1
|
Amini H, Namjoo AR, Narmi MT, Mardi N, Narimani S, Naturi O, Khosrowshahi ND, Rahbarghazi R, Saghebasl S, Hashemzadeh S, Nouri M. Exosome-bearing hydrogels and cardiac tissue regeneration. Biomater Res 2023; 27:99. [PMID: 37803483 PMCID: PMC10559618 DOI: 10.1186/s40824-023-00433-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 09/18/2023] [Indexed: 10/08/2023] Open
Abstract
BACKGROUND In recent years, cardiovascular disease in particular myocardial infarction (MI) has become the predominant cause of human disability and mortality in the clinical setting. The restricted capacity of adult cardiomyocytes to proliferate and restore the function of infarcted sites is a challenging issue after the occurrence of MI. The application of stem cells and byproducts such as exosomes (Exos) has paved the way for the alleviation of cardiac tissue injury along with conventional medications in clinics. However, the short lifespan and activation of alloreactive immune cells in response to Exos and stem cells are the main issues in patients with MI. Therefore, there is an urgent demand to develop therapeutic approaches with minimum invasion for the restoration of cardiac function. MAIN BODY Here, we focused on recent data associated with the application of Exo-loaded hydrogels in ischemic cardiac tissue. Whether and how the advances in tissue engineering modalities have increased the efficiency of whole-based and byproducts (Exos) therapies under ischemic conditions. The integration of nanotechnology and nanobiology for designing novel smart biomaterials with therapeutic outcomes was highlighted. CONCLUSION Hydrogels can provide suitable platforms for the transfer of Exos, small molecules, drugs, and other bioactive factors for direct injection into the damaged myocardium. Future studies should focus on the improvement of physicochemical properties of Exo-bearing hydrogel to translate for the standard treatment options.
Collapse
Affiliation(s)
- Hassan Amini
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of General and Vascular Surgery, Tabriz University of Medical Sciences, Tabriz, 51548/53431, Iran
| | - Atieh Rezaei Namjoo
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Taghavi Narmi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Narges Mardi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samaneh Narimani
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ozra Naturi
- Department of Organic and Biochemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran
| | - Nafiseh Didar Khosrowshahi
- Stem Cell and Tissue Engineering Research Laboratory, Sahand University of Technology, Tabriz, 51335-1996, Iran
| | - 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, 51548/53431, Iran.
| | - Solmaz Saghebasl
- 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, 51548/53431, Iran.
| | - Shahriar Hashemzadeh
- Department of General and Vascular Surgery, Tabriz University of Medical Sciences, Tabriz, 51548/53431, Iran.
| | - Mohammad Nouri
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
2
|
Feng L, Lo H, Hong Z, Zheng J, Yan Y, Ye Z, Chen X, Pan X. Microglial LRRK2-mediated NFATc1 attenuates α-synuclein immunotoxicity in association with CX3CR1-induced migration and the lysosome-initiated degradation. Glia 2023; 71:2266-2284. [PMID: 37300531 DOI: 10.1002/glia.24422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/22/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023]
Abstract
Synucleinopathies refer to a range of neurodegenerative diseases caused by abnormal α-synuclein (α-Syn) deposition, including Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA). Their pathogenesis is strongly linked to microglial dysfunction and neuroinflammation, which involves the leucine-rich-repeat kinase 2 (LRRK2)-regulated nuclear factor of activated T-cells (NFAT). Of the NFAT family, NFATc1 has been found to be increasingly translocated into the nucleus in α-syn stimulation. However, the specific role of NFATc1-mediated intracellular signaling in PD remains elusive in regulating microglial functions. In the current study, we crossbred LRRK2 or NFATc1 conditional knockout mice with Lyz2Cre mice to generate mice with microglia-specific deletion of LRRK2 or NFATc1, and by stereotactic injection of fibrillary α-Syn, we generated PD models in these mice. We found that LRRK2 deficiency enhanced microglial phagocytosis in the mice after α-Syn exposure and that genetic inhibition of NFATc1 markedly diminished phagocytosis and α-Syn elimination. We further demonstrated that LRRK2 negatively regulated NFATc1 in α-Syn-treated microglia, in which microglial LRRK2-deficiency facilitated NFATc1 nuclear translocation, CX3CR1 upregulation, and microglia migration. Additionally, NFATc1 translocation upregulated the expression of Rab7 and promoted the formation of late lysosomes, resulting in α-Syn degradation. In contrast, the microglial NFATc1 deficiency impaired CX3CR1 upregulation and the formation of Rab7-mediated late lysosomes. These findings highlight the critical role of NFATc1 in modulating microglial migration and phagocytosis, in which the LRRK2-NFATc1 signaling pathway regulates the expression of microglial CX3CR1 and endocytic degradative Rab7 to attenuate α-synuclein immunotoxicity.
Collapse
Affiliation(s)
- Linjuan Feng
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Hsuan Lo
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhaoxiang Hong
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Neurology, The University of HongKong Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Jiahao Zheng
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Yuhong Yan
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Zucheng Ye
- Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, Fuzhou, China
| | - Xiaochun Chen
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Xiaodong Pan
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, Fuzhou, China
| |
Collapse
|
3
|
Smith M, Zhang L, Jin Y, Yang M, Bade A, Gillis KD, Jana S, Bypaneni RN, Glass TE, Lin H. A Turn-On Fluorescent Amino Acid Sensor Reveals Chloroquine's Effect on Cellular Amino Acids via Inhibiting Cathepsin L. ACS CENTRAL SCIENCE 2023; 9:980-991. [PMID: 37252359 PMCID: PMC10214525 DOI: 10.1021/acscentsci.2c01325] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Indexed: 05/31/2023]
Abstract
Maintaining homeostasis of metabolites such as amino acids is critical for cell survival. Dysfunction of nutrient balance can result in human diseases such as diabetes. Much remains to be discovered about how cells transport, store, and utilize amino acids due to limited research tools. Here we developed a novel, pan-amino acid fluorescent turn-on sensor, NS560. It detects 18 of the 20 proteogenic amino acids and can be visualized in mammalian cells. Using NS560, we identified amino acids pools in lysosomes, late endosomes, and surrounding the rough endoplasmic reticulum. Interestingly, we observed amino acid accumulation in large cellular foci after treatment with chloroquine, but not with other autophagy inhibitors. Using a biotinylated photo-cross-linking chloroquine analog and chemical proteomics, we identified Cathepsin L (CTSL) as the chloroquine target leading to the amino acid accumulation phenotype. This study establishes NS560 as a useful tool to study amino acid regulation, identifies new mechanisms of action of chloroquine, and demonstrates the importance of CTSL regulation of lysosomes.
Collapse
Affiliation(s)
- Michael
R. Smith
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York 14853, United States
| | - Le Zhang
- Department
of Chemistry, University of Missouri, Columbia, Missouri 65211, United States
| | - Yizhen Jin
- Graduate
Program of Biochemistry, Molecular and Cell Biology, Department of
Chemistry and Chemical Biology, Cornell
University, Ithaca, New York 14853, United
States
| | - Min Yang
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York 14853, United States
| | - Anusha Bade
- Department
of Chemistry, University of Missouri, Columbia, Missouri 65211, United States
| | - Kevin D. Gillis
- Dalton
Cardiovascular Research Center, Department of Bioengineering and Department
of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri 65211, United States
| | - Sadhan Jana
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York 14853, United States
| | - Ramesh Naidu Bypaneni
- Department
of Chemistry, University of Missouri, Columbia, Missouri 65211, United States
| | - Timothy E. Glass
- Department
of Chemistry, University of Missouri, Columbia, Missouri 65211, United States
| | - Hening Lin
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York 14853, United States
- Howard
Hughes Medical Institute, Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
4
|
Tyagi S, Sarveswaran N, Higerd-Rusli GP, Liu S, Dib-Hajj FB, Waxman SG, Dib-Hajj SD. Conserved but not critical: Trafficking and function of NaV1.7 are independent of highly conserved polybasic motifs. Front Mol Neurosci 2023; 16:1161028. [PMID: 37008789 PMCID: PMC10060856 DOI: 10.3389/fnmol.2023.1161028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/02/2023] [Indexed: 03/18/2023] Open
Abstract
Non-addictive treatment of chronic pain represents a major unmet clinical need. Peripheral voltage-gated sodium (NaV) channels are an attractive target for pain therapy because they initiate and propagate action potentials in primary afferents that detect and transduce noxious stimuli. NaV1.7 sets the gain on peripheral pain-signaling neurons and is the best validated peripheral ion channel involved in human pain, and previous work has shown that it is transported in vesicles in sensory axons which also carry Rab6a, a small GTPase known to be involved in vesicular packaging and axonal transport. Understanding the mechanism of the association between Rab6a and NaV1.7 could inform therapeutic modalities to decrease trafficking of NaV1.7 to the distal axonal membrane. Polybasic motifs (PBM) have been shown to regulate Rab-protein interactions in a variety of contexts. In this study, we explored whether two PBMs in the cytoplasmic loop that joins domains I and II of human NaV1.7 were responsible for association with Rab6a and regulate axonal trafficking of the channel. Using site-directed mutagenesis we generated NaV1.7 constructs with alanine substitutions in the two PBMs. Voltage-clamp recordings showed that the constructs retain wild-type like gating properties. Optical Pulse-chase Axonal Long-distance (OPAL) imaging in live sensory axons shows that mutations of these PBMs do not affect co-trafficking of Rab6a and NaV1.7, or the accumulation of the channel at the distal axonal surface. Thus, these polybasic motifs are not required for interaction of NaV1.7 with the Rab6a GTPase, or for trafficking of the channel to the plasma membrane.
Collapse
Affiliation(s)
- Sidharth Tyagi
- Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, United States
- Center for Neuroscience and Regeneration Research, West Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Nivedita Sarveswaran
- Center for Neuroscience and Regeneration Research, West Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Grant P. Higerd-Rusli
- Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, United States
- Center for Neuroscience and Regeneration Research, West Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Shujun Liu
- Center for Neuroscience and Regeneration Research, West Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Fadia B. Dib-Hajj
- Center for Neuroscience and Regeneration Research, West Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Stephen G. Waxman
- Center for Neuroscience and Regeneration Research, West Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, United States
- *Correspondence: Stephen G. Waxman,
| | - Sulayman D. Dib-Hajj
- Center for Neuroscience and Regeneration Research, West Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, United States
- Sulayman D. Dib-Hajj,
| |
Collapse
|
5
|
Haque MS, Emi Y, Sakaguchi M. A conserved WXXE motif is an apical delivery determinant of ABC transporter C subfamily isoforms. Cell Struct Funct 2023; 48:71-82. [PMID: 36696993 PMCID: PMC10721954 DOI: 10.1247/csf.22049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 07/15/2022] [Indexed: 01/25/2023] Open
Abstract
ATP-binding cassette transporter isoform C7 (ABCC7), also designated as cystic fibrosis transmembrane conductance regulator (CFTR), is exclusively targeted to the apical plasma membrane of polarized epithelial cells. Although the apical localization of ABCC7 in epithelia is crucial for the Cl- excretion into lumens, the mechanism regulating its apical localization is poorly understood. In the present study, an apical localization determinant was identified in the N-terminal 80-amino acid long cytoplasmic region of ABCC7 (NT80). In HepG2 cells, overexpression of NT80 significantly disturbed the apical expression of ABCC7 in a competitive manner, suggesting the presence of a sorting determinant in this region. Deletion analysis identified a potential sorting information within a 20-amino acid long peptide (aa 41-60) of NT80. Alanine scanning mutagenesis of this region in full-length ABCC7 further narrowed down the apical localization determinant to four amino acids, W57DRE60. This WDRE sequence was conserved among vertebrate ABCC7 orthologs. Site-directed mutagenesis showed that W57 and E60 were critical for the apical expression of ABCC7, confirming a novel apical sorting determinant of ABCC7. Furthermore, a WXXE motif (tryptophan and glutamic acid residues with two-amino acid spacing) was found to be conserved among the N-terminal regions of apically localized ABCC members with 12-TM configuration. The significance of the WXXE motif was demonstrated for proper trafficking of ABCC4 to the apical plasma membrane.Key words: apical plasma membrane, sorting, ATP-binding cassette transporter, CFTR, MRP4.
Collapse
Affiliation(s)
- Md Shajedul Haque
- Graduate School of Science, University of Hyogo, Harima Science Park City, Hyogo 678-1297, Japan
| | - Yoshikazu Emi
- Graduate School of Science, University of Hyogo, Harima Science Park City, Hyogo 678-1297, Japan
| | - Masao Sakaguchi
- Graduate School of Science, University of Hyogo, Harima Science Park City, Hyogo 678-1297, Japan
| |
Collapse
|
6
|
Frank D, Moussi CJ, Ulferts S, Lorenzen L, Schwan C, Grosse R. Vesicle-Associated Actin Assembly by Formins Promotes TGFβ-Induced ANGPTL4 Trafficking, Secretion and Cell Invasion. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204896. [PMID: 36691769 PMCID: PMC10037683 DOI: 10.1002/advs.202204896] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/01/2022] [Indexed: 06/17/2023]
Abstract
Vesicle trafficking has emerged as an important process driving tumor progression through various mechanisms. Transforming growth factor beta (TGFβ)-mediated secretion of Angiopoietin-like 4 (ANGPTL4) is important for cancer development. Here, Formin-like 2 (FMNL2) is identified to be necessary for ANGPTL4 trafficking and secretion in response to TGFβ. Protein kinase C (PKC)-dependent phosphorylation of FMNL2 downstream of TGFβ stimulation is required for cancer cell invasion as well as ANGPTL4 vesicle trafficking and secretion. Moreover, using super resolution microscopy, ANGPTL4 trafficking is actin-dependent with FMNL2 directly polymerizing actin at ANGPTL4-containing vesicles, which are associated with Rab8a and myosin Vb. This work uncovers a formin-controlled mechanism that transiently polymerizes actin directly at intracellular vesicles to facilitate their mobility. This mechanism may be important for the regulation of cancer cell metastasis and tumor progression.
