1
|
Adnani L, Meehan B, Kim M, Choi D, Rudd CE, Riazalhosseini Y, Rak J. Immune cell infiltration into brain tumor microenvironment is mediated by Rab27-regulated vascular wall integrity. SCIENCE ADVANCES 2025; 11:eadr6940. [PMID: 40408475 PMCID: PMC12101492 DOI: 10.1126/sciadv.adr6940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 04/22/2025] [Indexed: 05/25/2025]
Abstract
Aggressive brain tumors often exhibit immunologically 'cold' microenvironment, where the vascular barrier impedes effective immunotherapy in poorly understood ways. Tumor vasculature also plays a pivotal role in immunoregulation and antitumor immunity. Here, we show that small GTPase Rab27 controls the vascular morphogenesis and permeability for blood content and immune effectors. Thus, in Rab27a/b double knock out (Rab27-dKO) mice, the brain vasculature is abnormally scarce, while the blood vessels become dysmorphic and hyperpermeable in the context of brain tumors, including syngeneic glioblastoma. These defects are reflected in rearrangements of endothelial cell subpopulations with underlying diminution of venous endothelial subtype along with changes in gene and protein expression. Notably, Rab27-dKO brain endothelial cells exhibit deficient tight junctions, whereby they enable large-scale extravasation of cytotoxic T cells into the tumor mass. We show that Rab27-regulated vascular T cell infiltration can be exploited to enhance adoptive T cell therapy in syngeneic brain tumors.
Collapse
Affiliation(s)
- Lata Adnani
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Brian Meehan
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Minjun Kim
- Victor Phillip Dahdaleh Institute of Genomic Medicine at McGill University. McGill University Department of Human Genetics, Montreal, QC, Canada
| | - Dongsic Choi
- Department of Biochemistry, College of Medicine, Soonchunhyang University, Cheonan, Chungcheongnam 31151, Republic of Korea
| | - Christopher E. Rudd
- Division of Immunology-Oncology Research Center, Maisonneuve-Rosemont Hospital, Montreal, QC H1T 2M4, Canada
- Département de Medicine, Université de Montréal, Montreal, QC H3C 3J7, Canada
- Centre for Translational Research in Cancer, McGill University, Montreal, QC, Canada
| | - Yasser Riazalhosseini
- Victor Phillip Dahdaleh Institute of Genomic Medicine at McGill University. McGill University Department of Human Genetics, Montreal, QC, Canada
| | - Janusz Rak
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- Department of Pediatrics, McGill University, Montreal, QC H4A 3J1, Canada
| |
Collapse
|
2
|
Powell JS, Larregina AT, Shufesky WJ, Sullivan ML, Stolz DB, Gould SJ, Camirand G, Catz SD, Watkins SC, Sadovsky Y, Morelli AE. Fetoplacental extracellular vesicles deliver conceptus-derived antigens to maternal secondary lymphoid tissues for immune recognition. JCI Insight 2025; 10:e186335. [PMID: 40401522 DOI: 10.1172/jci.insight.186335] [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: 09/06/2024] [Accepted: 04/09/2025] [Indexed: 05/23/2025] Open
Abstract
Pregnancy is an immunological paradox where despite a competent maternal immune system, regulatory mechanisms at the fetoplacental interface and maternal secondary lymphoid tissues (SLTs) circumvent rejection of semi-allogeneic concepti. Small extracellular vesicles (sEVs) are a vehicle for intercellular communication; nevertheless, the role of fetoplacental sEVs in transport of antigens to maternal SLTs has not been conclusively demonstrated. Using mice in which the conceptus generates fluoroprobe-tagged sEVs shed by the plasma membrane or released from the endocytic compartment, we show that fetoplacental sEVs are delivered to immune cells in the maternal spleen. Injection of sEVs from placentas of females impregnated with Act-mOVA B6 males elicited suboptimal activation of OVA-specific CD8+ OT-I T cells in virgin females as occurs during pregnancy. Furthermore, when OVA+ concepti were deficient in Rab27a, a protein required for sEV secretion, OT-I cell proliferation in the maternal spleen was decreased. Proteomics analysis revealed that mouse trophoblast sEVs were enriched in antiinflammatory and immunosuppressive mediators. Translational relevance was tested in humanized mice injected using sEVs from cultures of human trophoblasts. Our findings show that sEVs deliver fetoplacental antigens to the mother's SLTs that are recognized by maternal T cells. Alterations of such a mechanism may lead to pregnancy disorders.
Collapse
Affiliation(s)
- Juliana S Powell
- T.E. Starzl Transplantation Institute, Department of Surgery
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens Research Institute
| | - Adriana T Larregina
- Department of Dermatology
- Department of Immunology
- McGowan Institute for Regenerative Medicine; and
| | | | - Mara Lg Sullivan
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Donna Beer Stolz
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Stephen J Gould
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Geoffrey Camirand
- T.E. Starzl Transplantation Institute, Department of Surgery
- Department of Immunology
| | - Sergio D Catz
- The Scripps Research Institute, La Jolla, California, USA
| | - Simon C Watkins
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yoel Sadovsky
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens Research Institute
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Adrian E Morelli
- T.E. Starzl Transplantation Institute, Department of Surgery
- Department of Immunology
| |
Collapse
|
3
|
Hu S, Chen G, Luo A, Zhao H, Li D, Peng B, Du J, Luo D. Mechanism of LINC01018/miR-182-5p/Rab27B in the immune escape through PD-L1-mediated CD8 + T cell suppression in glioma. Biol Direct 2025; 20:61. [PMID: 40399992 PMCID: PMC12093642 DOI: 10.1186/s13062-025-00651-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 04/15/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND Glioma is a malignant tumor associated with poorer prognosis. This study aims to elucidate the mechanism of LINC01018/miR-182-5p/Rab27B axis in PD-L1-mediated CD8+ T cell suppression in the progression of gliomas. METHODS LINC01018, miR-182-5p, and Rab27B expression levels in glioblastoma tissues were measured. The proportion of infiltrating macrophages and monocytes and CD8+ T cell function were assessed. The relationship between miR-182-5p and Rab27B was analyzed. Glioma cell activity, invasion, and migration were measured. The expression of E-cadherin, N-cadherin, Vimentin, PD-L1, iNOS, and CD206 was determined. Glioma cell-derived EVs were isolated, and the co-localization of Rab27B and PD-L1 and the binding of Rab27B to PD-L1 were analyzed. The endocytosis of EVs by microglia was assayed. The impact of LINC01018/miR-182-5p/Rab27B on glioma growth was observed. The function of macrophages and CD8+ T cells in tumors was analyzed. RESULTS Rab27B was downregulated, and infiltrating macrophages and monocytes were increased in glioblastoma. miR-182-5p inhibited Rab27B expression. Rab27B knockdown reverses the inhibitory effect of LINC01018 overexpression on glioma cell growth. Glioma cells-derived EVs with low Rab27B expression carried more PD-L1 to increase PD-L1 expression and M2 polarization in microglia. LINC01018 overexpression reduced macrophages in orthotopic tumors. CD8+ T cell numbers showed no significant difference, but TIM-3 increased and IFN-γ decreased. miR-182-5p inhibition enhanced the therapeutic effect of anti-PD-L1, which was reversed after glioma cell-derived EVs. CONCLUSION LINC01018 promotes PD-L1-mediated CD8+ T cell suppression via the miR-182-5p/Rab27B axis in glioma cell-derived EVs, thereby contributing to immune escape in gliomas.
Collapse
Affiliation(s)
- Su Hu
- Department of Neurosurgery, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Guoshuo Chen
- Department of Intervention, the Affiliated Cancer Hospital, Guangzhou Institute of Cancer Research, Guangzhou Medical University, Guangzhou, 510095, China
| | - Aiping Luo
- Department of Radiology, the Affiliated Cancer Hospital, Guangzhou Institute of Cancer Research, Guangzhou Medical University, Guangzhou, 510095, China
| | - Hailin Zhao
- Department of Neurosurgery, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Dan Li
- Department of Neurosurgery, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Biao Peng
- Department of Neurosurgery, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China.
| | - Jike Du
- Department of Medical Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China.
| | - Dongdong Luo
- Department of Neurosurgery, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China.
| |
Collapse
|
4
|
Tanaka M, Nakamura T. Role of the RAB27/SYTL Axis in Tumor Microenvironment Construction. Cancer Sci 2025. [PMID: 40319893 DOI: 10.1111/cas.70096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/12/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025] Open
Abstract
Crosstalk between cancer cells and the tumor microenvironment (TME) is a key event in malignant progression and metastasis. The secretion of bioactive substances by cancer cells remodels the TME, affecting the activities of its components, including blood vessels, mesenchymal cells, and immune cells. These substances are effectively delivered through intracellular trafficking and exocytosis of cytoplasmic vesicles. The small guanosine triphosphatase (GTPase) RAB27 and its effectors, synaptotagmin-like (SYTL) family proteins, play essential roles in vesicle trafficking. Our recent research demonstrates the upregulation of RAB27A/B and SYTL1/2 in alveolar soft part sarcoma and acute myeloid leukemia. This enhanced trafficking promotes angiogenesis and the occupation of leukemia cells in the bone marrow niche. This review focuses on the role of the RAB27/SYTL axis in various cancer types associated with TME modifications, with a discussion on its importance as a therapeutic target.
