1
|
Guo Z, He L, Wang W, Tian S, Lin R. FUT2-dependent fucosylation of LAMP1 promotes the apoptosis of colorectal cancer cells by regulating the autophagy-lysosomal pathway. Cancer Lett 2025; 619:217643. [PMID: 40112906 DOI: 10.1016/j.canlet.2025.217643] [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: 12/23/2024] [Revised: 03/06/2025] [Accepted: 03/15/2025] [Indexed: 03/22/2025]
Abstract
Fucosyltransferase 2 (FUT2) is an enzyme that adds fucose to proteins or lipids via α-1,2-fucosylation in the intestinal mucosa. While FUT2 deficiency is linked to increased susceptibility to inflammatory bowel disease (IBD), its role in colorectal cancer (CRC) is unclear, and the molecular mechanisms involved remain largely unknown. We established an azoxymethane (AOM) and dextran sulfate sodium (DSS) model to induce CRC. FUT2 expression was assessed in human CRC tissues, AOM/DSS-induced mouse models, and CRC cell lines using qRT-PCR, western blotting, and UEA-I staining. FUT2 knockout (FUT2△IEC) mice were treated with AOM/DSS, and FUT2-overexpressing CRC cells were created to evaluate the effects of FUT2 on apoptosis in both in vitro and in vivo settings through Western blot analyses and functional assays. N-glycoproteomics, UEA-I chromatography, and co-immunoprecipitation were utilized to identify regulatory mechanisms and target fucosylated proteins. FUT2 expression and α-1,2-fucosylation were significantly decreased in CRC. FUT2 deficiency worsened AOM/DSS-induced CRC and reduced tumor apoptosis, while FUT2 overexpression induced apoptosis and inhibited proliferation in CRC cells and xenografts. Mechanistically, FUT2 appears to suppress autophagy by impairing lysosomal function and directly targeting and fucosylating LAMP1, contributing to lysosomal dysfunction. Our study reveals a fucosylation-dependent antitumor mechanism of FUT2 in CRC, suggesting potential therapeutic strategies for CRC treatment.
Collapse
Affiliation(s)
- Zijun Guo
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lingnan He
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong New Area, Shanghai, China
| | - Weijun Wang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuxin Tian
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rong Lin
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
2
|
Su D, He J, Yuan W, Duan R, Du B, Shi Y, Liu L, Li X. Effect and potential mechanism of modified citrus pectin in 3D printing-based cartilage tissue engineering. Int J Biol Macromol 2025; 311:144022. [PMID: 40345290 DOI: 10.1016/j.ijbiomac.2025.144022] [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: 09/23/2024] [Revised: 04/20/2025] [Accepted: 05/05/2025] [Indexed: 05/11/2025]
Abstract
Modified citrus pectin (MCP) is widely used as a dietary supplement in the food and pharmaceutical industries with pleiotropic bioactivities. Particularly, MCP has effects on chondroprotection and phenotype maintenance of chondrocyte. Here, after confirming its chondroprotective effect in a partial-thickness articular cartilage (AC) defect, the distributions of MCP in cartilage and chondrocytes were investigated using fluorescence labeled-MCP. Then the potential of it in cartilage tissue engineering (CTE) was studied using 3D-printed scaffolds of hybrid hydrogel (GelMA/HAMA/MCP) of MCP, methacryloydylated-gelatin (GelMA) and hyaluronic acid (HAMA). Finally, the mechanism of the scaffolds on chondrogenesis were analyzed through transcriptome sequencing. It was found that MCP could penetrate cartilage, enter chondrocytes and accumulate in lysosome by binding with Gal-3. MCP could promote the proliferation and maintain the phenotype of chondrocytes in continuous passage culture. Mechanistically, MCP-based scaffold could upregulate the expression of genes of chondrogenic markers and growth factors, downregulate genes related to inflammation or degeneration mediators, and modulate autophagy pathways to maintain homeostasis of chondrocytes. Ultimately, MCP-based scaffolds could support chondrocytes adhesion, proliferation, ECM deposition, and enhance the chondrogenesis of the engineered cartilage. Together, our results demonstrate that MCP has great potential in three-dimensional bioprinting (3DBP)-based CTE to enhance the cartilage regeneration.
