1
|
Perron T, Boissan M, Bièche I, Courtois L, Dingli F, Loew D, Chouchène M, Colasse S, Levy L, Prunier C. CYYR1 promotes the degradation of the E3 ubiquitin ligase WWP1 and is associated with favorable prognosis in breast cancer. J Biol Chem 2024; 300:107601. [PMID: 39059493 PMCID: PMC11399591 DOI: 10.1016/j.jbc.2024.107601] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/26/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Ubiquitination plays a crucial role in cellular homeostasis by regulating the degradation, localization, and activity of proteins, ensuring proper cell function and balance. Among E3 ubiquitin ligases, WW domain-containing protein 1 (WWP1) is implicated in cell proliferation, survival, and apoptosis. Notably WWP1 is frequently amplified in breast cancer and associated with poor prognosis. Here, we identify the protein cysteine and tyrosine-rich protein 1 (CYYR1) that had previously no assigned function, as a regulator of WWP1 activity and stability. We show that CYYR1 binds to the WW domains of the E3 ubiquitin ligase WWP1 through its PPxY motifs. This interaction triggers K63-linked autoubiquitination and subsequent degradation of WWP1. We furthermore demonstrate that CYYR1 localizes to late endosomal vesicles and directs polyubiquitinated WWP1 toward lysosomal degradation through binding to ANKyrin repeat domain-containing protein 13 A (ANKRD13A). Moreover, we found that CYYR1 expression attenuates breast cancer cell growth in anchorage-dependent and independent colony formation assays in a PPxY-dependent manner. Finally, we highlight that CYYR1 expression is significantly decreased in breast cancer and is associated with beneficial clinical outcome. Taken together our study suggests tumor suppressor properties for CYYR1 through regulation of WWP1 autoubiquitination and lysosomal degradation.
Collapse
Affiliation(s)
- Tiphaine Perron
- Sorbonne Université, INSERM UMR_S 938, Centre de Recherche Saint-Antoine, CRSA, Paris, France
| | - Mathieu Boissan
- Sorbonne Université, INSERM UMR_S 938, Centre de Recherche Saint-Antoine, CRSA, Paris, France; APHP, Hôpitaux Universitaires Pitié-Salpêtrière-Charles Foix, Laboratoire de Biochimie Endocrinienne et Oncologique, Oncobiologie Cellulaire et Moléculaire, Paris, France
| | - Ivan Bièche
- Department of Genetics, Institut Curie, Université Paris Cité, Paris, France
| | - Laura Courtois
- Department of Genetics, Institut Curie, Université Paris Cité, Paris, France
| | - Florent Dingli
- CurieCoreTech Mass Spectrometry Proteomics, Institut Curie, PSL Research University, Paris, France
| | - Damarys Loew
- CurieCoreTech Mass Spectrometry Proteomics, Institut Curie, PSL Research University, Paris, France
| | - Mouna Chouchène
- Sorbonne Université, INSERM UMR_S 938, Centre de Recherche Saint-Antoine, CRSA, Paris, France
| | - Sabrina Colasse
- Sorbonne Université, INSERM UMR_S 938, Centre de Recherche Saint-Antoine, CRSA, Paris, France
| | - Laurence Levy
- Sorbonne Université, INSERM UMR_S 938, Centre de Recherche Saint-Antoine, CRSA, Paris, France.
| | - Céline Prunier
- Sorbonne Université, INSERM UMR_S 938, Centre de Recherche Saint-Antoine, CRSA, Paris, France.
| |
Collapse
|
2
|
Xu D, Li Y, Yin S, Huang F. Strategies to address key challenges of metallacycle/metallacage-based supramolecular coordination complexes in biomedical applications. Chem Soc Rev 2024; 53:3167-3204. [PMID: 38385584 DOI: 10.1039/d3cs00926b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Owing to their capacity for dynamically linking two or more functional molecules, supramolecular coordination complexes (SCCs), exemplified by two-dimensional (2D) metallacycles and three-dimensional (3D) metallacages, have gained increasing significance in biomedical applications. However, their inherent hydrophobicity and self-assembly driven by heavy metal ions present common challenges in their applications. These challenges can be overcome by enhancing the aqueous solubility and in vivo circulation stability of SCCs, alongside minimizing their side effects during treatment. Addressing these challenges is crucial for advancing the fundamental research of SCCs and their subsequent clinical translation. In this review, drawing on extensive contemporary research, we offer a thorough and systematic analysis of the strategies employed by SCCs to surmount these prevalent yet pivotal obstacles. Additionally, we explore further potential challenges and prospects for the broader application of SCCs in the biomedical field.
Collapse
Affiliation(s)
- Dongdong Xu
- Key Laboratory of Organosilicon Chemistry and Materials Technology of Ministry of Education, College of Materials, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, P. R. China.
| | - Yang Li
- Key Laboratory of Organosilicon Chemistry and Materials Technology of Ministry of Education, College of Materials, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, P. R. China.
| | - Shouchun Yin
- Key Laboratory of Organosilicon Chemistry and Materials Technology of Ministry of Education, College of Materials, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, P. R. China.
| | - Feihe Huang
- Stoddart Institute of Molecular Science, Department of Chemistry, Zhejiang University, Hangzhou 310058, P. R. China.
- Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311215, P. R. China
| |
Collapse
|
3
|
Proctor JR, Wong H. Time-dependent clearance can confound exposure-response analysis of therapeutic antibodies: A comprehensive review of the current literature. Clin Transl Sci 2024; 17:e13676. [PMID: 37905360 PMCID: PMC10766027 DOI: 10.1111/cts.13676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/15/2023] [Accepted: 10/23/2023] [Indexed: 11/02/2023] Open
Abstract
Exposure-response (ER) analysis is used to optimize dose and dose regimens during clinical development. Characterization of relationships between drug exposure and efficacy or safety outcomes can be utilized to make dose adjustments that improve patient response. Therapeutic antibodies typically show predictable pharmacokinetics (PK) but can exhibit clearance that decreases over time due to treatment. Moreover, time-dependent changes in clearance are frequently associated with drug response, with larger decreases in clearance and increased exposure seen in patients who respond to treatment. This often confounds traditional ER analysis, as drug response influences exposure rather than the reverse. In this review, we survey published population PK analyses for reported time-dependent drug clearance effects across 158 therapeutic antibodies approved or in regulatory review. We describe the mechanisms by which time-dependent clearance can arise, and evaluate trends in frequency, magnitude, and time scale of changes in clearance with respect to indication, mechanistic interpretation of time-dependence, and PK modeling techniques employed. We discuss the modeling and simulation strategies commonly used to characterize time-dependent clearance, and examples where time-dependent clearance has impeded ER analysis. A case study using population model simulation was explored to interrogate the impact of time-dependent clearance on ER analysis and how it can lead to spurious conclusions. Overall, time-dependent clearance arises frequently among therapeutic antibodies and has spurred erroneous conclusions in ER analysis. Appropriate PK modeling techniques aid in identifying and characterizing temporal shifts in exposure that may impede accurate ER assessment and successful dose optimization.
Collapse
Affiliation(s)
- Jeffrey R. Proctor
- Faculty of Pharmaceutical SciencesThe University of British ColumbiaVancouverBritish ColumbiaCanada
| | - Harvey Wong
- Faculty of Pharmaceutical SciencesThe University of British ColumbiaVancouverBritish ColumbiaCanada
| |
Collapse
|
4
|
Zheng H, Li G, Min J, Xu X, Huang W. Lysosome and related protein degradation technologies. Drug Discov Today 2023; 28:103767. [PMID: 37708931 DOI: 10.1016/j.drudis.2023.103767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 08/31/2023] [Accepted: 09/07/2023] [Indexed: 09/16/2023]
Abstract
Recently, targeted protein degradation technologies based on lysosomal pathways have been developed. Lysosome-based targeted protein degradation technology has a broad range of substrates and the potential to degrade intracellular and extracellular proteins, protein aggregates, damaged organelles and non-protein molecules. Thus, they hold great promise for drug R&D. This study has focused on the biogenesis of lysosomes, their basic functions, lysosome-associated diseases and targeted protein degradation technologies through the lysosomal pathway. In addition, we thoroughly examine the potential applications and limitations of this technology and engage in insightful discussions on potential avenues for future research. Our primary objective is to foster preclinical research on this technology and facilitate its successful clinical implementation.
Collapse
Affiliation(s)
- Hongmei Zheng
- Affiliated Yongkang First People's Hospital and School of Pharmacy, Hangzhou Medical College, Hangzhou 310014, Zhejiang, China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, School of Pharmacy, Hangzhou Medical College, Hangzhou 310014, Zhejiang, China; Key Discipline of Zhejiang Province in Public Health and Preventive Medicine (First Class, Category A), Hangzhou Medical College, China
| | - Gangjian Li
- Affiliated Yongkang First People's Hospital and School of Pharmacy, Hangzhou Medical College, Hangzhou 310014, Zhejiang, China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, School of Pharmacy, Hangzhou Medical College, Hangzhou 310014, Zhejiang, China; Key Discipline of Zhejiang Province in Public Health and Preventive Medicine (First Class, Category A), Hangzhou Medical College, China
| | - Jingli Min
- Affiliated Yongkang First People's Hospital and School of Pharmacy, Hangzhou Medical College, Hangzhou 310014, Zhejiang, China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, School of Pharmacy, Hangzhou Medical College, Hangzhou 310014, Zhejiang, China; Key Discipline of Zhejiang Province in Public Health and Preventive Medicine (First Class, Category A), Hangzhou Medical College, China
| | - Xiangwei Xu
- Affiliated Yongkang First People's Hospital and School of Pharmacy, Hangzhou Medical College, Hangzhou 310014, Zhejiang, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.
| | - Wenhai Huang
- Affiliated Yongkang First People's Hospital and School of Pharmacy, Hangzhou Medical College, Hangzhou 310014, Zhejiang, China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, School of Pharmacy, Hangzhou Medical College, Hangzhou 310014, Zhejiang, China; Key Discipline of Zhejiang Province in Public Health and Preventive Medicine (First Class, Category A), Hangzhou Medical College, China.
| |
Collapse
|
5
|
Lysosomes as a Target of Anticancer Therapy. Int J Mol Sci 2023; 24:ijms24032176. [PMID: 36768500 PMCID: PMC9916765 DOI: 10.3390/ijms24032176] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 01/14/2023] [Accepted: 01/19/2023] [Indexed: 01/25/2023] Open
Abstract
Lysosomes are organelles containing acidic hydrolases that are responsible for lysosomal degradation and the maintenance of cellular homeostasis. They play an important role in autophagy, as well as in various cell death pathways, such as lysosomal and apoptotic death. Various agents, including drugs, can induce lysosomal membrane permeability, resulting in the translocation of acidic hydrolases into the cytoplasm, which promotes lysosomal-mediated death. This type of death may be of great importance in anti-cancer therapy, as both cancer cells with disturbed pathways leading to apoptosis and drug-resistant cells can undergo it. Important compounds that damage the lysosomal membrane include lysosomotropic compounds, antihistamines, immunosuppressants, DNA-damaging drugs, chemotherapeutics, photosensitizers and various plant compounds. An interesting approach in the treatment of cancer and the search for ways to overcome the chemoresistance of cancer cells may also be combining lysosomotropic compounds with targeted modulators of autophagy to induce cell death. These compounds may be an alternative in oncological treatment, and lysosomes may become a promising therapeutic target for many diseases, including cancer. Understanding the functional relationships between autophagy and apoptosis and the possibilities of their regulation, both in relation to normal and cancer cells, can be used to develop new and more effective anticancer therapies.