Collapse
Affiliation(s)
- Dennis Frank
- Institute of Experimental and Clinical Pharmacology and ToxicologyMedical FacultyUniversity of Freiburg79104FreiburgGermany
| | - Christel Jessica Moussi
- Institute of Experimental and Clinical Pharmacology and ToxicologyMedical FacultyUniversity of Freiburg79104FreiburgGermany
- Deutsche Forschungsgemeinschaft Research Training GroupMembrane Plasticity in Tissue Development and RemodelingUniversity of Marburg35037MarburgGermany
| | - Svenja Ulferts
- Institute of Experimental and Clinical Pharmacology and ToxicologyMedical FacultyUniversity of Freiburg79104FreiburgGermany
| | - Lina Lorenzen
- Institute of Experimental and Clinical Pharmacology and ToxicologyMedical FacultyUniversity of Freiburg79104FreiburgGermany
| | - Carsten Schwan
- Institute of Experimental and Clinical Pharmacology and ToxicologyMedical FacultyUniversity of Freiburg79104FreiburgGermany
| | - Robert Grosse
- Institute of Experimental and Clinical Pharmacology and ToxicologyMedical FacultyUniversity of Freiburg79104FreiburgGermany
- Centre for Integrative Biological Signalling Studies – CIBSS79104FreiburgGermany
| |
Collapse
|
7
|
Moretto E, Miozzo F, Longatti A, Bonnet C, Coussen F, Jaudon F, Cingolani LA, Passafaro M. The tetraspanin TSPAN5 regulates AMPAR exocytosis by interacting with the AP4 complex. eLife 2023; 12:76425. [PMID: 36795458 PMCID: PMC9934860 DOI: 10.7554/elife.76425] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/25/2023] [Indexed: 02/17/2023] Open
Abstract
Intracellular trafficking of AMPA receptors is a tightly regulated process which involves several adaptor proteins, and is crucial for the activity of excitatory synapses both in basal conditions and during synaptic plasticity. We found that, in rat hippocampal neurons, an intracellular pool of the tetraspanin TSPAN5 promotes exocytosis of AMPA receptors without affecting their internalisation. TSPAN5 mediates this function by interacting with the adaptor protein complex AP4 and Stargazin and possibly using recycling endosomes as a delivery route. This work highlights TSPAN5 as a new adaptor regulating AMPA receptor trafficking.
Collapse
Affiliation(s)
- Edoardo Moretto
- Institute of Neuroscience, CNRVedano al LambroItaly,NeuroMI Milan Center for Neuroscience, University of Milano-BicoccaMilanItaly
| | | | | | - Caroline Bonnet
- University of Bordeaux, Interdisciplinary Institute for NeuroscienceBordeauxFrance
| | - Francoise Coussen
- University of Bordeaux, Interdisciplinary Institute for NeuroscienceBordeauxFrance
| | - Fanny Jaudon
- Department of Life Sciences, University of TriesteTriesteItaly,IRCCS Ospedale Policlinico San MartinoGenoaItaly
| | - Lorenzo A Cingolani
- Department of Life Sciences, University of TriesteTriesteItaly,Center for Synaptic Neuroscience and Technology (NSYN), Istituto Italiano di Tecnologia (IIT)GenoaItaly
| | - Maria Passafaro
- Institute of Neuroscience, CNRVedano al LambroItaly,NeuroMI Milan Center for Neuroscience, University of Milano-BicoccaMilanItaly
| |
Collapse
|
8
|
Enrich C, Lu A, Tebar F, Rentero C, Grewal T. Ca 2+ and Annexins - Emerging Players for Sensing and Transferring Cholesterol and Phosphoinositides via Membrane Contact Sites. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1422:393-438. [PMID: 36988890 DOI: 10.1007/978-3-031-21547-6_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Maintaining lipid composition diversity in membranes from different organelles is critical for numerous cellular processes. However, many lipids are synthesized in the endoplasmic reticulum (ER) and require delivery to other organelles. In this scenario, formation of membrane contact sites (MCS) between neighbouring organelles has emerged as a novel non-vesicular lipid transport mechanism. Dissecting the molecular composition of MCS identified phosphoinositides (PIs), cholesterol, scaffolding/tethering proteins as well as Ca2+ and Ca2+-binding proteins contributing to MCS functioning. Compelling evidence now exists for the shuttling of PIs and cholesterol across MCS, affecting their concentrations in distinct membrane domains and diverse roles in membrane trafficking. Phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) at the plasma membrane (PM) not only controls endo-/exocytic membrane dynamics but is also critical in autophagy. Cholesterol is highly concentrated at the PM and enriched in recycling endosomes and Golgi membranes. MCS-mediated cholesterol transfer is intensely researched, identifying MCS dysfunction or altered MCS partnerships to correlate with de-regulated cellular cholesterol homeostasis and pathologies. Annexins, a conserved family of Ca2+-dependent phospholipid binding proteins, contribute to tethering and untethering events at MCS. In this chapter, we will discuss how Ca2+ homeostasis and annexins in the endocytic compartment affect the sensing and transfer of cholesterol and PIs across MCS.
Collapse
Affiliation(s)
- Carlos Enrich
- Departament de Biomedicina, Unitat de Biologia Cel⋅lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
- Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain.
| | - Albert Lu
- Departament de Biomedicina, Unitat de Biologia Cel⋅lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Francesc Tebar
- Departament de Biomedicina, Unitat de Biologia Cel⋅lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Carles Rentero
- Departament de Biomedicina, Unitat de Biologia Cel⋅lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Thomas Grewal
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
9
|
Čada Š, Vondálová Blanářová O, Gömoryová K, Mikulová A, Bačovská P, Zezula N, Kumari Jadaun A, Janovská P, Plešingerová H, Bryja V. Role of casein kinase 1 in the amoeboid migration of B-cell leukemic and lymphoma cells: A quantitative live imaging in the confined environment. Front Cell Dev Biol 2022; 10:911966. [PMID: 36561363 PMCID: PMC9763939 DOI: 10.3389/fcell.2022.911966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 11/03/2022] [Indexed: 12/12/2022] Open
Abstract
The migratory properties of leukemic cells are commonly associated with their pathological potential and can significantly affect the disease progression. While the research in immunopathology mostly employed powerful indirect methods such as flow cytometry, these cells were rarely observed directly using live imaging microscopy. This is especially true for the malignant cells of the B-cell lineage, such as those originating from chronic lymphocytic leukemia (CLL) and mantle cell lymphoma (MCL). In this study, we employed open-source image analysis tools to automatically and quantitatively describe the amoeboid migration of four B-cell leukemic and lymphoma cell lines and primary CLL cells. To avoid the effect of the shear stress of the medium on these usually non-adherent cells, we have confined the cells using a modified under-agarose assay. Surprisingly, the behavior of tested cell lines differed substantially in terms of basal motility or response to chemokines and VCAM1 stimulation. Since casein kinase 1 (CK1) was reported as a regulator of B-cell migration and a promoter of CLL, we looked at the effects of CK1 inhibition in more detail. Migration analysis revealed that CK1 inhibition induced rapid negative effects on the migratory polarity of these cells, which was quantitatively and morphologically distinct from the effect of ROCK inhibition. We have set up an assay that visualizes endocytic vesicles in the uropod and facilitates morphological analysis. This assay hints that the effect of CK1 inhibition might be connected to defects in polarized intracellular transport. In summary, 1) we introduce and validate a pipeline for the imaging and quantitative assessment of the amoeboid migration of CLL/MCL cells, 2) we provide evidence that the assay is sensitive enough to mechanistically study migration defects identified by the transwell assay, and 3) we describe the polarity defects induced by inhibition or deletion of CK1ε.
Collapse
Affiliation(s)
- Štěpán Čada
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | | | - Kristína Gömoryová
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Antónia Mikulová
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Petra Bačovská
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Nikodém Zezula
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Alka Kumari Jadaun
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Pavlína Janovská
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Hana Plešingerová
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia,Department of Internal Medicine—Hematology and Oncology, University Hospital Brno, Brno, Czechia
| | - Vítězslav Bryja
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia,Department of Cytokinetics, Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czechia,*Correspondence: Vítězslav Bryja,
| |
Collapse
|
10
|
Keable R, Hu S, Pfundstein G, Kozlova I, Su F, Du X, Yang H, Gunnersen J, Schachner M, Leshchyns'ka I, Sytnyk V. The BACE1-generated C-terminal fragment of the neural cell adhesion molecule 2 (NCAM2) promotes BACE1 targeting to Rab11-positive endosomes. Cell Mol Life Sci 2022; 79:555. [PMID: 36251052 PMCID: PMC9576659 DOI: 10.1007/s00018-022-04575-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/16/2022] [Accepted: 09/27/2022] [Indexed: 11/08/2022]
Abstract
Beta-site amyloid precursor protein cleaving enzyme 1 (BACE1), also known as β-secretase, is an aspartic protease. The sorting of this enzyme into Rab11-positive recycling endosomes regulates the BACE1-mediated cleavage of its substrates, however, the mechanisms underlying this targeting remain poorly understood. The neural cell adhesion molecule 2 (NCAM2) is a substrate of BACE1. We show that BACE1 cleaves NCAM2 in cultured hippocampal neurons and NCAM2-transfected CHO cells. The C-terminal fragment of NCAM2 that comprises the intracellular domain and a small portion of NCAM2’s extracellular domain, associates with BACE1. This association is not affected in cells with inhibited endocytosis, indicating that the interaction of NCAM2 and BACE1 precedes the targeting of BACE1 from the cell surface to endosomes. In neurons and CHO cells, this fragment and BACE1 co-localize in Rab11-positive endosomes. Overexpression of full-length NCAM2 or a recombinant NCAM2 fragment containing the transmembrane and intracellular domains but lacking the extracellular domain leads to an increase in BACE1 levels in these organelles. In NCAM2-deficient neurons, the levels of BACE1 are increased at the cell surface and reduced in intracellular organelles. These effects are correlated with increased levels of the soluble extracellular domain of BACE1 in the brains of NCAM2-deficient mice, suggesting increased shedding of BACE1 from the cell surface. Of note, shedding of the extracellular domain of Sez6, a protein cleaved exclusively by BACE1, is reduced in NCAM2-deficient animals. These results indicate that the BACE1-generated fragment of NCAM2 regulates BACE1 activity by promoting the targeting of BACE1 to Rab11-positive endosomes.
Collapse
Affiliation(s)
- Ryan Keable
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Shangfeng Hu
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Grant Pfundstein
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Irina Kozlova
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Feifei Su
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Ximing Du
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Hongyuan Yang
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Jenny Gunnersen
- Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Melitta Schachner
- Department of Cell Biology and Neuroscience, Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ, 08554, USA
| | - Iryna Leshchyns'ka
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Vladimir Sytnyk
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, 2052, Australia.
| |
Collapse
|
11
|
Medjmedj A, Ngalle-Loth A, Clemençon R, Hamacek J, Pichon C, Perche F. In Cellulo and In Vivo Comparison of Cholesterol, Beta-Sitosterol and Dioleylphosphatidylethanolamine for Lipid Nanoparticle Formulation of mRNA. NANOMATERIALS 2022; 12:nano12142446. [PMID: 35889670 PMCID: PMC9317807 DOI: 10.3390/nano12142446] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 12/10/2022]
Abstract
Lipid Nanoparticles (LNPs) are a leading class of mRNA delivery systems. LNPs are made of an ionizable lipid, a polyethyleneglycol (PEG)-lipid conjugate and helper lipids. The success of LNPs is due to proprietary ionizable lipids and appropriate helper lipids. Using a benchmark lipid (D-Lin-MC3) we compared the ability of three helper lipids to transfect dendritic cells in cellulo and in vivo. Studies revealed that the choice of helper lipid does not influence the transfection efficiency of immortalized cells but, LNPs prepared with DOPE (dioleylphosphatidylethanolamine) and β-sitosterol were more efficient for mRNA transfection in murine dendritic cells than LNPs containing DSPC (distearoylphosphatidylcholine). This higher potency of DOPE and β-sitosterol LNPs for mRNA expression was also evident in vivo but only at low mRNA doses. Overall, these data provide valuable insight for the design of novel mRNA LNP vaccines.
Collapse
Affiliation(s)
- Ayoub Medjmedj
- Centre de Biophysique Moléculaire, UPR4301 CNRS, Rue Charles Sadron, 45071 Orléans, France; (A.M.); (A.N.-L.); (R.C.); (J.H.); (C.P.)
| | - Albert Ngalle-Loth
- Centre de Biophysique Moléculaire, UPR4301 CNRS, Rue Charles Sadron, 45071 Orléans, France; (A.M.); (A.N.-L.); (R.C.); (J.H.); (C.P.)
| | - Rudy Clemençon
- Centre de Biophysique Moléculaire, UPR4301 CNRS, Rue Charles Sadron, 45071 Orléans, France; (A.M.); (A.N.-L.); (R.C.); (J.H.); (C.P.)
| | - Josef Hamacek
- Centre de Biophysique Moléculaire, UPR4301 CNRS, Rue Charles Sadron, 45071 Orléans, France; (A.M.); (A.N.-L.); (R.C.); (J.H.); (C.P.)
- Centre de Biophysique Moléculaire, University of Orléans, 45100 Orléans, France
| | - Chantal Pichon
- Centre de Biophysique Moléculaire, UPR4301 CNRS, Rue Charles Sadron, 45071 Orléans, France; (A.M.); (A.N.-L.); (R.C.); (J.H.); (C.P.)
- Centre de Biophysique Moléculaire, University of Orléans, 45100 Orléans, France
| | - Federico Perche
- Centre de Biophysique Moléculaire, UPR4301 CNRS, Rue Charles Sadron, 45071 Orléans, France; (A.M.); (A.N.-L.); (R.C.); (J.H.); (C.P.)