Collapse
Affiliation(s)
- Danning Su
- The Key Laboratory of Biomedical Material of Tianjin, State Key Laboratory of Advanced Medical Materials and Devices, Biomedical Barriers Research Center, Chinese Academy of Medical Sciences & Peking Union Medical College Institute of Biomedical Engineering, Tianjin 300192, PR China
| | - Jiayue He
- The Key Laboratory of Biomedical Material of Tianjin, State Key Laboratory of Advanced Medical Materials and Devices, Biomedical Barriers Research Center, Chinese Academy of Medical Sciences & Peking Union Medical College Institute of Biomedical Engineering, Tianjin 300192, PR China
| | - Wenlong Yuan
- The Key Laboratory of Biomedical Material of Tianjin, State Key Laboratory of Advanced Medical Materials and Devices, Biomedical Barriers Research Center, Chinese Academy of Medical Sciences & Peking Union Medical College Institute of Biomedical Engineering, Tianjin 300192, PR China
| | - Ruiping Duan
- The Key Laboratory of Biomedical Material of Tianjin, State Key Laboratory of Advanced Medical Materials and Devices, Biomedical Barriers Research Center, Chinese Academy of Medical Sciences & Peking Union Medical College Institute of Biomedical Engineering, Tianjin 300192, PR China
| | - Bo Du
- The Key Laboratory of Biomedical Material of Tianjin, State Key Laboratory of Advanced Medical Materials and Devices, Biomedical Barriers Research Center, Chinese Academy of Medical Sciences & Peking Union Medical College Institute of Biomedical Engineering, Tianjin 300192, PR China
| | - Yanping Shi
- School of Chemistry and Chemical Engineering, Tianjin University of Technology, Tianjin, 300384, PR China
| | - Lingrong Liu
- The Key Laboratory of Biomedical Material of Tianjin, State Key Laboratory of Advanced Medical Materials and Devices, Biomedical Barriers Research Center, Chinese Academy of Medical Sciences & Peking Union Medical College Institute of Biomedical Engineering, Tianjin 300192, PR China
| | - Xuemin Li
- The Key Laboratory of Biomedical Material of Tianjin, State Key Laboratory of Advanced Medical Materials and Devices, Biomedical Barriers Research Center, Chinese Academy of Medical Sciences & Peking Union Medical College Institute of Biomedical Engineering, Tianjin 300192, PR China.
| |
Collapse
|
3
|
Jiang M, Bianchi F, van den Bogaart G. Protonophore activity of short-chain fatty acids induces their intracellular accumulation and acidification. FEBS Lett 2025. [PMID: 40325954 DOI: 10.1002/1873-3468.70064] [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: 01/21/2025] [Revised: 03/27/2025] [Accepted: 04/13/2025] [Indexed: 05/07/2025]
Abstract
Short-chain fatty acids (SCFAs), produced by dietary fiber fermentation in the colon, play essential roles in cellular metabolism, with butyrate notably modulating immune responses and epigenetic regulation. Their production contributes to an acidic colonic environment where protonated SCFAs permeate membranes, leading to intracellular acidification and SCFA accumulation. Using our method to measure intracellular pH, we investigated how extracellular pH influences butyrate-induced acidification and immunomodulatory effects in human macrophages. Our data show that butyrate accumulates and acidifies cells at acidic extracellular pH due to the permeability of its protonated form. While inflammatory cytokine production was mildly influenced by extracellular pH, butyrate-induced histone acetylation exhibited a pH dependence, underscoring the importance of considering extracellular pH when assessing the SCFA's functions.
Collapse
Affiliation(s)
- Muwei Jiang
- Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands
| | - Frans Bianchi
- Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands
| | - Geert van den Bogaart
- Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
4
|
Qiao Q, Yin W, Wu X, Wu S, Ruan Y, Xu N, Li J, Wu ZS, Liu X, Xu Z. Two-Color Single-Molecule Blinking Ratiometricity: A Functional Super-Resolution Imaging Approach for Resolving Lysosomal pH and Dynamics. Angew Chem Int Ed Engl 2025; 64:e202503916. [PMID: 40055999 DOI: 10.1002/anie.202503916] [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: 02/17/2025] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 03/25/2025]
Abstract
Fluorescence super-resolution microscopy has enabled nanoscale imaging of intracellular structures, but it remains challenging to simultaneously achieve structural imaging and quantitative functional characterization, such as pH measurement, within the same region. Here, we introduce two-color single-molecule blinking ratiometricity (2C-SMBR), a novel method that integrates structural and functional imaging with single-molecule precision. By loading lysosomes with two pH-dependent spontaneously blinking fluorophores of distinct colors, 2C-SMBR leverages single-molecule localization of either fluorophore to achieve nanoscale structural imaging of lysosomes, whereas the ratiometric analysis of blinking dynamics between the two fluorophores provides quantitative pH measurement at the single-lysosome level. This dual-color ratiometric approach at the single-molecule level enables precise quantification of lysosomal pH with exceptional spatiotemporal resolution. Using 2C-SMBR, we reveal that lysosomal pH is highly heterogeneous at the single-lysosome level, with distinct subpopulations exhibiting diverse pH values. Our measurements show a pH range of 4.0-6.0 within lysosomes, with perinuclear lysosomes averaging a pH of approximately 4.88, whereas peripheral lysosomes average around 5.64. Crucially, 2C-SMBR enables real-time correlation between lysosomal dynamics and pH changes, overcoming a key limitation of super-resolution imaging. This approach not only advances nanoscale organelle characterization but also provides mechanistic insights into lysosomal physiology and function.