Collapse
|
6
|
Abstract
Lysosomes are acidic membrane-bound organelles that use hydrolytic enzymes to break down material through pathways such as endocytosis, phagocytosis, mitophagy, and autophagy. To function properly, intralysosomal environments are strictly controlled by a set of integral membrane proteins such as ion channels and transporters. Potassium ion (K+) channels are a large and diverse family of membrane proteins that control K+ flux across both the plasma membrane and intracellular membranes. In the plasma membrane, they are essential in both excitable and non-excitable cells for the control of membrane potential and cell signaling. However, our understanding of intracellular K+ channels is very limited. In this review, we summarize the recent development in studies of K+ channels in the lysosome. We focus on their characterization, potential roles in maintaining lysosomal membrane potential and lysosomal function, and pathological implications.
Collapse
Affiliation(s)
- Peng Huang
- Collaborative Innovation Center for Biomedicine, School of Clinical Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Mengnan Xu
- Department of Physiology and Biophysics, Dalhousie University, Sir Charles Tupper Medical Building, Halifax, NS, Canada
| | - Yi Wu
- Collaborative Innovation Center for Biomedicine, School of Clinical Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Alia Kazim Rizvi Syeda
- Department of Physiology and Biophysics, Dalhousie University, Sir Charles Tupper Medical Building, Halifax, NS, Canada
| | - Xian-Ping Dong
- Department of Physiology and Biophysics, Dalhousie University, Sir Charles Tupper Medical Building, Halifax, NS, Canada.
| |
Collapse
|
7
|
Nanotechnology for DNA and RNA delivery. Nanomedicine (Lond) 2023. [DOI: 10.1016/b978-0-12-818627-5.00008-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023] Open
|
8
|
Lou X, Ren TB, Chen H, Huan SY, Yuan L, Zhang XB. High-fidelity imaging of lysosomal enzyme through in situ ordered assembly of small molecular fluorescent probes. Biomaterials 2022; 287:121657. [PMID: 35853360 DOI: 10.1016/j.biomaterials.2022.121657] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/21/2022] [Accepted: 06/24/2022] [Indexed: 11/27/2022]
Abstract
As an organelle in cells, lysosomes play an important role in the degradation of biological macromolecules and pathogens. To elucidate the function of lysosomes in normal or disease states, recently, various fluorescent probes have been reported for imaging lysosomal analytes. However, because of the particularity of the lysosomal environment, most of the reported lysosomal fluorescent probes suffered from a series of practical issues such as easy diffusion, low detection signal-to-background ratio and false signal. To address these issues, based on an optimized in situ ordered assembly solid-state fluorophore HDPQ, we herein put forward a new strategy for the design of lysosomal enzymes probes. As a proof concept, we synthesized a fluorescent probe HDPQ-GLU for lysosomal enzyme β-glucuronidase (GLU). Experiment results displayed that compared with general lysosomal probe, the novel lysosomal probe not only exhibited excellent anti-pH interference ability and high signal-to-noise ratio in aqueous solution, but also has excellent long-term in situ imaging ability in the living system. Using this probe, we have realized high-fidelity and long-term in situ tracking GLU in lysosomes of living cells and evaluated the dynamic changes of GLU during the growth period of zebrafish. We anticipate that the new strategy based on the novel in situ ordered assembly solid-state fluorophore HDPQ may be a potential platform for developing fluorescent probes for high-fidelity imaging of lysosomal enzymes.
Collapse
Affiliation(s)
- Xiaofeng Lou
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, PR China
| | - Tian-Bing Ren
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, PR China.
| | - Haoming Chen
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, PR China
| | - Shuang-Yan Huan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, PR China
| | - Lin Yuan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, PR China
| | - Xiao-Bing Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, PR China.
| |
Collapse
|
9
|
Inhibition of lipid kinase PIKfyve reveals a role for phosphatase Inpp4b in the regulation of PI(3)P-mediated lysosome dynamics through VPS34 activity. J Biol Chem 2022; 298:102187. [PMID: 35760104 PMCID: PMC9304791 DOI: 10.1016/j.jbc.2022.102187] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 11/21/2022] Open
Abstract
Lysosome membranes contain diverse phosphoinositide (PtdIns) lipids that coordinate lysosome function and dynamics. The PtdIns repertoire on lysosomes is tightly regulated by the actions of diverse PtdIns kinases and phosphatases; however, specific roles for PtdIns in lysosomal functions and dynamics are currently unclear and require further investigation. It was previously shown that PIKfyve, a lipid kinase that synthesizes PtdIns(3,5)P2 from PtdIns(3)P, controls lysosome "fusion-fission" cycle dynamics, autophagosome turnover, and endocytic cargo delivery. Furthermore, INPP4B, a PtdIns 4-phosphatase that hydrolyzes PtdIns(3,4)P2 to form PtdIns(3)P, is emerging as a cancer-associated protein with roles in lysosomal biogenesis and other lysosomal functions. Here, we investigated the consequences of disrupting PIKfyve function in Inpp4b-deficient mouse embryonic fibroblasts. Through confocal fluorescence imaging, we observed the formation of massively enlarged lysosomes, accompanied by exacerbated reduction of endocytic trafficking, disrupted lysosome fusion-fission dynamics, and inhibition of autophagy. Finally, HPLC scintillation quantification of 3H-myo-inositol labelled phosphoinositides and phosphoinositide immunofluorescence staining, we observed that lysosomal PtdIns(3)P levels were significantly elevated in Inpp4b-deficient cells due to the hyperactivation of phosphatidylinositol 3-kinase catalytic subunit VPS34 enzymatic activity. In conclusion, our study identifies a novel signaling axis that maintains normal lysosomal homeostasis and dynamics, which includes the catalytic functions of Inpp4b, PIKfyve, and VPS34.
Collapse
|
10
|
Molnár M, Sőth Á, Simon-Vecsei Z. Pathways of integrins in the endo-lysosomal system. Biol Futur 2022; 73:171-185. [DOI: 10.1007/s42977-022-00120-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 04/09/2022] [Indexed: 12/13/2022]
Abstract
AbstractIn this review, we present recent scientific advances about integrin trafficking in the endo-lysosomal system. In the last few years, plenty of new information has emerged about the endo-lysosomal system, integrins, and the mechanism, how exactly the intracellular trafficking of integrins is regulated. We review the internalization and recycling pathways of integrins, and we provide information about the possible ways of lysosomal degradation through the endosomal and autophagic system. The regulation of integrin internalization and recycling proved to be a complex process worth studying. Trafficking of integrins, together with the regulation of their gene expression, defines cellular adhesion and cellular migration through bidirectional signalization and ligand binding. Thus, any malfunction in this system can potentially (but not necessarily) lead to tumorigenesis or metastasis. Hence, extensive examinations of integrins in the endo-lysosomal system raise the possibility to identify potential new medical targets. Furthermore, this knowledge can also serve as a basis for further determination of integrin signaling- and adhesion-related processes.
Collapse
|
11
|
Arad E, Jelinek R, Rapaport H. Amyloid fishing: β-Amyloid adsorption using tailor-made coated titania nanoparticles. Colloids Surf B Biointerfaces 2022; 212:112374. [PMID: 35121429 DOI: 10.1016/j.colsurfb.2022.112374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 11/19/2022]
Abstract
Amyloidoses are a family of diseases characterized by abnormal protein folding that leads to fibril aggregates, amyloids. Extensive research efforts are devoted to developing inhibitors to amyloid aggregates. Here we set to explore functionalized titania (TiO2) nanoparticles (NPs) as potential amyloid inhibiting agents. TiO2 NPs were coated by a catechol derivative, dihydroxy-phenylalanine propanoic acid (DPA), and further conjugated to the amyloids' specific dye Congo-Red (CR). TiO2-DPA-CR NPs were found to target mature fibrils of β-amyloid (Aβ). Moreover, coated NPs incubated with Aβ proteins suppressed amyloid fibrillation. TiO2-DPA-CR were found to target amyloids in solution and induce their sedimentation upon centrifugation. This work demonstrates the potential utilization of TiO2-DPA NPs for labeling and facilely separating from solution mature amyloid fibrils.
Collapse
Affiliation(s)
- Elad Arad
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Raz Jelinek
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Hanna Rapaport
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel.
| |
Collapse
|
12
|
Martucci LL, Cancela JM. Neurophysiological functions and pharmacological tools of acidic and non-acidic Ca2+ stores. Cell Calcium 2022; 104:102582. [DOI: 10.1016/j.ceca.2022.102582] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/07/2022] [Accepted: 03/23/2022] [Indexed: 02/08/2023]
|
13
|
A novel and modified fluorescent amphiphilic block copolymer simultaneously targeting to lysosomes and lipid droplets for cell imaging with large Stokes shift. Eur Polym J 2022. [DOI: 10.1016/j.eurpolymj.2022.111030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
14
|
Foster HE, Ventura Santos C, Carter AP. A cryo-ET survey of microtubules and intracellular compartments in mammalian axons. J Cell Biol 2022; 221:e202103154. [PMID: 34878519 PMCID: PMC7612188 DOI: 10.1083/jcb.202103154] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 09/28/2021] [Accepted: 11/16/2021] [Indexed: 12/16/2022] Open
Abstract
The neuronal axon is packed with cytoskeletal filaments, membranes, and organelles, many of which move between the cell body and axon tip. Here, we used cryo-electron tomography to survey the internal components of mammalian sensory axons. We determined the polarity of the axonal microtubules (MTs) by combining subtomogram classification and visual inspection, finding MT plus and minus ends are structurally similar. Subtomogram averaging of globular densities in the MT lumen suggests they have a defined structure, which is surprising given they likely contain the disordered protein MAP6. We found the endoplasmic reticulum in axons is tethered to MTs through multiple short linkers. We surveyed membrane-bound cargos and describe unexpected internal features such as granules and broken membranes. In addition, we detected proteinaceous compartments, including numerous virus-like capsid particles. Our observations outline novel features of axonal cargos and MTs, providing a platform for identification of their constituents.
