1
|
Deri E, Kumar Ojha S, Kartawy M, Khaliulin I, Amal H. Multi-omics study reveals differential expression and phosphorylation of autophagy-related proteins in autism spectrum disorder. Sci Rep 2025; 15:10878. [PMID: 40158064 PMCID: PMC11954894 DOI: 10.1038/s41598-025-95860-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
Our multi-omics study investigated the molecular mechanisms underlying autism spectrum disorder (ASD) using Shank3Δ4-22 and Cntnap2-/- mouse models. Through global- and phospho- proteomics of the mouse cortex, we focused on shared molecular changes and found that autophagy was particularly affected in both models. Global proteomics identified a small number of differentially expressed proteins that significantly impact postsynaptic components and synaptic function, including key pathways such as mTOR signaling. Phosphoproteomics revealed unique phosphorylation sites in autophagy-related proteins such as ULK2, RB1CC1, ATG16L1, and ATG9, suggesting that altered phosphorylation patterns contribute to impaired autophagic flux in ASD. SH-SY5Y cells with SHANK3 gene deletion showed elevated LC3-II and p62 levels, indicating autophagosome accumulation and autophagy initiation, while the reduced level of the lysosomal activity marker LAMP1 suggested impaired autophagosome-lysosome fusion. The study highlights the involvement of reactive nitrogen species and nitric oxide (NO) on autophagy disruption. Importantly, inhibition of neuronal NO synthase (nNOS) by 7-NI normalized autophagy markers levels in the SH-SY5Y cells and primary cultured neurons. We have previously shown that nNOS inhibition improved synaptic and behavioral phenotypes in Shank3Δ4-22 and Cntnap2-/- mouse models. Our multi-omics study reveals differential expression and phosphorylation of autophagy-related proteins in ASD but further investigation is needed to prove the full involvement of autophagy in ASD. Our study underscores the need for further examination into the functional consequences of the identified phosphorylation sites, which may offer potential novel therapeutic autophagy-related targets for ASD treatment.
Collapse
Affiliation(s)
- Eden Deri
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shashank Kumar Ojha
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Maryam Kartawy
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Igor Khaliulin
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, MA, USA.
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Wang L, Sooram B, Kumar R, Schedin-Weiss S, Tjernberg LO, Winblad B. Tau degradation in Alzheimer's disease: Mechanisms and therapeutic opportunities. Alzheimers Dement 2025; 21:e70048. [PMID: 40109019 PMCID: PMC11923393 DOI: 10.1002/alz.70048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/05/2025] [Accepted: 02/05/2025] [Indexed: 03/22/2025]
Abstract
In Alzheimer's disease (AD), tau undergoes abnormal post-translational modifications and aggregations. Impaired intracellular degradation pathways further exacerbate the accumulation of pathological tau. A new strategy - targeted protein degradation - recently emerged as a modality in drug discovery where bifunctional molecules bring the target protein close to the degradation machinery to promote clearance. Since 2016, this strategy has been applied to tau pathologies and attracted broad interest in academia and the pharmaceutical industry. However, a systematic review of recent studies on tau degradation mechanisms is lacking. Here we review tau degradation mechanisms (the ubiquitin-proteasome system and the autophagy-lysosome pathway), their dysfunction in AD, and tau-targeted degraders, such as proteolysis-targeting chimeras and autophagy-targeting chimeras. We emphasize the need for a continuous exploration of tau degradation mechanisms and provide a future perspective for developing tau-targeted degraders, encouraging researchers to work on new treatment options for AD patients. HIGHLIGHTS: Post-translational modifications, aggregation, and mutations affect tau degradation. A vicious circle exists between impaired degradation pathways and tau pathologies. Ubiquitin plays an important role in complex degradation pathways. Tau-targeted degraders provide promising strategies for novel AD treatment.
Collapse
Affiliation(s)
- Lisha Wang
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Banesh Sooram
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Rajnish Kumar
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, India
| | - Sophia Schedin-Weiss
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Lars O Tjernberg
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Bengt Winblad
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Huddinge, Sweden
| |
Collapse
|
3
|
Han L, Chen W, Li J, Zhao Y, Zong Y, He Z, Du R. Palmatine improves cognitive dysfunction in Alzheimer's disease model rats through autophagy pathway and regulation of gut microbiota. Brain Res 2024; 1835:148932. [PMID: 38609032 DOI: 10.1016/j.brainres.2024.148932] [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: 07/18/2023] [Revised: 03/17/2024] [Accepted: 04/09/2024] [Indexed: 04/14/2024]
Abstract
Alzheimer's disease (AD) is a primary degenerative encephalopathy that first appeared as a decline in memory and learning skills. Over time, the condition's severity grew. Palmatine (Pal) alleviates Alzheimer's disease symptoms, which has neuroprotective benefits. Numerous investigations have demonstrated a close relationship among AD and gut structure changes. The aim of the research was investigating whether the improvement of Pal on AD is linked to regulating gut flora and autophagy. First, we used Aβ1-40 to induce apoptosis in HT22 cells. After Pal treatment, apoptosis can be improved. Then, We used bilateral intracranial hippocampal injection of Aβ1-40 for establishing the AD model, after treatment with Pal, the morris water maze experiment and eight-arm maze test demonstrated that Pal enhanced the AD rats' capacity for learning and memory, HE staining illustrated that Pal improved the morphological abnormalities of brain cells and gut tissue damage. Pal reduced the death of hippocampus neurons, as shown by Nissl staining. Pal substantially reduced Tau hyperphosphorylation and Aβ accumulation in the brain, according to immunohistochemical labelling. Pal improved the expression of LC3, Beclin 1, AMPK, and suppressed the expression of mTOR and P62, as validated by RT-qPCR and immunofluorescence labelling. This suggests that Pal's treatment of AD may be associated with the control of the AMPK/mTOR autophagy signalling system. 16S rRNA sequencing and short-chain fatty acids (SCFAs) content detection analysis illustrated that Pal has the potential to enhance the content of SCFAs, reverse the alterations in gut microorganisms. It has been showed by the study that Pal could improve AD by activating autophagy signaling pathway and improving gut barrier changes.
Collapse
Affiliation(s)
- Lu Han
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Weijia Chen
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Jianming Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Yan Zhao
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Ying Zong
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Zhongmei He
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China; Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education of China, Changchun 130118, China; Jilin Provincial Engineering Research Center for Efficient Breeding and Product Development of Sika Deer of China, Changchun 130118, China.
| | - Rui Du
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China; Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education of China, Changchun 130118, China; Jilin Provincial Engineering Research Center for Efficient Breeding and Product Development of Sika Deer of China, Changchun 130118, China.
| |
Collapse
|
4
|
Schreiber T, Koll N, Padberg C, de los Reyes B, Quinting T, Malyshkina A, Metzen E, Sutter K, Fandrey J, Winning S. Reduced vacuolar ATPase protects mice from Friend virus infection - an unintended but instructive effect in Hif-2afl mice. J Cell Sci 2024; 137:jcs261893. [PMID: 38856651 PMCID: PMC11234382 DOI: 10.1242/jcs.261893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 05/20/2024] [Indexed: 06/11/2024] Open
Abstract
During acute viral infections, innate immune cells invade inflamed tissues and face hypoxic areas. Hypoxia-inducible factors (HIFs) adapt cellular responses towards these conditions. We wanted to investigate the effects of a loss of HIF-2α in macrophages during acute Friend murine leukemia retrovirus (FV) infection in C57BL/6 mice using a Cre/loxP system. Remarkably, mice with floxed Hif-2a (Hif-2afl; Hif-2a is also known as Epas1) did not show any signs of FV infection independent of Cre activity. This prevented a detailed analysis of the role of macrophage HIF-2α for FV infection but allowed us to study a model of unexpected FV resistance. Hif-2afl mice showed a significant decrease in the expression of the Atp6v1e2 gene encoding for the E2 subunit of the vacuolar H+-ATPase, which resulted in a decreased acidification of lysosomes and limited virus entry into the cell. These findings highlight that the insertion of loxP sites is not always without functional consequences and has established a phenotype in the floxed Hif-2a mouse, which is not only unexpected, but unwanted and is of relevance for the use of this mouse strain in (at least virus) experiments.