Collapse
Affiliation(s)
- Miwa Tanaka
- Division of Cancer Epigenomics, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
- Department of Experimental Pathology, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Takuro Nakamura
- Department of Experimental Pathology, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
5
|
Li J, Kemper T, Broering R, Lin Y, Wang X, Lu M. Amphisome plays a role in HBV production and release through the endosomal and autophagic pathways. Hepatol Commun 2025; 9:e0654. [PMID: 40079732 PMCID: PMC11908759 DOI: 10.1097/hc9.0000000000000654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 01/08/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND Autophagic and endosomal pathways coordinately contribute to HBV virions and subviral particles (SVPs) production. To date, limited evidence supports that HBV and exosomes have a common pathway for their biogenesis and secretion. The final steps of HBV production and release have not yet been well studied. METHODS We examined the production and release of HBV virions and SVPs by using GW4869 (N,N'-Bis[4-(4,5-dihydro-1H-imidazol-2-yl)phenyl]-3,3'-pht hal amide dihydrochloride), a small molecule inhibiting ceramide-mediated inward membrane budding. Neutral sphingomyelinase, the target of GW4869, and RAB27A and -B, 2 small GTPases involved in exosome release control, were silenced using gene silencing to confirm the results obtained. Western blot, immunofluorescence staining, and confocal microscopy were applied. RESULTS GW4869 inhibited HBV virion release, causing their accumulation along with SVPs in hepatocytes. This triggered cellular endoplasmic reticulum stress, leading to protein kinase B-mechanistic target of rapamycin kinase signaling pathway inactivation. GW4869 treatment increased autophagosome formation and impaired autophagic degradation by blocking autophagosome-lysosome fusion. Consequently, HBsAg is increasingly localized to autophagosomes and late endosomes/multivesicular bodies. Silencing neutral sphingomyelinase yielded consistent results. Similarly, RAB27A silencing inhibited HBV virion and SVP secretion, causing their accumulation within hepatoma cells. Notably, GW4869 treatment, as well as RAB27A and -B silencing, increased the presence of LC3+CD63+HBsAg+ complexes. CONCLUSIONS Our results demonstrate the involvement of the autophagosome-late endosome/multivesicular bodies-exosome axis in regulating HBV production and release, highlighting amphisomes as a potential platform for HBV release.
Collapse
Affiliation(s)
- Jia Li
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Thekla Kemper
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ruth Broering
- Department of Gastroenterology, Hepatology and Transplant Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Yong Lin
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Xueyu Wang
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Mengji Lu
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
6
|
Horodecka K, Czernek L, Pęczek Ł, Klink M. Revealing the role of RAB27 in HER receptor family expression and signaling in melanoma cells. Cell Commun Signal 2025; 23:118. [PMID: 40038749 PMCID: PMC11877929 DOI: 10.1186/s12964-025-02064-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/25/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Alterations in signalling pathways fuel the growth and progression of melanoma. Therefore, understanding these processes is essential for developing effective treatment strategies. RAB27A and RAB27B are known to possess oncogenic effects by modulating cancer cell proliferation, invasion and drug resistance in various types of cancer, including melanoma. These proteins are mostly acknowledged as coordinators of the vesicular trafficking, however, their function in cellular signaling is less recognized. Therefore we aimed to investigate the relationship between RAB27 and oncogenic or signalling proteins in melanoma cells. METHODS We generated RAB27A knockout (KO) in SkMel28, A375, and patient-derived DMBC12 melanoma cell lines. Additionally, a double RAB27A/B knockout (dKO) A375 cell line was created. Firstly, we applied the Proteome Profiler array to identify proteins differentially expressed upon RAB27A/B loss. Subsequently, we picked selected specific proteins for a further in-depth analysis using RT-PCR, Western blot, and flow cytometry. RESULTS We found that silencing RAB27 markedly decreased the levels of various intracellular proteins linked with proliferation, invasion, angiogenesis, adhesion, or EMT at a cell-line dependent level. Among others, we observed a link between the expression of RAB27 and EGFR, HER2 and HER3. Altered levels of HER receptors disturbed the downstream signaling pathways by reducing the phosphorylation of AKT and ERK1/2 proteins. CONCLUSIONS Our findings present novel, previously unpublished data on the relationship between HER family receptor expression and potential activity, and the involvement of RAB27 in melanoma cells.
Collapse
Affiliation(s)
- Katarzyna Horodecka
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112, Lodz, 90-363, Poland.
| | - Liliana Czernek
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112, Lodz, 90-363, Poland
| | - Łukasz Pęczek
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112, Lodz, 90-363, Poland
| | - Magdalena Klink
- Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106 Str., Lodz, 93-232, Poland.
| |
Collapse
|
7
|
Li X, Qiao F, Guo J, Jiang T, Lou H, Li H, Xie G, Wu H, Wang W, Pei R, Liu S, Ye M, Li J, Huang S, Zhang M, Ma C, Huang Y, Xu S, Li X, Sun X, Yu J, Fok KL, Duan S, Chen H. In situ architecture of the intercellular organelle reservoir between epididymal epithelial cells by volume electron microscopy. Nat Commun 2025; 16:1664. [PMID: 39955273 PMCID: PMC11830104 DOI: 10.1038/s41467-025-56807-9] [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: 01/11/2024] [Accepted: 01/31/2025] [Indexed: 02/17/2025] Open
Abstract
Mammalian epididymal epithelial cells are crucial for sperm maturation. Historically, vacuole-like ultrastructures in epididymal epithelial cells were observed via transmission electron microscopy but were undefined. Here, we utilize volume electron microscopy (vEM) to generate 3D reconstructions of epididymal epithelial cells and identify these vacuoles as intercellular organelle reservoirs (IORs) in the lateral intercellular space (LIS), which contains protein aggregates, autophagosomes, lysosome-related organelles and mitochondrial residues. Immunolabelling of organelle markers such as P62, LC3, LAMP1 and TOMM20 confirm these findings. The IOR size or number varies across four epididymal regions and decreases with age. Rab27a mutant mice exhibit reduced IORs in the caput epididymis and a subfertility phenotype, suggesting the involvement of Rab27a in the formation of IORs. Furthermore, we observe the presence of IORs between intestinal epithelial cells besides epididymis. Amino acid transporters at IOR edges suggest dynamic protein recycling. Our findings reveal that the IOR is an important structure critical for organelle turnover and recycling outside epithelial cells with limited self-degradation capabilities.
Collapse
Affiliation(s)
- Xia Li
- Institute of Special Environmental Medicine and Medical School, Nantong University and Guangzhou Women and Children's Medical Center, GMU-GIBH Joint School of Life Science, Guangzhou Medical University, Guangzhou, PR China
| | - Feng Qiao
- Institute of Special Environmental Medicine and Medical School, Nantong University and Guangzhou Women and Children's Medical Center, GMU-GIBH Joint School of Life Science, Guangzhou Medical University, Guangzhou, PR China
| | - Jiansheng Guo
- School of Brain Science and Brain Medicine and Center of Cryo-Electron Microscopy, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, PR China
| | - Ting Jiang
- Institute of Special Environmental Medicine and Medical School, Nantong University and Guangzhou Women and Children's Medical Center, GMU-GIBH Joint School of Life Science, Guangzhou Medical University, Guangzhou, PR China
| | - Huifang Lou
- School of Brain Science and Brain Medicine and Center of Cryo-Electron Microscopy, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, PR China
| | - Huixia Li
- Institute of Special Environmental Medicine and Medical School, Nantong University and Guangzhou Women and Children's Medical Center, GMU-GIBH Joint School of Life Science, Guangzhou Medical University, Guangzhou, PR China
| | - Gangcai Xie
- Institute of Special Environmental Medicine and Medical School, Nantong University and Guangzhou Women and Children's Medical Center, GMU-GIBH Joint School of Life Science, Guangzhou Medical University, Guangzhou, PR China
| | - Hangjun Wu
- School of Brain Science and Brain Medicine and Center of Cryo-Electron Microscopy, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, PR China
| | - Weizhen Wang
- Institute of Special Environmental Medicine and Medical School, Nantong University and Guangzhou Women and Children's Medical Center, GMU-GIBH Joint School of Life Science, Guangzhou Medical University, Guangzhou, PR China
| | - Ruoyu Pei
- Institute of Special Environmental Medicine and Medical School, Nantong University and Guangzhou Women and Children's Medical Center, GMU-GIBH Joint School of Life Science, Guangzhou Medical University, Guangzhou, PR China
| | - Sha Liu
- Institute of Special Environmental Medicine and Medical School, Nantong University and Guangzhou Women and Children's Medical Center, GMU-GIBH Joint School of Life Science, Guangzhou Medical University, Guangzhou, PR China
| | - Mei Ye
- Institute of Special Environmental Medicine and Medical School, Nantong University and Guangzhou Women and Children's Medical Center, GMU-GIBH Joint School of Life Science, Guangzhou Medical University, Guangzhou, PR China
| | - Jin Li
- Institute of Special Environmental Medicine and Medical School, Nantong University and Guangzhou Women and Children's Medical Center, GMU-GIBH Joint School of Life Science, Guangzhou Medical University, Guangzhou, PR China
| | - Shiqin Huang
- Institute of Special Environmental Medicine and Medical School, Nantong University and Guangzhou Women and Children's Medical Center, GMU-GIBH Joint School of Life Science, Guangzhou Medical University, Guangzhou, PR China
| | - Mengya Zhang
- Institute of Special Environmental Medicine and Medical School, Nantong University and Guangzhou Women and Children's Medical Center, GMU-GIBH Joint School of Life Science, Guangzhou Medical University, Guangzhou, PR China
| | - Chaoye Ma
- Institute of Special Environmental Medicine and Medical School, Nantong University and Guangzhou Women and Children's Medical Center, GMU-GIBH Joint School of Life Science, Guangzhou Medical University, Guangzhou, PR China
| | - Yiwen Huang
- Institute of Special Environmental Medicine and Medical School, Nantong University and Guangzhou Women and Children's Medical Center, GMU-GIBH Joint School of Life Science, Guangzhou Medical University, Guangzhou, PR China
| | - Shushu Xu
- Institute of Special Environmental Medicine and Medical School, Nantong University and Guangzhou Women and Children's Medical Center, GMU-GIBH Joint School of Life Science, Guangzhou Medical University, Guangzhou, PR China
| | - Xiaofeng Li
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, No. 1120 Lianhua Road, Futian District, Shenzhen, PR China
| | - Xiao Sun
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Jun Yu
- Institute of Special Environmental Medicine and Medical School, Nantong University and Guangzhou Women and Children's Medical Center, GMU-GIBH Joint School of Life Science, Guangzhou Medical University, Guangzhou, PR China
| | - Kin Lam Fok
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Shumin Duan
- School of Brain Science and Brain Medicine and Center of Cryo-Electron Microscopy, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, PR China.