- Correspondence: ; Tel.: +33-2-38-25-55-44
| |
Collapse
|
12
|
Yap CC, Digilio L, McMahon LP, Wang T, Winckler B. Dynein Is Required for Rab7-Dependent Endosome Maturation, Retrograde Dendritic Transport, and Degradation. J Neurosci 2022; 42:4415-4434. [PMID: 35474277 PMCID: PMC9172292 DOI: 10.1523/jneurosci.2530-21.2022] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/30/2022] [Accepted: 04/15/2022] [Indexed: 11/21/2022] Open
Abstract
In all cell types, endocytosed cargo is transported along a set of endosomal compartments, which are linked maturationally from early endosomes (EEs) via late endosomes (LEs) to lysosomes. Lysosomes are critical for degradation of proteins that enter through endocytic as well as autophagic pathways. Rab7 is the master regulator of early-to-late endosome maturation, motility, and fusion with lysosomes. We previously showed that most degradative lysosomes are localized in the soma and in the first 25 µm of the dendrite and that bulk degradation of dendritic membrane proteins occurs in/near the soma. Dendritic late endosomes therefore move retrogradely in a Rab7-dependent manner for fusion with somatic lysosomes. We now used cultured E18 rat hippocampal neurons of both sexes to determine which microtubule motor is responsible for degradative flux of late endosomes. Based on multiple approaches (inhibiting dynein/dynactin itself or inhibiting dynein recruitment to endosomes by expressing the C-terminus of the Rab7 effector, RILP), we now demonstrate that net retrograde flux of late endosomes in dendrites is supported by dynein. Inhibition of dynein also delays maturation of somatic endosomes, as evidenced by excessive accumulation of Rab7. In addition, degradation of dendritic cargos is inhibited. Our results also suggest that GDP-GTP cycling of Rab7 appears necessary not only for endosomal maturation but also for fusion with lysosomes subsequent to arrival in the soma. In conclusion, Rab7-dependent dynein/dynactin recruitment to dendritic endosomes plays multifaceted roles in dendritic endosome maturation as well as retrograde transport of late endosomes to sustain normal degradative flux.SIGNIFICANCE STATEMENT Lysosomes are critical for degradation of membrane and extracellular proteins that enter through endocytosis. Lysosomes are also the endpoint of autophagy and thus responsible for protein and organelle homeostasis. Endosomal-lysosomal dysfunction is linked to neurodegeneration and aging. We identify roles in dendrites for two proteins with links to human diseases, Rab7 and dynein. Our previous work identified a process that requires directional retrograde transport in dendrites, namely, efficient degradation of short-lived membrane proteins. Based on multiple approaches, we demonstrate that Rab7-dependent recruitment of dynein motors supports net retrograde transport to lysosomes and is needed for endosome maturation. Our data also suggest that GDP-GTP cycling of Rab7 is required for fusion with lysosomes and degradation, subsequent to arrival in the soma.
Collapse
Affiliation(s)
- Chan Choo Yap
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia 22908
| | - Laura Digilio
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia 22908
| | - Lloyd P McMahon
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia 22908
| | - Tuanlao Wang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian Province 361005, China
| | - Bettina Winckler
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia 22908
| |
Collapse
|
13
|
Lie PPY, Yoo L, Goulbourne CN, Berg MJ, Stavrides P, Huo C, Lee JH, Nixon RA. Axonal transport of late endosomes and amphisomes is selectively modulated by local Ca 2+ efflux and disrupted by PSEN1 loss of function. SCIENCE ADVANCES 2022; 8:eabj5716. [PMID: 35486730 PMCID: PMC9054012 DOI: 10.1126/sciadv.abj5716] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Dysfunction and mistrafficking of organelles in autophagy- and endosomal-lysosomal pathways are implicated in neurodegenerative diseases. Here, we reveal selective vulnerability of maturing degradative organelles (late endosomes/amphisomes) to disease-relevant local calcium dysregulation. These organelles undergo exclusive retrograde transport in axons, with occasional pauses triggered by regulated calcium efflux from agonist-evoked transient receptor potential cation channel mucolipin subfamily member 1 (TRPML1) channels-an effect greatly exaggerated by exogenous agonist mucolipin synthetic agonist 1 (ML-SA1). Deacidification of degradative organelles, as seen after Presenilin 1 (PSEN1) loss of function, induced pathological constitutive "inside-out" TRPML1 hyperactivation, slowing their transport comparably to ML-SA1 and causing accumulation in dystrophic axons. The mechanism involved calcium-mediated c-Jun N-terminal kinase (JNK) activation, which hyperphosphorylated dynein intermediate chain (DIC), reducing dynein activity. Blocking TRPML1 activation, JNK activity, or DIC1B serine-80 phosphorylation reversed transport deficits in PSEN1 knockout neurons. Our results, including features demonstrated in Alzheimer-mutant PSEN1 knockin mice, define a mechanism linking dysfunction and mistrafficking in lysosomal pathways to neuritic dystrophy under neurodegenerative conditions.
Collapse
Affiliation(s)
- Pearl P. Y. Lie
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
- Department of Psychiatry, New York University Langone Medical Center, New York, NY 10016, USA
| | - Lang Yoo
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
- Department of Psychiatry, New York University Langone Medical Center, New York, NY 10016, USA
| | - Chris N. Goulbourne
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
| | - Martin J. Berg
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
| | - Philip Stavrides
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
| | - Chunfeng Huo
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
| | - Ju-Hyun Lee
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
- Department of Psychiatry, New York University Langone Medical Center, New York, NY 10016, USA
| | - Ralph A. Nixon
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
- Department of Psychiatry, New York University Langone Medical Center, New York, NY 10016, USA
- Department of Cell Biology, New York University Langone Medical Center, New York, NY 10016, USA
- NYU Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA
- Corresponding author.
| |
Collapse
|
14
|
Christensen JR, Kendrick AA, Truong JB, Aguilar-Maldonado A, Adani V, Dzieciatkowska M, Reck-Peterson SL. Cytoplasmic dynein-1 cargo diversity is mediated by the combinatorial assembly of FTS-Hook-FHIP complexes. eLife 2021; 10:74538. [PMID: 34882091 PMCID: PMC8730729 DOI: 10.7554/elife.74538] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/08/2021] [Indexed: 11/13/2022] Open
Abstract
In eukaryotic cells, intracellular components are organized by the microtubule motors cytoplasmic dynein-1 (dynein) and kinesins, which are linked to cargos via adaptor proteins. While ~40 kinesins transport cargo toward the plus end of microtubules, a single dynein moves cargo in the opposite direction. How dynein transports a wide variety of cargos remains an open question. The FTS–Hook–FHIP (‘FHF’) cargo adaptor complex links dynein to cargo in humans and fungi. As human cells have three Hooks and four FHIP proteins, we hypothesized that the combinatorial assembly of different Hook and FHIP proteins could underlie dynein cargo diversity. Using proteomic approaches, we determine the protein ‘interactome’ of each FHIP protein. Live-cell imaging and biochemical approaches show that different FHF complexes associate with distinct motile cargos. These complexes also move with dynein and its cofactor dynactin in single-molecule in vitro reconstitution assays. Complexes composed of FTS, FHIP1B, and Hook1/Hook3 colocalize with Rab5-tagged early endosomes via a direct interaction between FHIP1B and GTP-bound Rab5. In contrast, complexes composed of FTS, FHIP2A, and Hook2 colocalize with Rab1A-tagged ER-to-Golgi cargos and FHIP2A is involved in the motility of Rab1A tubules. Our findings suggest that combinatorial assembly of different FTS–Hook–FHIP complexes is one mechanism dynein uses to achieve cargo specificity.
Collapse
Affiliation(s)
- Jenna R Christensen
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, United States
| | - Agnieszka A Kendrick
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, United States
| | - Joey B Truong
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, United States
| | | | - Vinit Adani
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, United States
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, United States
| | - Samara L Reck-Peterson
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, United States
| |
Collapse
|
15
|
Su X, You X, Luo H, Liang K, Chen L, Tian W, Ye Z, He J. Community-Acquired Respiratory Distress Syndrome Toxin: Unique Exotoxin for M. pneumoniae. Front Microbiol 2021; 12:766591. [PMID: 34867898 PMCID: PMC8640204 DOI: 10.3389/fmicb.2021.766591] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/19/2021] [Indexed: 11/13/2022] Open
Abstract
Mycoplasma pneumoniae infection often causes respiratory diseases in humans, particularly in children and adults with atypical pneumonia and community-acquired pneumonia (CAP), and is often exacerbated by co-infection with other lung diseases, such as asthma, bronchitis, and chronic obstructive pulmonary disorder. Community-acquired respiratory distress syndrome toxin (CARDS TX) is the only exotoxin produced by M. pneumoniae and has been extensively studied for its ADP-ribosyltransferase (ADPRT) activity and cellular vacuolization properties. Additionally, CARDS TX induces inflammatory responses, resulting in cell swelling, nuclear lysis, mucus proliferation, and cell vacuolization. CARDS TX enters host cells by binding to the host receptor and is then reverse transported to the endoplasmic reticulum to exert its pathogenic effects. In this review, we focus on the structural characteristics, functional activity, distribution and receptors, mechanism of cell entry, and inflammatory response of CARDS TX was examined. Overall, the findings of this review provide a theoretical basis for further investigation of the mechanism of M. pneumoniae infection and the development of clinical diagnosis and vaccines.
Collapse
Affiliation(s)
- Xiaoling Su
- The Affiliated Nanhua Hospital, Department of Clinical Laboratory, Hengyang Medical School, University of South China, Hengyang, China
| | - Xiaoxing You
- Institute of Pathogenic Biology, Hengyang Medical School, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Haodang Luo
- The Affiliated Nanhua Hospital, Department of Clinical Laboratory, Hengyang Medical School, University of South China, Hengyang, China
| | - Keying Liang
- The Affiliated Nanhua Hospital, Department of Clinical Laboratory, Hengyang Medical School, University of South China, Hengyang, China
| | - Li Chen
- The Affiliated Nanhua Hospital, Department of Clinical Laboratory, Hengyang Medical School, University of South China, Hengyang, China
| | - Wei Tian
- The Affiliated Nanhua Hospital, Department of Clinical Laboratory, Hengyang Medical School, University of South China, Hengyang, China
| | - Zufeng Ye
- The Affiliated Nanhua Hospital, Department of Clinical Laboratory, Hengyang Medical School, University of South China, Hengyang, China
| | - Jun He
- The Affiliated Nanhua Hospital, Department of Clinical Laboratory, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
16
|
Podinovskaia M, Prescianotto-Baschong C, Buser DP, Spang A. A novel live-cell imaging assay reveals regulation of endosome maturation. eLife 2021; 10:e70982. [PMID: 34846303 PMCID: PMC8635980 DOI: 10.7554/elife.70982] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 11/09/2021] [Indexed: 12/12/2022] Open
Abstract
Cell-cell communication is an essential process in life, with endosomes acting as key organelles for regulating uptake and secretion of signaling molecules. Endocytosed material is accepted by the sorting endosome where it either is sorted for recycling or remains in the endosome as it matures to be degraded in the lysosome. Investigation of the endosome maturation process has been hampered by the small size and rapid movement of endosomes in most cellular systems. Here, we report an easy versatile live-cell imaging assay to monitor endosome maturation kinetics, which can be applied to a variety of mammalian cell types. Acute ionophore treatment led to enlarged early endosomal compartments that matured into late endosomes and fused with lysosomes to form endolysosomes. Rab5-to-Rab7 conversion and PI(3)P formation and turn over were recapitulated with this assay and could be observed with a standard widefield microscope. We used this approach to show that Snx1 and Rab11-positive recycling endosome recruitment occurred throughout endosome maturation and was uncoupled from Rab conversion. In contrast, efficient endosomal acidification was dependent on Rab conversion. The assay provides a powerful tool to further unravel various aspects of endosome maturation.
Collapse
Affiliation(s)
| | | | | | - Anne Spang
- Biozentrum, University of BaselBaselSwitzerland
| |
Collapse
|
17
|
Radaszkiewicz T, Nosková M, Gömöryová K, Vondálová Blanářová O, Radaszkiewicz KA, Picková M, Víchová R, Gybeľ T, Kaiser K, Demková L, Kučerová L, Bárta T, Potěšil D, Zdráhal Z, Souček K, Bryja V. RNF43 inhibits WNT5A-driven signaling and suppresses melanoma invasion and resistance to the targeted therapy. eLife 2021; 10:65759. [PMID: 34702444 PMCID: PMC8550759 DOI: 10.7554/elife.65759] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 09/28/2021] [Indexed: 12/30/2022] Open
Abstract
RNF43 is an E3 ubiquitin ligase and known negative regulator of WNT/β-catenin signaling. We demonstrate that RNF43 is also a regulator of noncanonical WNT5A-induced signaling in human cells. Analysis of the RNF43 interactome using BioID and immunoprecipitation showed that RNF43 can interact with the core receptor complex components dedicated to the noncanonical Wnt pathway such as ROR1, ROR2, VANGL1, and VANGL2. RNF43 triggers VANGL2 ubiquitination and proteasomal degradation and clathrin-dependent internalization of ROR1 receptor and inhibits ROR2 activation. These activities of RNF43 are physiologically relevant and block pro-metastatic WNT5A signaling in melanoma. RNF43 inhibits responses to WNT5A, which results in the suppression of invasive properties of melanoma cells. Furthermore, RNF43 prevented WNT5A-assisted development of resistance to BRAF V600E and MEK inhibitors. Next, RNF43 acted as melanoma suppressor and improved response to targeted therapies in vivo. In line with these findings, RNF43 expression decreases during melanoma progression and RNF43-low patients have a worse prognosis. We conclude that RNF43 is a newly discovered negative regulator of WNT5A-mediated biological responses that desensitizes cells to WNT5A.
Collapse
Affiliation(s)
- Tomasz Radaszkiewicz
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Michaela Nosková
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Kristína Gömöryová
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Olga Vondálová Blanářová
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | | | - Markéta Picková
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic.,Department of Cytokinetics, Institute of Biophysics CAS, Brno, Czech Republic.,International Clinical Research Center FNUSA-ICRC, Brno, Czech Republic
| | - Ráchel Víchová
- Department of Cytokinetics, Institute of Biophysics CAS, Brno, Czech Republic
| | - Tomáš Gybeľ
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Karol Kaiser
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Lucia Demková
- Laboratory of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Bratislava, Slovakia
| | - Lucia Kučerová
- Laboratory of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Bratislava, Slovakia
| | - Tomáš Bárta
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic.,Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - David Potěšil
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Zbyněk Zdráhal
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Karel Souček
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic.,Department of Cytokinetics, Institute of Biophysics CAS, Brno, Czech Republic.,International Clinical Research Center FNUSA-ICRC, Brno, Czech Republic
| | - Vítězslav Bryja
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic.,Department of Cytokinetics, Institute of Biophysics CAS, Brno, Czech Republic
| |
Collapse
|
18
|
Revising Endosomal Trafficking under Insulin Receptor Activation. Int J Mol Sci 2021; 22:ijms22136978. [PMID: 34209489 PMCID: PMC8268289 DOI: 10.3390/ijms22136978] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/25/2021] [Accepted: 06/27/2021] [Indexed: 02/06/2023] Open
Abstract
The endocytosis of ligand-bound receptors and their eventual recycling to the plasma membrane (PM) are processes that have an influence on signalling activity and therefore on many cell functions, including migration and proliferation. Like other tyrosine kinase receptors (TKR), the insulin receptor (INSR) has been shown to be endocytosed by clathrin-dependent and -independent mechanisms. Once at the early endosome (EE), the sorting of the receptor, either to the late endosome (LE) for degradation or back to the PM through slow or fast recycling pathways, will determine the intensity and duration of insulin effects. Both the endocytic and the endosomic pathways are regulated by many proteins, the Arf and Rab families of small GTPases being some of the most relevant. Here, we argue for a specific role for the slow recycling route, whilst we review the main molecular mechanisms involved in INSR endocytosis, sorting and recycling, as well as their possible role in cell functions.