Collapse
Affiliation(s)
- Timm Schreiber
- Institute of Physiology, University of Duisburg-Essen, 45147 Essen, Germany
- Institute of Physiology, Pathophysiology and Toxicology and Center for Biomedical Education and Research (ZBAF), University of Witten/Herdecke, 58455 Witten, Germany
| | - Nora Koll
- Institute of Physiology, University of Duisburg-Essen, 45147 Essen, Germany
| | - Claudia Padberg
- Institute of Physiology, University of Duisburg-Essen, 45147 Essen, Germany
| | - Buena de los Reyes
- Institute of Physiology, University of Duisburg-Essen, 45147 Essen, Germany
| | - Theresa Quinting
- Institute of Physiology, University of Duisburg-Essen, 45147 Essen, Germany
| | - Anna Malyshkina
- Institute of Physiology, University of Duisburg-Essen, 45147 Essen, Germany
| | - Eric Metzen
- Institute of Physiology, University of Duisburg-Essen, 45147 Essen, Germany
| | - Kathrin Sutter
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
- Institute for Research on HIV and AIDS-associated Diseases, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Joachim Fandrey
- Institute of Physiology, University of Duisburg-Essen, 45147 Essen, Germany
| | - Sandra Winning
- Institute of Physiology, University of Duisburg-Essen, 45147 Essen, Germany
| |
Collapse
|
5
|
Liu H, Li C, Zhang X, Chen H, Zhang Q, Zeng Y, Zheng S, Zou J, Zhao Y, Zheng X, Huang G, Zeng Q. BMSC-Exosomes attenuate ALP dysfunction by restoring lysosomal function via the mTOR/TFEB Axis to reduce cerebral ischemia-reperfusion injury. Exp Neurol 2024; 376:114726. [PMID: 38403042 DOI: 10.1016/j.expneurol.2024.114726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 01/02/2024] [Accepted: 02/12/2024] [Indexed: 02/27/2024]
Abstract
BACKGROUND The complex pathophysiological changes following cerebral ischemia-reperfusion injury (CIRI) include the accumulation of defective proteins and damaged organelles, which cause massive neuron demise. To preserve cellular homeostasis, the autophagy-lysosomal pathway (ALP) is crucial for neurons to dispose of these substances. Many studies have shown that bone mesenchymal stem cell exosomes (BMSC-Exos) can reduce CIRI. However, the specific mechanisms have not been well elucidated, a fact that limits its widespread clinical use. This study aimed to clarify whether BMSC-Exos could attenuate ALP dysfunction by restoring lysosomal function after CIRI via inhibiting mTOR and then activating TFEB nucleus translocation. METHODS In this study, Flow cytometry, Nanoparticle tracking analysis (NTA), Transmission electron microscope (TEM), and Western blot were used to identify the BMSCs and BMSC-Exos used in this experiment as conforming to the requirements. In vivo experiments, SD rats were modeled with temporary middle cerebral artery occlusion (tMCAO), and BMSC-Exos was injected into the tail vein 2 h after modeling. Triphenyl tetrazolium chloride (TTC) staining, modified neurological severity scores (mNSS), corner turn test, and rotating rod test were used to detect neurological deficits in rats after BMSC-Exos intervention. Western blot and Immunofluorescence were used to detect ALP, transcription factor EB(TFEB) nucleus translocation, and mammalian target of rapamycin (mTOR) change at different time points after modeling and after BMSC-Exos intervention. In vitro experiments, pheochromocytoma cells (PC12) cells were subjected to oxygen-glucose deprivation and reperfusion (OGD/R) modeling to mimic CIRI, and were respectively intervened with BMSC-Exos, BMSC-Exos + MHY 1485 (the mTOR agonist), Rapamycin (the mTOR inhibitor). CCK8, Western blot, and Immunofluorescence were used to detect PC12 cell survival, TFEB nucleus translocation, and cathepsin B(CTSB) Immunofluorescence intensity. RESULTS We found that ALP dysfunction occurred 72 h after tMCAO, and BMSC-Exos can attenuate ALP dysfunction by restoring lysosomal function. Next, we examined TFEB nucleus translocation and the expression of mTOR, a key regulator of translocation. We found that BMSC-Exos could inhibit mTOR and activate TFEB nucleus translocation. Additional in vitro tests revealed that BMSC-Exos could increase PC12 cell survival after OGD/R, activating TFEB nucleus translocation and enhancing the fluorescence intensity of CTSB, which in turn could be reversed by the mTOR agonist, MHY1485. This effect was similar to another mTOR inhibitor, Rapamycin. CONCLUSION BMSC-Exos could attenuate ALP dysfunction by restoring lysosomal function after CIRI by inhibiting mTOR and then promoting TFEB nucleus translocation.
Collapse
Affiliation(s)
- Haining Liu
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China; School of Rehabilitation Sciences, Southern Medical University, Guangdong 510515, China
| | - Chen Li
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China; School of Rehabilitation Sciences, Southern Medical University, Guangdong 510515, China; Department of Rehabilitation Medicine, Hunan Provincial People's Hospital, Hunan Normal University, Changsha 410016, China
| | - Xiaofeng Zhang
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China; School of Rehabilitation Sciences, Southern Medical University, Guangdong 510515, China
| | - Hui Chen
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China; School of Rehabilitation Sciences, Southern Medical University, Guangdong 510515, China
| | - Qi Zhang
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China; School of Rehabilitation Sciences, Southern Medical University, Guangdong 510515, China
| | - Yuting Zeng
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China; School of Rehabilitation Sciences, Southern Medical University, Guangdong 510515, China
| | - Shuqi Zheng
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China; School of Rehabilitation Sciences, Southern Medical University, Guangdong 510515, China
| | - Jihua Zou
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China; School of Rehabilitation Sciences, Southern Medical University, Guangdong 510515, China; The Hong Kong Polytechnic University, Faculty of Health and Social Sciences, Hong Kong
| | - Yijin Zhao
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China; School of Rehabilitation Sciences, Southern Medical University, Guangdong 510515, China
| | - Xiaoyan Zheng
- School of Rehabilitation Sciences, Southern Medical University, Guangdong 510515, China.
| | - Guozhi Huang
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China; School of Rehabilitation Sciences, Southern Medical University, Guangdong 510515, China.
| | - Qing Zeng
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China; School of Rehabilitation Sciences, Southern Medical University, Guangdong 510515, China.
| |
Collapse
|
6
|
Noblejas-López MDM, Tébar-García D, López-Rosa R, Alcaraz-Sanabria A, Cristóbal-Cueto P, Pinedo-Serrano A, Rivas-García L, Galán-Moya EM. TACkling Cancer by Targeting Selective Protein Degradation. Pharmaceutics 2023; 15:2442. [PMID: 37896202 PMCID: PMC10610449 DOI: 10.3390/pharmaceutics15102442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/04/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
Targeted protein degradation has emerged as an alternative therapy against cancer, offering several advantages over traditional inhibitors. The new degrader drugs provide different therapeutic strategies: they could cross the phospholipid bilayer membrane by the addition of specific moieties to extracellular proteins. On the other hand, they could efficiently improve the degradation process by the generation of a ternary complex structure of an E3 ligase. Herein, we review the current trends in the use of TAC-based technologies (TACnologies), such as PROteolysis TArgeting Chimeras (PROTAC), PHOtochemically TArgeting Chimeras (PHOTAC), CLIck-formed Proteolysis TArgeting Chimeras (CLIPTAC), AUtophagy TArgeting Chimeras (AUTAC), AuTophagosome TEthering Compounds (ATTEC), LYsosome-TArgeting Chimeras (LYTAC), and DeUBiquitinase TArgeting Chimeras (DUBTAC), in experimental development and their progress towards clinical applications.
Collapse
Affiliation(s)
- María del Mar Noblejas-López
- Centro Regional de Investigaciones Biomédicas (CRIB), Campus de Albacete, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (M.d.M.N.-L.); (D.T.-G.); (R.L.-R.); (A.A.-S.); (P.C.-C.); (A.P.-S.)
- Unidad de Investigación, Complejo Hospitalario Universitario de Albacete, 02008 Albacete, Spain
| | - David Tébar-García
- Centro Regional de Investigaciones Biomédicas (CRIB), Campus de Albacete, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (M.d.M.N.-L.); (D.T.-G.); (R.L.-R.); (A.A.-S.); (P.C.-C.); (A.P.-S.)
- Unidad de Investigación, Complejo Hospitalario Universitario de Albacete, 02008 Albacete, Spain
| | - Raquel López-Rosa
- Centro Regional de Investigaciones Biomédicas (CRIB), Campus de Albacete, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (M.d.M.N.-L.); (D.T.-G.); (R.L.-R.); (A.A.-S.); (P.C.-C.); (A.P.-S.)
