cancers and is also known to interact with dopamine-(D(2)-like) receptors. The soluble structure of the channel has been resolved, but the exact physiological role of CLIC6, biophysical characterization, and the membrane structure remain unknown. Here, we aimed to characterize the biophysical properties of this channel using a patch-clamp approach. To determine the biophysical properties of CLIC6, we expressed CLIC6 in HEK-293 cells. On ectopic expression, CLIC6 localizes to the plasma membrane of HEK-293 cells. We established the biophysical properties of CLIC6 by using electrophysiological approaches. Using various anions and potassium (K+) solutions, we determined that CLIC6 is more permeable to chloride-(Cl-) as compared to bromide-(Br-), fluoride-(F-), and K+ ions. In the whole-cell configuration, the CLIC6 currents were inhibited after the addition of 10 μM of IAA-94 (CLIC-specific blocker). CLIC6 was also found to be regulated by pH and redox potential. We demonstrate that the histidine residue at 648 (H648) in the C terminus and cysteine residue in the N terminus (C487) are directly involved in the pH-induced conformational change and redox regulation of CLIC6, respectively. Using qRT-PCR, we identified that CLIC6 is most abundant in the lung and brain, and we recorded the CLIC6 current in mouse lung epithelial cells. Overall, we have determined the biophysical properties of CLIC6 and established it as a Cl- channel.
Collapse
Affiliation(s)
- Veronica Loyo-Celis
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Devendra Patel
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Shridhar Sanghvi
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, USA; Department of Molecular Cellular and Developmental Biology, The Ohio State University, Columbus, Ohio, USA
| | - Kamalpreet Kaur
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Devasena Ponnalagu
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, USA; Department of Pharmacology, The University of Washington, Seattle, Washington, USA
| | - Yang Zheng
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Sahej Bindra
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Harmeet Rireika Bhachu
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Isabelle Deschenes
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | | | - Harpreet Singh
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, USA; Department of Molecular Cellular and Developmental Biology, The Ohio State University, Columbus, Ohio, USA.
| |
Collapse
|
14
|
Lam GT, Sorvina A, Martini C, Prabhakaran S, Ung BSY, Lazniewska J, Moore CR, Beck AR, Hopkins AM, Johnson IRD, Caruso MC, Hickey SM, Brooks RD, Jackett L, Karageorgos L, Foster-Smith EJ, Malone V, Klebe S, O'Leary JJ, Brooks DA, Logan JM. Altered endosomal-lysosomal biogenesis in melanoma. Neoplasia 2023; 43:100924. [PMID: 37562257 PMCID: PMC10423698 DOI: 10.1016/j.neo.2023.100924] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 08/03/2023] [Indexed: 08/12/2023]
Abstract
Cutaneous melanoma is the deadliest form of skin neoplasm and its high mortality rates could be averted by early accurate detection. While the detection of melanoma is currently reliant upon melanin visualisation, research into melanosome biogenesis, as a key driver of pathogenesis, has not yielded technology that can reliably distinguish between atypical benign, amelanotic and melanotic lesions. The endosomal-lysosomal system has important regulatory roles in cancer cell biology, including a specific functional role in melanosome biogenesis. Herein, the involvement of the endosomal-lysosomal system in melanoma was examined by pooled secondary analysis of existing gene expression datasets. A set of differentially expressed endosomal-lysosomal genes was identified in melanoma, which were interconnected by biological function. To illustrate the protein expression of the dysregulated genes, immunohistochemistry was performed on samples from patients with cutaneous melanoma to reveal candidate markers. This study demonstrated the dysregulation of Syntenin-1, Sortilin and Rab25 may provide a differentiating feature between cutaneous melanoma and squamous cell carcinoma, while IGF2R may indicate malignant propensity in these skin cancers.
Collapse
Affiliation(s)
- Giang T Lam
- Clinical and Health Sciences, Cancer Research Institute, University of South Australia, Adelaide, SA, Australia
| | - Alexandra Sorvina
- Clinical and Health Sciences, Cancer Research Institute, University of South Australia, Adelaide, SA, Australia
| | - Carmela Martini
- Clinical and Health Sciences, Cancer Research Institute, University of South Australia, Adelaide, SA, Australia
| | - Sarita Prabhakaran
- Department of Anatomical Pathology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Ben S-Y Ung
- Clinical and Health Sciences, Cancer Research Institute, University of South Australia, Adelaide, SA, Australia
| | - Joanna Lazniewska
- Clinical and Health Sciences, Cancer Research Institute, University of South Australia, Adelaide, SA, Australia
| | - Courtney R Moore
- Clinical and Health Sciences, Cancer Research Institute, University of South Australia, Adelaide, SA, Australia
| | - Andrew R Beck
- Clinical and Health Sciences, Cancer Research Institute, University of South Australia, Adelaide, SA, Australia
| | - Ashley M Hopkins
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Ian R D Johnson
- Clinical and Health Sciences, Cancer Research Institute, University of South Australia, Adelaide, SA, Australia
| | - Maria C Caruso
- Clinical and Health Sciences, Cancer Research Institute, University of South Australia, Adelaide, SA, Australia
| | - Shane M Hickey
- Clinical and Health Sciences, Cancer Research Institute, University of South Australia, Adelaide, SA, Australia
| | - Robert D Brooks
- Clinical and Health Sciences, Cancer Research Institute, University of South Australia, Adelaide, SA, Australia
| | - Louise Jackett
- Anatomical Pathology Department, Austin Hospital, Melbourne, Vic, Australia
| | - Litsa Karageorgos
- Clinical and Health Sciences, Cancer Research Institute, University of South Australia, Adelaide, SA, Australia
| | | | - Victoria Malone
- Department of Histopathology, Trinity College Dublin, Ireland
| | - Sonja Klebe
- Department of Anatomical Pathology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia; Department of Surgical Pathology, SA Pathology at Flinders Medical Centre, Adelaide, SA, Australia
| | - John J O'Leary
- Department of Histopathology, Trinity College Dublin, Ireland
| | - Douglas A Brooks
- Clinical and Health Sciences, Cancer Research Institute, University of South Australia, Adelaide, SA, Australia
| | - Jessica M Logan
- Clinical and Health Sciences, Cancer Research Institute, University of South Australia, Adelaide, SA, Australia.
| |
Collapse
|
15
|
Wilson B, Flett C, Gemperle J, Lawless C, Hartshorn M, Hinde E, Harrison T, Chastney M, Taylor S, Allen J, Norman JC, Zacharchenko T, Caswell PT. Proximity labelling identifies pro-migratory endocytic recycling cargo and machinery of the Rab4 and Rab11 families. J Cell Sci 2023; 136:jcs260468. [PMID: 37232246 PMCID: PMC10323252 DOI: 10.1242/jcs.260468] [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: 07/28/2022] [Accepted: 05/18/2023] [Indexed: 05/27/2023] Open
Abstract
Endocytic recycling controls the return of internalised cargoes to the plasma membrane to coordinate their positioning, availability and downstream signalling. The Rab4 and Rab11 small GTPase families regulate distinct recycling routes, broadly classified as fast recycling from early endosomes (Rab4) and slow recycling from perinuclear recycling endosomes (Rab11), and both routes handle a broad range of overlapping cargoes to regulate cell behaviour. We adopted a proximity labelling approach, BioID, to identify and compare the protein complexes recruited by Rab4a, Rab11a and Rab25 (a Rab11 family member implicated in cancer aggressiveness), revealing statistically robust protein-protein interaction networks of both new and well-characterised cargoes and trafficking machinery in migratory cancer cells. Gene ontological analysis of these interconnected networks revealed that these endocytic recycling pathways are intrinsically connected to cell motility and cell adhesion. Using a knock-sideways relocalisation approach, we were further able to confirm novel links between Rab11, Rab25 and the ESCPE-1 and retromer multiprotein sorting complexes, and identify new endocytic recycling machinery associated with Rab4, Rab11 and Rab25 that regulates cancer cell migration in the 3D matrix.
Collapse
Affiliation(s)
- Beverley Wilson
- Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, M13 9PT, UK
| | - Chloe Flett
- Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, M13 9PT, UK
| | - Jakub Gemperle
- Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, M13 9PT, UK
| | - Craig Lawless
- Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, M13 9PT, UK
| | - Matthew Hartshorn
- Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, M13 9PT, UK
| | - Eleanor Hinde
- Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, M13 9PT, UK
| | - Tess Harrison
- Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, M13 9PT, UK
| | - Megan Chastney
- Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, M13 9PT, UK
| | - Sarah Taylor
- Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, M13 9PT, UK
| | - Jennifer Allen
- Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, M13 9PT, UK
| | - Jim C. Norman
- Cancer Research UK Beatson Institute, Glasgow, G61 1BD, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Thomas Zacharchenko
- Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, M13 9PT, UK
| | - Patrick T. Caswell
- Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, M13 9PT, UK
| |
Collapse
|
16
|
Transcription factor EB regulates phosphatidylinositol-3-phosphate levels that control lysosome positioning in the bladder cancer model. Commun Biol 2023; 6:114. [PMID: 36709383 PMCID: PMC9884284 DOI: 10.1038/s42003-023-04501-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 01/18/2023] [Indexed: 01/29/2023] Open
Abstract
Lysosomes orchestrate degradation and recycling of exogenous and endogenous material thus controlling cellular homeostasis. Little is known how this organelle changes during cancer. Here we investigate the intracellular landscape of lysosomes in a cellular model of bladder cancer. Employing standardized cell culture on micropatterns we identify a phenotype of peripheral lysosome positioning prevailing in bladder cancer cell lines but not normal urothelium. We show that lysosome positioning is controlled by phosphatidylinositol-3-phosphate (PtdIns3P) levels on endomembranes which recruit FYVE-domain containing proteins for lysosomal dispersion. We identify transcription factor EB (TFEB) as an upstream regulator of PtdIns3P production by VPS34 that is activated in aggressive bladder cancer cells with peripheral lysosomes. This conceptually clarifies the dual role of TFEB as regulator of endosomal maturation and autophagy, two distinct processes controlled by PtdIns3P. Altogether, our findings uncover peripheral lysosome positioning, resulting from PtdIns3P production downstream of TFEB activation, as a potential biomarker for bladder cancer.
