1
|
Mannherz HG, Budde H, Jarkas M, Hassoun R, Malek-Chudzik N, Mazur AJ, Skuljec J, Pul R, Napirei M, Hamdani N. Reorganization of the actin cytoskeleton during the formation of neutrophil extracellular traps (NETs). Eur J Cell Biol 2024; 103:151407. [PMID: 38555846 DOI: 10.1016/j.ejcb.2024.151407] [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: 08/10/2023] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 04/02/2024] Open
Abstract
We analyzed actin cytoskeleton alterations during NET extrusion by neutrophil-like dHL-60 cells and human neutrophils in the absence of DNase1 containing serum to avoid chromatin degradation and microfilament disassembly. NET-formation by dHL-60 cells and neutrophils was induced by Ionomycin or phorbol-12-myristat-13-acetate (PMA). Subsequent staining with anti-actin and TRITC-phalloidin showed depolymerization of the cortical F-actin at spatially confined areas, the NET extrusion sites, effected by transient activation of the monooxygenase MICAL-1 supported by the G-actin binding proteins cofilin, profilin, thymosin ß4 and probably the F-actin fragmenting activity of gelsolin and/or its fragments, which also decorated the formed NETs. MICAL-1 itself appeared to be proteolyzed by neutrophil elastase possibly to confine its activity to the NET-extrusion area. The F-actin oxidization activity of MICAL-1 is inhibited by Levosimendan leading to reduced NET-formation. Anti-gasdermin-D immunohistochemistry showed a cytoplasmic distribution in non-stimulated cells. After stimulation the NET-extrusion pore displayed reduced anti-gasdermin-D staining but accumulated underneath the plasma membrane of the remaining cell body. A similar distribution was observed for myosin that concentrated together with cortical F-actin along the periphery of the remaining cell body suggesting force production by acto-myosin interactions supporting NET expulsion as indicated by the inhibitory action of the myosin ATPase inhibitor blebbistatin. Isolated human neutrophils displayed differences in their content of certain cytoskeletal proteins. After stimulation neutrophils with high gelsolin content preferentially formed "cloud"-like NETs, whereas those with low or no gelsolin formed long "filamentous" NETs.
Collapse
Affiliation(s)
- Hans Georg Mannherz
- Department of Anatomy and Molecular Embryology, Medical Faculty, Ruhr-University Bochum, Germany; Department of Cellular and Translational Physiology, Institute of Physiology, Medical Faculty, Ruhr-University Bochum, and Molecular and Experimental Cardiology, Institute for Research and Education, St. Josef Hospital, Clinics of the Ruhr-University Bochum, Germany.
| | - Heidi Budde
- Department of Cellular and Translational Physiology, Institute of Physiology, Medical Faculty, Ruhr-University Bochum, and Molecular and Experimental Cardiology, Institute for Research and Education, St. Josef Hospital, Clinics of the Ruhr-University Bochum, Germany.
| | - Muhammad Jarkas
- Department of Cellular and Translational Physiology, Institute of Physiology, Medical Faculty, Ruhr-University Bochum, and Molecular and Experimental Cardiology, Institute for Research and Education, St. Josef Hospital, Clinics of the Ruhr-University Bochum, Germany.
| | - Roua Hassoun
- Department of Cellular and Translational Physiology, Institute of Physiology, Medical Faculty, Ruhr-University Bochum, and Molecular and Experimental Cardiology, Institute for Research and Education, St. Josef Hospital, Clinics of the Ruhr-University Bochum, Germany.
| | - Natalia Malek-Chudzik
- Department of Chemical Biology and Bioimaging, Faculty of Chemistry, University of Wroclaw, Poland.
| | - Antonina J Mazur
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Poland.
| | - Jelena Skuljec
- Department of Neurology, University Medicine Essen, Germany; Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Medicine Essen, Germany.
| | - Refik Pul
- Department of Neurology, University Medicine Essen, Germany; Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Medicine Essen, Germany.
| | - Markus Napirei
- Department of Anatomy and Molecular Embryology, Medical Faculty, Ruhr-University Bochum, Germany
| | - Nazha Hamdani
- Department of Cellular and Translational Physiology, Institute of Physiology, Medical Faculty, Ruhr-University Bochum, and Molecular and Experimental Cardiology, Institute for Research and Education, St. Josef Hospital, Clinics of the Ruhr-University Bochum, Germany; Department of Physiology, University Maastricht, Maastricht, the Netherlands; HCEMM-SU Cardiovascular Comorbidities Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest 1089, Hungary.
| |
Collapse
|
2
|
He L, Pan X, Wang X, Cao Y, Chen P, Du C, Huang D. Rab6c is a new target of miR‑218 that can promote the progression of bladder cancer. Mol Med Rep 2021; 24:792. [PMID: 34515321 DOI: 10.3892/mmr.2021.12432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 07/08/2021] [Indexed: 11/05/2022] Open
Abstract
Bladder cancer has high morbidity and mortality rates among the male genitourinary system tumor types. MicroRNA‑218 (miR‑218) is associated with the development of a variety of cancer types, including bladder cancer. Rab6c is a member of the Rab family and is involved in drug resistance in MCF7 cells. The aim of the present study was to clarify the relationship between Rab6c and miR‑218 in bladder cancer cell lines. In this study, the expression levels of miR‑218 and Rab6c were evaluated via reverse transcription‑quantitative PCR and western blotting, respectively. The association between Rab6c and miR‑218 was recognized via TargetScan analysis and dual luciferase reporter gene detection. Cell proliferation was analyzed using Cell Counting Kit‑8 and colony formation assays, and the invasive ability was measured via Transwell assays. Rab6c was highly expressed in bladder cancer, while miR‑218 had abnormally low expression in bladder cancer. In addition, there was a mutual regulation between Rab6c and miR‑218 in bladder cancer. It was found that overexpression of Rab6c significantly enhanced the proliferation, colony formation and invasion of T24 and EJ cells. Furthermore, miR‑218 overexpression blocked the promoting effects of Rab6c on the malignant behavior of bladder cancer cells. Thus, Rab6c promotes the proliferation and invasion of bladder cancer cells, while miR‑218 has the opposite effect, which may provide a novel insight for the treatment of bladder cancer.