| | - Hao Chen
- Institute of Special Environmental Medicine and Medical School, Nantong University and Guangzhou Women and Children's Medical Center, GMU-GIBH Joint School of Life Science, Guangzhou Medical University, Guangzhou, PR China.
- Key Laboratory of Reproductive Health Diseases Research and Translation of Ministry of Education & Key Laboratory of Human Reproductive Medicine and Genetic Research of Hainan Province & Hainan Provincial Clinical Research Center for Thalassemia, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, Hainan, China.
| |
Collapse
|
8
|
Sokolowski EK, Kursawe R, Selvam V, Bhuiyan RM, Thibodeau A, Zhao C, Spracklen CN, Ucar D, Stitzel ML. Multi-omic human pancreatic islet endoplasmic reticulum and cytokine stress response mapping provides type 2 diabetes genetic insights. Cell Metab 2024; 36:2468-2488.e7. [PMID: 39383866 PMCID: PMC11798411 DOI: 10.1016/j.cmet.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 06/14/2024] [Accepted: 09/10/2024] [Indexed: 10/11/2024]
Abstract
Endoplasmic reticulum (ER) and inflammatory stress responses contribute to islet dysfunction in type 2 diabetes (T2D). Comprehensive genomic understanding of these human islet stress responses and whether T2D-associated genetic variants modulate them is lacking. Here, comparative transcriptome and epigenome analyses of human islets exposed ex vivo to these stressors revealed 30% of expressed genes and 14% of islet cis-regulatory elements (CREs) as stress responsive, modulated largely in an ER- or cytokine-specific fashion. T2D variants overlapped 86 stress-responsive CREs, including 21 induced by ER stress. We linked the rs6917676-T T2D risk allele to increased islet ER-stress-responsive CRE accessibility and allele-specific β cell nuclear factor binding. MAP3K5, the ER-stress-responsive putative rs6917676 T2D effector gene, promoted stress-induced β cell apoptosis. Supporting its pro-diabetogenic role, MAP3K5 expression correlated inversely with human islet β cell abundance and was elevated in T2D β cells. This study provides genome-wide insights into human islet stress responses and context-specific T2D variant effects.
Collapse
Affiliation(s)
- Eishani K Sokolowski
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Romy Kursawe
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Vijay Selvam
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Redwan M Bhuiyan
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Asa Thibodeau
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Chi Zhao
- Department of Biostatistics and Epidemiology, University of Massachusetts, Amherst, Amherst, MA 01003, USA
| | - Cassandra N Spracklen
- Department of Biostatistics and Epidemiology, University of Massachusetts, Amherst, Amherst, MA 01003, USA
| | - Duygu Ucar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06032, USA; Institute of Systems Genomics, University of Connecticut, Farmington, CT 06032, USA.
| | - Michael L Stitzel
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06032, USA; Institute of Systems Genomics, University of Connecticut, Farmington, CT 06032, USA.
| |
Collapse
|
9
|
Wang X, Nie X, Wang H, Ren Z. Roles of small GTPases in cardiac hypertrophy (Review). Mol Med Rep 2024; 30:208. [PMID: 39301654 PMCID: PMC11425065 DOI: 10.3892/mmr.2024.13332] [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: 04/10/2024] [Accepted: 08/20/2024] [Indexed: 09/22/2024] Open
Abstract
Cardiac hypertrophy results from the heart reacting and adapting to various pathological stimuli and its persistent development is a major contributing factor to heart failure. However, the molecular mechanisms of cardiac hypertrophy remain unclear. Small GTPases in the Ras, Rho, Rab, Arf and Ran subfamilies exhibit GTPase activity and play crucial roles in regulating various cellular responses. Previous studies have shown that Ras, Rho and Rab are closely linked to cardiac hypertrophy and that their overexpression can induce cardiac hypertrophy. Here, we review the functions of small GTPases in cardiac hypertrophy and provide additional insights and references for the prevention and treatment of cardiac hypertrophy.
Collapse
Affiliation(s)
- Xin Wang
- School of Mathematics and Statistics, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Xinwen Nie
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
- School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Hao Wang
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
- School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Zhanhong Ren
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| |
Collapse
|
10
|
Huang J, Guo J, Jia R. N6-Methyladenosine Methyltransferase Component KIAA1429 Is a Potential Target of Cancer Therapy. Biomolecules 2024; 14:1319. [PMID: 39456252 PMCID: PMC11506059 DOI: 10.3390/biom14101319] [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: 09/15/2024] [Revised: 10/11/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
N6-methyladenosine (m6A), the most abundant RNA modification in eukaryotes, has a crucial impact on tumorigenesis. KIAA1429 is the key component of the m6A methyltransferase complex, in which KIAA1429 functions as a scaffold to bridge the catalytic core proteins. KIAA1429 is often overexpressed in malignances, associated with patient prognosis, and required for tumorigenesis. KIAA1429 regulates the expression of a number of tumor-associated genes in an m6A -dependent manner, and thus, contributes to cell proliferation, migration, drug resistance, tumor formation and metastasis. This review focuses on recent progress in the understanding of roles and mechanisms of KIAA1429 in cancers, and offers ideas for potential anti-cancer therapeutic methods by targeting KIAA1429.
Collapse
Affiliation(s)
- Junjun Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China; (J.H.); (J.G.)
| | - Jihua Guo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China; (J.H.); (J.G.)
- Department of Endodontics, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
| | - Rong Jia
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China; (J.H.); (J.G.)
| |
Collapse
|
11
|
Fritzen L, Wienken K, Wagner L, Kurtyka M, Vogel K, Körbelin J, Weggen S, Fricker G, Pietrzik CU. Truncated mini LRP1 transports cargo from luminal to basolateral side across the blood brain barrier. Fluids Barriers CNS 2024; 21:74. [PMID: 39289695 PMCID: PMC11409491 DOI: 10.1186/s12987-024-00573-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/02/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND The most crucial area to focus on when thinking of novel pathways for drug delivery into the CNS is the blood brain barrier (BBB). A number of nanoparticulate formulations have been shown in earlier research to target receptors at the BBB and transport therapeutics into the CNS. However, no mechanism for CNS entrance and movement throughout the CNS parenchyma has been proposed yet. Here, the truncated mini low-density lipoprotein receptor-related protein 1 mLRP1_DIV* was presented as blood to brain transport carrier, exemplified by antibodies and immunoliposomes using a systematic approach to screen the receptor and its ligands' route across endothelial cells in vitro. METHODS The use of mLRP1_DIV* as liposomal carrier into the CNS was validated based on internalization and transport assays across an in vitro model of the BBB using hcMEC/D3 and bEnd.3 cells. Trafficking routes of mLRP1_DIV* and corresponding cargo across endothelial cells were analyzed using immunofluorescence. Modulation of γ-secretase activity by immunoliposomes loaded with the γ-secretase modulator BB25 was investigated in co-cultures of bEnd.3 mLRP1_DIV* cells and CHO cells overexpressing human amyloid precursor protein (APP) and presenilin 1 (PSEN1). RESULTS We showed that while expressed in vitro, mLRP1_DIV* transports both, antibodies and functionalized immunoliposomes from luminal to basolateral side across an in vitro model of the BBB, followed by their mLRP1_DIV* dependent release of the cargo. Importantly, functionalized liposomes loaded with the γ-secretase modulator BB25 were demonstrated to effectively reduce toxic Aß42 peptide levels after mLRP1_DIV* mediated transport across a co-cultured endothelial monolayer. CONCLUSION Together, the data strongly suggest mLRP1_DIV* as a promising tool for drug delivery into the CNS, as it allows a straight transport of cargo from luminal to abluminal side across an endothelial monolayer and it's release into brain parenchyma in vitro, where it exhibits its intended therapeutic effect.
