1
|
Bezrukova AI, Basharova KS, Emelyanov AK, Rybakov AV, Miliukhina IV, Pchelina SN, Usenko TS. Autophagy Process in Parkinson's Disease Depends on Mutations in the GBA1 and LRRK2 Genes. Biochem Genet 2025:10.1007/s10528-025-11125-z. [PMID: 40388077 DOI: 10.1007/s10528-025-11125-z] [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: 01/09/2025] [Accepted: 04/28/2025] [Indexed: 05/20/2025]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder characterized by the loss of dopaminergic neurons and abnormal aggregation of the alpha-synuclein protein. Disruption of the autophagy-lysosomal pathway is closely associated with PD pathogenesis. Here, using western-blot analysis we assessed the level of autophagy-related proteins, including phosphorylated mTOR (p-mTOR), phosphorylated RPS6 (p-RPS6), beclin-1 (BECN1), LC3B, p62, and cathepsin D (CTSD) in macrophages derived from peripheral blood mononuclear cells (PBMC-derived macrophages) of GBA1-PD (p.N370S/N, p.L444P/N), LRRK2-PD (p.G2019S/N), idiopathic PD (iPD) patients, and healthy controls. Our findings revealed mutation-specific disruptions in autophagy pathways among PD patients. In p.N370S-GBA1-PD, PBMC-derived macrophages exhibited elevated levels of p-RPS6, BECN1, LC3B-II and decreased mature form of CTSD levels suggesting more active mTOR-dependent autophagy initiation alongside potential autophagosome accumulation that may lead to downregulation of lysosomal degradation. p.L444P-GBA1-PD PBMC-derived macrophages showed increased levels of p-RPS6 and BECN1, coupled with decreased p62 levels and stable mature form of CTSD and LC3B-II, indicative of enhanced autophagy flux driven by mTOR activity without evident lysosomal dysfunction. In p.G2019S-LRRK2-PD patients, PBMC-derived macrophages demonstrated elevated p-RPS6, LC3B-II, and mature CTSD levels, alongside reduced p62 levels. These changes suggest higher basal autophagosome abundance in steady-state autophagy and turnover, potentially driven by lysosomal alterations rather than direct mTOR dysregulation. These mutation-dependent differences highlight distinct autophagy dynamics in GBA1-PD and LRRK2-PD, underscoring the critical role of genetic mutations in modulating PD pathogenesis. Our results emphasize the necessity for subtype-specific therapeutic strategies targeting autophagy and other mTOR-regulated pathways to address the heterogeneity of PD mechanisms.
Collapse
Affiliation(s)
- A I Bezrukova
- Petersburg Nuclear Physics Institute named by B.P.Konstantinov of NRC «Kurchatov Institute», 1, mkr. Orlova roshcha, 188300, Gatchina, Russia
- Pavlov First Saint Petersburg State Medical University, 6-8 Lva Tolstogo Street, 197022, Saint Petersburg, Russia
| | - K S Basharova
- Petersburg Nuclear Physics Institute named by B.P.Konstantinov of NRC «Kurchatov Institute», 1, mkr. Orlova roshcha, 188300, Gatchina, Russia
- Pavlov First Saint Petersburg State Medical University, 6-8 Lva Tolstogo Street, 197022, Saint Petersburg, Russia
| | - A K Emelyanov
- Petersburg Nuclear Physics Institute named by B.P.Konstantinov of NRC «Kurchatov Institute», 1, mkr. Orlova roshcha, 188300, Gatchina, Russia
- Pavlov First Saint Petersburg State Medical University, 6-8 Lva Tolstogo Street, 197022, Saint Petersburg, Russia
| | - A V Rybakov
- Institute of the Human Brain, Russian Academy of Sciences (RAS), 9 Akademika Pavlova Street, Saint Petersburg, Russia
| | - I V Miliukhina
- Institute of the Human Brain, Russian Academy of Sciences (RAS), 9 Akademika Pavlova Street, Saint Petersburg, Russia
| | - S N Pchelina
- Petersburg Nuclear Physics Institute named by B.P.Konstantinov of NRC «Kurchatov Institute», 1, mkr. Orlova roshcha, 188300, Gatchina, Russia
- Pavlov First Saint Petersburg State Medical University, 6-8 Lva Tolstogo Street, 197022, Saint Petersburg, Russia
| | - T S Usenko
- Petersburg Nuclear Physics Institute named by B.P.Konstantinov of NRC «Kurchatov Institute», 1, mkr. Orlova roshcha, 188300, Gatchina, Russia.
- Pavlov First Saint Petersburg State Medical University, 6-8 Lva Tolstogo Street, 197022, Saint Petersburg, Russia.
| |
Collapse
|
2
|
He X, Tian S, Bu L, Zhao X, Zheng L, Zhang P, Guo R, Ma M. Cathepsin D inhibits AGEs-induced phenotypic transformation in vascular smooth muscle cells. Sci Rep 2025; 15:11502. [PMID: 40181129 PMCID: PMC11968932 DOI: 10.1038/s41598-025-96038-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 03/25/2025] [Indexed: 04/05/2025] Open
Abstract
This study investigates the role of Cathepsin D (CTSD) in diabetic vascular complications, particularly its impact on the phenotypic transformation of vascular smooth muscle cells (VSMCs) induced by advanced glycation end-products (AGEs), and explores its potential molecular mechanisms. CTSD was overexpressed in VSMCs using lentiviral vectors. Various methods, including CCK-8, immunofluorescence, SA-β-Gal staining, EdU assay, scratch assay, cell cycle analysis, and Western blotting, were employed to assess VSMC viability, proliferation, migration, senescence, and apoptosis. Additionally, transcriptomic and metabolomic analyses were conducted to investigate the molecular mechanisms underlying CTSD overexpression in VSMCs. AGEs treatment significantly inhibited CTSD expression in VSMCs, leading to reduced cell viability, enhanced proliferation and migration, increased senescence, and apoptosis. In contrast, overexpression of CTSD effectively inhibited AGEs-induced VSMCs proliferation, migration, senescence, and apoptosis. Combined transcriptomic and metabolomic analyses suggested that CTSD may affect VSMCs phenotypic transformation by inhibiting the glycolysis pathway. This study highlights the critical role of CTSD in the phenotypic transformation of VSMCs induced by AGEs and provides a new perspective for cardiovascular and cerebrovascular disease treatment. CTSD may emerge as a novel therapeutic target, though its specific molecular mechanisms and clinical application prospects in VSMCs phenotypic transformation require further investigation.
Collapse
Affiliation(s)
- Xingmin He
- Fenyang College of Shanxi Medical University, Fenyang, 032200, Shanxi, China
| | - Songhao Tian
- Department of Medical Laboratory Science, Fenyang College of Shanxi Medical University, Fenyang, 032200, Shanxi, China
| | - Lixia Bu
- Department of Geratology, Fenyang Hospital of Shanxi Province, Fenyang, 032200, Shanxi, China
| | - Xinna Zhao
- Research Office, Fenyang Hospital of Shanxi Province, Fenyang, 032200, Shanxi, China
| | - Liqiang Zheng
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200000, China
| | - Peigang Zhang
- Department of Cardiothoracic Surgery, Lvliang People's Hospital, Li Shi, 033000, Shanxi, China
| | - Renwei Guo
- Department of Cardiology, Fenyang Hospital of Shanxi Province, Fenyang, 032200, Shanxi, China.
| | - Mingfeng Ma
- Department of Cardiology, Fenyang Hospital of Shanxi Province, Fenyang, 032200, Shanxi, China.
- Department of Internal Medicine, Fenyang College of Shanxi Medical University, Fenyang, 032200, Shanxi, China.
| |
Collapse
|
3
|
Zhang Y, Du B, Zou M, Peng B, Rao Y. Neuronal Ceroid Lipofuscinosis-Concepts, Classification, and Avenues for Therapy. CNS Neurosci Ther 2025; 31:e70261. [PMID: 39925015 PMCID: PMC11808193 DOI: 10.1111/cns.70261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/26/2025] [Accepted: 01/28/2025] [Indexed: 02/11/2025] Open
Abstract
Neuronal ceroid lipofuscinosis (NCL) is a group of neurodegenerative lysosomal storage disorders characterized by excessive accumulation of lysosomal lipofuscin. Thirteen subtypes of NCL have been identified, each associated with distinct genes encoding various transmembrane proteins, secretory proteins, or lysosomal enzymes. Clinically, NCL manifests in infants through vision impairment, motor and cognitive dysfunctions, epilepsy, and premature death. The pathological complexity of NCL has hindered the development of effective clinical protocols. Current treatment modalities, including enzyme replacement therapy, pharmacological approaches, gene therapy, and stem cell therapy, have demonstrated limited efficacy. However, emerging evidence suggests a significant relationship between NCL and microglial cells, highlighting the potential of novel microglial cell replacement therapies. This review comprehensively examines the pathogenic genes associated with various NCL subtypes, elucidating their roles, clinical presentations, and corresponding mouse models. Especially, we thoroughly discuss the advances in the clinical study of potential therapeutics, which crucially calls for early diagnosis and treatment more than ever.
Collapse
Affiliation(s)
- Yuheng Zhang
- Department of Neurology, Zhongshan Hospital, Laboratory Animal CenterFudan UniversityShanghaiChina
- Children’s Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory DiseasesFudan UniversityShanghaiChina
| | - Bingying Du
- Children’s Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory DiseasesFudan UniversityShanghaiChina
- Department of NeurologyThe First Affiliated Hospital of Naval Medical UniversityShanghaiChina
| | - Miaozhan Zou
- Department of Neurology, Zhongshan Hospital, Laboratory Animal CenterFudan UniversityShanghaiChina
- Children’s Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory DiseasesFudan UniversityShanghaiChina
| | - Bo Peng
- Children’s Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory DiseasesFudan UniversityShanghaiChina
| | - Yanxia Rao
- Department of Neurology, Zhongshan Hospital, Laboratory Animal CenterFudan UniversityShanghaiChina
| |
Collapse
|
4
|
Xiong N, Du Y, Huang C, Yan Q, Zhao L, Yang C, Sun Q, Gao Z, Wang C, Zhan J, Zhang H, Wang S, Ye Y, Li Y, Shen Z. N-glycosylation Modification of CTSD Affects Liver Metastases in Colorectal Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411740. [PMID: 39716927 PMCID: PMC11831497 DOI: 10.1002/advs.202411740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/01/2024] [Indexed: 12/25/2024]
Abstract
Liver metastasis is the primary factor contributing to unfavorable prognosis in colorectal cancer (CRC). Although N-glycosylation is implicated in metastasis, there is a notable paucity of comprehensive studies addressing the N-glycosylation proteomics associated with liver metastasis in CRC. In this study, N-glycosylated proteins and N-glycosylation sites of differential expression between primary lesions and paired liver metastatic lesions are identified. Cathepsin D (CTSD) is further screened as a potentially pivotal N-glycosylated protein in CRC liver metastasis. Glycosyltransferases complex DDOST and STT3B can regulate N-glycosylation modification at residue 263 of CTSD (a protease), thereby affecting CTSD protease to lyse ACADM. ACADM can regulate ferroptosis-related proteins (ACSL4, SLC7A11, and GPX4) to further influence the invasion and metastasis of CRC cells. This newly discovered mechanism provides potential therapeutic targets for CRC treatment and insights for controlling CRC progression and metastasis.
Collapse
Affiliation(s)
- Nan Xiong
- Department of Gastroenterological SurgeryPeking University People's HospitalBeijing100044China
- Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment ResearchBeijing100044China
- Laboratory of Surgical OncologyPeking University People's HospitalBeijing100044China
| | - Yan Du
- Department of Gastroenterological SurgeryPeking University People's HospitalBeijing100044China
- Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment ResearchBeijing100044China
- Laboratory of Surgical OncologyPeking University People's HospitalBeijing100044China
| | - Chuncui Huang
- Key Laboratory of Epigenetic Regulation and InterventionInstitute of BiophysicsChinese Academy of Sciences15 Datun RoadBeijing100101China
- University of Chinese Academy of Sciences19 Yuquan RoadBeijing100049China
| | - Quanyi Yan
- Western Institute of Health Data Science28 High Tech AvenueChongqing401329China
| | - Long Zhao
- Department of Gastroenterological SurgeryPeking University People's HospitalBeijing100044China
- Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment ResearchBeijing100044China
- Laboratory of Surgical OncologyPeking University People's HospitalBeijing100044China
| | - Changjiang Yang
- Department of Gastroenterological SurgeryPeking University People's HospitalBeijing100044China
- Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment ResearchBeijing100044China
- Laboratory of Surgical OncologyPeking University People's HospitalBeijing100044China
| | - Qing Sun
- Key Laboratory of Epigenetic Regulation and InterventionInstitute of BiophysicsChinese Academy of Sciences15 Datun RoadBeijing100101China
- University of Chinese Academy of Sciences19 Yuquan RoadBeijing100049China
| | - Zhidong Gao
- Department of Gastroenterological SurgeryPeking University People's HospitalBeijing100044China
- Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment ResearchBeijing100044China
- Laboratory of Surgical OncologyPeking University People's HospitalBeijing100044China
| | - Caihong Wang
- Department of Gastroenterological SurgeryPeking University People's HospitalBeijing100044China
- Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment ResearchBeijing100044China
- Laboratory of Surgical OncologyPeking University People's HospitalBeijing100044China
| | - Jun Zhan
- Program for Cancer and Cell BiologyDepartment of Human AnatomyHistology and EmbryologySchool of Basic Medical SciencesPeking University Health Science CenterBeijing100191China
| | - Hongquan Zhang
- Program for Cancer and Cell BiologyDepartment of Human AnatomyHistology and EmbryologySchool of Basic Medical SciencesPeking University Health Science CenterBeijing100191China
| | - Shan Wang
- Department of Gastroenterological SurgeryPeking University People's HospitalBeijing100044China
- Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment ResearchBeijing100044China
- Laboratory of Surgical OncologyPeking University People's HospitalBeijing100044China
| | - Yingjiang Ye
- Department of Gastroenterological SurgeryPeking University People's HospitalBeijing100044China
- Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment ResearchBeijing100044China
- Laboratory of Surgical OncologyPeking University People's HospitalBeijing100044China
| | - Yan Li
- Key Laboratory of Epigenetic Regulation and InterventionInstitute of BiophysicsChinese Academy of Sciences15 Datun RoadBeijing100101China
- University of Chinese Academy of Sciences19 Yuquan RoadBeijing100049China
| | - Zhanlong Shen
- Department of Gastroenterological SurgeryPeking University People's HospitalBeijing100044China
- Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment ResearchBeijing100044China
- Laboratory of Surgical OncologyPeking University People's HospitalBeijing100044China
| |
Collapse
|
5
|
Szentgyörgyi V, Lueck LM, Overwijn D, Ritz D, Zoeller N, Schmidt A, Hondele M, Spang A, Bakhtiar S. Arf1-dependent LRBA recruitment to Rab4 endosomes is required for endolysosome homeostasis. J Cell Biol 2024; 223:e202401167. [PMID: 39325073 PMCID: PMC11449124 DOI: 10.1083/jcb.202401167] [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: 02/03/2024] [Revised: 07/15/2024] [Accepted: 08/06/2024] [Indexed: 09/27/2024] Open
Abstract
Deleterious mutations in the lipopolysaccharide responsive beige-like anchor protein (LRBA) gene cause severe childhood immune dysregulation. The complexity of the symptoms involving multiple organs and the broad range of unpredictable clinical manifestations of LRBA deficiency complicate the choice of therapeutic interventions. Although LRBA has been linked to Rab11-dependent trafficking of the immune checkpoint protein CTLA-4, its precise cellular role remains elusive. We show that LRBA, however, only slightly colocalizes with Rab11. Instead, LRBA is recruited by members of the small GTPase Arf protein family to the TGN and to Rab4+ endosomes, where it controls intracellular traffic. In patient-derived fibroblasts, loss of LRBA led to defects in the endosomal pathway promoting the accumulation of enlarged endolysosomes and lysosome secretion. Thus, LRBA appears to regulate flow through the endosomal system on Rab4+ endosomes. Our data strongly suggest functions of LRBA beyond CTLA-4 trafficking and provide a conceptual framework to develop new therapies for LRBA deficiency.