Collapse
Affiliation(s)
- Long He
- Department of Urology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu 225003, P.R. China
| | - Xiang Pan
- Department of Urology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu 225003, P.R. China
| | - Xialu Wang
- Key Laboratory of Pattern Recognition in Liaoning, School of Medical Devices, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
| | - Yuhua Cao
- Department of The Second Cadre Ward, General Hospital of Northern Theater Command, National Center for Clinical Research of Geriatric Diseases, Shenyang, Liaoning 157099, P.R. China
| | - Peng Chen
- Department of Urology, General Hospital of Northern Theater Command, Shenyang, Liaoning 110013, P.R. China
| | - Cheng Du
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, Liaoning 110013, P.R. China
| | - Daifa Huang
- Department of The Second Cadre Ward, General Hospital of Northern Theater Command, National Center for Clinical Research of Geriatric Diseases, Shenyang, Liaoning 157099, P.R. China
| |
Collapse
|
3
|
Khera L, Vinik Y, Maina F, Lev S. The AXL-PYK2-PKCα axis as a nexus of stemness circuits in TNBC. Life Sci Alliance 2021; 4:4/6/e202000985. [PMID: 33785524 PMCID: PMC8046419 DOI: 10.26508/lsa.202000985] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/18/2021] [Accepted: 03/22/2021] [Indexed: 12/21/2022] Open
Abstract
A clinically relevant AXL-PYK2-PKCα axis where PYK2 and PKCα act as signaling nodes and functionally cooperate to converge stemness promoting pathways and regulate Oct4 and Nanog pluripotent TFs. Cancer stem cells (CSCs) are implicated in tumor initiation, metastasis and drug resistance, and considered as attractive targets for cancer therapy. Here we identified a clinically relevant signaling nexus mediated by AXL receptor, PYK2 and PKCα and show its impact on stemness in TNBC. AXL, PYK2, and PKCα expression correlates with stemness signature in basal-like breast cancer patients, and their depletion in multiple mesenchymal TNBC cell lines markedly reduced the number of mammosphere-forming cells and cells harboring CSCs characteristic markers. Knockdown of PYK2 reduced the levels of AXL, PKCα, FRA1, and PYK2 proteins, and similar trend was obtained upon PKCα depletion. PYK2 depletion decreased AXL transcription through feedback loops mediated by FRA1 and TAZ, whereas PKCα inhibition induced redistribution of AXL to endosomal/lysosomal compartment and enhanced its degradation. PYK2 and PKCα cooperate at a convergence point of multiple stemness-inducing pathways to regulate AXL levels and concomitantly the levels/activation of STAT3, TAZ, FRA1, and SMAD3 as well as the pluripotent transcription factors Nanog and Oct4. Induction of stemness in TNBC sensitized cells to PYK2 and PKCα inhibition suggesting that targeting the AXL-PYK2-PKCα circuit could be an efficient strategy to eliminate CSCs in TNBC.
Collapse
Affiliation(s)
- Lohit Khera
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Yaron Vinik
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Flavio Maina
- Aix Marseille University, Le Centre National de la Recherche Scientifique (CNRS), Developmental Biology Institute of Marseille (IBDM) Unité Mixte de Recherche (UMR) 7288, Marseille, France
| | - Sima Lev
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
4
|
Bansal P, Antil N, Kumar M, Yamaryo-Botté Y, Rawat RS, Pinto S, Datta KK, Katris NJ, Botté CY, Prasad TSK, Sharma P. Protein kinase TgCDPK7 regulates vesicular trafficking and phospholipid synthesis in Toxoplasma gondii. PLoS Pathog 2021; 17:e1009325. [PMID: 33635921 PMCID: PMC7909640 DOI: 10.1371/journal.ppat.1009325] [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: 11/27/2020] [Accepted: 01/20/2021] [Indexed: 02/06/2023] Open
Abstract
Apicomplexan parasites are causative agents of major human diseases. Calcium Dependent Protein Kinases (CDPKs) are crucial components for the intracellular development of apicomplexan parasites and are thus considered attractive drug targets. CDPK7 is an atypical member of this family, which initial characterization suggested to be critical for intracellular development of both Apicomplexa Plasmodium falciparum and Toxoplasma gondii. However, the mechanisms via which it regulates parasite replication have remained unknown. We performed quantitative phosphoproteomics of T. gondii lacking TgCDPK7 to identify its parasitic targets. Our analysis lead to the identification of several putative TgCDPK7 substrates implicated in critical processes like phospholipid (PL) synthesis and vesicular trafficking. Strikingly, phosphorylation of TgRab11a via TgCDPK7 was critical for parasite intracellular development and protein trafficking. Lipidomic analysis combined with biochemical and cellular studies confirmed that TgCDPK7 regulates phosphatidylethanolamine (PE) levels in T. gondii. These studies provide novel insights into the regulation of these processes that are critical for parasite development by TgCDPK7. In this study, we demonstrate that protein kinase TgCDPK7 regulates cellular processes like vesicular trafficking and the synthesis of phospholipids, which are critical for the development of the parasite Toxoplasma gondii. It regulates the localization of a small GTPase TgRab11a by phosphorylating it at a specific site, which is critical for trafficking of important parasite proteins and is important for parasite division. TgCDPK7 may regulate key enzymes involved biogenesis of phosphatidylethanolamine, which may contribute to the synthesis of this important phospholipid. These and other studies shed light on a novel signaling pathway in apicomplexan parasite Toxoplasma gondii.
