1
|
Chou CC, Vest R, Prado MA, Wilson-Grady J, Paulo JA, Shibuya Y, Moran-Losada P, Lee TT, Luo J, Gygi SP, Kelly JW, Finley D, Wernig M, Wyss-Coray T, Frydman J. Proteostasis and lysosomal repair deficits in transdifferentiated neurons of Alzheimer's disease. Nat Cell Biol 2025; 27:619-632. [PMID: 40140603 PMCID: PMC11991917 DOI: 10.1038/s41556-025-01623-y] [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: 05/21/2024] [Accepted: 01/21/2025] [Indexed: 03/28/2025]
Abstract
Ageing is the most prominent risk factor for Alzheimer's disease (AD). However, the cellular mechanisms linking neuronal proteostasis decline to the characteristic aberrant protein deposits in the brains of patients with AD remain elusive. Here we develop transdifferentiated neurons (tNeurons) from human dermal fibroblasts as a neuronal model that retains ageing hallmarks and exhibits AD-linked vulnerabilities. Remarkably, AD tNeurons accumulate proteotoxic deposits, including phospho-tau and amyloid β, resembling those in APP mouse brains and the brains of patients with AD. Quantitative tNeuron proteomics identify ageing- and AD-linked deficits in proteostasis and organelle homeostasis, most notably in endosome-lysosomal components. Lysosomal deficits in aged tNeurons, including constitutive lysosomal damage and ESCRT-mediated lysosomal repair defects, are exacerbated in AD tNeurons and linked to inflammatory cytokine secretion and cell death. Providing support for the centrality of lysosomal deficits in AD, compounds ameliorating lysosomal function reduce amyloid β deposits and cytokine secretion. Thus, the tNeuron model system reveals impaired lysosomal homeostasis as an early event of ageing and AD.
Collapse
Affiliation(s)
- Ching-Chieh Chou
- Department of Biology, Stanford University, Stanford, CA, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| | - Ryan Vest
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
- Department of Neurology and Neurological Sciences and The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA
- Qinotto Inc., San Carlos, CA, USA
| | - Miguel A Prado
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | | | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Yohei Shibuya
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Patricia Moran-Losada
- Department of Neurology and Neurological Sciences and The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Ting-Ting Lee
- Department of Biology, Stanford University, Stanford, CA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Jian Luo
- Palo Alto Veterans Institute for Research Inc. (PAVIR), Palo Alto, CA, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Jeffery W Kelly
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Daniel Finley
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Marius Wernig
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences and The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Judith Frydman
- Department of Biology, Stanford University, Stanford, CA, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Genetics, Stanford University, Stanford, CA, USA.
| |
Collapse
|
2
|
Deshpande RP, Wu K, Wu SY, Tyagi A, Smith EC, Kim JW, Watabe K. MHC-I upregulation by macbecin II in the solid tumors potentiates the effect of active immunotherapy. EMBO Mol Med 2025; 17:797-822. [PMID: 40087501 PMCID: PMC11982318 DOI: 10.1038/s44321-025-00213-7] [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: 08/06/2024] [Revised: 02/21/2025] [Accepted: 02/28/2025] [Indexed: 03/17/2025] Open
Abstract
We aimed to restore MHC-I expression on the surface of solid tumors including breast cancer and melanoma cells to regain sensitivity to immunotherapy and suppress metastatic progression. We screened a natural compound library and identified macbecin II as a reagent that upregulates MHC-I expression and induces antigen-dependent cell death in pre-invasive and invasive breast cancer models. Furthermore, we employed active immunotherapy using engineered small extracellular vesicles from dendritic cells (DCs) as a tumor vaccine (IL2-ep13nsEV) in combination with macbecin II for personalized breast cancer treatment. We found that macbecin II induced MHC-I-dependent antigen presentation and that IL2-ep13nsEV synergized with macbecin II inducing cell death, reducing metastasis, and boosting immune cell infiltration. In addition, macbecin II potentiated the effects of anti-PD-1 immunotherapy in suppressing tumor growth and metastasis. Mechanistically, macbecin II upregulated MHC-I expression post-translationally by rescuing it from lysosomal degradation. Our findings revealed a strong efficacy of macbecin II in regulating MHC-I expression and following antigen-dependent cell death. Therefore, combining active immunotherapies and macbecin II represents an effective strategy to prevent growth and progression of solid tumors including breast cancer and melanoma.
Collapse
Affiliation(s)
- Ravindra Pramod Deshpande
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Kerui Wu
- University of North Carolina, Greensboro, NC, 27412, USA
| | - Shih-Ying Wu
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Abhishek Tyagi
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Eleanor C Smith
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Jee-Won Kim
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Kounosuke Watabe
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
3
|
Mark JR, Tansey MG. Immune cell metabolic dysfunction in Parkinson's disease. Mol Neurodegener 2025; 20:36. [PMID: 40128809 PMCID: PMC11934562 DOI: 10.1186/s13024-025-00827-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 03/07/2025] [Indexed: 03/26/2025] Open
Abstract
Parkinson's disease (PD) is a multi-system disorder characterized histopathologically by degeneration of dopaminergic neurons in the substantia nigra pars compacta. While the etiology of PD remains multifactorial and complex, growing evidence suggests that cellular metabolic dysfunction is a critical driver of neuronal death. Defects in cellular metabolism related to energy production, oxidative stress, metabolic organelle health, and protein homeostasis have been reported in both neurons and immune cells in PD. We propose that these factors act synergistically in immune cells to drive aberrant inflammation in both the CNS and the periphery in PD, contributing to a hostile inflammatory environment which renders certain subsets of neurons vulnerable to degeneration. This review highlights the overlap between established neuronal metabolic deficits in PD with emerging findings in central and peripheral immune cells. By discussing the rapidly expanding literature on immunometabolic dysfunction in PD, we aim to draw attention to potential biomarkers and facilitate future development of immunomodulatory strategies to prevent or delay the progression of PD.
Collapse
Affiliation(s)
- Julian R Mark
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Malú Gámez Tansey
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA.
- Department of Neurology and Norman Fixel Institute for Neurological Diseases, University of Florida Health, Gainesville, FL, 32608, USA.
| |
Collapse
|
4
|
Fu Y, Zhang J, Qin R, Ren Y, Zhou T, Han B, Liu B. Activating autophagy to eliminate toxic protein aggregates with small molecules in neurodegenerative diseases. Pharmacol Rev 2025; 77:100053. [PMID: 40187044 DOI: 10.1016/j.pharmr.2025.100053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 12/05/2024] [Indexed: 04/07/2025] Open
Abstract
Neurodegenerative diseases (NDs), such as Alzheimer disease, Parkinson disease, Huntington disease, amyotrophic lateral sclerosis, and frontotemporal dementia, are well known to pose formidable challenges for their treatment due to their intricate pathogenesis and substantial variability among patients, including differences in environmental exposures and genetic predispositions. One of the defining characteristics of NDs is widely reported to be the buildup of misfolded proteins. For example, Alzheimer disease is marked by amyloid beta and hyperphosphorylated Tau aggregates, whereas Parkinson disease exhibits α-synuclein aggregates. Amyotrophic lateral sclerosis and frontotemporal dementia exhibit TAR DNA-binding protein 43, superoxide dismutase 1, and fused-in sarcoma protein aggregates, and Huntington disease involves mutant huntingtin and polyglutamine aggregates. These misfolded proteins are the key biomarkers of NDs and also serve as potential therapeutic targets, as they can be addressed through autophagy, a process that removes excess cellular inclusions to maintain homeostasis. Various forms of autophagy, including macroautophagy, chaperone-mediated autophagy, and microautophagy, hold a promise in eliminating toxic proteins implicated in NDs. In this review, we focus on elucidating the regulatory connections between autophagy and toxic proteins in NDs, summarizing the cause of the aggregates, exploring their impact on autophagy mechanisms, and discussing how autophagy can regulate toxic protein aggregation. Moreover, we underscore the activation of autophagy as a potential therapeutic strategy across different NDs and small molecules capable of activating autophagy pathways, such as rapamycin targeting the mTOR pathway to clear α-synuclein and Sertraline targeting the AMPK/mTOR/RPS6KB1 pathway to clear Tau, to further illustrate their potential in NDs' therapeutic intervention. Together, these findings would provide new insights into current research trends and propose small-molecule drugs targeting autophagy as promising potential strategies for the future ND therapies. SIGNIFICANCE STATEMENT: This review provides an in-depth overview of the potential of activating autophagy to eliminate toxic protein aggregates in the treatment of neurodegenerative diseases. It also elucidates the fascinating interrelationships between toxic proteins and the process of autophagy of "chasing and escaping" phenomenon. Moreover, the review further discusses the progress utilizing small molecules to activate autophagy to improve the efficacy of therapies for neurodegenerative diseases by removing toxic protein aggregates.
Collapse
Affiliation(s)
- Yuqi Fu
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, China; Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jin Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China; School of Pharmaceutical Sciences of Medical School, Shenzhen University, Shenzhen, China
| | - Rui Qin
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yueting Ren
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China; Department of Brain Science, Faculty of Medicine, Imperial College, London, UK
| | - Tingting Zhou
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China; Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Second Military Medical University, Shanghai, China.