Collapse
Affiliation(s)
| | | | | | - Danilo Ritz
- Biozentrum, University of Basel, Basel, Switzerland
| | - Nadja Zoeller
- Dermatology, Goethe University Frankfurt, Frankfurt, Germany
| | | | | | - Anne Spang
- Biozentrum, University of Basel, Basel, Switzerland
| | - Shahrzad Bakhtiar
- Department of Pediatrics, Goethe-University Frankfurt, Frankfurt, Germany
| |
Collapse
|
6
|
Wu S, Lin W. The physiological role of the unfolded protein response in the nervous system. Neural Regen Res 2024; 19:2411-2420. [PMID: 38526277 PMCID: PMC11090440 DOI: 10.4103/1673-5374.393105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 12/12/2023] [Indexed: 03/26/2024] Open
Abstract
The unfolded protein response (UPR) is a cellular stress response pathway activated when the endoplasmic reticulum, a crucial organelle for protein folding and modification, encounters an accumulation of unfolded or misfolded proteins. The UPR aims to restore endoplasmic reticulum homeostasis by enhancing protein folding capacity, reducing protein biosynthesis, and promoting protein degradation. It also plays a pivotal role in coordinating signaling cascades to determine cell fate and function in response to endoplasmic reticulum stress. Recent research has highlighted the significance of the UPR not only in maintaining endoplasmic reticulum homeostasis but also in influencing various physiological processes in the nervous system. Here, we provide an overview of recent findings that underscore the UPR's involvement in preserving the function and viability of neuronal and myelinating cells under physiological conditions, and highlight the critical role of the UPR in brain development, memory storage, retinal cone development, myelination, and maintenance of myelin thickness.
Collapse
Affiliation(s)
- Shuangchan Wu
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Wensheng Lin
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
7
|
Yao Q, The E, Nedumaran B, Zhai Y, Ao L, Fullerton DA, Meng X. Cathepsin D elevates the fibrocalcific activity in human aortic valve cells through the ERK1/2-Sox9 pathway. Front Cardiovasc Med 2024; 11:1410862. [PMID: 39380629 PMCID: PMC11458440 DOI: 10.3389/fcvm.2024.1410862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 09/05/2024] [Indexed: 10/10/2024] Open
Abstract
Background Human Aortic valve interstitial cells (AVICs) from calcific aortic valve disease (CAVD)-affected valves exhibit elevated fibrocalcific activity although the underlying mechanism remains incompletely understood. This study aimed to identify endogenous factors that promote aortic valve fibrocalcification. Methods and results Proteomic analysis found increased cathepsin D levels in AVICs from CAVD-affected valves compared to AVICs from normal valves, and this finding was validated by immunoblotting. ELISA assay identified exacerbated release of cathepsin D by AVICs of diseased valves. Recombinant human cathepsin D upregulated the expression of runt-related transcription factor 2 (Runx2), alkaline phosphatase (ALP), collagen I and collagen IV in human AVICs, resulting in the deposition of calcium and collagen. Blocking of the ERK1/2-Sox9 signaling pathway markedly reduced the pro-fibrocalcific effect of cathepsin D. Moreover, normal AVICs express and release greater levels of cathepsin D when exposed to soluble matrilin 2. Knockdown of cathepsin D attenuated the fibrocalcific response induced by soluble matrilin 2. Conclusion AVICs of diseased aortic valves produce and release greater levels of cathepsin D that exerts a pro-fibrocalcific effect on AVICs through the ERK1/2-Sox9 pathway. Soluble matrilin 2 up-regulates cathepsin D to elevate AVIC fibrocalcific activity. Over-expression of cathepsin D in the aortic valve may enhance the pathobiological activities in AVICs.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xianzhong Meng
- Department of Surgery, University of Colorado, Denver, CO, United States
| |
Collapse
|
8
|
Shiro Y, Katayama S, Tsukamoto H, Yamazaki T. Pro-cathepsin D prevents aberrant protein aggregation dependent on endoplasmic reticulum protein CLN6. Mol Genet Metab 2024; 143:108539. [PMID: 39032464 DOI: 10.1016/j.ymgme.2024.108539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/15/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024]
Abstract
We previously expressed a chimeric protein in which the small heat-shock protein αB-crystallin (αBC) is fused at its N-terminus to the C-terminus of the first transmembrane segment of the endoplasmic reticulum (ER) protein mitsugumin 23 and confirmed its localization to the ER. Moreover, overexpression of this N-terminally modified αBC was shown to prevent the aggregation of the coexpressed R120G αBC variant, which is highly aggregation-prone and associated with the hereditary myopathy αB-crystallinopathy. To uncover a molecular mechanism by which the ER-anchored αBC negatively regulates the protein aggregation, we isolated proteins that bind to the ER-anchored αBC and identified the lysosomal protease cathepsin D (CTSD) as one such interacting protein. Proteolytically active CTSD is produced by multi-step processing of pro-cathepsin D (proCTSD), which is initially synthesized in the ER and delivered to lysosomes. When overexpressed, CTSD itself prevented the coexpressed R120G αBC variant from aggregating. This anti-aggregate activity was also elicited upon overexpression of the W383C CTSD variant, which is predominantly sequestered in the ER and consequently remains unprocessed, suggesting that proCTSD, rather than mature CTSD, serves to suppress the aggregation of the R120G αBC variant. Meanwhile, overexpression of the A58V CTSD variant, which is identical to wild-type CTSD except for the Ala58Val substitution within the pro-peptide, did not suppress the protein aggregation, indicating that the integrity of the pro-peptide is required for proCTSD to exert its anti-aggregate activity. Based on our previous finding that overexpression of the ER transmembrane protein CLN6 (ceroid-lipofuscinosis, neuronal 6), identified as an interacting protein of the ER-anchored αBC, prevents the R120G αBC variant from aggregating, the CLN6-proCTSD coupling was hypothesized to underpin the functionality of proCTSD within the ER. Indeed, CTSD, when overexpressed in CLN6-depleted cells, was unable to exert its anti-aggregate activity, supporting our view. Collectively, we show here that proCTSD prevents the protein aggregation through the functional association with CLN6 in the microenvironment surrounding the ER membrane, shedding light on a novel aspect of proCTSD and its potential involvement in CTSD-related disorders characterized by the accumulation of aberrant protein aggregates.
Collapse
Affiliation(s)
- Yuki Shiro
- Department of Molecular Cell Biology and Medicine, Graduate School of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan
| | - Syouichi Katayama
- Department of Molecular Cell Biology and Medicine, Graduate School of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan
| | - Haruka Tsukamoto
- Department of Molecular Cell Biology and Medicine, Graduate School of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan
| | - Tetsuo Yamazaki
- Department of Molecular Cell Biology and Medicine, Graduate School of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan.
| |
Collapse
|
9
|
Wang H, Chen G, Gong Q, Wu J, Chen P. Primary immunodeficiency-related genes and varicella-zoster virus reactivation syndrome: a Mendelian randomization study. Front Immunol 2024; 15:1403429. [PMID: 39253091 PMCID: PMC11381235 DOI: 10.3389/fimmu.2024.1403429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 08/08/2024] [Indexed: 09/11/2024] Open
Abstract
Background Currently, evidence regarding the causal relationship between primary immunodeficiency-related genes and varicella-zoster virus reactivation syndrome is limited and inconsistent. Therefore, this study employs Mendelian randomization (MR) methodology to investigate the causal relationship between the two. Methods This study selected 110 single-nucleotide polymorphisms (SNPs) of primary immunodeficiency-related genes as instrumental variables (IVs). Genetic associations of primary immunodeficiency-related genes were derived from recent genome-wide association studies (GWAS) data on human plasma protein levels and circulating immune cells. Data on genes associated with varicella-zoster virus reactivation syndrome were obtained from the GWAS Catalog and FINNGEN database, primarily analyzed using inverse variance weighting (IVW) and sensitivity analysis. Results Through MR analysis, we identified 9 primary immunodeficiency-related genes causally associated with herpes zoster and its subsequent neuralgia; determined causal associations of 20 primary immunodeficiency-related genes with three vascular lesions (stroke, cerebral aneurysm, giant cell arteritis); revealed causal associations of 10 primary immunodeficiency-related genes with two ocular diseases (retinopathy, keratitis); additionally, three primary immunodeficiency-related genes each were associated with encephalitis, cranial nerve palsy, and gastrointestinal infections. Conclusions This study discovers a certain association between primary immunodeficiency-related genes and varicella-zoster virus reactivation syndrome, yet further investigations are warranted to explore the specific mechanisms underlying these connections.
Collapse
Affiliation(s)
- Hao Wang
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Guanglei Chen
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Qian Gong
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jing Wu
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Peng Chen
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| |
Collapse
|
10
|
Xu J, Zhao C, Kang Y. The Formation and Renewal of Photoreceptor Outer Segments. Cells 2024; 13:1357. [PMID: 39195247 PMCID: PMC11352558 DOI: 10.3390/cells13161357] [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: 06/30/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
The visual system is essential for humans to perceive the environment. In the retina, rod and cone photoreceptor neurons are the initial sites where vision forms. The apical region of both cone and rod photoreceptors contains a light-sensing organelle known as the outer segment (OS), which houses tens of thousands of light-sensitive opsins. The OSs of photoreceptors are not static; they require rhythmic renewal to maintain normal physiological functions. Disruptions in OS renewal can lead to various genetic disorders, such as retinitis pigmentosa (RP). Understanding the patterns and molecular mechanisms of photoreceptor OS renewal remains one of the most intriguing topics in visual biology. This review aims to elucidate the structure of photoreceptor OSs, the molecular mechanisms underlying photoreceptor OS renewal, and the retinal diseases resulting from defects in this renewal process. Additionally, we will explore retinal diseases related to photoreceptor OS renewal and potential therapeutic strategies, concluding with a discussion on future research directions for OS renewal.
Collapse
Affiliation(s)
- Jingjin Xu
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (J.X.); (C.Z.)
- MoE Key Laboratory of Evolution and Marine Biodiversity, Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China
| | - Chengtian Zhao
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (J.X.); (C.Z.)
- MoE Key Laboratory of Evolution and Marine Biodiversity, Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China
| | - Yunsi Kang
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (J.X.); (C.Z.)
- MoE Key Laboratory of Evolution and Marine Biodiversity, Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China
| |
Collapse
|
11
|
Bonini S, Winter D. Two-Step Enrichment Facilitates Background Reduction for Proteomic Analysis of Lysosomes. J Proteome Res 2024; 23:3393-3403. [PMID: 38967832 DOI: 10.1021/acs.jproteome.4c00053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/06/2024]
Abstract
Lysosomes constitute the main degradative compartment of most mammalian cells and are involved in various cellular functions. Most of them are catalyzed by lysosomal proteins, which typically are low abundant, complicating their analysis by mass spectrometry-based proteomics. To increase analytical performance and to enable profiling of lysosomal content, lysosomes are often enriched. Two approaches have gained popularity in recent years, namely, superparamagnetic iron oxide nanoparticles (SPIONs) and immunoprecipitation from cells overexpressing a 3xHA-tagged version of TMEM192 (TMEM-IP). The effect of these approaches on the lysosomal proteome has not been investigated to date. We addressed this topic through a combination of both techniques and proteomic analysis of lysosome-enriched fractions. For SPIONs treatment, we identified altered cellular iron homeostasis and moderate changes of the lysosomal proteome. For overexpression of TMEM192, we observed more pronounced effects in lysosomal protein expression, especially for lysosomal membrane proteins and those involved in protein trafficking. Furthermore, we established a combined strategy based on the sequential enrichment of lysosomes with SPIONs and TMEM-IP. This enabled increased purity of lysosome-enriched fractions and, through TMEM-IP-based lysosome enrichment from SPIONs flow-through and eluate fractions, additional insights into the properties of individual approaches. All data are available via ProteomeXchange with PXD048696.
Collapse
Affiliation(s)
- Sara Bonini
- Institute for Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Bonn 53115, Germany
| | - Dominic Winter
- Institute for Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Bonn 53115, Germany
| |
Collapse
|
12
|
Liu XH, Liu XT, Wu Y, Li SA, Ren KD, Cheng M, Huang B, Yang Y, Liu PP. Broadening Horizons: Exploring the Cathepsin Family as Therapeutic Targets for Alzheimer's Disease. Aging Dis 2024:AD.2024.0456. [PMID: 39122455 DOI: 10.14336/ad.2024.0456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 06/02/2024] [Indexed: 08/12/2024] Open
Abstract
Alzheimer's disease (AD) is an intricate neurodegenerative disorder characterized by the accumulation of misfolded proteins, including beta-amyloid (Aβ) and tau, leading to cognitive decline. Despite decades of research, the precise mechanisms underlying its onset and progression remain elusive. Cathepsins are a family of lysosomal enzymes that play vital roles in cellular processes, including protein degradation and regulation of immune responses. Emerging evidence suggests that cathepsins may be involved in AD pathogenesis. Cathepsins can influence the activation of microglia and astrocytes, the resident immune cells in the brain. However, cathepsin dysfunction may lead to the accumulation of misfolded proteins, notably Aβ and tau. In addition, dysregulated cathepsin activity may induce an exaggerated immune response, promoting chronic inflammation and neuronal dysfunction in patients with AD. By unraveling the classification, functions, and roles of cathepsins in AD's pathogenesis, this review sheds light on their intricate involvement in this devastating disease. Targeting cathepsin activity could be a promising and novel approach for mitigating the pathological processes that contribute to AD, providing new avenues for its treatment and prevention.
Collapse
Affiliation(s)
- Xiao-Hui Liu
- Clinical Systems Biology Laboratories, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- The Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiao-Tong Liu
- Clinical Laboratory, the First Hospital of Yongnian District, Yongnian, Hebei, China
| | - Yue Wu
- Clinical Systems Biology Laboratories, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- The Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Shu-Ang Li
- Clinical Systems Biology Laboratories, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Kai-Di Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Meng Cheng
- Translational Medical Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Bing Huang
- Brain Function and Disease Laboratory, Shantou University Medical College, Shantou, China
| | - Yang Yang
- Clinical Systems Biology Laboratories, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Pei-Pei Liu
- Clinical Systems Biology Laboratories, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
13
|
Liu Y, Gu Y, Chen Y, Wang X, Zhou G, Li J, Wang M, Fang S, Yang Y. Translocational attenuation mediated by the PERK-SRP14 axis is a protective mechanism of unfolded protein response. Cell Rep 2024; 43:114402. [PMID: 38943644 DOI: 10.1016/j.celrep.2024.114402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 05/06/2024] [Accepted: 06/11/2024] [Indexed: 07/01/2024] Open
Abstract
The unfolded protein response (UPR) relieves endoplasmic reticulum (ER) stress through multiple strategies, including reducing protein synthesis, increasing protein folding capabilities, and enhancing misfolded protein degradation. After a multi-omics analysis, we find that signal recognition particle 14 (SRP14), an essential component of the SRP, is markedly reduced in cells undergoing ER stress. Further experiments indicate that SRP14 reduction requires PRKR-like ER kinase (PERK)-mediated eukaryotic translation initiation factor 2α (eIF2α) phosphorylation but is independent of ATF4 or ATF3 transcription factors. The decrease of SRP14 correlates with reduced translocation of fusion proteins and endogenous cathepsin D. Enforced expression of an SRP14 variant with elongation arrest capability prevents the reduced translocation of cathepsin D in stressed cells, whereas an SRP14 mutant without the activity does not. Finally, overexpression of SRP14 augments the UPR and aggravates ER-stress-induced cell death. These data suggest that translocational attenuation mediated by the PERK-SRP14 axis is a protective measure for the UPR to mitigate ER stress.
Collapse
Affiliation(s)
- Yaofu Liu
- China Regional Research Centre, International Centre for Genetic Engineering and Biotechnology (ICGEB), Taizhou, Jiangsu 225316, China; School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China; Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuexi Gu
- China Regional Research Centre, International Centre for Genetic Engineering and Biotechnology (ICGEB), Taizhou, Jiangsu 225316, China
| | - Ying Chen
- Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu 215123, China
| | - Xuan Wang
- China Regional Research Centre, International Centre for Genetic Engineering and Biotechnology (ICGEB), Taizhou, Jiangsu 225316, China
| | - Guangfeng Zhou
- China Regional Research Centre, International Centre for Genetic Engineering and Biotechnology (ICGEB), Taizhou, Jiangsu 225316, China
| | - Jing Li
- Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu 215123, China
| | - Mu Wang
- Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu 215123, China.
| | - Shengyun Fang
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Yili Yang
- China Regional Research Centre, International Centre for Genetic Engineering and Biotechnology (ICGEB), Taizhou, Jiangsu 225316, China.
| |
Collapse
|
14
|
Huang J, Yu Y, Pang D, Li C, Wei Q, Cheng Y, Cui Y, Ou R, Shang H. Lnc-HIBADH-4 Regulates Autophagy-Lysosome Pathway in Amyotrophic Lateral Sclerosis by Targeting Cathepsin D. Mol Neurobiol 2024; 61:4768-4782. [PMID: 38135852 PMCID: PMC11236912 DOI: 10.1007/s12035-023-03835-5] [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: 08/24/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is the most prevalent and lethal class of severe motor neuron diseases (MND) with no efficacious treatment. The pathogenic mechanisms underlying ALS remain unclear. Nearly 90% of patients exhibit sporadic onset (sALS). Therefore, elucidating the pathophysiology of ALS is imperative. Long non-coding RNA (lncRNA) is a large class of non-coding RNAs that regulate transcription, translation, and post-translational processes. LncRNAs contribute to the pathogenesis of diverse neurodegenerative disorders and hold promise as targets for interference in the realm of neurodegeneration. However, the mechanisms of which lncRNAs are involved in ALS have not been thoroughly investigated. We identified and validated a downregulated lncRNA, lnc-HIBADH-4, in ALS which correlated with disease severity and overall survival. Lnc-HIBADH-4 acted as a "molecular sponge" regulating lysosomal function through the lnc-HIBADH-4/miR-326/CTSD pathway, thereby impacting autophagy-lysosome dynamics and the levels of cell proliferation and apoptosis. Therefore, this study discovered and revealed the role of lnc-HIBADH-4 in the pathogenesis of ALS. With further research, lnc-HIBADH-4 is expected to provide a new biomarker in the diagnosis and treatment of ALS.