Collapse
Affiliation(s)
- Priyanka Bansal
- Eukaryotic Gene Expression laboratory, National Institute of Immunology, New Delhi, India
| | - Neelam Antil
- Institute of Bioinformatics, International Tech Park, Bangalore, India
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, India
| | - Manish Kumar
- Eukaryotic Gene Expression laboratory, National Institute of Immunology, New Delhi, India
- Institute of Bioinformatics, International Tech Park, Bangalore, India
| | - Yoshiki Yamaryo-Botté
- ApicoLipid Team, Institute of Advanced Biosciences, CNRS UMR5309, INSERM U1209, Université Grenoble Alpes, Grenoble, France
| | - Rahul Singh Rawat
- Eukaryotic Gene Expression laboratory, National Institute of Immunology, New Delhi, India
| | - Sneha Pinto
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Keshava K. Datta
- Institute of Bioinformatics, International Tech Park, Bangalore, India
| | - Nicholas J. Katris
- ApicoLipid Team, Institute of Advanced Biosciences, CNRS UMR5309, INSERM U1209, Université Grenoble Alpes, Grenoble, France
| | - Cyrille Y. Botté
- ApicoLipid Team, Institute of Advanced Biosciences, CNRS UMR5309, INSERM U1209, Université Grenoble Alpes, Grenoble, France
| | - T. S. Keshava Prasad
- Institute of Bioinformatics, International Tech Park, Bangalore, India
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
- NIMHANS IOB Proteomics and Bioinformatics Laboratory, Neurobiology Research Centre, National Institute of Mental Health and Neuro Sciences, Bangalore, Karnataka, India
| | - Pushkar Sharma
- Eukaryotic Gene Expression laboratory, National Institute of Immunology, New Delhi, India
- * E-mail:
| |
Collapse
|
5
|
Macias A, de la Cruz A, Peraza DA, de Benito-Bueno A, Gonzalez T, Valenzuela C. K V1.5-K Vβ1.3 Recycling Is PKC-Dependent. Int J Mol Sci 2021; 22:ijms22031336. [PMID: 33572906 PMCID: PMC7866247 DOI: 10.3390/ijms22031336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/12/2021] [Accepted: 01/26/2021] [Indexed: 11/26/2022] Open
Abstract
KV1.5 channel function is modified by different regulatory subunits. KVβ1.3 subunits assemble with KV1.5 channels and induce a fast and incomplete inactivation. Inhibition of PKC abolishes the KVβ1.3-induced fast inactivation, decreases the amplitude of the current KV1.5–KVβ1.3 and modifies their pharmacology likely due to changes in the traffic of KV1.5–KVβ1.3 channels in a PKC-dependent manner. In order to analyze this hypothesis, HEK293 cells were transfected with KV1.5–KVβ1.3 channels, and currents were recorded by whole-cell configuration of the patch-clamp technique. The presence of KV1.5 in the membrane was analyzed by biotinylation techniques, live cell imaging and confocal microscopy approaches. PKC inhibition resulted in a decrease of 33 ± 7% of channels in the cell surface due to reduced recycling to the plasma membrane, as was confirmed by confocal microscopy. Live cell imaging indicated that PKC inhibition almost abolished the recycling of the KV1.5–KVβ1.3 channels, generating an accumulation of channels into the cytoplasm. All these results suggest that the trafficking regulation of KV1.5–KVβ1.3 channels is dependent on phosphorylation by PKC and, therefore, they could represent a clinically relevant issue, mainly in those diseases that exhibit modifications in PKC activity.
Collapse
Affiliation(s)
- Alvaro Macias
- Instituto de Investigaciones Biomédicas Madrid CSIC-UAM. C/Arturo Duperier 4, 28029 Madrid, Spain; (A.d.l.C.); (D.A.P.); (A.d.B.-B.); (T.G.)
- CIBER Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Correspondence: (A.M.); (C.V.); Tel.: +34-91-453-1200 (A.M.); +34-91-585-4493 (C.V.)
| | - Alicia de la Cruz
- Instituto de Investigaciones Biomédicas Madrid CSIC-UAM. C/Arturo Duperier 4, 28029 Madrid, Spain; (A.d.l.C.); (D.A.P.); (A.d.B.-B.); (T.G.)
| | - Diego A. Peraza
- Instituto de Investigaciones Biomédicas Madrid CSIC-UAM. C/Arturo Duperier 4, 28029 Madrid, Spain; (A.d.l.C.); (D.A.P.); (A.d.B.-B.); (T.G.)
- CIBER Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Angela de Benito-Bueno
- Instituto de Investigaciones Biomédicas Madrid CSIC-UAM. C/Arturo Duperier 4, 28029 Madrid, Spain; (A.d.l.C.); (D.A.P.); (A.d.B.-B.); (T.G.)
- CIBER Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Teresa Gonzalez
- Instituto de Investigaciones Biomédicas Madrid CSIC-UAM. C/Arturo Duperier 4, 28029 Madrid, Spain; (A.d.l.C.); (D.A.P.); (A.d.B.-B.); (T.G.)
- CIBER Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Department of Biochemistry, School of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), 28029 Madrid, Spain
| | - Carmen Valenzuela
- Instituto de Investigaciones Biomédicas Madrid CSIC-UAM. C/Arturo Duperier 4, 28029 Madrid, Spain; (A.d.l.C.); (D.A.P.); (A.d.B.-B.); (T.G.)
- CIBER Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Correspondence: (A.M.); (C.V.); Tel.: +34-91-453-1200 (A.M.); +34-91-585-4493 (C.V.)
| |
Collapse
|
6
|
Homma Y, Hiragi S, Fukuda M. Rab family of small GTPases: an updated view on their regulation and functions. FEBS J 2021; 288:36-55. [PMID: 32542850 PMCID: PMC7818423 DOI: 10.1111/febs.15453] [Citation(s) in RCA: 270] [Impact Index Per Article: 67.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/27/2020] [Accepted: 06/11/2020] [Indexed: 12/13/2022]
Abstract
The Rab family of small GTPases regulates intracellular membrane trafficking by orchestrating the biogenesis, transport, tethering, and fusion of membrane-bound organelles and vesicles. Like other small GTPases, Rabs cycle between two states, an active (GTP-loaded) state and an inactive (GDP-loaded) state, and their cycling is catalyzed by guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs). Because an active form of each Rab localizes on a specific organelle (or vesicle) and recruits various effector proteins to facilitate each step of membrane trafficking, knowing when and where Rabs are activated and what effectors Rabs recruit is crucial to understand their functions. Since the discovery of Rabs, they have been regarded as one of the central hubs for membrane trafficking, and numerous biochemical and genetic studies have revealed the mechanisms of Rab functions in recent years. The results of these studies have included the identification and characterization of novel GEFs, GAPs, and effectors, as well as post-translational modifications, for example, phosphorylation, of Rabs. Rab functions beyond the simple effector-recruiting model are also emerging. Furthermore, the recently developed CRISPR/Cas technology has enabled acceleration of knockout analyses in both animals and cultured cells and revealed previously unknown physiological roles of many Rabs. In this review article, we provide the most up-to-date and comprehensive lists of GEFs, GAPs, effectors, and knockout phenotypes of mammalian Rabs and discuss recent findings in regard to their regulation and functions.