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Bo Liu
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, China; Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
5
|
Zeng J, Indajang J, Pitt D, Lo CH. Lysosomal acidification impairment in astrocyte-mediated neuroinflammation. J Neuroinflammation 2025; 22:72. [PMID: 40065324 PMCID: PMC11892208 DOI: 10.1186/s12974-025-03410-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 03/05/2025] [Indexed: 03/14/2025] Open
Abstract
Astrocytes are a major cell type in the central nervous system (CNS) that play a key role in regulating homeostatic functions, responding to injuries, and maintaining the blood-brain barrier. Astrocytes also regulate neuronal functions and survival by modulating myelination and degradation of pathological toxic protein aggregates. Astrocytes have recently been proposed to possess both autophagic activity and active phagocytic capability which largely depend on sufficiently acidified lysosomes for complete degradation of cellular cargos. Defective lysosomal acidification in astrocytes impairs their autophagic and phagocytic functions, resulting in the accumulation of cellular debris, excessive myelin and lipids, and toxic protein aggregates, which ultimately contributes to the propagation of neuroinflammation and neurodegenerative pathology. Restoration of lysosomal acidification in impaired astrocytes represent new neuroprotective strategy and therapeutic direction. In this review, we summarize pathogenic factors, including neuroinflammatory signaling, metabolic stressors, myelin and lipid mediated toxicity, and toxic protein aggregates, that contribute to lysosomal acidification impairment and associated autophagic and phagocytic dysfunction in astrocytes. We discuss the role of lysosomal acidification dysfunction in astrocyte-mediated neuroinflammation primarily in the context of neurodegenerative diseases along with other brain injuries. We then highlight re-acidification of impaired lysosomes as a therapeutic strategy to restore autophagic and phagocytic functions as well as lysosomal degradative capacity in astrocytes. We conclude by providing future perspectives on the role of astrocytes as phagocytes and their crosstalk with other CNS cells to impart neurodegenerative or neuroprotective effects.
Collapse
Affiliation(s)
- Jialiu Zeng
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA.
- Interdisciplinary Neuroscience Program, Syracuse University, Syracuse, NY, 13244, USA.
| | - Jonathan Indajang
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - David Pitt
- Department of Neurology, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Chih Hung Lo
- Interdisciplinary Neuroscience Program, Syracuse University, Syracuse, NY, 13244, USA.
- Department of Biology, Syracuse University, Syracuse, NY, 13244, USA.
| |
Collapse
|
6
|
Sanyal A, Scanavachi G, Somerville E, Saminathan A, Nair A, Bango Da Cunha Correia RF, Aylan B, Sitarska E, Oikonomou A, Hatzakis NS, Kirchhausen T. Neuronal constitutive endolysosomal perforations enable α-synuclein aggregation by internalized PFFs. J Cell Biol 2025; 224:e202401136. [PMID: 39714357 DOI: 10.1083/jcb.202401136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/12/2024] [Accepted: 11/14/2024] [Indexed: 12/24/2024] Open
Abstract
Endocytosis, required for the uptake of receptors and their ligands, can also introduce pathological aggregates such as α-synuclein (α-syn) in Parkinson's Disease. We show here the unexpected presence of intrinsically perforated endolysosomes in neurons, suggesting involvement in the genesis of toxic α-syn aggregates induced by internalized preformed fibrils (PFFs). Aggregation of endogenous α-syn in late endosomes and lysosomes of human iPSC-derived neurons (iNs), seeded by internalized α-syn PFFs, caused the death of the iNs but not of the parental iPSCs and non-neuronal cells. Live-cell imaging of iNs showed constitutive perforations in ∼5% of their endolysosomes. These perforations, identified by 3D electron microscopy in iNs and CA1 pyramidal neurons and absent in non-neuronal cells, may facilitate cytosolic access of endogenous α-syn to PFFs in the lumen of endolysosomes, triggering aggregation. Inhibiting the PIKfyve phosphoinositol kinase reduced α-syn aggregation and associated iN death, even with ongoing PFF endolysosomal entry, suggesting that maintaining endolysosomal integrity might afford a therapeutic strategy to counteract synucleinopathies.
Collapse
Affiliation(s)
- Anwesha Sanyal
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital , Boston, MA, USA
| | - Gustavo Scanavachi
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital , Boston, MA, USA
| | - Elliott Somerville
- Program in Cellular and Molecular Medicine, Boston Children's Hospital , Boston, MA, USA
| | - Anand Saminathan
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital , Boston, MA, USA
| | - Athul Nair
- Program in Cellular and Molecular Medicine, Boston Children's Hospital , Boston, MA, USA
| | - Ricardo F Bango Da Cunha Correia
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital , Boston, MA, USA
| | - Beren Aylan
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital , Boston, MA, USA
| | - Ewa Sitarska
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital , Boston, MA, USA
| | | | - Nikos S Hatzakis
- Department of Chemistry, University of Copenhagen, Copenhagen, Denmark
| | - Tom Kirchhausen
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital , Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
7
|
Hao M, Chu J, Zhang T, Yin T, Gu Y, Liang W, Ji W, Zhuang J, Liu Y, Gao J, Yin Y. Nanomaterials-mediated lysosomal regulation: a robust protein-clearance approach for the treatment of Alzheimer's disease. Neural Regen Res 2025; 20:424-439. [PMID: 38819046 PMCID: PMC11317947 DOI: 10.4103/nrr.nrr-d-23-01736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 01/04/2024] [Accepted: 02/20/2024] [Indexed: 06/01/2024] Open
Abstract
Alzheimer's disease is a debilitating, progressive neurodegenerative disorder characterized by the progressive accumulation of abnormal proteins, including amyloid plaques and intracellular tau tangles, primarily within the brain. Lysosomes, crucial intracellular organelles responsible for protein degradation, play a key role in maintaining cellular homeostasis. Some studies have suggested a link between the dysregulation of the lysosomal system and pathogenesis of neurodegenerative diseases, including Alzheimer's disease. Restoring the normal physiological function of lysosomes hold the potential to reduce the pathological burden and improve the symptoms of Alzheimer's disease. Currently, the efficacy of drugs in treating Alzheimer's disease is limited, with major challenges in drug delivery efficiency and targeting. Recently, nanomaterials have gained widespread use in Alzheimer's disease drug research owing to their favorable physical and chemical properties. This review aims to provide a comprehensive overview of recent advances in using nanomaterials (polymeric nanomaterials, nanoemulsions, and carbon-based nanomaterials) to enhance lysosomal function in treating Alzheimer's disease. This review also explores new concepts and potential therapeutic strategies for Alzheimer's disease through the integration of nanomaterials and modulation of lysosomal function. In conclusion, this review emphasizes the potential of nanomaterials in modulating lysosomal function to improve the pathological features of Alzheimer's disease. The application of nanotechnology to the development of Alzheimer's disease drugs brings new ideas and approaches for future treatment of this disease.
Collapse
Affiliation(s)
- Mengqi Hao
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Jianjian Chu
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
| | - Tinglin Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Tong Yin
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
| | - Yuankai Gu
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
| | - Wendanqi Liang
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Wenbo Ji
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
| | - Jianhua Zhuang
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
| | - Yan Liu
- Department of Clinical Pharmacy, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - You Yin
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
8
|
Austin MC, Muralidharan C, Roy S, Crowder JJ, Piganelli JD, Linnemann AK. Dysfunctional β-cell autophagy induces β-cell stress and enhances islet immunogenicity. Front Immunol 2025; 16:1504583. [PMID: 39944686 PMCID: PMC11814175 DOI: 10.3389/fimmu.2025.1504583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 01/09/2025] [Indexed: 02/16/2025] Open
Abstract
Background Type 1 Diabetes (T1D) is caused by a combination of genetic and environmental factors that trigger autoimmune-mediated destruction of pancreatic β-cells. Defects in β-cell stress response pathways such as autophagy may play an important role in activating and/or exacerbating the immune response in disease development. Previously, we discovered that β-cell autophagy is impaired prior to the onset of T1D, implicating this pathway in T1D pathogenesis. Aims To assess the role of autophagy in β-cell health and survival, and whether defects in autophagy render islets more immunogenic. Methods We knocked out the critical autophagy enzyme, ATG7, in the β-cells of mice (ATG7Δβ-cell) then monitored blood glucose, performed glucose tolerance tests, and evaluated bulk islet mRNA and protein. We also assessed MHC-I expression and presence of CD45+ immune cells in ATG7Δβ-cell islets and evaluated how impaired autophagy affects EndoC-βH1 HLA-I expression under basal and IFNα stimulated conditions. Lastly, we co-cultured ATG7Δβ-cell islet cells with diabetogenic BDC2.5 helper T cells and evaluated T cell activation. Results We found that all ATG7Δβ-cell mice developed diabetes between 11-15 weeks of age. Gene ontology analysis revealed a significant upregulation of pathways involved in inflammatory processes, response to ER stress, and the ER-associated degradation pathway. Interestingly, we also observed upregulation of proteins involved in MHC-I presentation, suggesting that defective β-cell autophagy may alter the immunopeptidome, or antigen repertoire, and enhance β-cell immune visibility. In support of this hypothesis, we observed increased MHC-I expression and CD45+ immune cells in ATG7Δβ-cell islets. We also demonstrate that HLA-I is upregulated in EndoC β-cells when autophagic degradation is inhibited. This effect was observed under both basal and IFNα stimulated conditions. Conversely, a stimulator of lysosome acidification/function, C381, decreased HLA-I expression. Lastly, we showed that in the presence of islet cells with defective autophagy, there is enhanced BDC2.5 T cell activation. Conclusions Our findings demonstrate that β-cell autophagy is critical to cell survival/function. Defective β-cell autophagy induces ER stress, alters pathways of antigen production, and enhances MHC-I/HLA-I presentation to surveilling immune cells. Overall, our results suggest that defects in autophagy make β-cells more susceptible to immune attack and destruction.