Collapse
|
19
|
Vos M, Klein C. The Importance of Drosophila melanogaster Research to UnCover Cellular Pathways Underlying Parkinson's Disease. Cells 2021; 10:579. [PMID: 33800736 PMCID: PMC7998316 DOI: 10.3390/cells10030579] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/01/2021] [Accepted: 03/05/2021] [Indexed: 12/11/2022] Open
Abstract
Parkinson's disease (PD) is a complex neurodegenerative disorder that is currently incurable. As a consequence of an incomplete understanding of the etiology of the disease, therapeutic strategies mainly focus on symptomatic treatment. Even though the majority of PD cases remain idiopathic (~90%), several genes have been identified to be causative for PD, facilitating the generation of animal models that are a good alternative to study disease pathways and to increase our understanding of the underlying mechanisms of PD. Drosophila melanogaster has proven to be an excellent model in these studies. In this review, we will discuss the different PD models in flies and key findings identified in flies in different affected pathways in PD. Several molecular changes have been identified, of which mitochondrial dysfunction and a defective endo-lysosomal pathway emerge to be the most relevant for PD pathogenesis. Studies in flies have significantly contributed to our knowledge of how disease genes affect and interact in these pathways enabling a better understanding of the disease etiology and providing possible therapeutic targets for the treatment of PD, some of which have already resulted in clinical trials.
Collapse
Affiliation(s)
- Melissa Vos
- Institute of Neurogenetics, University of Luebeck, Ratzeburger Allee 160, Building 67, 23562 Luebeck, Germany
| | - Christine Klein
- Institute of Neurogenetics, University of Luebeck, Ratzeburger Allee 160, Building 67, 23562 Luebeck, Germany
| |
Collapse
|
20
|
Hilterbrand AT, Daly RE, Heldwein EE. Contributions of the Four Essential Entry Glycoproteins to HSV-1 Tropism and the Selection of Entry Routes. mBio 2021; 12:e00143-21. [PMID: 33653890 PMCID: PMC8092210 DOI: 10.1128/mbio.00143-21] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 01/22/2021] [Indexed: 01/02/2023] Open
Abstract
Herpes simplex viruses (HSV-1 and HSV-2) encode up to 16 envelope proteins, four of which are essential for entry. However, whether these four proteins alone are sufficient to dictate the broad cellular tropism of HSV-1 and the selection of different cell type-dependent entry routes is unknown. To begin addressing this, we previously pseudotyped vesicular stomatitis virus (VSV), lacking its native glycoprotein G, with only the four essential entry glycoproteins of HSV-1: gB, gH, gL, and gD. This novel VSVΔG-BHLD pseudotype recapitulated several important features of HSV-1 entry: the requirement for gB, gH, gL, gD, and a cellular receptor and sensitivity to anti-gB and anti-gH/gL neutralizing antibodies. However, due to the use of a single cell type in that study, the tropism of the VSVΔG-BHLD pseudotype was not investigated. Here, we show that the cellular tropism of the pseudotype is severely limited compared to that of wild-type HSV-1 and that its entry pathways differ from the native HSV-1 entry pathways. To test the hypothesis that other HSV-1 envelope proteins may contribute to HSV-1 tropism, we generated a derivative pseudotype containing the HSV-1 glycoprotein C (VSVΔG-BHLD-gC) and observed a gC-dependent increase in entry efficiency in two cell types. We propose that the pseudotyping platform developed here has the potential to uncover functional contributions of HSV-1 envelope proteins to entry in a gain-of-function manner.IMPORTANCE Herpes simplex viruses (HSV-1 and HSV-2) contain up to 16 different proteins in their envelopes. Four of these, glycoproteins gB, gD, gH, and gL, are termed essential with regard to entry, whereas the rest are typically referred to as nonessential based on the entry phenotypes of the respective single genetic deletions. However, the single-gene deletion approach, which relies on robust loss-of-function phenotypes, may be confounded by functional redundancies among the many HSV-1 envelope proteins. We have developed a pseudotyping platform in which the essential four entry glycoproteins are isolated from the rest, which can be added back individually for systematic gain-of-function entry experiments. Here, we show the utility of this platform for dissecting the contributions of HSV envelope proteins, both the essential four and the remaining dozen (using gC as an example), to HSV entry.
Collapse
Affiliation(s)
- Adam T Hilterbrand
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Raecliffe E Daly
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
- Graduate Program in Cellular, Molecular, and Developmental Biology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Ekaterina E Heldwein
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
- Graduate Program in Cellular, Molecular, and Developmental Biology, Tufts University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
21
|
Melendez J, Sieiro D, Salgado D, Morin V, Dejardin MJ, Zhou C, Mullen AC, Marcelle C. TGFβ signalling acts as a molecular brake of myoblast fusion. Nat Commun 2021; 12:749. [PMID: 33531476 PMCID: PMC7854724 DOI: 10.1038/s41467-020-20290-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 11/21/2020] [Indexed: 01/30/2023] Open
Abstract
Fusion of nascent myoblasts to pre-existing myofibres is critical for skeletal muscle growth and repair. The vast majority of molecules known to regulate myoblast fusion are necessary in this process. Here, we uncover, through high-throughput in vitro assays and in vivo studies in the chicken embryo, that TGFβ (SMAD2/3-dependent) signalling acts specifically and uniquely as a molecular brake on muscle fusion. While constitutive activation of the pathway arrests fusion, its inhibition leads to a striking over-fusion phenotype. This dynamic control of TGFβ signalling in the embryonic muscle relies on a receptor complementation mechanism, prompted by the merging of myoblasts with myofibres, each carrying one component of the heterodimer receptor complex. The competence of myofibres to fuse is likely restored through endocytic degradation of activated receptors. Altogether, this study shows that muscle fusion relies on TGFβ signalling to regulate its pace.
Collapse
Affiliation(s)
- Julie Melendez
- Institut NeuroMyoGène (INMG), University Claude Bernard Lyon1, CNRS UMR 5310, INSERM U1217, Lyon, France
| | - Daniel Sieiro
- Institut NeuroMyoGène (INMG), University Claude Bernard Lyon1, CNRS UMR 5310, INSERM U1217, Lyon, France
- Australian Regenerative Medicine Institute (ARMI), Monash University, Clayton, VIC, Australia
- Plexus Ventures LLC, Boston, MA, USA
| | - David Salgado
- Australian Regenerative Medicine Institute (ARMI), Monash University, Clayton, VIC, Australia
- Marseille Medical Genetics (MMG), Aix Marseille University, INSERM U1251, Marseille, France
| | - Valérie Morin
- Institut NeuroMyoGène (INMG), University Claude Bernard Lyon1, CNRS UMR 5310, INSERM U1217, Lyon, France
| | - Marie-Julie Dejardin
- Institut NeuroMyoGène (INMG), University Claude Bernard Lyon1, CNRS UMR 5310, INSERM U1217, Lyon, France
| | - Chan Zhou
- Gastrointestinal Unit, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Alan C Mullen
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA
| | - Christophe Marcelle
- Institut NeuroMyoGène (INMG), University Claude Bernard Lyon1, CNRS UMR 5310, INSERM U1217, Lyon, France.
- Australian Regenerative Medicine Institute (ARMI), Monash University, Clayton, VIC, Australia.
| |
Collapse
|
22
|
Huizing M, Gahl WA. Inherited disorders of lysosomal membrane transporters. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2020; 1862:183336. [PMID: 32389669 PMCID: PMC7508925 DOI: 10.1016/j.bbamem.2020.183336] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/01/2020] [Accepted: 04/28/2020] [Indexed: 02/07/2023]
Abstract
Disorders caused by defects in lysosomal membrane transporters form a distinct subgroup of lysosomal storage disorders (LSDs). To date, defects in only 10 lysosomal membrane transporters have been associated with inherited disorders. The clinical presentations of these diseases resemble the phenotypes of other LSDs; they are heterogeneous and often present in children with neurodegenerative manifestations. However, for pathomechanistic and therapeutic studies, lysosomal membrane transport defects should be distinguished from LSDs caused by defective hydrolytic enzymes. The involved proteins differ in function, localization, and lysosomal targeting, and the diseases themselves differ in their stored material and therapeutic approaches. We provide an overview of the small group of disorders of lysosomal membrane transporters, emphasizing discovery, pathomechanism, clinical features, diagnostic methods and therapeutic aspects. We discuss common aspects of lysosomal membrane transporter defects that can provide the basis for preclinical research into these disorders.
Collapse
Affiliation(s)
- Marjan Huizing
- Human Biochemical Genetics Section, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - William A Gahl
- Human Biochemical Genetics Section, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
23
|
A trimeric Rab7 GEF controls NPC1-dependent lysosomal cholesterol export. Nat Commun 2020; 11:5559. [PMID: 33144569 PMCID: PMC7642327 DOI: 10.1038/s41467-020-19032-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 09/18/2020] [Indexed: 12/26/2022] Open
Abstract
Cholesterol import in mammalian cells is mediated by the LDL receptor pathway. Here, we perform a genome-wide CRISPR screen using an endogenous cholesterol reporter and identify >100 genes involved in LDL-cholesterol import. We characterise C18orf8 as a core subunit of the mammalian Mon1-Ccz1 guanidine exchange factor (GEF) for Rab7, required for complex stability and function. C18orf8-deficient cells lack Rab7 activation and show severe defects in late endosome morphology and endosomal LDL trafficking, resulting in cellular cholesterol deficiency. Unexpectedly, free cholesterol accumulates within swollen lysosomes, suggesting a critical defect in lysosomal cholesterol export. We find that active Rab7 interacts with the NPC1 cholesterol transporter and licenses lysosomal cholesterol export. This process is abolished in C18orf8-, Ccz1- and Mon1A/B-deficient cells and restored by a constitutively active Rab7. The trimeric Mon1-Ccz1-C18orf8 (MCC) GEF therefore plays a central role in cellular cholesterol homeostasis coordinating Rab7 activation, endosomal LDL trafficking and NPC1-dependent lysosomal cholesterol export. Lysosomes play an important role in cellular LDL-cholesterol uptake. Here, the authors perform a genome-wide genetic screen for cholesterol regulators and identify C18orf8 as a conserved subunit of a trimeric Rab7 GEF that controls LDL trafficking and NPC1-dependent lysosomal cholesterol export.
Collapse
|
24
|
SNX27-Mediated Recycling of Neuroligin-2 Regulates Inhibitory Signaling. Cell Rep 2020; 29:2599-2607.e6. [PMID: 31775031 PMCID: PMC6899438 DOI: 10.1016/j.celrep.2019.10.096] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 08/22/2019] [Accepted: 10/24/2019] [Indexed: 01/11/2023] Open
Abstract
GABAA receptors mediate fast inhibitory transmission in the brain, and their number can be rapidly up- or downregulated to alter synaptic strength. Neuroligin-2 plays a critical role in the stabilization of synaptic GABAA receptors and the development and maintenance of inhibitory synapses. To date, little is known about how the amount of neuroligin-2 at the synapse is regulated and whether neuroligin-2 trafficking affects inhibitory signaling. Here, we show that neuroligin-2, when internalized to endosomes, co-localizes with SNX27, a brain-enriched cargo-adaptor protein that facilitates membrane protein recycling. Direct interaction between the PDZ domain of SNX27 and PDZ-binding motif in neuroligin-2 enables membrane retrieval of neuroligin-2, thus enhancing synaptic neuroligin-2 clusters. Furthermore, SNX27 knockdown has the opposite effect. SNX27-mediated up- and downregulation of neuroligin-2 surface levels affects inhibitory synapse composition and signaling strength. Taken together, we show a role for SNX27-mediated recycling of neuroligin-2 in maintenance and signaling of the GABAergic synapse.
Collapse
|
25
|
The Role of Vesicle Trafficking Defects in the Pathogenesis of Prion and Prion-Like Disorders. Int J Mol Sci 2020; 21:ijms21197016. [PMID: 32977678 PMCID: PMC7582986 DOI: 10.3390/ijms21197016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/15/2020] [Accepted: 09/21/2020] [Indexed: 11/26/2022] Open
Abstract
Prion diseases are fatal and transmissible neurodegenerative diseases in which the cellular form of the prion protein ‘PrPc’, misfolds into an infectious and aggregation prone isoform termed PrPSc, which is the primary component of prions. Many neurodegenerative diseases, like Alzheimer’s disease, Parkinson’s disease, and polyglutamine diseases, such as Huntington’s disease, are considered prion-like disorders because of the common characteristics in the propagation and spreading of misfolded proteins that they share with the prion diseases. Unlike prion diseases, these are non-infectious outside experimental settings. Many vesicular trafficking impairments, which are observed in prion and prion-like disorders, favor the accumulation of the pathogenic amyloid aggregates. In addition, many of the vesicular trafficking impairments that arise in these diseases, turn out to be further aggravating factors. This review offers an insight into the currently known vesicular trafficking defects in these neurodegenerative diseases and their implications on disease progression. These findings suggest that these impaired trafficking pathways may represent similar therapeutic targets in these classes of neurodegenerative disorders.
Collapse
|
26
|
A High-Content Screen Identifies TPP1 and Aurora B as Regulators of Axonal Mitochondrial Transport. Cell Rep 2020; 28:3224-3237.e5. [PMID: 31533043 PMCID: PMC6937139 DOI: 10.1016/j.celrep.2019.08.035] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 05/12/2019] [Accepted: 08/09/2019] [Indexed: 12/22/2022] Open
Abstract
Dysregulated axonal trafficking of mitochondria is linked to neurodegenerative disorders. We report a high-content screen for small-molecule regulators of the axonal transport of mitochondria. Six compounds enhanced mitochondrial transport in the sub-micromolar range, acting via three cellular targets: F-actin, Tripeptidyl peptidase 1 (TPP1), or Aurora Kinase B (AurKB). Pharmacological inhibition or small hairpin RNA (shRNA) knockdown of each target promotes mitochondrial axonal transport in rat hippocampal neurons and induced pluripotent stem cell (iPSC)-derived human cortical neurons and enhances mitochondrial transport in iPSC-derived motor neurons from an amyotrophic lateral sclerosis (ALS) patient bearing one copy of SOD1A4V mutation. Our work identifies druggable regulators of axonal transport of mitochondria, provides broadly applicable methods for similar image-based screens, and suggests that restoration of proper axonal trafficking of mitochondria can be achieved in human ALS neurons. Shlevkov et al. establish a high-content screen for enhancers of axonal mitochondrial trafficking. Identified compounds act through three cellular targets: F-Actin, Tripeptidyl peptidase 1, and Aurora Kinase B. Motor neurons derived from a SOD1+/A4VALS patient have decreased mitochondrial motility, which can be reversed by inhibitors of these targets.