Collapse
Affiliation(s)
- Jingxuan Huang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Diseases Center, West China Hospital, Sichuan University, No.37, Guoxue Lane, Chengdu, 610041, Sichuan, China
| | - Yujiao Yu
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Diseases Center, West China Hospital, Sichuan University, No.37, Guoxue Lane, Chengdu, 610041, Sichuan, China
| | - Dejiang Pang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Diseases Center, West China Hospital, Sichuan University, No.37, Guoxue Lane, Chengdu, 610041, Sichuan, China
| | - Chunyu Li
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Diseases Center, West China Hospital, Sichuan University, No.37, Guoxue Lane, Chengdu, 610041, Sichuan, China
| | - Qianqian Wei
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Diseases Center, West China Hospital, Sichuan University, No.37, Guoxue Lane, Chengdu, 610041, Sichuan, China
| | - Yangfan Cheng
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Diseases Center, West China Hospital, Sichuan University, No.37, Guoxue Lane, Chengdu, 610041, Sichuan, China
| | - Yiyuan Cui
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Diseases Center, West China Hospital, Sichuan University, No.37, Guoxue Lane, Chengdu, 610041, Sichuan, China
| | - Ruwei Ou
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Diseases Center, West China Hospital, Sichuan University, No.37, Guoxue Lane, Chengdu, 610041, Sichuan, China
| | - Huifang Shang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Diseases Center, West China Hospital, Sichuan University, No.37, Guoxue Lane, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
15
|
Ng ESY, Hu J, Jiang Z, Radu RA. Impaired cathepsin D in retinal pigment epithelium cells mediates Stargardt disease pathogenesis. FASEB J 2024; 38:e23720. [PMID: 38837708 PMCID: PMC11296957 DOI: 10.1096/fj.202400210rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 05/12/2024] [Accepted: 05/21/2024] [Indexed: 06/07/2024]
Abstract
Recessive Stargardt disease (STGD1) is an inherited juvenile maculopathy caused by mutations in the ABCA4 gene, for which there is no suitable treatment. Loss of functional ABCA4 in the retinal pigment epithelium (RPE) alone, without contribution from photoreceptor cells, was shown to induce STGD1 pathology. Here, we identified cathepsin D (CatD), the primary RPE lysosomal protease, as a key molecular player contributing to endo-lysosomal dysfunction in STGD1 using a newly developed "disease-in-a-dish" RPE model from confirmed STGD1 patients. Induced pluripotent stem cell (iPSC)-derived RPE originating from three STGD1 patients exhibited elevated lysosomal pH, as previously reported in Abca4-/- mice. CatD protein maturation and activity were impaired in RPE from STGD1 patients and Abca4-/- mice. Consequently, STGD1 RPE cells have reduced photoreceptor outer segment degradation and abnormal accumulation of α-synuclein, the natural substrate of CatD. Furthermore, dysfunctional ABCA4 in STGD1 RPE cells results in intracellular accumulation of autofluorescent material and phosphatidylethanolamine (PE). The altered distribution of PE associated with the internal membranes of STGD1 RPE cells presumably compromises LC3-associated phagocytosis, contributing to delayed endo-lysosomal degradation activity. Drug-mediated re-acidification of lysosomes in the RPE of STGD1 restores CatD functional activity and reduces the accumulation of immature CatD protein loads. This preclinical study validates the contribution of CatD deficiencies to STGD1 pathology and provides evidence for an efficacious therapeutic approach targeting RPE cells. Our findings support a cell-autonomous RPE-driven pathology, informing future research aimed at targeting RPE cells to treat ABCA4-mediated retinopathies.
Collapse
Affiliation(s)
- Eunice Sze Yin Ng
- UCLA Stein Eye Institute, University of California, Los Angeles, CA 90095, USA
- Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
- Molecular, Cellular, and Integrative Physiology Interdepartmental Program, University of California, Los Angeles, CA 90095, USA
| | - Jane Hu
- UCLA Stein Eye Institute, University of California, Los Angeles, CA 90095, USA
- Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Zhichun Jiang
- UCLA Stein Eye Institute, University of California, Los Angeles, CA 90095, USA
- Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Roxana A. Radu
- UCLA Stein Eye Institute, University of California, Los Angeles, CA 90095, USA
- Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
16
|
Ji H, Han Y, Danyang Jie, Yue Li, Hailan Yang, Sun H, You C, Xiao A, Liu Y. Decoding the biology and clinical implication of neutrophils in intracranial aneurysm. Ann Clin Transl Neurol 2024; 11:958-972. [PMID: 38317016 PMCID: PMC11021671 DOI: 10.1002/acn3.52014] [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: 08/20/2023] [Revised: 10/08/2023] [Accepted: 01/11/2024] [Indexed: 02/07/2024] Open
Abstract
OBJECTIVE Abundant neutrophils have been identified in both ruptured and unruptured intracranial aneurysm (IA) domes, with their function and clinical implication being poorly characterized. MATERIALS AND METHODS We employed single-cell RNA sequencing (scRNA-Seq) datasets of both human and murine model, and external bulk mRNA sequencing datasets to thoroughly explore the features and functional heterogeneous of neutrophils infiltrating the IA dome. RESULTS We found that both unruptured and ruptured IA dome contain a substantial population of neutrophils, characterized by FCGR3B, G0S2, CSF3R, and CXCR2. These cells exhibited heterogeneity in terms of function and differentiation. Despite similar transcriptional activation, neutrophils in IA dome expressed a repertoire of gene programs that mimicked transcriptomic alterations observed from bone marrow to peripheral blood, showing self-similarity. In addition, the recruitment of neutrophils in unruptured IA was primarily mediated by monocytes/macrophages, and once ruptured, both neutrophils, and a specific subset of inflammatory smooth muscle cells (SMCs) were involved in the process. The receiver operator characteristic curve (ROC) analysis indicated that distinct neutrophil subclusters were associated with IA formation and rupture, respectively. By reviewing current studies, we found that neutrophils play a detrimental role to IA wall integrity through secreting specific ligands, ferroptosis driven by ALOX5AP and PTGS2, and the formation of neutrophil extracellular traps (NETs) mediated by PADI4. INTERPRETATION This study delineated the biology and potential clinical implications of neutrophils in IA dome and provided a reliable basis for future researches.
Collapse
Affiliation(s)
- Hang Ji
- Department of Neurosurgery, West China HospitalSichuan UniversityNo. 37 Guoxue LaneChengduSichuanChina
| | - Yujing Han
- Plevic Floor Disorders Centre, West China Tianfu HospitalSichuan UniversityNo. 3966, Tianfu AvenueChengduSichuanChina
| | - Danyang Jie
- Department of Neurosurgery, West China HospitalSichuan UniversityNo. 37 Guoxue LaneChengduSichuanChina
| | - Yue Li
- Department of Neurosurgery, West China HospitalSichuan UniversityNo. 37 Guoxue LaneChengduSichuanChina
| | - Hailan Yang
- Department of Neurosurgery, West China HospitalSichuan UniversityNo. 37 Guoxue LaneChengduSichuanChina
| | - Haogeng Sun
- Department of Neurosurgery, West China HospitalSichuan UniversityNo. 37 Guoxue LaneChengduSichuanChina
| | - Chao You
- Department of Neurosurgery, West China HospitalSichuan UniversityNo. 37 Guoxue LaneChengduSichuanChina
| | - Anqi Xiao
- Department of Neurosurgery, West China HospitalSichuan UniversityNo. 37 Guoxue LaneChengduSichuanChina
| | - Yi Liu
- Department of Neurosurgery, West China HospitalSichuan UniversityNo. 37 Guoxue LaneChengduSichuanChina
| |
Collapse
|
17
|
Zhao D, Huang ZK, Liang Y, Li ZJ, Zhang XW, Li KH, Wu H, Zhang XD, Li CS, An D, Sun X, An MX, Shi JX, Bao YJ, Tian L, Wang DF, Wu AH, Chen YH, Zhao WD. Monocytes Release Pro-Cathepsin D to Drive Blood-to-Brain Transcytosis in Diabetes. Circ Res 2024; 134:e17-e33. [PMID: 38420756 DOI: 10.1161/circresaha.123.323622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 02/15/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND Microvascular complications are the major outcome of type 2 diabetes progression, and the underlying mechanism remains to be determined. METHODS High-throughput RNA sequencing was performed using human monocyte samples from controls and diabetes. The transgenic mice expressing human CTSD (cathepsin D) in the monocytes was constructed using CD68 promoter. In vivo 2-photon imaging, behavioral tests, immunofluorescence, transmission electron microscopy, Western blot analysis, vascular leakage assay, and single-cell RNA sequencing were performed to clarify the phenotype and elucidate the molecular mechanism. RESULTS Monocytes expressed high-level CTSD in patients with type 2 diabetes. The transgenic mice expressing human CTSD in the monocytes showed increased brain microvascular permeability resembling the diabetic microvascular phenotype, accompanied by cognitive deficit. Mechanistically, the monocytes release nonenzymatic pro-CTSD to upregulate caveolin expression in brain endothelium triggering caveolae-mediated transcytosis, without affecting the paracellular route of brain microvasculature. The circulating pro-CTSD activated the caveolae-mediated transcytosis in brain endothelial cells via its binding with low-density LRP1 (lipoprotein receptor-related protein 1). Importantly, genetic ablation of CTSD in the monocytes exhibited a protective effect against the diabetes-enhanced brain microvascular transcytosis and the diabetes-induced cognitive impairment. CONCLUSIONS These findings uncover the novel role of circulatory pro-CTSD from monocytes in the pathogenesis of cerebral microvascular lesions in diabetes. The circulatory pro-CTSD is a potential target for the intervention of microvascular complications in diabetes.
Collapse
Affiliation(s)
- Dan Zhao
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
- Department of Neurosurgery, the First Affiliated Hospital of China Medical University, Shenyang, China (D.Z., K.-H.L., X.-D.Z., Y.-J.B.)
| | - Zeng-Kang Huang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Yu Liang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Zhi-Jun Li
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Xue-Wei Zhang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Kun-Hang Li
- Department of Neurosurgery, the First Affiliated Hospital of China Medical University, Shenyang, China (D.Z., K.-H.L., X.-D.Z., Y.-J.B.)
| | - Hao Wu
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Xu-Dong Zhang
- Department of Neurosurgery, the First Affiliated Hospital of China Medical University, Shenyang, China (D.Z., K.-H.L., X.-D.Z., Y.-J.B.)
| | - Chen-Sheng Li
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Dong An
- School of Mechanical Engineering, Shenyang Jianzhu University, China (D.A.)
| | - Xue Sun
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Ming-Xin An
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Jun-Xiu Shi
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Yi-Jun Bao
- Department of Neurosurgery, the First Affiliated Hospital of China Medical University, Shenyang, China (D.Z., K.-H.L., X.-D.Z., Y.-J.B.)
| | - Li Tian
- Department of Gerontology (L.T., D.-F.W.), Shengjing Hospital of China Medical University, Shenyang, China
| | - Di-Fei Wang
- Department of Gerontology (L.T., D.-F.W.), Shengjing Hospital of China Medical University, Shenyang, China
| | - An-Hua Wu
- Department of Neurosurgery (A.-H.W.), Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu-Hua Chen
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Wei-Dong Zhao
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| |
Collapse
|
18
|
Anjum F, Kaushik K, Salam A, Yadav A, Nandi CK. Super-Resolution Microscopy Unveils Synergistic Structural Changes of Organelles Upon Point Mutation. Adv Biol (Weinh) 2024; 8:e2300399. [PMID: 38053236 DOI: 10.1002/adbi.202300399] [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: 08/03/2023] [Revised: 11/02/2023] [Indexed: 12/07/2023]
Abstract
Ethyl methanesulphonate (EMS), is a widely used chemical mutagen that causes high-frequency germline null mutation by inserting an alkyl group into the nucleotide guanine in eukaryotic cells. The effect of EMS on the dynamics of the aneuploid genome, increased cellular instability, and carcinogenicity in relation to benign and malignant tumors are reported, but the molecular level understanding of morphological changes of higher-order chromatin structure has poorly been understood. This is due to a lack of sufficient resolution in conventional microscopic techniques to see small structures below the diffraction limit. Here, using super-resolution radial fluctuation, a largely fragmented, decompaction, and less dense heterochromatin structure upon EMS treatment to HEK 293A cells without any change in nuclear DNA domains is observed. This result suggests an early stage of carcinogenicity happened due to the point mutation. In addition, the distinct structural changes with an elongated morphology of lysosomes are also observed. On the other hand, fragmented and increased heterogeneous populations with an increased cytoplasmic occupancy of mitochondria are observed.
Collapse
Affiliation(s)
- Farhan Anjum
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh, 175005, India
| | - Kush Kaushik
- School of Chemical Sciences, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh, 175005, India
| | - Abdul Salam
- School of Chemical Sciences, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh, 175005, India
| | - Aditya Yadav
- School of Chemical Sciences, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh, 175005, India
| | - Chayan Kanti Nandi
- School of Chemical Sciences, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh, 175005, India
| |
Collapse
|
19
|
Swift IJ, Sjödin S, Gobom J, Brinkmalm A, Blennow K, Zetterberg H, Rohrer JD, Sogorb-Esteve A. Differential patterns of lysosomal dysfunction are seen in the clinicopathological forms of primary progressive aphasia. J Neurol 2024; 271:1277-1285. [PMID: 37917233 PMCID: PMC10896779 DOI: 10.1007/s00415-023-12063-9] [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: 08/15/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 11/04/2023]
Abstract
Increasing evidence implicates endo-lysosomal dysfunction in frontotemporal dementia (FTD). 18 proteins were quantified using a mass spectrometry assay panel in the cerebrospinal fluid of 36 people with the language variant of FTD, primary progressive aphasia (PPA) (including 13 with non-fluent variant (nfvPPA), 11 with semantic variant (svPPA), and 12 with logopenic variant (lvPPA)) and 19 healthy controls. The concentrations of the cathepsins (B, D, F, L1, and Z) as well as AP-2 complex subunit beta, ganglioside GM2 activator, beta-hexosaminidase subunit beta, tissue alpha L-fucosidase, and ubiquitin were decreased in nfvPPA compared with controls. In contrast, the concentrations of amyloid beta A4 protein, cathepsin Z, and dipeptidyl peptidase 2 were decreased in svPPA compared with controls. No proteins were abnormal in lvPPA. These results indicate a differential alteration of lysosomal proteins in the PPA variants, suggesting those with non-Alzheimer's pathologies are more likely to show abnormal lysosomal function.
Collapse
Affiliation(s)
- Imogen J Swift
- UK Dementia Research Institute at University College London, UCL Queen Square Institute of Neurology, University College London, London, UK
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Simon Sjödin
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Johan Gobom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Ann Brinkmalm
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- UK Dementia Research Institute at University College London, UCL Queen Square Institute of Neurology, University College London, London, UK
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Jonathan D Rohrer
- UK Dementia Research Institute at University College London, UCL Queen Square Institute of Neurology, University College London, London, UK
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Aitana Sogorb-Esteve
- UK Dementia Research Institute at University College London, UCL Queen Square Institute of Neurology, University College London, London, UK.