Collapse
Affiliation(s)
- Matthew C. Austin
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Charanya Muralidharan
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Saptarshi Roy
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Justin J. Crowder
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jon D. Piganelli
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Amelia K. Linnemann
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
- Indiana Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
9
|
Deivasigamani S, Thekkan S, Vergara HM, Conolly O, Cosden M, Phan T, Smith S, Marcus J, Uslaner J, Venkat D, Drolet RE, Krishnan Y, Modi S. Multimodal Blood-Based Biomarker Panel Reveals Altered Lysosomal Ionic Content in Alzheimer's Disease. ACS Chem Biol 2025; 20:137-152. [PMID: 39699875 DOI: 10.1021/acschembio.4c00602] [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: 12/20/2024]
Abstract
Lysosomal storage disorders (LSDs) and adult neurodegenerative disorders like Alzheimer's disease (AD) share various clinical and pathophysiological features. LSDs are characterized by impaired lysosomal activity caused by mutations in key proteins and enzymes. While lysosomal dysfunction is also linked to AD pathogenesis, its precise role in disease onset or progression remains unclear. Lysosomal ionic homeostasis is recognized as a key feature of many LSDs, but it has not been clinically linked with AD pathology. Thus, investigating whether this regulation is disrupted in AD is important, as it could lead to new therapeutic targets and biomarkers for this multifactorial disease. Here, using two-ion mapping (2-IM) technology, we quantitatively profiled lysosomal pH and Ca2+ in blood-derived monocytes from AD patients and age-matched controls and correlated lysosome ionicity with age and key markers of AD pathology, namely, amyloid deposits, tauopathy, neurodegeneration, and inflammation. Together, the data show that the ionic milieu of lysosomes is dysregulated in monocytes of AD patients and correlates with key plasma biomarkers of AD. Using a machine learning model based on the above parameters, we describe a proof-of-concept combinatorial biomarker platform that accurately distinguishes between patients with AD and control participants with an area under the curve of >96%. Our study introduces a convenient, noninvasive platform with the potential to diagnose Alzheimer's disease based on fluid, cellular, and molecular biomarkers. Further, these findings highlight the potential for investigating therapeutic mechanisms capable of restoring lysosome ionic homeostasis to ameliorate AD.
Collapse
Affiliation(s)
| | | | | | | | - Mali Cosden
- Neuroscience Department, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Thienlong Phan
- Neuroscience Department, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Sean Smith
- Neuroscience Department, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Jacob Marcus
- Neuroscience Department, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Jason Uslaner
- Neuroscience Department, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | | | - Robert E Drolet
- Neuroscience Department, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Yamuna Krishnan
- Esya Ltd., 84 Wood Lane, London W12 0BZ, U.K
- Department of Chemistry, The University of Chicago, Chicago, Illinois 60637, United States
- Neuroscience Institute, The University of Chicago, Chicago, Illinois 60637, United States
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, Illinois 60637, United States
| | - Souvik Modi
- Esya Ltd., 84 Wood Lane, London W12 0BZ, U.K
| |
Collapse
|
10
|
Chou CC, Vest R, Prado MA, Wilson-Grady J, Paulo JA, Shibuya Y, Moran-Losada P, Lee TT, Luo J, Gygi SP, Kelly JW, Finley D, Wernig M, Wyss-Coray T, Frydman J. Proteostasis and lysosomal repair deficits in transdifferentiated neurons of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.03.27.534444. [PMID: 37034684 PMCID: PMC10081252 DOI: 10.1101/2023.03.27.534444] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Aging is the most prominent risk factor for Alzheimer's disease (AD). However, the cellular mechanisms linking neuronal proteostasis decline to the characteristic aberrant protein deposits in AD brains remain elusive. Here, we develop transdifferentiated neurons (tNeurons) from human dermal fibroblasts as a neuronal model that retains aging hallmarks and exhibits AD-linked vulnerabilities. Remarkably, AD tNeurons accumulate proteotoxic deposits, including phospho-Tau and Aβ, resembling those in AD patient and APP mouse brains. Quantitative tNeuron proteomics identify aging and AD-linked deficits in proteostasis and organelle homeostasis, most notably in endosome-lysosomal components. Lysosomal deficits in aged tNeurons, including constitutive lysosomal damage and ESCRT-mediated lysosomal repair defects, are exacerbated in AD tNeurons and linked to inflammatory cytokine secretion and cell death. Supporting lysosomal deficits' centrality in AD, compounds ameliorating lysosomal function reduce Aβ deposits and cytokine secretion. Thus, the tNeuron model system reveals impaired lysosomal homeostasis as an early event of aging and AD.
Collapse
Affiliation(s)
- Ching-Chieh Chou
- Department of Biology, Stanford University, Stanford, California, USA
| | - Ryan Vest
- Department of Chemical Engineering, Stanford University, Stanford, California, USA
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California, USA and The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA
- Qinotto, Inc. San Carlos, California, USA
| | - Miguel A. Prado
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Joshua Wilson-Grady
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Joao A. Paulo
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Yohei Shibuya
- Departments of Pathology, Stanford University School of Medicine, Stanford, California, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Patricia Moran-Losada
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California, USA and The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA
| | - Ting-Ting Lee
- Department of Biology, Stanford University, Stanford, California, USA
| | - Jian Luo
- Palo Alto Veterans Institute for Research, Inc. (PAVIR), Palo Alto, California, USA
| | - Steven P. Gygi
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Jeffery W. Kelly
- Department of Chemistry, The Scripps Research Institute, La Jolla, California, USA
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California, USA
| | - Daniel Finley
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Marius Wernig
- Departments of Pathology, Stanford University School of Medicine, Stanford, California, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Tony Wyss-Coray
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California, USA and The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA
| | - Judith Frydman
- Department of Biology, Stanford University, Stanford, California, USA
- Department of Genetics, Stanford University, Stanford, California, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| |
Collapse
|
11
|
Su L, Hu P, Luo X, Ding H, Zhang R, Qian Y, Qi S, Tian X, Ling W. Development and Application of a BODIPY Carbazole Derivative Probe for Lysosomal Imaging: Insights into Lysosomal Dynamics and Dysfunction in Inflammation-Related Diseases. ACS APPLIED MATERIALS & INTERFACES 2025; 17:607-616. [PMID: 39688339 DOI: 10.1021/acsami.4c17607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Inflammation is crucial in neurodegenerative and chronic diseases, including Alzheimer disease (AD) and liver fibrosis. To gain a deeper understanding of lysosomal functions in cellular physiology and disease mechanisms, we developed a carbazole-based BODIPY lysosomal probe, designated LysoI. This probe specifically targets lysosomes within 15 min and exhibits a Stokes shift of approximately 180 nm, enabling continuous incubation for up to 5 h without the need for washing steps. Interestingly, LysoI remained effective for long-term imaging, even up to 24 h poststaining. Despite varying pH values and conditions, such as autophagy, apoptosis, and inflammation, it consistently provides excellent lysosomal imaging. Notably, inflammation disrupts lysosomal morphology and motility, as evidenced by an increased size, a decrease in number, and a reduction in movement speed, as observed with LysoI. Furthermore, lysosomal rupture and impaired clearance may exacerbate inflammation and contribute to cellular apoptosis. These findings suggest that lysosomal dysfunction is closely associated with disease progression; therefore, protection and repair targeting lysosomes may offer promising strategies for treating inflammation-related diseases.
Collapse
Affiliation(s)
- Liping Su
- Department of Medical Ultrasound, State Key Laboratory of Biotherapy, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, West China Hospital of Sichuan University, Sichuan University, Chengdu 610000, Sichuan, People's Republic of China
| | - Panyi Hu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, People's Republic of China
| | - Xinmei Luo
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and National Collaborative Innovation Center, Chengdu 610041, People's Republic of China
| | - Haitao Ding
- Department of Medical Ultrasound, State Key Laboratory of Biotherapy, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, West China Hospital of Sichuan University, Sichuan University, Chengdu 610000, Sichuan, People's Republic of China
| | - Rundong Zhang
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and National Collaborative Innovation Center, Chengdu 610041, People's Republic of China
| | - Yeben Qian
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, People's Republic of China
| | - Shiqian Qi
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, and National Collaborative Innovation Center, Chengdu 610041, People's Republic of China
| | - Xiaohe Tian
- Department of Medical Ultrasound, State Key Laboratory of Biotherapy, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, West China Hospital of Sichuan University, Sichuan University, Chengdu 610000, Sichuan, People's Republic of China
| | - Wenwu Ling
- Department of Medical Ultrasound, State Key Laboratory of Biotherapy, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, West China Hospital of Sichuan University, Sichuan University, Chengdu 610000, Sichuan, People's Republic of China
| |
Collapse
|
12
|
Zhang H, Liu J, Yuan W, Zhang Q, Luo X, Li Y, Peng Y, Feng J, Liu X, Chen J, Zhou Y, Lv J, Zhou N, Ma J, Tang K, Huang B. Ammonia-induced lysosomal and mitochondrial damage causes cell death of effector CD8 + T cells. Nat Cell Biol 2024; 26:1892-1902. [PMID: 39261719 DOI: 10.1038/s41556-024-01503-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 08/15/2024] [Indexed: 09/13/2024]
Abstract
Ammonia is thought to be a cytotoxin and its increase in the blood impairs cell function. However, whether and how this toxin triggers cell death under pathophysiological conditions remains unclear. Here we show that ammonia induces a distinct form of cell death in effector T cells. We found that rapidly proliferating T cells use glutaminolysis to release ammonia in the mitochondria, which is then translocated to and stored in the lysosomes. Excessive ammonia accumulation increases lysosomal pH and results in the termination of lysosomal ammonia storage and ammonia reflux into mitochondria, leading to mitochondrial damage and cell death, which is characterized by lysosomal alkalization, mitochondrial swelling and impaired autophagic flux. Inhibition of glutaminolysis or blocking lysosomal alkalization prevents ammonia-induced T cell death and improves T cell-based antitumour immunotherapy. These findings identify a distinct form of cell death that differs from previously known mechanisms.