Collapse
Affiliation(s)
- Laura Fritzen
- Molecular Neurodegeneration, Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University of Mainz, Duesbergweg 6, 55099, Mainz, Germany.
| | - Katharina Wienken
- Institute for Pharmacy and Molecular Biotechnology, University Heidelberg, Heidelberg, Germany
| | - Lelia Wagner
- Institute for Pharmacy and Molecular Biotechnology, University Heidelberg, Heidelberg, Germany
| | - Magdalena Kurtyka
- Molecular Neurodegeneration, Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University of Mainz, Duesbergweg 6, 55099, Mainz, Germany
| | - Katharina Vogel
- Department of Neuropathology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Jakob Körbelin
- Department for Oncology and Hematology, University Medical Center Hamburg-Eppendorf, Hubertus Wald Cancer Center, Hamburg, Germany
| | - Sascha Weggen
- Department of Neuropathology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Gert Fricker
- Institute for Pharmacy and Molecular Biotechnology, University Heidelberg, Heidelberg, Germany
| | - Claus U Pietrzik
- Molecular Neurodegeneration, Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University of Mainz, Duesbergweg 6, 55099, Mainz, Germany.
| |
Collapse
|
12
|
Jang E, Ho TWW, Brumell JH, Lefebvre F, Wang C, Lee WL. IL-1β Induces LDL Transcytosis by a Novel Pathway Involving LDLR and Rab27a. Arterioscler Thromb Vasc Biol 2024; 44:2053-2068. [PMID: 38989581 DOI: 10.1161/atvbaha.124.320940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 06/26/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND In early atherosclerosis, circulating LDLs (low-density lipoproteins) traverse individual endothelial cells by an active process termed transcytosis. The CANTOS trial (Canakinumab Antiinflammatory Thrombosis Outcome Study) treated advanced atherosclerosis using a blocking antibody for IL-1β (interleukin-1β); this significantly reduced cardiovascular events. However, whether IL-1β regulates early disease, particularly LDL transcytosis, remains unknown. METHODS We used total internal reflection fluorescence microscopy to quantify transcytosis by human coronary artery endothelial cells exposed to IL-1β. To investigate transcytosis in vivo, we injected wild-type and knockout mice with IL-1β and LDL to visualize acute LDL deposition in the aortic arch. RESULTS Exposure to picomolar concentrations of IL-1β induced transcytosis of LDL but not of albumin by human coronary artery endothelial cells. Surprisingly, expression of the 2 known receptors for LDL transcytosis, ALK-1 (activin receptor-like kinase-1) and SR-BI (scavenger receptor BI), was unchanged or decreased. Instead, IL-1β increased the expression of the LDLR (LDL receptor); this was unexpected because LDLR is not required for LDL transcytosis. Overexpression of LDLR had no effect on basal LDL transcytosis. However, knockdown of LDLR abrogated the effect of IL-1β on transcytosis rates while the depletion of Cav-1 (caveolin-1) did not. Since LDLR was necessary but overexpression had no effect, we reasoned that another player must be involved. Using public RNA sequencing data to curate a list of Rab (Ras-associated binding) GTPases affected by IL-1β, we identified Rab27a. Overexpression of Rab27a alone had no effect on basal transcytosis, but its knockdown prevented induction by IL-1β. This was phenocopied by depletion of the Rab27a effector JFC1 (synaptotagmin-like protein 1). In vivo, IL-1β increased LDL transcytosis in the aortic arch of wild-type but not Ldlr-/- or Rab27a-deficient mice. The JFC1 inhibitor nexinhib20 also blocked IL-1β-induced LDL accumulation in the aorta. CONCLUSIONS IL-1β induces LDL transcytosis by a distinct pathway requiring LDLR and Rab27a; this route differs from basal transcytosis. We speculate that induction of transcytosis by IL-1β may contribute to the acceleration of early disease.
Collapse
Affiliation(s)
- Erika Jang
- Keenan Centre for Biomedical Research, St. Michael's Hospital, Toronto, ON, Canada (E.J., T.W.W.H., C.W., W.L.L.)
- Department of Laboratory Medicine and Pathobiology (E.J., T.W.W.H., W.L.L.), University of Toronto, ON, Canada
| | - Tse Wing Winnie Ho
- Keenan Centre for Biomedical Research, St. Michael's Hospital, Toronto, ON, Canada (E.J., T.W.W.H., C.W., W.L.L.)
- Department of Laboratory Medicine and Pathobiology (E.J., T.W.W.H., W.L.L.), University of Toronto, ON, Canada
| | - John H Brumell
- Cell Biology Program, Hospital for Sick Children, Toronto, ON, Canada (J.H.B.)
| | - François Lefebvre
- Canadian Centre for Computational Genomics, McGill University, Montreal, QC, Canada (F.L.)
| | - Changsen Wang
- Keenan Centre for Biomedical Research, St. Michael's Hospital, Toronto, ON, Canada (E.J., T.W.W.H., C.W., W.L.L.)
| | - Warren L Lee
- Keenan Centre for Biomedical Research, St. Michael's Hospital, Toronto, ON, Canada (E.J., T.W.W.H., C.W., W.L.L.)
- Department of Laboratory Medicine and Pathobiology (E.J., T.W.W.H., W.L.L.), University of Toronto, ON, Canada
- Department of Biochemistry (W.L.L.), University of Toronto, ON, Canada
- Department of Medicine and the Interdepartmental Division of Critical Care Medicine (W.L.L.), University of Toronto, ON, Canada
| |
Collapse
|
13
|
Leng K, Rooney B, McCarthy F, Xia W, Rose IVL, Bax S, Chin M, Fathi S, Herrington KA, Leonetti M, Kao A, Fancy SPJ, Elias JE, Kampmann M. mTOR activation induces endolysosomal remodeling and nonclassical secretion of IL-32 via exosomes in inflammatory reactive astrocytes. J Neuroinflammation 2024; 21:198. [PMID: 39118084 PMCID: PMC11312292 DOI: 10.1186/s12974-024-03165-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 07/03/2024] [Indexed: 08/10/2024] Open
Abstract
Astrocytes respond and contribute to neuroinflammation by adopting inflammatory reactive states. Although recent efforts have characterized the gene expression signatures associated with these reactive states, the cell biology underlying inflammatory reactive astrocyte phenotypes remains under-explored. Here, we used CRISPR-based screening in human iPSC-derived astrocytes to identify mTOR activation a driver of cytokine-induced endolysosomal system remodeling, manifesting as alkalinization of endolysosomal compartments, decreased autophagic flux, and increased exocytosis of certain endolysosomal cargos. Through endolysosomal proteomics, we identified and focused on one such cargo-IL-32, a disease-associated pro-inflammatory cytokine not present in rodents, whose secretion mechanism is not well understood. We found that IL-32 was partially secreted in extracellular vesicles likely to be exosomes. Furthermore, we found that IL-32 was involved in the polarization of inflammatory reactive astrocyte states and was upregulated in astrocytes in multiple sclerosis lesions. We believe that our results advance our understanding of cell biological pathways underlying inflammatory reactive astrocyte phenotypes and identify potential therapeutic targets.
Collapse
Affiliation(s)
- Kun Leng
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA.
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA.
- Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA, USA.
| | - Brendan Rooney
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
| | | | - Wenlong Xia
- Departments of Neurology and Pediatrics, School of Medicine, University of California, San Francisco, CA, USA
| | - Indigo V L Rose
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Sophie Bax
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Marcus Chin
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- Small Molecule Discovery Center, Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Saeed Fathi
- Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA, USA
| | - Kari A Herrington
- Center for Advanced Microscopy, University of California, San Francisco, San Francisco, CA, USA
| | | | - Aimee Kao
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Stephen P J Fancy
- Departments of Neurology and Pediatrics, School of Medicine, University of California, San Francisco, CA, USA
| | | | - Martin Kampmann
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA.
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
14
|
Schöl M, Schempp R, Hennig T, Wigger D, Schumacher F, Kleuser B, Stigloher C, van Ham M, Jänsch L, Schneider-Schaulies S, Dölken L, Avota E. Dynamic changes in the proximitome of neutral sphingomyelinase-2 (nSMase2) in TNFα stimulated Jurkat cells. Front Immunol 2024; 15:1435701. [PMID: 39044828 PMCID: PMC11263205 DOI: 10.3389/fimmu.2024.1435701] [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: 05/20/2024] [Accepted: 06/17/2024] [Indexed: 07/25/2024] Open
Abstract
Ceramides generated by the activity of the neutral sphingomyelinase 2 (nSMase2) play a pivotal role in stress responses in mammalian cells. Dysregulation of sphingolipid metabolism has been implicated in numerous inflammation-related pathologies. However, its influence on inflammatory cytokine-induced signaling is yet incompletely understood. Here, we used proximity labeling to explore the plasma membrane proximal protein network of nSMase2 and TNFα-induced changes thereof. We established Jurkat cells stably expressing nSMase2 C-terminally fused to the engineered ascorbate peroxidase 2 (APEX2). Removal of excess biotin phenol substantially improved streptavidin-based affinity purification of biotinylated proteins. Using our optimized protocol, we determined nSMase2-proximal biotinylated proteins and their changes within the first 5 min of TNFα stimulation by quantitative mass spectrometry. We observed significant dynamic changes in the nSMase2 microenvironment in response to TNFα stimulation consistent with rapid remodeling of protein networks. Our data confirmed known nSMase2 interactors and revealed that the recruitment of most proteins depended on nSMase2 enzymatic activity. We measured significant enrichment of proteins related to vesicle-mediated transport, including proteins of recycling endosomes, trans-Golgi network, and exocytic vesicles in the proximitome of enzymatically active nSMase2 within the first minutes of TNFα stimulation. Hence, the nSMase2 proximal network and its TNFα-induced changes provide a valuable resource for further investigations into the involvement of nSMase2 in the early signaling pathways triggered by TNFα.