Collapse
|
27
|
Joshi DC, Zhang CL, Babujee L, Vevea JD, August BK, Sheng ZH, Chapman ER, Gomez TM, Chiu SY. Inappropriate Intrusion of an Axonal Mitochondrial Anchor into Dendrites Causes Neurodegeneration. Cell Rep 2020; 29:685-696.e5. [PMID: 31618636 PMCID: PMC6884150 DOI: 10.1016/j.celrep.2019.09.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 07/30/2019] [Accepted: 09/05/2019] [Indexed: 01/21/2023] Open
Abstract
Syntaphilin (SNPH) is a major mitochondrial anchoring protein targeted to axons and excluded from dendrites. In this study, we provide in vivo evidence that this spatial specificity is lost in Shiverer (Shi) mice, a model for progressive multiple sclerosis (MS), resulting in inappropriate intrusion of SNPH into dendrites of cerebellar Purkinje cells with neurodegenerative consequences. Thus, reconstituting dendritic SNPH intrusion in SNPH-KO mice by viral transduction greatly sensitizes Purkinje cells to excitotoxicity when the glutamatergic climbing fibers are stimulated. Finally, we demonstrate in vitro that overexpression of SNPH in dendrites compromises neuronal viability by inducing N-methyl-D-aspartate (NMDA) excitotoxicity, reducing mitochondrial calcium uptake, and interfering with quality control of mitochondria by blocking somal mitophagy. Collectively, we propose that inappropriate immobilization of dendritic mitochondria by SNPH intrusion produces excitotoxicity and suggest that interception of dendritic SNPH intrusion is a therapeutic strategy to combat neurodegeneration.
Collapse
Affiliation(s)
- Dinesh C Joshi
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Chuan-Li Zhang
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Lavanya Babujee
- Department of Plant Pathology, College of Agricultural and Life Sciences, University of Wisconsin, Madison, WI, USA
| | - Jason D Vevea
- Howard Hughes Medical Institute and Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Benjamin K August
- Medical School Electron Microscope Facility, University of Wisconsin, Madison, WI, USA
| | - Zu-Hang Sheng
- Synaptic Functions Section, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Edwin R Chapman
- Howard Hughes Medical Institute and Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Timothy M Gomez
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Shing Yan Chiu
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
28
|
Kounatidis I, Stanifer ML, Phillips MA, Paul-Gilloteaux P, Heiligenstein X, Wang H, Okolo CA, Fish TM, Spink MC, Stuart DI, Davis I, Boulant S, Grimes JM, Dobbie IM, Harkiolaki M. 3D Correlative Cryo-Structured Illumination Fluorescence and Soft X-ray Microscopy Elucidates Reovirus Intracellular Release Pathway. Cell 2020; 182:515-530.e17. [PMID: 32610083 PMCID: PMC7391008 DOI: 10.1016/j.cell.2020.05.051] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/14/2020] [Accepted: 05/28/2020] [Indexed: 01/15/2023]
Abstract
Imaging of biological matter across resolution scales entails the challenge of preserving the direct and unambiguous correlation of subject features from the macroscopic to the microscopic level. Here, we present a correlative imaging platform developed specifically for imaging cells in 3D under cryogenic conditions by using X-rays and visible light. Rapid cryo-preservation of biological specimens is the current gold standard in sample preparation for ultrastructural analysis in X-ray imaging. However, cryogenic fluorescence localization methods are, in their majority, diffraction-limited and fail to deliver matching resolution. We addressed this technological gap by developing an integrated, user-friendly platform for 3D correlative imaging of cells in vitreous ice by using super-resolution structured illumination microscopy in conjunction with soft X-ray tomography. The power of this approach is demonstrated by studying the process of reovirus release from intracellular vesicles during the early stages of infection and identifying intracellular virus-induced structures.
Collapse
Affiliation(s)
- Ilias Kounatidis
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot OX11 0DE, UK
| | - Megan L Stanifer
- Department of Infectious Diseases, Molecular Virology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Michael A Phillips
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot OX11 0DE, UK; Micron Advanced Imaging Consortium, Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK; Division of Structural Biology, The Henry Wellcome Building for Genomic Medicine, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - Perrine Paul-Gilloteaux
- Université de Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France; Nantes Université, CHU Nantes, Inserm, CNRS, SFR Santé, Inserm UMS 016, CNRS UMS3556, Nantes, France
| | | | - Hongchang Wang
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot OX11 0DE, UK
| | - Chidinma A Okolo
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot OX11 0DE, UK
| | - Thomas M Fish
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot OX11 0DE, UK
| | - Matthew C Spink
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot OX11 0DE, UK
| | - David I Stuart
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot OX11 0DE, UK; Division of Structural Biology, The Henry Wellcome Building for Genomic Medicine, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - Ilan Davis
- Micron Advanced Imaging Consortium, Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Steeve Boulant
- Department of Infectious Diseases, Virology, Heidelberg University Hospital, 69120 Heidelberg, Germany; Research Group "Cellular polarity and viral infection," German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Jonathan M Grimes
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot OX11 0DE, UK; Division of Structural Biology, The Henry Wellcome Building for Genomic Medicine, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - Ian M Dobbie
- Micron Advanced Imaging Consortium, Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Maria Harkiolaki
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot OX11 0DE, UK.
| |
Collapse
|
29
|
Silva BSC, DiGiovanni L, Kumar R, Carmichael RE, Kim PK, Schrader M. Maintaining social contacts: The physiological relevance of organelle interactions. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118800. [PMID: 32712071 PMCID: PMC7377706 DOI: 10.1016/j.bbamcr.2020.118800] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/12/2020] [Accepted: 07/19/2020] [Indexed: 02/07/2023]
Abstract
Membrane-bound organelles in eukaryotic cells form an interactive network to coordinate and facilitate cellular functions. The formation of close contacts, termed "membrane contact sites" (MCSs), represents an intriguing strategy for organelle interaction and coordinated interplay. Emerging research is rapidly revealing new details of MCSs. They represent ubiquitous and diverse structures, which are important for many aspects of cell physiology and homeostasis. Here, we provide a comprehensive overview of the physiological relevance of organelle contacts. We focus on mitochondria, peroxisomes, the Golgi complex and the plasma membrane, and discuss the most recent findings on their interactions with other subcellular organelles and their multiple functions, including membrane contacts with the ER, lipid droplets and the endosomal/lysosomal compartment.
Collapse
Affiliation(s)
- Beatriz S C Silva
- College of Life and Environmental Sciences, Biosciences, University of Exeter, Exeter EX4 4QD, Devon, UK
| | - Laura DiGiovanni
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada; Department of Biochemistry, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Rechal Kumar
- College of Life and Environmental Sciences, Biosciences, University of Exeter, Exeter EX4 4QD, Devon, UK
| | - Ruth E Carmichael
- College of Life and Environmental Sciences, Biosciences, University of Exeter, Exeter EX4 4QD, Devon, UK.
| | - Peter K Kim
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada; Department of Biochemistry, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| | - Michael Schrader
- College of Life and Environmental Sciences, Biosciences, University of Exeter, Exeter EX4 4QD, Devon, UK.
| |
Collapse
|
30
|
Meneses-Salas E, García-Melero A, Kanerva K, Blanco-Muñoz P, Morales-Paytuvi F, Bonjoch J, Casas J, Egert A, Beevi SS, Jose J, Llorente-Cortés V, Rye KA, Heeren J, Lu A, Pol A, Tebar F, Ikonen E, Grewal T, Enrich C, Rentero C. Annexin A6 modulates TBC1D15/Rab7/StARD3 axis to control endosomal cholesterol export in NPC1 cells. Cell Mol Life Sci 2020; 77:2839-2857. [PMID: 31664461 PMCID: PMC7326902 DOI: 10.1007/s00018-019-03330-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/30/2019] [Accepted: 10/01/2019] [Indexed: 01/23/2023]
Abstract
Cholesterol accumulation in late endosomes is a prevailing phenotype of Niemann-Pick type C1 (NPC1) mutant cells. Likewise, annexin A6 (AnxA6) overexpression induces a phenotype reminiscent of NPC1 mutant cells. Here, we demonstrate that this cellular cholesterol imbalance is due to AnxA6 promoting Rab7 inactivation via TBC1D15, a Rab7-GAP. In NPC1 mutant cells, AnxA6 depletion and eventual Rab7 activation was associated with peripheral distribution and increased mobility of late endosomes. This was accompanied by an enhanced lipid accumulation in lipid droplets in an acyl-CoA:cholesterol acyltransferase (ACAT)-dependent manner. Moreover, in AnxA6-deficient NPC1 mutant cells, Rab7-mediated rescue of late endosome-cholesterol export required the StAR-related lipid transfer domain-3 (StARD3) protein. Electron microscopy revealed a significant increase of membrane contact sites (MCS) between late endosomes and ER in NPC1 mutant cells lacking AnxA6, suggesting late endosome-cholesterol transfer to the ER via Rab7 and StARD3-dependent MCS formation. This study identifies AnxA6 as a novel gatekeeper that controls cellular distribution of late endosome-cholesterol via regulation of a Rab7-GAP and MCS formation.
Collapse
Affiliation(s)
- Elsa Meneses-Salas
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036, Barcelona, Spain
- Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain
| | - Ana García-Melero
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036, Barcelona, Spain
| | - Kristiina Kanerva
- Faculty of Medicine, Anatomy, University of Helsinki, 00014, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, 00290, Helsinki, Finland
| | - Patricia Blanco-Muñoz
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036, Barcelona, Spain
- Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain
| | - Frederic Morales-Paytuvi
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036, Barcelona, Spain
- Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain
| | - Júlia Bonjoch
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036, Barcelona, Spain
- Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain
| | - Josefina Casas
- Research Unit on BioActive Molecules (RUBAM), Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain
| | - Antonia Egert
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, 2006, Australia
| | - Syed S Beevi
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, 2006, Australia
| | - Jaimy Jose
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, 2006, Australia
| | - Vicenta Llorente-Cortés
- Lipids and Cardiovascular Pathology Group, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
- CIBERCV, Institute of Health Carlos III, Madrid, Spain
- Biomedical Research Institute of Barcelona-CSIC, Barcelona, Spain
| | - Kerry-Anne Rye
- School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Joerg Heeren
- Department of Biochemistry and Molecular Biology II: Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Albert Lu
- Department of Biochemistry, Stanford University School of Medicine, Stanford, USA
| | - Albert Pol
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036, Barcelona, Spain
- Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avaçats (ICREA), 08010, Barcelona, Spain
| | - Francesc Tebar
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036, Barcelona, Spain
- Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain
| | - Elina Ikonen
- Faculty of Medicine, Anatomy, University of Helsinki, 00014, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, 00290, Helsinki, Finland
| | - Thomas Grewal
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, 2006, Australia.
| | - Carlos Enrich
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036, Barcelona, Spain.
- Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain.
| | - Carles Rentero
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036, Barcelona, Spain.
- Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain.
| |
Collapse
|
31
|
Overeem AW, Li Q, Qiu Y, Cartón‐García F, Leng C, Klappe K, Dronkers J, Hsiao N, Wang J, Arango D, van Ijzendoorn SC. A Molecular Mechanism Underlying Genotype-Specific Intrahepatic Cholestasis Resulting From MYO5B Mutations. Hepatology 2020; 72:213-229. [PMID: 31750554 PMCID: PMC7496772 DOI: 10.1002/hep.31002] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 10/17/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Progressive familial intrahepatic cholestasis (PFIC) 6 has been associated with missense but not biallelic nonsense or frameshift mutations in MYO5B, encoding the motor protein myosin Vb (myoVb). This genotype-phenotype correlation and the mechanism through which MYO5B mutations give rise to PFIC are not understood. The aim of this study was to determine whether the loss of myoVb or expression of patient-specific myoVb mutants can be causally related to defects in canalicular protein localization and, if so, through which mechanism. APPROACH AND RESULTS We demonstrate that the cholestasis-associated substitution of the proline at amino acid position 600 in the myoVb protein to a leucine (P660L) caused the intracellular accumulation of bile canalicular proteins in vesicular compartments. Remarkably, the knockout of MYO5B in vitro and in vivo produced no canalicular localization defects. In contrast, the expression of myoVb mutants consisting of only the tail domain phenocopied the effects of the Myo5b-P660L mutation. Using additional myoVb and rab11a mutants, we demonstrate that motor domain-deficient myoVb inhibited the formation of specialized apical recycling endosomes and that its disrupting effect on the localization of canalicular proteins was dependent on its interaction with active rab11a and occurred at the trans-Golgi Network/recycling endosome interface. CONCLUSIONS Our results reveal a mechanism through which MYO5B motor domain mutations can cause the mislocalization of canalicular proteins in hepatocytes which, unexpectedly, does not involve myoVb loss-of-function but, as we propose, a rab11a-mediated gain-of-toxic function. The results explain why biallelic MYO5B mutations that affect the motor domain but not those that eliminate myoVb expression are associated with PFIC6.