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
20
|
Chun YHP, Tan C, Villanueva O, Colley ME, Quintanilla TJ, Basiouny MS, Hartel CA, Critchfield CS, Bach SBH, Fajardo RJ, Pham CD. Overexpression of ameloblastin in secretory ameloblasts results in demarcated, hypomineralized opacities in enamel. Front Physiol 2024; 14:1233391. [PMID: 38274050 PMCID: PMC10808694 DOI: 10.3389/fphys.2023.1233391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 11/01/2023] [Indexed: 01/27/2024] Open
Abstract
Introduction: Developmental defects of the enamel manifest before tooth eruption and include amelogenesis imperfecta, a rare disease of underlying gene mutations, and molar-incisor hypomineralization (MIH), a prevalent disease in children originating from environmental and epigenetic factors. MIH enamel presents as the abnormal enamel marked by loss of translucency, demarcation between the healthy and affected enamel, and reduced mineral content. The pathophysiology of opaque, demarcated enamel lesions is not understood; however, the retention of enamel proteins in the matrix has been suggested. Ameloblastin (Ambn) is an enamel protein of the secreted calcium-binding phosphoproteins (SCPPs) critical for enamel formation. When the Ambn gene is mutated or deleted, teeth are affected by hypoplastic amelogenesis imperfecta. Methods: In this study, enamel formation in mice was analyzed when transgenic Ambn was overexpressed from the amelogenin promoter encoding full-length Ambn. Ambn was under- and overexpressed at six increasing concentrations in separate mouse lines. Results: Mice overexpressing Ambn displayed opaque enamel at low concentrations and demarcated lesions at high concentrations. The severity of enamel lesions increased starting from the inner enamel close to the dentino-enamel junction (DEJ) to span the entire width of the enamel layer in demarcated areas. Associated with the opaque enamel were 17-kDa Ambn cleavage products, a prolonged secretory stage, and a thin basement membrane in the maturation stage. Ambn accumulations found in the innermost enamel close to the DEJ and the mineralization front correlated with reduced mineral content. Demarcated enamel lesions were associated with Ambn species of 17 kDa and higher, prolonged secretory and transition stages, a thin basement membrane, and shortened maturation stages. Hypomineralized opacities were delineated against the surrounding mineralized enamel and adjacent to ameloblasts detached from the enamel surface. Inefficient Ambn cleavage, loss of contact between ameloblasts, and the altered basement membrane curtailed the endocytic activity; thus, enamel proteins remained unresorbed in the matrix. Ameloblasts have the ability to distinguish between Ambn concentration and Ambn cleavage products through finely tuned feedback mechanisms. The under- or overexpression of Ambn in murine secretory ameloblasts results in either hypoplastic amelogenesis imperfecta or hypomineralization with opaque or sharply demarcated boundaries of lesions, similar to MIH.
Collapse
Affiliation(s)
- Yong-Hee Patricia Chun
- Department of Periodontics, School of Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- Department of Cell Systems and Anatomy, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- Department of Molecular Medicine, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Chunyan Tan
- Department of Periodontics, School of Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Omar Villanueva
- Department of Periodontics, School of Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Madeline E. Colley
- Department of Chemistry, University of Texas San Antonio, San Antonio, TX, United States
- Department of Biochemistry, Vanderbilt University, Nashville, TN, United States
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN, United States
| | - Travis J. Quintanilla
- Department of Periodontics, School of Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Mohamed S. Basiouny
- Department of Periodontics, School of Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Caldonia A. Hartel
- Department of Periodontics, School of Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Cameron S. Critchfield
- Department of Periodontics, School of Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Stephan B. H. Bach
- Department of Chemistry, University of Texas San Antonio, San Antonio, TX, United States
| | - Roberto J. Fajardo
- Department of Clinical and Applied Science Education, School of Osteopathic Medicine, University of the Incarnate Word, San Antonio, TX, United States
| | - Cong-Dat Pham
- Department of Periodontics, School of Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
21
|
Cook AA, Leung TCS, Rice M, Nachman M, Zadigue-Dube É, Watt AJ. Endosomal dysfunction contributes to cerebellar deficits in spinocerebellar ataxia type 6. eLife 2023; 12:RP90510. [PMID: 38084749 PMCID: PMC10715727 DOI: 10.7554/elife.90510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023] Open
Abstract
Spinocerebellar ataxia type 6 (SCA6) is a rare disease that is characterized by cerebellar dysfunction. Patients have progressive motor coordination impairment, and postmortem brain tissue reveals degeneration of cerebellar Purkinje cells and a reduced level of cerebellar brain-derived neurotrophic factor (BDNF). However, the pathophysiological changes underlying SCA6 are not fully understood. We carried out RNA-sequencing of cerebellar vermis tissue in a mouse model of SCA6, which revealed widespread dysregulation of genes associated with the endo-lysosomal system. Since disruption to endosomes or lysosomes could contribute to cellular deficits, we examined the endo-lysosomal system in SCA6. We identified alterations in multiple endosomal compartments in the Purkinje cells of SCA6 mice. Early endosomes were enlarged, while the size of the late endosome compartment was reduced. We also found evidence for impaired trafficking of cargo to the lysosomes. As the proper functioning of the endo-lysosomal system is crucial for the sorting and trafficking of signaling molecules, we wondered whether these changes could contribute to previously identified deficits in signaling by BDNF and its receptor tropomyosin kinase B (TrkB) in SCA6. Indeed, we found that the enlarged early endosomes in SCA6 mice accumulated both BDNF and TrkB. Furthermore, TrkB recycling to the cell membrane in recycling endosomes was reduced, and the late endosome transport of BDNF for degradation was impaired. Therefore, mis-trafficking due to aberrant endo-lysosomal transport and function could contribute to SCA6 pathophysiology through alterations to BDNF-TrkB signaling, as well as mishandling of other signaling molecules. Deficits in early endosomes and BDNF localization were rescued by chronic administration of a TrkB agonist, 7,8-dihydroxyflavone, that we have previously shown restores motor coordination and cerebellar TrkB expression. The endo-lysosomal system is thus both a novel locus of pathophysiology in SCA6 and a promising therapeutic target.
Collapse
Affiliation(s)
- Anna A Cook
- Biology Department, McGill UniversityMontrealCanada
| | | | - Max Rice
- Biology Department, McGill UniversityMontrealCanada
- Department of Biological Sciences, Columbia UniversityNew YorkUnited States
| | - Maya Nachman
- Biology Department, McGill UniversityMontrealCanada
| | | | | |
Collapse
|
22
|
Deng C, Li M, Liu Y, Yan C, He Z, Chen ZY, Zhu H. Cholesterol Oxidation Products: Potential Adverse Effect and Prevention of Their Production in Foods. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:18645-18659. [PMID: 38011512 DOI: 10.1021/acs.jafc.3c05158] [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: 11/29/2023]
Abstract
Cholesterol oxidation products (COPs) are a group of substances formed during food processing. COPs in diet is a health concern because they may affect human health in association with the risk of various diseases including atherosclerosis, Alzheimer's disease, age-related macular degeneration, diabetes, and chronic gastrointestinal inflammatory colitis. Production of COPs in foods can be affected by many factors such as temperature, pH, light, oxygen, water, carbohydrates, fatty acids, proteins, and metal cations. The key issue is preventing its generation in foods. Some COPs can also be produced in vivo by both nonenzymatic and enzymatic-catalyzed oxidation reactions. Currently, a number of natural antioxidants such as catechins, flavonoids, and other polyphenols have been proven to inhibit the generation of COPs. In addition, measures taken during food processing can also minimize the production of COPs, such as the Maillard reaction and marinating food with plant polyphenol-rich seasonings. In conclusion, a comprehensive approach encompassing the suppression on COPs generation and implementation of processing measures is imperative to safeguard human health against the production of COPs in the food chain.
Collapse
Affiliation(s)
- Chuanling Deng
- School of Food Science and Engineering/Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing/National Technical Center (Foshan) for Quality Control of Famous and Special Agricultural Products (CAQS-GAP-KZZX043), Foshan University, Foshan 528000, Guangdong China
| | - Mingxuan Li
- School of Food Science and Engineering/Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing/National Technical Center (Foshan) for Quality Control of Famous and Special Agricultural Products (CAQS-GAP-KZZX043), Foshan University, Foshan 528000, Guangdong China
- School of Life Sciences, South China Agricultural University, Guangzhou 510000, Guangdong China
| | - Yang Liu
- School of Food Science and Engineering/Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing/National Technical Center (Foshan) for Quality Control of Famous and Special Agricultural Products (CAQS-GAP-KZZX043), Foshan University, Foshan 528000, Guangdong China
| | - Chi Yan
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, NT 999077, Hong Kong China
| | - Zouyan He
- School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi China
| | - Zhen-Yu Chen
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, NT 999077, Hong Kong China
| | - Hanyue Zhu
- School of Food Science and Engineering/Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing/National Technical Center (Foshan) for Quality Control of Famous and Special Agricultural Products (CAQS-GAP-KZZX043), Foshan University, Foshan 528000, Guangdong China
| |
Collapse
|
23
|
Pham TNM, Perumal N, Manicam C, Basoglu M, Eimer S, Fuhrmann DC, Pietrzik CU, Clement AM, Körschgen H, Schepers J, Behl C. Adaptive responses of neuronal cells to chronic endoplasmic reticulum (ER) stress. Redox Biol 2023; 67:102943. [PMID: 37883843 PMCID: PMC10618786 DOI: 10.1016/j.redox.2023.102943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/16/2023] [Accepted: 10/19/2023] [Indexed: 10/28/2023] Open
Abstract
Accumulation of misfolded proteins or perturbation of calcium homeostasis leads to endoplasmic reticulum (ER) stress and is linked to the pathogenesis of neurodegenerative diseases. Hence, understanding the ability of neuronal cells to cope with chronic ER stress is of fundamental interest. Interestingly, several brain areas uphold functions that enable them to resist challenges associated with neurodegeneration. Here, we established novel clonal mouse hippocampal (HT22) cell lines that are resistant to prolonged (chronic) ER stress induced by thapsigargin (TgR) or tunicamycin (TmR) as in vitro models to study the adaption to ER stress. Morphologically, we observed a significant increase in vesicular und autophagosomal structures in both resistant lines and 'giant lysosomes', especially striking in TgR cells. While autophagic activity increased under ER stress, lysosomal function appeared slightly impaired; in both cell lines, we observed enhanced ER-phagy. However, proteomic analyses revealed that various protein clusters and signaling pathways were differentially regulated in TgR versus TmR cells in response to chronic ER stress. Additionally, bioenergetic analyses in both resistant cell lines showed a shift toward aerobic glycolysis ('Warburg effect') and a defective complex I of the oxidative phosphorylation (OXPHOS) machinery. Furthermore, ER stress-resistant cells differentially activated the unfolded protein response (UPR) comprising IRE1α and ATF6 pathways. These findings display the wide portfolio of adaptive responses of neuronal cells to chronic ER stress. ER stress-resistant neuronal cells could be the basis to uncover molecular modulators of adaptation, resistance, and neuroprotection as potential pharmacological targets for preventing neurodegeneration.
Collapse
Affiliation(s)
- Thu Nguyen Minh Pham
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Natarajan Perumal
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Caroline Manicam
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Marion Basoglu
- Department of Structural Cell Biology, Institute for Cell Biology and Neuroscience, Goethe University, Frankfurt am Main, Germany
| | - Stefan Eimer
- Department of Structural Cell Biology, Institute for Cell Biology and Neuroscience, Goethe University, Frankfurt am Main, Germany
| | - Dominik C Fuhrmann
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Claus U Pietrzik
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Albrecht M Clement
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Hagen Körschgen
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Jana Schepers
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Christian Behl
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
24
|
Krebs N, Bock C, Tebben J, Mark FC, Lucassen M, Lannig G, Pörtner HO. Evolutionary Adaptation of Protein Turnover in White Muscle of Stenothermal Antarctic Fish: Elevated Cold Compensation at Reduced Thermal Responsiveness. Biomolecules 2023; 13:1507. [PMID: 37892189 PMCID: PMC10605280 DOI: 10.3390/biom13101507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/06/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
Protein turnover is highly energy consuming and overall relates to an organism's growth performance varying largely between species, e.g., due to pre-adaptation to environmental characteristics such as temperature. Here, we determined protein synthesis rates and capacity of protein degradation in white muscle of the cold stenothermal Antarctic eelpout (Pachycara brachycephalum) and its closely related temperate counterpart, the eurythermal common eelpout (Zoarces viviparus). Both species were exposed to acute warming (P. brachycephalum, 0 °C + 2 °C day-1; Z. viviparus, 4 °C + 3 °C day-1). The in vivo protein synthesis rate (Ks) was monitored after injection of 13C-phenylalanine, and protein degradation capacity was quantified by measuring the activity of cathepsin D in vitro. Untargeted metabolic profiling by nuclear magnetic resonance (NMR) spectroscopy was used to identify the metabolic processes involved. Independent of temperature, the protein synthesis rate was higher in P. brachycephalum (Ks = 0.38-0.614 % day-1) than in Z. viviparus (Ks= 0.148-0.379% day-1). Whereas protein synthesis remained unaffected by temperature in the Antarctic species, protein synthesis in Z. viviparus increased to near the thermal optimum (16 °C) and tended to fall at higher temperatures. Most strikingly, capacities for protein degradation were about ten times higher in the Antarctic compared to the temperate species. These differences are mirrored in the metabolic profiles, with significantly higher levels of complex and essential amino acids in the free cytosolic pool of the Antarctic congener. Together, the results clearly indicate a highly cold-compensated protein turnover in the Antarctic eelpout compared to its temperate confamilial. Constant versus variable environments are mirrored in rigid versus plastic functional responses of the protein synthesis machinery.
Collapse
Affiliation(s)
- Nina Krebs
- Department of Integrative Ecophysiology, Alfred Wegener Institute, Helmholtz Centre for Polar and Marine Research, Am Handelshafen 12, 27570 Bremerhaven, Germany; (C.B.); (F.C.M.); (M.L.); (G.L.)
| | - Christian Bock
- Department of Integrative Ecophysiology, Alfred Wegener Institute, Helmholtz Centre for Polar and Marine Research, Am Handelshafen 12, 27570 Bremerhaven, Germany; (C.B.); (F.C.M.); (M.L.); (G.L.)
| | - Jan Tebben
- Department of Ecological Chemistry, Alfred Wegener Institute, Helmholtz Centre for Polar and Marine Research, Am Handelshafen 12, 27570 Bremerhaven, Germany;
| | - Felix C. Mark
- Department of Integrative Ecophysiology, Alfred Wegener Institute, Helmholtz Centre for Polar and Marine Research, Am Handelshafen 12, 27570 Bremerhaven, Germany; (C.B.); (F.C.M.); (M.L.); (G.L.)
| | - Magnus Lucassen
- Department of Integrative Ecophysiology, Alfred Wegener Institute, Helmholtz Centre for Polar and Marine Research, Am Handelshafen 12, 27570 Bremerhaven, Germany; (C.B.); (F.C.M.); (M.L.); (G.L.)
| | - Gisela Lannig
- Department of Integrative Ecophysiology, Alfred Wegener Institute, Helmholtz Centre for Polar and Marine Research, Am Handelshafen 12, 27570 Bremerhaven, Germany; (C.B.); (F.C.M.); (M.L.); (G.L.)
| | - Hans-Otto Pörtner
- Department of Integrative Ecophysiology, Alfred Wegener Institute, Helmholtz Centre for Polar and Marine Research, Am Handelshafen 12, 27570 Bremerhaven, Germany; (C.B.); (F.C.M.); (M.L.); (G.L.)
| |
Collapse
|
25
|
Maurer J, Grouzmann E, Eugster PJ. Tutorial review for peptide assays: An ounce of pre-analytics is worth a pound of cure. J Chromatogr B Analyt Technol Biomed Life Sci 2023; 1229:123904. [PMID: 37832388 DOI: 10.1016/j.jchromb.2023.123904] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/04/2023] [Accepted: 10/05/2023] [Indexed: 10/15/2023]
Abstract
The recent increase in peptidomimetic-based medications and the growing interest in peptide hormones has brought new attention to the quantification of peptides for diagnostic purposes. Indeed, the circulating concentrations of peptide hormones in the blood provide a snapshot of the state of the body and could eventually lead to detecting a particular health condition. Although extremely useful, the quantification of such molecules, preferably by liquid chromatography coupled to mass spectrometry, might be quite tricky. First, peptides are subjected to hydrolysis, oxidation, and other post-translational modifications, and, most importantly, they are substrates of specific and nonspecific proteases in biological matrixes. All these events might continue after sampling, changing the peptide hormone concentrations. Second, because they include positively and negatively charged groups and hydrophilic and hydrophobic residues, they interact with their environment; these interactions might lead to a local change in the measured concentrations. A phenomenon such as nonspecific adsorption to lab glassware or materials has often a tremendous effect on the concentration and needs to be controlled with particular care. Finally, the circulating levels of peptides might be low (pico- or femtomolar range), increasing the impact of the aforementioned effects and inducing the need for highly sensitive instruments and well-optimized methods. Thus, despite the extreme diversity of these peptides and their matrixes, there is a common challenge for all the assays: the need to keep concentrations unchanged from sampling to analysis. While significant efforts are often placed on optimizing the analysis, few studies consider in depth the impact of pre-analytical steps on the results. By working through practical examples, this solution-oriented tutorial review addresses typical pre-analytical challenges encountered during the development of a peptide assay from the standpoint of a clinical laboratory. We provide tips and tricks to avoid pitfalls as well as strategies to guide all new developments. Our ultimate goal is to increase pre-analytical awareness to ensure that newly developed peptide assays produce robust and accurate results.