- Unidad de Investigación, Complejo Hospitalario Universitario de Albacete, 02008 Albacete, Spain
| | - Ana Alcaraz-Sanabria
- Centro Regional de Investigaciones Biomédicas (CRIB), Campus de Albacete, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (M.d.M.N.-L.); (D.T.-G.); (R.L.-R.); (A.A.-S.); (P.C.-C.); (A.P.-S.)
- Unidad de Investigación, Complejo Hospitalario Universitario de Albacete, 02008 Albacete, Spain
| | - Pablo Cristóbal-Cueto
- Centro Regional de Investigaciones Biomédicas (CRIB), Campus de Albacete, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (M.d.M.N.-L.); (D.T.-G.); (R.L.-R.); (A.A.-S.); (P.C.-C.); (A.P.-S.)
| | - Alejandro Pinedo-Serrano
- Centro Regional de Investigaciones Biomédicas (CRIB), Campus de Albacete, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (M.d.M.N.-L.); (D.T.-G.); (R.L.-R.); (A.A.-S.); (P.C.-C.); (A.P.-S.)
| | - Lorenzo Rivas-García
- Centro Regional de Investigaciones Biomédicas (CRIB), Campus de Albacete, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (M.d.M.N.-L.); (D.T.-G.); (R.L.-R.); (A.A.-S.); (P.C.-C.); (A.P.-S.)
| | - Eva M. Galán-Moya
- Centro Regional de Investigaciones Biomédicas (CRIB), Campus de Albacete, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (M.d.M.N.-L.); (D.T.-G.); (R.L.-R.); (A.A.-S.); (P.C.-C.); (A.P.-S.)
- Unidad de Investigación, Complejo Hospitalario Universitario de Albacete, 02008 Albacete, Spain
- Facultad de Enfermería, Campus de Albacete, Universidad de Castilla-La Mancha, 02006 Albacete, Spain
| |
Collapse
|
7
|
Obergasteiger J, Castonguay AM, Pizzi S, Magnabosco S, Frapporti G, Lobbestael E, Baekelandt V, Hicks AA, Pramstaller PP, Gravel C, Corti C, Lévesque M, Volta M. The small GTPase Rit2 modulates LRRK2 kinase activity, is required for lysosomal function and protects against alpha-synuclein neuropathology. NPJ Parkinsons Dis 2023; 9:44. [PMID: 36973269 PMCID: PMC10042831 DOI: 10.1038/s41531-023-00484-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 03/06/2023] [Indexed: 03/29/2023] Open
Abstract
In Parkinson's disease (PD) misfolded alpha-synuclein (aSyn) accumulates in the substantia nigra, where dopaminergic neurons are progressively lost. The mechanisms underlying aSyn pathology are still unclear, but they are hypothesized to involve the autophagy-lysosome pathway (ALP). LRRK2 mutations are a major cause of familial and sporadic PD, and LRRK2 kinase activity has been shown to be involved in pS129-aSyn inclusion modulation. We observed selective downregulation of the novel PD risk factor RIT2 in vitro and in vivo. Rit2 overexpression in G2019S-LRRK2 cells rescued ALP abnormalities and diminished aSyn inclusions. In vivo, viral mediated overexpression of Rit2 operated neuroprotection against AAV-A53T-aSyn. Furthermore, Rit2 overexpression prevented the A53T-aSyn-dependent increase of LRRK2 kinase activity in vivo. On the other hand, reduction of Rit2 levels leads to defects in the ALP, similar to those induced by the G2019S-LRRK2 mutation. Our data indicate that Rit2 is required for correct lysosome function, inhibits overactive LRRK2 to ameliorate ALP impairment, and counteracts aSyn aggregation and related deficits. Targeting Rit2 could represent an effective strategy to combat neuropathology in familial and idiopathic PD.
Collapse
Affiliation(s)
- Julia Obergasteiger
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Volta 21, 39100, Bolzano, Italy
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, CERVO Brain Research Centre, 2601 Chemin de la Canardiere, Quebec, QC, Canada
| | - Anne-Marie Castonguay
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, CERVO Brain Research Centre, 2601 Chemin de la Canardiere, Quebec, QC, Canada
| | - Sara Pizzi
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Volta 21, 39100, Bolzano, Italy
| | - Stefano Magnabosco
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Volta 21, 39100, Bolzano, Italy
| | - Giulia Frapporti
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Volta 21, 39100, Bolzano, Italy
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Evy Lobbestael
- Department of Neurosciences, KU Leuven, Herestraat 49 bus 1023, 3000, Leuven, Belgium
| | - Veerle Baekelandt
- Department of Neurosciences, KU Leuven, Herestraat 49 bus 1023, 3000, Leuven, Belgium
| | - Andrew A Hicks
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Volta 21, 39100, Bolzano, Italy
| | - Peter P Pramstaller
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Volta 21, 39100, Bolzano, Italy
| | - Claude Gravel
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, CERVO Brain Research Centre, 2601 Chemin de la Canardiere, Quebec, QC, Canada
| | - Corrado Corti
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Volta 21, 39100, Bolzano, Italy
| | - Martin Lévesque
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, CERVO Brain Research Centre, 2601 Chemin de la Canardiere, Quebec, QC, Canada.
| | - Mattia Volta
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Volta 21, 39100, Bolzano, Italy.
| |
Collapse
|
8
|
Kim KR, Park SE, Hong JY, Koh JY, Cho DH, Hwang JJ, Kim YH. Zinc enhances autophagic flux and lysosomal function through transcription factor EB activation and V-ATPase assembly. Front Cell Neurosci 2022; 16:895750. [PMID: 36246521 PMCID: PMC9558701 DOI: 10.3389/fncel.2022.895750] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
The stimulation of autophagy or lysosomes has been considered therapeutic for neurodegenerative disorders because the accumulation of misfolded proteins is commonly observed in the brains of individuals with these diseases. Although zinc is known to play critical roles in the functions of lysosomes and autophagy, the mechanism behind this regulatory relationship remains unclear. Therefore, in this study, we examined which mechanism is involved in zinc-mediated activation of autophagy and lysosome. Exposure to zinc at a sub-lethal concentration activated autophagy in a concentration-dependent manner in mRFP-GFP-LC3-expressing H4 glioma cells. Zinc also rescued the blocking of autophagic flux arrested by pharmaceutical de-acidification. Co-treatment with zinc attenuated the chloroquine (CQ)-induced increase in the number and size of mRFP-GFP-LC3 puncta in H4 cells and accumulation of p62 by CQ or ammonium chloride in both H4 and mouse cerebrocortical cultures. Zinc rapidly induced the expression of cathepsin B (CTSB) and cathepsin D (CTSD), representative lysosomal proteases in neurons, which appeared likely to be mediated by transcription factor EB (TFEB). We observed the translocation of TFEB from neurite to nucleus and the dephosphorylation of TFEB by zinc. The addition of cycloheximide, a chemical inhibitor of protein synthesis, inhibited the activity of CTSB and CTSD at 8 h after zinc exposure but not at 1 h, indicating that only late lysosomal activation was dependent on the synthesis of CTSB and CTSD proteins. At the very early time point, the activation of cathepsins was mediated by an increased assembly of V-ATPase on lysosomes and resultant lysosomal acidification. Finally, considering that P301L mutation in tau protein causes frontotemporal dementia through aggressive tau accumulation, we investigated whether zinc reduces the accumulation of protein aggregates in SK-N-BE(2)-C neuroblastoma cells expressing wild-type tau or mutant P301L-tau. Zinc markedly attenuated the levels of phosphorylated tau and total tau as well as p62 in both wild-type and mutant tau-overexpressing cells. We also observed that zinc was more effective than rapamycin at inducing TFEB-dependent CTSB and CTSD expression and V-ATPase-dependent lysosomal acidification and CTSB/CTSD activation. These results suggest that the regulation of zinc homeostasis could be a new approach for developing treatments for neurodegenerative diseases, including Alzheimer’s and Parkinson’s.