Collapse
|
17
|
Huet-Calderwood C, Rivera-Molina F, Toomre D, Calderwood DA. Use of Ecto-Tagged Integrins to Monitor Integrin Exocytosis and Endocytosis. Methods Mol Biol 2023; 2608:17-38. [PMID: 36653699 PMCID: PMC9999384 DOI: 10.1007/978-1-0716-2887-4_2] [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] [Indexed: 01/19/2023]
Abstract
Controlled exocytosis and endocytosis of integrin adhesion receptors is required for normal cell adhesion, migration, and signaling. In this chapter, we describe the design of functional β1 integrins carrying extracellular fluorescent or chemically traceable tags (ecto-tag) and methods for their use to image β1 integrin trafficking in cells. We provide approaches to generate cells in which endogenous β1 integrins are replaced by ecto-tagged integrins containing a pH-sensitive fluorophore pHluorin or a HaloTag and describe strategies using photobleaching, selective extracellular/intracellular labeling, and chase, quenching, and blocking to reveal β1 integrin exocytosis, endocytosis, and recycling by live total internal reflection fluorescence (TIRF) microscopy.
Collapse
Affiliation(s)
- Clotilde Huet-Calderwood
- Departments of Pharmacology, Yale University School of Medicine, Yale University, New Haven, CT, USA
| | - Felix Rivera-Molina
- Departments of Cell Biology, Yale University School of Medicine, Yale University, New Haven, CT, USA
| | - Derek Toomre
- Departments of Cell Biology, Yale University School of Medicine, Yale University, New Haven, CT, USA
| | - David A Calderwood
- Departments of Pharmacology, Yale University School of Medicine, Yale University, New Haven, CT, USA.
- Departments of Cell Biology, Yale University School of Medicine, Yale University, New Haven, CT, USA.
| |
Collapse
|
18
|
Integrin receptor trafficking in health and disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 196:271-302. [PMID: 36813362 DOI: 10.1016/bs.pmbts.2022.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Integrins are a family of 24 different heterodimers that are indispensable for multicellular life. Cell polarity, adhesion and migration are controlled by integrins delivered to the cell surface which in turn is regulated by the exo- and endocytic trafficking of integrins. The deep integration between trafficking and cell signaling determines the spatial and temporal output from any biochemical cue. Integrin trafficking plays a key role in development and many pathological conditions, especially cancer. Several novel regulators of integrin traffic have been discovered in recent times, including a novel class of integrin carrying vesicles, the intracellular nanovesicles (INVs). The tight regulation of trafficking pathways by cell signaling, where kinases phosphorylate key small GTPases in the trafficking pathway enable coordination of cell response to the extracellular milieu. Integrin heterodimer expression and trafficking differ in different tissues and contexts. In this Chapter, we discuss recent studies on integrin trafficking and its contribution to normal physiological and pathophysiological states.
Collapse
|
19
|
Kohil A, Amir SS, Behrens A, Khan OM. A small Rho GTPase RAB25 with a potential role in chemotherapy resistance in pancreatic cancer. Cancer Biomark 2022; 36:133-145. [PMID: 36565104 DOI: 10.3233/cbm-220214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDA) is one of the major human health challenges with minimal therapeutic benefits due to its late detection, and de novo - and acquired chemotherapy resistance. OBJECTIVE In this work we unravel the potential pro-survival role of RAB25 in pancreatic cancer chemotherapy resistance and aim to identify if RAB25 is a prognostic marker of patients' survival in PDA. METHODS We used RNA sequencing, shRNA mediated gene knockdown, BioGRID open repository of CRISPR screens (ORCS), GEPIA, kmplot.com, and cBioPortal.org databases to identify the role of RAB25 in PDA cell proliferation, chemotherapy response, expression in tumour versus normal tissues, and overall patients' survival. RESULTS RNA sequencing show Rab25 to be one of the top upregulated genes in gemcitabine resistance mouse PDA cells. Knockdown of Rab25 in these cells enhanced gemcitabine toxicity. In addition, re-analysis of previously published CRISPR/Cas9 data confirm RAB25 to be responsible for chemotherapy resistance in KRASG12D mutant human pancreatic cancer cell line. Finally, we used publicly available TCGA datasets and identify the upregulation of RAB25 in tumour tissues compared to the adjacent normal tissue, co-occurrence of KRASG12 mutations with RAB25 amplifications, and poor patients' survival in cohorts with higher mRNA expression of RAB25. CONCLUSION RAB25 expression is a prognostic marker for patient's survival and gemcitabine resistance in PDA.
Collapse
Affiliation(s)
- Amira Kohil
- Biological and Biomedical Sciences, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Sayeda S Amir
- Biological and Biomedical Sciences, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Axel Behrens
- The Francis Crick Institute, London, UK.,Cancer Stem Cell Team, Institute of Cancer Research, London, UK
| | - Omar M Khan
- Biological and Biomedical Sciences, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| |
Collapse
|
20
|
Structure-based discovery and in vitro validation of inhibitors of chloride intracellular channel 4 protein. Comput Struct Biotechnol J 2022; 21:688-701. [PMID: 36659928 PMCID: PMC9826898 DOI: 10.1016/j.csbj.2022.12.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 12/25/2022] Open
Abstract
The use of computer-aided methods have continued to propel accelerated drug discovery across various disease models, interestingly allowing the specific inhibition of pathogenic targets. Chloride Intracellular Channel Protein 4 (CLIC4) is a novel class of intracellular ion channel highly implicated in tumor and vascular biology. It regulates cell proliferation, apoptosis and angiogenesis; and is involved in multiple pathologic signaling pathways. Absence of specific inhibitors however impedes its advancement to translational research. Here, we integrate structural bioinformatics and experimental research approaches for the discovery and validation of small-molecule inhibitors of CLIC4. High-affinity allosteric binders were identified from a library of 1615 Food and Drug Administration (FDA)-approved drugs via a high-performance computing-powered blind-docking approach, resulting in the selection of amphotericin B and rapamycin. NMR assays confirmed the binding and conformational disruptive effects of both drugs while they also reversed stress-induced membrane translocation of CLIC4 and inhibited endothelial cell migration. Structural and dynamics simulation studies further revealed that the inhibitory mechanisms of these compounds were hinged on the allosteric modulation of the catalytic glutathione (GSH)-like site loop and the extended catalytic β loop which may elicit interference with the catalytic activities of CLIC4. Structure-based insights from this study provide the basis for the selective targeting of CLIC4 to treat the associated pathologies.
Collapse
Key Words
- A9C, 9-Anthracenecarboxylic acid
- AMPhB, Amphotericin B
- Ad, Adenovirus
- Allosteric inhibition
- Bad, BCL2 associated agonist of cell death
- Bcl-2, B-cell lymphoma 2
- Bcl-xL, B-cell lymphoma-extra large
- CDK, Cyclin-dependent kinases
- CLIC, Chloride intracellular channel protein
- Chloride intracellular channel protein 4
- Computational high-throughput screening
- DAPI, 4′,6-diamidino-2-phenylindole
- DIDS, 4,4′-Diisothiocyano-2,2′-stilbenedisulfonic acid
- DMSO, Dimethyl sulfoxide
- DOPE, Discrete optimized protein energy
- GPU, Graphics Processing Unit
- GSH-like catalytic site
- GST, glutathione S-transferases
- GUI, Graphical User Interface
- HEPES, (4-(2-hydroxyethyl)− 1-piperazineethanesulfonic acid;
- HIF, Hypoxia-inducible factor
- HSQC, Heteronuclear single quantum coherence spectroscopy
- HUVEC, Human umbilical vein endothelial cells
- IKKβ, Inhibitor of nuclear kappa-B-kinase subunit beta
- JNK, c-Jun N-terminal kinase
- MKK6, Mitogen-activated protein kinase kinase-6
- MOI, Multiplicity of infection
- NF-κB, Nuclear factor kappa-light-chain-enhancer of activated B cells
- NMR, Nuclear magnetic resonance
- NPT, The constant-temperature, constant-pressure ensemble
- NaCL, Sodium chloride
- Nuclear magnetic resonance
- PAH, Pulmonary arterial hypertension
- RAPA, Rapamycin
- SASA, Solvent accessible surface area
- SEK1, Dual specificity mitogen-activated protein kinase kinase 4
- Smad, Suppressor of Mothers against Decapentaplegic
- Structure-based drug discovery
- TEV, Tobacco etch virus
- TIP3P, Transferable intermolecular potential 3 P
- TROSY, Transverse relaxation optimized spectroscopy
- UCSF, University of California, San Francisco
- VEGF, Vascular endothelial growth factor
- p38, Mitogen activated protein kinases
Collapse
|
21
|
Gao N, Zheng Q, Wang Y, Li X, Li Z, Xiao H. Wun2-mediated integrin recycling promotes apoptotic cell clearance in Drosophila melanogaster. Cell Death Differ 2022; 29:2545-2561. [PMID: 35840760 PMCID: PMC9751302 DOI: 10.1038/s41418-022-01039-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 06/25/2022] [Accepted: 07/04/2022] [Indexed: 01/31/2023] Open
Abstract
Apoptotic cell (AC) clearance is a complex process in which phagocytes recognize, engulf, and digest ACs during organismal development and tissue homeostasis. Impaired efferocytosis results in developmental defects and autoimmune diseases. In the current study, we performed RNA-sequencing to systematically identify regulators involved in the phagocytosis of ACs by Drosophila melanogaster macrophage-like S2 cells, followed by targeted RNA interference screening. Wunen2 (Wun2), a homolog of mammalian lipid phosphate phosphatase (LPP), was deemed as required for efferocytosis both in vitro and in vivo. However, efferocytosis was independent of Wun2 phosphatase activity. Proteomic analysis further revealed that Rab11 and its effector Rip11 are interaction partners of Wun2. Therefore, Wun2 collaborates with Rip11 and Rab11 to mediate efficient recycling of the phagocytic receptor βν integrin subunit to the plasma membrane. The loss of Wun2 results in the routing of βv integrin subunit (Itgbn) into lysosomes, leading to its degradation. The deficiency of βv integrin subunit on the cell surface leads to aberrant and disorganized actin cytoskeleton, thereby influencing the formation of macrophage pseudopodia toward ACs and thus failure to engulf them. The findings of this study provide insights that clarify how phagocytes coordinate AC signals and adopt a precise mechanism for the maintenance of engulfment receptors at their cell membrane surface to regulate efferocytosis.