Collapse
Affiliation(s)
- Jonathan Maurer
- Service of Clinical Pharmacology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Eric Grouzmann
- Service of Clinical Pharmacology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Philippe J Eugster
- Service of Clinical Pharmacology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
26
|
Liu P, Karim MR, Covelo A, Yue Y, Lee MK, Lin W. The UPR Maintains Proteostasis and the Viability and Function of Hippocampal Neurons in Adult Mice. Int J Mol Sci 2023; 24:11542. [PMID: 37511300 PMCID: PMC10380539 DOI: 10.3390/ijms241411542] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/05/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
The unfolded protein response (UPR), which comprises three branches: PERK, ATF6α, and IRE1, is a major mechanism for maintaining cellular proteostasis. Many studies show that the UPR is a major player in regulating neuron viability and function in various neurodegenerative diseases; however, its role in neurodegeneration is highly controversial. Moreover, while evidence suggests activation of the UPR in neurons under normal conditions, deficiency of individual branches of the UPR has no major effect on brain neurons in animals. It remains unclear whether or how the UPR participates in regulating neuronal proteostasis under normal and disease conditions. To determine the physiological role of the UPR in neurons, we generated mice with double deletion of PERK and ATF6α in neurons. We found that inactivation of PERK and ATF6α in neurons caused lysosomal dysfunction (as evidenced by decreased expression of the V0a1 subunit of v-ATPase and decreased activation of cathepsin D), impairment of autophagic flux (as evidenced by increased ratio of LC3-II/LC3-I and increased p62 level), and accumulation of p-tau and Aβ42 in the hippocampus, and led to impairment of spatial memory, impairment of hippocampal LTP, and hippocampal degeneration in adult mice. These results suggest that the UPR is required for maintaining neuronal proteostasis (particularly tau and Aβ homeostasis) and the viability and function of neurons in the hippocampus of adult mice.
Collapse
Affiliation(s)
- Pingting Liu
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
- Institute for Translational Neuroscience, University of Minnesota, 2101 6th Street SE, WMBB4-140, Minneapolis, MN 55455, USA
| | - Md Razaul Karim
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
- Institute for Translational Neuroscience, University of Minnesota, 2101 6th Street SE, WMBB4-140, Minneapolis, MN 55455, USA
| | - Ana Covelo
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Yuan Yue
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
- Institute for Translational Neuroscience, University of Minnesota, 2101 6th Street SE, WMBB4-140, Minneapolis, MN 55455, USA
| | - Michael K Lee
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
- Institute for Translational Neuroscience, University of Minnesota, 2101 6th Street SE, WMBB4-140, Minneapolis, MN 55455, USA
| | - Wensheng Lin
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
- Institute for Translational Neuroscience, University of Minnesota, 2101 6th Street SE, WMBB4-140, Minneapolis, MN 55455, USA
| |
Collapse
|
27
|
Zhang KR, Jankowski CSR, Marshall R, Nair R, Más Gómez N, Alnemri A, Liu Y, Erler E, Ferrante J, Song Y, Bell BA, Baumann BH, Sterling J, Anderson B, Foshe S, Roof J, Fazelinia H, Spruce LA, Chuang JZ, Sung CH, Dhingra A, Boesze-Battaglia K, Chavali VRM, Rabinowitz JD, Mitchell CH, Dunaief JL. Oxidative stress induces lysosomal membrane permeabilization and ceramide accumulation in retinal pigment epithelial cells. Dis Model Mech 2023; 16:dmm050066. [PMID: 37401371 PMCID: PMC10399446 DOI: 10.1242/dmm.050066] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 06/23/2023] [Indexed: 07/05/2023] Open
Abstract
Oxidative stress has been implicated in the pathogenesis of age-related macular degeneration, the leading cause of blindness in older adults, with retinal pigment epithelium (RPE) cells playing a key role. To better understand the cytotoxic mechanisms underlying oxidative stress, we used cell culture and mouse models of iron overload, as iron can catalyze reactive oxygen species formation in the RPE. Iron-loading of cultured induced pluripotent stem cell-derived RPE cells increased lysosomal abundance, impaired proteolysis and reduced the activity of a subset of lysosomal enzymes, including lysosomal acid lipase (LIPA) and acid sphingomyelinase (SMPD1). In a liver-specific Hepc (Hamp) knockout murine model of systemic iron overload, RPE cells accumulated lipid peroxidation adducts and lysosomes, developed progressive hypertrophy and underwent cell death. Proteomic and lipidomic analyses revealed accumulation of lysosomal proteins, ceramide biosynthetic enzymes and ceramides. The proteolytic enzyme cathepsin D (CTSD) had impaired maturation. A large proportion of lysosomes were galectin-3 (Lgals3) positive, suggesting cytotoxic lysosomal membrane permeabilization. Collectively, these results demonstrate that iron overload induces lysosomal accumulation and impairs lysosomal function, likely due to iron-induced lipid peroxides that can inhibit lysosomal enzymes.
Collapse
Affiliation(s)
- Kevin R. Zhang
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Connor S. R. Jankowski
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Rayna Marshall
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rohini Nair
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Néstor Más Gómez
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ahab Alnemri
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yingrui Liu
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elizabeth Erler
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Julia Ferrante
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ying Song
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Brent A. Bell
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bailey H. Baumann
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jacob Sterling
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Brandon Anderson
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sierra Foshe
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jennifer Roof
- CHOP-PENN Proteomics Core Facility, The Children's Hospital of Philadelphia Research Institute, Philadelphia, PA 19104, USA
| | - Hossein Fazelinia
- CHOP-PENN Proteomics Core Facility, The Children's Hospital of Philadelphia Research Institute, Philadelphia, PA 19104, USA
| | - Lynn A. Spruce
- CHOP-PENN Proteomics Core Facility, The Children's Hospital of Philadelphia Research Institute, Philadelphia, PA 19104, USA
| | - Jen-Zen Chuang
- Department of Ophthalmology, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ching-Hwa Sung
- Department of Ophthalmology, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Anuradha Dhingra
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kathleen Boesze-Battaglia
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Venkata R. M. Chavali
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joshua D. Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Claire H. Mitchell
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joshua L. Dunaief
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
28
|
Li W, Pergande MR, Crutchfield CA, Searle BC, Backlund PS, Picache JA, Burkert K, Yanjanin-Farhat NM, Blank PS, Toth CL, Wassif CA, Porter FD, Cologna SM. A differential proteomics study of cerebrospinal fluid from individuals with Niemann-Pick disease, Type C1. Proteomics 2023; 23:e2200378. [PMID: 36638187 PMCID: PMC10918788 DOI: 10.1002/pmic.202200378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/18/2022] [Accepted: 01/03/2023] [Indexed: 01/15/2023]
Abstract
Niemann-Pick, type C1 (NPC1) is a fatal, neurodegenerative disease, which belongs to the family of lysosomal diseases. In NPC1, endo/lysosomal accumulation of unesterified cholesterol and sphingolipids arise from improper intracellular trafficking resulting in multi-organ dysfunction. With the proximity between the brain and cerebrospinal fluid (CSF), performing differential proteomics provides a means to shed light to changes occurring in the brain. In this study, CSF samples obtained from NPC1 individuals and unaffected controls were used for protein biomarker identification. A subset of these individuals with NPC1 are being treated with miglustat, a glycosphingolipid synthesis inhibitor. Of the 300 identified proteins, 71 proteins were altered in individuals with NPC1 compared to controls including cathepsin D, and members of the complement family. Included are a report of 10 potential markers for monitoring therapeutic treatment. We observed that pro-neuropeptide Y (NPY) was significantly increased in NPC1 individuals relative to healthy controls; however, individuals treated with miglustat displayed levels comparable to healthy controls. In further investigation, NPY levels in a NPC1 mouse model corroborated our findings. We posit that NPY could be a potential therapeutic target for NPC1 due to its multiple roles in the central nervous system such as attenuating neuroinflammation and reducing excitotoxicity.
Collapse
Affiliation(s)
- Wenping Li
- Department of Chemistry, University of Illinois Chicago
| | | | - Christopher A. Crutchfield
- Eunice Kennedy Shriver, National Institute of Child Health and Human Development, National Institutes of Health
| | - Brian C. Searle
- Department of Biomedical Informatics, The Ohio State University Medical Center
| | - Peter S. Backlund
- Eunice Kennedy Shriver, National Institute of Child Health and Human Development, National Institutes of Health
| | - Jaqueline A. Picache
- Eunice Kennedy Shriver, National Institute of Child Health and Human Development, National Institutes of Health
| | - Kathryn Burkert
- Eunice Kennedy Shriver, National Institute of Child Health and Human Development, National Institutes of Health
| | - Nicole M. Yanjanin-Farhat
- Eunice Kennedy Shriver, National Institute of Child Health and Human Development, National Institutes of Health
| | - Paul S. Blank
- Eunice Kennedy Shriver, National Institute of Child Health and Human Development, National Institutes of Health
| | - Cynthia L. Toth
- Eunice Kennedy Shriver, National Institute of Child Health and Human Development, National Institutes of Health
| | - Christopher A. Wassif
- Eunice Kennedy Shriver, National Institute of Child Health and Human Development, National Institutes of Health
| | - Forbes D. Porter
- Eunice Kennedy Shriver, National Institute of Child Health and Human Development, National Institutes of Health
| | | |
Collapse
|
29
|
Zhuang Q, Zhang Y, Zhu Y, He L, Kang C, Ke P, Lin H, Xiong Y, Huang X. Maintenance of cathepsin D-dependent autophagy-lysosomal function protects against cardiac ischemia/reperfusion injury. Biochem Biophys Res Commun 2023; 667:1-9. [PMID: 37201357 DOI: 10.1016/j.bbrc.2023.04.105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/20/2023] [Accepted: 04/27/2023] [Indexed: 05/20/2023]
Abstract
Cardiac ischemia/reperfusion(I/R) induced-cardiac vascular endothelial injury is an important pathological process that appears in the early stage of cardiac I/R injury. The autophagy-lysosomal pathway is essential for the maintenance of cellular homeostasis. However, in cardiac I/R injury, the role of the autophagy-lysosomal pathway is controversial. The present study aimed to use oxygen-glucose deprivation/oxygen-glucose resupply(OGD/OGR) in human coronary artery endothelial cells(HCAECs) with I/R injury to assess the role of the autophagy-lysosomal pathway in I/R-induced endothelial injury. The results revealed lysosomal dysfunction and impaired autophagic flux in endothelial cells exposed to OGD/OGR. Meanwhile, our data showed that the levels of cathepsin D(CTSD) decreased time-dependently. Knockdown of CTSD caused lysosomal dysfunction and impaired autophagic flux. Conversely, restoration of CTSD levels protected HCAECs against OGD/OGR induced-defects in autophagy-lysosomal function and cellular damage. Our findings indicated that I/R induced-impaired autophagic flux, rather than excessive autophagic initiation, mediates endothelial cells injury. The maintenance of autophagy-lysosomal function is critical to protect endothelial cells against I/R injury, and CTSD is a key regulator. Thus, strategies focused on restoring CTSD function are potentially novel treatments for cardiac reperfusion injury.
Collapse
Affiliation(s)
- Qizhen Zhuang
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Yang Zhang
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Yanting Zhu
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Lina He
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Chunmin Kang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou, 510120, China
| | - Peifeng Ke
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou, 510120, China
| | - Haibiao Lin
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou, 510120, China
| | - Yujuan Xiong
- Department of Laboratory Medicine, Panyu Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, 511400, China.
| | - Xianzhang Huang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou, 510120, China.
| |
Collapse
|
30
|
Ikari N, Arakawa H. Identification of a mitochondrial targeting sequence in cathepsin D and its localization in mitochondria. Biochem Biophys Res Commun 2023; 655:25-34. [PMID: 36921448 DOI: 10.1016/j.bbrc.2023.03.016] [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: 02/21/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/10/2023]
Abstract
Cathepsin D (CTSD) is a major lysosomal protease harboring an N-terminal signal peptide (amino acids 1-20) to enable vesicular transport from endoplasmic reticulum to lysosomes. Here, we report the possibility of a mitochondrial targeting sequence and mitochondrial localization of CTSD in cells. Live-cell imaging analysis with C-terminal enhanced green fluorescent protein-tagged CTSD (EGFP-CTSD) indicated that CTSD localizes to mitochondria. CTSD amino acids 21-35 are responsible for its mitochondrial localization, which exhibit typical features of mitochondrial targeting sequences, and are evolutionarily conserved. A proteinase K protection assay and sucrose gradient analysis showed that a small population of endogenous CTSD molecules exists in mitochondria. These results suggest that CTSD is a dual-targeted protein that may localize in both lysosomes and mitochondria.
Collapse
Affiliation(s)
- Naoki Ikari
- Division of Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, Japan
| | - Hirofumi Arakawa
- Division of Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, Japan.
| |
Collapse
|
31
|
Takano N, Hiramoto M, Yamada Y, Kokuba H, Tokuhisa M, Hino H, Miyazawa K. Azithromycin, a potent autophagy inhibitor for cancer therapy, perturbs cytoskeletal protein dynamics. Br J Cancer 2023; 128:1838-1849. [PMID: 36871041 PMCID: PMC10147625 DOI: 10.1038/s41416-023-02210-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 02/07/2023] [Accepted: 02/15/2023] [Indexed: 03/06/2023] Open
Abstract
BACKGROUND Autophagy plays an important role in tumour cell growth and survival and also promotes resistance to chemotherapy. Hence, autophagy has been targeted for cancer therapy. We previously reported that macrolide antibiotics including azithromycin (AZM) inhibit autophagy in various types of cancer cells in vitro. However, the underlying molecular mechanism for autophagy inhibition remains unclear. Here, we aimed to identify the molecular target of AZM for inhibiting autophagy. METHODS We identified the AZM-binding proteins using AZM-conjugated magnetic nanobeads for high-throughput affinity purification. Autophagy inhibitory mechanism of AZM was analysed by confocal microscopic and transmission electron microscopic observation. The anti-tumour effect with autophagy inhibition by oral AZM administration was assessed in the xenografted mice model. RESULTS We elucidated that keratin-18 (KRT18) and α/β-tubulin specifically bind to AZM. Treatment of the cells with AZM disrupts intracellular KRT18 dynamics, and KRT18 knockdown resulted in autophagy inhibition. Additionally, AZM treatment suppresses intracellular lysosomal trafficking along the microtubules for blocking autophagic flux. Oral AZM administration suppressed tumour growth while inhibiting autophagy in tumour tissue. CONCLUSIONS As drug-repurposing, our results indicate that AZM is a potent autophagy inhibitor for cancer treatment, which acts by directly interacting with cytoskeletal proteins and perturbing their dynamics.
Collapse
Affiliation(s)
- Naoharu Takano
- Department of Biochemistry, Tokyo Medical University, Tokyo, Japan.
| | - Masaki Hiramoto
- Department of Biochemistry, Tokyo Medical University, Tokyo, Japan
| | - Yumiko Yamada
- Department of Biochemistry, Tokyo Medical University, Tokyo, Japan
| | - Hiroko Kokuba
- Laboratory of Electron Microscopy, Tokyo Medical University, Tokyo, Japan
| | - Mayumi Tokuhisa
- Department of Biochemistry, Tokyo Medical University, Tokyo, Japan
| | - Hirotsugu Hino
- Department of Biochemistry, Tokyo Medical University, Tokyo, Japan
| | - Keisuke Miyazawa
- Department of Biochemistry, Tokyo Medical University, Tokyo, Japan.
| |
Collapse
|
32
|
Chen YH, Chen WY, Yu CL, Tsai CY, Hsieh SC. Gouty arthritis involves impairment of autophagic degradation via cathepsin D inactivation-mediated lysosomal dysfunction that promotes apoptosis in macrophages. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166703. [PMID: 37001704 DOI: 10.1016/j.bbadis.2023.166703] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 03/03/2023] [Accepted: 03/22/2023] [Indexed: 03/31/2023]
Abstract
This study examined autophagy-lysosome pathway (ALP) perturbations in synovial monocytes/macrophages from patients with gouty arthritis (GA) and the associations of ALP perturbations with cell death. Synovial fluid mononuclear cells (SFMCs) and synovial tissues (STs) from patients with GA, as well as monosodium urate (MSU) crystal-exposed macrophages, underwent immunoblotting, quantitative polymerase chain reaction, and immunofluorescence analyses of markers linked to the ALP (microtubule-associated protein 1 light chain 3B [LC3B], p62, cathepsin D [CTSD], and lysosome-associated membrane protein 2 [LAMP2]) and cell death (caspase-3). GA STs underwent immunohistochemistry and immunofluorescence analyses to determine the distributions of LC3B-positive autophagosomes and macrophages. GA SFMCs and STs exhibited impaired autophagic degradation, indicated by elevated levels of LC3B and p62, along with CTSD upregulation and caspase-3 activation. Macrophages from GA STs exhibited significant accumulation of LC3B-positive autophagosomes. The temporal effects of MSU crystals on the ALP and the associations of these effects with cell death were investigated using a macrophage model of GA. MSU crystal-exposed macrophages exhibited early (2 h) autophagosome formation but later (6-24 h) autophagic flux impairment, demonstrated by p62 accumulation, lysosomal inhibitor failure to increase LC3B accumulation, and LC3B colocalization with p62. These macrophages exhibited autophagic flux impairment because of CTSD inactivation-mediated lysosomal dysfunction, which caused immature CTSD to accumulate within damaged LAMP2-positive lysosomes. This accumulation coincided with caspase-3-dependent cell death (24 h) that was unaffected by CTSD inhibition. These findings indicate that GA involves MSU crystal-induced impairment of autophagic degradation via CTSD inactivation-mediated lysosomal dysfunction, which promotes apoptosis in macrophages.