Collapse
Affiliation(s)
- Ki-Ryeong Kim
- Department of Integrative Bioscience and Biotechnology, Sejong University, Seoul, South Korea
| | - Sang Eun Park
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, South Korea
| | - Ji-Ye Hong
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, South Korea
| | - Jae-Young Koh
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
- Neuronal Injury Lab, Biomedical Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, South Korea
| | - Dong-Hyung Cho
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu, South Korea
| | - Jung Jin Hwang
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, South Korea
- Jung Jin Hwang,
| | - Yang-Hee Kim
- Department of Integrative Bioscience and Biotechnology, Sejong University, Seoul, South Korea
- *Correspondence: Yang-Hee Kim,
| |
Collapse
|
9
|
Cai L, Yoon JD, Hwang SU, Lee J, Kim E, Kim M, Hyun SY, Choi H, Oh D, Jeon Y, Hyun SH. Exploring the mechanism of trehalose: dual functions of PI3K/Akt and VPS34/mTOR pathways in porcine oocytes and cumulus cells†. Biol Reprod 2022; 107:432-445. [PMID: 35348612 DOI: 10.1093/biolre/ioac060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 10/25/2021] [Accepted: 03/18/2022] [Indexed: 11/14/2022] Open
Abstract
Autophagy, an intracellular recycling system, is essential for the meiotic maturation of porcine oocytes. Multiple studies, sought to reveal the precise mechanism employed, commonly used autophagy inducers, such as rapamycin, which is a mammalian target of rapamycin (mTOR) inhibitor. However, it has a limitation as mTOR plays various roles in cell growth and metabolism beyond autophagy. Trehalose has been reported as a novel mTOR-independent autophagy inducer in many cells. Furthermore, our previous study demonstrated that trehalose supplementation during in vitro maturation of porcine oocytes improves the developmental competence of parthenogenetic embryos possibly via autophagic activation, whereas the underlying mechanisms remain unclear. Therefore, the aim of this study was to address this issue. In this study, we found that trehalose plays a role as an autophagy activator by autophagic flux assay and determined that it promotes PI3K/Akt inhibition and VPS34/mTOR activation by immunoblotting, both in cumulus cells (CCs) and oocytes. However, it is interesting to note that these effects caused by trehalose were worked totally varying between CCs and oocytes. In CCs, the autophagy was activated through the improvement of lysosomal function/autophagic clearance viability by upregulation of coordinated lysosomal expression and regulation genes via PI3K/Akt inhibition. Whereas in oocytes, autophagy was activated via VPS34 induction which directly influences autophagosome formation, and the precise meiotic process was ensured via Akt inhibition and mTOR activation. Taken together, this study provided evidence that trehalose could be used as an autophagy inductor during porcine oocyte maturation based on the revealed mechanism.
Collapse
Affiliation(s)
- Lian Cai
- College of Veterinary Medicine, Chungbuk National University, Cheongju, 28644, South Korea.,Institute for Stem cell & Regenerative Medicine (ISCRM), Chungbuk National University, Chengju 28644, Republic of Korea.,Graduate School of Veterinary Biosecurity and Protection, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Junchul David Yoon
- College of Veterinary Medicine, Chungbuk National University, Cheongju, 28644, South Korea.,Institute for Stem cell & Regenerative Medicine (ISCRM), Chungbuk National University, Chengju 28644, Republic of Korea
| | - Seon-Ung Hwang
- College of Veterinary Medicine, Chungbuk National University, Cheongju, 28644, South Korea.,Institute for Stem cell & Regenerative Medicine (ISCRM), Chungbuk National University, Chengju 28644, Republic of Korea
| | - Joohyeong Lee
- College of Veterinary Medicine, Chungbuk National University, Cheongju, 28644, South Korea.,Institute for Stem cell & Regenerative Medicine (ISCRM), Chungbuk National University, Chengju 28644, Republic of Korea
| | - Eunhye Kim
- College of Veterinary Medicine, Chungbuk National University, Cheongju, 28644, South Korea.,Graduate School of Veterinary Biosecurity and Protection, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Mirae Kim
- College of Veterinary Medicine, Chungbuk National University, Cheongju, 28644, South Korea.,Institute for Stem cell & Regenerative Medicine (ISCRM), Chungbuk National University, Chengju 28644, Republic of Korea
| | - Saang-Yoon Hyun
- College of Fisheries Sciences, Pukyong National University, Busan 48513, Republic of Korea
| | - Hyerin Choi
- College of Veterinary Medicine, Chungbuk National University, Cheongju, 28644, South Korea.,Institute for Stem cell & Regenerative Medicine (ISCRM), Chungbuk National University, Chengju 28644, Republic of Korea
| | - Dongjin Oh
- College of Veterinary Medicine, Chungbuk National University, Cheongju, 28644, South Korea.,Institute for Stem cell & Regenerative Medicine (ISCRM), Chungbuk National University, Chengju 28644, Republic of Korea
| | - Yubyeol Jeon
- Department of Theriogenology and Reproductive Biotechnology, College of Veterinary Medicine and Bio-safety Research Institute, Jeonbuk National University, Iksan 54596, Republic of Korea
| | - Sang-Hwan Hyun
- College of Veterinary Medicine, Chungbuk National University, Cheongju, 28644, South Korea.,Institute for Stem cell & Regenerative Medicine (ISCRM), Chungbuk National University, Chengju 28644, Republic of Korea.,Graduate School of Veterinary Biosecurity and Protection, Chungbuk National University, Cheongju 28644, Republic of Korea
| |
Collapse
|
10
|
Sepúlveda D, Cisternas-Olmedo M, Arcos J, Nassif M, Vidal RL. Contribution of Autophagy-Lysosomal Pathway in the Exosomal Secretion of Alpha-Synuclein and Its Impact in the Progression of Parkinson’s Disease. Front Mol Neurosci 2022; 15:805087. [PMID: 35250476 PMCID: PMC8891570 DOI: 10.3389/fnmol.2022.805087] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 01/07/2022] [Indexed: 01/07/2023] Open
Abstract
Parkinson’s disease (PD) is caused by the degeneration of dopaminergic neurons due to an accumulation of intraneuronal abnormal alpha-synuclein (α-syn) protein aggregates. It has been reported that the levels of exosomal α-syn of neuronal origin in plasma correlate significantly with motor dysfunction, highlighting the exosomes containing α-syn as a potential biomarker of PD. In addition, it has been found that the selective autophagy-lysosomal pathway (ALP) contributes to the secretion of misfolded proteins involved in neurodegenerative diseases. In this review, we describe the evidence that supports the relationship between the ALP and α-syn exosomal secretion on the PD progression and its implications in the diagnosis and progression of this pathology.
Collapse
Affiliation(s)
- Denisse Sepúlveda
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
- Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Marisol Cisternas-Olmedo
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
- Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Javiera Arcos
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
- Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Melissa Nassif
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
- Escuela de Biotecnología, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - René L. Vidal
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
- Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
- *Correspondence: René L. Vidal,
| |
Collapse
|
11
|
Zhou X, Suo F, Haslinger K, Quax WJ. Artemisinin-Type Drugs in Tumor Cell Death: Mechanisms, Combination Treatment with Biologics and Nanoparticle Delivery. Pharmaceutics 2022; 14:395. [PMID: 35214127 PMCID: PMC8875250 DOI: 10.3390/pharmaceutics14020395] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 02/04/2023] Open
Abstract
Artemisinin, the most famous anti-malaria drug initially extracted from Artemisia annua L., also exhibits anti-tumor properties in vivo and in vitro. To improve its solubility and bioavailability, multiple derivatives have been synthesized. However, to reveal the anti-tumor mechanism and improve the efficacy of these artemisinin-type drugs, studies have been conducted in recent years. In this review, we first provide an overview of the effect of artemisinin-type drugs on the regulated cell death pathways, which may uncover novel therapeutic approaches. Then, to overcome the shortcomings of artemisinin-type drugs, we summarize the recent advances in two different therapeutic approaches, namely the combination therapy with biologics influencing regulated cell death, and the use of nanocarriers as drug delivery systems. For the former approach, we discuss the superiority of combination treatments compared to monotherapy in tumor cells based on their effects on regulated cell death. For the latter approach, we give a systematic overview of nanocarrier design principles used to deliver artemisinin-type drugs, including inorganic-based nanoparticles, liposomes, micelles, polymer-based nanoparticles, carbon-based nanoparticles, nanostructured lipid carriers and niosomes. Both approaches have yielded promising findings in vitro and in vivo, providing a strong scientific basis for further study and upcoming clinical trials.