Collapse
Affiliation(s)
- Ning Gao
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
- Medical College of Yan'an University, Yan'an, Shaanxi, 716000, China
| | - Qian Zheng
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Yanzhe Wang
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Xiaowen Li
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Zhi Li
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Hui Xiao
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China.
| |
Collapse
|
22
|
He Z, Wang J, Xu J, Jiang X, Liu X, Jiang J. Dynamic regulation of KIF15 phosphorylation and acetylation promotes focal adhesions disassembly in pancreatic cancer. Cell Death Dis 2022; 13:896. [PMID: 36280663 PMCID: PMC9592618 DOI: 10.1038/s41419-022-05338-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022]
Abstract
Pancreatic cancer (PC) is prone to distant metastasis in the early stage, which is attributed to the strong migration ability of tumor cells. Focal adhesion turnover is essential for cancer cell metastasis, and the integrin recycling process is a key activation pathway for focal adhesion depolymerization. To identify the key motor protein involving in the integrin β1 recycling, we screened kinesin proteins involved in integrin β1 recycling using a kinesin family siRNA library and identified kinesin family 15 (KIF15) as a key regulator. KIF15 was upregulated in metastasis PC tissues and promoted PC cell migration and invasion. We identified KIF15 as a key component mediating integrin β1/FAK signaling that accelerated FA disassembly in a FAK-Y397-dependent manner. KIF15 recruited PI3K-C2α to promote integrin β1/FAK signaling and FA disassembly in a RAB11A-dependent manner. The C-terminal tail of KIF15 is required for the PI3K-C2α interaction and RAB11A activation. In addition, we also found that SIRT1-mediated acetylation of KIF15 is essential for KIF15 phosphorylation, which is the key activation event in motor protein function. Together, these findings indicate that KIF15 interacts with PI3K-C2α to promote FA turnover in PC cells by controlling the endosome recycling of integrin β1 in a SIRT1 acetylation modification-dependent manner, eventually promoting focal adhesions turnover and distant metastasis in PC.
Collapse
Affiliation(s)
- Zhiwei He
- grid.412632.00000 0004 1758 2270Department of Hepatic-Biliary Surgery, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Jie Wang
- grid.412632.00000 0004 1758 2270Department of Hepatic-Biliary Surgery, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Jian Xu
- grid.412632.00000 0004 1758 2270Department of Hepatic-Biliary Surgery, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Xueyi Jiang
- grid.412632.00000 0004 1758 2270Department of Hepatic-Biliary Surgery, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Xinyuan Liu
- grid.412632.00000 0004 1758 2270Department of Hepatic-Biliary Surgery, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Jianxin Jiang
- grid.412632.00000 0004 1758 2270Department of Hepatic-Biliary Surgery, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| |
Collapse
|
23
|
Ozaki S, Mikami K, Kunieda T, Tanaka J. Chloride Intracellular Channel Proteins (CLICs) and Malignant Tumor Progression: A Focus on the Preventive Role of CLIC2 in Invasion and Metastasis. Cancers (Basel) 2022; 14:cancers14194890. [PMID: 36230813 PMCID: PMC9562003 DOI: 10.3390/cancers14194890] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 10/03/2022] [Accepted: 10/03/2022] [Indexed: 11/27/2022] Open
Abstract
Simple Summary Although chloride intracellular channel proteins (CLICs) have been identified as ion channel proteins, their true functions are still elusive. Recent in silico analyses show that CLICs may be prognostic markers in cancer. This review focuses on CLIC2 that plays preventive roles in malignant cell invasion and metastasis. CLIC2 is secreted extracellularly and binds to matrix metalloproteinase 14 (MMP14), while inhibiting its activity. As a result, CLIC2 may contribute to the development/maintenance of junctions between blood vessel endothelial cells and the inhibition of invasion and metastasis of tumor cells. CLIC2 may be a novel therapeutic target for malignancies. Abstract CLICs are the dimorphic protein present in both soluble and membrane fractions. As an integral membrane protein, CLICs potentially possess ion channel activity. However, it is not fully clarified what kinds of roles CLICs play in physiological and pathological conditions. In vertebrates, CLICs are classified into six classes: CLIC1, 2, 3, 4, 5, and 6. Recently, in silico analyses have revealed that the expression level of CLICs may have prognostic significance in cancer. In this review, we focus on CLIC2, which has received less attention than other CLICs, and discuss its role in the metastasis and invasion of malignant tumor cells. CLIC2 is expressed at higher levels in benign tumors than in malignant ones, most likely preventing tumor cell invasion into surrounding tissues. CLIC2 is also expressed in the vascular endothelial cells of normal tissues and maintains their intercellular adhesive junctions, presumably suppressing the hematogenous metastasis of malignant tumor cells. Surprisingly, CLIC2 is localized in secretory granules and secreted into the extracellular milieu. Secreted CLIC2 binds to MMP14 and inhibits its activity, leading to suppressed MMP2 activity. CLIC4, on the other hand, promotes MMP14 activity. These findings challenge the assumption that CLICs are ion channels, implying that they could be potential new targets for the treatment of malignant tumors.
Collapse
Affiliation(s)
- Saya Ozaki
- Department of Neurosurgery, Graduate School of Medicine, Ehime University, Toon 791-0295, Japan
- Department of Neurosurgery, National Cerebral and Cardiovascular Center Hospital, Suita 564-8565, Japan
- Correspondence: (S.O.); (J.T.)
| | - Kanta Mikami
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Toon 791-0295, Japan
| | - Takeharu Kunieda
- Department of Neurosurgery, Graduate School of Medicine, Ehime University, Toon 791-0295, Japan
| | - Junya Tanaka
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Toon 791-0295, Japan
- Correspondence: (S.O.); (J.T.)
| |
Collapse
|
24
|
Mahmutefendić Lučin H, Blagojević Zagorac G, Marcelić M, Lučin P. Host Cell Signatures of the Envelopment Site within Beta-Herpes Virions. Int J Mol Sci 2022; 23:9994. [PMID: 36077391 PMCID: PMC9456339 DOI: 10.3390/ijms23179994] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 11/26/2022] Open
Abstract
Beta-herpesvirus infection completely reorganizes the membrane system of the cell. This system is maintained by the spatiotemporal arrangement of more than 3000 cellular proteins that continuously adapt the configuration of membrane organelles according to cellular needs. Beta-herpesvirus infection establishes a new configuration known as the assembly compartment (AC). The AC membranes are loaded with virus-encoded proteins during the long replication cycle and used for the final envelopment of the newly formed capsids to form infectious virions. The identity of the envelopment membranes is still largely unknown. Electron microscopy and immunofluorescence studies suggest that the envelopment occurs as a membrane wrapping around the capsids, similar to the growth of phagophores, in the area of the AC with the membrane identities of early/recycling endosomes and the trans-Golgi network. During wrapping, host cell proteins that define the identity and shape of these membranes are captured along with the capsids and incorporated into the virions as host cell signatures. In this report, we reviewed the existing information on host cell signatures in human cytomegalovirus (HCMV) virions. We analyzed the published proteomes of the HCMV virion preparations that identified a large number of host cell proteins. Virion purification methods are not yet advanced enough to separate all of the components of the rich extracellular material, including the large amounts of non-vesicular extracellular particles (NVEPs). Therefore, we used the proteomic data from large and small extracellular vesicles (lEVs and sEVs) and NVEPs to filter out the host cell proteins identified in the viral proteomes. Using these filters, we were able to narrow down the analysis of the host cell signatures within the virions and determine that envelopment likely occurs at the membranes derived from the tubular recycling endosomes. Many of these signatures were also found at the autophagosomes, suggesting that the CMV-infected cell forms membrane organelles with phagophore growth properties using early endosomal host cell machinery that coordinates endosomal recycling.
Collapse
Affiliation(s)
| | | | | | - Pero Lučin
- Department of Physiology, Immunology and Pathophysiology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| |
Collapse
|
25
|
Meecham A, Cutmore LC, Protopapa P, Rigby LG, Marshall JF. Ligand-bound integrin αvβ6 internalisation and trafficking. Front Cell Dev Biol 2022; 10:920303. [PMID: 36092709 PMCID: PMC9448872 DOI: 10.3389/fcell.2022.920303] [Citation(s) in RCA: 6] [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: 04/14/2022] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
The integrin αvβ6 is expressed at low levels in most normal healthy tissue but is very often upregulated in a disease context including cancer and fibrosis. Integrins use endocytosis and trafficking as a means of regulating their surface expression and thus their functions, however little is known of how this process is regulated in the context of αvβ6. As αvβ6 is a major target for the development of therapeutics in cancer and fibrosis, understanding these dynamics is critical in the development of αvβ6-targeted therapies. Following development of a flow cytometry-based assay to measure ligand (A20FMDV2 or LAP)-bound αvβ6 endocytosis, an siRNA screen was performed to identify which genes were responsible for internalising αvβ6. These data identified 15 genes (DNM2, CBLB, DNM3, CBL, EEA1, CLTC, ARFGAP3, CAV1, CYTH2, CAV3, CAV2, IQSEC1, AP2M1, TSG101) which significantly decreased endocytosis, predominantly within dynamin-dependent pathways. Inhibition of these dynamin-dependent pathways significantly reduced αvβ6-dependent migration (αvβ6-specific migration was 547 ± 128 under control conditions, reduced to 225 ± 73 with clathrin inhibition, and 280 ± 51 with caveolin inhibition). Colocalization studies of αvβ6 with endosome markers revealed that up to 6 h post-internalisation of ligand, αvβ6 remains in Rab11-positive endosomes in a perinuclear location, with no evidence of αvβ6 degradation up to 48 h post exposure to A20FMDV2. Additionally, 60% of ligand-bound αvβ6 was recycled back to the surface by 6 h. With studies ongoing using conjugated A20FMDV2 to therapeutically target αvβ6 in cancer and fibrosis, these data have important implications. Binding of A20FMDV2 seemingly removes much of the αvβ6 from the cell membrane, and upon its recycling, a large fraction appears to still be in the ligand-bound state. While these results are observed with A20FMDV2, these data will be of value in the design of αvβ6-specific therapeutics and potentially the types of therapeutic load.