Collapse
|
15
|
Wang Z, Lv J, Yu P, Qu Y, Zhou Y, Zhou L, Zhu Q, Li S, Song J, Deng W, Gao R, Liu Y, Liu J, Tong WM, Qin C, Huang B. SARS-CoV-2 treatment effects induced by ACE2-expressing microparticles are explained by the oxidized cholesterol-increased endosomal pH of alveolar macrophages. Cell Mol Immunol 2022; 19:210-221. [PMID: 34983944 PMCID: PMC8724656 DOI: 10.1038/s41423-021-00813-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/28/2021] [Indexed: 01/02/2023] Open
Abstract
Exploring the cross-talk between the immune system and advanced biomaterials to treat SARS-CoV-2 infection is a promising strategy. Here, we show that ACE2-overexpressing A549 cell-derived microparticles (AO-MPs) are a potential therapeutic agent against SARS-CoV-2 infection. Intranasally administered AO-MPs dexterously navigate the anatomical and biological features of the lungs to enter the alveoli and are taken up by alveolar macrophages (AMs). Then, AO-MPs increase the endosomal pH but decrease the lysosomal pH in AMs, thus escorting bound SARS-CoV-2 from phago-endosomes to lysosomes for degradation. This pH regulation is attributable to oxidized cholesterol, which is enriched in AO-MPs and translocated to endosomal membranes, thus interfering with proton pumps and impairing endosomal acidification. In addition to promoting viral degradation, AO-MPs also inhibit the proinflammatory phenotype of AMs, leading to increased treatment efficacy in a SARS-CoV-2-infected mouse model without side effects. These findings highlight the potential use of AO-MPs to treat SARS-CoV-2-infected patients and showcase the feasibility of MP therapies for combatting emerging respiratory viruses in the future.
Collapse
Affiliation(s)
- Zhenfeng Wang
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, 100005, China
| | - Jiadi Lv
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, 100005, China
| | - Pin Yu
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, CAMS and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Yajin Qu
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, CAMS and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Yabo Zhou
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, 100005, China
| | - Li Zhou
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, 100005, China
| | - Qiangqiang Zhu
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, 100005, China
| | - Shunshun Li
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, 100005, China
| | - Jiangping Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, CAMS and Peking Union Medical College, Beijing, China
| | - Wei Deng
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, CAMS and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Ran Gao
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, CAMS and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Yuying Liu
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, 100005, China
| | - Jiangning Liu
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, CAMS and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Wei-Min Tong
- Department of Pathology, Institute of Basic Medical Sciences, CAMS and Peking Union Medical College, Beijing, China
| | - Chuan Qin
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, CAMS and Comparative Medicine Center, Peking Union Medical College, Beijing, China.
| | - Bo Huang
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, 100005, China.
- Department of Biochemistry & Molecular Biology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430030, China.
| |
Collapse
|
16
|
Wu Y, Xu M, Wang P, Syeda AKR, Huang P, Dong XP. Lysosomal potassium channels. Cell Calcium 2022; 102:102536. [PMID: 35016151 DOI: 10.1016/j.ceca.2022.102536] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 12/21/2022]
Abstract
The lysosome is an important membrane-bound acidic organelle that is regarded as the degradative center as well as multifunctional signaling hub. It digests unwanted macromolecules, damaged organelles, microbes, and other materials derived from endocytosis, autophagy, and phagocytosis. To function properly, the ionic homeostasis and membrane potential of the lysosome are strictly regulated by transporters and ion channels. As the most abundant cation inside the cell, potassium ions (K+) are vital for lysosomal membrane potential and lysosomal calcium (Ca2+) signaling. However, our understanding about how lysosomal K+homeostasis is regulated and what are the functions of K+in the lysosome is very limited. Currently, two lysosomal K+channels have been identified: large-conductance Ca2+-activated K+channel (BK) and transmembrane Protein 175 (TMEM175). In this review, we summarize recent development in our understanding of K+ homeostasis and K+channels in the lysosome. We hope to guide the readers into a more in-depth discussion of lysosomal K+ channels in lysosomal physiology and human diseases.
Collapse
Affiliation(s)
- Yi Wu
- Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Rd, Shanghai 201318, China; School of Pharmacy, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Rd, Shanghai 201318, China
| | - Mengnan Xu
- Department of Physiology and Biophysics, Dalhousie University, Sir Charles Tupper Medical Building, 5850 College Street, Halifax, Nova NS B3H 4R2, Canada
| | - Pingping Wang
- Department of Physiology and Biophysics, Dalhousie University, Sir Charles Tupper Medical Building, 5850 College Street, Halifax, Nova NS B3H 4R2, Canada
| | - Alia Kazim Rizvi Syeda
- Department of Physiology and Biophysics, Dalhousie University, Sir Charles Tupper Medical Building, 5850 College Street, Halifax, Nova NS B3H 4R2, Canada
| | - Peng Huang
- Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Rd, Shanghai 201318, China; School of Clinical Medicine, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Rd, Shanghai 201318, China.
| | - Xian-Ping Dong
- Department of Physiology and Biophysics, Dalhousie University, Sir Charles Tupper Medical Building, 5850 College Street, Halifax, Nova NS B3H 4R2, Canada.
| |
Collapse
|
17
|
Mahapatra KK, Mishra SR, Behera BP, Patil S, Gewirtz DA, Bhutia SK. The lysosome as an imperative regulator of autophagy and cell death. Cell Mol Life Sci 2021; 78:7435-7449. [PMID: 34716768 PMCID: PMC11071813 DOI: 10.1007/s00018-021-03988-3] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/02/2021] [Accepted: 10/14/2021] [Indexed: 02/07/2023]
Abstract
Lysosomes are single membrane-bound organelles containing acid hydrolases responsible for the degradation of cellular cargo and maintenance of cellular homeostasis. Lysosomes could originate from pre-existing endolysosomes or autolysosomes, acting as a critical juncture between autophagy and endocytosis. Stress that triggers lysosomal membrane permeabilization can be altered by ESCRT complexes; however, irreparable damage to the membrane results in the induction of a selective lysosomal degradation pathway, specifically lysophagy. Lysosomes play an indispensable role in different types of autophagy, including microautophagy, macroautophagy, and chaperone-mediated autophagy, and various cell death pathways such as lysosomal cell death, apoptotic cell death, and autophagic cell death. In this review, we discuss lysosomal reformation, maintenance, and degradation pathways following the involvement of the lysosome in autophagy and cell death, which are related to several pathophysiological conditions observed in humans.
Collapse
Affiliation(s)
- Kewal Kumar Mahapatra
- Department of Life Science, Cancer and Cell Death Laboratory, National Institute of Technology Rourkela, Rourkela, Odisha, 769008, India
| | - Soumya Ranjan Mishra
- Department of Life Science, Cancer and Cell Death Laboratory, National Institute of Technology Rourkela, Rourkela, Odisha, 769008, India
| | - Bishnu Prasad Behera
- Department of Life Science, Cancer and Cell Death Laboratory, National Institute of Technology Rourkela, Rourkela, Odisha, 769008, India
| | - Shankargouda Patil
- Division of Oral Pathology, Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Jazan University, Jazan, Saudi Arabia
| | - David A Gewirtz
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, USA
| | - Sujit Kumar Bhutia
- Department of Life Science, Cancer and Cell Death Laboratory, National Institute of Technology Rourkela, Rourkela, Odisha, 769008, India.
| |
Collapse
|
18
|
Huang L, Li H, Ye Z, Xu Q, Fu Q, Sun W, Qi W, Yue J. Berbamine inhibits Japanese encephalitis virus (JEV) infection by compromising TPRMLs-mediated endolysosomal trafficking of low-density lipoprotein receptor (LDLR). Emerg Microbes Infect 2021; 10:1257-1271. [PMID: 34102949 PMCID: PMC8238074 DOI: 10.1080/22221751.2021.1941276] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 05/06/2021] [Accepted: 06/06/2021] [Indexed: 12/24/2022]
Abstract
Japanese encephalitis virus (JEV), a member of the Flavivirus genus, is an important pathogen that causes human and animal infectious diseases in Asia. So far, no effective antiviral agents are available to treat JEV infection. Here, we found that LDLR is a host factor required for JEV entry. Berbamine significantly decreases the level of LDLR at the plasma membrane by inducing the secretion of LDLR via extracellular vesicles (EVs), thereby inhibiting JEV infection. Mechanistically, berbamine blocks TRPMLs (Ca2+ permeable non-selective cation channels in endosomes and lysosomes) to compromise the endolysosomal trafficking of LDLR. This leads to the increased secretion of LDLR via EVs and the concomitant decrease in its level at the plasma membrane, thereby rendering cells resistant to JEV infection. Berbamine also protects mice from the lethal challenge of JEV. In summary, these results indicate that berbamine is an effective anti-JEV agent by preventing JEV entry.
Collapse
Affiliation(s)
- Lihong Huang
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, People's Republic of China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, People's Republic of China
| | - Huanan Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, People's Republic of China
| | - Zuodong Ye
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, People's Republic of China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, People's Republic of China
| | - Qiang Xu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, People's Republic of China
| | - Qiang Fu
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, People's Republic of China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, People's Republic of China
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, People's Republic of China
| | - Wei Sun
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, People's Republic of China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, People's Republic of China
| | - Wenbao Qi
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, People's Republic of China
| | - Jianbo Yue
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, People's Republic of China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, People's Republic of China
- City University of Hong Kong Chengdu Research Institute, Chengdu, People's Republic of China
| |
Collapse
|
19
|
Saffi GT, Tang E, Mamand S, Inpanathan S, Fountain A, Salmena L, Botelho RJ. Reactive oxygen species prevent lysosome coalescence during PIKfyve inhibition. PLoS One 2021; 16:e0259313. [PMID: 34813622 PMCID: PMC8610251 DOI: 10.1371/journal.pone.0259313] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 10/16/2021] [Indexed: 11/19/2022] Open
Abstract
Lysosomes are terminal, degradative organelles of the endosomal pathway that undergo repeated fusion-fission cycles with themselves, endosomes, phagosomes, and autophagosomes. Lysosome number and size depends on balanced fusion and fission rates. Thus, conditions that favour fusion over fission can reduce lysosome numbers while enlarging their size. Conversely, favouring fission over fusion may cause lysosome fragmentation and increase their numbers. PIKfyve is a phosphoinositide kinase that generates phosphatidylinositol-3,5-bisphosphate to modulate lysosomal functions. PIKfyve inhibition causes an increase in lysosome size and reduction in lysosome number, consistent with lysosome coalescence. This is thought to proceed through reduced lysosome reformation and/or fission after fusion with endosomes or other lysosomes. Previously, we observed that photo-damage during live-cell imaging prevented lysosome coalescence during PIKfyve inhibition. Thus, we postulated that lysosome fusion and/or fission dynamics are affected by reactive oxygen species (ROS). Here, we show that ROS generated by various independent mechanisms all impaired lysosome coalescence during PIKfyve inhibition and promoted lysosome fragmentation during PIKfyve re-activation. However, depending on the ROS species or mode of production, lysosome dynamics were affected distinctly. H2O2 impaired lysosome motility and reduced lysosome fusion with phagosomes, suggesting that H2O2 reduces lysosome fusogenecity. In comparison, inhibitors of oxidative phosphorylation, thiol groups, glutathione, or thioredoxin, did not impair lysosome motility but instead promoted clearance of actin puncta on lysosomes formed during PIKfyve inhibition. Additionally, actin depolymerizing agents prevented lysosome coalescence during PIKfyve inhibition. Thus, we discovered that ROS can generally prevent lysosome coalescence during PIKfyve inhibition using distinct mechanisms depending on the type of ROS.