Collapse
Affiliation(s)
- Huafeng Zhang
- Department of Pathology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan, China
| | - Jincheng Liu
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Wu Yuan
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qian Zhang
- Department of Pathology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Luo
- Department of Pathology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yonggang Li
- Hubei Provincial Key Laboratory for Applied Toxicology, Hubei Provincial Center for Disease Control and Prevention, Wuhan, China
| | - Yue'e Peng
- State Key Laboratory of Biogeology and Environmental Geology, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, China
| | - Jingyu Feng
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyu Liu
- Department of Pathology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Chen
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yabo Zhou
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiadi Lv
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nannan Zhou
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingwei Ma
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ke Tang
- Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan, China
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Huang
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
13
|
Li L, Sun B, Harris OA, Luo J. TGF-β Signaling in Microglia: A Key Regulator of Development, Homeostasis and Reactivity. Biomedicines 2024; 12:2468. [PMID: 39595034 PMCID: PMC11592028 DOI: 10.3390/biomedicines12112468] [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: 09/27/2024] [Revised: 10/18/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
Microglia, the resident immune cells of the central nervous system (CNS), are crucial for normal brain development and function. They become reactive in response to brain injury and disease, a process known as microglial reactivity. This reactivity, along with microglial homeostasis, is tightly regulated by the local microenvironment and interactions with surrounding cells. The TGF-β signaling pathway plays an essential role in this regulation. Recent genetic studies employing microglia-specific manipulation of the TGF-β signaling pathway have shed light on its significance in microglial development, homeostasis and reactivity. This review provides an updated overview of how TGF-β signaling modulates microglial function and reactivity, contributing to our understanding of microglial biology in health and disease.
Collapse
Affiliation(s)
- Lulin Li
- Palo Alto Veterans Institute for Research, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Bryan Sun
- Palo Alto Veterans Institute for Research, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Odette A. Harris
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Polytrauma System of Care, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Jian Luo
- Palo Alto Veterans Institute for Research, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
- Polytrauma System of Care, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| |
Collapse
|
14
|
Li L, Nguyen A, Zhao B, Vest R, Yerra L, Sun B, Luo J. Small Molecule Drug C381 Attenuates Brain Vascular Damage Following Repetitive Mild Traumatic Injury. Neurotrauma Rep 2024; 5:1016-1026. [PMID: 39464529 PMCID: PMC11499285 DOI: 10.1089/neur.2024.0060] [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] [Indexed: 10/29/2024] Open
Abstract
Traumatic brain injury (TBI) remains a significant public health concern, with no effective therapeutic interventions to ameliorate the enduring consequences. The prevailing understanding of TBI pathophysiology indicates a central role for vascular dysfunction. Transforming growth factor-β (TGF-β) is a multifunctional cytokine crucial for vascular development. Aberrant TGF-β signaling is implicated in vascular pathologies associated with various neurological conditions. We recently developed a novel small molecule drug, C381, a TGF-β activator with the ability to restore lysosomal function. Here we used a mouse model of repetitive mild TBI (mTBI) to examine whether C381 would attenuate vascular injury. We first employed RNA-seq analysis to investigate the gene expression patterns associated with mTBI and evaluated the therapeutic potential of C381 in mitigating these changes. Our results demonstrate distinct mTBI-related gene expression signatures, prominently implicating pathways related to vascular integrity and endothelial function. Notably, treatment with C381 reversed these mTBI-induced gene expression changes. Immunohistochemical analysis further corroborated these findings, revealing that C381 treatment attenuated vascular damage in mTBI-affected brain tissue. These findings strongly support the potential clinical usefulness of C381 as a novel therapeutic intervention for mTBI.
Collapse
Affiliation(s)
- Lulin Li
- Palo Alto Veterans Institute for Research, VA Palo Alto Health Care System, Palo Alto, California, USA
| | - Andy Nguyen
- Palo Alto Veterans Institute for Research, VA Palo Alto Health Care System, Palo Alto, California, USA
| | - Brian Zhao
- Palo Alto Veterans Institute for Research, VA Palo Alto Health Care System, Palo Alto, California, USA
| | - Ryan Vest
- Department of Chemical Engineering, Stanford University, Stanford, California, USA
| | - Lakshmi Yerra
- Palo Alto Veterans Institute for Research, VA Palo Alto Health Care System, Palo Alto, California, USA
| | - Bryan Sun
- Palo Alto Veterans Institute for Research, VA Palo Alto Health Care System, Palo Alto, California, USA
| | - Jian Luo
- Palo Alto Veterans Institute for Research, VA Palo Alto Health Care System, Palo Alto, California, USA
- Polytrauma System of Care, VA Palo Alto Health Care System, Palo Alto, California, USA
| |
Collapse
|
15
|
Zhao CC, Peng S, Wang JR, Hou X, Zhao Y, Huang F. Azobenzene-based liposomes with nanomechanical action for cytosolic chemotherapeutic drug delivery. Colloids Surf B Biointerfaces 2024; 245:114198. [PMID: 39236362 DOI: 10.1016/j.colsurfb.2024.114198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/21/2024] [Accepted: 08/30/2024] [Indexed: 09/07/2024]
Abstract
The stimuli-responsive nano-carriers are at the forefront of research in nanotechnology and materials science. These advanced systems are designed to alter their physicochemical properties upon exposure to specific stimuli, enabling controllable and targeted delivery of therapeutic agents. Nevertheless, limited endosomal escape reduces the drug bioavailability in clinical use. We herein report azobenzene (Azo)-based liposomes, prepared by co-assembling the photoisomerizable cationic Azo lipids and helper lipids, which achieve controllable doxorubicin (Dox) release and enhanced cytosolic transport upon light irradiation. Azo lipids undergo reversible isomerization between cis-isomers and trans-isomer when received UV and visible (Vis) light irradiation, causing liposomal membrane permeability changes for controlled drug release. Moreover, the nanomechanical action created by the isomerization of Azo lipids promotes the endosomal escape of the liposomes. DSPC-Azo liposomes, with minimal Dox leakage, showed significant tumor cell killing upon irradiation. For in vivo study, we co-encapsulated the upconverting nanoparticles (UCNPs), which can convert the near-infrared (NIR) light into UV/Vis emissions, facilitating Azo units activation. UCNP/Dox-loaded DSPC-Azo liposomes inhibited tumor growth under NIR irradiation in a 4T1 tumor-bearing mouse model.
Collapse
Affiliation(s)
- Cui-Cui Zhao
- Department of VIP Ward, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy (Tianjin), Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China
| | - Shiyu Peng
- State Key Laboratory of Advanced Medicals and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Jialiang Rachel Wang
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, United States
| | - Xiaoxue Hou
- State Key Laboratory of Advanced Medicals and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Yu Zhao
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, United States.
| | - Fan Huang
- State Key Laboratory of Advanced Medicals and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China.
| |
Collapse
|
16
|
Jia ZC, Yang X, Wu YK, Li M, Das D, Chen MX, Wu J. The Art of Finding the Right Drug Target: Emerging Methods and Strategies. Pharmacol Rev 2024; 76:896-914. [PMID: 38866560 PMCID: PMC11334170 DOI: 10.1124/pharmrev.123.001028] [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: 08/21/2023] [Revised: 05/28/2024] [Accepted: 05/31/2024] [Indexed: 06/14/2024] Open
Abstract
Drug targets are specific molecules in biological tissues and body fluids that interact with drugs. Drug target discovery is a key component of drug discovery and is essential for the development of new drugs in areas such as cancer therapy and precision medicine. Traditional in vitro or in vivo target discovery methods are time-consuming and labor-intensive, limiting the pace of drug discovery. With the development of modern discovery methods, the discovery and application of various emerging technologies have greatly improved the efficiency of drug discovery, shortened the cycle time, and reduced the cost. This review provides a comprehensive overview of various emerging drug target discovery strategies, including computer-assisted approaches, drug affinity response target stability, multiomics analysis, gene editing, and nonsense-mediated mRNA degradation, and discusses the effectiveness and limitations of the various approaches, as well as their application in real cases. Through the review of the aforementioned contents, a general overview of the development of novel drug targets and disease treatment strategies will be provided, and a theoretical basis will be provided for those who are engaged in pharmaceutical science research. SIGNIFICANCE STATEMENT: Target-based drug discovery has been the main approach to drug discovery in the pharmaceutical industry for the past three decades. Traditional drug target discovery methods based on in vivo or in vitro validation are time-consuming and costly, greatly limiting the development of new drugs. Therefore, the development and selection of new methods in the drug target discovery process is crucial.