Collapse
Affiliation(s)
- Amelia Meecham
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
- University of California, San Diego, San Diego, CA, United States
| | - Lauren C. Cutmore
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Pantelitsa Protopapa
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Lauren G. Rigby
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - John F. Marshall
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
26
|
Kuo IY, Hsieh CH, Kuo WT, Chang CP, Wang YC. Recent advances in conventional and unconventional vesicular secretion pathways in the tumor microenvironment. J Biomed Sci 2022; 29:56. [PMID: 35927755 PMCID: PMC9354273 DOI: 10.1186/s12929-022-00837-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 07/18/2022] [Indexed: 11/18/2022] Open
Abstract
All cells in the changing tumor microenvironment (TME) need a class of checkpoints to regulate the balance among exocytosis, endocytosis, recycling and degradation. The vesicular trafficking and secretion pathways regulated by the small Rab GTPases and their effectors convey cell growth and migration signals and function as meditators of intercellular communication and molecular transfer. Recent advances suggest that Rab proteins govern conventional and unconventional vesicular secretion pathways by trafficking widely diverse cargoes and substrates in remodeling TME. The mechanisms underlying the regulation of conventional and unconventional vesicular secretion pathways, their action modes and impacts on the cancer and stromal cells have been the focus of much attention for the past two decades. In this review, we discuss the current understanding of vesicular secretion pathways in TME. We begin with an overview of the structure, regulation, substrate recognition and subcellular localization of vesicular secretion pathways. We then systematically discuss how the three fundamental vesicular secretion processes respond to extracellular cues in TME. These processes are the conventional protein secretion via the endoplasmic reticulum-Golgi apparatus route and two types of unconventional protein secretion via extracellular vesicles and secretory autophagy. The latest advances and future directions in vesicular secretion-involved interplays between tumor cells, stromal cell and host immunity are also described.
Collapse
Affiliation(s)
- I-Ying Kuo
- Department of Pharmacology, College of Medicine, National Cheng Kung University, No.1, University Road, Tainan, 701, Taiwan.,Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Hsiung Hsieh
- Department of Pharmacology, College of Medicine, National Cheng Kung University, No.1, University Road, Tainan, 701, Taiwan
| | - Wan-Ting Kuo
- Department of Pharmacology, College of Medicine, National Cheng Kung University, No.1, University Road, Tainan, 701, Taiwan.,Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, No.1, University Road, Tainan, 701, Taiwan
| | - Chih-Peng Chang
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, No.1, University Road, Tainan, 701, Taiwan. .,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| | - Yi-Ching Wang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, No.1, University Road, Tainan, 701, Taiwan. .,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
27
|
Linking Late Endosomal Cholesterol with Cancer Progression and Anticancer Drug Resistance. Int J Mol Sci 2022; 23:ijms23137206. [PMID: 35806209 PMCID: PMC9267071 DOI: 10.3390/ijms23137206] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 06/22/2022] [Accepted: 06/25/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer cells undergo drastic metabolic adaptions to cover increased bioenergetic needs, contributing to resistance to therapies. This includes a higher demand for cholesterol, which often coincides with elevated cholesterol uptake from low-density lipoproteins (LDL) and overexpression of the LDL receptor in many cancers. This implies the need for cancer cells to accommodate an increased delivery of LDL along the endocytic pathway to late endosomes/lysosomes (LE/Lys), providing a rapid and effective distribution of LDL-derived cholesterol from LE/Lys to other organelles for cholesterol to foster cancer growth and spread. LDL-cholesterol exported from LE/Lys is facilitated by Niemann–Pick Type C1/2 (NPC1/2) proteins, members of the steroidogenic acute regulatory-related lipid transfer domain (StARD) and oxysterol-binding protein (OSBP) families. In addition, lysosomal membrane proteins, small Rab GTPases as well as scaffolding proteins, including annexin A6 (AnxA6), contribute to regulating cholesterol egress from LE/Lys. Here, we summarize current knowledge that links upregulated activity and expression of cholesterol transporters and related proteins in LE/Lys with cancer growth, progression and treatment outcomes. Several mechanisms on how cellular distribution of LDL-derived cholesterol from LE/Lys influences cancer cell behavior are reviewed, some of those providing opportunities for treatment strategies to reduce cancer progression and anticancer drug resistance.
Collapse
|
28
|
Larocque G, Royle SJ. Integrating intracellular nanovesicles into integrin trafficking pathways and beyond. Cell Mol Life Sci 2022; 79:335. [PMID: 35657500 PMCID: PMC9166830 DOI: 10.1007/s00018-022-04371-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 04/28/2022] [Accepted: 05/11/2022] [Indexed: 12/24/2022]
Abstract
Membrane traffic controls the movement of proteins and lipids from one cellular compartment to another using a system of transport vesicles. Intracellular nanovesicles (INVs) are a newly described class of transport vesicles. These vesicles are small, carry diverse cargo, and are involved in multiple trafficking steps including anterograde traffic and endosomal recycling. An example of a biological process that they control is cell migration and invasion, due to their role in integrin recycling. In this review, we describe what is known so far about these vesicles. We discuss how INVs may integrate into established membrane trafficking pathways using integrin recycling as an example. We speculate where in the cell INVs have the potential to operate and we identify key questions for future investigation.
Collapse
Affiliation(s)
| | - Stephen J Royle
- Centre for Mechanochemical Cell Biology, Warwick Medical School, Gibbet Hill Road, Coventry, CV4 7AL, UK.
| |
Collapse
|
29
|
Ariano C, Riganti C, Corà D, Valdembri D, Mana G, Astanina E, Serini G, Bussolino F, Doronzo G. TFEB controls integrin-mediated endothelial cell adhesion by the regulation of cholesterol metabolism. Angiogenesis 2022; 25:471-492. [PMID: 35545719 PMCID: PMC9519734 DOI: 10.1007/s10456-022-09840-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 04/18/2022] [Indexed: 11/30/2022]
Abstract
The dynamic integrin-mediated adhesion of endothelial cells (ECs) to the surrounding ECM is fundamental for angiogenesis both in physiological and pathological conditions, such as embryonic development and cancer progression. The dynamics of EC-to-ECM adhesions relies on the regulation of the conformational activation and trafficking of integrins. Here, we reveal that oncogenic transcription factor EB (TFEB), a known regulator of lysosomal biogenesis and metabolism, also controls a transcriptional program that influences the turnover of ECM adhesions in ECs by regulating cholesterol metabolism. We show that TFEB favors ECM adhesion turnover by promoting the transcription of genes that drive the synthesis of cholesterol, which promotes the aggregation of caveolin-1, and the caveolin-dependent endocytosis of integrin β1. These findings suggest that TFEB might represent a novel target for the pharmacological control of pathological angiogenesis and bring new insights in the mechanism sustaining TFEB control of endocytosis.
Collapse
Affiliation(s)
- Camilla Ariano
- Department of Oncology, University of Torino, Candiolo, Italy.,Candiolo Cancer Institute- FPO-IRCCS, Candiolo, Italy
| | - Chiara Riganti
- Department of Oncology, University of Torino, Torino, Italy
| | - Davide Corà
- Department of Translational Medicine, Piemonte Orientale University, Novara, Italy.,Center for Translational Research on Autoimmune and Allergic Diseases-CAAD, Novara, Italy
| | - Donatella Valdembri
- Department of Oncology, University of Torino, Candiolo, Italy.,Candiolo Cancer Institute- FPO-IRCCS, Candiolo, Italy
| | - Giulia Mana
- Department of Oncology, University of Torino, Candiolo, Italy.,Candiolo Cancer Institute- FPO-IRCCS, Candiolo, Italy
| | - Elena Astanina
- Department of Oncology, University of Torino, Candiolo, Italy.,Candiolo Cancer Institute- FPO-IRCCS, Candiolo, Italy
| | - Guido Serini
- Department of Oncology, University of Torino, Candiolo, Italy.,Candiolo Cancer Institute- FPO-IRCCS, Candiolo, Italy
| | - Federico Bussolino
- Department of Oncology, University of Torino, Candiolo, Italy. .,Candiolo Cancer Institute- FPO-IRCCS, Candiolo, Italy.
| | - Gabriella Doronzo
- Department of Oncology, University of Torino, Candiolo, Italy. .,Candiolo Cancer Institute- FPO-IRCCS, Candiolo, Italy.
| |
Collapse
|
30
|
Molnár M, Sőth Á, Simon-Vecsei Z. Pathways of integrins in the endo-lysosomal system. Biol Futur 2022; 73:171-185. [DOI: 10.1007/s42977-022-00120-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 04/09/2022] [Indexed: 12/13/2022]
Abstract
AbstractIn this review, we present recent scientific advances about integrin trafficking in the endo-lysosomal system. In the last few years, plenty of new information has emerged about the endo-lysosomal system, integrins, and the mechanism, how exactly the intracellular trafficking of integrins is regulated. We review the internalization and recycling pathways of integrins, and we provide information about the possible ways of lysosomal degradation through the endosomal and autophagic system. The regulation of integrin internalization and recycling proved to be a complex process worth studying. Trafficking of integrins, together with the regulation of their gene expression, defines cellular adhesion and cellular migration through bidirectional signalization and ligand binding. Thus, any malfunction in this system can potentially (but not necessarily) lead to tumorigenesis or metastasis. Hence, extensive examinations of integrins in the endo-lysosomal system raise the possibility to identify potential new medical targets. Furthermore, this knowledge can also serve as a basis for further determination of integrin signaling- and adhesion-related processes.
Collapse
|
31
|
Alves LS, Marques ARA, Padrão N, Carvalho FA, Ramalho J, Lopes CS, Soares MIL, Futter CE, Pinho E Melo TMVD, Santos NC, Vieira OV. Cholesteryl hemiazelate causes lysosome dysfunction impacting vascular smooth muscle cell homeostasis. J Cell Sci 2022; 135:272202. [PMID: 34528688 DOI: 10.1242/jcs.254631] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 09/07/2021] [Indexed: 01/07/2023] Open
Abstract
In atherosclerotic lesions, vascular smooth muscle cells (VSMCs) represent half of the foam cell population, which is characterized by an aberrant accumulation of undigested lipids within lysosomes. Loss of lysosome function impacts VSMC homeostasis and disease progression. Understanding the molecular mechanisms underlying lysosome dysfunction in these cells is, therefore, crucial. We identify cholesteryl hemiazelate (ChA), a stable oxidation end-product of cholesteryl-polyunsaturated fatty acid esters, as an inducer of lysosome malfunction in VSMCs. ChA-treated VSMCs acquire a foam-cell-like phenotype, characterized by enlarged lysosomes full of ChA and neutral lipids. The lysosomes are perinuclear and exhibit degradative capacity and cargo exit defects. Lysosome luminal pH is also altered. Even though the transcriptional response machinery and autophagy are not activated by ChA, the addition of recombinant lysosomal acid lipase (LAL) is able to rescue lysosome dysfunction. ChA significantly affects VSMC proliferation and migration, impacting atherosclerosis. In summary, this work shows that ChA is sufficient to induce lysosomal dysfunction in VSMCs, that, in ChA-treated VSMCs, neither lysosome biogenesis nor autophagy are triggered, and, finally, that recombinant LAL can be a therapeutic approach for lysosomal dysfunction.