Collapse
Affiliation(s)
- Yuta Homma
- Laboratory of Membrane Trafficking MechanismsDepartment of Integrative Life SciencesGraduate School of Life SciencesTohoku UniversitySendaiJapan
| | - Shu Hiragi
- Laboratory of Membrane Trafficking MechanismsDepartment of Integrative Life SciencesGraduate School of Life SciencesTohoku UniversitySendaiJapan
| | - Mitsunori Fukuda
- Laboratory of Membrane Trafficking MechanismsDepartment of Integrative Life SciencesGraduate School of Life SciencesTohoku UniversitySendaiJapan
| |
Collapse
|
7
|
Waschbüsch D, Khan AR. Phosphorylation of Rab GTPases in the regulation of membrane trafficking. Traffic 2020; 21:712-719. [PMID: 32969543 PMCID: PMC7756361 DOI: 10.1111/tra.12765] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/17/2020] [Accepted: 09/17/2020] [Indexed: 12/15/2022]
Abstract
Rab GTPases are master regulators of membrane trafficking in eukaryotic cells. Phosphorylation of Rab GTPases was characterized in the 1990s and there have been intermittent reports of its relevance to Rab functions. Phosphorylation as a regulatory mechanism has gained prominence through the identification of Rabs as physiological substrates of leucine‐rich repeat kinase 2 (LRRK2). LRRK2 is a Ser/Thr kinase that is associated with inherited and sporadic forms of Parkinson disease. In recent years, numerous kinases and their associated signaling pathways have been identified that lead to phosphorylation of Rabs. These emerging studies suggest that serine/threonine and tyrosine phosphorylation of Rabs may be a widespread and under‐appreciated mechanism for controlling their membrane trafficking functions. Here we survey current knowledge of Rab phosphorylation and discuss models for how this post‐translational mechanism exerts control of membrane trafficking.
Collapse
Affiliation(s)
- Dieter Waschbüsch
- School of Biochemistry and Immunology, Trinity College, Dublin, Ireland
| | - Amir R Khan
- School of Biochemistry and Immunology, Trinity College, Dublin, Ireland.,Division of Newborn Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
8
|
Stephens DC, Powell TW, Taraska JW, Harris DA. Imaging the rapid yet transient accumulation of regulatory lipids, lipid kinases, and protein kinases during membrane fusion, at sites of exocytosis of MMP-9 in MCF-7 cells. Lipids Health Dis 2020; 19:195. [PMID: 32829709 PMCID: PMC7444259 DOI: 10.1186/s12944-020-01374-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/17/2020] [Indexed: 11/10/2022] Open
Abstract
Background The regulation of exocytosis is physiologically vital in cells and requires a variety of distinct proteins and lipids that facilitate efficient, fast, and timely release of secretory vesicle cargo. Growing evidence suggests that regulatory lipids act as important lipid signals and regulate various biological processes including exocytosis. Though functional roles of many of these regulatory lipids has been linked to exocytosis, the dynamic behavior of these lipids during membrane fusion at sites of exocytosis in cell culture remains unknown. Methods Total internal reflection fluorescence microscopy (TIRF) was used to observe the spatial organization and temporal dynamics (i.e. spatial positioning and timing patterns) of several lipids, and accessory proteins, like lipid kinases and protein kinases, in the form of protein kinase C (PRKC) associated with sites of exocytosis of matrix metalloproteinase-9 (MMP-9) in living MCF-7 cancer cells. Results Following stimulation with phorbol myristate acetate (PMA) to promote exocytosis, a transient accumulation of several distinct regulatory lipids, lipid kinases, and protein kinases at exocytic sites was observed. This transient accumulation centered at the time of membrane fusion is followed by a rapid diffusion away from the fusion sites. Additionally, the synthesis of these regulatory lipids, degradation of these lipids, and the downstream effectors activated by these lipids, are also achieved by the recruitment and accumulation of key enzymes at exocytic sites (during the moment of cargo release). This includes key enzymes like lipid kinases, protein kinases, and phospholipases that facilitate membrane fusion and exocytosis of MMP-9. Conclusions This work suggests that these regulatory lipids and associated effector proteins are locally synthesized and/or recruited to sites of exocytosis, during membrane fusion and cargo release. More importantly, their enrichment at fusion sites serves as an important spatial and temporal organizing “element” defining individual exocytic sites.
Collapse
Affiliation(s)
- Dominique C Stephens
- Department of Chemistry, Howard University, 525 College Street NW, Washington, D.C, 20059, USA
| | - Tyrel W Powell
- Department of Chemistry, Howard University, 525 College Street NW, Washington, D.C, 20059, USA
| | - Justin W Taraska
- Laboratory of Molecular Biophysics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Dinari A Harris
- Department of Chemistry, Howard University, 525 College Street NW, Washington, D.C, 20059, USA.
| |
Collapse
|
9
|
Nazarenko I. Extracellular Vesicles: Recent Developments in Technology and Perspectives for Cancer Liquid Biopsy. Recent Results Cancer Res 2020; 215:319-344. [PMID: 31605237 DOI: 10.1007/978-3-030-26439-0_17] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Extracellular micro- and nanoscale membrane vesicles produced by different cells progressively attract the attention of the scientific community. They function as mediators of intercellular communication and transport genetic material and signaling molecules between the cells. In the context of keeping homeostasis, the extracellular vesicles contribute to the regulation of various systemic and local processes. Vesicles released by the tumor and activated stromal cells exhibit multiple functions including support of tumor growth, preparation of the pre-metastatic niches, and immune suppression. Considerable progress has been made regarding the criteria of classification of the vesicles according to their origin, content, and function: Exosomes, microvesicles, also referred to as microparticles or ectosomes, and large oncosomes were defined as actively released vesicles. Additionally, apoptotic bodies represented by a highly heterogeneous population of particles produced during apoptosis, the programmed cell death, should be considered. Because the majority of isolation techniques do not allow the separation of different types of vesicles, a joined term "extracellular vesicles" (EVs) was recommended by the ISEV community for the definition of vesicles isolated from either the cell culture supernatants or the body fluids. Because EV content reflects the content of the cell of origin, multiple studies on EVs from body fluids in the context of cancer diagnosis, prediction, and prognosis were performed, actively supporting their high potential as a biomarker source. Here, we review the leading achievements in EV analysis from body fluids, defined as EV-based liquid biopsy, and provide an overview of the main EV constituents: EV surface proteins, intravesicular soluble proteins, EV RNA including mRNA and miRNA, and EV DNA as potential biomarkers. Furthermore, we discuss recent developments in technology for quantitative EV analysis in the clinical setting and future perspectives toward miniaturized high-precision liquid biopsy approaches.