Collapse
Affiliation(s)
- Golam T. Saffi
- Molecular Science Graduate Program, Ryerson University, Toronto, Ontario, Canada
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Evan Tang
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada
| | - Sami Mamand
- Molecular Science Graduate Program, Ryerson University, Toronto, Ontario, Canada
- Polytechnic Research Center, Erbil Polytechnic University, Kurdistan Regional Government, Erbil, Kurdistan
| | - Subothan Inpanathan
- Molecular Science Graduate Program, Ryerson University, Toronto, Ontario, Canada
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada
| | - Aaron Fountain
- Molecular Science Graduate Program, Ryerson University, Toronto, Ontario, Canada
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada
| | - Leonardo Salmena
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Roberto J. Botelho
- Molecular Science Graduate Program, Ryerson University, Toronto, Ontario, Canada
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada
| |
Collapse
|
20
|
Fountain A, Inpanathan S, Alves P, Verdawala MB, Botelho RJ. Phagosome maturation in macrophages: Eat, digest, adapt, and repeat. Adv Biol Regul 2021; 82:100832. [PMID: 34717137 DOI: 10.1016/j.jbior.2021.100832] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 10/06/2021] [Indexed: 11/30/2022]
Abstract
Phagocytosis is a dynamic process that requires an intricate interplay between phagocytic receptors, membrane lipids, and numerous signalling proteins and their effectors, to coordinate the engulfment of a bound particle. These particles are diverse in their physico-chemical properties such as size and shape and include bacteria, fungi, apoptotic cells, living tumour cells, and abiotic particles. Once engulfed, these particles are enclosed within a phagosome, which undergoes a striking transformation referred to as phagosome maturation, which will ultimately lead to the processing and degradation of the enclosed particulate. In this review, we focus on recent advancements in phagosome maturation in macrophages, highlighting new discoveries and emerging themes. Such advancements include identification of new GTPases and their effectors and the intricate spatio-temporal dynamics of phosphoinositides in governing phagosome maturation. We then explore phagosome fission and recycling, the emerging role of membrane contact sites, and delve into mechanisms of phagosome resolution to recycle and reform lysosomes. We further illustrate how phagosome maturation is context-dependent, subject to the type of particle, phagocytic receptors, the phagocytes and their state of activation during phagocytosis. Lastly, we discuss how phagosomes serve as signalling platforms to help phagocytes adapt to their environmental conditions. Overall, this review aims to cover recent findings, identify emerging themes, and highlight current challenges and directions to improve our understanding of phagosome maturation in macrophages.
Collapse
Affiliation(s)
- Aaron Fountain
- Department of Chemistry and Biology and Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B2K3, Canada; Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B2K3, Canada
| | - Subothan Inpanathan
- Department of Chemistry and Biology and Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B2K3, Canada; Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B2K3, Canada
| | - Patris Alves
- Department of Chemistry and Biology and Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B2K3, Canada; Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B2K3, Canada
| | - Munira B Verdawala
- Department of Chemistry and Biology and Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B2K3, Canada
| | - Roberto J Botelho
- Department of Chemistry and Biology and Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B2K3, Canada; Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B2K3, Canada.
| |
Collapse
|
21
|
Kamalkazemi E, Abedi-Gaballu F, Mohammad Hosseini TF, Mohammadi A, Mansoori B, Dehghan G, Baradaran B, Sheibani N. Glimpse into Cellular Internalization and Intracellular Trafficking of Lipid-Based Nanoparticles in Cancer Cells. Anticancer Agents Med Chem 2021; 22:1897-1912. [PMID: 34488605 DOI: 10.2174/1871520621666210906101421] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 06/14/2021] [Accepted: 06/27/2021] [Indexed: 11/22/2022]
Abstract
Lipid-based nanoparticles as drug delivery carriers have been mainly used for delivery of anti-cancer therapeutic agents. Lipid-based nanoparticles, due to their smaller particle size and similarity to cell membranes, are readily internalized into cancer cells. Interestingly, cancer cells also overexpress receptors for specific ligands including folic acid, hyaluronic acid, and transferrin on their surface. This allows the use of these ligands for surface modification of the lipid-based nanoparticle. These modifications then allow the specific recognition of these ligand-coated nanoparticles by their receptors on cancer cells allowing the targeted gradual intracellular accumulation of the functionalized nanoplatforms. These interactions could eventually enhance the internalization of desired drugs via increasing ligand-receptor mediated cellular uptake of the nanoplatforms. The cellular internalization of the nanoplatforms also varies and depends on their physicochemical properties including particle size, zeta potential, and shape. The cellular uptake is also influenced by the types of ligand internalization pathway utilized by cells such as phagocytosis, macropinocytosis, and multiple endocytosis pathways. In this review, we will classify and discuss lipid based nanoparticles engineered to express specific ligands, and are recognized by their receptors on cancer cell, and their cellular internalization pathways. Moreover, the intracellular fate of nanoparticles decorated with specific ligands and the best internalization pathways (caveolae mediated endocytosis) for safe cargo delivery will be discussed.
Collapse
Affiliation(s)
- Elham Kamalkazemi
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz. Iran
| | | | | | - Ali Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Gholamreza Dehghan
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz. Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Nader Sheibani
- Departments of Ophthalmology and Visual Sciences, Biomedical Engineering, and Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI . United States
| |
Collapse
|
22
|
Pham TD. Fuzzy Recurrence Exponents of Subcellular-Nanostructure Dynamics in Time-lapse Confocal Imaging. IEEE Trans Nanobioscience 2021; 20:497-506. [PMID: 34398761 DOI: 10.1109/tnb.2021.3105533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Studying the dynamics of nanostructures in the intracellular space is important because it allows gaining insights into the mechanism of complex biological functions of organelles. Understanding such dynamical processes can contribute to the development of nanomedicine for the diagnosis and treatment of many diseases caused by the interaction of multiple genes and environmental factors. Here a quantitative measure of spatial-temporal dynamics of nanostructures within a cell line in the context of nonlinear dynamics is introduced, where early endosomes, late endosomes, and lysosomes recorded by time-lapse confocal imaging are examined. The mathematical derivation of the proposed technique is based on the concept of recurrence dynamics and sequential rate of change over time. The quantification introduced as fuzzy recurrence exponents can be generalized for characterizing the dynamics of experimental evolutions in other nanostructures of living cells captured under the optical microscope.
Collapse
|
23
|
Nelson BN, Beakley SG, Posey S, Conn B, Maritz E, Seshu J, Wozniak KL. Antifungal activity of dendritic cell lysosomal proteins against Cryptococcus neoformans. Sci Rep 2021; 11:13619. [PMID: 34193926 PMCID: PMC8245489 DOI: 10.1038/s41598-021-92991-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 06/14/2021] [Indexed: 12/17/2022] Open
Abstract
Cryptococcal meningitis is a life-threatening disease among immune compromised individuals that is caused by the opportunistic fungal pathogen Cryptococcus neoformans. Previous studies have shown that the fungus is phagocytosed by dendritic cells (DCs) and trafficked to the lysosome where it is killed by both oxidative and non-oxidative mechanisms. While certain molecules from the lysosome are known to kill or inhibit the growth of C. neoformans, the lysosome is an organelle containing many different proteins and enzymes that are designed to degrade phagocytosed material. We hypothesized that multiple lysosomal components, including cysteine proteases and antimicrobial peptides, could inhibit the growth of C. neoformans. Our study identified the contents of the DC lysosome and examined the anti-cryptococcal properties of different proteins found within the lysosome. Results showed several DC lysosomal proteins affected the growth of C. neoformans in vitro. The proteins that killed or inhibited the fungus did so in a dose-dependent manner. Furthermore, the concentration of protein needed for cryptococcal inhibition was found to be non-cytotoxic to mammalian cells. These data show that many DC lysosomal proteins have antifungal activity and have potential as immune-based therapeutics.
Collapse
Affiliation(s)
- Benjamin N Nelson
- Department of Microbiology and Molecular Genetics, Oklahoma State University, 307 Life Science East, Stillwater, OK, 74078, USA
| | - Savannah G Beakley
- Department of Microbiology and Molecular Genetics, Oklahoma State University, 307 Life Science East, Stillwater, OK, 74078, USA
| | - Sierra Posey
- Department of Microbiology and Molecular Genetics, Oklahoma State University, 307 Life Science East, Stillwater, OK, 74078, USA
| | - Brittney Conn
- Department of Microbiology and Molecular Genetics, Oklahoma State University, 307 Life Science East, Stillwater, OK, 74078, USA
| | - Emma Maritz
- Department of Microbiology and Molecular Genetics, Oklahoma State University, 307 Life Science East, Stillwater, OK, 74078, USA
| | - Janakiram Seshu
- Department of Biology, South Texas Center for Emerging Infectious Diseases, San Antonio, TX, USA
| | - Karen L Wozniak
- Department of Microbiology and Molecular Genetics, Oklahoma State University, 307 Life Science East, Stillwater, OK, 74078, USA.
| |
Collapse
|
24
|
Luther DC, Jeon T, Goswami R, Nagaraj H, Kim D, Lee YW, Rotello VM. Protein Delivery: If Your GFP (or Other Small Protein) Is in the Cytosol, It Will Also Be in the Nucleus. Bioconjug Chem 2021; 32:891-896. [PMID: 33872490 PMCID: PMC8508718 DOI: 10.1021/acs.bioconjchem.1c00103] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Intracellular protein delivery is a transformative tool for biologics research and medicine. Delivery into the cytosol allows proteins to diffuse throughout the cell and access subcellular organelles. Inefficient delivery caused by endosomal entrapment is often misidentified as cytosolic delivery. This inaccuracy muddles what should be a key checkpoint in assessing delivery efficiency. Green fluorescent protein (GFP) is a robust cargo small enough to passively diffuse from the cytosol into the nucleus. Fluorescence of GFP in the nucleus is a direct readout for cytosolic access and effective delivery. Here, we highlight recent examples from the literature for the accurate assessment of cytosolic protein delivery using GFP fluorescence in the cytosol and nucleus.