Collapse
Affiliation(s)
- Liliana S Alves
- Chronic Diseases Research Centre (CEDOC), NOVA Medical School, NOVA University Lisbon, 1169-056 Lisboa, Portugal
| | - André R A Marques
- Chronic Diseases Research Centre (CEDOC), NOVA Medical School, NOVA University Lisbon, 1169-056 Lisboa, Portugal
| | - Nuno Padrão
- Chronic Diseases Research Centre (CEDOC), NOVA Medical School, NOVA University Lisbon, 1169-056 Lisboa, Portugal
| | - Filomena A Carvalho
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa 1649-028, Lisboa, Portugal
| | - José Ramalho
- Chronic Diseases Research Centre (CEDOC), NOVA Medical School, NOVA University Lisbon, 1169-056 Lisboa, Portugal
| | - Catarina S Lopes
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa 1649-028, Lisboa, Portugal
| | - Maria I L Soares
- CQC and Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal
| | - Clare E Futter
- Department of Cell Biology, UCL Institute of Ophthalmology, London EC1V 9EL, UK
| | | | - Nuno C Santos
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa 1649-028, Lisboa, Portugal
| | - Otília V Vieira
- Chronic Diseases Research Centre (CEDOC), NOVA Medical School, NOVA University Lisbon, 1169-056 Lisboa, Portugal
| |
Collapse
|
32
|
Roblek M, Bicher J, van Gogh M, György A, Seeböck R, Szulc B, Damme M, Olczak M, Borsig L, Siekhaus DE. The Solute Carrier MFSD1 Decreases the Activation Status of β1 Integrin and Thus Tumor Metastasis. Front Oncol 2022; 12:777634. [PMID: 35211397 PMCID: PMC8861502 DOI: 10.3389/fonc.2022.777634] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 01/13/2022] [Indexed: 11/17/2022] Open
Abstract
Solute carriers are increasingly recognized as participating in a plethora of pathologies, including cancer. We describe here the involvement of the orphan solute carrier Major Facilitator Superfamily Domain-containing protein 1 (MFSD1) in the regulation of tumor cell migration. Loss of MFSD1 enabled higher levels of metastasis in experimental and spontaneous metastasis mouse models. We identified an increased migratory potential in MFSD1−/− tumor cells which was mediated by increased focal adhesion turnover, reduced stability of mature inactive β1 integrin, and the resulting increased integrin activation index. We show that MFSD1 promoted recycling to the cell surface of endocytosed inactive β1 integrin and thereby protected β1 integrin from proteolytic degradation; this led to dampening of the integrin activation index. Furthermore, downregulation of MFSD1 expression was observed during the early steps of tumorigenesis, and higher MFSD1 expression levels correlate with a better cancer patient prognosis. In sum, we describe a requirement for endolysosomal MFSD1 in efficient β1 integrin recycling to suppress tumor cell dissemination.
Collapse
Affiliation(s)
- Marko Roblek
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Julia Bicher
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Merel van Gogh
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Attila György
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Rita Seeböck
- Institute of Clinical Pathology, University Hospital St. Polten, St. Polten, Austria
| | - Bozena Szulc
- Laboratory of Biochemistry, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Markus Damme
- Institute of Biochemistry, University of Kiel, Kiel, Germany
| | - Mariusz Olczak
- Laboratory of Biochemistry, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Lubor Borsig
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Daria E Siekhaus
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| |
Collapse
|
33
|
Chloride Channels and Transporters: Roles beyond Classical Cellular Homeostatic pH or Ion Balance in Cancers. Cancers (Basel) 2022; 14:cancers14040856. [PMID: 35205604 PMCID: PMC8870652 DOI: 10.3390/cancers14040856] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/28/2022] [Accepted: 02/06/2022] [Indexed: 01/04/2023] Open
Abstract
Simple Summary Roles of chloride-associated transporters have been raised in various cancers. Although complicated ion movements, crosstalk among channels/transporters through homeostatic electric regulation, difficulties with experimental implementation such as activity measurement of intracellular location were disturbed to verify the precise modulation of channels/transporters, recently defined cancerous function and communication with tumor microenvironment of chloride channels/transporters should be highlighted beyond classical homeostatic ion balance. Chloride-associated transporters as membrane-associated components of chloride movement, regulations of transmembrane member 16A, calcium-activated chloride channel regulators, transmembrane member 206, chloride intracellular channels, voltage-gated chloride channels, cystic fibrosis transmembrane conductance regulator, voltage-dependent anion channel, volume-regulated anion channel, and chloride-bicarbonate exchangers are discussed. Abstract The canonical roles of chloride channels and chloride-associated transporters have been physiologically determined; these roles include the maintenance of membrane potential, pH balance, and volume regulation and subsequent cellular functions such as autophagy and cellular proliferative processes. However, chloride channels/transporters also play other roles, beyond these classical function, in cancerous tissues and under specific conditions. Here, we focused on the chloride channel-associated cancers and present recent advances in understanding the environments of various types of cancer caused by the participation of many chloride channel or transporters families and discuss the challenges and potential targets for cancer treatment. The modulation of chloride channels/transporters might promote new aspect of cancer treatment strategies.
Collapse
|
34
|
Jiang X, Yang L, Gao Q, Liu Y, Feng X, Ye S, Yang Z. The Role of RAB GTPases and Its Potential in Predicting Immunotherapy Response and Prognosis in Colorectal Cancer. Front Genet 2022; 13:828373. [PMID: 35154286 PMCID: PMC8833848 DOI: 10.3389/fgene.2022.828373] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 01/10/2022] [Indexed: 12/29/2022] Open
Abstract
Background: Colorectal cancer (CRC) is the third most common cancer worldwide, in which aberrant activation of the RAS signaling pathway appears frequently. RAB proteins (RABs) are the largest Ras small GTPases superfamily that regulates intracellular membrane trafficking pathways. The dysregulation of RABs have been found in various diseases including cancers. Compared with other members of Ras families, the roles of RABs in colorectal cancer are less well understood. Methods: We analyzed the differential expression and clinicopathological association of RABs in CRC using RNA sequencing and genotyping datasets from TCGA samples. Moreover, the biological function of RAB17 and RAB34 were investigated in CRC cell lines and patient samples. Results: Of the 62 RABs we analyzed in CRC, seven (RAB10, RAB11A, RAB15, RAB17, RAB19, RAB20, and RAB25) were significantly upregulated, while six (RAB6B, RAB9B, RAB12, RAB23, RAB31, and RAB34) were significantly downregulated in tumor tissues as compared to normal. We found that the upregulated-RABs, which were highly expressed in metabolic activated CRC subtype (CMS3), are associated with cell cycle related pathways enrichment and positively correlated with the mismatch repair (MMR) genes in CRC, implying their role in regulating cell metabolism and tumor growth. While, high expression of the downregulated-RABs were significantly associated with poor prognostic CRC mesenchymal subtypes (CMS4), immune checkpoint genes, and tumor infiltrating immune cells, indicating their role in predicting prognosis and immunotherapy efficacy. Interestingly, though RAB34 mRNA is downregulated in CRC, its high expression is significantly associated with poor prognosis. In vitro experiments showed that RAB17 overexpression can promote cell proliferation via cell cycle regulation. While, RAB34 overexpression can promote cell migration and invasion and is associated with PD-L1/PD-L2 expression increase in CRC cells. Conclusions: Our study showed that RABs may play important roles in regulating cell cycle and immune-related pathways, therefore might be potential biomarkers in predicting prognosis and immunotherapy response in CRC.
Collapse
|
35
|
Sheikh A, Alhakamy NA, Md S, Kesharwani P. Recent Progress of RGD Modified Liposomes as Multistage Rocket Against Cancer. Front Pharmacol 2022; 12:803304. [PMID: 35145405 PMCID: PMC8822168 DOI: 10.3389/fphar.2021.803304] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/22/2021] [Indexed: 12/24/2022] Open
Abstract
Cancer is a life-threatening disease, contributing approximately 9.4 million deaths worldwide. To address this challenge, scientific researchers have investigated molecules that could act as speed-breakers for cancer. As an abiotic drug delivery system, liposomes can hold both hydrophilic and lipophilic drugs, which promote a controlled release, accumulate in the tumor microenvironment, and achieve elongated half-life with an enhanced safety profile. To further improve the safety and impair the off-target effect, the surface of liposomes could be modified in a way that is easily identified by cancer cells, promotes uptake, and facilitates angiogenesis. Integrins are overexpressed on cancer cells, which upon activation promote downstream cell signaling and eventually activate specific pathways, promoting cell growth, proliferation, and migration. RGD peptides are easily recognized by integrin over expressed cells. Just like a multistage rocket, ligand anchored liposomes can be selectively recognized by target cells, accumulate at the specific site, and finally, release the drug in a specific and desired way. This review highlights the role of integrin in cancer development, so gain more insights into the phenomenon of tumor initiation and survival. Since RGD is recognized by the integrin family, the fate of RGD has been demonstrated after its binding with the acceptor’s family. The role of RGD based liposomes in targeting various cancer cells is also highlighted in the paper.