Collapse
|
33
|
Aguilera AC, Leiva N, Alvarez PA, Pulcini G, Pereyra LL, Morales CR, Sosa MÁ, Carvelli L. Sortilin knock-down alters the expression and distribution of cathepsin D and prosaposin and up-regulates the cation-dependent mannose-6-phosphate receptor in rat epididymal cells. Sci Rep 2023; 13:3461. [PMID: 36859404 PMCID: PMC9977780 DOI: 10.1038/s41598-023-29157-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/31/2023] [Indexed: 03/03/2023] Open
Abstract
The selective transport to lysosomes can be mediated by either mannose-6-phosphate receptors (CD-MPR and CI-MPR) or sortilin. In mammalian epididymis, some lysosomal proteins are secreted into the lumen through unknown mechanisms. To investigate the underlying mechanisms of lysosomal protein transport in epididymal cells we studied the expression and distribution of cathepsin D (CatD) and prosaposin (PSAP) in a sortilin knocked down RCE-1 epididymal cell line (RCE-1 KD) in comparison with non-transfected RCE-1 cells. In RCE-1 cells, CatD was found in the perinuclear zone and co-localize with sortilin, whereas in RCE-1 KD cells, the expression, distribution and processing of the enzyme were altered. In turn, PSAP accumulated intracellularly upon sortilin knock-down and redistributed from LAMP-1-positive compartment to a perinuclear location, remaining co-localized with CatD. Interestingly, the sortilin knock-down induced CD-MPR overexpression and a redistribution of the receptor from the perinuclear zone to a dispersed cytoplasmic location, accompanied by an increased co-localization with CatD. The increase in CD-MPR could result from a compensatory response for the proper delivery of CatD to lysosomes in epididymal cells. The intracellular pathway taken by lysosomal proteins could be an approach for addressing further studies to understand the mechanism of exocytosis and therefore the role of these proteins in the epididymis.
Collapse
Affiliation(s)
- Andrea Carolina Aguilera
- CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, M5500, Mendoza, Argentina.,Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, M5500, Mendoza, Argentina
| | - Natalia Leiva
- CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, M5500, Mendoza, Argentina.,Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, M5500, Mendoza, Argentina
| | - Pablo Ariel Alvarez
- Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, M5500, Mendoza, Argentina
| | - Georgina Pulcini
- IHEM-CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, M5500, Mendoza, Argentina
| | - Laura Lucía Pereyra
- IHEM-CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, M5500, Mendoza, Argentina
| | | | - Miguel Ángel Sosa
- Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, M5500, Mendoza, Argentina.,IHEM-CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, M5500, Mendoza, Argentina
| | - Lorena Carvelli
- Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, M5500, Mendoza, Argentina. .,IHEM-CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, M5500, Mendoza, Argentina.
| |
Collapse
|
34
|
Cao M, Xue T, Huo H, Zhang X, Wang NN, Yan X, Li C. Spatial transcriptomes and microbiota reveal immune mechanism that respond to pathogen infection in the posterior intestine of Sebastes schlegelii. Open Biol 2023; 13:220302. [PMID: 36974664 PMCID: PMC9944294 DOI: 10.1098/rsob.220302] [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: 10/02/2022] [Accepted: 01/25/2023] [Indexed: 02/25/2023] Open
Abstract
The intestine is a site of immune cell priming at birth. Therefore, spatial transcriptomes were performed to define how the transcriptomic landscape was spatially organized in the posterior intestine of Sebastes schlegelii following Edwardsiella piscicida infection. In the healthy condition, we identified a previously unappreciated molecular regionalization of the posterior intestine. Following bacterial infection, most immune-related genes were identified in mucosa layer. Moreover, investigation of immune-related genes and genes in immune-related KEGG pathways based on spatial transcriptomes shed light on which sections of these genes are in the posterior intestine. Meanwhile, the high expression of genes related to regeneration also indicated that the posterior intestine was responding to the invasion of pathogens by constantly proliferating new cells. In addition, the increasing microbiota communities indicated that these bacteria maintained posterior intestine integrity and shaped the mucosal immune system. Taken together, spatial transcriptomes and microbiota compositions have significant implications for understanding the immune mechanism that responds to E. piscicida infection in the posterior intestine of S. schlegelii, which also provides a theoretical basis for the spatial distribution of immune genes and changes in bacterial flora in other teleosts in the process of resisting pathogens.
Collapse
Affiliation(s)
- Min Cao
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Ting Xue
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Huijun Huo
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Xiaoyan Zhang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Ning Ning Wang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Xu Yan
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Chao Li
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| |
Collapse
|
35
|
D'Acunzo P, Ungania JM, Kim Y, Barreto BR, DeRosa S, Pawlik M, Canals-Baker S, Erdjument-Bromage H, Hashim A, Goulbourne CN, Neubert TA, Saito M, Sershen H, Levy E. Cocaine perturbs mitovesicle biology in the brain. J Extracell Vesicles 2023; 12:e12301. [PMID: 36691887 PMCID: PMC9871795 DOI: 10.1002/jev2.12301] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 12/06/2022] [Accepted: 12/26/2022] [Indexed: 01/25/2023] Open
Abstract
Cocaine, an addictive psychostimulant, has a broad mechanism of action, including the induction of a wide range of alterations in brain metabolism and mitochondrial homeostasis. Our group recently identified a subpopulation of non-microvesicular, non-exosomal extracellular vesicles of mitochondrial origin (mitovesicles) and developed a method to isolate mitovesicles from brain parenchyma. We hypothesised that the generation and secretion of mitovesicles is affected by mitochondrial abnormalities induced by chronic cocaine exposure. Mitovesicles from the brain extracellular space of cocaine-administered mice were enlarged and more numerous when compared to controls, supporting a model in which mitovesicle biogenesis is enhanced in the presence of mitochondrial alterations. This interrelationship was confirmed in vitro. Moreover, cocaine affected mitovesicle protein composition, causing a functional alteration in mitovesicle ATP production capacity. These data suggest that mitovesicles are previously unidentified players in the biology of cocaine addiction and that target therapies to fine-tune brain mitovesicle functionality may be beneficial to mitigate the effects of chronic cocaine exposure.
Collapse
Affiliation(s)
- Pasquale D'Acunzo
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, New York, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York, New York, USA
| | - Jonathan M Ungania
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, New York, USA
| | - Yohan Kim
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, New York, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York, New York, USA
| | - Bryana R Barreto
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, New York, USA
| | - Steven DeRosa
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, New York, USA
| | - Monika Pawlik
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, New York, USA
| | - Stefanie Canals-Baker
- Division of Neurochemistry, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, New York, USA
| | - Hediye Erdjument-Bromage
- Department of Cell Biology, New York University Grossman School of Medicine, New York, New York, USA
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Grossman School of Medicine, New York, New York, USA
| | - Audrey Hashim
- Division of Neurochemistry, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, New York, USA
| | - Chris N Goulbourne
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, New York, USA
| | - Thomas A Neubert
- Department of Cell Biology, New York University Grossman School of Medicine, New York, New York, USA
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Grossman School of Medicine, New York, New York, USA
| | - Mariko Saito
- Department of Psychiatry, New York University Grossman School of Medicine, New York, New York, USA
- Division of Neurochemistry, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, New York, USA
| | - Henry Sershen
- Department of Psychiatry, New York University Grossman School of Medicine, New York, New York, USA
- Division of Neurochemistry, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, New York, USA
| | - Efrat Levy
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, New York, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York, New York, USA
- Department of Biochemistry & Molecular Pharmacology, New York University Grossman School of Medicine, New York, New York, USA
- NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
36
|
Pinto AT, Machado AB, Osório H, Pinto ML, Vitorino R, Justino G, Santa C, Castro F, Cruz T, Rodrigues C, Lima J, Sousa JLR, Cardoso AP, Figueira R, Monteiro A, Marques M, Manadas B, Pauwels J, Gevaert K, Mareel M, Rocha S, Duarte T, Oliveira MJ. Macrophage Resistance to Ionizing Radiation Exposure Is Accompanied by Decreased Cathepsin D and Increased Transferrin Receptor 1 Expression. Cancers (Basel) 2022; 15:270. [PMID: 36612268 PMCID: PMC9818572 DOI: 10.3390/cancers15010270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/06/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
PURPOSE To identify a molecular signature of macrophages exposed to clinically relevant ionizing radiation (IR) doses, mirroring radiotherapy sessions. METHODS Human monocyte-derived macrophages were exposed to 2 Gy/ fraction/ day for 5 days, mimicking one week of cancer patient's radiotherapy. Protein expression profile by proteomics was performed. RESULTS A gene ontology analysis revealed that radiation-induced protein changes are associated with metabolic alterations, which were further supported by a reduction of both cellular ATP levels and glucose uptake. Most of the radiation-induced deregulated targets exhibited a decreased expression, as was the case of cathepsin D, a lysosomal protease associated with cell death, which was validated by Western blot. We also found that irradiated macrophages exhibited an increased expression of the transferrin receptor 1 (TfR1), which is responsible for the uptake of transferrin-bound iron. TfR1 upregulation was also found in tumor-associated mouse macrophages upon tumor irradiation. In vitro irradiated macrophages also presented a trend for increased divalent metal transporter 1 (DMT1), which transports iron from the endosome to the cytosol, and a significant increase in iron release. CONCLUSIONS Irradiated macrophages present lower ATP levels and glucose uptake, and exhibit decreased cathepsin D expression, while increasing TfR1 expression and altering iron metabolism.
Collapse
Affiliation(s)
- Ana Teresa Pinto
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB–Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), Universidade de Aveiro, 3810-193 Aveiro, Portugal
| | - Ana Beatriz Machado
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB–Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- Champalimaud Centre for the Unknown, Fundação Champalimaud, 1400-038 Lisboa, Portugal
| | - Hugo Osório
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IPATIMUP–Instituto de Patologia e Imunologia Molecular da Universidade do Porto, 4200-135 Porto, Portugal
- Departament of Pathology, Faculdade de Medicina, Universidade do Porto, 4200-319 Porto, Portugal
| | - Marta Laranjeiro Pinto
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB–Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Rui Vitorino
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), Universidade de Aveiro, 3810-193 Aveiro, Portugal
| | - Gonçalo Justino
- Centro de Química Estrutural–Institute of Molecular Sciences, Instituto Superior Técnico, Universidade Técnica de Lisboa, 1049-001 Lisboa, Portugal
| | - Cátia Santa
- CNC–Center for Neuroscience and Cell Biology, Universidade de Coimbra, 3004-504 Coimbra, Portugal
- Institute for Interdisciplinary Research (III), Universidade de Coimbra, 3030-789 Coimbra, Portugal
| | - Flávia Castro
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB–Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Tânia Cruz
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB–Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Carla Rodrigues
- REQUIMTE–LAQV, Chemistry Department, NOVA School of Science and Technology, Universidade de Lisboa, 2829-516 Caparica, Portugal
| | - Jorge Lima
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IPATIMUP–Instituto de Patologia e Imunologia Molecular da Universidade do Porto, 4200-135 Porto, Portugal
| | - José Luís R. Sousa
- Personal Health Data Science Group, Sano-Centre for Computational Personalised Medicine, 30-054 Krakow, Poland
| | - Ana Patrícia Cardoso
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB–Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Rita Figueira
- Radiotherapy Service, Centro Hospitalar Universitário São João (CHUSJ), EPE, 4200-319 Porto, Portugal
| | - Armanda Monteiro
- Radiotherapy Service, Centro Hospitalar Universitário São João (CHUSJ), EPE, 4200-319 Porto, Portugal
| | - Margarida Marques
- Radiotherapy Service, Centro Hospitalar Universitário São João (CHUSJ), EPE, 4200-319 Porto, Portugal
| | - Bruno Manadas
- Institute for Interdisciplinary Research (III), Universidade de Coimbra, 3030-789 Coimbra, Portugal
| | - Jarne Pauwels
- VIB-UGent Center for Medical Biotechnology, 9052 Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, 9052 Ghent, Belgium
| | - Kris Gevaert
- VIB-UGent Center for Medical Biotechnology, 9052 Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, 9052 Ghent, Belgium
| | - Marc Mareel
- Department of Radiation Oncology and Experimental Cancer Research, Ghent University Hospital, 9000 Ghent, Belgium
| | - Sónia Rocha
- Institute of System, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3 GE, UK
| | - Tiago Duarte
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC–Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Maria José Oliveira
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB–Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- Departament of Pathology, Faculdade de Medicina, Universidade do Porto, 4200-319 Porto, Portugal
| |
Collapse
|
37
|
Lucchino V, Scaramuzzino L, Scalise S, Lo Conte M, Zannino C, Benedetto GL, Aguglia U, Ferlazzo E, Cuda G, Parrotta EI. Insights into the Genetic Profile of Two Siblings Affected by Unverricht-Lundborg Disease Using Patient-Derived hiPSCs. Cells 2022; 11:3491. [PMID: 36359887 PMCID: PMC9655992 DOI: 10.3390/cells11213491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 11/29/2023] Open
Abstract
Unverricht-Lundborg disease (ULD), also known as progressive myoclonic epilepsy 1 (EPM1), is a rare autosomal recessive neurodegenerative disorder characterized by a complex symptomatology that includes action- and stimulus-sensitive myoclonus and tonic-clonic seizures. The main cause of the onset and development of ULD is a repeat expansion of a dodecamer sequence localized in the promoter region of the gene encoding cystatin B (CSTB), an inhibitor of lysosomal proteases. Although this is the predominant mutation found in most patients, the physio-pathological mechanisms underlying the disease complexity remain largely unknown. In this work, we used patient-specific iPSCs and their neuronal derivatives to gain insight into the molecular and genetic machinery responsible for the disease in two Italian siblings affected by different phenotypes of ULD. Specifically, fragment length analysis on amplified CSTB promoters found homozygous status for dodecamer expansion in both patients and showed that the number of dodecamer repeats is the same in both. Furthermore, the luciferase reporter assay showed that the CSTB promoter activity was similarly reduced in both lines compared to the control. This information allowed us to draw important conclusions: (1) the phenotypic differences of the patients do not seem to be strictly dependent on the genetic mutation around the CSTB gene, and (2) that some other molecular mechanisms, not yet clearly identified, might be taken into account. In line with the inhibitory role of cystatin B on cathepsins, molecular investigations performed on iPSCs-derived neurons showed an increased expression of lysosomal cathepsins (B, D, and L) and a reduced expression of CSTB protein. Intriguingly, the increase in cathepsin expression does not appear to be correlated with the residual amount of CSTB, suggesting that other mechanisms, in addition to the regulation of cathepsins, could be involved in the pathological complexity of the disease.
Collapse
Affiliation(s)
- Valeria Lucchino
- Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy
| | - Luana Scaramuzzino
- Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy
| | - Stefania Scalise
- Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy
| | - Michela Lo Conte
- Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy
| | - Clara Zannino
- Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy
| | - Giorgia Lucia Benedetto
- Department of Medical and Surgical Sciences, University Magna Graecia, 88100 Catanzaro, Italy
| | - Umberto Aguglia
- Department of Medical and Surgical Sciences, University Magna Graecia, 88100 Catanzaro, Italy
| | - Edoardo Ferlazzo
- Department of Medical and Surgical Sciences, University Magna Graecia, 88100 Catanzaro, Italy
| | - Giovanni Cuda
- Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy
| | | |
Collapse
|
38
|
Sirikaew N, Pruksakorn D, Chaiyawat P, Chutipongtanate S. Mass Spectrometric-Based Proteomics for Biomarker Discovery in Osteosarcoma: Current Status and Future Direction. Int J Mol Sci 2022; 23:ijms23179741. [PMID: 36077137 PMCID: PMC9456544 DOI: 10.3390/ijms23179741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/23/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Due to a lack of novel therapies and biomarkers, the clinical outcomes of osteosarcoma patients have not significantly improved for decades. The advancement of mass spectrometry (MS), peptide quantification, and downstream pathway analysis enables the investigation of protein profiles across a wide range of input materials, from cell culture to long-term archived clinical specimens. This can provide insight into osteosarcoma biology and identify candidate biomarkers for diagnosis, prognosis, and stratification of chemotherapy response. In this review, we provide an overview of proteomics studies of osteosarcoma, indicate potential biomarkers that might be promising therapeutic targets, and discuss the challenges and opportunities of mass spectrometric-based proteomics in future osteosarcoma research.