Collapse
Affiliation(s)
- Zi-Chang Jia
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, China (Z.-C.J., X.Y., Y.-K.W., M.-X.C., J.W.); The Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee (D.D.); and State Key Laboratory of Crop Biology, College of Life Science, Shandong Agricultural University, Taian, Shandong, China (M.L.)
| | - Xue Yang
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, China (Z.-C.J., X.Y., Y.-K.W., M.-X.C., J.W.); The Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee (D.D.); and State Key Laboratory of Crop Biology, College of Life Science, Shandong Agricultural University, Taian, Shandong, China (M.L.)
| | - Yi-Kun Wu
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, China (Z.-C.J., X.Y., Y.-K.W., M.-X.C., J.W.); The Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee (D.D.); and State Key Laboratory of Crop Biology, College of Life Science, Shandong Agricultural University, Taian, Shandong, China (M.L.)
| | - Min Li
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, China (Z.-C.J., X.Y., Y.-K.W., M.-X.C., J.W.); The Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee (D.D.); and State Key Laboratory of Crop Biology, College of Life Science, Shandong Agricultural University, Taian, Shandong, China (M.L.)
| | - Debatosh Das
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, China (Z.-C.J., X.Y., Y.-K.W., M.-X.C., J.W.); The Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee (D.D.); and State Key Laboratory of Crop Biology, College of Life Science, Shandong Agricultural University, Taian, Shandong, China (M.L.) ;
| | - Mo-Xian Chen
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, China (Z.-C.J., X.Y., Y.-K.W., M.-X.C., J.W.); The Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee (D.D.); and State Key Laboratory of Crop Biology, College of Life Science, Shandong Agricultural University, Taian, Shandong, China (M.L.) ;
| | - Jian Wu
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, China (Z.-C.J., X.Y., Y.-K.W., M.-X.C., J.W.); The Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee (D.D.); and State Key Laboratory of Crop Biology, College of Life Science, Shandong Agricultural University, Taian, Shandong, China (M.L.) ;
| |
Collapse
|
17
|
Sun M, Cao Y, Cheng J, Xu D, Li F, Wang J, Ge Y, Liu Y, Long X, Guo W, Liu J, Fu S. Stigmasterol Activates the mTOR Signaling Pathway by Inhibiting ORP5 Ubiquitination to Promote Milk Synthesis in Bovine Mammary Epithelial Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:14769-14785. [PMID: 38912664 DOI: 10.1021/acs.jafc.4c03243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
Stigmasterol (ST), a phytosterol found in food, has various biological activities. However, the effect of ST on milk synthesis in dairy cows remains unclear. Therefore, bovine primary mammary epithelial cells (BMECs) were isolated, cultured, and treated with ST to determine the effect of ST on milk synthesis. The study revealed that 10 μM ST significantly increased milk synthesis in BMECs by activating the mammalian target of rapamycin (mTOR) signaling pathway. Further investigation revealed that this activation depends on the regulatory role of oxysterol binding protein 5 (ORP5). ST induces the translocation of ORP5 from the cytoplasm to the lysosome, interacts with the mTOR, recruits mTOR to target the lysosomal surface, and promotes the activation of the mTOR signaling pathway. Moreover, ST was found to increase ORP5 protein levels by inhibiting its degradation via the ubiquitin-proteasome pathway. Specifically, the E3 ubiquitin ligase membrane-associated cycle-CH-type finger 4 (MARCH4) promotes the ubiquitination and subsequent degradation of ORP5. ST mitigates the interaction between MARCH4 and ORP5, thereby enhancing the structural stability of ORP5 and reducing its ubiquitination. In summary, ST stabilizes ORP5 by inhibiting the interaction between MARCH4 and ORP5, thereby activating mTOR signaling pathway and enhancing milk synthesis.
Collapse
Affiliation(s)
- Mingyang Sun
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yu Cao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Ji Cheng
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Dianwen Xu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Feng Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Jiaxin Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yusong Ge
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yuhao Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xiaoyu Long
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Wenjin Guo
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Juxiong Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Shoupeng Fu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| |
Collapse
|
18
|
Singh MK, Shin Y, Ju S, Han S, Kim SS, Kang I. Comprehensive Overview of Alzheimer's Disease: Etiological Insights and Degradation Strategies. Int J Mol Sci 2024; 25:6901. [PMID: 39000011 PMCID: PMC11241648 DOI: 10.3390/ijms25136901] [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: 05/21/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder and affects millions of individuals globally. AD is associated with cognitive decline and memory loss that worsens with aging. A statistical report using U.S. data on AD estimates that approximately 6.9 million individuals suffer from AD, a number projected to surge to 13.8 million by 2060. Thus, there is a critical imperative to pinpoint and address AD and its hallmark tau protein aggregation early to prevent and manage its debilitating effects. Amyloid-β and tau proteins are primarily associated with the formation of plaques and neurofibril tangles in the brain. Current research efforts focus on degrading amyloid-β and tau or inhibiting their synthesis, particularly targeting APP processing and tau hyperphosphorylation, aiming to develop effective clinical interventions. However, navigating this intricate landscape requires ongoing studies and clinical trials to develop treatments that truly make a difference. Genome-wide association studies (GWASs) across various cohorts identified 40 loci and over 300 genes associated with AD. Despite this wealth of genetic data, much remains to be understood about the functions of these genes and their role in the disease process, prompting continued investigation. By delving deeper into these genetic associations, novel targets such as kinases, proteases, cytokines, and degradation pathways, offer new directions for drug discovery and therapeutic intervention in AD. This review delves into the intricate biological pathways disrupted in AD and identifies how genetic variations within these pathways could serve as potential targets for drug discovery and treatment strategies. Through a comprehensive understanding of the molecular underpinnings of AD, researchers aim to pave the way for more effective therapies that can alleviate the burden of this devastating disease.
Collapse
Affiliation(s)
- Manish Kumar Singh
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yoonhwa Shin
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Songhyun Ju
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sunhee Han
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sung Soo Kim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Insug Kang
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
19
|
Chou CC, Vest R, Prado MA, Wilson-Grady J, Paulo JA, Shibuya Y, Moran-Losada P, Lee TT, Luo J, Gygi SP, Kelly JW, Finley D, Wernig M, Wyss-Coray T, Frydman J. Human tNeurons reveal aging-linked proteostasis deficits driving Alzheimer's phenotypes. RESEARCH SQUARE 2024:rs.3.rs-4407236. [PMID: 38853828 PMCID: PMC11160905 DOI: 10.21203/rs.3.rs-4407236/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Aging is a prominent risk factor for Alzheimer's disease (AD), but the cellular mechanisms underlying neuronal phenotypes remain elusive. Both accumulation of amyloid plaques and neurofibrillary tangles in the brain1 and age-linked organelle deficits2-7 are proposed as causes of AD phenotypes but the relationship between these events is unclear. Here, we address this question using a transdifferentiated neuron (tNeuron) model directly from human dermal fibroblasts. Patient-derived tNeurons retain aging hallmarks and exhibit AD-linked deficits. Quantitative tNeuron proteomic analyses identify aging and AD-linked deficits in proteostasis and organelle homeostasis, particularly affecting endosome-lysosomal components. The proteostasis and lysosomal homeostasis deficits in aged tNeurons are exacerbated in sporadic and familial AD tNeurons, promoting constitutive lysosomal damage and defects in ESCRT-mediated repair. We find deficits in neuronal lysosomal homeostasis lead to inflammatory cytokine secretion, cell death and spontaneous development of Aß and phospho-Tau deposits. These proteotoxic inclusions co-localize with lysosomes and damage markers and resemble inclusions in brain tissue from AD patients and APP-transgenic mice. Supporting the centrality of lysosomal deficits driving AD phenotypes, lysosome-function enhancing compounds reduce AD-associated cytokine secretion and Aβ deposits. We conclude that proteostasis and organelle deficits are upstream initiating factors leading to neuronal aging and AD phenotypes.
Collapse
Affiliation(s)
- Ching-Chieh Chou
- Department of Biology, Stanford University, Stanford, California, USA
| | - Ryan Vest
- Department of Chemical Engineering, Stanford University, Stanford, California, USA
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California, USA
- Qinotto, Inc. San Carlos, California, USA
| | - Miguel A. Prado
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Joshua Wilson-Grady
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Joao A. Paulo
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Yohei Shibuya
- Departments of Pathology, Stanford University School of Medicine, Stanford, California, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Patricia Moran-Losada
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California, USA
| | - Ting-Ting Lee
- Department of Biology, Stanford University, Stanford, California, USA
| | - Jian Luo
- Palo Alto Veterans Institute for Research, Inc. (PAVIR), Palo Alto, California, USA
| | - Steven P. Gygi
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Jeffery W. Kelly
- Department of Chemistry, The Scripps Research Institute, La Jolla, California, USA
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California, USA
| | - Daniel Finley
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Marius Wernig
- Departments of Pathology, Stanford University School of Medicine, Stanford, California, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Tony Wyss-Coray
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California, USA
| | - Judith Frydman
- Department of Biology, Stanford University, Stanford, California, USA
- Department of Genetics, Stanford University, Stanford, California, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Lead contact
| |
Collapse
|
20
|
Sung W, Noh MY, Nahm M, Kim YS, Ki CS, Kim YE, Kim HJ, Kim SH. Progranulin haploinsufficiency mediates cytoplasmic TDP-43 aggregation with lysosomal abnormalities in human microglia. J Neuroinflammation 2024; 21:47. [PMID: 38347588 PMCID: PMC10863104 DOI: 10.1186/s12974-024-03039-1] [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: 12/21/2023] [Accepted: 02/07/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND Progranulin (PGRN) haploinsufficiency due to progranulin gene (GRN) variants can cause frontotemporal dementia (FTD) with aberrant TAR DNA-binding protein 43 (TDP-43) accumulation. Despite microglial burden with TDP-43-related pathophysiology, direct microglial TDP-43 pathology has not been clarified yet, only emphasized in neuronal pathology. Thus, the objective of this study was to investigate TDP-43 pathology in microglia of patients with PGRN haploinsufficiency. METHODS To design a human microglial cell model with PGRN haploinsufficiency, monocyte-derived microglia (iMGs) were generated from FTD-GRN patients carrying pathogenic or likely pathogenic variants (p.M1? and p.W147*) and three healthy controls. RESULTS iMGs from FTD-GRN patients with PGRN deficiency exhibited severe neuroinflammation phenotype and failure to maintain their homeostatic molecular signatures, along with impaired phagocytosis. In FTD-GRN patients-derived iMGs, significant cytoplasmic TDP-43 aggregation and accumulation of lipid droplets with profound lysosomal abnormalities were observed. These pathomechanisms were mediated by complement C1q activation and upregulation of pro-inflammatory cytokines. CONCLUSIONS Our study provides considerable cellular and molecular evidence that loss-of-function variants of GRN in human microglia can cause microglial dysfunction with abnormal TDP-43 aggregation induced by inflammatory milieu as well as the impaired lysosome. Elucidating the role of microglial TDP-43 pathology in intensifying neuroinflammation in individuals with FTD due to PGRN deficiency and examining consequential effects on microglial dysfunction might yield novel insights into the mechanisms underlying FTD and neurodegenerative disorders.