Collapse
Affiliation(s)
- Afsana Sheikh
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Nabil A. Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Shadab Md
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
- *Correspondence: Prashant Kesharwani,
| |
Collapse
|
36
|
Sharma P, Parveen S, Datta S. Measuring Plasma Membrane Recycling Using Microscopic and Biochemical Approaches. Methods Mol Biol 2022; 2473:237-257. [PMID: 35819770 DOI: 10.1007/978-1-0716-2209-4_17] [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: 06/15/2023]
Abstract
The endocytic pathway has an intricate network of vesicular compartments carrying a variety of proteins referred to as cargoes. Endosomal trafficking is exclusively required to transport these cargoes through various intracellular routes for their delivery to the site of action. Among these, recycling of cargoes to the plasma membrane is a crucial pathway for the efficient functioning of the cell. Hence, endosomal cargo recycling assays are crucial to gain insight into the molecular mechanism governing recycling of the cargoes and in turn to understand their key role in maintaining cellular physiology. These assays have been efficiently utilized to study the recycling of adhesion molecules, transporters, channels, receptors, and so on to the plasma membrane. The basic methodology involves labelling of the cargo at the surface, allowing its internalization followed by direct or indirect measurement of the amount of the cargo recycled back to the plasma membrane. These microscopy-based and biochemical methods can be used as a tool to study the role of various trafficking or signaling molecules on the cell surface involved with the recycling of the membrane proteins, by altering their expression either by silencing or overexpressing the gene.
Collapse
Affiliation(s)
- Priyanka Sharma
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, India.
| | - Sameena Parveen
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, India
| | - Sunando Datta
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, India
| |
Collapse
|
37
|
Zhang J, Jiang Z, Shi A. Rab GTPases: The principal players in crafting the regulatory landscape of endosomal trafficking. Comput Struct Biotechnol J 2022; 20:4464-4472. [PMID: 36051867 PMCID: PMC9418685 DOI: 10.1016/j.csbj.2022.08.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 08/05/2022] [Accepted: 08/07/2022] [Indexed: 11/16/2022] Open
Abstract
After endocytosis, diverse cargos are sorted into endosomes and directed to various destinations, including extracellular macromolecules, membrane lipids, and membrane proteins. Some cargos are returned to the plasma membrane via endocytic recycling. In contrast, others are delivered to the Golgi apparatus through the retrograde pathway, while the rest are transported to late endosomes and eventually to lysosomes for degradation. Rab GTPases are major regulators that ensure cargos are delivered to their proper destinations. Rabs are localized to distinct endosomes and play predominant roles in membrane budding, vesicle formation and motility, vesicle tethering, and vesicle fusion by recruiting effectors. The cascades between Rabs via shared effectors or the recruitment of Rab activators provide an additional layer of spatiotemporal regulation of endocytic trafficking. Notably, several recent studies have indicated that disorders of Rab-mediated endocytic transports are closely associated with diseases such as immunodeficiency, cancer, and neurological disorders.
Collapse
|
38
|
Horikawa M, Sabe H, Onodera Y. Dual roles of AMAP1 in the transcriptional regulation and intracellular trafficking of carbonic anhydrase IX. Transl Oncol 2022; 15:101258. [PMID: 34742153 PMCID: PMC8577137 DOI: 10.1016/j.tranon.2021.101258] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 10/20/2021] [Accepted: 10/25/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND The cell-surface enzyme carbonic anhydrase IX (CAIX/CA9) promotes tumor growth, survival, invasion, and metastasis, mainly via its pH-regulating functions. Owing to its tumor-specific expression, CAIX-targeting antibodies/chemicals are utilized for therapeutic and diagnostic purposes. However, mechanisms of CAIX trafficking, which affects such CAIX-targeting modalities remain unclear. In this study, roles of the AMAP1-PRKD2 pathway, which mediates integrin recycling of invasive cancer cells, in CAIX trafficking were investigated. METHODS Using highly invasive MDA-MB-231 breast cancer cells, the physical association and colocalization of endogenous proteins were analyzed by immunoprecipitation and immunofluorescence, protein/mRNA levels were quantified by western blotting/qPCR, and cell-surface transport and intracellular/extracellular pH regulation were measured by biotin-labeling and fluorescent dye-based assays, respectively. The correlation between mRNA levels and patients' prognoses was analyzed using a TCGA breast cancer dataset. RESULTS AMAP1 associated with the CAIX protein complex, and they colocalized at the plasma membrane and tubulovesicular structures. AMAP1 knockdown reduced total/surface CAIX, induced its lysosomal accumulation and degradation, and affected intracellular/extracellular pH. PRKD2 knockdown excluded AMAP1 from the CAIX complex and reduced total CAIX in a lysosome-dependent manner. Unexpectedly, AMAP1 knockdown also reduced CAIX mRNA. AMAP1 interacted with PIAS3, which stabilizes HIF-1α, a transcriptional regulator of CA9. AMAP1 knockdown inhibited the PIAS3-HIF-1α interaction and destabilized the HIF-1α protein. High-ASAP1 (AMAP1-encoding gene) together with high-PIAS3 correlated with high-CA9 and an unfavorable prognosis in breast cancer. CONCLUSION The AMAP1-PRKD2 pathway regulates CAIX trafficking, and modulates its total/surface expression. The AMAP1-PIAS3 interaction augments CA9 transcription by stabilizing HIF-1α, presumably contributing to an unfavorable prognosis.
Collapse
Affiliation(s)
- Mei Horikawa
- Department of Molecular Biology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15W7 Kita-ku, Sapporo, Hokkaido 060-8638, Japan
| | - Hisataka Sabe
- Department of Molecular Biology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15W7 Kita-ku, Sapporo, Hokkaido 060-8638, Japan.
| | - Yasuhito Onodera
- Department of Molecular Biology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15W7 Kita-ku, Sapporo, Hokkaido 060-8638, Japan; Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, N15W7 Kita-ku, Sapporo, Hokkaido 060-8638, Japan.
| |
Collapse
|
39
|
Rabas N, Palmer S, Mitchell L, Ismail S, Gohlke A, Riley JS, Tait SW, Gammage P, Soares LL, Macpherson IR, Norman JC. PINK1 drives production of mtDNA-containing extracellular vesicles to promote invasiveness. J Cell Biol 2021; 220:e202006049. [PMID: 34623384 PMCID: PMC8641410 DOI: 10.1083/jcb.202006049] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/29/2021] [Accepted: 09/15/2021] [Indexed: 12/11/2022] Open
Abstract
The cystine-glutamate antiporter, xCT, supports a glutathione synthesis program enabling cancer cells to cope with metabolically stressful microenvironments. Up-regulated xCT, in combination with glutaminolysis, leads to increased extracellular glutamate, which promotes invasive behavior by activating metabotropic glutamate receptor 3 (mGluR3). Here we show that activation of mGluR3 in breast cancer cells activates Rab27-dependent release of extracellular vesicles (EVs), which can transfer invasive characteristics to "recipient" tumor cells. These EVs contain mitochondrial DNA (mtDNA), which is packaged via a PINK1-dependent mechanism. We highlight mtDNA as a key EV cargo necessary and sufficient for intercellular transfer of invasive behavior by activating Toll-like receptor 9 in recipient cells, and this involves increased endosomal trafficking of pro-invasive receptors. We propose that an EV-mediated mechanism, through which altered cellular metabolism in one cell influences endosomal trafficking in other cells, is key to generation and dissemination of pro-invasive microenvironments during mammary carcinoma progression.
Collapse
Affiliation(s)
| | - Sarah Palmer
- Beatson Institute for Cancer Research, Glasgow, UK
| | | | | | | | - Joel S. Riley
- Beatson Institute for Cancer Research, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Stephen W.G. Tait
- Beatson Institute for Cancer Research, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Payam Gammage
- Beatson Institute for Cancer Research, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Leandro Lemgruber Soares
- Glasgow Imaging Facility, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Iain R. Macpherson
- Beatson Institute for Cancer Research, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Jim C. Norman
- Beatson Institute for Cancer Research, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
40
|
Hernandez SJ, Fote G, Reyes-Ortiz AM, Steffan JS, Thompson LM. Cooperation of cell adhesion and autophagy in the brain: Functional roles in development and neurodegenerative disease. Matrix Biol Plus 2021; 12:100089. [PMID: 34786551 PMCID: PMC8579148 DOI: 10.1016/j.mbplus.2021.100089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/11/2021] [Accepted: 10/18/2021] [Indexed: 12/19/2022] Open
Abstract
Cellular adhesive connections directed by the extracellular matrix (ECM) and maintenance of cellular homeostasis by autophagy are seemingly disparate functions that are molecularly intertwined, each regulating the other. This is an emerging field in the brain where the interplay between adhesion and autophagy functions at the intersection of neuroprotection and neurodegeneration. The ECM and adhesion proteins regulate autophagic responses to direct protein clearance and guide regenerative programs that go awry in brain disorders. Concomitantly, autophagic flux acts to regulate adhesion dynamics to mediate neurite outgrowth and synaptic plasticity with functional disruption contributed by neurodegenerative disease. This review highlights the cooperative exchange between cellular adhesion and autophagy in the brain during health and disease. As the mechanistic alliance between adhesion and autophagy has been leveraged therapeutically for metastatic disease, understanding overlapping molecular functions that direct the interplay between adhesion and autophagy might uncover therapeutic strategies to correct or compensate for neurodegeneration.