Collapse
Affiliation(s)
- David C. Luther
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
- These authors contributed equally
| | - Taewon Jeon
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, 230 Stockbridge Road, Amherst, MA 01003, USA
- These authors contributed equally
| | - Ritabrita Goswami
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Harini Nagaraj
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Dongkap Kim
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Department of Chemistry, Hanyang University, Seoul 04763, Republic of Korea
| | - Yi-Wei Lee
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| |
Collapse
|
25
|
Qi J, Xing Y, Liu Y, Wang MM, Wei X, Sui Z, Ding L, Zhang Y, Lu C, Fei YH, Liu N, Chen R, Wu M, Wang L, Zhong Z, Wang T, Liu Y, Wang Y, Liu J, Xu H, Guo F, Wang W. MCOLN1/TRPML1 finely controls oncogenic autophagy in cancer by mediating zinc influx. Autophagy 2021; 17:4401-4422. [PMID: 33890549 DOI: 10.1080/15548627.2021.1917132] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Macroautophagy/autophagy is elevated to ensure the high demand for nutrients for the growth of cancer cells. Here we demonstrated that MCOLN1/TRPML1 is a pharmaceutical target of oncogenic autophagy in cancers such as pancreatic cancer, breast cancer, gastric cancer, malignant melanoma, and glioma. First, we showed that activating MCOLN1, by increasing expression of the channel or using the MCOLN1 agonists, ML-SA5 or MK6-83, arrests autophagic flux by perturbing fusion between autophagosomes and lysosomes. Second, we demonstrated that MCOLN1 regulates autophagy by mediating the release of zinc from the lysosome to the cytosol. Third, we uncovered that zinc influx through MCOLN1 blocks the interaction between STX17 (syntaxin 17) in the autophagosome and VAMP8 in the lysosome and thereby disrupting the fusion process that is determined by the two SNARE proteins. Furthermore, we demonstrated that zinc influx originating from the extracellular fluid arrests autophagy by the same mechanism as lysosomal zinc, confirming the fundamental function of zinc as a participant in membrane trafficking. Last, we revealed that activating MCOLN1 with the agonists, ML-SA5 or MK6-83, triggers cell death of a number of cancer cells by evoking autophagic arrest and subsequent apoptotic response and cell cycle arrest, with little or no effect observed on normal cells. Consistent with the in vitro results, administration of ML-SA5 in Patu 8988 t xenograft mice profoundly suppresses tumor growth and improves survival. These results establish that a lysosomal cation channel, MCOLN1, finely controls oncogenic autophagy in cancer by mediating zinc influx into the cytosol.
Collapse
Affiliation(s)
- Jiansong Qi
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China.,Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Canada
| | - Yanhong Xing
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Yucheng Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Meng-Meng Wang
- Department of Otolaryngology and Neck Surgery, The Sleep Medicine Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiangqing Wei
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Zhongheng Sui
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Lin Ding
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Yang Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Chen Lu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Yuan-Hui Fei
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Nan Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Rong Chen
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Mengmei Wu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Lijuan Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Zhenyu Zhong
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Ting Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Yifan Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Yuqing Wang
- Department of Medicine and Biosystemic Science, Faculty of Medicine, Kyushu University, Fukuoka, Japan
| | - Jiamei Liu
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Haoxing Xu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, USA
| | - Feng Guo
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Wuyang Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
26
|
Lv J, Wang Z, Qu Y, Zhu H, Zhu Q, Tong W, Bao L, Lv Q, Cong J, Li D, Deng W, Yu P, Song J, Tong WM, Liu J, Liu Y, Qin C, Huang B. Distinct uptake, amplification, and release of SARS-CoV-2 by M1 and M2 alveolar macrophages. Cell Discov 2021; 7:24. [PMID: 33850112 PMCID: PMC8043100 DOI: 10.1038/s41421-021-00258-1] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 03/11/2021] [Indexed: 01/17/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) invades the alveoli, where abundant alveolar macrophages (AMs) reside. How AMs respond to SARS-CoV-2 invasion remains elusive. Here, we show that classically activated M1 AMs facilitate viral spread; however, alternatively activated M2 AMs limit the spread. M1 AMs utilize cellular softness to efficiently take up SARS-CoV-2. Subsequently, the invaded viruses take over the endo-lysosomal system to escape. M1 AMs have a lower endosomal pH, favoring membrane fusion and allowing the entry of viral RNA from the endosomes into the cytoplasm, where the virus achieves replication and is packaged to be released. In contrast, M2 AMs have a higher endosomal pH but a lower lysosomal pH, thus delivering the virus to lysosomes for degradation. In hACE2 transgenic mouse model, M1 AMs are found to facilitate SARS-CoV-2 infection of the lungs. These findings provide insights into the complex roles of AMs during SARS-CoV-2 infection, along with potential therapeutic targets.
Collapse
Affiliation(s)
- Jiadi Lv
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College, Beijing 100005, China
| | - Zhenfeng Wang
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College, Beijing 100005, China
| | - Yajin Qu
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, CAMS and Comparative Medicine Center, Peking Union Medical College, Beijing 100005, China
| | - Hua Zhu
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, CAMS and Comparative Medicine Center, Peking Union Medical College, Beijing 100005, China
| | - Qiangqiang Zhu
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College, Beijing 100005, China
| | - Wei Tong
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, CAMS and Comparative Medicine Center, Peking Union Medical College, Beijing 100005, China
| | - Linlin Bao
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, CAMS and Comparative Medicine Center, Peking Union Medical College, Beijing 100005, China
| | - Qi Lv
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, CAMS and Comparative Medicine Center, Peking Union Medical College, Beijing 100005, China
| | - Ji Cong
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, CAMS and Comparative Medicine Center, Peking Union Medical College, Beijing 100005, China
| | - Dan Li
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, CAMS and Comparative Medicine Center, Peking Union Medical College, Beijing 100005, China
| | - Wei Deng
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, CAMS and Comparative Medicine Center, Peking Union Medical College, Beijing 100005, China
| | - Pin Yu
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, CAMS and Comparative Medicine Center, Peking Union Medical College, Beijing 100005, China
| | - Jiangping Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, CAMS and Peking Union Medical College, Beijing 100005, China
| | - Wei-Min Tong
- Department of Pathology, Institute of Basic Medical Sciences, CAMS and Peking Union Medical College, Beijing 100005, China
| | - Jiangning Liu
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, CAMS and Comparative Medicine Center, Peking Union Medical College, Beijing 100005, China.
| | - Yuying Liu
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College, Beijing 100005, China. .,Clinical Immunology Center, CAMS, Beijing 100005, China.
| | - Chuan Qin
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, CAMS and Comparative Medicine Center, Peking Union Medical College, Beijing 100005, China.
| | - Bo Huang
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College, Beijing 100005, China. .,Clinical Immunology Center, CAMS, Beijing 100005, China. .,Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
27
|
Shi W, Lu D, Wu C, Li M, Ding Z, Li Y, Chen B, Lin X, Su W, Shao X, Xia Z, Fang L, Liu K, Li H. Coibamide A kills cancer cells through inhibiting autophagy. Biochem Biophys Res Commun 2021; 547:52-58. [PMID: 33592379 DOI: 10.1016/j.bbrc.2021.01.112] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 02/06/2023]
Abstract
Natural products are useful tools for biological mechanism research and drug discovery. Due to the excellent tumor cell growth inhibitory profile and sub-nanomolar potency, Coibamide A (CA), an N-methyl-stabilized depsipeptide isolated from marine cyanobacterium, has been considered as a promising lead compound for cancer treatment. However, the molecular anti-cancer mechanism of the action of CA remains unclear. Here, we showed that CA treatment induced caspase-independent cell death in breast cancer cells. CA treatment also led to severe lysosome defects, which was ascribed to the impaired glycosylation of lysosome membrane protein LAMP1 and LAMP2. As a consequence, the autophagosome-lysosome fusion was blocked upon CA treatment. In addition, we presented evidence that this autophagy defect partially contributed to the CA treatment-induced tumor cell death. Together, our work uncovers a novel mechanism underlying the anti-cancer action of CA, which will promote its further application for cancer therapy.