Collapse
Affiliation(s)
- Qinglong Qiao
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Wenting Yin
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Xia Wu
- Fluorescence Research Group, Singapore University of Technology and Design, Somapah Road, Singapore, 487372, Singapore
| | - Shaowei Wu
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yiyan Ruan
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ning Xu
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Jin Li
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Zhong-Shuai Wu
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Xiaogang Liu
- Fluorescence Research Group, Singapore University of Technology and Design, Somapah Road, Singapore, 487372, Singapore
| | - Zhaochao Xu
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
5
|
P K, Wilson H, Silswal A, Mishra L, Bhattacharya D, Mishra M, Koner AL. Morpholine Anchored Fluorogenic Toolkit: Unveiled Disease Allied Protein Fibrillation in Lysosomal Compartment of Live-Cell and Drosophila Models. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2404008. [PMID: 39690871 DOI: 10.1002/smll.202404008] [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: 05/16/2024] [Revised: 11/30/2024] [Indexed: 12/19/2024]
Abstract
The aberrant accumulation of cytotoxic protein aggregates is a hallmark of various neurodegenerative and non-neurodegenerative ailments, necessitating the development of sensitive and selective tools for their detection. Herein, we report a series of morpholine-anchored fluorescent probes, denoted as SC-nmor (n = 2, 4, 6), designed for facile visualization of protein aggregates. These probes display notable changes in their photophysical properties upon binding with protein aggregates, owing to their high sensitivity to the fibrillar microenvironment. Specifically, the SC-4mor probe demonstrates strong selectivity for aggregated insulin proteins over native insulin, accompanied by a significant enhancement in fluorescence lifetime. Live-cell imaging reveals an exclusive localization at the lysosomal compartment. This feature enables the visualization of lysosomal accumulated protein fibrils induced with pepstatin A. Additionally, in vivo assessments on genetically mutated and dietary-modified Drosophila melanogaster, representing neurodegenerative and non-neurodegenerative disease models, demonstrate staining of protein aggregates. The enhanced emission from the eye lobes of Aβ-mutated and HSD brain samples, suggesting that SC-4mor can exhibit adequate retention in the brain with minimal biological toxicity. SC-4mor also shows its capability to cross the blood-brain barrier in mice model. Consequently, SC-4mor emerges as a promising marker for detecting and monitoring neurotoxic protein fibrillation in live cells and animal models, offering potential insights into the pathogenesis and progression of protein aggregation.
Collapse
Affiliation(s)
- Kavyashree P
- Bionanotechnology Laboratory, Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, Madhya Pradesh, 462066, India
| | - Harry Wilson
- Bionanotechnology Laboratory, Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, Madhya Pradesh, 462066, India
| | - Akshay Silswal
- Bionanotechnology Laboratory, Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, Madhya Pradesh, 462066, India
| | - Lopamudra Mishra
- Neural Developmental Biology Laboratory, Department of Life Science, National Institute of Technology (NIT), Rourkela, Odisha, 769008, India
| | - Debapriya Bhattacharya
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, Madhya Pradesh, 462066, India
| | - Monalisa Mishra
- Neural Developmental Biology Laboratory, Department of Life Science, National Institute of Technology (NIT), Rourkela, Odisha, 769008, India
| | - Apurba Lal Koner
- Bionanotechnology Laboratory, Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, Madhya Pradesh, 462066, India
| |
Collapse
|
6
|
Kakanj P, Bonse M, Kshirsagar A, Gökmen A, Gaedke F, Sen A, Mollá B, Vogelsang E, Schauss A, Wodarz A, Pla-Martín D. Retromer promotes the lysosomal turnover of mtDNA. SCIENCE ADVANCES 2025; 11:eadr6415. [PMID: 40184468 PMCID: PMC11970507 DOI: 10.1126/sciadv.adr6415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 02/28/2025] [Indexed: 04/06/2025]
Abstract
Mitochondrial DNA (mtDNA) is exposed to multiple insults produced by normal cellular function. Upon mtDNA replication stress, the mitochondrial genome transfers to endosomes for degradation. Using proximity biotinylation, we found that mtDNA stress leads to the rewiring of the mitochondrial proximity proteome, increasing mitochondria's association with lysosomal and vesicle-related proteins. Among these, the retromer complex, particularly VPS35, plays a pivotal role by extracting mitochondrial components. The retromer promotes the formation of mitochondrial-derived vesicles shuttled to lysosomes. The mtDNA, however, directly shuttles to a recycling organelle in a BAX-dependent manner. Moreover, using a Drosophila model carrying a long deletion on the mtDNA (ΔmtDNA), we found that ΔmtDNA activates a specific transcriptome profile to counteract mitochondrial damage. Here, Vps35 expression restores mtDNA homoplasmy and alleviates associated defects. Hence, we demonstrate the existence of a previously unknown quality control mechanism for the mitochondrial matrix and the essential role of lysosomes in mtDNA turnover to relieve mtDNA damage.