Collapse
Affiliation(s)
- Marie Schöl
- Institute for Virology and Immunobiology, University of Wuerzburg, Würzburg, Germany
| | - Rebekka Schempp
- Institute for Virology and Immunobiology, University of Wuerzburg, Würzburg, Germany
| | - Thomas Hennig
- Institute for Virology and Immunobiology, University of Wuerzburg, Würzburg, Germany
| | - Dominik Wigger
- Institute of Pharmacy, Department of Pharmacology & Toxicology, Freie Universität Berlin, Berlin, Germany
| | - Fabian Schumacher
- Institute of Pharmacy, Department of Pharmacology & Toxicology, Freie Universität Berlin, Berlin, Germany
| | - Burkhard Kleuser
- Institute of Pharmacy, Department of Pharmacology & Toxicology, Freie Universität Berlin, Berlin, Germany
| | - Christian Stigloher
- Imaging Core Facility, Biocenter, University of Wuerzburg, Würzburg, Germany
| | - Marco van Ham
- Cellular Proteome Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Lothar Jänsch
- Cellular Proteome Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | - Lars Dölken
- Institute of Virology, Medizinische Hochschule Hannover, Hannover, Germany
| | - Elita Avota
- Institute for Virology and Immunobiology, University of Wuerzburg, Würzburg, Germany
| |
Collapse
|
15
|
Zakeri Z, Heiderzadeh M, Kocaarslan A, Metin E, Hosseini Karimi SN, Saghati S, Vural A, Akyoldaş G, Baysal K, Yağcı Y, Gürsoy-Özdemir Y, Taşoğlu S, Rahbarghazi R, Sokullu E. Exosomes encapsulated in hydrogels for effective central nervous system drug delivery. Biomater Sci 2024; 12:2561-2578. [PMID: 38602364 DOI: 10.1039/d3bm01055d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
The targeted delivery of pharmacologically active molecules, metabolites, and growth factors to the brain parenchyma has become one of the major challenges following the onset of neurodegeneration and pathological conditions. The therapeutic effect of active biomolecules is significantly impaired after systemic administration in the central nervous system (CNS) because of the blood-brain barrier (BBB). Therefore, the development of novel therapeutic approaches capable of overcoming these limitations is under discussion. Exosomes (Exo) are nano-sized vesicles of endosomal origin that have a high distribution rate in biofluids. Recent advances have introduced Exo as naturally suitable bio-shuttles for the delivery of neurotrophic factors to the brain parenchyma. In recent years, many researchers have attempted to regulate the delivery of Exo to target sites while reducing their removal from circulation. The encapsulation of Exo in natural and synthetic hydrogels offers a valuable strategy to address the limitations of Exo, maintaining their integrity and controlling their release at a desired site. Herein, we highlight the current and novel approaches related to the application of hydrogels for the encapsulation of Exo in the field of CNS tissue engineering.
Collapse
Affiliation(s)
- Ziba Zakeri
- Research Center for Translational Medicine (KUTTAM), Koç University, Rumeli Feneri, 34450, Istanbul, Sariyer, Turkey.
| | - Morteza Heiderzadeh
- Research Center for Translational Medicine (KUTTAM), Koç University, Rumeli Feneri, 34450, Istanbul, Sariyer, Turkey.
| | - Azra Kocaarslan
- Chemistry Department, Faculty of Science, İstanbul Technical University, İstanbul, Turkey
| | - Ecem Metin
- Research Center for Translational Medicine (KUTTAM), Koç University, Rumeli Feneri, 34450, Istanbul, Sariyer, Turkey.
| | | | - Sepideh Saghati
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Atay Vural
- Research Center for Translational Medicine (KUTTAM), Koç University, Rumeli Feneri, 34450, Istanbul, Sariyer, Turkey.
- Department of Neurology, School of Medicine, KoÒ« University, Istanbul 34450, Turkey
| | - Göktuğ Akyoldaş
- Department of Neurosurgery, Koç University Hospital, Istanbul 34450, Turkey
| | - Kemal Baysal
- Research Center for Translational Medicine (KUTTAM), Koç University, Rumeli Feneri, 34450, Istanbul, Sariyer, Turkey.
- Department of Biochemistry, School of Medicine, Koç University, Istanbul 34450, Turkey
| | - Yusuf Yağcı
- Chemistry Department, Faculty of Science, İstanbul Technical University, İstanbul, Turkey
| | - Yasemin Gürsoy-Özdemir
- Research Center for Translational Medicine (KUTTAM), Koç University, Rumeli Feneri, 34450, Istanbul, Sariyer, Turkey.
- Department of Neurology, School of Medicine, KoÒ« University, Istanbul 34450, Turkey
| | - Savaş Taşoğlu
- Research Center for Translational Medicine (KUTTAM), Koç University, Rumeli Feneri, 34450, Istanbul, Sariyer, Turkey.
- Mechanical Engineering Department, School of Engineering, Koç University, Rumeli Feneri, 34450, Istanbul, Sariyer, Turkey
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Emel Sokullu
- Research Center for Translational Medicine (KUTTAM), Koç University, Rumeli Feneri, 34450, Istanbul, Sariyer, Turkey.
- Biophysics Department, Koç University School of Medicine, Rumeli Feneri, 34450, Istanbul, Sariyer, Turkey
| |
Collapse
|
16
|
Zhu Y, Hui Q, Zhang Z, Fu H, Qin Y, Zhao Q, Li Q, Zhang J, Guo L, He W, Han C. Advancements in the study of synaptic plasticity and mitochondrial autophagy relationship. J Neurosci Res 2024; 102:e25309. [PMID: 38400573 DOI: 10.1002/jnr.25309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024]
Abstract
Synapses serve as the points of communication between neurons, consisting primarily of three components: the presynaptic membrane, synaptic cleft, and postsynaptic membrane. They transmit signals through the release and reception of neurotransmitters. Synaptic plasticity, the ability of synapses to undergo structural and functional changes, is influenced by proteins such as growth-associated proteins, synaptic vesicle proteins, postsynaptic density proteins, and neurotrophic growth factors. Furthermore, maintaining synaptic plasticity consumes more than half of the brain's energy, with a significant portion of this energy originating from ATP generated through mitochondrial energy metabolism. Consequently, the quantity, distribution, transport, and function of mitochondria impact the stability of brain energy metabolism, thereby participating in the regulation of fundamental processes in synaptic plasticity, including neuronal differentiation, neurite outgrowth, synapse formation, and neurotransmitter release. This article provides a comprehensive overview of the proteins associated with presynaptic plasticity, postsynaptic plasticity, and common factors between the two, as well as the relationship between mitochondrial energy metabolism and synaptic plasticity.
Collapse
Affiliation(s)
- Yousong Zhu
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Qinlong Hui
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Zheng Zhang
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Hao Fu
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Yali Qin
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Qiong Zhao
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Qinqing Li
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Junlong Zhang
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
| | - Lei Guo
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
| | - Wenbin He
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
| | - Cheng Han
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| |
Collapse
|
17
|
Welsh JA, Goberdhan DCI, O'Driscoll L, Buzas EI, Blenkiron C, Bussolati B, Cai H, Di Vizio D, Driedonks TAP, Erdbrügger U, Falcon‐Perez JM, Fu Q, Hill AF, Lenassi M, Lim SK, Mahoney MG, Mohanty S, Möller A, Nieuwland R, Ochiya T, Sahoo S, Torrecilhas AC, Zheng L, Zijlstra A, Abuelreich S, Bagabas R, Bergese P, Bridges EM, Brucale M, Burger D, Carney RP, Cocucci E, Colombo F, Crescitelli R, Hanser E, Harris AL, Haughey NJ, Hendrix A, Ivanov AR, Jovanovic‐Talisman T, Kruh‐Garcia NA, Ku'ulei‐Lyn Faustino V, Kyburz D, Lässer C, Lennon KM, Lötvall J, Maddox AL, Martens‐Uzunova ES, Mizenko RR, Newman LA, Ridolfi A, Rohde E, Rojalin T, Rowland A, Saftics A, Sandau US, Saugstad JA, Shekari F, Swift S, Ter‐Ovanesyan D, Tosar JP, Useckaite Z, Valle F, Varga Z, van der Pol E, van Herwijnen MJC, Wauben MHM, Wehman AM, Williams S, Zendrini A, Zimmerman AJ, MISEV Consortium, Théry C, Witwer KW. Minimal information for studies of extracellular vesicles (MISEV2023): From basic to advanced approaches. J Extracell Vesicles 2024; 13:e12404. [PMID: 38326288 PMCID: PMC10850029 DOI: 10.1002/jev2.12404] [Citation(s) in RCA: 1148] [Impact Index Per Article: 1148.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 02/09/2024] Open
Abstract
Extracellular vesicles (EVs), through their complex cargo, can reflect the state of their cell of origin and change the functions and phenotypes of other cells. These features indicate strong biomarker and therapeutic potential and have generated broad interest, as evidenced by the steady year-on-year increase in the numbers of scientific publications about EVs. Important advances have been made in EV metrology and in understanding and applying EV biology. However, hurdles remain to realising the potential of EVs in domains ranging from basic biology to clinical applications due to challenges in EV nomenclature, separation from non-vesicular extracellular particles, characterisation and functional studies. To address the challenges and opportunities in this rapidly evolving field, the International Society for Extracellular Vesicles (ISEV) updates its 'Minimal Information for Studies of Extracellular Vesicles', which was first published in 2014 and then in 2018 as MISEV2014 and MISEV2018, respectively. The goal of the current document, MISEV2023, is to provide researchers with an updated snapshot of available approaches and their advantages and limitations for production, separation and characterisation of EVs from multiple sources, including cell culture, body fluids and solid tissues. In addition to presenting the latest state of the art in basic principles of EV research, this document also covers advanced techniques and approaches that are currently expanding the boundaries of the field. MISEV2023 also includes new sections on EV release and uptake and a brief discussion of in vivo approaches to study EVs. Compiling feedback from ISEV expert task forces and more than 1000 researchers, this document conveys the current state of EV research to facilitate robust scientific discoveries and move the field forward even more rapidly.