Collapse
Affiliation(s)
- Sarah J. Hernandez
- Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
| | - Gianna Fote
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA
| | - Andrea M. Reyes-Ortiz
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA
| | - Joan S. Steffan
- Psychaitry and Human Behavior, University of California Irvine, Irvine, CA 92697, USA
- Institute of Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92617, USA
| | - Leslie M. Thompson
- Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA
- Psychaitry and Human Behavior, University of California Irvine, Irvine, CA 92697, USA
- Institute of Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92617, USA
| |
Collapse
|
41
|
Derivation and Comprehensive Analysis of Aging Patterns in Patients with Bladder Cancer. DISEASE MARKERS 2021; 2021:3385058. [PMID: 34721733 PMCID: PMC8553474 DOI: 10.1155/2021/3385058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 09/29/2021] [Indexed: 12/24/2022]
Abstract
Background Aging is an essential risk factor for cancer. However, aging-related genes (ARGs) have not been comprehensively analyzed in bladder cancer (BC). Therefore, the study is aimed at derivating a risk stratification system for BC patients based on ARGs. Methods Public databases were used to acquire ARGs sets, transcriptome files, and clinical data. The “limma” package was then used to screen for differential ARGs while also using univariate Cox regression analysis to explore for prognostic ARGs. The “ConsensusClusterPlus” package was used to perform aging patterns in BC patients based on the above prognostic ARGs. Subsequently, aging patterns were investigated in survival prediction, mutation landscape, immunotherapy, immunological checkpoints, and immune microenvironment. We likewise utilized gene enrichment analysis to explore the biological functions that were behind the findings. To construct a risk signature and nonogram for prognostic prediction, we used LASSO and Cox regression analysis based on differential genes in aging patterns. In addition, we plotted a nomogram and validate the accuracy of the risk signature in GEO and TCGA cohorts. We explored the possible biological mechanism using GSEA analysis and preliminarily identified a hub gene using PPI network. Finally, we validated the expression of hub gene in BC cell lines. Results We screened 84 downregulated ARGs, 74 upregulated ARGs, and 32 prognostic ARGs in the human aging genome resource. The aging patterns based on prognostic genes had excellent survival prediction (p < 0.001) and discriminatory ability in 405 BC patients. In addition, we found no significant differences in aging patterns in mutation analysis, which were all characterized by TP53, TTN, and KMT2D mutations. It is worth noting that cluster B in the aging patterns has a better response to immunotherapy and a more active immune microenvironment (p < 0.05). In addition, gene enrichment analysis showed that aging patterns may be related to biological processes such as Staphylococcus aureus infection, phagosome, and cytokine-cytokine receptor interaction. Subsequently, we constructed a risk signature based on 16 differential genes from different aging patterns and had good survival prediction ability in both GEO and TCGA cohort. Specifically, survival analysis revealed a significantly shorter survival time in the high-risk group than in the low-risk group (TCGA and GEO, p < 0.001). In addition, AUC values in the ROC analysis predicted 1, 3, and 5 years in TCGA cohort that are 0.713, 0.714, and 0.738, respectively. AUC values predicted 1, 3, and 5 years in GEO cohort that are 0.606, 0.663, and 0.718, respectively. There is no doubt that risk score was an independent prognostic factor from results of multivariate Cox regression analysis in BC patients (p < 0.001). There were also significant differences in immune cell infiltration, immune checkpoint, and immune score between the two groups (p < 0.05), but it should not be ignored that the correlation with the HLA expression was weak. Finally, we identified and validated CLIC3 as a hub gene that may be involved in the Wnt signaling pathway, etc. Conclusion We provided robust evidences that aging patterns based on ARGs can guide targeted therapy and survival prediction in BC patients.
Collapse
|
42
|
Le AH, Yelland T, Paul NR, Fort L, Nikolaou S, Ismail S, Machesky LM. CYRI-A limits invasive migration through macropinosome formation and integrin uptake regulation. J Cell Biol 2021; 220:e202012114. [PMID: 34165494 PMCID: PMC8236918 DOI: 10.1083/jcb.202012114] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 05/16/2021] [Accepted: 05/29/2021] [Indexed: 12/11/2022] Open
Abstract
The Scar/WAVE complex drives actin nucleation during cell migration. Interestingly, the same complex is important in forming membrane ruffles during macropinocytosis, a process mediating nutrient uptake and membrane receptor trafficking. Mammalian CYRI-B is a recently described negative regulator of the Scar/WAVE complex by RAC1 sequestration, but its other paralogue, CYRI-A, has not been characterized. Here, we implicate CYRI-A as a key regulator of macropinosome formation and integrin internalization. We find that CYRI-A is transiently recruited to nascent macropinosomes, dependent on PI3K and RAC1 activity. CYRI-A recruitment precedes RAB5A recruitment but follows sharply after RAC1 and actin signaling, consistent with it being a local inhibitor of actin polymerization. Depletion of both CYRI-A and -B results in enhanced surface expression of the α5β1 integrin via reduced internalization. CYRI depletion enhanced migration, invasion, and anchorage-independent growth in 3D. Thus, CYRI-A is a dynamic regulator of macropinocytosis, functioning together with CYRI-B to regulate integrin trafficking.
Collapse
Affiliation(s)
- Anh Hoang Le
- Cancer Research UK Beatson Institute, Bearsden, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Bearsden, Glasgow, UK
| | - Tamas Yelland
- Cancer Research UK Beatson Institute, Bearsden, Glasgow, UK
| | - Nikki R. Paul
- Cancer Research UK Beatson Institute, Bearsden, Glasgow, UK
| | - Loic Fort
- Cancer Research UK Beatson Institute, Bearsden, Glasgow, UK
- Department of Cell and Developmental Biology, Medical Research Building III, Vanderbilt University, Nashville, TN
| | - Savvas Nikolaou
- Cancer Research UK Beatson Institute, Bearsden, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Bearsden, Glasgow, UK
| | - Shehab Ismail
- Cancer Research UK Beatson Institute, Bearsden, Glasgow, UK
| | - Laura M. Machesky
- Cancer Research UK Beatson Institute, Bearsden, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Bearsden, Glasgow, UK
| |
Collapse
|
43
|
Ballesteros‐Álvarez J, Andersen JK. mTORC2: The other mTOR in autophagy regulation. Aging Cell 2021; 20:e13431. [PMID: 34250734 PMCID: PMC8373318 DOI: 10.1111/acel.13431] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/15/2021] [Accepted: 06/24/2021] [Indexed: 12/13/2022] Open
Abstract
The mechanistic target of rapamycin (mTOR) has gathered significant attention as a ubiquitously expressed multimeric kinase with key implications for cell growth, proliferation, and survival. This kinase forms the central core of two distinct complexes, mTORC1 and mTORC2, which share the ability of integrating environmental, nutritional, and hormonal cues but which regulate separate molecular pathways that result in different cellular responses. Particularly, mTORC1 has been described as a major negative regulator of endosomal biogenesis and autophagy, a catabolic process that degrades intracellular components and organelles within the lysosomes and is thought to play a key role in human health and disease. In contrast, the role of mTORC2 in the regulation of autophagy has been considerably less studied despite mounting evidence this complex may regulate autophagy in a different and perhaps complementary manner to that of mTORC1. Genetic ablation of unique subunits is currently being utilized to study the differential effects of the two mTOR complexes. RICTOR is the best‐described subunit specific to mTORC2 and as such has become a useful tool for investigating the specific actions of this complex. The development of complex‐specific inhibitors for mTORC2 is also an area of intense interest. Studies to date have demonstrated that mTORC1/2 complexes each signal to a variety of exclusive downstream molecules with distinct biological roles. Pinpointing the particular effects of these downstream effectors is crucial toward the development of novel therapies aimed at accurately modulating autophagy in the context of human aging and disease.
Collapse
|
44
|
Larocque G, Moore DJ, Sittewelle M, Kuey C, Hetmanski JHR, La-Borde PJ, Wilson BJ, Clarke NI, Caswell PT, Royle SJ. Intracellular nanovesicles mediate α5β1 integrin trafficking during cell migration. J Cell Biol 2021; 220:212493. [PMID: 34287617 PMCID: PMC8298100 DOI: 10.1083/jcb.202009028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 05/29/2021] [Accepted: 06/30/2021] [Indexed: 12/30/2022] Open
Abstract
Membrane traffic is an important regulator of cell migration through the endocytosis and recycling of cell surface receptors such as integrin heterodimers. Intracellular nanovesicles (INVs) are transport vesicles that are involved in multiple membrane trafficking steps, including the recycling pathway. The only known marker for INVs is tumor protein D54 (TPD54/TPD52L2), a member of the TPD52-like protein family. Overexpression of TPD52-like family proteins in cancer has been linked to poor prognosis and an aggressive metastatic phenotype, which suggests cell migration may be altered under these conditions. Here, we show that TPD54 directly binds membrane and associates with INVs via a conserved positively charged motif in its C terminus. We describe how other TPD52-like proteins are also associated with INVs, and we document the Rab GTPase complement of all INVs. Depletion of TPD52-like proteins inhibits cell migration and invasion, while their overexpression boosts motility. We show that inhibition of migration is likely due to altered recycling of α5β1 integrins in INVs.
Collapse
Affiliation(s)
- Gabrielle Larocque
- Centre for Mechanochemical Cell Biology, Warwick Medical School, Coventry, UK
| | - Daniel J Moore
- Centre for Mechanochemical Cell Biology, Warwick Medical School, Coventry, UK
| | - Méghane Sittewelle
- Centre for Mechanochemical Cell Biology, Warwick Medical School, Coventry, UK
| | - Cansu Kuey
- Centre for Mechanochemical Cell Biology, Warwick Medical School, Coventry, UK
| | - Joseph H R Hetmanski
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Penelope J La-Borde
- Centre for Mechanochemical Cell Biology, Warwick Medical School, Coventry, UK
| | - Beverley J Wilson
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Nicholas I Clarke
- Centre for Mechanochemical Cell Biology, Warwick Medical School, Coventry, UK
| | - Patrick T Caswell
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Stephen J Royle
- Centre for Mechanochemical Cell Biology, Warwick Medical School, Coventry, UK
| |
Collapse
|
45
|
Wang H, An J, He S, Liao C, Wang J, Tuo B. Chloride intracellular channels as novel biomarkers for digestive system tumors (Review). Mol Med Rep 2021; 24:630. [PMID: 34278487 DOI: 10.3892/mmr.2021.12269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 05/19/2021] [Indexed: 11/06/2022] Open
Abstract
Digestive system malignant tumors are common tumors, and the traditional treatment methods for these tumors include surgical resection, radiotherapy, chemotherapy, and molecularly targeted drugs. However, diagnosis remains challenging, and the early detection of postoperative recurrence is complicated. Therefore, it is necessary to explore novel biomarkers to facilitate clinical diagnosis and treatment. Accumulating evidence supports the crucial role of chloride channels in the development of multiple types of cancers. Given that chloride channels are widely expressed and involved in cell proliferation, apoptosis and cell cycle, among other processes, they may serve as a promising diagnostic and therapeutic target. Chloride intracellular channels (CLICs) are a class of chloride channels that are upregulated or downregulated in certain types of cancer. Furthermore, in certain cases, during cell cycle progression, the localization and function of the cytosolic form of the transmembrane proteins of CLICs are also altered, which may provide a key target for cancer therapy. The aim of the present review was to focus on CLICs as biomarkers for digestive system tumors.