Collapse
Affiliation(s)
- Parisa Kakanj
- Institute of Genetics, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Mari Bonse
- Institute of Physiology, University Clinics and Faculty of Medicine, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Arya Kshirsagar
- Institute of Biochemistry and Molecular Biology, University Clinics and Faculty of Medicine, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Aylin Gökmen
- Institute of Physiology, University Clinics and Faculty of Medicine, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Felix Gaedke
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Ayesha Sen
- Institute of Biochemistry and Molecular Biology, University Clinics and Faculty of Medicine, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | | | - Elisabeth Vogelsang
- Department of Molecular Cell Biology, Institute I for Anatomy. University Clinics and Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Astrid Schauss
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Andreas Wodarz
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Department of Molecular Cell Biology, Institute I for Anatomy. University Clinics and Faculty of Medicine, University of Cologne, Cologne, Germany
| | - David Pla-Martín
- Institute of Physiology, University Clinics and Faculty of Medicine, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Institute of Biochemistry and Molecular Biology, University Clinics and Faculty of Medicine, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
7
|
Shen YZ, Yao YD, Li HL, Li Y, Hu YC. CTSO and HLA-DQA1 as biomarkers in sepsis-associated ARDS: insights from RNA sequencing and immune infiltration analysis. BMC Infect Dis 2025; 25:326. [PMID: 40055592 PMCID: PMC11887161 DOI: 10.1186/s12879-025-10726-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 02/27/2025] [Indexed: 05/13/2025] Open
Abstract
The onset of sepsis frequently coincides with acute respiratory distress syndrome (ARDS), which constitutes a significant contributor to severe acid-base disturbances in septic patients. In the pathogenesis of sepsis, it conducts a crucial role. lysosomal metabolic disorders and immune imbalance conduct a pivotal role. Despite extensive research into the alterations in immune status during sepsis, few studies have been reported to thoroughly examine the association between lysosomes and sepsis. As a result, this study is predominantly Intended to delve into the link between lysosome-related genes and alterations in the lysosome in the immune microenvironment from the standpoint of bioinformatics in sepsis. The Registration Number was ChiCTR1900021261. Registration Date is 2019/02/04. Method Sepsis data source: Sepsis data was collected from previous clinical data and sequencing results (Originated from BGI Shenzhen Co., Ltd.) and the GO database was utilized for data collection of lysosome-related genes. Differential expression genes (DEGs) were screened on clinical sequencing data by employing IDEP 0.93 software subsequent to quality control. Afterwards, enrichment analysis was conducted by adopting Gene Set Enrichment Analysis (GSEA) and Weighted Gene Co expression Network Analysis (WGCNA), followed by cross referencing of lysosomal genes to identify DEGs associated with lysosomes. GO and KEGG pathway analysis wereperformed subsequently. The genes obtained from PLSGs and WGCNA by Creating a PPI network entails the following steps: the points were intersected at first. Afterwards, CytoHubba and MCODE analysis were performed by utilizing cytoscape software. Next, the intersection was taken to confirm Hub gene sequences, and subsequently the central DEGs tightly associated with existing CTD scores. Notwithstanding the fact that the causes of sepsis are multifaceted, ARDS can often trigger the development of sepsis in numerous cases. Simultaneously, with an aim to predict transcription factor levels in the central nervous system, Cytoscape software was adopted DEGs and to find relevant target miRNAs in the miRWalk database, and a correlated regulatory network was established accordingly. The SEPSIS immune infiltration model was constructed by employing ImmuCellAI software. Afterwards, the association between DEGs and immune microenvironment abundance was constructed by adopting Spearman's method. Last but not least, it is worth noting that single-cell sequencing has been validated as a method to analyze hub gene expression in immune cells of sepsis