Collapse
Affiliation(s)
- Irina Nazarenko
- Institute for Infection Prevention and Hospital Epidemiology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany. .,German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
10
|
Roma MG, Barosso IR, Miszczuk GS, Crocenzi FA, Pozzi EJS. Dynamic Localization of Hepatocellular Transporters: Role in Biliary Excretion and Impairment in Cholestasis. Curr Med Chem 2019; 26:1113-1154. [DOI: 10.2174/0929867325666171205153204] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 09/06/2017] [Accepted: 09/07/2017] [Indexed: 12/25/2022]
Abstract
Bile flow generation is driven by the vectorial transfer of osmotically active compounds from sinusoidal blood into a confined space, the bile canaliculus. Hence, localization of hepatocellular transporters relevant to bile formation is crucial for bile secretion. Hepatocellular transporters are localized either in the plasma membrane or in recycling endosomes, from where they can be relocated to the plasma membrane on demand, or endocytosed when the demand decreases. The balance between endocytic internalization/ exocytic targeting to/from this recycling compartment is therefore the main determinant of the hepatic capability to generate bile, and to dispose endo- and xenobiotics. Furthermore, the exacerbated endocytic internalization is a common pathomechanisms in both experimental and human cholestasis; this results in bile secretory failure and, eventually, posttranslational transporter downregulation by increased degradation. This review summarizes the proposed structural mechanisms accounting for this pathological condition (e.g., alteration of function, localization or expression of F-actin or F-actin/transporter cross-linking proteins, and switch to membrane microdomains where they can be readily endocytosed), and the mediators implicated (e.g., triggering of “cholestatic” signaling transduction pathways). Lastly, we discussed the efficacy to counteract the cholestatic failure induced by transporter internalization of a number of therapeutic experimental approaches based upon the use of compounds that trigger exocytic targetting of canalicular transporters (e.g., cAMP, tauroursodeoxycholate). This therapeutics may complement treatments aimed to transcriptionally improve transporter expression, by affording proper localization and membrane stability to the de novo synthesized transporters.
Collapse
Affiliation(s)
- Marcelo G. Roma
- Instituto de Fisiologia Experimental (IFISE) - Facultad de Ciencias Bioquimicas y Farmaceuticas (CONICET - U.N.R.), S2002LRL, Rosario, Argentina
| | - Ismael R. Barosso
- Instituto de Fisiologia Experimental (IFISE) - Facultad de Ciencias Bioquimicas y Farmaceuticas (CONICET - U.N.R.), S2002LRL, Rosario, Argentina
| | - Gisel S. Miszczuk
- Instituto de Fisiologia Experimental (IFISE) - Facultad de Ciencias Bioquimicas y Farmaceuticas (CONICET - U.N.R.), S2002LRL, Rosario, Argentina
| | - Fernando A. Crocenzi
- Instituto de Fisiologia Experimental (IFISE) - Facultad de Ciencias Bioquimicas y Farmaceuticas (CONICET - U.N.R.), S2002LRL, Rosario, Argentina
| | - Enrique J. Sánchez Pozzi
- Instituto de Fisiologia Experimental (IFISE) - Facultad de Ciencias Bioquimicas y Farmaceuticas (CONICET - U.N.R.), S2002LRL, Rosario, Argentina
| |
Collapse
|
11
|
Tzeng HT, Li TH, Tang YA, Tsai CH, Frank Lu PJ, Lai WW, Chiang CW, Wang YC. Phosphorylation of Rab37 by protein kinase C alpha inhibits the exocytosis function and metastasis suppression activity of Rab37. Oncotarget 2017; 8:108556-108570. [PMID: 29312551 PMCID: PMC5752464 DOI: 10.18632/oncotarget.20998] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 09/05/2017] [Indexed: 01/31/2023] Open
Abstract
We previously identified a novel Rab small GTPase protein, Rab37, which plays a critical role in regulating exocytosis of secreted glycoproteins, tissue inhibitor of metalloproteinases 1 (TIMP1) to suppress lung cancer metastasis. Patients with preserved Rab37 protein expression were associated with better prognosis. However, a significant number of the patients with preserved Rab37 expression showed poor survival. In addition, the molecular mechanism for the regulation of Rab37-mediated exocytosis remained to be further identified. Therefore, we investigated the molecular mechanism underlying the dysregulation of Rab37-mediated exocytosis and metastasis suppression. Here, we report a novel mechanism for Rab37 inactivation by phosphorylation. Lung cancer patients with preserved Rab37, low TIMP1, and high PKCα expression profile correlate with worse progression-free survival examined by Kaplan-Meier survival, suggesting that PKCα overexpression leads to dysfunction of Rab37. This PKCα-Rab37-TIMP1 expression profile predicts the poor outcome by multivariate Cox regression analysis. We also show that Rab37 is phosphorylated by protein kinase Cα (PKCα) at threonine 172 (T172), leading to attenuation of its GTP-bound state, and impairment of the Rab37-mediated exocytosis of TIMP1, and thus reduces its suppression activity on lung cancer cell motility. We further demonstrate that PKCα reduces vesicle colocalization of Rab37 and TIMP1, and therefore inhibits Rab37-mediated TIMP1 trafficking. Moreover, Phospho-mimetic aspartate substitution mutant T172D of Rab37 significantly promotes tumor metastasis in vivo. Our findings reveal a novel regulation of Rab37 activity by PKCα-mediated phosphorylation which inhibits exocytic transport of TIMP1 and thereby enhances lung tumor metastasis.
Collapse
Affiliation(s)
- Hong-Tai Tzeng
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tsung-Hsin Li
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yen-An Tang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Agency for Science, Technology and Research (ASTAR), Singapore
| | - Chung-Han Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Jung Frank Lu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wu-Wei Lai
- Division of Thoracic Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chi-Wu Chiang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Ching Wang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
12
|
Zhai X, Leo MD, Jaggar JH. Endothelin-1 Stimulates Vasoconstriction Through Rab11A Serine 177 Phosphorylation. Circ Res 2017; 121:650-661. [PMID: 28696251 DOI: 10.1161/circresaha.117.311102] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/06/2017] [Accepted: 07/07/2017] [Indexed: 01/18/2023]
Abstract
RATIONALE Large-conductance calcium-activated potassium channels (BK) are composed of pore-forming BKα and auxiliary β1 subunits in arterial smooth muscle cells (myocytes). Vasoconstrictors, including endothelin-1 (ET-1), inhibit myocyte BK channels, leading to contraction, but mechanisms involved are unclear. Recent evidence indicates that BKα is primarily plasma membrane localized, whereas the cellular location of β1 can be rapidly altered by Rab11A-positive recycling endosomes. Whether vasoconstrictors regulate the multisubunit composition of surface BK channels to stimulate contraction is unclear. OBJECTIVE Test the hypothesis that ET-1 inhibits BK channels by altering BKα and β1 surface trafficking in myocytes, identify mechanisms involved, and determine functional significance in myocytes of small cerebral arteries. METHODS AND RESULTS ET-1, through activation of PKC (protein kinase C), reduced surface β1 abundance and the proximity of β1 to surface BKα in myocytes. In contrast, ET-1 did not alter surface BKα, total β1, or total BKα proteins. ET-1 stimulated Rab11A phosphorylation, which reduced Rab11A activity. Rab11A serine 177 was identified as a high-probability PKC phosphorylation site. Expression of a phosphorylation-incapable Rab11A construct (Rab11A S177A) blocked the ET-1-induced Rab11A phosphorylation, reduction in Rab11A activity, and decrease in surface β1 protein. ET-1 inhibited single BK channels and transient BK currents in myocytes and stimulated vasoconstriction via a PKC-dependent mechanism that required Rab11A S177. In contrast, NO-induced Rab11A activation, surface trafficking of β1 subunits, BK channel and transient BK current activation, and vasodilation did not involve Rab11A S177. CONCLUSIONS ET-1 stimulates PKC-mediated phosphorylation of Rab11A at serine 177, which inhibits Rab11A and Rab11A-dependent surface trafficking of β1 subunits. The decrease in surface β1 subunits leads to a reduction in BK channel calcium-sensitivity, inhibition of transient BK currents, and vasoconstriction. We describe a unique mechanism by which a vasoconstrictor inhibits BK channels and identify Rab11A serine 177 as a modulator of arterial contractility.