Collapse
Affiliation(s)
| | | | - Kristina Haslinger
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (X.Z.); (F.S.)
| | - Wim J. Quax
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (X.Z.); (F.S.)
| |
Collapse
|
12
|
Filippone A, Li JG, Praticò D. VPS35 Downregulation Alters Degradation Pathways in Neuronal Cells. J Alzheimers Dis 2021; 84:1079-1089. [PMID: 34602481 DOI: 10.3233/jad-210701] [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] [Indexed: 12/11/2022]
Abstract
BACKGROUND The vacuolar protein sorting 35 (VPS35) is the main component of the retromer recognition core complex system which regulates intracellular cargo protein sorting and trafficking. Downregulation of VPS35 has been linked to the pathogenesis of neurodegenerative disorders such Alzheimer's and Parkinson's diseases via endosome dysregulation. OBJECTIVE Here we show that the genetic manipulation of VPS35 affects intracellular degradation pathways. METHODS A neuronal cell line expressing human APP Swedish mutant was used. VPS35 silencing was performed treating cells with VPS35 siRNA or Ctr siRNA for 72 h. RESULTS Downregulation of VPS35 was associated with alteration of autophagy flux and intracellular accumulation of acidic and ubiquitinated aggregates suggesting that dysfunction of the retromer recognition core leads to a significant alteration in both pathways. CONCLUSION Taken together, our data demonstrate that besides cargo sorting and trafficking, VPS35 by supporting the integral function of the retromer complex system plays an important role also as a critical regulator of intracellular degradation pathways.
Collapse
Affiliation(s)
- Alessia Filippone
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Jian-Guo Li
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Domenico Praticò
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
13
|
Rana T, Behl T, Sehgal A, Mehta V, Singh S, Bhatia S, Al-Harrasi A, Bungau S. Exploring the Role of Autophagy Dysfunction in Neurodegenerative Disorders. Mol Neurobiol 2021; 58:4886-4905. [PMID: 34212304 DOI: 10.1007/s12035-021-02472-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/21/2021] [Indexed: 12/12/2022]
Abstract
Autophagy is a catabolic pathway by which misfolded proteins or damaged organelles are engulfed by autophagosomes and then transported to lysosomes for degradation. Recently, a great improvement has been done to explain the molecular mechanisms and roles of autophagy in several important cellular metabolic processes. Besides being a vital clearance pathway or a cell survival pathway in response to different stresses, autophagy dysfunction, either upregulated or down-regulated, has been suggested to be linked with numerous neurodegenerative disorders like Alzheimer's disease, Parkinson's disease, Huntington's disease, and Amyotrophic lateral sclerosis. Impairment at different stages of autophagy results in the formation of large protein aggregates and damaged organelles, which leads to the onset and progression of different neurodegenerative disorders. This article elucidates the recent progress about the role of autophagy in neurodegenerative disorders and explains how autophagy dysfunction is linked with the pathogenesis of such disorders as well as the novel potential autophagy-associated therapies for treating them.
Collapse
Affiliation(s)
- Tarapati Rana
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
- Government Pharmacy College, Seraj, Mandi, Himachal Pradesh, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Vineet Mehta
- Government College of Pharmacy, Rohru, Distt. Shimla, Himachal Pradesh, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Saurabh Bhatia
- Amity Institute of Pharmacy, Amity University, Haryana, India
- Natural & Medical Sciences Research Centre, University of Nizwa, Nizwa, Oman
| | - Ahmed Al-Harrasi
- Natural & Medical Sciences Research Centre, University of Nizwa, Nizwa, Oman
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
14
|
Eshaq RS, Harris NR. The role of tumor necrosis factor-α and interferon-γ in the hyperglycemia-induced ubiquitination and loss of platelet endothelial cell adhesion molecule-1 in rat retinal endothelial cells. Microcirculation 2021; 28:e12717. [PMID: 34008903 PMCID: PMC10078990 DOI: 10.1111/micc.12717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 05/03/2021] [Accepted: 05/11/2021] [Indexed: 11/25/2022]
Abstract
OBJECTIVE This study aimed to investigate the role of the hyperglycemia-induced increase in tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ) in the ubiquitination and degradation of platelet endothelial cell adhesion molecule-1 (PECAM-1) in the diabetic retina. METHODS Type I diabetes was induced in rats by the injection of streptozotocin, with age-matched non-diabetic rats as controls. Primary rat retinal microvascular endothelial cells were grown in normal or high glucose media for 6 days or in normal glucose media for 24 h with addition of TNF-α and/or IFN-γ. PECAM-1, TNF-α, IFN-γ, and ubiquitin levels were assessed using Western blotting, immunofluorescence, and immunoprecipitation assays. Additionally, proteasome activity was assessed both in vivo and in vitro. RESULTS Under hyperglycemic conditions, total ubiquitination levels in the retina and RRMECs, and PECAM-1 ubiquitination levels in RRMECs, were significantly increased. Additionally, TNF-α and IFN-γ levels were significantly increased under hyperglycemic conditions. PECAM-1 levels in RRMECs treated with TNF-α and/or IFN-γ were significantly decreased. Moreover, there was a significant decrease in proteasome activity in the diabetic retina, hyperglycemic RRMECs, and RRMECs treated with TNF-α or IFN-γ. CONCLUSION Tumor necrosis factor-α and IFN-γ may contribute to the hyperglycemia-induced loss of PECAM-1 in retinal endothelial cells, possibly by upregulating PECAM-1 ubiquitination.
Collapse
Affiliation(s)
- Randa S Eshaq
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA
| | - Norman R Harris
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA
| |
Collapse
|
15
|
Wang J, Zhou CJ, Khodabukus A, Tran S, Han SO, Carlson AL, Madden L, Kishnani PS, Koeberl DD, Bursac N. Three-dimensional tissue-engineered human skeletal muscle model of Pompe disease. Commun Biol 2021; 4:524. [PMID: 33953320 PMCID: PMC8100136 DOI: 10.1038/s42003-021-02059-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 03/31/2021] [Indexed: 01/24/2023] Open
Abstract
In Pompe disease, the deficiency of the lysosomal enzyme acid alpha-glucosidase (GAA) causes skeletal and cardiac muscle weakness, respiratory failure, and premature death. While enzyme replacement therapy using recombinant human GAA (rhGAA) can significantly improve patient outcomes, detailed disease mechanisms and incomplete therapeutic effects require further studies. Here we report a three-dimensional primary human skeletal muscle ("myobundle") model of infantile-onset Pompe disease (IOPD) that recapitulates hallmark pathological features including reduced GAA enzyme activity, elevated glycogen content and lysosome abundance, and increased sensitivity of muscle contractile function to metabolic stress. In vitro treatment of IOPD myobundles with rhGAA or adeno-associated virus (AAV)-mediated hGAA expression yields increased GAA activity and robust glycogen clearance, but no improvements in stress-induced functional deficits. We also apply RNA sequencing analysis to the quadriceps of untreated and AAV-treated GAA-/- mice and wild-type controls to establish a Pompe disease-specific transcriptional signature and reveal novel disease pathways. The mouse-derived signature is enriched in the transcriptomic profile of IOPD vs. healthy myobundles and partially reversed by in vitro rhGAA treatment, further confirming the utility of the human myobundle model for studies of Pompe disease and therapy.
Collapse
Affiliation(s)
- Jason Wang
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Chris J Zhou
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Sabrina Tran
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Sang-Oh Han
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Aaron L Carlson
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Lauran Madden
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Priya S Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Dwight D Koeberl
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
16
|
Li Y, Jacox LA, Coats S, Kwon J, Xue P, Tang N, Rui Z, Wang X, Kim YI, Wu TJ, Lee YT, Wong SW, Chien CH, Cheng CW, Gross R, Lin FC, Tseng H, Martinez J, Ko CC. Roles of autophagy in orthodontic tooth movement. Am J Orthod Dentofacial Orthop 2021; 159:582-593. [PMID: 33771430 PMCID: PMC10911631 DOI: 10.1016/j.ajodo.2020.01.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 01/01/2020] [Accepted: 01/01/2020] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Orthodontic tooth movement (OTM) relies on efficient remodeling of alveolar bone. While a well-controlled inflammatory response is essential during OTM, the mechanism regulating inflammation is unknown. Autophagy, a conserved catabolic pathway, has been shown to protect cells from excess inflammation in disease states. We hypothesize that autophagy plays a role in regulating inflammation during OTM. METHODS A split-mouth design was used to force load molars in adult male mice, carrying a GFP-LC3 transgene for in vivo detection of autophagy. Confocal microscopy, Western blot, and quantitative polymerase chain reaction analyses were used to evaluate autophagy activation in tissues of loaded and control molars at time points after force application. Rapamycin, a Food and Drug Administration-approved immunosuppressant, was injected to evaluate induction of autophagy. RESULTS Autophagy activity increases shortly after loading, primarily on the compression side of the tooth, and is closely associated with inflammatory cytokine expression and osteoclast recruitment. Daily administration of rapamycin, an autophagy activator, led to reduced tooth movement and osteoclast recruitment, suggesting that autophagy downregulates the inflammatory response and bone turnover during OTM. CONCLUSIONS This is the first demonstration that shows that autophagy is induced by orthodontic loading and plays a role during OTM, likely via negative regulation of inflammatory response and bone turnover. Exploring roles of autophagy in OTM holds great promise, as aberrant autophagy is associated with periodontal disease and its related systemic inflammatory disorders.