Collapse
Affiliation(s)
- Wenli Shi
- School of Life and Pharmaceutical Sciences, Hainan University, Haikou, 570228, China; Hainan Key Laboratory for Sustainable Utilization of Tropical Bioresources, College of Tropical Crops, Hainan University, Haikou, 570228, China; Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Danyi Lu
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Institute of Molecular Rhythm and Metabolism, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Chunlei Wu
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Meiqing Li
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhihao Ding
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yanyan Li
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Binghua Chen
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xian Lin
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Wu Su
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Ximing Shao
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Zhihui Xia
- Hainan Key Laboratory for Sustainable Utilization of Tropical Bioresources, College of Tropical Crops, Hainan University, Haikou, 570228, China
| | - Lijing Fang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Ke Liu
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Hongchang Li
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
28
|
Gu Y, Princely Abudu Y, Kumar S, Bissa B, Choi SW, Jia J, Lazarou M, Eskelinen E, Johansen T, Deretic V. Mammalian Atg8 proteins regulate lysosome and autolysosome biogenesis through
SNARE
s. EMBO J 2019; 38. [DOI: https:/doi.org/10.15252/embj.2019101994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 09/13/2019] [Indexed: 12/19/2023] Open
Affiliation(s)
- Yuexi Gu
- Autophagy, Inflammation and Metabolism (AIM) Center of Biomedical Research Excellence University of New Mexico Health Sciences Center Albuquerque NM USA
- Department of Molecular Genetics and Microbiology University of New Mexico Health Sciences Center Albuquerque NM USA
| | - Yakubu Princely Abudu
- Molecular Cancer Research Group Institute of Medical Biology University of Tromsø‐The Arctic University of Norway Tromsø Norway
| | - Suresh Kumar
- Autophagy, Inflammation and Metabolism (AIM) Center of Biomedical Research Excellence University of New Mexico Health Sciences Center Albuquerque NM USA
- Department of Molecular Genetics and Microbiology University of New Mexico Health Sciences Center Albuquerque NM USA
| | - Bhawana Bissa
- Autophagy, Inflammation and Metabolism (AIM) Center of Biomedical Research Excellence University of New Mexico Health Sciences Center Albuquerque NM USA
- Department of Molecular Genetics and Microbiology University of New Mexico Health Sciences Center Albuquerque NM USA
| | - Seong Won Choi
- Department of Molecular Genetics and Microbiology University of New Mexico Health Sciences Center Albuquerque NM USA
| | - Jingyue Jia
- Autophagy, Inflammation and Metabolism (AIM) Center of Biomedical Research Excellence University of New Mexico Health Sciences Center Albuquerque NM USA
- Department of Molecular Genetics and Microbiology University of New Mexico Health Sciences Center Albuquerque NM USA
| | - Michael Lazarou
- Department of Biochemistry and Molecular Biology Biomedicine Discovery Institute Monash University Melbourne Australia
| | | | - Terje Johansen
- Molecular Cancer Research Group Institute of Medical Biology University of Tromsø‐The Arctic University of Norway Tromsø Norway
| | - Vojo Deretic
- Autophagy, Inflammation and Metabolism (AIM) Center of Biomedical Research Excellence University of New Mexico Health Sciences Center Albuquerque NM USA
- Department of Molecular Genetics and Microbiology University of New Mexico Health Sciences Center Albuquerque NM USA
| |
Collapse
|
29
|
Gu Y, Princely Abudu Y, Kumar S, Bissa B, Choi SW, Jia J, Lazarou M, Eskelinen E, Johansen T, Deretic V. Mammalian Atg8 proteins regulate lysosome and autolysosome biogenesis through SNAREs. EMBO J 2019; 38:e101994. [PMID: 31625181 PMCID: PMC6856626 DOI: 10.15252/embj.2019101994] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 09/07/2019] [Accepted: 09/13/2019] [Indexed: 12/14/2022] Open
Abstract
Mammalian homologs of yeast Atg8 protein (mAtg8s) are important in autophagy, but their exact mode of action remains ill-defined. Syntaxin 17 (Stx17), a SNARE with major roles in autophagy, was recently shown to bind mAtg8s. Here, we identified LC3-interacting regions (LIRs) in several SNAREs that broaden the landscape of the mAtg8-SNARE interactions. We found that Syntaxin 16 (Stx16) and its cognate SNARE partners all have LIR motifs and bind mAtg8s. Knockout of Stx16 caused defects in lysosome biogenesis, whereas a Stx16 and Stx17 double knockout completely blocked autophagic flux and decreased mitophagy, pexophagy, xenophagy, and ribophagy. Mechanistic analyses revealed that mAtg8s and Stx16 control several properties of lysosomal compartments including their function as platforms for active mTOR. These findings reveal a broad direct interaction of mAtg8s with SNAREs with impact on membrane remodeling in eukaryotic cells and expand the roles of mAtg8s to lysosome biogenesis.
Collapse
Affiliation(s)
- Yuexi Gu
- Autophagy, Inflammation and Metabolism (AIM) Center of Biomedical Research ExcellenceUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
- Department of Molecular Genetics and MicrobiologyUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
| | - Yakubu Princely Abudu
- Molecular Cancer Research GroupInstitute of Medical BiologyUniversity of Tromsø‐The Arctic University of NorwayTromsøNorway
| | - Suresh Kumar
- Autophagy, Inflammation and Metabolism (AIM) Center of Biomedical Research ExcellenceUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
- Department of Molecular Genetics and MicrobiologyUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
| | - Bhawana Bissa
- Autophagy, Inflammation and Metabolism (AIM) Center of Biomedical Research ExcellenceUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
- Department of Molecular Genetics and MicrobiologyUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
| | - Seong Won Choi
- Department of Molecular Genetics and MicrobiologyUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
| | - Jingyue Jia
- Autophagy, Inflammation and Metabolism (AIM) Center of Biomedical Research ExcellenceUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
- Department of Molecular Genetics and MicrobiologyUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
| | - Michael Lazarou
- Department of Biochemistry and Molecular BiologyBiomedicine Discovery InstituteMonash UniversityMelbourneAustralia
| | | | - Terje Johansen
- Molecular Cancer Research GroupInstitute of Medical BiologyUniversity of Tromsø‐The Arctic University of NorwayTromsøNorway
| | - Vojo Deretic
- Autophagy, Inflammation and Metabolism (AIM) Center of Biomedical Research ExcellenceUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
- Department of Molecular Genetics and MicrobiologyUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
| |
Collapse
|
30
|
The peripheral vesicles gather multivesicular bodies with different behavior during the Giardia intestinalis life cycle. J Struct Biol 2019; 207:301-311. [DOI: 10.1016/j.jsb.2019.07.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 06/05/2019] [Accepted: 07/01/2019] [Indexed: 11/17/2022]
|
31
|
Yin J, Peng M, Lin W. Tracking lysosomal polarity variation in inflamed, obese, and cancer mice guided by a fluorescence sensing strategy. Chem Commun (Camb) 2019; 55:11063-11066. [PMID: 31454009 DOI: 10.1039/c9cc04739e] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Elucidating lysosome polarity effect in complicated biosystems was impeded with the deficiency of lacking multi-disease models for researching the relation between lysosomal polarity and diseases. So far, dissecting the abnormal lysosome polarity in the inflamed and obese living mice has not been realized. To overcome this challenge, a robust probe MND-Lys was proposed for monitoring lysosomal polarity with two-photon emission. Using the probe, monitoring the intrinsic polarity variance in embryos and adult zebrafish has been achieved for the first time. Moreover, besides obviously discriminating tumors from normal ones, the probe also enabled tracing polarity changes in inflammatory and obese mice for the first time. The unique tracking and distinguishing polarity in lysosome make the probe a promising agent for fluorescence visualization studies of LD-lysosome related bioprocess and metabolism diseases.
Collapse
Affiliation(s)
- Junling Yin
- Institute of Fluorescent Probes for Biological Imaging, School of Chemistry and Chemical Engineering, School of Materials Science and Engineering, University of Jinan, Jinan, Shandong 250022, P. R. China.
| | - Min Peng
- Institute of Fluorescent Probes for Biological Imaging, School of Chemistry and Chemical Engineering, School of Materials Science and Engineering, University of Jinan, Jinan, Shandong 250022, P. R. China.
| | - Weiying Lin
- Institute of Fluorescent Probes for Biological Imaging, School of Chemistry and Chemical Engineering, School of Materials Science and Engineering, University of Jinan, Jinan, Shandong 250022, P. R. China.
| |
Collapse
|
32
|
Lyu C, Cai X. A GFP-tagged version of the pseudorabies virus protein UL56 localizes to the Golgi and trans-Golgi network through a predicted C-terminal leucine-rich helix in transfected cells. Virol J 2019; 16:81. [PMID: 31221185 PMCID: PMC6585060 DOI: 10.1186/s12985-019-1191-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 06/12/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Pseudorabies virus (PRV) protein UL56 (pUL56) has been implicated in viral dissemination and virulence in vivo. However, the properties of PRV pUL56 remain largely unknown. In the present study, we aim to investigate the subcellular localization of pUL56 and the underlying molecular basis in transfected cells. METHODS Constructs of N-terminal green fluorescent protein (GFP) fused pUL56 and its truncations were employed for investigating subcellular localization and further identifying amino acids crucial for pUL56 localization in transfected Vero cells. Finally, the identified amino acids were replaced with alanine for confirming if these mutations could impair the specific localization of pUL56. RESULTS The pUL56 predominantly localized at the Golgi and trans-Golgi network (TGN) through its predicted C-terminal transmembrane helix in transfected Vero cells. A Golgi-associated protein Rab6a, independent of interaction with pUL56, was significantly downregulated by pUL56. Further, we found three truncated pUL56 C-terminal fragments (174-184, 175-185 and 191-195) could restrict GFP in the perinuclear region, respectively. Within these truncations, the 174proline (P), 181leucine (L), 185L and 191L were essential for maintaining perinuclear accumulation, thus suggesting an important role of leucine. Alanine (A) mutagenesis assays were employed to generate a series of pUL56 C-terminal mutants on the basis of leucine. Finally, a pUL56 mutant M10 (174P/A-177L/A-181L/A-185L/A-191L/A-194L/A-195I/A-196-197L/A-200L/A) lost Golgi-TGN localization. Thus, our data revealed that the leucine-rich transmembrane helix was responsible for pUL56 Golgi-TGN localization and retention, probably through specific intracellular membrane insertion. CONCLUSION Our data indicated that the C-terminal transmembrane helix was responsible for the Golgi-TGN localization of pUL56. In addition, the leucines within C-terminal transmembrane helix were essential for maintaining pUL56 Golgi-TGN retention in cells. Further, the pUL56 can induce downregulation of Golgi-associated protein Rab6a.
Collapse
Affiliation(s)
- Chuang Lyu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Haping Road No.678, Xiang Fang District, Harbin, 150069, Heilongjiang, China
| | - Xuehui Cai
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Haping Road No.678, Xiang Fang District, Harbin, 150069, Heilongjiang, China.
| |
Collapse
|
33
|
Sharma D, Otto G, Warren EC, Beesley P, King JS, Williams RSB. Gamma secretase orthologs are required for lysosomal activity and autophagic degradation in Dictyostelium discoideum, independent of PSEN (presenilin) proteolytic function. Autophagy 2019; 15:1407-1418. [PMID: 30806144 PMCID: PMC6613883 DOI: 10.1080/15548627.2019.1586245] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mutations in the γ-secretase complex are strongly associated with familial Alzheimer disease. Both proteolytic and non-proteolytic functions for the γ-secretase complex have been previously described in mammalian model organisms, but their relative contributions to disease pathology remain unclear. Here, we dissect the roles of orthologs of the γ-secretase components in the model system Dictyostelium, focusing on endocytosis, lysosomal activity and autophagy. In this model, we show that the orthologs of PSEN (psenA and psenB), Ncstn (nicastrin) and Aph-1 (gamma-secretase subunit Aph-1), are necessary for optimal fluid-phase uptake by macropinocytosis and in multicellular development under basic pH conditions. Disruption of either psenA/B or Aph-1 proteins also leads to disrupted phagosomal proteolysis as well as decreased autophagosomal acidification and autophagic flux. This indicates a general defect in lysosomal trafficking and degradation, which we show leads to the accumulation of ubiquitinated protein aggregates in cells lacking psenA/B and Aph-1 proteins. Importantly, we find that all the endocytic defects observed in Dictyostelium PSEN ortholog mutants can be fully rescued by proteolytically inactive Dictyostelium psenB and human PSEN1 proteins. Our data therefore demonstrates an evolutionarily conserved non-proteolytic role for presenilin, and γ-secretase component orthologs, in maintaining Dictyostelium lysosomal trafficking and autophagy. Abbreviations: Atg8: autophagy protein 8a; Aph-1: gamma-secretase subunit Aph-1; crtA: calreticulin; ER: endoplasmic reticulum; GFP: green fluorescent protein; GSK3B: glycogen synthase kinase 3 beta; Ncstn: nicastrin; PSEN1: presenilin 1; psenA and psenB: Dictyostelium presenilin A and B; TRITC; tetramethylrhodamine isothiocyanate.