Collapse
Affiliation(s)
- Nutnicha Sirikaew
- Musculoskeletal Science and Translational Research (MSTR) Center, Department of Orthopedics, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Dumnoensun Pruksakorn
- Musculoskeletal Science and Translational Research (MSTR) Center, Department of Orthopedics, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Parunya Chaiyawat
- Musculoskeletal Science and Translational Research (MSTR) Center, Department of Orthopedics, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Correspondence: (P.C.); (S.C.)
| | - Somchai Chutipongtanate
- Division of Epidemiology, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Correspondence: (P.C.); (S.C.)
| |
Collapse
|
39
|
Zhang Z, Nakata E, Shibano Y, Morii T. FRET-based cathepsin probes for simultaneous detection of cathepsin B and D activities. Chembiochem 2022; 23:e202200319. [PMID: 35929606 DOI: 10.1002/cbic.202200319] [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: 06/06/2022] [Revised: 08/05/2022] [Indexed: 11/09/2022]
Abstract
Fluorescent cathepsin probes were prepared by modification of peptidic substrates for cathepsin B (CTSB) and cathepsin D (CTSD) with FRET pairs. Fluorophores with distinguishable emission characteristics were applied to CTSB and CTSD probes with their appropriate quenchers to simultaneously monitor the activity of CTSB and/or CTSD. Conjugation of both the CTSB and CTSD probes with short single-stranded DNA drastically increased their reactivity to cathepsins over the parent probes possibly by improving their solubility. The activity of CTSB and CTSD were simultaneously detected by using these orthogonal FRET-based cathepsin probes.
Collapse
Affiliation(s)
- Zhengxiao Zhang
- Kyoto University: Kyoto Daigaku, Institute of Advanced Energy, Gokasyo, 611-0011, Uji, JAPAN
| | - Eiji Nakata
- Kyoto University: Kyoto Daigaku, Institute of Advanced Energy, Gokasyo, 611-0011, Uji, JAPAN
| | - Yuya Shibano
- Kyoto University - Uji Campus: Kyoto Daigaku - Uji Campus, Institute of Advanced energy, Gokasyo, 6110011, Uji, JAPAN
| | - Takashi Morii
- Kyoto University: Kyoto Daigaku, Institute of Advanced Energy, Gokasyo, 611-0011, Uji, JAPAN
| |
Collapse
|
40
|
Hoffman HK, Aguilar RS, Clark AR, Groves NS, Pezeshkian N, Bruns MM, van Engelenburg SB. Endocytosed HIV-1 Envelope Glycoprotein Traffics to Rab14 + Late Endosomes and Lysosomes to Regulate Surface Levels in T-Cell Lines. J Virol 2022; 96:e0076722. [PMID: 35770989 PMCID: PMC9327703 DOI: 10.1128/jvi.00767-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 06/06/2022] [Indexed: 11/20/2022] Open
Abstract
Production of infectious HIV-1 particles requires incorporation of the viral envelope glycoprotein (Env) at the plasma membrane (PM) of infected CD4+ T cells. Env trafficking to the PM exposes viral epitopes that can be exploited by the host immune system; however, HIV-1 can evade this response by endocytosis of excess Env from the PM. The fate of Env after internalization remains unclear, with evidence suggesting several different vesicular trafficking steps may be involved, including recycling pathways. To date, there have been very few studies documenting the trafficking pathways of native Env in infected T cells. Furthermore, it remains unclear whether there are T-cell-specific endosomal pathways regulating the fate of endocytic Env. Here, we use a pulse-labeling approach with a monovalent anti-Env Fab probe to characterize the trafficking of internalized Env within infected CD4+ T-cell lines, together with CRISPR/Cas9-mediated endogenous protein tagging, to assess the role of host cell Rab GTPases in Env trafficking. We show that endocytosed Env traffics to Rab14+ compartments that possess hallmarks of late endosomes and lysosomes. We also demonstrate that Env can recycle back to the PM, although we find that recycling does not occur at high rates when compared to the model recycling protein transferrin. These results help to resolve open questions about the fate and relevance of endocytosed Env in HIV-infected cells and suggest a novel role for Rab14 in a cell-type-specific late-endosomal/lysosomal trafficking pathway in T cells. IMPORTANCE HIV-1 envelope glycoprotein (Env) evades immune neutralization through many mechanisms. One immune evasion strategy may result from the internalization of excess surface-exposed Env to prevent antibody-dependent cellular cytotoxicity or neutralization. Characterization of the fate of endocytosed Env is critical to understand which vesicular pathways could be targeted to promote display of Env epitopes to the immune system. In this study, we characterize the endocytic fate of native Env, expressed from infected human T-cell lines. We demonstrate that Env is rapidly trafficked to a late-endosome/lysosome-like compartment and can be recycled to the cell surface for incorporation into virus assembly sites. This study implicates a novel intracellular compartment, marked by host-cell Rab14 GTPases, for the sequestration of Env. Therapeutic approaches aimed at mobilizing this intracellular pool of Env could lead to stronger immune control of HIV-1 infection via antibody-dependent cell-mediated cytotoxicity.
Collapse
Affiliation(s)
- Huxley K. Hoffman
- Molecular and Cellular Biophysics Program, Department of Biological Sciences, University of Denver, Denver, Colorado, USA
| | - Rebekah S. Aguilar
- Molecular and Cellular Biophysics Program, Department of Biological Sciences, University of Denver, Denver, Colorado, USA
| | - Austin R. Clark
- Molecular and Cellular Biophysics Program, Department of Biological Sciences, University of Denver, Denver, Colorado, USA
| | - Nicholas S. Groves
- Molecular and Cellular Biophysics Program, Department of Biological Sciences, University of Denver, Denver, Colorado, USA
| | - Nairi Pezeshkian
- Molecular and Cellular Biophysics Program, Department of Biological Sciences, University of Denver, Denver, Colorado, USA
| | - Merissa M. Bruns
- Molecular and Cellular Biophysics Program, Department of Biological Sciences, University of Denver, Denver, Colorado, USA
| | - Schuyler B. van Engelenburg
- Molecular and Cellular Biophysics Program, Department of Biological Sciences, University of Denver, Denver, Colorado, USA
| |
Collapse
|
41
|
Drobny A, Prieto Huarcaya S, Dobert J, Kluge A, Bunk J, Schlothauer T, Zunke F. The role of lysosomal cathepsins in neurodegeneration: Mechanistic insights, diagnostic potential and therapeutic approaches. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119243. [PMID: 35217144 DOI: 10.1016/j.bbamcr.2022.119243] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 12/12/2022]
Abstract
Lysosomes are ubiquitous organelles with a fundamental role in maintaining cellular homeostasis by mediating degradation and recycling processes. Cathepsins are the most abundant lysosomal hydrolyses and are responsible for the bulk degradation of various substrates. A correct autophagic function is essential for neuronal survival, as most neurons are post-mitotic and thus susceptible to accumulate cellular components. Increasing evidence suggests a crucial role of the lysosome in neurodegeneration as a key regulator of aggregation-prone and disease-associated proteins, such as α-synuclein, β-amyloid and huntingtin. Particularly, alterations in lysosomal cathepsins CTSD, CTSB and CTSL can contribute to the pathogenesis of neurodegenerative diseases as seen for neuronal ceroid lipofuscinosis, synucleinopathies (Parkinson's disease, Dementia with Lewy Body and Multiple System Atrophy) as well as Alzheimer's and Huntington's disease. In this review, we provide an overview of recent evidence implicating CTSD, CTSB and CTSL in neurodegeneration, with a special focus on the role of these enzymes in α-synuclein metabolism. In addition, we summarize the potential role of lysosomal cathepsins as clinical biomarkers in neurodegenerative diseases and discuss potential therapeutic approaches by targeting lysosomal function.
Collapse
Affiliation(s)
- Alice Drobny
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | | | - Jan Dobert
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Annika Kluge
- Department of Neurology, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Josina Bunk
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | | | - Friederike Zunke
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
42
|
Tanaka T, Warner BM, Michael DG, Nakamura H, Odani T, Yin H, Atsumi T, Noguchi M, Chiorini JA. LAMP3 inhibits autophagy and contributes to cell death by lysosomal membrane permeabilization. Autophagy 2022; 18:1629-1647. [PMID: 34802379 PMCID: PMC9298453 DOI: 10.1080/15548627.2021.1995150] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 10/06/2021] [Accepted: 10/14/2021] [Indexed: 01/18/2023] Open
Abstract
ABBREVIATIONS A253-control: A253 control for LAMP3 stable overexpression; A253- LAMP3: A253 LAPM3 stable overexpression; CASP1: caspase 1; CASP3: caspase 3; CHX: cycloheximide; CTSB: cathepsin B; CTSD: cathepsin D; CQ: chloroquine; DCs: dendritic cells; ER: endoplasmic reticulum; LGALS3: galectin 3; HCV: hepatitis C virus; HSG-control: HSG control for LAMP3 stable overexpression; HSG-LAMP3: HSG LAMP3 stable overexpression; HSP: heat shock protein; HTLV-1: human T-lymphocyte leukemia virus-1; IXA: ixazomib; LAMP: lysosomal associated membrane protein; MHC: major histocompatibility complex; mAb: monoclonal antibody; OE: overexpression; pepA: pepstatin A; pAb: polyclonal antibody; pSS: primary Sjögren syndrome; qRT-PCR: quantitative real- time reverse transcriptase polymerase chain reaction; SLE: systemic lupus erythematosus; SS: Sjögren syndrome; UPR: unfolded protein response; V-ATPase: vacuolar-type proton- translocating ATPase; Y-VAD: Ac-YVAD-cmk; Z-DEVD; Z-DEVD-fmk; Z-VAD: Z-VAD- fmk.
Collapse
Affiliation(s)
- Tsutomu Tanaka
- Adeno-Associated Virus Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Blake M. Warner
- Salivary Disorders Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Drew G. Michael
- Adeno-Associated Virus Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Hiroyuki Nakamura
- Adeno-Associated Virus Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Toshio Odani
- Adeno-Associated Virus Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Hongen Yin
- Adeno-Associated Virus Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Tatsuya Atsumi
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine Hokkaido University, Sapporo, Japan
| | - Masayuki Noguchi
- Division of Cancer Biology, Institute for Genetic Medicine Hokkaido University, Sapporo, Japan
| | - John A. Chiorini
- Adeno-Associated Virus Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
43
|
Skopál A, Kéki T, Tóth PÁ, Csóka B, Koscsó B, Németh ZH, Antonioli L, Ivessa A, Ciruela F, Virág L, Haskó G, Kókai E. Cathepsin D interacts with adenosine A 2A receptors in mouse macrophages to modulate cell surface localization and inflammatory signaling. J Biol Chem 2022; 298:101888. [PMID: 35367412 PMCID: PMC9065627 DOI: 10.1016/j.jbc.2022.101888] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 11/30/2022] Open
Abstract
Adenosine A2A receptor (A2AR)–dependent signaling in macrophages plays a key role in the regulation of inflammation. However, the processes regulating A2AR targeting to the cell surface and degradation in macrophages are incompletely understood. For example, the C-terminal domain of the A2AR and proteins interacting with it are known to regulate receptor recycling, although it is unclear what role potential A2AR-interacting partners have in macrophages. Here, we aimed to identify A2AR-interacting partners in macrophages that may effect receptor trafficking and activity. To this end, we performed a yeast two-hybrid screen using the C-terminal tail of A2AR as the “bait” and a macrophage expression library as the “prey.” We found that the lysosomal protease cathepsin D (CtsD) was a robust hit. The A2AR–CtsD interaction was validated in vitro and in cellular models, including RAW 264.7 and mouse peritoneal macrophage (IPMΦ) cells. We also demonstrated that the A2AR is a substrate of CtsD and that the blockade of CtsD activity increases the density and cell surface targeting of A2AR in macrophages. Conversely, we demonstrate that A2AR activation prompts the maturation and enzymatic activity of CtsD in macrophages. In summary, we conclude that CtsD is a novel A2AR-interacting partner and thus describe molecular and functional interplay that may be crucial for adenosine-mediated macrophage regulation in inflammatory processes.
Collapse
Affiliation(s)
- Adrienn Skopál
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Doctoral School of Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamás Kéki
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Doctoral School of Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Péter Á Tóth
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Balázs Csóka
- Department of Anesthesiology, Columbia University, New York, New York, USA
| | - Balázs Koscsó
- Department of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Zoltán H Németh
- Department of Anesthesiology, Columbia University, New York, New York, USA; Department of Surgery, Morristown Medical Center, Morristown, New Jersey, USA
| | - Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Andreas Ivessa
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain; Neuropharmacology and Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, L'Hospitalet de Llobregat, Barcelona, Spain
| | - László Virág
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; MTA-DE Cell Biology and Signaling Research Group, University of Debrecen, Debrecen, Hungary
| | - György Haskó
- Department of Anesthesiology, Columbia University, New York, New York, USA.
| | - Endre Kókai
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
44
|
Prieto Huarcaya S, Drobny A, Marques ARA, Di Spiezio A, Dobert JP, Balta D, Werner C, Rizo T, Gallwitz L, Bub S, Stojkovska I, Belur NR, Fogh J, Mazzulli JR, Xiang W, Fulzele A, Dejung M, Sauer M, Winner B, Rose-John S, Arnold P, Saftig P, Zunke F. Recombinant pro-CTSD (cathepsin D) enhances SNCA/α-Synuclein degradation in α-Synucleinopathy models. Autophagy 2022; 18:1127-1151. [PMID: 35287553 PMCID: PMC9196656 DOI: 10.1080/15548627.2022.2045534] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Parkinson disease (PD) is a neurodegenerative disorder characterized by the abnormal intracellular accumulation of SNCA/α-synuclein. While the exact mechanisms underlying SNCA pathology are not fully understood, increasing evidence suggests the involvement of autophagy as well as lysosomal deficiencies. Because CTSD (cathepsin D) has been proposed to be the major lysosomal protease involved in SNCA degradation, its deficiency has been linked to the presence of insoluble SNCA conformers in the brain of mice and humans as well as to the transcellular transmission of SNCA aggregates. We here postulate that SNCA degradation can be enhanced by the application of the recombinant human proform of CTSD (rHsCTSD). Our results reveal that rHsCTSD is efficiently endocytosed by neuronal cells, correctly targeted to lysosomes and matured to an enzymatically active protease. In dopaminergic neurons derived from induced pluripotent stem cells (iPSC) of PD patients harboring the A53T mutation within the SNCA gene, we confirm the reduction of insoluble SNCA after treatment with rHsCTSD. Moreover, we demonstrate a decrease of pathological SNCA conformers in the brain and within primary neurons of a ctsd-deficient mouse model after dosing with rHsCTSD. Boosting lysosomal CTSD activity not only enhanced SNCA clearance in human and murine neurons as well as tissue, but also restored endo-lysosome and autophagy function. Our findings indicate that CTSD is critical for SNCA clearance and function. Thus, enzyme replacement strategies utilizing CTSD may also be of therapeutic interest for the treatment of PD and other synucleinopathies aiming to decrease the SNCA burden.Abbreviations: aa: amino acid; SNCA/α-synuclein: synuclein alpha; APP: amyloid beta precursor protein; BBB: blood brain barrier; BF: basal forebrain; CBB: Coomassie Brilliant Blue; CLN: neuronal ceroid lipofuscinosis; CNL10: neuronal ceroid lipofuscinosis type 10; Corr.: corrected; CTSD: cathepsin D; CTSB: cathepsin B; DA: dopaminergic; DA-iPSn: induced pluripotent stem cell-derived dopaminergic neurons; dox: doxycycline; ERT: enzyme replacement therapy; Fx: fornix, GBA/β-glucocerebrosidase: glucosylceramidase beta; h: hour; HC: hippocampus; HT: hypothalamus; i.c.: intracranially; IF: immunofluorescence; iPSC: induced pluripotent stem cell; KO: knockout; LAMP1: lysosomal associated membrane protein 1; LSDs: lysosomal storage disorders; MAPT: microtubule associated protein tau; M6P: mannose-6-phosphate; M6PR: mannose-6-phosphate receptor; MB: midbrain; mCTSD: mature form of CTSD; neurofil.: neurofilament; PD: Parkinson disease; proCTSD: proform of CTSD; PRNP: prion protein; RFU: relative fluorescence units; rHsCTSD: recombinant human proCTSD; SAPC: Saposin C; SIM: structured illumination microscopy; T-insol: Triton-insoluble; T-sol: Triton-soluble; TEM: transmission electron microscopy, TH: tyrosine hydroxylase; Thal: thalamus.