Collapse
Affiliation(s)
- Wonjae Sung
- Department of Neurology, College of Medicine, Hanyang University, 222, Wangsimni-Ro, Seongdong-Gu, Seoul, 04763, Republic of Korea
| | - Min-Young Noh
- Department of Neurology, College of Medicine, Hanyang University, 222, Wangsimni-Ro, Seongdong-Gu, Seoul, 04763, Republic of Korea
| | - Minyeop Nahm
- Dementia Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Yong Sung Kim
- Department of Neurology, College of Medicine, Hanyang University, 222, Wangsimni-Ro, Seongdong-Gu, Seoul, 04763, Republic of Korea
| | | | - Young-Eun Kim
- Department of Laboratory Medicine, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Hee-Jin Kim
- Department of Neurology, College of Medicine, Hanyang University, 222, Wangsimni-Ro, Seongdong-Gu, Seoul, 04763, Republic of Korea
| | - Seung Hyun Kim
- Department of Neurology, College of Medicine, Hanyang University, 222, Wangsimni-Ro, Seongdong-Gu, Seoul, 04763, Republic of Korea.
| |
Collapse
|
21
|
Sanyal A, Scanavachi G, Somerville E, Saminathan A, Nair A, Oikonomou A, Hatzakis NS, Kirchhausen T. Constitutive Endolysosomal Perforation in Neurons allows Induction of α-Synuclein Aggregation by Internalized Pre-Formed Fibrils. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.30.573738. [PMID: 38260258 PMCID: PMC10802249 DOI: 10.1101/2023.12.30.573738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The endocytic pathway is both an essential route of molecular uptake in cells and a potential entry point for pathology-inducing cargo. The cell-to-cell spread of cytotoxic aggregates, such as those of α-synuclein (α-syn) in Parkinson's Disease (PD), exemplifies this duality. Here we used a human iPSC-derived induced neuronal model (iNs) prone to death mediated by aggregation in late endosomes and lysosomes of endogenous α-syn, seeded by internalized pre-formed fibrils of α-syn (PFFs). This PFF-mediated death was not observed with parental iPSCs or other non-neuronal cells. Using live-cell optical microscopy to visualize the read out of biosensors reporting endo-lysosome wounding, we discovered that up to about 10% of late endosomes and lysosomes in iNs exhibited spontaneous constitutive perforations, regardless of the presence of internalized PFFs. This wounding, absent in parental iPSCs and non-neuronal cells, corresponded to partial damage by nanopores in the limiting membranes of a subset of endolysosomes directly observed by volumetric focused ion beam scanning electron microscopy (FIB-SEM) in iNs and in CA1 pyramidal neurons from mouse brain, and not found in iPSCs or in other non-neuronal cells in culture or in mouse liver and skin. We suggest that the compromised limiting membranes in iNs and neurons in general are the primary conduit for cytosolic α-syn to access PFFs entrapped within endo-lysosomal lumens, initiating PFF-mediated α-syn aggregation. Significantly, eradicating the intrinsic endolysosomal perforations in iNs by inhibiting the endosomal Phosphatidylinositol-3-Phosphate/Phosphatidylinositol 5-Kinase (PIKfyve kinase) using Apilimod or Vacuolin-1 markedly reduced PFF-induced α-syn aggregation, despite PFFs continuing to enter the endolysosomal compartment. Crucially, this intervention also diminished iN death associated with PFF incubation. Our results reveal the surprising presence of intrinsically perforated endo-lysosomes in neurons, underscoring their crucial early involvement in the genesis of toxic α-syn aggregates induced by internalized PFFs. This discovery offers a basis for employing PIKfyve kinase inhibition as a potential therapeutic strategy to counteract synucleinopathies.
Collapse
Affiliation(s)
- Anwesha Sanyal
- Department of Cell Biology, Harvard Medical School, 200 Longwood Ave, Boston, MA 02115, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, 200 Longwood Ave, Boston, MA 02115, USA
| | - Gustavo Scanavachi
- Department of Cell Biology, Harvard Medical School, 200 Longwood Ave, Boston, MA 02115, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, 200 Longwood Ave, Boston, MA 02115, USA
| | - Elliott Somerville
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, 200 Longwood Ave, Boston, MA 02115, USA
| | - Anand Saminathan
- Department of Cell Biology, Harvard Medical School, 200 Longwood Ave, Boston, MA 02115, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, 200 Longwood Ave, Boston, MA 02115, USA
| | - Athul Nair
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, 200 Longwood Ave, Boston, MA 02115, USA
| | | | - Nikos S. Hatzakis
- Department of Chemistry University of Copenhagen, 2100 Copenhagen, Denmark
| | - Tom Kirchhausen
- Department of Cell Biology, Harvard Medical School, 200 Longwood Ave, Boston, MA 02115, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, 200 Longwood Ave, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, 200 Longwood Ave, Boston, MA 02115, USA
| |
Collapse
|
22
|
Zhao Y, Ye Z, Song D, Wich D, Gao S, Khirallah J, Xu Q. Nanomechanical action opens endo-lysosomal compartments. Nat Commun 2023; 14:6645. [PMID: 37863882 PMCID: PMC10589329 DOI: 10.1038/s41467-023-42280-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 10/05/2023] [Indexed: 10/22/2023] Open
Abstract
Endo-lysosomal escape is a highly inefficient process, which is a bottleneck for intracellular delivery of biologics, including proteins and nucleic acids. Herein, we demonstrate the design of a lipid-based nanoscale molecular machine, which achieves efficient cytosolic transport of biologics by destabilizing endo-lysosomal compartments through nanomechanical action upon light irradiation. We fabricate lipid-based nanoscale molecular machines, which are designed to perform mechanical movement by consuming photons, by co-assembling azobenzene lipidoids with helper lipids. We show that lipid-based nanoscale molecular machines adhere onto the endo-lysosomal membrane after entering cells. We demonstrate that continuous rotation-inversion movement of Azo lipidoids triggered by ultraviolet/visible irradiation results in the destabilization of the membranes, thereby transporting cargoes, such as mRNAs and Cre proteins, to the cytoplasm. We find that the efficiency of cytosolic transport is improved about 2.1-fold, compared to conventional intracellular delivery systems. Finally, we show that lipid-based nanoscale molecular machines are competent for cytosolic transport of tumour antigens into dendritic cells, which induce robust antitumour activity in a melanoma mouse model.
Collapse
Affiliation(s)
- Yu Zhao
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Zhongfeng Ye
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Donghui Song
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Douglas Wich
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Shuliang Gao
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Jennifer Khirallah
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA.
| |
Collapse
|
23
|
Eberwein AE, Kulkarni SS, Rushton E, Broadie K. Glycosphingolipids are linked to elevated neurotransmission and neurodegeneration in a Drosophila model of Niemann Pick type C. Dis Model Mech 2023; 16:dmm050206. [PMID: 37815467 PMCID: PMC10581387 DOI: 10.1242/dmm.050206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 09/27/2023] [Indexed: 10/11/2023] Open
Abstract
The lipid storage disease Niemann Pick type C (NPC) causes neurodegeneration owing primarily to loss of NPC1. Here, we employed a Drosophila model to test links between glycosphingolipids, neurotransmission and neurodegeneration. We found that Npc1a nulls had elevated neurotransmission at the glutamatergic neuromuscular junction (NMJ), which was phenocopied in brainiac (brn) mutants, impairing mannosyl glucosylceramide (MacCer) glycosylation. Npc1a; brn double mutants had the same elevated synaptic transmission, suggesting that Npc1a and brn function within the same pathway. Glucosylceramide (GlcCer) synthase inhibition with miglustat prevented elevated neurotransmission in Npc1a and brn mutants, further suggesting epistasis. Synaptic MacCer did not accumulate in the NPC model, but GlcCer levels were increased, suggesting that GlcCer is responsible for the elevated synaptic transmission. Null Npc1a mutants had heightened neurodegeneration, but no significant motor neuron or glial cell death, indicating that dying cells are interneurons and that elevated neurotransmission precedes neurodegeneration. Glycosphingolipid synthesis mutants also had greatly heightened neurodegeneration, with similar neurodegeneration in Npc1a; brn double mutants, again suggesting that Npc1a and brn function in the same pathway. These findings indicate causal links between glycosphingolipid-dependent neurotransmission and neurodegeneration in this NPC disease model.