Collapse
Affiliation(s)
- Devdutt Sharma
- a School of Biological Sciences , Royal Holloway, University of London , Egham , UK
| | - Grant Otto
- a School of Biological Sciences , Royal Holloway, University of London , Egham , UK
| | - Eleanor C Warren
- a School of Biological Sciences , Royal Holloway, University of London , Egham , UK
| | - Philip Beesley
- a School of Biological Sciences , Royal Holloway, University of London , Egham , UK
| | - Jason S King
- b Department of Biomedical Sciences , University of Sheffield , Sheffield , UK
| | - Robin S B Williams
- a School of Biological Sciences , Royal Holloway, University of London , Egham , UK
| |
Collapse
|
34
|
KDEL receptor regulates secretion by lysosome relocation- and autophagy-dependent modulation of lipid-droplet turnover. Nat Commun 2019; 10:735. [PMID: 30760704 PMCID: PMC6374470 DOI: 10.1038/s41467-019-08501-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 01/09/2019] [Indexed: 02/01/2023] Open
Abstract
Inter-organelle signalling has essential roles in cell physiology encompassing cell metabolism, aging and temporal adaptation to external and internal perturbations. How such signalling coordinates different organelle functions within adaptive responses remains unknown. Membrane traffic is a fundamental process in which membrane fluxes need to be sensed for the adjustment of cellular requirements and homeostasis. Studying endoplasmic reticulum-to-Golgi trafficking, we found that Golgi-based, KDEL receptor-dependent signalling promotes lysosome repositioning to the perinuclear area, involving a complex process intertwined to autophagy, lipid-droplet turnover and Golgi-mediated secretion that engages the microtubule motor protein dynein-LRB1 and the autophagy cargo receptor p62/SQSTM1. This process, here named ‘traffic-induced degradation response for secretion’ (TIDeRS) discloses a cellular mechanism by which nutrient and membrane sensing machineries cooperate to sustain Golgi-dependent protein secretion. Inter-organelle signaling coordinates adaptive responses via currently unknown mechanisms. Here, Tapia et al. show that KDEL signaling repositions lysosomes in a complex process termed ‘traffic-induced degradation response for secretion’ (TIDeRS) that connects multiple pathways and Golgi secretion.
Collapse
|
35
|
3D reconstruction of Trypanosoma cruzi-macrophage interaction shows the recruitment of host cell organelles towards parasitophorous vacuoles during its biogenesis. J Struct Biol 2019; 205:133-146. [DOI: 10.1016/j.jsb.2018.12.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 12/18/2018] [Accepted: 12/20/2018] [Indexed: 02/06/2023]
|
36
|
Grabon A, Bankaitis VA, McDermott MI. The interface between phosphatidylinositol transfer protein function and phosphoinositide signaling in higher eukaryotes. J Lipid Res 2018; 60:242-268. [PMID: 30504233 DOI: 10.1194/jlr.r089730] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/12/2018] [Indexed: 12/22/2022] Open
Abstract
Phosphoinositides are key regulators of a large number of diverse cellular processes that include membrane trafficking, plasma membrane receptor signaling, cell proliferation, and transcription. How a small number of chemically distinct phosphoinositide signals are functionally amplified to exert specific control over such a diverse set of biological outcomes remains incompletely understood. To this end, a novel mechanism is now taking shape, and it involves phosphatidylinositol (PtdIns) transfer proteins (PITPs). The concept that PITPs exert instructive regulation of PtdIns 4-OH kinase activities and thereby channel phosphoinositide production to specific biological outcomes, identifies PITPs as central factors in the diversification of phosphoinositide signaling. There are two evolutionarily distinct families of PITPs: the Sec14-like and the StAR-related lipid transfer domain (START)-like families. Of these two families, the START-like PITPs are the least understood. Herein, we review recent insights into the biochemical, cellular, and physiological function of both PITP families with greater emphasis on the START-like PITPs, and we discuss the underlying mechanisms through which these proteins regulate phosphoinositide signaling and how these actions translate to human health and disease.
Collapse
Affiliation(s)
- Aby Grabon
- E. L. Wehner-Welch Laboratory, Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843-1114
| | - Vytas A Bankaitis
- E. L. Wehner-Welch Laboratory, Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843-1114
| | - Mark I McDermott
- E. L. Wehner-Welch Laboratory, Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843-1114
| |
Collapse
|
37
|
Bertman KA, Abeywickrama CS, Baumann HJ, Alexander N, McDonald L, Shriver LP, Konopka M, Pang Y. A fluorescent flavonoid for lysosome detection in live cells under "wash free" conditions. J Mater Chem B 2018; 6:5050-5058. [PMID: 32254534 DOI: 10.1039/c8tb00325d] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Lysosomes are vital organelles in living cells, which have acidic environments (pH 4.0-5.0) where macrobiomolecules and malfunctioning organelles are broken down into monomers by hydrolase activity. The majority of the currently reported fluorescent probes for detecting lysosomes suffer from small Stokes shifts (Δλ < 20 nm) and higher cytotoxicity due to an "alkalinizing effect". An interesting flavonoid-based lysosome probe is synthesized by introducing a morpholine moiety onto the flavonoid skeleton. This new probe has shown excellent selectivity to detect lysosomes in MO3.13 oligodendrocytes and normal human lung fibroblast cell lines. Probes 1a and 1b have shown excellent fluorescence quantum yield (φfl up to 0.43 in non-aqueous solvents) and large Stokes shifts (120-150 nm). These new fluorescent probes also exhibit a large quantum yield difference from an aqueous to organic environment, making them potentially useful as "wash-free" stains for visualizing lysosomes. Cell viability evaluation of these probes shows excellent biocompatibility with the median lethal concentration being LC50 ≈ 50 μM.
Collapse
|
38
|
Hayashi T, Shinagawa M, Kawano T, Iwasaki T. Drug delivery using polyhistidine peptide-modified liposomes that target endogenous lysosome. Biochem Biophys Res Commun 2018; 501:648-653. [DOI: 10.1016/j.bbrc.2018.05.037] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 05/06/2018] [Indexed: 12/25/2022]
|
39
|
Han T, Deng H, Qiu Z, Zhao Z, Zhang H, Zou H, Leung NLC, Shan G, Elsegood MRJ, Lam JWY, Tang BZ. Facile Multicomponent Polymerizations toward Unconventional Luminescent Polymers with Readily Openable Small Heterocycles. J Am Chem Soc 2018; 140:5588-5598. [DOI: 10.1021/jacs.8b01991] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Ting Han
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Institute for Advanced Study, Division of Biomedical Engineering and Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- HKUST-Shenzhen Research Institute, No. 9 Yuexing first RD, South Area, Hi-tech Park, Nanshan, Shenzhen 518057, China
| | - Haiqin Deng
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Institute for Advanced Study, Division of Biomedical Engineering and Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- HKUST-Shenzhen Research Institute, No. 9 Yuexing first RD, South Area, Hi-tech Park, Nanshan, Shenzhen 518057, China
| | - Zijie Qiu
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Institute for Advanced Study, Division of Biomedical Engineering and Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- HKUST-Shenzhen Research Institute, No. 9 Yuexing first RD, South Area, Hi-tech Park, Nanshan, Shenzhen 518057, China
| | - Zheng Zhao
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Institute for Advanced Study, Division of Biomedical Engineering and Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- HKUST-Shenzhen Research Institute, No. 9 Yuexing first RD, South Area, Hi-tech Park, Nanshan, Shenzhen 518057, China
| | - Haoke Zhang
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Institute for Advanced Study, Division of Biomedical Engineering and Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- HKUST-Shenzhen Research Institute, No. 9 Yuexing first RD, South Area, Hi-tech Park, Nanshan, Shenzhen 518057, China
| | - Hang Zou
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Institute for Advanced Study, Division of Biomedical Engineering and Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- HKUST-Shenzhen Research Institute, No. 9 Yuexing first RD, South Area, Hi-tech Park, Nanshan, Shenzhen 518057, China
| | - Nelson L. C. Leung
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Institute for Advanced Study, Division of Biomedical Engineering and Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- HKUST-Shenzhen Research Institute, No. 9 Yuexing first RD, South Area, Hi-tech Park, Nanshan, Shenzhen 518057, China
| | - Guogang Shan
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Institute for Advanced Study, Division of Biomedical Engineering and Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- HKUST-Shenzhen Research Institute, No. 9 Yuexing first RD, South Area, Hi-tech Park, Nanshan, Shenzhen 518057, China
| | - Mark R. J. Elsegood
- Chemistry Department, Loughborough University, Loughborough, Leicestershire LE11 3TU, U.K
| | - Jacky W. Y. Lam
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Institute for Advanced Study, Division of Biomedical Engineering and Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- HKUST-Shenzhen Research Institute, No. 9 Yuexing first RD, South Area, Hi-tech Park, Nanshan, Shenzhen 518057, China
| | - Ben Zhong Tang
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Institute for Advanced Study, Division of Biomedical Engineering and Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- HKUST-Shenzhen Research Institute, No. 9 Yuexing first RD, South Area, Hi-tech Park, Nanshan, Shenzhen 518057, China
- China NSFC Center for Luminescence from Molecular Aggregates, SCUT-HKUST Joint Research Institute, State Key Laboratory of Luminescent Materials and Devices, South China University of Technology, Guangzhou 510640, China
| |
Collapse
|
40
|
Pancreatic alpha cells in diabetic rats express active GLP-1 receptor: Endosomal co-localization of GLP-1/GLP-1R complex functioning through intra-islet paracrine mechanism. Sci Rep 2018; 8:3725. [PMID: 29487355 PMCID: PMC5829082 DOI: 10.1038/s41598-018-21751-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 02/09/2018] [Indexed: 01/03/2023] Open
Abstract
Glucagon-like peptide-1 (GLP-1) stimulates insulin secretion from pancreatic beta cells and suppresses glucagon secretion from alpha cells. It remains controversial, however, whether GLP-1 receptor (GLP-1R) is expressed in mature alpha cells. In this study, unlike previous studies using non-diabetic animals, we demonstrated using diabetic model rats and confocal laser scanning microscopy that the GLP-1/GLP-1R complex was located in the endosome of diabetic islets. In addition, we showed that GLP-1 and GLP-1R co-localized with various endosomal markers and adenylate cyclase in the alpha cells of diabetic rats. Diabetic rats had endosomal signaling pathway but normal rats had classical signaling pathway for activated GLP-1R. Furthermore, we performed pancreatic perfusion to assess the functional activity of GLP-1R when stimulated by exendin-4 (EX4). In a pancreas perfusion study, EX4 significantly stimulated glucagon secretion in diabetic rats but not normal rats. However, such glucagon secretion was immediately suppressed, probably due to concomitantly secreted insulin. The GLP-1/GLP-1R complex appears to function through an intra-islet paracrine mechanism in diabetic conditions which could explain, at least in part, the mechanism of paradoxical hyperglucagonaemia in type 2 diabetes.