Collapse
Affiliation(s)
| | - Alice Drobny
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (Fau), Erlangen, Germany
| | - André R A Marques
- iNOVA4Health, Chronic Diseases Research Center (CEDOC), Nova Medical School, Nms, Nova University Lisbon, Lisboa, Portugal
| | | | - Jan Philipp Dobert
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (Fau), Erlangen, Germany
| | - Denise Balta
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (Fau), Erlangen, Germany
| | - Christian Werner
- Department of Biotechnology and Biophysics, University of Würzburg, Biocenter, Am Hubland, Würzburg, Germany
| | - Tania Rizo
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Lisa Gallwitz
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Simon Bub
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (Fau), Erlangen, Germany
| | - Iva Stojkovska
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois USA
| | - Nandkishore R Belur
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois USA
| | | | - Joseph R Mazzulli
- Department of Biotechnology and Biophysics, University of Würzburg, Biocenter, Am Hubland, Würzburg, Germany
| | - Wei Xiang
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (Fau), Erlangen, Germany
| | - Amitkumar Fulzele
- Institute of Molecular Biology (IMB), Ackermannweg 4, Mainz, Germany
| | - Mario Dejung
- Institute of Molecular Biology (IMB), Ackermannweg 4, Mainz, Germany
| | - Markus Sauer
- Department of Biotechnology and Biophysics, University of Würzburg, Biocenter, Am Hubland, Würzburg, Germany
| | - Beate Winner
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Philipp Arnold
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Paul Saftig
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Friederike Zunke
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (Fau), Erlangen, Germany
| |
Collapse
|
45
|
Liu J, Bassal M, Schlichting S, Braren I, Di Spiezio A, Saftig P, Bartsch U. Intravitreal gene therapy restores the autophagy-lysosomal pathway and attenuates retinal degeneration in cathepsin D-deficient mice. Neurobiol Dis 2022; 164:105628. [PMID: 35033660 DOI: 10.1016/j.nbd.2022.105628] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/07/2022] [Accepted: 01/10/2022] [Indexed: 11/16/2022] Open
Abstract
Loss of vision due to progressive retinal degeneration is a hallmark of neuronal ceroid lipofuscinoses (NCL), a group of fatal neurodegenerative lysosomal storage diseases. Enzyme substitution therapies represent promising treatment options for NCLs caused by dysfunctions of soluble lysosomal enzymes. Here, we compared the efficacy of a cell-based enzyme substitution strategy and a gene therapy approach to attenuate the retinal pathology in cathepsin D- (CTSD) deficient mice, an animal model of CLN10 disease. Levels of enzymatically active CTSD in mutant retinas were significantly higher after an adeno-associated virus vector-mediated CTSD transfer to retinal glial cells and retinal pigment epithelial cells than after intravitreal transplantations of a CTSD overexpressing clonal neural stem cell line. In line with this finding, the gene therapy treatment restored the disrupted autophagy-lysosomal pathway more effectively than the cell-based approach, as indicated by a complete clearance of storage, significant attenuation of lysosomal hypertrophy, and normalized levels of the autophagy marker sequestosome 1/p62 and microtubule-associated protein 1 light chain 3-II. While the cell-based treatment did not prevent the rapidly progressing loss of various retinal cell types, the gene therapy approach markedly attenuated retinal degeneration as demonstrated by a pronounced rescue of photoreceptor cells and rod bipolar cells.
Collapse
Affiliation(s)
- Junling Liu
- Department of Ophthalmology, Experimental Ophthalmology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Mahmoud Bassal
- Department of Ophthalmology, Experimental Ophthalmology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Stefanie Schlichting
- Department of Ophthalmology, Experimental Ophthalmology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Ingke Braren
- Vector Facility, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | | | - Paul Saftig
- Institute of Biochemistry, Christian-Albrechts-University Kiel, 24118 Kiel, Germany
| | - Udo Bartsch
- Department of Ophthalmology, Experimental Ophthalmology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| |
Collapse
|
46
|
Hatamiya S, Miyara M, Kotake Y. Tributyltin inhibits autophagy by decreasing lysosomal acidity in SH-SY5Y cells. Biochem Biophys Res Commun 2022; 592:31-37. [PMID: 35016149 DOI: 10.1016/j.bbrc.2021.12.118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/13/2021] [Accepted: 12/30/2021] [Indexed: 12/29/2022]
Abstract
Tributyltin (TBT) is an environmental pollutant that remains in marine sediments and is toxic to mammals. For example, TBT elicits neurotoxic and immunosuppressive effects on rats. However, it is not entirely understood how TBT causes toxicity. Autophagy plays a pivotal role in protein quality control and eliminates aggregated proteins and damaged organelles. We previously reported that TBT dephosphorylates mammalian target of rapamycin (mTOR), which may be involved in enhancement of autophagosome synthesis, in primary cultures of cortical neurons. Autophagosomes can accumulate due to enhancement of autophagosome synthesis or inhibition of autophagic degradation, and we did not clarify whether TBT alters autophagic flux. Here, we investigated the mechanism by which TBT causes accumulation of autophagosomes in SH-SY5Y cells. TBT inhibited autophagy without affecting autophagosome-lysosome fusion before it caused cell death. TBT dramatically decreased the acidity of lysosomes without affecting lysosomal membrane integrity. TBT decreased the mature protein level of cathepsin B, and this may be related to the decrease in lysosomal acidity. These results suggest that TBT inhibits autophagic degradation by decreasing lysosomal acidity. Autophagy impairment may be involved in the mechanism underlying neuronal death and/or T-cell-dependent thymus atrophy induced by TBT.
Collapse
Affiliation(s)
- Shunichi Hatamiya
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima, 734-8553, Japan
| | - Masatsugu Miyara
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima, 734-8553, Japan.
| | - Yaichiro Kotake
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima, 734-8553, Japan.
| |
Collapse
|
47
|
Krohn P, Rega LR, Harvent M, Festa BP, Taranta A, Luciani A, Dewulf J, Cremonesi A, Camassei FD, Hanson JVM, Gerth-Kahlert C, Emma F, Berquez M, Devuyst O. OUP accepted manuscript. Hum Mol Genet 2022; 31:2262-2278. [PMID: 35137071 PMCID: PMC9262394 DOI: 10.1093/hmg/ddac033] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/21/2022] [Accepted: 02/02/2022] [Indexed: 11/14/2022] Open
Abstract
Recessive mutations in the CTNS gene encoding the lysosomal transporter cystinosin cause cystinosis, a lysosomal storage disease leading to kidney failure and multisystem manifestations. A Ctns knockout mouse model recapitulates features of cystinosis, but the delayed onset of kidney manifestations, phenotype variability and strain effects limit its use for mechanistic and drug development studies. To provide a better model for cystinosis, we generated a Ctns knockout rat model using CRISPR/Cas9 technology. The Ctns−/− rats display progressive cystine accumulation and crystal formation in multiple tissues including kidney, liver and thyroid. They show an early onset and progressive loss of urinary solutes, indicating generalized proximal tubule dysfunction, with development of typical swan-neck lesions, tubulointerstitial fibrosis and kidney failure, and decreased survival. The Ctns−/− rats also present crystals in the cornea, and bone and liver defects, as observed in patients. Mechanistically, the loss of cystinosin induces a phenotype switch associating abnormal proliferation and dedifferentiation, loss of apical receptors and transporters, and defective lysosomal activity and autophagy in the cells. Primary cultures of proximal tubule cells derived from the Ctns−/− rat kidneys confirmed the key changes caused by cystine overload, including reduced endocytic uptake, increased proliferation and defective lysosomal dynamics and autophagy. The novel Ctns−/− rat model and derived proximal tubule cell system provide invaluable tools to investigate the pathogenesis of cystinosis and to accelerate drug discovery.
Collapse
Affiliation(s)
- Patrick Krohn
- Institute of Physiology, University of Zurich, Zurich 8057, Switzerland
| | - Laura Rita Rega
- Renal Diseases Research Unit, Genetics and Rare Diseases Research Area, Bambino Gesù Children’s Hospital, IRCCS, Rome 00165, Italy
| | - Marianne Harvent
- Institute of Physiology, University of Zurich, Zurich 8057, Switzerland
| | | | - Anna Taranta
- Renal Diseases Research Unit, Genetics and Rare Diseases Research Area, Bambino Gesù Children’s Hospital, IRCCS, Rome 00165, Italy
| | | | - Joseph Dewulf
- Department of Laboratory Medicine, Cliniques universitaires Saint Luc, UCLouvain, Brussels 1200, Belgium
- Department of Biochemistry, de Duve Institute, UCLouvain, Brussels 1200, Belgium
| | - Alessio Cremonesi
- Division of Clinical Chemistry and Biochemistry, University Children’s Hospital Zurich, Zurich 8032, Switzerland
| | | | - James V M Hanson
- Department of Ophthalmology, University Hospital Zurich and University of Zurich, Zurich 8091, Switzerland
| | - Christina Gerth-Kahlert
- Department of Ophthalmology, University Hospital Zurich and University of Zurich, Zurich 8091, Switzerland
| | - Francesco Emma
- Renal Diseases Research Unit, Genetics and Rare Diseases Research Area, Bambino Gesù Children’s Hospital, IRCCS, Rome 00165, Italy
- Department of Pediatric Subspecialties, Division of Nephrology, Children’s Hospital Bambino Gesù, IRCCS, Rome 00165, Italy
| | - Marine Berquez
- To whom correspondence should be addressed at: University of Zurich, Mechanisms of Inherited Kidney Disorders Group, Winterthurerstrasse 190, Zurich 8057, Switzerland. Tel: +41 (0)44 635 51 07; (Marine Berquez); Tel: +41 (0)44 635 50 82; Fax: +41 (0)44 635 68 14; (Olivier Devuyst)
| | - Olivier Devuyst
- To whom correspondence should be addressed at: University of Zurich, Mechanisms of Inherited Kidney Disorders Group, Winterthurerstrasse 190, Zurich 8057, Switzerland. Tel: +41 (0)44 635 51 07; (Marine Berquez); Tel: +41 (0)44 635 50 82; Fax: +41 (0)44 635 68 14; (Olivier Devuyst)
| |
Collapse
|
48
|
Moraes JDN, Francisco AF, Dill LM, Diniz RS, Oliveira CSD, Silva TMRD, Caldeira CADS, Corrêa EDA, Coutinho-Neto A, Zanchi FB, Fontes MRDM, Soares AM, Calderon LDA. New multienzymatic complex formed between human cathepsin D and snake venom phospholipase A2. J Venom Anim Toxins Incl Trop Dis 2022; 28:e20220002. [DOI: 10.1590/1678-9199-jvatitd-2022-0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 08/16/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
| | - Aleff Ferreira Francisco
- São Paulo State University (UNESP), Brazil; Oswaldo Cruz Foundation (FIOCRUZ), Brazil; National Institute of Science and Technology of Epidemiology of the Western Amazon, Brazil; Smart Active Ingredients Lab (SAIL), Brazil
| | | | - Rafaela Souza Diniz
- Oswaldo Cruz Foundation (FIOCRUZ), Brazil; Federal University of Rondônia (UNIR), Brazil; Oswaldo Cruz Foundation (FIOCRUZ), Brazil; National Institute of Science and Technology of Epidemiology of the Western Amazon, Brazil
| | | | | | | | | | | | - Fernando Berton Zanchi
- Federal University of Rondônia (UNIR), Brazil; Oswaldo Cruz Foundation (FIOCRUZ), Brazil
| | | | - Andreimar Martins Soares
- Oswaldo Cruz Foundation (FIOCRUZ), Brazil; National Institute of Science and Technology of Epidemiology of the Western Amazon, Brazil; São Lucas University Center (UniSL), Brazil
| | - Leonardo de Azevedo Calderon
- Oswaldo Cruz Foundation (FIOCRUZ), Brazil; Federal University of Rondônia (UNIR), Brazil; Smart Active Ingredients Lab (SAIL), Brazil; Aparicio Carvalho University Center (FIMCA), Brazil
| |
Collapse
|
49
|
Lisowski C, Dias J, Costa S, Silva RJ, Mano M, Eulalio A. Dysregulated endolysosomal trafficking in cells arrested in the G 1 phase of the host cell cycle impairs Salmonella vacuolar replication. Autophagy 2021; 18:1785-1800. [PMID: 34781820 DOI: 10.1080/15548627.2021.1999561] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Modulation of the host cell cycle has emerged as a common theme among the pathways regulated by bacterial pathogens, arguably to promote host cell colonization. However, in most cases the exact benefit ensuing from such interference to the infection process remains unclear. Previously, we have shown that Salmonella actively induces G2/M arrest of host cells, and that infection is severely inhibited in cells arrested in G1. In this study, we demonstrate that Salmonella vacuolar replication is inhibited in host cells blocked in G1, whereas the cytosolic replication of the closely related pathogen Shigella is not affected. Mechanistically, we show that cells arrested in G1, but not cells arrested in G2, present dysregulated endolysosomal trafficking, displaying an abnormal accumulation of vesicles positive for late endosomal and lysosomal markers. In addition, the macroautophagic/autophagic flux and degradative lysosomal function are strongly impaired. This endolysosomal trafficking dysregulation results in sustained activation of the SPI-1 type III secretion system and lack of vacuole repair by the autophagy pathway, ultimately compromising the maturation and integrity of the Salmonella-containing vacuole. As such, Salmonella is released in the host cytosol. Collectively, our findings demonstrate that the modulation of the host cell cycle occurring during Salmonella infection is related to a disparity in the permissivity of cells arrested in G1 and G2/M, due to their intrinsic characteristics.
Collapse
Affiliation(s)
- Clivia Lisowski
- Host RNA Metabolism Group, Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany
| | - Jane Dias
- RNA & Infection Laboratory, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Functional Genomics and RNA-based Therapeutics Laboratory, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,PhD Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Susana Costa
- RNA & Infection Laboratory, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Functional Genomics and RNA-based Therapeutics Laboratory, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,PhD Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Ricardo Jorge Silva
- Functional Genomics and RNA-based Therapeutics Laboratory, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,PhD Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Miguel Mano
- Functional Genomics and RNA-based Therapeutics Laboratory, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Ana Eulalio
- Host RNA Metabolism Group, Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany.,RNA & Infection Laboratory, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
50
|
Loss of Christianson Syndrome Na +/H + Exchanger 6 (NHE6) Causes Abnormal Endosome Maturation and Trafficking Underlying Lysosome Dysfunction in Neurons. J Neurosci 2021; 41:9235-9256. [PMID: 34526390 PMCID: PMC8570832 DOI: 10.1523/jneurosci.1244-20.2021] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/05/2021] [Accepted: 08/07/2021] [Indexed: 01/08/2023] Open
Abstract
Loss-of-function mutations in endosomal Na+/H+ exchanger 6 (NHE6) cause the X-linked neurologic disorder Christianson syndrome. Patients exhibit symptoms associated with both neurodevelopmental and neurodegenerative abnormalities. While loss of NHE6 has been shown to overacidify the endosome lumen, and is associated with endolysosome neuropathology, NHE6-mediated mechanisms in endosome trafficking and lysosome function have been understudied. Here, we show that NHE6-null mouse neurons demonstrate worsening lysosome function with time in culture, likely as a result of defective endosome trafficking. NHE6-null neurons exhibit overall reduced lysosomal proteolysis despite overacidification of the endosome and lysosome lumen. Akin to Nhx1 mutants in Saccharomyces cerevisiae, we observe decreased endosome-lysosome fusion in NHE6-null neurons. Also, we find premature activation of pH-dependent cathepsin D (CatD) in endosomes. While active CatD is increased in endosomes, CatD activation and CatD protein levels are reduced in the lysosome. Protein levels of another mannose 6-phosphate receptor (M6PR)-dependent enzyme, β-N-acetylglucosaminidase, were also decreased in lysosomes of NHE6-null neurons. M6PRs accumulate in late endosomes, suggesting defective M6PR recycling and retromer function in NHE6-null neurons. Finally, coincident with decreased endosome-lysosome fusion, using total internal reflection fluorescence, we also find a prominent increase in fusion between endosomal multivesicular bodies and the plasma membrane, indicating enhanced exosome secretion from NHE6-null neurons. In summary, in addition to overacidification of endosomes and lysosomes, loss of NHE6 leads to defects in endosome maturation and trafficking, including enhanced exosome release, contributing to lysosome deficiency and potentially leading to neurodegenerative disease. SIGNIFICANCE STATEMENT Loss-of-function mutations in the endosomal Na+/H+ exchanger 6 (NHE6) cause Christianson syndrome, an X-linked neurologic disorder. Loss of NHE6 has been shown to overacidify endosomes; however, endosome trafficking mechanisms have been understudied, and the mechanisms leading to neurodegeneration are largely unknown. In NHE6-null mouse neurons in vitro, we find worsening lysosome function with days in culture. Notably, pH-dependent lysosome enzymes, such as cathepsin D, have reduced activity in lysosomes yet increased, precocious activity in endosomes in NHE6-null neurons. Further, endosomes show reduced fusion to lysosomes, and increased fusion to the plasma membrane with increased exosome release. This study identifies new mechanisms involving defective endosome maturation and trafficking that impair lysosome function in Christianson syndrome, likely contributing to neurodegeneration.
Collapse
|