Collapse
Affiliation(s)
- Xue Zhai
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis
| | - M Dennis Leo
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis
| | - Jonathan H Jaggar
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis.
| |
Collapse
|
13
|
Alfonzo-Méndez MA, Hernández-Espinosa DA, Carmona-Rosas G, Romero-Ávila MT, Reyes-Cruz G, García-Sáinz JA. Protein Kinase C Activation Promotes α 1B-Adrenoceptor Internalization and Late Endosome Trafficking through Rab9 Interaction. Role in Heterologous Desensitization. Mol Pharmacol 2017; 91:296-306. [PMID: 28082304 DOI: 10.1124/mol.116.106583] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 01/09/2017] [Indexed: 12/25/2022] Open
Abstract
Upon agonist stimulation, α1B-adrenergic receptors couple to Gq proteins, calcium signaling and protein kinase C activation; subsequently, the receptors are phosphorylated, desensitized, and internalized. Internalization seems to involve scaffolding proteins, such as β-arrestin and clathrin. However, the fine mechanisms that participate remain unsolved. The roles of protein kinase C and the small GTPase, Rab9, in α1B-AR vesicular traffic were investigated by studying α1B-adrenergic receptor-Rab protein interactions, using Förster resonance energy transfer (FRET), confocal microscopy, and intracellular calcium quantitation. In human embryonic kidney 293 cells overexpressing Discosoma spp. red fluorescent protein (DsRed)-tagged α1B-ARs and enhanced green fluorescent protein--tagged Rab proteins, pharmacological protein kinase C activation mimicked α1B-AR traffic elicited by nonrelated agents, such as sphingosine 1-phosphate (i.e., transient α1B-AR-Rab5 FRET signal followed by a sustained α1B-AR-Rab9 interaction), suggesting brief receptor localization in early endosomes and transfer to late endosomes. This latter interaction was abrogated by blocking protein kinase C activity, resulting in receptor retention at the plasma membrane. Similar effects were observed when a dominant-negative Rab9 mutant (Rab9-GDP) was employed. When α1B-adrenergic receptors that had been mutated at protein kinase C phosphorylation sites (S396A, S402A) were used, phorbol ester-induced desensitization of the calcium response was markedly decreased; however, interaction with Rab9 was only partially decreased and internalization was observed in response to phorbol esters and sphingosine 1-phosphate. Finally, Rab9-GDP expression did not affect adrenergic-mediated calcium response but abolished receptor traffic and altered desensitization. Data suggest that protein kinase C modulates α1B-adrenergic receptor transfer to late endosomes and that Rab9 regulates this process and participates in G protein-mediated signaling turn-off.
Collapse
Affiliation(s)
- Marco A Alfonzo-Méndez
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México (M.A.A.-M., D.A.H.-E., G.C.-R., M.T.R.-A., J.A.G.-S.) and Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional-CINVESTAV, Col. San Pedro Zacatenco, Ciudad de México (G.R.-C.)
| | - David A Hernández-Espinosa
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México (M.A.A.-M., D.A.H.-E., G.C.-R., M.T.R.-A., J.A.G.-S.) and Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional-CINVESTAV, Col. San Pedro Zacatenco, Ciudad de México (G.R.-C.)
| | - Gabriel Carmona-Rosas
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México (M.A.A.-M., D.A.H.-E., G.C.-R., M.T.R.-A., J.A.G.-S.) and Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional-CINVESTAV, Col. San Pedro Zacatenco, Ciudad de México (G.R.-C.)
| | - M Teresa Romero-Ávila
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México (M.A.A.-M., D.A.H.-E., G.C.-R., M.T.R.-A., J.A.G.-S.) and Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional-CINVESTAV, Col. San Pedro Zacatenco, Ciudad de México (G.R.-C.)
| | - Guadalupe Reyes-Cruz
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México (M.A.A.-M., D.A.H.-E., G.C.-R., M.T.R.-A., J.A.G.-S.) and Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional-CINVESTAV, Col. San Pedro Zacatenco, Ciudad de México (G.R.-C.)
| | - J Adolfo García-Sáinz
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México (M.A.A.-M., D.A.H.-E., G.C.-R., M.T.R.-A., J.A.G.-S.) and Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional-CINVESTAV, Col. San Pedro Zacatenco, Ciudad de México (G.R.-C.)
| |
Collapse
|
14
|
Abstract
A large group of small Rab GTPases which mediate secretory and endosomal membrane transport, as well as autophagosome biogenesis, are essential components of vesicle trafficking machinery. Specific Rab protein together with the cognate effectors coordinates the dynamics of trafficking pathway and determines the cargo proteins destination. Functional impairments of Rab proteins by mutations or post-translational modifications disrupting the regulatory network of vesicle trafficking have been implicated in tumorigenesis. Therefore, the vesicle transport regulators play essential roles in the mediation of cancer cell biology, including uncontrolled cell growth, invasion and metastasis. The context-dependent role of the same Rab to act as either an oncoprotein or tumor suppressor in different cancers is found. Such discrepancies may be due in part to the interaction of specific Rab protein with different effectors or cargos in various tumors. Here, we review recent advances in the roles of Rab GTPases in communicating with other effectors in tumor progression. In this review, we also emphasize dysregulation of Rab-mediated membrane delivery shifting normal cell behaviors toward malignancy. Thus, recovery of the dysregulated vesicle trafficking systems in cancer cells may provide future directions for potential strategy to restrain tumor progression.