Collapse
Affiliation(s)
- Joshua A. Welsh
- Translational Nanobiology Section, Laboratory of PathologyNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Deborah C. I. Goberdhan
- Nuffield Department of Women's and Reproductive HealthUniversity of Oxford, Women's Centre, John Radcliffe HospitalOxfordUK
| | - Lorraine O'Driscoll
- School of Pharmacy and Pharmaceutical SciencesTrinity College DublinDublinIreland
- Trinity Biomedical Sciences InstituteTrinity College DublinDublinIreland
- Trinity St. James's Cancer InstituteTrinity College DublinDublinIreland
| | - Edit I. Buzas
- Department of Genetics, Cell‐ and ImmunobiologySemmelweis UniversityBudapestHungary
- HCEMM‐SU Extracellular Vesicle Research GroupSemmelweis UniversityBudapestHungary
- HUN‐REN‐SU Translational Extracellular Vesicle Research GroupSemmelweis UniversityBudapestHungary
| | - Cherie Blenkiron
- Faculty of Medical and Health SciencesThe University of AucklandAucklandNew Zealand
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health SciencesUniversity of TurinTurinItaly
| | | | - Dolores Di Vizio
- Department of Surgery, Division of Cancer Biology and TherapeuticsCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Tom A. P. Driedonks
- Department CDL ResearchUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Uta Erdbrügger
- University of Virginia Health SystemCharlottesvilleVirginiaUSA
| | - Juan M. Falcon‐Perez
- Exosomes Laboratory, Center for Cooperative Research in BiosciencesBasque Research and Technology AllianceDerioSpain
- Metabolomics Platform, Center for Cooperative Research in BiosciencesBasque Research and Technology AllianceDerioSpain
- IKERBASQUE, Basque Foundation for ScienceBilbaoSpain
| | - Qing‐Ling Fu
- Otorhinolaryngology Hospital, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Extracellular Vesicle Research and Clinical Translational CenterThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Andrew F. Hill
- Institute for Health and SportVictoria UniversityMelbourneAustralia
| | - Metka Lenassi
- Faculty of MedicineUniversity of LjubljanaLjubljanaSlovenia
| | - Sai Kiang Lim
- Institute of Molecular and Cell Biology (IMCB)Agency for Science, Technology and Research (A*STAR)SingaporeSingapore
- Paracrine Therapeutics Pte. Ltd.SingaporeSingapore
- Department of Surgery, YLL School of MedicineNational University SingaporeSingaporeSingapore
| | - Mỹ G. Mahoney
- Thomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Sujata Mohanty
- Stem Cell FacilityAll India Institute of Medical SciencesNew DelhiIndia
| | - Andreas Möller
- Chinese University of Hong KongHong KongHong Kong S.A.R.
- QIMR Berghofer Medical Research InstituteBrisbaneAustralia
| | - Rienk Nieuwland
- Laboratory of Experimental Clinical Chemistry, Amsterdam University Medical Centers, Location AMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Vesicle Center, Amsterdam University Medical Centers, Location AMCUniversity of AmsterdamAmsterdamThe Netherlands
| | | | - Susmita Sahoo
- Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Ana C. Torrecilhas
- Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, Departamento de Ciências Farmacêuticas, Instituto de Ciências Ambientais, Químicas e FarmacêuticasUniversidade Federal de São Paulo (UNIFESP) Campus DiademaDiademaBrazil
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Andries Zijlstra
- Department of PathologyVanderbilt University Medical CenterNashvilleTennesseeUSA
- GenentechSouth San FranciscoCaliforniaUSA
| | - Sarah Abuelreich
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Reem Bagabas
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Paolo Bergese
- Department of Molecular and Translational MedicineUniversity of BresciaBresciaItaly
- Center for Colloid and Surface Science (CSGI)FlorenceItaly
- National Center for Gene Therapy and Drugs based on RNA TechnologyPaduaItaly
| | - Esther M. Bridges
- Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordUK
| | - Marco Brucale
- Consiglio Nazionale delle Ricerche ‐ Istituto per lo Studio dei Materiali NanostrutturatiBolognaItaly
- Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande InterfaseFlorenceItaly
| | - Dylan Burger
- Kidney Research CentreOttawa Hopsital Research InstituteOttawaCanada
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaCanada
- School of Pharmaceutical SciencesUniversity of OttawaOttawaCanada
| | - Randy P. Carney
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
| | - Emanuele Cocucci
- Division of Pharmaceutics and Pharmacology, College of PharmacyThe Ohio State UniversityColumbusOhioUSA
- Comprehensive Cancer CenterThe Ohio State UniversityColumbusOhioUSA
| | - Federico Colombo
- Division of Pharmaceutics and Pharmacology, College of PharmacyThe Ohio State UniversityColumbusOhioUSA
| | - Rossella Crescitelli
- Sahlgrenska Center for Cancer Research, Department of Surgery, Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Edveena Hanser
- Department of BiomedicineUniversity Hospital BaselBaselSwitzerland
- Department of BiomedicineUniversity of BaselBaselSwitzerland
| | | | - Norman J. Haughey
- Departments of Neurology and PsychiatryJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - An Hendrix
- Laboratory of Experimental Cancer Research, Department of Human Structure and RepairGhent UniversityGhentBelgium
- Cancer Research Institute GhentGhentBelgium
| | - Alexander R. Ivanov
- Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical BiologyNortheastern UniversityBostonMassachusettsUSA
| | - Tijana Jovanovic‐Talisman
- Department of Cancer Biology and Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Nicole A. Kruh‐Garcia
- Bio‐pharmaceutical Manufacturing and Academic Resource Center (BioMARC)Infectious Disease Research Center, Colorado State UniversityFort CollinsColoradoUSA
| | - Vroniqa Ku'ulei‐Lyn Faustino
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Diego Kyburz
- Department of BiomedicineUniversity of BaselBaselSwitzerland
- Department of RheumatologyUniversity Hospital BaselBaselSwitzerland
| | - Cecilia Lässer
- Krefting Research Centre, Department of Internal Medicine and Clinical NutritionInstitute of Medicine at Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Kathleen M. Lennon
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Jan Lötvall
- Krefting Research Centre, Institute of Medicine at Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Adam L. Maddox
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Elena S. Martens‐Uzunova
- Erasmus MC Cancer InstituteUniversity Medical Center Rotterdam, Department of UrologyRotterdamThe Netherlands
| | - Rachel R. Mizenko
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
| | - Lauren A. Newman
- College of Medicine and Public HealthFlinders UniversityAdelaideAustralia
| | - Andrea Ridolfi
- Department of Physics and Astronomy, and LaserLaB AmsterdamVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Eva Rohde
- Department of Transfusion Medicine, University HospitalSalzburger Landeskliniken GmbH of Paracelsus Medical UniversitySalzburgAustria
- GMP Unit, Paracelsus Medical UniversitySalzburgAustria
- Transfer Centre for Extracellular Vesicle Theralytic Technologies, EV‐TTSalzburgAustria
| | - Tatu Rojalin
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
- Expansion Therapeutics, Structural Biology and BiophysicsJupiterFloridaUSA
| | - Andrew Rowland
- College of Medicine and Public HealthFlinders UniversityAdelaideAustralia
| | - Andras Saftics
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Ursula S. Sandau
- Department of Anesthesiology & Perioperative MedicineOregon Health & Science UniversityPortlandOregonUSA
| | - Julie A. Saugstad
- Department of Anesthesiology & Perioperative MedicineOregon Health & Science UniversityPortlandOregonUSA
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
- Celer DiagnosticsTorontoCanada
| | - Simon Swift
- Waipapa Taumata Rau University of AucklandAucklandNew Zealand
| | - Dmitry Ter‐Ovanesyan
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMassachusettsUSA
| | - Juan P. Tosar
- Universidad de la RepúblicaMontevideoUruguay
- Institut Pasteur de MontevideoMontevideoUruguay
| | - Zivile Useckaite
- College of Medicine and Public HealthFlinders UniversityAdelaideAustralia
| | - Francesco Valle
- Consiglio Nazionale delle Ricerche ‐ Istituto per lo Studio dei Materiali NanostrutturatiBolognaItaly
- Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande InterfaseFlorenceItaly
| | - Zoltan Varga
- Biological Nanochemistry Research GroupInstitute of Materials and Environmental Chemistry, Research Centre for Natural SciencesBudapestHungary
- Department of Biophysics and Radiation BiologySemmelweis UniversityBudapestHungary
| | - Edwin van der Pol
- Amsterdam Vesicle Center, Amsterdam University Medical Centers, Location AMCUniversity of AmsterdamAmsterdamThe Netherlands
- Biomedical Engineering and Physics, Amsterdam UMC, location AMCUniversity of AmsterdamAmsterdamThe Netherlands
- Laboratory of Experimental Clinical Chemistry, Amsterdam UMC, location AMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Martijn J. C. van Herwijnen
- Department of Biomolecular Health Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Marca H. M. Wauben
- Department of Biomolecular Health Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | | | | | - Andrea Zendrini
- Department of Molecular and Translational MedicineUniversity of BresciaBresciaItaly
- Center for Colloid and Surface Science (CSGI)FlorenceItaly
| | - Alan J. Zimmerman
- Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical BiologyNortheastern UniversityBostonMassachusettsUSA
| | | | - Clotilde Théry
- Institut Curie, INSERM U932PSL UniversityParisFrance
- CurieCoreTech Extracellular Vesicles, Institut CurieParisFrance
| | - Kenneth W. Witwer
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- EV Core Facility “EXCEL”, Institute for Basic Biomedical SciencesJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's DiseaseJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