Collapse
|
41
|
Abstract
Parkinson's disease is a progressive neurodegenerative disease characterized by Lewy body pathology of which the primary constituent is aggregated misfolded alpha-synuclein protein. Currently, there are no clinical therapies for treatment of the underlying alpha-synuclein dysfunction and accumulation, and the standard of care for patients with Parkinson's disease focuses only on symptom management, creating an immense therapeutic gap that needs to be filled. Defects in autophagy have been strongly implicated in Parkinson's disease. Here, we review evidence from human, mouse, and cell culture studies to briefly explain these defects in autophagy in Parkinson's disease and the necessity for autophagy to be carefully and precisely tuned to maintain neuron survival. We summarize recent experimental agents for treating alpha-synuclein accumulation in α-synuclein Parkinson's disease and related synucleinopathies. Most of the efforts for developing experimental agents have focused on immunotherapeutic strategies, but we discuss why those efforts are misplaced. Finally, we emphasize why increasing autophagy flux for alpha-synuclein clearance is the most promising therapeutic strategy. Activating autophagy has been successful in preclinical models of Parkinson's disease and yields promising results in clinical trials.
Collapse
Affiliation(s)
- Alan J Fowler
- Department of Neurology, Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, Room 203-C, Building D, 4000 Reservoir Rd. NW, Washington, DC, USA
| | - Charbel E-H Moussa
- Department of Neurology, Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, Room 203-C, Building D, 4000 Reservoir Rd. NW, Washington, DC, USA.
| |
Collapse
|
42
|
Methods for monitoring Ca 2+ and ion channels in the lysosome. Cell Calcium 2017; 64:20-28. [DOI: 10.1016/j.ceca.2016.12.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Revised: 12/07/2016] [Accepted: 12/07/2016] [Indexed: 12/22/2022]
|
43
|
Abstract
Across all kingdoms in the tree of life, calcium (Ca2+) is an essential element used by cells to respond and adapt to constantly changing environments. In multicellular organisms, it plays fundamental roles during fertilization, development and adulthood. The inability of cells to regulate Ca2+ can lead to pathological conditions that ultimately culminate in cell death. One such pathological condition is manifested in Parkinson's disease, the second most common neurological disorder in humans, which is characterized by the aggregation of the protein, α-synuclein. This Review discusses current evidence that implicates Ca2+ in the pathogenesis of Parkinson's disease. Understanding the mechanisms by which Ca2+ signaling contributes to the progression of this disease will be crucial for the development of effective therapies to combat this devastating neurological condition.
Collapse
Affiliation(s)
- Sofia V Zaichick
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Kaitlyn M McGrath
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Gabriela Caraveo
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
44
|
Kavčič N, Pegan K, Turk B. Lysosomes in programmed cell death pathways: from initiators to amplifiers. Biol Chem 2017; 398:289-301. [DOI: 10.1515/hsz-2016-0252] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 09/05/2016] [Indexed: 01/19/2023]
Abstract
Abstract
Lysosome is the central organelle for intracellular degradation of biological macromolecules and organelles. The material destined for degradation enters the lysosomes primarily via endocytosis, autophagy and phagocytosis, and is degraded through the concerted action of more than 50 lysosomal hydrolases. However, lysosomes are also linked with numerous other processes, including cell death, inflammasome activation and immune response, as well as with lysosomal secretion and cholesterol recycling. Among them programmed cell death pathways including apoptosis have received major attention. In most of these pathways, cell death was accompanied by lysosomal membrane permeabilization and release of lysosomal constituents with an involvement of lysosomal hydrolases, including the cathepsins. However, it is less clear, whether lysosomal membrane permeabilization is really critical for the initiation of cell death programme(s). Therefore, the role of lysosomal membrane permeabilization in various programmed cell death pathways is reviewed, as well as the mechanisms leading to it.
Collapse
|
45
|
Salem ESB, Fan GC. Pathological Effects of Exosomes in Mediating Diabetic Cardiomyopathy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 998:113-138. [PMID: 28936736 DOI: 10.1007/978-981-10-4397-0_8] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Diabetic subjects are at risk of developing cardiovascular disease, which accounts for 60-80% of diabetes-related mortality. Atherosclerosis is still considered as a leading cause of heart failure in diabetic patients, but it could also be an intrinsic and long-term effect of contractile cardiac cells malfunction, known as diabetic cardiomyopathy (DCM). Pathologically, this cardiac dysfunction is manifested by inflammation, apoptosis, fibrosis, hypertrophy and altered cardiomyocytes metabolism. However, the underlying molecular mechanisms of DCM pathophysiology are not clearly understood. Recent and several studies have suggested that exosomes are contributed to the regulation of cell-to-cell communication. Therefore, their in-depth investigation can interpret the complex pathophysiology of DCM. Structurally, exosomes are membrane-bounded vesicles (10-200 nm in diameter), which are actively released from all types of cells and detected in all biological fluids. They carry a wide array of bioactive molecules, including mRNAs, none-coding RNAs (e.g., microRNAs, lncRNAs, circRNAs, etc), proteins and lipids. Importantly, the abundance and nature of loaded molecules inside exosomes fluctuate with cell types and pathological conditions. This chapter summarizes currently available studies on the exosomes' role in the regulation of diabetic cardiomyopathy. Specifically, the advances on the pathological effects of exosomes in diabetic cardiomyopathy as well as the therapeutic potentials and perspectives are also discussed.
Collapse
Affiliation(s)
- Esam S B Salem
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, 5872 Care Mail Loc-0575, Cincinnati, OH, 45267, USA
| | - Guo-Chang Fan
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, 5872 Care Mail Loc-0575, Cincinnati, OH, 45267, USA.
| |
Collapse
|
46
|
Evans AM. Nanojunctions of the Sarcoplasmic Reticulum Deliver Site- and Function-Specific Calcium Signaling in Vascular Smooth Muscles. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2016; 78:1-47. [PMID: 28212795 DOI: 10.1016/bs.apha.2016.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Vasoactive agents may induce myocyte contraction, dilation, and the switch from a contractile to a migratory-proliferative phenotype(s), which requires changes in gene expression. These processes are directed, in part, by Ca2+ signals, but how different Ca2+ signals are generated to select each function is enigmatic. We have previously proposed that the strategic positioning of Ca2+ pumps and release channels at membrane-membrane junctions of the sarcoplasmic reticulum (SR) demarcates cytoplasmic nanodomains, within which site- and function-specific Ca2+ signals arise. This chapter will describe how nanojunctions of the SR may: (1) define cytoplasmic nanospaces about the plasma membrane, mitochondria, contractile myofilaments, lysosomes, and the nucleus; (2) provide for functional segregation by restricting passive diffusion and by coordinating active ion transfer within a given nanospace via resident Ca2+ pumps and release channels; (3) select for contraction, relaxation, and/or changes in gene expression; and (4) facilitate the switch in myocyte phenotype through junctional reorganization. This should serve to highlight the need for further exploration of cellular nanojunctions and the mechanisms by which they operate, that will undoubtedly open up new therapeutic horizons.
Collapse
Affiliation(s)
- A M Evans
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
47
|
Endocytic regulation of cytokine receptor signaling. Cytokine Growth Factor Rev 2016; 32:63-73. [DOI: 10.1016/j.cytogfr.2016.07.002] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 07/13/2016] [Indexed: 12/11/2022]
|
48
|
Shorter SA, Gollings AS, Gorringe-Pattrick MAM, Coakley JE, Dyer PDR, Richardson SCW. The potential of toxin-based drug delivery systems for enhanced nucleic acid therapeutic delivery. Expert Opin Drug Deliv 2016; 14:685-696. [DOI: 10.1080/17425247.2016.1227781] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
49
|
Xu W, Zeng Z, Jiang JH, Chang YT, Yuan L. Wahrnehmung der chemischen Prozesse in einzelnen Organellen mit niedermolekularen Fluoreszenzsonden. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201510721] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Wang Xu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering; Hunan University; Changsha 410082 Volksrepublik China
- Department of Chemistry and Medicinal Chemistry Programme; National University of Singapore; Singapore 117543 Singapur
- Laboratory of Bioimaging Probe Development, A*STAR; Singapur
- Department of Chemistry; Stanford University; USA
| | - Zebing Zeng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering; Hunan University; Changsha 410082 Volksrepublik China
| | - Jian-Hui Jiang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering; Hunan University; Changsha 410082 Volksrepublik China
| | - Young-Tae Chang
- Department of Chemistry and Medicinal Chemistry Programme; National University of Singapore; Singapore 117543 Singapur
- Laboratory of Bioimaging Probe Development, A*STAR; Singapur
| | - Lin Yuan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering; Hunan University; Changsha 410082 Volksrepublik China
| |
Collapse
|
50
|
Xu W, Zeng Z, Jiang JH, Chang YT, Yuan L. Discerning the Chemistry in Individual Organelles with Small-Molecule Fluorescent Probes. Angew Chem Int Ed Engl 2016; 55:13658-13699. [DOI: 10.1002/anie.201510721] [Citation(s) in RCA: 548] [Impact Index Per Article: 60.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Indexed: 12/22/2022]
Affiliation(s)
- Wang Xu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering; Hunan University; Changsha 410082 P.R. China
- Department of Chemistry and Medicinal Chemistry Programme; National University of Singapore; Singapore 117543 Singapore
- Laboratory of Bioimaging Probe Development, A*STAR; Singapore
- Department of Chemistry; Stanford University; USA
| | - Zebing Zeng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering; Hunan University; Changsha 410082 P.R. China
| | - Jian-Hui Jiang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering; Hunan University; Changsha 410082 P.R. China
| | - Young-Tae Chang
- Department of Chemistry and Medicinal Chemistry Programme; National University of Singapore; Singapore 117543 Singapore
- Laboratory of Bioimaging Probe Development, A*STAR; Singapore
| | - Lin Yuan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering; Hunan University; Changsha 410082 P.R. China
| |
Collapse
|