Collapse
Affiliation(s)
- Anna E. Eberwein
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN 37235, USA
| | - Swarat S. Kulkarni
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN 37235, USA
| | - Emma Rushton
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN 37235, USA
| | - Kendal Broadie
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN 37235, USA
- Department of Cell and Developmental Biology, Vanderbilt University and Medical Center, Nashville, TN 37235, USA
- Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, TN 37235, USA
- Kennedy Center for Research on Human Development, Vanderbilt University and Medical Center, Nashville, TN 37235, USA
| |
Collapse
|
24
|
de Wet S, Theart R, Loos B. Cogs in the autophagic machine-equipped to combat dementia-prone neurodegenerative diseases. Front Mol Neurosci 2023; 16:1225227. [PMID: 37720551 PMCID: PMC10500130 DOI: 10.3389/fnmol.2023.1225227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/31/2023] [Indexed: 09/19/2023] Open
Abstract
Neurodegenerative diseases are often characterized by hydrophobic inclusion bodies, and it may be the case that the aggregate-prone proteins that comprise these inclusion bodies are in fact the cause of neurotoxicity. Indeed, the appearance of protein aggregates leads to a proteostatic imbalance that causes various interruptions in physiological cellular processes, including lysosomal and mitochondrial dysfunction, as well as break down in calcium homeostasis. Oftentimes the approach to counteract proteotoxicity is taken to merely upregulate autophagy, measured by an increase in autophagosomes, without a deeper assessment of contributors toward effective turnover through autophagy. There are various ways in which autophagy is regulated ranging from the mammalian target of rapamycin (mTOR) to acetylation status of proteins. Healthy mitochondria and the intracellular energetic charge they preserve are key for the acidification status of lysosomes and thus ensuring effective clearance of components through the autophagy pathway. Both mitochondria and lysosomes have been shown to bear functional protein complexes that aid in the regulation of autophagy. Indeed, it may be the case that minimizing the proteins associated with the respective neurodegenerative pathology may be of greater importance than addressing molecularly their resulting inclusion bodies. It is in this context that this review will dissect the autophagy signaling pathway, its control and the manner in which it is molecularly and functionally connected with the mitochondrial and lysosomal system, as well as provide a summary of the role of autophagy dysfunction in driving neurodegenerative disease as a means to better position the potential of rapamycin-mediated bioactivities to control autophagy favorably.
Collapse
Affiliation(s)
- Sholto de Wet
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Rensu Theart
- Department of Electric and Electronic Engineering, Stellenbosch University, Stellenbosch, South Africa
| | - Ben Loos
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
25
|
Quick JD, Silva C, Wong JH, Lim KL, Reynolds R, Barron AM, Zeng J, Lo CH. Lysosomal acidification dysfunction in microglia: an emerging pathogenic mechanism of neuroinflammation and neurodegeneration. J Neuroinflammation 2023; 20:185. [PMID: 37543564 PMCID: PMC10403868 DOI: 10.1186/s12974-023-02866-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 07/30/2023] [Indexed: 08/07/2023] Open
Abstract
Microglia are the resident innate immune cells in the brain with a major role in orchestrating immune responses. They also provide a frontline of host defense in the central nervous system (CNS) through their active phagocytic capability. Being a professional phagocyte, microglia participate in phagocytic and autophagic clearance of cellular waste and debris as well as toxic protein aggregates, which relies on optimal lysosomal acidification and function. Defective microglial lysosomal acidification leads to impaired phagocytic and autophagic functions which result in the perpetuation of neuroinflammation and progression of neurodegeneration. Reacidification of impaired lysosomes in microglia has been shown to reverse neurodegenerative pathology in Alzheimer's disease. In this review, we summarize key factors and mechanisms contributing to lysosomal acidification impairment and the associated phagocytic and autophagic dysfunction in microglia, and how these defects contribute to neuroinflammation and neurodegeneration. We further discuss techniques to monitor lysosomal pH and therapeutic agents that can reacidify impaired lysosomes in microglia under disease conditions. Finally, we propose future directions to investigate the role of microglial lysosomal acidification in lysosome-mitochondria crosstalk and in neuron-glia interaction for more comprehensive understanding of its broader CNS physiological and pathological implications.
Collapse
Affiliation(s)
- Joseph D Quick
- Department of Integrative Biology and Physiology, Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Cristian Silva
- Faculty of Graduate Studies, University of Kelaniya, Kelaniya, Sri Lanka
| | - Jia Hui Wong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Kah Leong Lim
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Richard Reynolds
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Anna M Barron
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Jialiu Zeng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
| | - Chih Hung Lo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
26
|
Seidel M, Rajkumar S, Steffke C, Noeth V, Agarwal S, Roger K, Lipecka J, Ludolph A, Guerrera CI, Boeckers T, Catanese A. Propranolol reduces the accumulation of cytotoxic aggregates in C9orf72-ALS/FTD in vitro models. CURRENT RESEARCH IN NEUROBIOLOGY 2023; 5:100105. [PMID: 37576491 PMCID: PMC10412779 DOI: 10.1016/j.crneur.2023.100105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 06/23/2023] [Accepted: 07/26/2023] [Indexed: 08/15/2023] Open
Abstract
Mutations in the C9orf72 gene are the most common cause of familial amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). The pathogenetic mechanisms linked to this gene are a direct consequence of an aberrant intronic expansion of a GGGGCC hexanucleotide located between the 1a and 1b non-coding exons, which can be transcribed to form cytotoxic RNA foci or even translated into aggregation-prone dipeptide repeat proteins. Importantly, the abnormal length of these repeats affects also the expression levels of C9orf72 itself, which suggests haploinsufficiency as additional pathomechanism. Thus, it appears that both toxic gain of function and loss of function are distinct but still coexistent features contributing to the insurgence of the disease in case of C9orf72 mutations. In this study, we aimed at identifying a strategy to address both aspects of the C9orf72-related pathobiochemistry and provide proof-of-principle information for a better understanding of the mechanisms leading to neuronal loss. By using primary neurons overexpressing toxic poly(GA), the most abundant protein product of the GGGGCC repeats, we found that the antiarrhythmic drug propranolol could efficiently reduce the accumulation of aberrant aggregates and increase the survival of C9orf72-related cultures. Interestingly, the improved catabolism appeared to not depend on major degradative pathways such as autophagy and the proteasome. By analyzing the proteome of poly(GA)-expressing neurons after exposure to propranolol, we found that the drug increased lysosomal degradation through a mechanism directly involving C9orf72 protein, whose levels were increased after treatment. Further confirmation of the beneficial effect of the beta blocker on aggregates' accumulation and survival of hiPSC-derived C9orf72-mutant motoneurons strengthened the finding that addressing both facets of C9orf72 pathology might represent a valid strategy for the treatment of these ALS/FTD cases.
Collapse
Affiliation(s)
- Mira Seidel
- Institute of Anatomy and Cell Biology, Ulm University School of Medicine, Ulm, Germany
| | - Sandeep Rajkumar
- Institute of Anatomy and Cell Biology, Ulm University School of Medicine, Ulm, Germany
| | - Christina Steffke
- Institute of Anatomy and Cell Biology, Ulm University School of Medicine, Ulm, Germany
| | - Vivien Noeth
- Institute of Anatomy and Cell Biology, Ulm University School of Medicine, Ulm, Germany
- International Graduate School in Molecular Medicine, Ulm University, Ulm, Germany
| | - Shreya Agarwal
- Institute of Anatomy and Cell Biology, Ulm University School of Medicine, Ulm, Germany
- International Graduate School in Molecular Medicine, Ulm University, Ulm, Germany
| | - Kevin Roger
- Proteomics Platform Necker, Université Paris Cité - Structure Fédérative de Recherche Necker, INSERM US24/CNRS UAR3633, Paris, France
| | - Joanna Lipecka
- Proteomics Platform Necker, Université Paris Cité - Structure Fédérative de Recherche Necker, INSERM US24/CNRS UAR3633, Paris, France
| | - Albert Ludolph
- Department of Neurology, Ulm University School of Medicine, Ulm, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Ulm site, Ulm, Germany
| | - Chiara Ida Guerrera
- Proteomics Platform Necker, Université Paris Cité - Structure Fédérative de Recherche Necker, INSERM US24/CNRS UAR3633, Paris, France
| | - Tobias Boeckers
- Institute of Anatomy and Cell Biology, Ulm University School of Medicine, Ulm, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Ulm site, Ulm, Germany
| | - Alberto Catanese
- Institute of Anatomy and Cell Biology, Ulm University School of Medicine, Ulm, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Ulm site, Ulm, Germany
| |
Collapse
|
27
|
Lo CH, Zeng J. Defective lysosomal acidification: a new prognostic marker and therapeutic target for neurodegenerative diseases. Transl Neurodegener 2023; 12:29. [PMID: 37287072 PMCID: PMC10249214 DOI: 10.1186/s40035-023-00362-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 05/12/2023] [Indexed: 06/09/2023] Open
Abstract
Lysosomal acidification dysfunction has been implicated as a key driving factor in the pathogenesis of neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease. Multiple genetic factors have been linked to lysosomal de-acidification through impairing the vacuolar-type ATPase and ion channels on the organelle membrane. Similar lysosomal abnormalities are also present in sporadic forms of neurodegeneration, although the underlying pathogenic mechanisms are unclear and remain to be investigated. Importantly, recent studies have revealed early occurrence of lysosomal acidification impairment before the onset of neurodegeneration and late-stage pathology. However, there is a lack of methods for organelle pH monitoring in vivo and a dearth of lysosome-acidifying therapeutic agents. Here, we summarize and present evidence for the notion of defective lysosomal acidification as an early indicator of neurodegeneration and urge the critical need for technological advancement in developing tools for lysosomal pH monitoring and detection both in vivo and for clinical applications. We further discuss current preclinical pharmacological agents that modulate lysosomal acidification, including small molecules and nanomedicine, and their potential clinical translation into lysosome-targeting therapies. Both timely detection of lysosomal dysfunction and development of therapeutics that restore lysosomal function represent paradigm shifts in targeting neurodegenerative diseases.