18
|
Chon NL, Tran S, Miller CS, Lin H, Knight JD. A conserved electrostatic membrane-binding surface in synaptotagmin-like proteins revealed using molecular phylogenetic analysis and homology modeling. Protein Sci 2024; 33:e4850. [PMID: 38038838 PMCID: PMC10731544 DOI: 10.1002/pro.4850] [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: 07/12/2023] [Revised: 10/29/2023] [Accepted: 11/28/2023] [Indexed: 12/02/2023]
Abstract
Protein structure prediction has emerged as a core technology for understanding biomolecules and their interactions. Here, we combine homology-based structure prediction with molecular phylogenetic analysis to study the evolution of electrostatic membrane binding among the vertebrate synaptotagmin-like protein (Slp) family. Slp family proteins play key roles in the membrane trafficking of large dense-core secretory vesicles. Our previous experimental and computational study found that the C2A domain of Slp-4 (also called granuphilin) binds with high affinity to anionic phospholipids in the cytoplasmic leaflet of the plasma membrane through a large positively charged protein surface centered on a cluster of phosphoinositide-binding lysine residues. Because this surface contributes greatly to Slp-4 C2A domain membrane binding, we hypothesized that the net charge on the surface might be evolutionarily conserved. To test this hypothesis, the known C2A sequences of Slp-4 among vertebrates were organized by class (from mammalia to pisces) using molecular phylogenetic analysis. Consensus sequences for each class were then identified and used to generate homology structures, from which Poisson-Boltzmann electrostatic potentials were calculated. For comparison, homology structures and electrostatic potentials were also calculated for the five human Slp protein family members. The results demonstrate that the charge on the membrane-binding surface is highly conserved throughout the evolution of Slp-4, and more highly conserved than many individual residues among the human Slp family paralogs. Such molecular phylogenetic-driven computational analysis can help to describe the evolution of electrostatic interactions between proteins and membranes which are crucial for their function.
Collapse
Affiliation(s)
- Nara L. Chon
- Department of ChemistryUniversity of Colorado DenverDenverColoradoUSA
| | - Sherleen Tran
- Department of ChemistryUniversity of Colorado DenverDenverColoradoUSA
| | | | - Hai Lin
- Department of ChemistryUniversity of Colorado DenverDenverColoradoUSA
| | | |
Collapse
|
19
|
Menaceur C, Dusailly O, Gosselet F, Fenart L, Saint-Pol J. Vesicular Trafficking, a Mechanism Controlled by Cascade Activation of Rab Proteins: Focus on Rab27. BIOLOGY 2023; 12:1530. [PMID: 38132356 PMCID: PMC10740503 DOI: 10.3390/biology12121530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Abstract
Vesicular trafficking is essential for the cell to internalize useful proteins and soluble substances, for cell signaling or for the degradation of pathogenic elements such as bacteria or viruses. This vesicular trafficking also enables the cell to engage in secretory processes for the elimination of waste products or for the emission of intercellular communication vectors such as cytokines, chemokines and extracellular vesicles. Ras-related proteins (Rab) and their effector(s) are of crucial importance in all of these processes, and mutations/alterations to them have serious pathophysiological consequences. This review presents a non-exhaustive overview of the role of the major Rab involved in vesicular trafficking, with particular emphasis on their involvement in the biogenesis and secretion of extracellular vesicles, and on the role of Rab27 in various pathophysiological processes. Therefore, Rab and their effector(s) are central therapeutic targets, given their involvement in vesicular trafficking and their importance for cell physiology.
Collapse
Affiliation(s)
| | | | | | | | - Julien Saint-Pol
- Univ. Artois, UR 2465, Blood-Brain Barrier Laboratory (LBHE), F-62300 Lens, France; (C.M.); (O.D.); (F.G.); (L.F.)
| |
Collapse
|
20
|
Cay E, Sezer A, Karakulak V, Serbes M, Ozcan D, Bisgin A, Aygunes U, Sasmaz HI, Yucel SP, Toyran T, Altintas DU. Hemophagocytic lymphohistiocytosis in children with Griscelli syndrome type 2: genetics, laboratory findings and treatment. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL IMMUNOLOGY 2023; 12:140-152. [PMID: 38187365 PMCID: PMC10767195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 12/03/2023] [Indexed: 01/09/2024]
Abstract
Griscelli syndrome is a rare inherited autosomal recessive syndrome that causes immunodeficiency. Hemophagocytic lymphohistiocytosis (HLH), which is characterized by a high mortality rate, may develop because of Griscelli syndrome type 2 (GS2). We aimed to share our experience with the diagnosis and treatment methods of patients who developed HLH secondary to GS2. Patients with GS2 who were diagnosed and treated for HLH between 2017 and 2022 at the Cukurova University Division of Pediatric Allergy & Immunology and Division of Pediatric Hematology were included in the study. Microscopic examination of the hair shaft and next-generation sequencing for molecular genetic testing of RAB27A helped in the diagnosis of GS2. The first clinical presentation of 8 patients was HLH. One patient presented with CNS involvement and two patients presented with recurrent fever. Over 5 years, GS2 was diagnosed in 15 patients, of whom 11 (73.3%) developed HLH. The HLH-2004 protocol was used to treat these patients. Hematopoietic stem cell transplantation (HSCT) was performed in five patients who were matched with suitable donors. While all patients who underwent HSCT were alive, three patients who could not undergo HSCT because no donor could be found died. Deletion of CAAGC at nucleotides 514_518 in GS2 patients is associated with CNS involvement and a poor prognosis. HLH may be the first sign of presentation in patients with GS2. Although further research is needed, regardless of the conditioning regimen utilized, early HSCT remains the primary therapy option for preventing GS2-induced mortality in HLH.
Collapse
Affiliation(s)
- Ezgi Cay
- Department of Pediatrics, Faculty of Medicine, Balcali Hospital, Cukurova UniversityAdana, Turkey
| | - Ahmet Sezer
- Division of Pediatric Allergy and Immunology, Faculty of Medicine, Balcali Hospital, Cukurova UniversityAdana, Turkey
| | - Veysel Karakulak
- Division of Pediatric Allergy and Immunology, Faculty of Medicine, Balcali Hospital, Cukurova UniversityAdana, Turkey
| | - Mahir Serbes
- Division of Pediatric Allergy and Immunology, Faculty of Medicine, Balcali Hospital, Cukurova UniversityAdana, Turkey
| | - Dilek Ozcan
- Division of Pediatric Allergy and Immunology, Faculty of Medicine, Balcali Hospital, Cukurova UniversityAdana, Turkey
| | - Atil Bisgin
- Cukurova University, AGENTEM (Adana Genetic Diseases Diagnosis and Treatment Center)Adana, Turkey
- Department of Medical Genetics, Faculty of Medicine, Balcali Hospital, Cukurova UniversityAdana, Turkey
| | - Utku Aygunes
- Division of Pediatric Hematology, Oncology and BMT Unit, Acibadem Adana HospitalAdana, Turkey
| | - H Ilgen Sasmaz
- Division of Pediatric Hematology, Faculty of Medicine, Balcali Hospital, Cukurova UniversityAdana, Turkey
| | - Sevinc P Yucel
- Department of Biostatistics, Faculty of Medicine, Balcali Hospital, Cukurova UniversityAdana, Turkey
| | - Tugba Toyran
- Department of Pathology, Faculty of Medicine, Balcali Hospital, Cukurova UniversityAdana, Turkey
| | - Derya U Altintas
- Division of Pediatric Allergy and Immunology, Faculty of Medicine, Balcali Hospital, Cukurova UniversityAdana, Turkey
| |
Collapse
|
21
|
Chon NL, Tran S, Miller CS, Lin H, Knight JD. A Conserved Electrostatic Membrane-Binding Surface in Synaptotagmin-Like Proteins Revealed Using Molecular Phylogenetic Analysis and Homology Modeling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.13.548768. [PMID: 37502952 PMCID: PMC10369986 DOI: 10.1101/2023.07.13.548768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Protein structure prediction has emerged as a core technology for understanding biomolecules and their interactions. Here, we combine homology-based structure prediction with molecular phylogenetic analysis to study the evolution of electrostatic membrane binding among vertebrate synaptotagmin-like proteins (Slps). Slp family proteins play key roles in the membrane trafficking of large dense-core secretory vesicles. Our previous experimental and computational study found that the C2A domain of Slp-4 (also called granuphilin) binds with high affinity to anionic phospholipids in the cytoplasmic leaflet of the plasma membrane through a large positively charged protein surface centered on a cluster of phosphoinositide-binding lysine residues. Because this surface contributes greatly to Slp-4 C2A domain membrane binding, we hypothesized that the net charge on the surface might be evolutionarily conserved. To test this hypothesis, the known C2A sequences of Slp-4 among vertebrates were organized by class (from mammalia to pisces) using molecular phylogenetic analysis. Consensus sequences for each class were then identified and used to generate homology structures, from which Poisson-Boltzmann electrostatic potentials were calculated. For comparison, homology structures and electrostatic potentials were also calculated for the five human Slp protein family members. The results demonstrate that the charge on the membrane-binding surface is highly conserved throughout the evolution of Slp-4, and more highly conserved than many individual residues among the human Slp family paralogs. Such molecular phylogenetic-driven computational analysis can help to describe the evolution of electrostatic interactions between proteins and membranes which are crucial for their function.