Collapse
Affiliation(s)
- Hong-Tai Tzeng
- Department of Pharmacology, National Cheng Kung University, College of Medicine, No.1, University Road, Tainan, 70101, Taiwan, People's Republic of China
| | - Yi-Ching Wang
- Department of Pharmacology, National Cheng Kung University, College of Medicine, No.1, University Road, Tainan, 70101, Taiwan, People's Republic of China. .,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, No.1, University Road, Tainan, 70101, Taiwan, People's Republic of China.
| |
Collapse
|
15
|
Mauriat M, Leplé JC, Claverol S, Bartholomé J, Negroni L, Richet N, Lalanne C, Bonneu M, Coutand C, Plomion C. Quantitative Proteomic and Phosphoproteomic Approaches for Deciphering the Signaling Pathway for Tension Wood Formation in Poplar. J Proteome Res 2015; 14:3188-203. [PMID: 26112267 DOI: 10.1021/acs.jproteome.5b00140] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Trees adjust their growth following forced changes in orientation to re-establish a vertical position. In angiosperms, this adjustment involves the differential regulation of vascular cambial activity between the lower (opposite wood) and upper (tension wood) sides of the leaning stem. We investigated the molecular mechanisms leading to the formation of differential wood types through a quantitative proteomic and phosphoproteomic analysis on poplar subjected to a gravitropic stimulus. We identified and quantified 675 phosphopeptides, corresponding to 468 phosphoproteins, and 3 763 nonphosphorylated peptides, corresponding to 1 155 proteins, in the differentiating xylem of straight-growing trees (control) and trees subjected to a gravitational stimulus during 8 weeks. About 1% of the peptides were specific to a wood type (straight, opposite, or tension wood). Proteins quantified in more than one type of wood were more numerous: a mixed linear model showed 389 phosphopeptides and 556 proteins to differ in abundance between tension wood and opposite wood. Twenty-one percent of the phosphoproteins identified here were described in their phosphorylated form for the first time. Our analyses revealed remarkable developmental molecular plasticity, with wood type-specific phosphorylation events, and highlighted the involvement of different proteins in the biosynthesis of cell wall components during the formation of the three types of wood.
Collapse
Affiliation(s)
- Mélanie Mauriat
- †INRA, UMR 1202 BIOGECO, F-33610 Cestas, France.,‡Univ. Bordeaux, BIOGECO, UMR1202, F-33615 Pessac, France
| | - Jean-Charles Leplé
- §INRA, UR0588 AGPF, 2163 Avenue de la Pomme de Pin, CS 40001 Ardon, F-45075 Orléans Cedex 2, France
| | - Stéphane Claverol
- ⊥Plateforme Protéome, CGFB, Université Bordeaux Segalen, F-33076 Bordeaux, France
| | - Jérôme Bartholomé
- †INRA, UMR 1202 BIOGECO, F-33610 Cestas, France.,‡Univ. Bordeaux, BIOGECO, UMR1202, F-33615 Pessac, France
| | - Luc Negroni
- ⊥Plateforme Protéome, CGFB, Université Bordeaux Segalen, F-33076 Bordeaux, France
| | - Nicolas Richet
- §INRA, UR0588 AGPF, 2163 Avenue de la Pomme de Pin, CS 40001 Ardon, F-45075 Orléans Cedex 2, France
| | - Céline Lalanne
- †INRA, UMR 1202 BIOGECO, F-33610 Cestas, France.,‡Univ. Bordeaux, BIOGECO, UMR1202, F-33615 Pessac, France
| | - Marc Bonneu
- ⊥Plateforme Protéome, CGFB, Université Bordeaux Segalen, F-33076 Bordeaux, France
| | - Catherine Coutand
- ¶INRA, UMR 547 PIAF, 234 Avenue du Brézet, F-63100 Clermont-Ferrand, France.,∥Clermont Université, Université Blaise Pascal, UMR 547 PIAF, F-63100 Clermont-Ferrand, France
| | - Christophe Plomion
- †INRA, UMR 1202 BIOGECO, F-33610 Cestas, France.,‡Univ. Bordeaux, BIOGECO, UMR1202, F-33615 Pessac, France
| |
Collapse
|
16
|
Miller IV, Grunewald TGP. Tumour-derived exosomes: Tiny envelopes for big stories. Biol Cell 2015; 107:287-305. [PMID: 25923825 DOI: 10.1111/boc.201400095] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 04/24/2015] [Indexed: 12/14/2022]
Abstract
The discovery of exosomes, which are small, 30-100 nm sized extracellular vesicles that are released by virtual all cells, has initiated a rapidly expanding and vibrant research field. Current investigations are mainly directed toward the role of exosomes in intercellular communication and their potential value as biomarkers for a broad set of diseases. By horizontal transfer of molecular information such as micro RNAs, messenger RNAs or proteins, as well as by receptor-cell interactions, exosomes are capable to mediate the reprogramming of surrounding cells. Herein, we review how especially cancer cells take advantage of this mechanism to influence their microenvironment in favour of immune escape, therapy resistance, tumour growth and metastasis. Moreover, we provide a comprehensive microarray analysis (n > 1970) to study the expression patterns of genes known to be intimately involved in exosome biogenesis across 26 different cancer entities and a normal tissue atlas. Consistent with the elevated production of exosomes observed in cancer patient plasma, we found a significant overexpression especially of RAB27A, CHMP4C and SYTL4 in the corresponding cancer entities as compared to matched normal tissues. Finally, we discuss the immune-modulatory and anti-tumorigenic functions of exosomes as well as innovative approaches to specifically target the exosomal circuits in experimental cancer therapy.
Collapse
Affiliation(s)
- Isabella V Miller
- Department of Medicine II, Würzburg University Medical Centre, Würzburg, 97080, Germany
| | - Thomas G P Grunewald
- Laboratory for Paediatric Sarcoma Biology, Institute for Pathology of the LMU Munich, Munich, 80337, Germany
| |
Collapse
|
17
|
Park SW, Schonhoff CM, Webster CRL, Anwer MS. Rab11, but not Rab4, facilitates cyclic AMP- and tauroursodeoxycholate-induced MRP2 translocation to the plasma membrane. Am J Physiol Gastrointest Liver Physiol 2014; 307:G863-70. [PMID: 25190474 PMCID: PMC4200318 DOI: 10.1152/ajpgi.00457.2013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Rab proteins (Ras homologous for brain) play an important role in vesicle trafficking. Rab4 and Rab11 are involved in vesicular trafficking to the plasma membrane from early endosomes and recycling endosomes, respectively. Tauroursodeoxycholate (TUDC) and cAMP increase bile formation, in part, by increasing plasma membrane localization of multidrug resistance-associated protein 2 (MRP2). The goal of the present study was to determine the role of these Rab proteins in the trafficking of MRP2 by testing the hypothesis that Rab11 and/or Rab4 facilitate cAMP- and TUDC-induced MRP2 translocation to the plasma membrane. Studies were conducted in HuH-NTCP cells (HuH7 cells stably transfected with human NTCP), which constitutively express MRP2. HuH-NTCP cells were transfected with Rab11-WT and GDP-locked dominant inactive Rab11-GDP or with Rab4-GDP to study the role of Rab11 and Rab4. A biotinylation method and a GTP overlay assay were used to determine plasma membrane MRP2 and activation of Rab proteins (Rab11 and Rab4), respectively. Cyclic AMP and TUDC increased plasma membrane MRP2 and stimulated Rab11 activity. Plasma membrane translocation of MRP2 by cAMP and TUDC was increased and inhibited in cells transfected with Rab11-WT and Rab11-GDP, respectively. Cyclic AMP (previous study) and TUDC increased Rab4 activity. However, cAMP- and TUDC-induced increases in MRP2 were not inhibited by Rab4-GDP. Taken together, these results suggest that Rab11 is involved in cAMP- and TUDC-induced MRP2 translocation to the plasma membrane.