Collapse
Affiliation(s)
- Yina Li
- Department of Orthodontics, School of Dentistry, University of North Carolina, Chapel Hill, Nc
| | - Laura Anne Jacox
- Department of Orthodontics, School of Dentistry, University of North Carolina, Chapel Hill, Nc
| | - Shannon Coats
- Duke University Medical Center Greenspace, Durham, Nc
| | - Jane Kwon
- Oral and Craniofacial Health Sciences, School of Dentistry, University of North Carolina, Chapel Hill, Nc
| | - Peng Xue
- Department of Orthodontics, School of Dentistry, University of North Carolina, Chapel Hill, Nc
| | - Na Tang
- Oral and Craniofacial Health Sciences, School of Dentistry, University of North Carolina, Chapel Hill, Nc Department of Oral Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Zou Rui
- Oral and Craniofacial Health Sciences, School of Dentistry, University of North Carolina, Chapel Hill, Nc Department of Orthodontics, Stomatological Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xiaoyu Wang
- Oral and Craniofacial Health Sciences, School of Dentistry, University of North Carolina, Chapel Hill, Nc Department of Dentistry, Beijing Tiantan Hospital, Capital Medical University, Beijing, Beijing, China
| | - Yong-Il Kim
- Oral and Craniofacial Health Sciences, School of Dentistry, University of North Carolina, Chapel Hill, Nc Department of Orthodontics, School of Dentistry, Pusan National University, Yangsan, South Korea
| | - Te Ju Wu
- Oral and Craniofacial Health Sciences, School of Dentistry, University of North Carolina, Chapel Hill, Nc Department of Orthodontics, Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Yan-Ting Lee
- Oral and Craniofacial Health Sciences, School of Dentistry, University of North Carolina, Chapel Hill, Nc
| | - Sing Wai Wong
- Department of Periodontology, School of Dentistry, University of North Carolina, Chapel Hill, Nc
| | - Chia Hui Chien
- Oral and Craniofacial Health Sciences, School of Dentistry, University of North Carolina, Chapel Hill, Nc Division of Prosthodontics, Department of Dentistry, Tainan, Taiwan
| | - Chih-Wen Cheng
- Oral and Craniofacial Health Sciences, School of Dentistry, University of North Carolina, Chapel Hill, Nc Division of Prosthodontics, Department of Dentistry, Tainan, Taiwan
| | - Ryan Gross
- Department of Orthodontics, School of Dentistry, University of North Carolina, Chapel Hill, Nc
| | - Feng-Chang Lin
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, Nc
| | - Henry Tseng
- Duke University Medical Center Greenspace and Glaucoma Division, Duke Eye Center, Durham, Nc
| | - Jennifer Martinez
- National Institutes of Health, Bethesda, Ma National Institute of Environmental Health Sciences, Research Triangle Park, Durham, Nc
| | - Ching-Chang Ko
- Department of Orthodontics, School of Dentistry, University of North Carolina, Chapel Hill, Nc.
| |
Collapse
|
17
|
The Role of Exosomes in Lysosomal Storage Disorders. Biomolecules 2021; 11:biom11040576. [PMID: 33920837 PMCID: PMC8071119 DOI: 10.3390/biom11040576] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 12/15/2022] Open
Abstract
Exosomes, small membrane-bound organelles formed from endosomal membranes, represent a heterogenous source of biological and pathological biomarkers capturing the metabolic status of a cell. Exosomal cargo, including lipids, proteins, mRNAs, and miRNAs, can either act as inter-cellular messengers or are shuttled for autophagic/lysosomal degradation. Most cell types in the central nervous system (CNS) release exosomes, which serve as long and short distance communicators between neurons, astrocytes, oligodendrocytes, and microglia. Lysosomal storage disorders are diseases characterized by the accumulation of partially or undigested cellular waste. The exosomal content in these diseases is intrinsic to each individual disorder. Emerging research indicates that lysosomal dysfunction enhances exocytosis, and hence, in lysosomal disorders, exosomal secretion may play a role in disease pathogenesis. Furthermore, the unique properties of exosomes and their ability to carry cargo between adjacent cells and organs, and across the blood-brain barrier, make them attractive candidates for use as therapeutic delivery vehicles. Thus, understanding exosomal content and function may have utility in the treatment of specific lysosomal storage disorders. Since lysosomal dysfunction and the deficiency of at least one lysosomal enzyme, glucocerebrosidase, is associated with the development of parkinsonism, the study and use of exosomes may contribute to an improved understanding of Parkinson disease, potentially leading to new therapeutics.
Collapse
|
18
|
Khan N, Chen X, Geiger JD. Role of Endolysosomes in Severe Acute Respiratory Syndrome Coronavirus-2 Infection and Coronavirus Disease 2019 Pathogenesis: Implications for Potential Treatments. Front Pharmacol 2020; 11:595888. [PMID: 33324224 PMCID: PMC7723437 DOI: 10.3389/fphar.2020.595888] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 10/05/2020] [Indexed: 12/14/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is an enveloped, single-stranded RNA virus. Humans infected with SARS-CoV-2 develop a disease known as coronavirus disease 2019 (COVID-19) with symptoms and consequences including acute respiratory distress syndrome (ARDS), cardiovascular disorders, and death. SARS-CoV-2 appears to infect cells by first binding viral spike proteins with host protein angiotensin-converting enzyme 2 (ACE2) receptors; the virus is endocytosed following priming by transmembrane protease serine 2 (TMPRSS2). The process of virus entry into endosomes and its release from endolysosomes are key features of enveloped viruses. Thus, it is important to focus attention on the role of endolysosomes in SARS-CoV-2 infection. Indeed, coronaviruses are now known to hijack endocytic machinery to enter cells such that they can deliver their genome at replication sites without initiating host detection and immunological responses. Hence, endolysosomes might be good targets for developing therapeutic strategies against coronaviruses. Here, we focus attention on the involvement of endolysosomes in SARS-CoV-2 infection and COVID-19 pathogenesis. Further, we explore endolysosome-based therapeutic strategies to restrict SARS-CoV-2 infection and COVID-19 pathogenesis.
Collapse
Affiliation(s)
| | | | - Jonathan D. Geiger
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
| |
Collapse
|
19
|
Kawahata I, Fukunaga K. Degradation of Tyrosine Hydroxylase by the Ubiquitin-Proteasome System in the Pathogenesis of Parkinson's Disease and Dopa-Responsive Dystonia. Int J Mol Sci 2020; 21:ijms21113779. [PMID: 32471089 PMCID: PMC7312529 DOI: 10.3390/ijms21113779] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/19/2020] [Accepted: 05/25/2020] [Indexed: 12/16/2022] Open
Abstract
Nigrostriatal dopaminergic systems govern physiological functions related to locomotion, and their dysfunction leads to movement disorders, such as Parkinson’s disease and dopa-responsive dystonia (Segawa disease). Previous studies revealed that expression of the gene encoding nigrostriatal tyrosine hydroxylase (TH), a rate-limiting enzyme of dopamine biosynthesis, is reduced in Parkinson’s disease and dopa-responsive dystonia; however, the mechanism of TH depletion in these disorders remains unclear. In this article, we review the molecular mechanism underlying the neurodegeneration process in dopamine-containing neurons and focus on the novel degradation pathway of TH through the ubiquitin-proteasome system to advance our understanding of the etiology of Parkinson’s disease and dopa-responsive dystonia. We also introduce the relation of α-synuclein propagation with the loss of TH protein in Parkinson’s disease as well as anticipate therapeutic targets and early diagnosis of these diseases.