Collapse
Affiliation(s)
- Hui Wang
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Jiaxing An
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Suyu He
- The Fourth Department of the Digestive Disease Center, Suining Central Hospital, Suining, Sichuan 629000, P.R. China
| | - Chengcheng Liao
- Special Key Laboratory of Oral Disease Research, Higher Education Institution in Guizhou Province, School of Stomatology, Zunyi Medical University, Zunyi, Guizhou 563006, P.R. China
| | - Juan Wang
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Biguang Tuo
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| |
Collapse
|
46
|
Zhang C, Xie C, Lu Y. Local Anesthetic Lidocaine and Cancer: Insight Into Tumor Progression and Recurrence. Front Oncol 2021; 11:669746. [PMID: 34249706 PMCID: PMC8264592 DOI: 10.3389/fonc.2021.669746] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/03/2021] [Indexed: 12/11/2022] Open
Abstract
Cancer is a leading contributor to deaths worldwide. Surgery is the primary treatment for resectable cancers. Nonetheless, it also results in inflammatory response, angiogenesis, and stimulated metastasis. Local anesthetic lidocaine can directly and indirectly effect different cancers. The direct mechanisms are inhibiting proliferation and inducing apoptosis via regulating PI3K/AKT/mTOR and caspase-dependent Bax/Bcl2 signaling pathways or repressing cytoskeleton formation. Repression invasion, migration, and angiogenesis through influencing the activation of TNFα-dependent, Src-induced AKT/NO/ICAM and VEGF/PI3K/AKT signaling pathways. Moreover, the indirect influences are immune regulation, anti-inflammation, and postoperative pain relief. This review summarizes the latest evidence that revealed potential clinical benefits of lidocaine in cancer treatment to explore the probable molecular mechanisms and the appropriate dose.
Collapse
Affiliation(s)
- Caihui Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Cuiyu Xie
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yao Lu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Ambulatory Surgery Center, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
47
|
Podocalyxin in Normal Tissue and Epithelial Cancer. Cancers (Basel) 2021; 13:cancers13122863. [PMID: 34201212 PMCID: PMC8227556 DOI: 10.3390/cancers13122863] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 12/13/2022] Open
Abstract
Podocalyxin (PODXL), a glycosylated cell surface sialomucin of the CD34 family, is normally expressed in kidney podocytes, vascular endothelial cells, hematopoietic progenitors, mesothelium, as well as a subset of neurons. In the kidney, PODXL functions primarily as an antiadhesive molecule in podocyte epithelial cells, regulating adhesion and cell morphology, and playing an essential role in the development and function of the organ. Outside the kidney, PODXL plays subtle roles in tissue remodelling and development. Furthermore, many cancers, especially those that originated from the epithelium, have been reported to overexpress PODXL. Collective evidence suggests that PODXL overexpression is linked to poor prognosis, more aggressive tumour progression, unfavourable treatment outcomes, and possibly chemoresistance. This review summarises our current knowledge of PODXL in normal tissue function and epithelial cancer, with a particular focus on its underlying roles in cancer metastasis, likely involvement in chemoresistance, and potential use as a diagnostic and prognostic biomarker.
Collapse
|
48
|
Gibieža P, Petrikaitė V. The dual functions of Rab11 and Rab35 GTPases-regulation of cell division and promotion of tumorigenicity. Am J Cancer Res 2021; 11:1861-1872. [PMID: 34094658 PMCID: PMC8167671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/01/2021] [Indexed: 06/12/2023] Open
Abstract
The broad studies of cancer have led researchers to the creditable understanding of biological and environmental factors that make benign cells to become malignant, as well as the developmental aspects of the tumour cells, known as the "hallmarks of cancer". However, additional research is needed to uncover the features of cancer biology, which would allow to design new and more effective treatment strategies for cancer patients. Since RabGTPases and their effectors are frequently altered in cancer, their role in a regulation of cell division leading to the acquisition of cancer cell-like phenotype has drawn a lot of attention from different research groups in recent years. Both, Rab11 and Rab35 belong to a superfamily of small monomeric GTPases that regulate a diverse array of cellular functions. Lately, Rab11 and Rab35 were declared as oncogenic, and because of their association with abundant cellular functions, a linkage to the induction of cancer, has been proposed. Although the clear connection between the improper regulation of Rab11 or Rab35 and the initiation of tumorigenicity has only beginning to emerge, in this review we will discuss the newest findings regarding the participation of RabGTPases in a control of cell division and promotion of tumorigenesis, trying to link the actual function to the cancer causality.
Collapse
Affiliation(s)
- Paulius Gibieža
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences Kaunas, LT-50162, Lithuania
| | - Vilma Petrikaitė
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences Kaunas, LT-50162, Lithuania
| |
Collapse
|
49
|
Ludwig BS, Kessler H, Kossatz S, Reuning U. RGD-Binding Integrins Revisited: How Recently Discovered Functions and Novel Synthetic Ligands (Re-)Shape an Ever-Evolving Field. Cancers (Basel) 2021; 13:1711. [PMID: 33916607 PMCID: PMC8038522 DOI: 10.3390/cancers13071711] [Citation(s) in RCA: 131] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/22/2021] [Accepted: 03/29/2021] [Indexed: 12/19/2022] Open
Abstract
Integrins have been extensively investigated as therapeutic targets over the last decades, which has been inspired by their multiple functions in cancer progression, metastasis, and angiogenesis as well as a continuously expanding number of other diseases, e.g., sepsis, fibrosis, and viral infections, possibly also Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV-2). Although integrin-targeted (cancer) therapy trials did not meet the high expectations yet, integrins are still valid and promising targets due to their elevated expression and surface accessibility on diseased cells. Thus, for the future successful clinical translation of integrin-targeted compounds, revisited and innovative treatment strategies have to be explored based on accumulated knowledge of integrin biology. For this, refined approaches are demanded aiming at alternative and improved preclinical models, optimized selectivity and pharmacological properties of integrin ligands, as well as more sophisticated treatment protocols considering dose fine-tuning of compounds. Moreover, integrin ligands exert high accuracy in disease monitoring as diagnostic molecular imaging tools, enabling patient selection for individualized integrin-targeted therapy. The present review comprehensively analyzes the state-of-the-art knowledge on the roles of RGD-binding integrin subtypes in cancer and non-cancerous diseases and outlines the latest achievements in the design and development of synthetic ligands and their application in biomedical, translational, and molecular imaging approaches. Indeed, substantial progress has already been made, including advanced ligand designs, numerous elaborated pre-clinical and first-in-human studies, while the discovery of novel applications for integrin ligands remains to be explored.
Collapse
Affiliation(s)
- Beatrice S. Ludwig
- Department of Nuclear Medicine, University Hospital Klinikum Rechts der Isar and Central Institute for Translational Cancer Research (TranslaTUM), Technical University Munich, 81675 Munich, Germany;
| | - Horst Kessler
- Department of Chemistry, Institute for Advanced Study, Technical University Munich, 85748 Garching, Germany;
| | - Susanne Kossatz
- Department of Nuclear Medicine, University Hospital Klinikum Rechts der Isar and Central Institute for Translational Cancer Research (TranslaTUM), Technical University Munich, 81675 Munich, Germany;
- Department of Chemistry, Institute for Advanced Study, Technical University Munich, 85748 Garching, Germany;
| | - Ute Reuning
- Clinical Research Unit, Department of Obstetrics and Gynecology, University Hospital Klinikum Rechts der Isar, Technical University Munich, 81675 Munich, Germany
| |
Collapse
|
50
|
Roper JA, Wilkinson AL, Gower E, Slack RJ. Downregulation of the αv β6 Integrin via RGD Engagement Is Affinity and Time Dependent. J Pharmacol Exp Ther 2021; 376:273-280. [PMID: 33318076 DOI: 10.1124/jpet.120.000379] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 11/30/2020] [Indexed: 12/14/2022] Open
Abstract
The arginyl-glycinyl-aspartic acid (RGD) integrin alpha-v beta-6 (αvβ6) has been identified as playing a key role in the activation of transforming growth factor-β (TGFβ) that is hypothesized to be pivotal in the development of fibrosis and other diseases. In this study, αvβ6 small molecule inhibitors were characterized in a range of in vitro systems to determine affinity, kinetics, and duration of TGFβ inhibition. High αvβ6 binding affinity was shown to be correlated with slow dissociation kinetics. Compound 1 (high αvβ6 affinity, slow dissociation) and SC-68448 (low αvβ6 affinity, fast dissociation) induced concentration- and time-dependent internalization of αvβ6 in normal human bronchial epithelial (NHBE) cells. After washout, the αvβ6 cell surface repopulation was faster for SC-68448 compared with compound 1 In addition, αvβ6-dependent release of active TGFβ from NHBE cells was inhibited by compound 1 and SC-68448. After washout of SC-68448, release of active TGFβ was restored, whereas after washout of compound 1 the inhibition of TGFβ activation was maintained and only reversible in the presence of a lysosomal inhibitor (chloroquine). However, SC-68448 was able to reduce total levels of αvβ6 in NHBE cells if present continuously. These observations suggest αvβ6 can be degraded after high affinity RGD binding that sorts the integrin for lysosomal degradation after internalization, likely due to sustained engagement as a result of slow dissociation kinetics. In addition, the αvβ6 integrin can also be downregulated after sustained engagement of the RGD binding site with low affinity ligands that do not sort the integrin for immediate lysosomal degradation. SIGNIFICANCE STATEMENT: The fate of RGD integrin after ligand binding has not been widely investigated. Using the αvβ6 integrin as a case study, we have demonstrated that RGD-induced downregulation of αvβ6 is both affinity and time dependent. High affinity ligands induced downregulation via lysosomal degradation, likely due to slow dissociation, whereas sustained low affinity ligand engagement was only able to decrease αvβ6 expression over longer periods of time. Our study provides a potential unique mechanism for obtaining duration of action for drugs targeting integrins.
Collapse
Affiliation(s)
- James A Roper
- Fibrosis Discovery Performance Unit (DPU), Respiratory Therapy Area Unit (TAU), GlaxoSmithKline, Stevenage, Hertfordshire, United Kingdom
| | - Alex L Wilkinson
- Fibrosis Discovery Performance Unit (DPU), Respiratory Therapy Area Unit (TAU), GlaxoSmithKline, Stevenage, Hertfordshire, United Kingdom
| | - Elaine Gower
- Fibrosis Discovery Performance Unit (DPU), Respiratory Therapy Area Unit (TAU), GlaxoSmithKline, Stevenage, Hertfordshire, United Kingdom
| | - Robert J Slack
- Fibrosis Discovery Performance Unit (DPU), Respiratory Therapy Area Unit (TAU), GlaxoSmithKline, Stevenage, Hertfordshire, United Kingdom
| |
Collapse
|