Collapse
Affiliation(s)
- Arend W. Overeem
- Department of Biomedical Sciences of Cells and SystemsSection Molecular Cell BiologyUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| | - Qinghong Li
- Department of Biomedical Sciences of Cells and SystemsSection Molecular Cell BiologyUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| | - Yi‐Ling Qiu
- The Center for Pediatric Liver DiseasesChildren’s Hospital of Fudan UniversityShanghaiChina,Department of PediatricsJinshan Hospital of Fudan UniversityShanghaiChina
| | - Fernando Cartón‐García
- Group of Biomedical Research in Digestive Tract TumorsCIBBIM‐NanomedicineVall d’Hebron Research Institute (VHIR)Universitat Autònoma de Barcelona (UAB)Barcelona08035Spain
| | - Changsen Leng
- Department of Biomedical Sciences of Cells and SystemsSection Molecular Cell BiologyUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| | - Karin Klappe
- Department of Biomedical Sciences of Cells and SystemsSection Molecular Cell BiologyUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| | - Just Dronkers
- Department of Biomedical Sciences of Cells and SystemsSection Molecular Cell BiologyUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| | - Nai‐Hua Hsiao
- Department of Biomedical Sciences of Cells and SystemsSection Molecular Cell BiologyUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| | - Jian‐She Wang
- The Center for Pediatric Liver DiseasesChildren’s Hospital of Fudan UniversityShanghaiChina,Department of PediatricsJinshan Hospital of Fudan UniversityShanghaiChina
| | - Diego Arango
- Group of Biomedical Research in Digestive Tract TumorsCIBBIM‐NanomedicineVall d’Hebron Research Institute (VHIR)Universitat Autònoma de Barcelona (UAB)Barcelona08035Spain
| | - Sven C.D. van Ijzendoorn
- Department of Biomedical Sciences of Cells and SystemsSection Molecular Cell BiologyUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| |
Collapse
|
32
|
Rivero-Ríos P, Romo-Lozano M, Fasiczka R, Naaldijk Y, Hilfiker S. LRRK2-Related Parkinson's Disease Due to Altered Endolysosomal Biology With Variable Lewy Body Pathology: A Hypothesis. Front Neurosci 2020; 14:556. [PMID: 32581693 PMCID: PMC7287096 DOI: 10.3389/fnins.2020.00556] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/06/2020] [Indexed: 12/14/2022] Open
Abstract
Mutations in the gene encoding for leucine-rich repeat kinase 2 (LRRK2) are associated with both familial and sporadic Parkinson's disease (PD). LRRK2 encodes a large protein comprised of a GTPase and a kinase domain. All pathogenic variants converge on enhancing LRRK2 kinase substrate phosphorylation, and distinct LRRK2 kinase inhibitors are currently in various stages of clinical trials. Although the precise pathophysiological functions of LRRK2 remain largely unknown, PD-associated mutants have been shown to alter various intracellular vesicular trafficking pathways, especially those related to endolysosomal protein degradation events. In addition, biochemical studies have identified a subset of Rab proteins, small GTPases required for all vesicular trafficking steps, as substrate proteins for the LRRK2 kinase activity in vitro and in vivo. Therefore, it is crucial to evaluate the impact of such phosphorylation on neurodegenerative mechanisms underlying LRRK2-related PD, especially with respect to deregulated Rab-mediated endolysosomal membrane trafficking and protein degradation events. Surprisingly, a significant proportion of PD patients due to LRRK2 mutations display neuronal cell loss in the substantia nigra pars compacta in the absence of any apparent α-synuclein-containing Lewy body neuropathology. These findings suggest that endolysosomal alterations mediated by pathogenic LRRK2 per se are not sufficient to cause α-synuclein aggregation. Here, we will review current knowledge about the link between pathogenic LRRK2, Rab protein phosphorylation and endolysosomal trafficking alterations, and we will propose a testable working model whereby LRRK2-related PD may present with variable LB pathology.
Collapse
Affiliation(s)
- Pilar Rivero-Ríos
- Institute of Parasitology and Biomedicine "López-Neyra", Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain.,Life Sciences Institute, University of Michigan, Ann Arbor, MI, United States
| | - María Romo-Lozano
- Institute of Parasitology and Biomedicine "López-Neyra", Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain
| | - Rachel Fasiczka
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Yahaira Naaldijk
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Sabine Hilfiker
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| |
Collapse
|
33
|
Glycoprotein 5 Is Cleaved by Cathepsin E during Porcine Reproductive and Respiratory Syndrome Virus Membrane Fusion. J Virol 2020; 94:JVI.00097-20. [PMID: 32102888 DOI: 10.1128/jvi.00097-20] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 02/24/2020] [Indexed: 12/26/2022] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is a serious viral disease affecting the global swine industry. Its causative agent, PRRS virus (PRRSV), is an enveloped virus, and therefore membrane fusion between its envelope and host cell target membrane is critical for viral infection. Though much research has focused on PRRSV infection, the detailed mechanisms involved in its membrane fusion remain to be elucidated. In the present study, we performed confocal microscopy in combination with a constitutively active (CA) or dominant negative (DN) mutant, specific inhibitors, and small interfering RNAs (siRNAs), as well as multiple other approaches, to explore PRRSV membrane fusion. We first observed that PRRSV membrane fusion occurred in Rab11-recycling endosomes during early infection using labeled virions and subcellular markers. We further demonstrated that low pH and cathepsin E in Rab11-recycling endosomes are critical for PRRSV membrane fusion. Moreover, PRRSV glycoprotein 5 (GP5) is identified as being cleaved by cathepsin E during this process. Taken together, our findings provide in-depth information regarding PRRSV pathogenesis, which support a novel basis for the development of antiviral drugs and vaccines.IMPORTANCE PRRS, caused by PRRSV, is an economically critical factor in pig farming worldwide. As PRRSV is a lipid membrane-wrapped virus, merging of the PRRSV envelope with the host cell membrane is indispensable for viral infection. However, there is a lack of knowledge on its membrane fusion. Here, we first explored when and where PRRSV membrane fusion occurs. Furthermore, we determined which host cell factors were involved in the process. Importantly, PRRSV GP5 is shown to be cleaved by cathepsin E during membrane fusion. Our work not only provides information on PRRSV membrane fusion for the first time but also deepens our understanding of the molecular mechanisms of PRRSV infection, which provides a foundation for future applications in the prevention and control of PRRS.
Collapse
|
34
|
Colaco A, Kaya E, Adriaenssens E, Davis LC, Zampieri S, Fernández‐Suárez ME, Tan CY, Deegan PB, Porter FD, Galione A, Bembi B, Dardis A, Platt FM. Mechanistic convergence and shared therapeutic targets in Niemann-Pick disease. J Inherit Metab Dis 2020; 43:574-585. [PMID: 31707734 PMCID: PMC7317544 DOI: 10.1002/jimd.12191] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/31/2019] [Accepted: 11/08/2019] [Indexed: 01/07/2023]
Abstract
Niemann-Pick disease type C (NPC) and Tangier disease are genetically and clinically distinct rare inborn errors of metabolism. NPC is caused by defects in either NPC1 or NPC2; whereas Tangier disease is caused by a defect in ABCA1. Tangier disease is currently without therapy, whereas NPC can be treated with miglustat, a small molecule inhibitor of glycosphingolipid biosynthesis that slows the neurological course of the disease. When a Tangier disease patient was misdiagnosed with NPC and treated with miglustat, her symptoms improved. This prompted us to consider whether there is mechanistic convergence between these two apparently unrelated rare inherited metabolic diseases. In this study, we found that when ABCA1 is defective (Tangier disease) there is secondary inhibition of the NPC disease pathway, linking these two diseases at the level of cellular pathophysiology. In addition, this study further supports the hypothesis that miglustat, as well as other substrate reduction therapies, may be potential therapeutic agents for treating Tangier disease as fibroblasts from multiple Tangier patients were corrected by miglustat treatment.
Collapse
Affiliation(s)
| | - Ecem Kaya
- Department of PharmacologyUniversity of OxfordOxfordUK
| | | | | | | | | | - Chong Y. Tan
- Lysosomal Disorders UnitAddenbrooke's HospitalCambridgeUK
| | | | - Forbes D. Porter
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIHBethesdaMaryland
| | | | - Bruno Bembi
- University Hospital Santa Maria della MisericordiaUdineItaly
| | - Andrea Dardis
- University Hospital Santa Maria della MisericordiaUdineItaly
| | | |
Collapse
|
35
|
Duan X, Li Y, Yi K, Guo F, Wang H, Wu PH, Yang J, Mair DB, Morales EA, Kalab P, Wirtz D, Sun SX, Li R. Dynamic organelle distribution initiates actin-based spindle migration in mouse oocytes. Nat Commun 2020; 11:277. [PMID: 31937754 PMCID: PMC6959240 DOI: 10.1038/s41467-019-14068-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 12/05/2019] [Indexed: 12/26/2022] Open
Abstract
Migration of meiosis-I (MI) spindle from the cell center to a sub-cortical location is a critical step for mouse oocytes to undergo asymmetric meiotic cell division. In this study, we investigate the mechanism by which formin-2 (FMN2) orchestrates the initial movement of MI spindle. By defining protein domains responsible for targeting FMN2, we show that spindle-periphery localized FMN2 is required for spindle migration. The spindle-peripheral FMN2 nucleates short actin bundles from vesicles derived likely from the endoplasmic reticulum (ER) and concentrated in a layer outside the spindle. This layer is in turn surrounded by mitochondria. A model based on polymerizing actin filaments pushing against mitochondria, thus generating a counter force on the spindle, demonstrated an inherent ability of this system to break symmetry and evolve directional spindle motion. The model is further supported through experiments involving spatially biasing actin nucleation via optogenetics and disruption of mitochondrial distribution and dynamics. Mammalian oocytes divide asymmetrically during meiotic maturation. Here, the authors show that spindle movement away from oocyte center depends on actin filaments nucleated from the spindle periphery pushing against surrounding mitochondria, which polarizes spontaneously to produce directional spindle motion.
Collapse
Affiliation(s)
- Xing Duan
- Center for Cell Dynamics, Department of Cell Biology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD, 21205, USA.,Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Yizeng Li
- Department of Mechanical Engineering, Kennesaw State University, Marietta, GA, 30060, USA.,Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Kexi Yi
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO, 64110, USA
| | - Fengli Guo
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO, 64110, USA
| | - HaiYang Wang
- Center for Cell Dynamics, Department of Cell Biology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD, 21205, USA.,Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Pei-Hsun Wu
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Jing Yang
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Devin B Mair
- Center for Cell Dynamics, Department of Cell Biology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD, 21205, USA.,Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Edwin Angelo Morales
- Center for Cell Dynamics, Department of Cell Biology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD, 21205, USA
| | - Petr Kalab
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Denis Wirtz
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Sean X Sun
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Rong Li
- Center for Cell Dynamics, Department of Cell Biology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD, 21205, USA. .,Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA. .,Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, Singapore, 117411, Singapore.
| |
Collapse
|
36
|
Morad G, Carman CV, Hagedorn EJ, Perlin JR, Zon LI, Mustafaoglu N, Park TE, Ingber DE, Daisy CC, Moses MA. Tumor-Derived Extracellular Vesicles Breach the Intact Blood-Brain Barrier via Transcytosis. ACS NANO 2019; 13:13853-13865. [PMID: 31479239 PMCID: PMC7169949 DOI: 10.1021/acsnano.9b04397] [Citation(s) in RCA: 382] [Impact Index Per Article: 63.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The restrictive nature of the blood-brain barrier (BBB) creates a major challenge for brain drug delivery with current nanomedicines lacking the ability to cross the BBB. Extracellular vesicles (EVs) have been shown to contribute to the progression of a variety of brain diseases including metastatic brain cancer and have been suggested as promising therapeutics and drug delivery vehicles. However, the ability of native tumor-derived EVs to breach the BBB and the mechanism(s) involved in this process remain unknown. Here, we demonstrate that tumor-derived EVs can breach the intact BBB in vivo, and by using state-of-the-art in vitro and in vivo models of the BBB, we have identified transcytosis as the mechanism underlying this process. Moreover, high spatiotemporal resolution microscopy demonstrated that the endothelial recycling endocytic pathway is involved in this transcellular transport. We further identify and characterize the mechanism by which tumor-derived EVs circumvent the low physiologic rate of transcytosis in the BBB by decreasing the brain endothelial expression of rab7 and increasing the efficiency of their transport. These findings identify previously unknown mechanisms by which tumor-derived EVs breach an intact BBB during the course of brain metastasis and can be leveraged to guide and inform the development of drug delivery approaches to deliver therapeutic cargoes across the BBB for treatment of a variety of brain diseases including, but not limited to, brain malignancies.
Collapse
Affiliation(s)
- Golnaz Morad
- Vascular Biology Program, Boston Children’s Hospital, Boston, Massachusetts 02115, United States
- Department of Surgery, Harvard Medical School, Boston, Massachusetts 02115, United States
- Graduate School of Arts and Sciences, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Christopher V. Carman
- Molecular and Integrative Physiological Sciences Program, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, United States
| | - Elliott J. Hagedorn
- Stem Cell Program and Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts 02115, United States
- Department of Stem Cell and Regenerative Biology and Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts 02115, United States
| | - Julie R. Perlin
- Stem Cell Program and Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts 02115, United States
- Department of Stem Cell and Regenerative Biology and Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts 02115, United States
| | - Leonard I. Zon
- Stem Cell Program and Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts 02115, United States
- Department of Stem Cell and Regenerative Biology and Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts 02115, United States
| | - Nur Mustafaoglu
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, United States
| | - Tae-Eun Park
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, United States
- Ulsan National Institute of Science and Technology, Ulsan 44919, South Korea
| | - Donald E. Ingber
- Vascular Biology Program, Boston Children’s Hospital, Boston, Massachusetts 02115, United States
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, United States
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Cassandra C. Daisy
- Vascular Biology Program, Boston Children’s Hospital, Boston, Massachusetts 02115, United States
| | - Marsha A. Moses
- Vascular Biology Program, Boston Children’s Hospital, Boston, Massachusetts 02115, United States
- Department of Surgery, Harvard Medical School, Boston, Massachusetts 02115, United States
- Department of Surgery, Boston Children’s Hospital, Boston, Massachusetts 02115, United States
- Corresponding Author:
| |
Collapse
|
37
|
Gabas IM, Nielsen PE. Effective Cellular Delivery of Antisense Peptide Nucleic Acid by Conjugation to Guanidinylated Diaminobutanoic Acid-Based Peptide Dendrons. Biomacromolecules 2019; 21:472-483. [PMID: 31756087 DOI: 10.1021/acs.biomac.9b01227] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A series of amino- and guanidino-terminating 3- and 4-generation 2,4-diaminobutanoic acid (Dab) dendrons have been robustly synthesized on a solid phase and characterized as cellular delivery agents in antisense peptide nucleic acid (PNA) conjugates in the pLuc705 HeLa cell splice switching system. The dendron-PNA conjugates exhibited splice correction activity at one digit micromolar concentrations, and guanidino-terminating dendrons were significantly more effective than analogous amine terminating ones. Furthermore, introduction of lipophilic groups such as phenyl, alkyl, or fatty acids increased efficacy, but also increased cellular toxicity. Fluorescence microscopy analyses supported an endosomal uptake mechanism and furthermore predominantly showed colocalization with late endosomes and lysosomes. The robust solid phase synthesis should make such Dab-dendrons a useful platform for further in vitro as well as in vivo optimization.