Collapse
Affiliation(s)
- Se Won Park
- Department of Biomedical Sciences, Cummings School of Veterinary Medicine at Tufts University, North Grafton, Massachusetts; and
| | - Christopher M Schonhoff
- Department of Biomedical Sciences, Cummings School of Veterinary Medicine at Tufts University, North Grafton, Massachusetts; and
| | - Cynthia R L Webster
- Department of Clinical Sciences, Cummings School of Veterinary Medicine at Tufts University, North Grafton, Massachusetts
| | - M Sawkat Anwer
- Department of Biomedical Sciences, Cummings School of Veterinary Medicine at Tufts University, North Grafton, Massachusetts; and
| |
Collapse
|
18
|
Anwer MS. Role of protein kinase C isoforms in bile formation and cholestasis. Hepatology 2014; 60:1090-7. [PMID: 24700589 PMCID: PMC4141907 DOI: 10.1002/hep.27088] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 02/13/2014] [Indexed: 12/11/2022]
Abstract
Transhepatic solute transport provides the osmotic driving force for canalicular bile formation. Choleretic and cholestatic agents affect bile formation, in part, by altering plasma membrane localizations of transporters involved in bile formation. These short-term dynamic changes in transporter location are highly regulated posttranslational events requiring various cellular signaling pathways. Interestingly, both choleretic and cholestatic agents activate the same intracellular signaling kinases, such as phosphoinositide-3-kinase (PI3K), protein kinase C (PKC), and mitogen-activated protein kinase (MAPK). An emerging theme is that choleretic and cholestatic effects may be mediated by different isoforms of these kinases. This is most evident for PKC-mediated regulation of plasma membrane localization of Na+-taurocholate cotransporting polypeptide (NTCP) and multidrug resistance-associated protein 2 (MRP2) by conventional PKCα (cPKCα), novel PKCδ (nPKCδ), nPKCε, and atypical PKCζ (aPKCζ). aPKCζ may mediate choleretic effects by inserting NTCP into the plasma membrane, and nPKCε may mediate cholestatic effects by retrieving MRP2 from the plasma membrane. On the other hand, cPKCα and nPKCδ may be involved in choleretic, cholestatic, and anticholestatic effects by inserting, retrieving, and inhibiting retrieval of transporters, respectively. The effects of PKC isoforms may be mediated by phosphorylation of the transporters, actin binding proteins (radixin and myristoylated alanine-rich C kinase substrate), and Rab proteins. Human NTCP plays an important role in the entry of hepatitis B and D viruses into hepatocytes and consequent infection. Thus, PKCs, by regulating NTCP trafficking, may also play an important role in hepatic viral infections.
Collapse
Affiliation(s)
- M Sawkat Anwer
- Department of Biomedical Sciences, Cummings School of Veterinary Medicine at Tufts University, North Grafton, MA
| |
Collapse
|
19
|
Rapetti-Mauss R, O'Mahony F, Sepulveda FV, Urbach V, Harvey BJ. Oestrogen promotes KCNQ1 potassium channel endocytosis and postendocytic trafficking in colonic epithelium. J Physiol 2013; 591:2813-31. [PMID: 23529131 PMCID: PMC3690688 DOI: 10.1113/jphysiol.2013.251678] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 03/19/2013] [Indexed: 12/23/2022] Open
Abstract
The cAMP-regulated potassium channel KCNQ1:KCNE3 plays an essential role in transepithelial Cl(-) secretion. Recycling of K(+) across the basolateral membrane provides the driving force necessary to maintain apical Cl(-) secretion. The steroid hormone oestrogen (17β-oestradiol; E2), produces a female-specific antisecretory response in rat distal colon through the inhibition of the KCNQ1:KCNE3 channel. It has previously been shown that rapid inhibition of the channel conductance results from E2-induced uncoupling of the KCNE3 regulatory subunit from the KCNQ1 channel pore complex. The purpose of this study was to determine the mechanism required for sustained inhibition of the channel function. We found that E2 plays a role in regulation of KCNQ1 cell membrane abundance by endocytosis. Ussing chamber experiments have shown that E2 inhibits both Cl(-) secretion and KCNQ1 current in a colonic cell line, HT29cl.19A, when cultured as a confluent epithelium. Following E2 treatment, KCNQ1 was retrieved from the plasma membrane by a clathrin-mediated endocytosis, which involved the association between KCNQ1 and the clathrin adaptor, AP-2. Following endocytosis, KCNQ1 was accumulated in early endosomes. Following E2-induced endocytosis, rather than being degraded, KCNQ1 was recycled by a biphasic mechanism involving Rab4 and Rab11. Protein kinase Cδ and AMP-dependent kinase were rapidly phosphorylated in response to E2 on their activating phosphorylation sites, Ser643 and Thr172, respectively (as previously shown). Both kinases are necessary for the E2-induced endocytosis, because E2 failed to induce KCNQ1 internalization following pretreatment with specific inhibitors of both protein kinase Cδ and AMP-dependent kinase. The ubiquitin ligase Nedd4.2 binds KCNQ1 in response to E2 to induce channel internalization. This study has provided the first demonstration of hormonal regulation of KCNQ1 trafficking. In conclusion, we propose that internalization of KCNQ1 is a key event in the sustained antisecretory response to oestrogen.
Collapse
Affiliation(s)
- Raphael Rapetti-Mauss
- Department of Molecular Medicine, RCSI-ERC, Beaumont Hospital, PO Box 9063, Dublin 9, Ireland
| | | | | | | | | |
Collapse
|