Collapse
Affiliation(s)
- Nara L. Chon
- Department of Chemistry, University of Colorado Denver
| | - Sherleen Tran
- Department of Chemistry, University of Colorado Denver
| | | | - Hai Lin
- Department of Chemistry, University of Colorado Denver
| | | |
Collapse
|
22
|
Heckle LA, Kozminski KG. Osh-dependent and -independent Regulation of PI4P Levels During Polarized Growth of Saccharomyces cerevisiae. Mol Biol Cell 2023; 34:ar104. [PMID: 37556206 PMCID: PMC10559303 DOI: 10.1091/mbc.e23-03-0089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 07/03/2023] [Accepted: 07/31/2023] [Indexed: 08/11/2023] Open
Abstract
Polarized secretion facilitates polarized cell growth. For a secretory vesicle to dock at the plasma membrane, it must mature with a progressive association or dissociation of molecules that are, respectively, necessary for or inhibitory to vesicle docking, including an exchange of Rab GTPases. In current models, oxysterol-binding protein homologue 4 (Osh4p) establishes a phosphatidylinositol 4-phosphate (PI4P) gradient along the secretory trafficking pathway such that vesicles have higher PI4P levels after budding from the trans-Golgi relative to when vesicles arrive at the plasma membrane. In this study, using the lipid-binding domain P4M and live-cell imaging, we show that secretory vesicle-associated PI4P levels remain constant when vesicles traffic from the trans-Golgi to the plasma membrane. We also show that deletion of OSH4 does not alter vesicle-associated PI4P levels, though loss of any individual member of the OSH family or complete loss of OSH family function alters the intracellular distribution of PI4P. We propose a model in which the Rab GTPases Ypt32p and Sec4p remain associated with a secretory vesicle during trafficking, independent of PI4P levels and Osh4p. Together these data indicate the necessity of experiments revealing the location and timing of events required for vesicle maturation.
Collapse
Affiliation(s)
- Lindsay A. Heckle
- Department of Biology, University of Virginia, Charlottesville, VA 22904
| | - Keith G. Kozminski
- Department of Biology, University of Virginia, Charlottesville, VA 22904
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908
| |
Collapse
|
23
|
Izumi T. Multiple pathways and independent functional pools in insulin granule exocytosis. Genes Cells 2023; 28:471-481. [PMID: 37070774 PMCID: PMC11448364 DOI: 10.1111/gtc.13029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/04/2023] [Accepted: 04/05/2023] [Indexed: 04/19/2023]
Abstract
In contrast to synaptic vesicle exocytosis, secretory granule exocytosis follows a much longer time course, and thus allows for different prefusion states prior to stimulation. Indeed, total internal reflection fluorescence microscopy in living pancreatic β cells reveals that, prior to stimulation, either visible or invisible granules fuse in parallel during both early (first) and late (second) phases after glucose stimulation. Therefore, fusion occurs not only from granules predocked to the plasma membrane but also from those translocated from the cell interior during ongoing stimulation. Recent findings suggest that such heterogeneous exocytosis is conducted by a specific set of multiple Rab27 effectors that appear to operate on the same granule; namely, exophilin-8, granuphilin, and melanophilin play differential roles in distinct secretory pathways to final fusion. Furthermore, the exocyst, which is known to tether secretory vesicles to the plasma membrane in constitutive exocytosis, cooperatively functions with these Rab27 effectors in regulated exocytosis. In this review, the basic nature of insulin granule exocytosis will be described as a representative example of secretory granule exocytosis, followed by a discussion of the means by which different Rab27 effectors and the exocyst coordinate to regulate the entire exocytic processes in β cells.
Collapse
Affiliation(s)
- Tetsuro Izumi
- Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| |
Collapse
|
24
|
Zhao K, Matsunaga K, Mizuno K, Wang H, Okunishi K, Izumi T. Functional hierarchy among different Rab27 effectors involved in secretory granule exocytosis. eLife 2023; 12:82821. [PMID: 36803984 PMCID: PMC9988257 DOI: 10.7554/elife.82821] [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: 08/18/2022] [Accepted: 02/21/2023] [Indexed: 02/23/2023] Open
Abstract
The Rab27 effectors are known to play versatile roles in regulated exocytosis. In pancreatic beta cells, exophilin-8 anchors granules in the peripheral actin cortex, whereas granuphilin and melanophilin mediate granule fusion with and without stable docking to the plasma membrane, respectively. However, it is unknown whether these coexisting effectors function in parallel or in sequence to support the whole insulin secretory process. Here, we investigate their functional relationships by comparing the exocytic phenotypes in mouse beta cells simultaneously lacking two effectors with those lacking just one of them. Analyses of prefusion profiles by total internal reflection fluorescence microscopy suggest that melanophilin exclusively functions downstream of exophilin-8 to mobilize granules for fusion from the actin network to the plasma membrane after stimulation. The two effectors are physically linked via the exocyst complex. Downregulation of the exocyst component affects granule exocytosis only in the presence of exophilin-8. The exocyst and exophilin-8 also promote fusion of granules residing beneath the plasma membrane prior to stimulation, although they differentially act on freely diffusible granules and those stably docked to the plasma membrane by granuphilin, respectively. This is the first study to diagram the multiple intracellular pathways of granule exocytosis and the functional hierarchy among different Rab27 effectors within the same cell.
Collapse
Affiliation(s)
- Kunli Zhao
- Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma UniversityMaebashiJapan
| | - Kohichi Matsunaga
- Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma UniversityMaebashiJapan
| | - Kouichi Mizuno
- Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma UniversityMaebashiJapan
| | - Hao Wang
- Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma UniversityMaebashiJapan
| | - Katsuhide Okunishi
- Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma UniversityMaebashiJapan
| | - Tetsuro Izumi
- Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma UniversityMaebashiJapan
| |
Collapse
|
25
|
Mizuno K, Izumi T. Munc13b stimulus-dependently accumulates on granuphilin-mediated, docked granules prior to fusion. Cell Struct Funct 2022; 47:31-41. [PMID: 35387942 PMCID: PMC10511056 DOI: 10.1247/csf.22005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/30/2022] [Indexed: 11/11/2022] Open
Abstract
The Rab27 effector granuphilin plays an indispensable role in stable docking of secretory granules to the plasma membrane by interacting with the complex of Munc18-1 and the fusion-incompetent, closed form of syntaxins-1~3. Although this process prevents spontaneous granule exocytosis, those docked granules actively fuse in parallel with other undocked granules after stimulation. Therefore, it is postulated that the closed form of syntaxins must be converted into the fusion-competent open form in a stimulus-dependent manner. Although Munc13 family proteins are generally thought to prime docked vesicles by facilitating conformational change in syntaxins, it is unknown which isoform acts in granuphilin-mediated, docked granule exocytosis. In the present study, we show that, although both Munc13a and Munc13b are expressed in mouse pancreatic islets and their beta-cell line MIN6, the silencing of Munc13b, but not that of Munc13a, severely affects glucose-induced insulin secretion. Furthermore, Munc13b accumulates on a subset of granules beneath the plasma membrane just prior to fusion during stimulation, whereas Munc13a is translocated to the plasma membrane where granules do not exist. When fluorescently labeled granuphilin was introduced to discriminate between molecularly docked granules and other undocked granules in living cells, Munc13b downregulation was observed to preferentially decrease the fusion of granuphilin-positive granules immobilized to the plasma membrane. These findings suggest that Munc13b promotes insulin exocytosis by clustering on molecularly docked granules in a stimulus-dependent manner.Key words: docking, insulin, live cell imaging, priming, TIRF microscopy.
Collapse
Affiliation(s)
- Kouichi Mizuno
- Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi 371-8512, Japan
| | - Tetsuro Izumi
- Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi 371-8512, Japan
| |
Collapse
|