Collapse
Affiliation(s)
- Isabel Maicas Gabas
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, The Panum Institute , University of Copenhagen , Blegdamsvej 3 , 2200 Copenhagen , Denmark
| | - Peter E Nielsen
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, The Panum Institute , University of Copenhagen , Blegdamsvej 3 , 2200 Copenhagen , Denmark
| |
Collapse
|
38
|
de Araujo MEG, Liebscher G, Hess MW, Huber LA. Lysosomal size matters. Traffic 2019; 21:60-75. [PMID: 31808235 PMCID: PMC6972631 DOI: 10.1111/tra.12714] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/30/2019] [Accepted: 10/31/2019] [Indexed: 12/25/2022]
Abstract
Lysosomes are key cellular catabolic centers that also perform fundamental metabolic, signaling and quality control functions. Lysosomes are not static and they respond dynamically to intra‐ and extracellular stimuli triggering changes in organelle numbers, size and position. Such physical changes have a strong impact on lysosomal activity ultimately influencing cellular homeostasis. In this review, we summarize the current knowledge on lysosomal size regulation, on its physiological role(s) and association to several disease conditions.
Collapse
Affiliation(s)
- Mariana E G de Araujo
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Gudrun Liebscher
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Michael W Hess
- Institute of Histology and Embryology, Medical University of Innsbruck, Innsbruck, Austria
| | - Lukas A Huber
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria.,Austrian Drug Screening Institute, ADSI, Innsbruck, Austria
| |
Collapse
|
39
|
Leng C, Overeem AW, Cartón-Garcia F, Li Q, Klappe K, Kuipers J, Cui Y, Zuhorn IS, Arango D, van IJzendoorn SCD. Loss of MYO5B expression deregulates late endosome size which hinders mitotic spindle orientation. PLoS Biol 2019; 17:e3000531. [PMID: 31682603 PMCID: PMC6855566 DOI: 10.1371/journal.pbio.3000531] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 11/14/2019] [Accepted: 10/17/2019] [Indexed: 12/23/2022] Open
Abstract
Recycling endosomes regulate plasma membrane recycling. Recently, recycling endosome–associated proteins have been implicated in the positioning and orientation of the mitotic spindle and cytokinesis. Loss of MYO5B, encoding the recycling endosome–associated myosin Vb, is associated with tumor development and tissue architecture defects in the gastrointestinal tract. Whether loss of MYO5B expression affects mitosis is not known. Here, we demonstrate that loss of MYO5B expression delayed cytokinesis, perturbed mitotic spindle orientation, led to the misorientation of the plane of cell division during the course of mitosis, and resulted in the delamination of epithelial cells. Remarkably, the effects on spindle orientation, but not cytokinesis, were a direct consequence of physical hindrance by giant late endosomes, which were formed in a chloride channel–sensitive manner concomitant with a redistribution of chloride channels from the cell periphery to late endosomes upon loss of MYO5B. Rab7 availability was identified as a limiting factor for the development of giant late endosomes. In accordance, increasing rab7 availability corrected mitotic spindle misorientation and cell delamination in cells lacking MYO5B expression. In conclusion, we identified a novel role for MYO5B in the regulation of late endosome size control and identify the inability to control late endosome size as an unexpected novel mechanism underlying defects in cell division orientation and epithelial architecture. Loss of the recycling endosome-associated motor protein myosin Vb causes the formation of giant late endo-lysosomes; these in turn hinder the orientation of the mitotic spindle and chromosome segregation. Deregulated endosome size thus hampers faithful cell division.
Collapse
Affiliation(s)
- Changsen Leng
- Department of Biomedical Sciences of Cells and Systems, section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Arend W. Overeem
- Department of Biomedical Sciences of Cells and Systems, section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Fernando Cartón-Garcia
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Qinghong Li
- Department of Biomedical Sciences of Cells and Systems, section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Karin Klappe
- Department of Biomedical Sciences of Cells and Systems, section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Jeroen Kuipers
- Department of Biomedical Sciences of Cells and Systems, section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Yingying Cui
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Inge S. Zuhorn
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Diego Arango
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Sven C. D. van IJzendoorn
- Department of Biomedical Sciences of Cells and Systems, section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- * E-mail:
| |
Collapse
|
40
|
Enforced expression of phosphatidylinositol 4-phosphate 5-kinase homolog alters PtdIns(4,5)P 2 distribution and the localization of small G-proteins. Sci Rep 2019; 9:14789. [PMID: 31616009 PMCID: PMC6794296 DOI: 10.1038/s41598-019-51272-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 09/20/2019] [Indexed: 02/02/2023] Open
Abstract
The generation of phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P2) by phosphatidylinositol 4-phosphate 5-kinases (PIP5Ks) is essential for many functions including control of the cytoskeleton, signal transduction, and endocytosis. Due to its presence in the plasma membrane and anionic charge, PtdIns(4,5)P2, together with phosphatidylserine, provide the inner leaflet of the plasma membrane with a negative surface charge. This negative charge helps to define the identity of the plasma membrane, as it serves to recruit or regulate a multitude of peripheral and membrane proteins that contain polybasic domains or patches. Here, we determine that the phosphatidylinositol 4-phosphate 5-kinase homolog (PIPKH) alters the subcellular distribution of PtdIns(4,5)P2 by re-localizing the three PIP5Ks to endomembranes. We find a redistribution of the PIP5K family members to endomembrane structures upon PIPKH overexpression that is accompanied by accumulation of PtdIns(4,5)P2 and phosphatidylinositol 3,4,5-trisphosphate (PtdIns(3,4,5)P3). PIP5Ks are targeted to membranes in part due to electrostatic interactions; however, the interaction between PIPKH and PIP5K is maintained following hydrolysis of PtdIns(4,5)P2. Expression of PIPKH did not impair bulk endocytosis as monitored by FM4-64 uptake but did result in clustering of FM4-64 positive endosomes. Finally, we demonstrate that accumulation of polyphosphoinositides increases the negative surface charge of endosomes and in turn, leads to relocalization of surface charge probes as well as the polycationic proteins K-Ras and Rac1.
Collapse
|
41
|
Akin EJ, Higerd GP, Mis MA, Tanaka BS, Adi T, Liu S, Dib-Hajj FB, Waxman SG, Dib-Hajj SD. Building sensory axons: Delivery and distribution of Na V1.7 channels and effects of inflammatory mediators. SCIENCE ADVANCES 2019; 5:eaax4755. [PMID: 31681845 PMCID: PMC6810356 DOI: 10.1126/sciadv.aax4755] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 09/13/2019] [Indexed: 05/12/2023]
Abstract
Sodium channel NaV1.7 controls firing of nociceptors, and its role in human pain has been validated by genetic and functional studies. However, little is known about NaV1.7 trafficking or membrane distribution along sensory axons, which can be a meter or more in length. We show here with single-molecule resolution the first live visualization of NaV1.7 channels in dorsal root ganglia neurons, including long-distance microtubule-dependent vesicular transport in Rab6A-containing vesicles. We demonstrate nanoclusters that contain a median of 12.5 channels at the plasma membrane on axon termini. We also demonstrate that inflammatory mediators trigger an increase in the number of NaV1.7-carrying vesicles per axon, a threefold increase in the median number of NaV1.7 channels per vesicle and a ~50% increase in forward velocity. This remarkable enhancement of NaV1.7 vesicular trafficking and surface delivery under conditions that mimic a disease state provides new insights into the contribution of NaV1.7 to inflammatory pain.
Collapse
Affiliation(s)
- Elizabeth J. Akin
- Department of Neurology, Yale University, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Grant P. Higerd
- Department of Neurology, Yale University, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
- MD-PhD Program, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Malgorzata A. Mis
- Department of Neurology, Yale University, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Brian S. Tanaka
- Department of Neurology, Yale University, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Talia Adi
- Department of Neurology, Yale University, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Shujun Liu
- Department of Neurology, Yale University, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Fadia B. Dib-Hajj
- Department of Neurology, Yale University, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Stephen G. Waxman
- Department of Neurology, Yale University, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
- Corresponding author. (S.D.D.-H.); (S.G.W.)
| | - Sulayman D. Dib-Hajj
- Department of Neurology, Yale University, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
- Corresponding author. (S.D.D.-H.); (S.G.W.)
| |
Collapse
|
42
|
Degors IS, Wang C, Rehman ZU, Zuhorn IS. Carriers Break Barriers in Drug Delivery: Endocytosis and Endosomal Escape of Gene Delivery Vectors. Acc Chem Res 2019; 52:1750-1760. [PMID: 31243966 PMCID: PMC6639780 DOI: 10.1021/acs.accounts.9b00177] [Citation(s) in RCA: 269] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Indexed: 12/31/2022]
Abstract
Over the past decades, major efforts were undertaken to develop devices on a nanoscale level for the efficient and nontoxic delivery of molecules to tissues and cells, for the purpose of either diagnosis or treatment of disease. The application of such devices in drug delivery has proven to be beneficial for matters as diverse as drug solubility, drug targeting, controlled drug release, and transport of drugs across cellular barriers. Multiple nanotherapeutics have been approved for clinical treatment, and more products are being evaluated in preclinical and clinical trials. However, many biological barriers hinder the medical application of nanocarriers. There are two main classes of barriers that need to be overcome by drug nanocarriers: extracellular and intracellular barriers, both of which may capture and/or destroy therapeutics before they reach their target site. This Account discusses major biological barriers that are confronted by nanotherapeutics, following their systemic administration, focusing on cellular entry and endosomal escape of gene delivery vectors. The use of pH-responsive materials to overcome the endosomal barrier is addressed. Historically, cell biologists have studied the interaction between cells and pathogens in order to unveil the mechanisms of endocytosis and cell signaling. Meanwhile, it is becoming clear that cells may respond in similar ways to artificial drug delivery systems and, consequently, that knowledge on the cellular response against both pathogens and nanoparticulate systems will aid in the design of improved nanomedicine. A close collaboration between bioengineers and cell biologists will promote this development. At the same time, we have come to realize that tools that we use to study fundamental cellular processes, including metabolic inhibitors of endocytosis and overexpression/downregulation of proteins, may cause changes in cellular physiology. This calls for the implementation of refined methods to study nanocarrier-cell interactions, as is discussed in this Account. Finally, recent papers on the dynamics of cargo release from endosomes by means of live cell imaging have significantly advanced our understanding of the transfection process. They have initiated discussion (among others) on the limited number of endosomal escape events in transfection, and on the endosomal stage at which genetic cargo is most efficiently released. Advancements in imaging techniques, including super-resolution microscopy, in concert with techniques to label endogenous proteins and/or label proteins with synthetic fluorophores, will contribute to a more detailed understanding of nanocarrier-cell dynamics, which is imperative for the development of safe and efficient nanomedicine.
Collapse
Affiliation(s)
- Isabelle
M. S. Degors
- Department
of Biomedical Engineering, University Medical
Center Groningen, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Cuifeng Wang
- School
of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of
New Drug Design and Evaluation, Sun Yat-sen
University, Guangzhou 510006, P. R. China
| | - Zia Ur Rehman
- Department
of Biotechnology and Genetic Engineering, Kohat University of Sciences and Technology (KUST), Kohat 26000, Khyber Pakhtunkhwa, Pakistan
| | - Inge S. Zuhorn
- Department
of Biomedical Engineering, University Medical
Center Groningen, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| |
Collapse
|
43
|
Tower-Gilchrist C, Zlatic SA, Yu D, Chang Q, Wu H, Lin X, Faundez V, Chen P. Adaptor protein-3 complex is required for Vangl2 trafficking and planar cell polarity of the inner ear. Mol Biol Cell 2019; 30:2422-2434. [PMID: 31268833 PMCID: PMC6741063 DOI: 10.1091/mbc.e16-08-0592] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Planar cell polarity (PCP) regulates coordinated cellular polarity among neighboring cells to establish a polarity axis parallel to the plane of the tissue. Disruption in PCP results in a range of developmental anomalies and diseases. A key feature of PCP is the polarized and asymmetric localization of several membrane PCP proteins, which is essential to establish the polarity axis to orient cells coordinately. However, the machinery that regulates the asymmetric partition of PCP proteins remains largely unknown. In the present study, we show Van gogh-like 2 (Vangl2) in early and recycling endosomes as made evident by colocalization with diverse endosomal Rab proteins. Vangl2 biochemically interacts with adaptor protein-3 complex (AP-3). Using short hairpin RNA knockdown, we found that Vangl2 subcellular localization was modified in AP-3–depleted cells. Moreover, Vangl2 membrane localization within the cochlea is greatly reduced in AP-3–deficient mocha mice, which exhibit profound hearing loss. In inner ears from AP-3–deficient mocha mice, we observed PCP-dependent phenotypes, such as misorientation and deformation of hair cell stereociliary bundles and disorganization of hair cells characteristic of defects in convergent extension that is driven by PCP. These findings demonstrate a novel role of AP-3–mediated sorting mechanisms in regulating PCP proteins.
Collapse
Affiliation(s)
| | - Stephanie A Zlatic
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322
| | - Dehong Yu
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322.,Department of Otolaryngology-Head and Neck Surgery, Xinhua Hospital and Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai 200125, China
| | - Qing Chang
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322.,Department of Otolaryngology, Emory University School of Medicine, Atlanta, GA 30322
| | - Hao Wu
- Department of Otolaryngology-Head and Neck Surgery, Xinhua Hospital and Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai 200125, China
| | - Xi Lin
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322.,Department of Otolaryngology, Emory University School of Medicine, Atlanta, GA 30322
| | - Victor Faundez
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322
| | - Ping Chen
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322
| |
Collapse
|
44
|
Zunke F, Mazzulli JR. Modeling neuronopathic storage diseases with patient-derived culture systems. Neurobiol Dis 2019; 127:147-162. [PMID: 30790616 PMCID: PMC6588474 DOI: 10.1016/j.nbd.2019.01.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/23/2019] [Accepted: 01/25/2019] [Indexed: 02/08/2023] Open
Abstract
Lysosomes are organelles involved in the degradation and recycling of macromolecules, and play a critical role in sensing metabolic information in the cell. A class of rare metabolic diseases called lysosomal storage disorders (LSD) are characterized by lysosomal dysfunction and the accumulation of macromolecular substrates. The central nervous system appears to be particularly vulnerable to lysosomal dysfunction, since many LSDs are characterized by severe, widespread neurodegeneration with pediatric onset. Furthermore, variants in lysosomal genes are strongly associated with some common neurodegenerative disorders such as Parkinson's disease (PD). To better understand disease pathology and develop novel treatment strategies, it is critical to study the fundamental molecular disease mechanisms in the affected cell types that harbor endogenously expressed mutations. The discovery of methods for reprogramming of patient-derived somatic cells into induced pluripotent stem cells (iPSCs), and their differentiation into distinct neuronal and glial cell types, have provided novel opportunities to study mechanisms of lysosomal dysfunction within the relevant, vulnerable cell types. These models also expand our ability to develop and test novel therapeutic targets. We discuss recently developed methods for iPSC differentiation into distinct neuronal and glial cell types, while addressing the need for meticulous experimental techniques and parameters that are essential to accurately identify inherent cellular pathologies. iPSC models for neuronopathic LSDs and their relationship to sporadic age-related neurodegeneration are also discussed. These models should facilitate the discovery and development of personalized therapies in the future.