Collapse
Affiliation(s)
- Ichiro Kawahata
- Correspondence: (I.K.); (K.F.); Tel.: +81-22-795-6838 (I.K.); +81-22-795-6836 (K.F.); Fax: +81-22-795-6835 (I.K. & K.F.)
| | - Kohji Fukunaga
- Correspondence: (I.K.); (K.F.); Tel.: +81-22-795-6838 (I.K.); +81-22-795-6836 (K.F.); Fax: +81-22-795-6835 (I.K. & K.F.)
| |
Collapse
|
20
|
Martínez-García GG, Mariño G. Autophagy role in environmental pollutants exposure. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 172:257-291. [PMID: 32620245 DOI: 10.1016/bs.pmbts.2020.02.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
During the last decades, the potential harmfulness derived from the exposure to environmental pollutants has been largely demonstrated, with associated damages ranging from geno- and cyto-toxicity to tissue malfunction and alterations in organism physiology. Autophagy is an evolutionarily-conserved cellular mechanism essential for cellular homeostasis, which contributes to protect cells from a wide variety of intracellular and extracellular stressors. Due to its pivotal importance, its correct functioning is directly linked to cell, tissue and organismal fitness. Environmental pollutants, particularly industrial compounds, are able to impact autophagic flux, either by increasing it as a protective response, by blocking it, or by switching its protective role toward a pro-cell death mechanism. Thus, the understanding of the effects of chemicals exposure on autophagy has become highly relevant, offering new potential approaches for risk assessment, protection and preventive measures to counteract the detrimental effects of environmental pollutants on human health.
Collapse
Affiliation(s)
- Gemma G Martínez-García
- Laboratorio "Autofagia y Metabolismo", Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain; Departamento de Biología Funcional, Universidad de Oviedo, Oviedo, Spain
| | - Guillermo Mariño
- Laboratorio "Autofagia y Metabolismo", Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain; Departamento de Biología Funcional, Universidad de Oviedo, Oviedo, Spain.
| |
Collapse
|
21
|
Shi W, Liu W, Ma J, Lu J, Yang X, Wang J, Cao J, Tian Y, Yang H, Zhang L. The role of Atg7-mediated autophagy in ionizing radiation-induced neural stem cell damage. Gene 2020; 738:144485. [PMID: 32087272 DOI: 10.1016/j.gene.2020.144485] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 02/15/2020] [Accepted: 02/18/2020] [Indexed: 02/06/2023]
Abstract
Impairment of neurogenesis is thought to be one of the important mechanisms underlying radiation-induced cognitive decline. Self-renewal and differentiation of neural stem cells (NSCs) are important components of neurogenesis. It has been well established that autophagy plays an important role in neurodegenerative conditions, however, its role in radiation-induced cognitive decline remains unclear. Our previous studies have found that ionizing radiation (IR) induces autophagy in mouse neurons, and minocycline, an antibiotic that can cross the blood-brain barrier, protects neurons from radiation-induced apoptosis through promoting autophagy, thus may contribute to the improvement of mouse cognitive performance after whole-brain irradiation. In the present study, we investigated whether autophagy is involved in radiation-induced damage in self-renewal and differentiation of NSCs. We found that NSCs were extremely sensitive to IR. Irradiation induced autophagy in NSCs in a dose-dependent manner. Atg7 knockdown significantly decreased autophagy, thus increased the apoptosis levels in irradiated NSCs, suggesting that autophagy protected NSCs from radiation-induced apoptosis. Moreover, compared with the negative control NSCs, the neurosphere size was significantly reduced and the neuronal differentiation was notably inhibited in Atg7-deficient NSCs after irradiation, indicating that autophagy defect could exacerbate radiation-induced reduction in NSC self-renewal and differentiation potential. In conclusion, down-regulating autophagy by selective Atg7 knockdown in NSCs enhanced radiation-induced NSC damage, suggesting an important protective role of autophagy in maintaining neurogenesis. Along with the protective effect of autophagy on irradiated neurons, our results on NSCs not only shed the light on the involvement of autophagy in the development of radiation-induced cognitive decline, but also provided a potential target for preventing cognitive impairment after cranial radiation exposure.
Collapse
Affiliation(s)
- Wenyu Shi
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu Province 215004, PR China; Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu Province 215004, PR China
| | - Wei Liu
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu Province 215004, PR China; Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu Province 215004, PR China
| | - Jiayan Ma
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu Province 215004, PR China; Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu Province 215004, PR China
| | - Jiawei Lu
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu Province 215004, PR China; Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu Province 215004, PR China
| | - Xuejiao Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University/Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, 199 Renai Road, Suzhou Industrial Park, Suzhou, Jiangsu Province 215123, PR China
| | - Jingdong Wang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University/Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, 199 Renai Road, Suzhou Industrial Park, Suzhou, Jiangsu Province 215123, PR China
| | - Jianping Cao
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University/Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, 199 Renai Road, Suzhou Industrial Park, Suzhou, Jiangsu Province 215123, PR China
| | - Ye Tian
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu Province 215004, PR China; Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu Province 215004, PR China; Institute of Radiotherapy & Oncology of Soochow University, PR China
| | - Hongying Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University/Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, 199 Renai Road, Suzhou Industrial Park, Suzhou, Jiangsu Province 215123, PR China; Institute of Radiotherapy & Oncology of Soochow University, PR China.
| | - Liyuan Zhang
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu Province 215004, PR China; Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu Province 215004, PR China; Institute of Radiotherapy & Oncology of Soochow University, PR China.
| |
Collapse
|
22
|
Chen YB. Autophagy and its role in pulmonary hypertension. Aging Clin Exp Res 2019; 31:1027-1033. [PMID: 30406918 DOI: 10.1007/s40520-018-1063-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 10/19/2018] [Indexed: 01/16/2023]
Abstract
Pulmonary hypertension (PH) is a very common kind of pulmonary vascular disease, which can cause a heavier burden on patient's quality of life, even lead to death. Yet, the mechanism of PH is incomprehensive and not so clear nowadays. In recent years, more and more studies show that autophagy plays a pivotal role in the development of PH. Some modalities target on the formation or maturation of autophagosome that has emerged from our increasing knowledge of autophagy machinery, which may prevent or eliminate the process of PH. The deciphering of molecular selectivity of autophagy has also been a source of novel modulators that act specifically on selective forms of autophagy. Tremendous recent progress has opened a new possibility for modulating autophagy in complex diseases. Thus, autophagy may become a prospective choice for treatment of PH. Herein, we reviewed the literatures and discussed the role of autophagy in the development and treatment of PH.
Collapse
|
23
|
Tian L, Cheng F, Wang L, Qin W, Zou K, Chen J. CLE-10 from Carpesium abrotanoides L. Suppresses the Growth of Human Breast Cancer Cells (MDA-MB-231) In Vitro by Inducing Apoptosis and Pro-Death Autophagy Via the PI3K/Akt/mTOR Signaling Pathway. Molecules 2019; 24:molecules24061091. [PMID: 30897708 PMCID: PMC6471079 DOI: 10.3390/molecules24061091] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 03/15/2019] [Accepted: 03/16/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The antitumor activity of CLE-10 (4-epi-isoinuviscolide), a sesquiterpene lactone compound, isolated from Carpesium abrotanoides L. has rarely been reported. The aim of this study is to investigate the antitumor activity of CLE-10 and give a greater explanation of its underlying mechanisms. METHODS The cytotoxicity of CLE-10 was evaluated using MTT assay. Autophagy was detected by the formation of mRFP-GFP-LC3 fluorescence puncta and observed using transmission electron microscopy, while flow cytometry was employed to detect apoptosis. The protein expressions were detected through Western blotting. RESULTS CLE-10 induced pro-death autophagy and apoptosis in MDA-MB-231 cells by increasing the protein expression of LC3-II, p-ULK1, Bax, and Bad, as well as downregulating p-PI3K, p-Akt, p-mTOR, p62, LC3-I, Bcl-2, and Bcl-xl. CLE-10 that was pretreated with 3-methyladenine (3-MA) or chloroquine (CQ) weakened the upregulation of the protein expression of p-ULK1, or the downregulation of p62, p-mTOR, and decreased the level of cytotoxicity against MDA-MB-231 cells. Meanwhile, rapamycin enhanced the effect of CLE-10 on the expression of autophagy-related protein and its cytotoxicity, with the IC50 value of CLE-10 decreasing from 4.07 µM to 2.38 µM. CONCLUSION CLE-10 induced pro-death autophagy and apoptosis in MDA-MB-231 cells by upregulating the protein expressions of LC3-II, p-ULK1, Bax, and Bad and downregulating p-PI3K, p-Akt, p-mTOR, p62, Bcl-2, and Bcl-xl.