Collapse
Affiliation(s)
- Chih Hung Lo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore.
| | - Jialiu Zeng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore.
| |
Collapse
|
28
|
Niu Z, Tang G, Wang X, Yang X, Zhao Y, Wang Y, Liu Q, Zhang F, Zhao Y, Ding X, Hao X. Trigonochinene E promotes lysosomal biogenesis and enhances autophagy via TFEB/TFE3 in human degenerative NP cells against oxidative stress. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 112:154720. [PMID: 36868108 DOI: 10.1016/j.phymed.2023.154720] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 02/01/2023] [Accepted: 02/15/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Macroautophagy (henceforth autophagy) is the major form of autophagy, which delivers intracellular cargo to lysosomes for degradation. Considerable research has revealed that the impairment of lysosomal biogenesis and autophagic flux exacerbates the development of autophagy-related diseases. Therefore, reparative medicines restoring lysosomal biogenesis and autophagic flux in cells may have therapeutic potential against the increasing prevalence of these diseases. PURPOSE The aim of the present study was thus to explore the effect of trigonochinene E (TE), an aromatic tetranorditerpene isolated from Trigonostemon flavidus, on lysosomal biogenesis and autophagy and to elucidate the potential underlying mechanism. METHODS Four human cell lines, HepG2, nucleus pulposus (NP), HeLa and HEK293 cells were applied in this study. The cytotoxicity of TE was evaluated by MTT assay. Lysosomal biogenesis and autophagic flux induced by 40 μM TE were analyzed using gene transfer techniques, western blotting, real-time PCR and confocal microscopy. Immunofluorescence, immunoblotting and pharmacological inhibitors/activators were applied to determine the changes in the protein expression levels in mTOR, PKC, PERK, and IRE1α signaling pathways. RESULTS Our results showed that TE promotes lysosomal biogenesis and autophagic flux by activating the transcription factors of lysosomes, transcription factor EB (TFEB) and transcription factor E3 (TFE3). Mechanistically, TE induces TFEB and TFE3 nuclear translocation through an mTOR/PKC/ROS-independent and endoplasmic reticulum (ER) stress-mediated pathway. The PERK and IRE1α branches of ER stress are crucial for TE-induced autophagy and lysosomal biogenesis. Whereas TE activated PERK, which mediated calcineurin dephosphorylation of TFEB/TFE3, IRE1α was activated and led to inactivation of STAT3, which further enhanced autophagy and lysosomal biogenesis. Functionally, knockdown of TFEB or TFE3 impairs TE-induced lysosomal biogenesis and autophagic flux. Furthermore, TE-induced autophagy protects NP cells from oxidative stress to ameliorate intervertebral disc degeneration (IVDD). CONCLUSIONS Here, our study showed that TE can induce TFEB/TFE3-dependent lysosomal biogenesis and autophagy via the PERK-calcineurin axis and IRE1α-STAT3 axis. Unlike other agents regulating lysosomal biogenesis and autophagy, TE showed limited cytotoxicity, thereby providing a new direction for therapeutic opportunities to use TE to treat diseases with impaired autophagy-lysosomal pathways, including IVDD.
Collapse
Affiliation(s)
- Zhenpeng Niu
- School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou 550025, China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany Chinese Academy of Sciences, Kunming, Yunnan 650201, China; Research Unit of Chemical Biology of Natural Anti-Virus Products, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Guihua Tang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Xuenan Wang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany Chinese Academy of Sciences, Kunming, Yunnan 650201, China; Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Xu Yang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Yueqin Zhao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Yinyuan Wang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany Chinese Academy of Sciences, Kunming, Yunnan 650201, China; School of Life Sciences, Yunnan University, Kunming, Yunnan 650091, China
| | - Qin Liu
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, Guizhou 550014, China
| | - Fan Zhang
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Yuhan Zhao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany Chinese Academy of Sciences, Kunming, Yunnan 650201, China.
| | - Xiao Ding
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany Chinese Academy of Sciences, Kunming, Yunnan 650201, China; Research Unit of Chemical Biology of Natural Anti-Virus Products, Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Xiaojiang Hao
- School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou 550025, China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany Chinese Academy of Sciences, Kunming, Yunnan 650201, China; Research Unit of Chemical Biology of Natural Anti-Virus Products, Chinese Academy of Medical Sciences, Beijing 100730, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, Guizhou 550014, China.
| |
Collapse
|
29
|
Simon MJ, Logan T, DeVos SL, Di Paolo G. Lysosomal functions of progranulin and implications for treatment of frontotemporal dementia. Trends Cell Biol 2023; 33:324-339. [PMID: 36244875 DOI: 10.1016/j.tcb.2022.09.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/15/2022] [Accepted: 09/15/2022] [Indexed: 12/12/2022]
Abstract
Loss-of-function heterozygous mutations in GRN, the gene encoding progranulin (PGRN), were identified in patients with frontotemporal lobar degeneration (FTLD) almost two decades ago and are generally linked to reduced PGRN protein expression levels. Although initial characterization of PGRN function primarily focused on its role in extracellular signaling as a secreted protein, more recent studies revealed critical roles of PGRN in regulating lysosome function, including proteolysis and lipid degradation, consistent with its lysosomal localization. Emerging from these studies is the notion that PGRN regulates glucocerebrosidase activity via direct chaperone activities and via interaction with prosaposin (i.e., a key regulator of lysosomal sphingolipid-metabolizing enzymes), as well as with the anionic phospholipid bis(monoacylglycero)phosphate. This emerging lysosomal biology of PGRN identified novel and promising opportunities in therapeutic discovery as well as biomarker development.
Collapse
Affiliation(s)
| | - Todd Logan
- Denali Therapeutics, South San Francisco, CA, USA
| | | | | |
Collapse
|
30
|
Sahoo S, Padhy AA, Kumari V, Mishra P. Role of Ubiquitin-Proteasome and Autophagy-Lysosome Pathways in α-Synuclein Aggregate Clearance. Mol Neurobiol 2022; 59:5379-5407. [PMID: 35699874 DOI: 10.1007/s12035-022-02897-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 05/21/2022] [Indexed: 11/26/2022]
Abstract
Synuclein aggregation in neuronal cells is the primary underlying cause of synucleinopathies. Changes in gene expression patterns, structural modifications, and altered interactions with other cellular proteins often trigger aggregation of α-synuclein, which accumulates as oligomers or fibrils in Lewy bodies. Although fibrillar forms of α-synuclein are primarily considered pathological, recent studies have revealed that even the intermediate states of aggregates are neurotoxic, complicating the development of therapeutic interventions. Autophagy and ubiquitin-proteasome pathways play a significant role in maintaining the soluble levels of α-synuclein inside cells; however, the heterogeneous nature of the aggregates presents a significant bottleneck to its degradation by these cellular pathways. With studies focused on identifying the proteins that modulate synuclein aggregation and clearance, detailed mechanistic insights are emerging about the individual and synergistic effects of these degradation pathways in regulating soluble α-synuclein levels. In this article, we discuss the impact of α-synuclein aggregation on autophagy-lysosome and ubiquitin-proteasome pathways and the therapeutic strategies that target various aspects of synuclein aggregation or degradation via these pathways. Additionally, we also highlight the natural and synthetic compounds that have shown promise in alleviating the cellular damage caused due to synuclein aggregation.
Collapse
Affiliation(s)
- Subhashree Sahoo
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, India
| | - Amrita Arpita Padhy
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, India
| | - Varsha Kumari
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, India
| | - Parul Mishra
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, India.
| |
Collapse
|
31
|
Luo J. TGF-β as a Key Modulator of Astrocyte Reactivity: Disease Relevance and Therapeutic Implications. Biomedicines 2022; 10:1206. [PMID: 35625943 PMCID: PMC9138510 DOI: 10.3390/biomedicines10051206] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/12/2022] [Accepted: 05/20/2022] [Indexed: 02/06/2023] Open
Abstract
Astrocytes are essential for normal brain development and functioning. They respond to brain injury and disease through a process referred to as reactive astrogliosis, where the reactivity is highly heterogenous and context-dependent. Reactive astrocytes are active contributors to brain pathology and can exert beneficial, detrimental, or mixed effects following brain insults. Transforming growth factor-β (TGF-β) has been identified as one of the key factors regulating astrocyte reactivity. The genetic and pharmacological manipulation of the TGF-β signaling pathway in animal models of central nervous system (CNS) injury and disease alters pathological and functional outcomes. This review aims to provide recent understanding regarding astrocyte reactivity and TGF-β signaling in brain injury, aging, and neurodegeneration. Further, it explores how TGF-β signaling modulates astrocyte reactivity and function in the context of CNS disease and injury.
Collapse
Affiliation(s)
- Jian Luo
- Palo Alto Veterans Institute for Research, VAPAHCS, Palo Alto, CA 94304, USA
| |
Collapse
|