Collapse
Affiliation(s)
- Friederike Zunke
- Institute of Biochemistry, Christian-Albrechts-Universität zu Kiel, Kiel 24118, Germany.
| | - Joseph R Mazzulli
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
45
|
León M, Ferreira CR, Eberlin LS, Jarmusch AK, Pirro V, Rodrigues ACB, Favaron PO, Miglino MA, Cooks RG. Metabolites and Lipids Associated with Fetal Swine Anatomy via Desorption Electrospray Ionization - Mass Spectrometry Imaging. Sci Rep 2019; 9:7247. [PMID: 31076607 PMCID: PMC6510765 DOI: 10.1038/s41598-019-43698-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 04/27/2019] [Indexed: 12/11/2022] Open
Abstract
Chemical imaging by mass spectrometry (MS) has been largely used to study diseases in animals and humans, especially cancer; however, this technology has been minimally explored to study the complex chemical changes associated with fetal development. In this work, we report the histologically-compatible chemical imaging of small molecules by desorption electrospray ionization (DESI) - MS of a complete swine fetus at 50 days of gestation. Tissue morphology was unperturbed by morphologically-friendly DESI-MS analysis while allowing detection of a wide range of small molecules. We observed organ-dependent localization of lipids, e.g. a large diversity of phosphatidylserine lipids in brain compared to other organs, as well as metabolites such as N-acetyl-aspartic acid in the developing nervous system and N-acetyl-L-glutamine in the heart. Some lipids abundant in the lungs, such as PC(32:0) and PS(40:6), were similar to surfactant composition reported previously. Sulfatides were highly concentrated in the fetus liver, while hexoses were barely detected at this organ but were abundant in lung and heart. The chemical information on small molecules recorded via DESI-MS imaging coupled with traditional anatomical evaluation is a powerful source of bioanalytical information which reveals the chemical changes associated with embryonic and fetal development that, when disturbed, causes congenital diseases such as spina bifida and cleft palate.
Collapse
Affiliation(s)
- Marisol León
- Surgery Department, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil
| | - Christina R Ferreira
- Department of Chemistry and Center for Analytical Instrumentation Development, Purdue University, West Lafayette, IN, 47907, United States
| | - Livia S Eberlin
- Department of Chemistry, The University of Texas at Austin, Austin, TX, 78712, United States
| | - Alan K Jarmusch
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, 92093, United States
| | - Valentina Pirro
- Department of Chemistry and Center for Analytical Instrumentation Development, Purdue University, West Lafayette, IN, 47907, United States
| | - Ana Clara Bastos Rodrigues
- Surgery Department, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil
| | | | - Maria Angelica Miglino
- Surgery Department, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil
| | - R Graham Cooks
- Department of Chemistry and Center for Analytical Instrumentation Development, Purdue University, West Lafayette, IN, 47907, United States.
| |
Collapse
|
46
|
Williams ET, Glauser L, Tsika E, Jiang H, Islam S, Moore DJ. Parkin mediates the ubiquitination of VPS35 and modulates retromer-dependent endosomal sorting. Hum Mol Genet 2019; 27:3189-3205. [PMID: 29893854 DOI: 10.1093/hmg/ddy224] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/04/2018] [Indexed: 12/27/2022] Open
Abstract
Mutations in a number of genes cause familial forms of Parkinson's disease (PD), including mutations in the vacuolar protein sorting 35 ortholog (VPS35) and parkin genes. In this study, we identify a novel functional interaction between parkin and VPS35. We demonstrate that parkin interacts with and robustly ubiquitinates VPS35 in human neural cells. Familial parkin mutations are impaired in their ability to ubiquitinate VPS35. Parkin mediates the attachment of an atypical poly-ubiquitin chain to VPS35 with three lysine residues identified within the C-terminal region of VPS35 that are covalently modified by ubiquitin. Notably, parkin-mediated VPS35 ubiquitination does not promote the proteasomal degradation of VPS35. Furthermore, parkin does not influence the steady-state levels or turnover of VPS35 in neural cells and VPS35 levels are normal in the brains of parkin knockout mice. These data suggest that ubiquitination of VPS35 by parkin may instead serve a non-degradative cellular function potentially by regulating retromer-dependent sorting. Accordingly, we find that components of the retromer-associated WASH complex are markedly decreased in the brain of parkin knockout mice, suggesting that parkin may modulate WASH complex-dependent retromer sorting. Parkin gene silencing in primary cortical neurons selectively disrupts the vesicular sorting of the autophagy receptor ATG9A, a WASH-dependent retromer cargo. Parkin is not required for dopaminergic neurodegeneration induced by the expression of PD-linked D620N VPS35 in mice, consistent with VPS35 being located downstream of parkin function. Our data reveal a novel functional interaction of parkin with VPS35 that may be important for retromer-mediated endosomal sorting and PD.
Collapse
Affiliation(s)
- Erin T Williams
- Van Andel Institute Graduate School, Van Andel Research Institute, Grand Rapids, MI, USA.,Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Liliane Glauser
- Brain Mind Institute, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Elpida Tsika
- Brain Mind Institute, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,AC Immune SA, EPFL Innovation Park, Lausanne, Switzerland
| | - Haisong Jiang
- Institute for Cell Engineering and Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shariful Islam
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Darren J Moore
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| |
Collapse
|
47
|
Levanova N, Mattheis C, Carson D, To KN, Jank T, Frankel G, Aktories K, Schroeder GN. The Legionella effector LtpM is a new type of phosphoinositide-activated glucosyltransferase. J Biol Chem 2019; 294:2862-2879. [PMID: 30573678 PMCID: PMC6393602 DOI: 10.1074/jbc.ra118.005952] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 12/13/2018] [Indexed: 01/01/2023] Open
Abstract
Legionella pneumophila causes Legionnaires' disease, a severe form of pneumonia. L. pneumophila translocates more than 300 effectors into host cells via its Dot/Icm (Defective in organelle trafficking/Intracellular multiplication) type IV secretion system to enable its replication in target cells. Here, we studied the effector LtpM, which is encoded in a recombination hot spot in L. pneumophila Paris. We show that a C-terminal phosphoinositol 3-phosphate (PI3P)-binding domain, also found in otherwise unrelated effectors, targets LtpM to the Legionella-containing vacuole and to early and late endosomes. LtpM expression in yeast caused cytotoxicity. Sequence comparison and structural homology modeling of the N-terminal domain of LtpM uncovered a remote similarity to the glycosyltransferase (GT) toxin PaTox from the bacterium Photorhabdus asymbiotica; however, instead of the canonical DxD motif of GT-A type glycosyltransferases, essential for enzyme activity and divalent cation coordination, we found that a DxN motif is present in LtpM. Using UDP-glucose as sugar donor, we show that purified LtpM nevertheless exhibits glucohydrolase and autoglucosylation activity in vitro and demonstrate that PI3P binding activates LtpM's glucosyltransferase activity toward protein substrates. Substitution of the aspartate or the asparagine in the DxN motif abolished the activity of LtpM. Moreover, whereas all glycosyltransferase toxins and effectors identified so far depend on the presence of divalent cations, LtpM is active in their absence. Proteins containing LtpM-like GT domains are encoded in the genomes of other L. pneumophila isolates and species, suggesting that LtpM is the first member of a novel family of glycosyltransferase effectors employed to subvert hosts.
Collapse
Affiliation(s)
- Nadezhda Levanova
- From the Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, D-79104 Freiburg, Germany
| | - Corinna Mattheis
- the MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom, and
| | - Danielle Carson
- the MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom, and
| | - Ka-Ning To
- the MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom, and
| | - Thomas Jank
- From the Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, D-79104 Freiburg, Germany
| | - Gad Frankel
- the MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom, and
| | - Klaus Aktories
- From the Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, D-79104 Freiburg, Germany,
| | - Gunnar Neels Schroeder
- the MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom, and
- the Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7BL, Northern Ireland, United Kingdom
| |
Collapse
|
48
|
Wheeler S, Schmid R, Sillence DJ. Lipid⁻Protein Interactions in Niemann⁻Pick Type C Disease: Insights from Molecular Modeling. Int J Mol Sci 2019; 20:E717. [PMID: 30736449 PMCID: PMC6387118 DOI: 10.3390/ijms20030717] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 01/31/2019] [Accepted: 02/03/2019] [Indexed: 12/19/2022] Open
Abstract
The accumulation of lipids in the late endosomes and lysosomes of Niemann⁻Pick type C disease (NPCD) cells is a consequence of the dysfunction of one protein (usually NPC1) but induces dysfunction in many proteins. We used molecular docking to propose (a) that NPC1 exports not just cholesterol, but also sphingosine, (b) that the cholesterol sensitivity of big potassium channel (BK) can be traced to a previously unappreciated site on the channel's voltage sensor, (c) that transient receptor potential mucolipin 1 (TRPML1) inhibition by sphingomyelin is likely an indirect effect, and (d) that phosphoinositides are responsible for both the mislocalization of annexin A2 (AnxA2) and a soluble NSF (N-ethylmaleimide Sensitive Fusion) protein attachment receptor (SNARE) recycling defect. These results are set in the context of existing knowledge of NPCD to sketch an account of the endolysosomal pathology key to this disease.
Collapse
Affiliation(s)
- Simon Wheeler
- School of Pharmacy, De Montfort University, The Gateway, Leicester LE1 9BH, UK.
| | - Ralf Schmid
- Leicester Institute of Structural and Chemical Biology, Henry Wellcome Building, University of Leicester, Lancaster Road, Leicester LE1 7RH, UK.
| | - Dan J Sillence
- School of Pharmacy, De Montfort University, The Gateway, Leicester LE1 9BH, UK.
| |
Collapse
|
49
|
Rivero-Ríos P, Romo-Lozano M, Madero-Pérez J, Thomas AP, Biosa A, Greggio E, Hilfiker S. The G2019S variant of leucine-rich repeat kinase 2 (LRRK2) alters endolysosomal trafficking by impairing the function of the GTPase RAB8A. J Biol Chem 2019; 294:4738-4758. [PMID: 30709905 PMCID: PMC6442034 DOI: 10.1074/jbc.ra118.005008] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 01/30/2019] [Indexed: 12/25/2022] Open
Abstract
Mutations in the gene encoding for leucine-rich repeat kinase 2 (LRRK2) are a common cause of hereditary Parkinson's disease. LRRK2 regulates various intracellular vesicular trafficking pathways, including endolysosomal degradative events such as epidermal growth factor receptor (EGFR) degradation. Recent studies have revealed that a subset of RAB proteins involved in secretory and endocytic recycling are LRRK2 kinase substrates in vivo. However, the effects of LRRK2-mediated phosphorylation of these substrates on membrane trafficking remain unknown. Here, using an array of immunofluorescence and pulldown assays, we report that expression of active or phosphodeficient RAB8A variants rescues the G2019S LRRK2–mediated effects on endolysosomal membrane trafficking. Similarly, up-regulation of the RAB11–Rabin8–RAB8A cascade, which activates RAB8A, also reverted these trafficking deficits. Loss of RAB8A mimicked the effects of G2019S LRRK2 on endolysosomal trafficking and decreased RAB7A activity. Expression of pathogenic G2019S LRRK2 or loss of RAB8A interfered with EGFR degradation by causing its accumulation in a RAB4-positive endocytic compartment, which was accompanied by a deficit in EGFR recycling and was rescued upon expression of active RAB7A. Dominant-negative RAB7A expression resulted in similar deficits in EGF degradation, accumulation in a RAB4 compartment, and deficits in EGFR recycling, which were all rescued upon expression of active RAB8A. Taken together, these findings suggest that, by impairing RAB8A function, pathogenic G2019S LRRK2 deregulates endolysosomal transport and endocytic recycling events.
Collapse
Affiliation(s)
- Pilar Rivero-Ríos
- From the Institute of Parasitology and Biomedicine "López-Neyra," Consejo Superior de Investigaciones Científicas (CSIC), Avda del Conocimiento s/n, 18016 Granada, Spain
| | - María Romo-Lozano
- From the Institute of Parasitology and Biomedicine "López-Neyra," Consejo Superior de Investigaciones Científicas (CSIC), Avda del Conocimiento s/n, 18016 Granada, Spain
| | - Jesús Madero-Pérez
- From the Institute of Parasitology and Biomedicine "López-Neyra," Consejo Superior de Investigaciones Científicas (CSIC), Avda del Conocimiento s/n, 18016 Granada, Spain
| | - Andrew P Thomas
- the Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey 07103, and
| | - Alice Biosa
- the Department of Biology, University of Padova, Padova 35121, Italy
| | - Elisa Greggio
- the Department of Biology, University of Padova, Padova 35121, Italy
| | - Sabine Hilfiker
- From the Institute of Parasitology and Biomedicine "López-Neyra," Consejo Superior de Investigaciones Científicas (CSIC), Avda del Conocimiento s/n, 18016 Granada, Spain,
| |
Collapse
|
50
|
Enrich C, Rentero C, Grewal T, Futter CE, Eden ER. Cholesterol Overload: Contact Sites to the Rescue! CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2019; 2:2515256419893507. [PMID: 31858076 PMCID: PMC6923141 DOI: 10.1177/2515256419893507] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Delivery of low-density lipoprotein-derived cholesterol to the endoplasmic reticulum (ER) is essential for cholesterol homeostasis, yet the mechanism of this transport has largely remained elusive. Two recent reports shed some light on this process, uncovering a role for Niemann Pick type-C1 protein (NPC1) in the formation of membrane contact sites (MCS) between late endosomes (LE)/lysosomes (Lys) and the ER. Both studies identified a loss of MCS in cells lacking functional NPC1, where cholesterol accumulates in late endocytic organelles. Remarkably, and taking different approaches, both studies have made a striking observation that expansion of LE/Lys-ER MCS can rescue the cholesterol accumulation phenotype in NPC1 mutant or deficient cells. In both cases, the cholesterol was shown to be transported to the ER, demonstrating the importance of ER-LE/Lys contact sites in the direct transport of low-density lipoprotein-derived cholesterol to the ER.
Collapse
Affiliation(s)
- Carlos Enrich
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Spain
- Centre de Recerca Biomèdica CELLEX, Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Carles Rentero
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Spain
- Centre de Recerca Biomèdica CELLEX, Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Thomas Grewal
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia
| | | | | |
Collapse
|