Collapse
Affiliation(s)
- Li Tian
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China.
| | - Fan Cheng
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China.
| | - Lei Wang
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China.
| | - Wen Qin
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China.
| | - Kun Zou
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China.
| | - Jianfeng Chen
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China.
| |
Collapse
|
24
|
Song L, Rijal R, Karow M, Stumpf M, Hahn O, Park L, Insall R, Schröder R, Hofmann A, Clemen CS, Eichinger L. Expression of N471D strumpellin leads to defects in the endolysosomal system. Dis Model Mech 2018; 11:dmm033449. [PMID: 30061306 PMCID: PMC6177004 DOI: 10.1242/dmm.033449] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 07/09/2018] [Indexed: 12/11/2022] Open
Abstract
Hereditary spastic paraplegias (HSPs) are genetically diverse and clinically characterised by lower limb weakness and spasticity. The N471D and several other point mutations of human strumpellin (Str; also known as WASHC5), a member of the Wiskott-Aldrich syndrome protein and SCAR homologue (WASH) complex, have been shown to cause a form of HSP known as spastic paraplegia 8 (SPG8). To investigate the molecular functions of wild-type (WT) and N417D Str, we generated Dictyostelium Str- cells and ectopically expressed StrWT-GFP or StrN471D-GFP in Str- and WT cells. Overexpression of both proteins apparently caused a defect in cell division, as we observed a clear increase in multinucleate cells. Real-time PCR analyses revealed no transcriptional changes in WASH complex subunits in Str- cells, but western blots showed a twofold decrease in the SWIP subunit. GFP-trap experiments in conjunction with mass-spectrometric analysis revealed many previously known, as well as new, Str-interacting proteins, and also proteins that no longer bind to StrN471D At the cellular level, Str- cells displayed defects in cell growth, phagocytosis, macropinocytosis, exocytosis and lysosomal function. Expression of StrWT-GFP in Str- cells rescued all observed defects. In contrast, expression of StrN471D-GFP could not rescue lysosome morphology and exocytosis of indigestible material. Our results underscore a key role for the WASH complex and its core subunit, Str, in the endolysosomal system, and highlight the fundamental importance of the Str N471 residue for maintaining lysosome morphology and dynamics. Our data indicate that the SPG8-causing N471D mutation leads to a partial loss of Str function in the endolysosomal system. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Lin Song
- Center for Biochemistry, Institute of Biochemistry I, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Ramesh Rijal
- Center for Biochemistry, Institute of Biochemistry I, Medical Faculty, University of Cologne, 50931 Cologne, Germany
- Department of Biology, Texas A&M University, College Station, TX 3258, USA
| | - Malte Karow
- Center for Biochemistry, Institute of Biochemistry I, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Maria Stumpf
- Center for Biochemistry, Institute of Biochemistry I, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Oliver Hahn
- Max Planck Institute for Biology of Ageing, Biological Mechanisms of Ageing, 50931 Cologne, Germany
| | - Laura Park
- CR-UK Beatson Institute, Institute of Cancer Sciences, Glasgow University, Glasgow G12 8QQ, UK
| | - Robert Insall
- CR-UK Beatson Institute, Institute of Cancer Sciences, Glasgow University, Glasgow G12 8QQ, UK
| | - Rolf Schröder
- Institute of Neuropathology, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Andreas Hofmann
- Structural Chemistry Program, Eskitis Institute, Griffith University, N75 Don Young Road, Nathan, QLD 4111, Australia
- Faculty of Veterinary Science, The University of Melbourne, Parkville, VIC 3030, Australia
| | - Christoph S Clemen
- Center for Biochemistry, Institute of Biochemistry I, Medical Faculty, University of Cologne, 50931 Cologne, Germany
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789 Bochum, Germany
| | - Ludwig Eichinger
- Center for Biochemistry, Institute of Biochemistry I, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
25
|
Lei L, Tzekov R, Li H, McDowell JH, Gao G, Smith WC, Tang S, Kaushal S. Inhibition or Stimulation of Autophagy Affects Early Formation of Lipofuscin-Like Autofluorescence in the Retinal Pigment Epithelium Cell. Int J Mol Sci 2017; 18:ijms18040728. [PMID: 28353645 PMCID: PMC5412314 DOI: 10.3390/ijms18040728] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 03/23/2017] [Accepted: 03/24/2017] [Indexed: 12/22/2022] Open
Abstract
The accumulation of lipofuscin in the retinal pigment epithelium (RPE) is dependent on the effectiveness of photoreceptor outer segment material degradation. This study explored the role of autophagy in the fate of RPE lipofuscin degradation. After seven days of feeding with either native or modified rod outer segments, ARPE-19 cells were treated with enhancers or inhibitors of autophagy and the autofluorescence was detected by fluorescence-activated cell sorting. Supplementation with different types of rod outer segments increased lipofuscin-like autofluorescence (LLAF) after the inhibition of autophagy, while the induction of autophagy (e.g., application of rapamycin) decreased LLAF. The effects of autophagy induction were further confirmed by Western blotting, which showed the conversion of LC3-I to LC3-II, and by immunofluorescence microscopy, which detected the lysosomal activity of the autophagy inducers. We also monitored LLAF after the application of several autophagy inhibitors by RNA-interference and confocal microscopy. The results showed that, in general, the inhibition of the autophagy-related proteins resulted in an increase in LLAF when cells were fed with rod outer segments, which further confirms the effect of autophagy in the fate of RPE lipofuscin degradation. These results emphasize the complex role of autophagy in modulating RPE autofluorescence and confirm the possibility of the pharmacological clearance of RPE lipofuscin by small molecules.
Collapse
Affiliation(s)
- Lei Lei
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, No.54 South Xianlie Road, Guangzhou 510060, China.
- Department of Ophthalmology, University of Massachusetts Medical School, 381 Plantation Street, Worcester, MA 01605, USA.
| | - Radouil Tzekov
- Department of Ophthalmology, University of Massachusetts Medical School, 381 Plantation Street, Worcester, MA 01605, USA.
- Department of Ophthalmology, University of South Florida, 13127 USF Magnolia Drive, Tampa, FL 33612, USA.
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL 34243, USA.
| | - Huapeng Li
- Gene Therapy Center, University of Massachusetts Medical School, 381 Plantation Street, Worcester, MA 01605, USA.
| | - J Hugh McDowell
- The Department of Ophthalmology, University of Florida Health Science Center, 1600 SW Archer Road, Gainesville, FL 32610, USA.
| | - Guangping Gao
- Gene Therapy Center, University of Massachusetts Medical School, 381 Plantation Street, Worcester, MA 01605, USA.
| | - W Clay Smith
- The Department of Ophthalmology, University of Florida Health Science Center, 1600 SW Archer Road, Gainesville, FL 32610, USA.
| | - Shibo Tang
- Aier School of Ophthalmology, Central South University, Floor 4, New Century Building, 198# Furong Middle Road, Changsha 410015, China.
| | - Shalesh Kaushal
- Department of Ophthalmology, University of Massachusetts Medical School, 381 Plantation Street, Worcester, MA 01605, USA.
- VRMI, 6205 NW 81st Drive, Gainesville, FL 32653, USA.
| |
Collapse
|
26
|
Chronic Iron Overload Results in Impaired Bacterial Killing of THP-1 Derived Macrophage through the Inhibition of Lysosomal Acidification. PLoS One 2016; 11:e0156713. [PMID: 27244448 PMCID: PMC4886970 DOI: 10.1371/journal.pone.0156713] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 05/18/2016] [Indexed: 01/19/2023] Open
Abstract
Iron is essential for living organisms and the disturbance of iron homeostasis is associated with altered immune function. Additionally, bacterial infections can cause major complications in instances of chronic iron overload, such as patients with transfusion-dependent thalassemia. Monocytes and macrophages play important roles in maintaining systemic iron homoeostasis and in defense against invading pathogens. However, the effect of iron overload on the function of monocytes and macrophages is unclear. We elucidated the effects of chronic iron overload on human monocytic cell line (THP-1) and THP-1 derived macrophages (TDM) by continuously exposing them to high levels of iron (100 μM) to create I-THP-1 and I-TDM, respectively. Our results show that iron overload did not affect morphology or granularity of I-THP-1, but increased the granularity of I-TDM. Bactericidal assays for non-pathogenic E. coli DH5α, JM109 and pathogenic P. aeruginosa all revealed decreased efficiency with increasing iron concentration in I-TDM. The impaired P. aeruginosa killing ability of human primary monocyte derived macrophages (hMDM) was also found when cells are cultured in iron contained medium. Further studies on the bactericidal activity of I-TDM revealed lysosomal dysfunction associated with the inhibition of lysosomal acidification resulting in increasing lysosomal pH, the impairment of post-translational processing of cathepsins (especially cathepsin D), and decreased autophagic flux. These findings may explain the impaired innate immunity of thalassemic patients with chronic iron overload, suggesting the manipulation of lysosomal function as a novel therapeutic approach.
Collapse
|