patients, enabling the selection of key genes that are closely associated with predictive outcomes. Result When acute respiratory distress syndrome (ARDS) is present, the differentially expressed genes (DEGs) are implicated in lysosomal metabolism and the regulation of the immune microenvironment. Six hub DEGs were bound up with sepsis or was attributable to the examinations. On top of that, it was determined that the patients had acute respiratory distress syndrome. The associated immune analysis illustrated a remarkable augment in T cell infiltration in the immune microenvironment of sepsis, while the infiltration relative to DC was reduced at certain level. Positive correlations were found between the two by employing Spearman analysis between hub DEGs and the regulatory role of immune cells. Moreover, it was universally acknowledged that anti-inflammatory immune cells were responsible for the negative correlation. On the basis of single-cell sequencing, it has been determined that CTSO and HLA-DQA1 were expressed in immune cells in sepsis. Aside from that, the survival-death curve direction suggested that they could be utilized as core genes for predicting sepsis-related prognosis analysis. Conclusion An analysis of this study demonstrates the interaction between sepsis lysosome-related metabolism and changes by understanding the pathogenesis of immune cells in the microenvironment. On this basis, we can develop new clinical diagnostics and therapeutic approaches of sepsis and identifying drug targets. Nonetheless, ARDS and sepsis can differ simply by the difference in site of infection; as the etiology of numerous ARDS cases is quite complex, progression to sepsis can occur if infection exacerbates or other complications arise, meeting the diagnostic criteria of sepsis 3.0.
Collapse
Affiliation(s)
- Yu Zhou Shen
- Department of Emergency Medicine, The Affiliated Hospital of Southwest Medical University, Lu Zhou, People's Republic of China
| | - Yan Dong Yao
- Department of Emergency Medicine, The Affiliated Hospital of Southwest Medical University, Lu Zhou, People's Republic of China
| | - Hai Li Li
- Department of Emergency Medicine, The Affiliated Hospital of Southwest Medical University, Lu Zhou, People's Republic of China
| | - Yang Li
- Department of Emergency Medicine, The Affiliated Hospital of Southwest Medical University, Lu Zhou, People's Republic of China
| | - Ying Chun Hu
- Department of Emergency Medicine, The Affiliated Hospital of Southwest Medical University, Lu Zhou, People's Republic of China.
| |
Collapse
|
8
|
Xu Y, Wang Q, Wang J, Qian C, Wang Y, Lu S, Song L, He Z, Liu W, Wan W. The cGAS-STING pathway activates transcription factor TFEB to stimulate lysosome biogenesis and pathogen clearance. Immunity 2025; 58:309-325.e6. [PMID: 39689715 DOI: 10.1016/j.immuni.2024.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/27/2024] [Accepted: 11/14/2024] [Indexed: 12/19/2024]
Abstract
Induction of autophagy is an ancient function of the cyclic GMP-AMP (cGAMP) synthase (cGAS)-stimulator of interferon genes (STING) pathway through which autophagic cargoes are delivered to lysosomes for degradation. However, whether lysosome function is also modulated by the cGAS-STING pathway remains unknown. Here, we discovered that the cGAS-STING pathway upregulated lysosomal activity by stimulating lysosome biogenesis independently of the downstream protein kinase TANK-binding kinase 1 (TBK1). STING activation enhanced lysosome biogenesis through inducing the nuclear translocation of transcription factor EB (TFEB) as well as its paralogs transcription factor E3 (TFE3) and microphthalmia-associated transcription factor (MITF). STING-induced lipidation of GABA type A receptor-associated protein (GABARAP), an autophagy-related protein, on STING vesicles was responsible for TFEB activation. Membrane-bound GABARAP sequestered the GTPase-activating protein folliculin (FLCN) and FLCN-interacting protein (FNIP) complex to block its function toward the Rag GTPases Ras-related GTP-binding C and D (RagC and RagD), abolishing mechanistic target of rapamycin (mTOR) complex 1 (mTORC1)-dependent phosphorylation and inactivation of TFEB. Functionally, STING-induced lysosome biogenesis within cells facilitated the clearance of cytoplasmic DNA and invading pathogens. Thus, our findings reveal that induction of lysosome biogenesis is another important function of the cGAS-STING pathway.
Collapse
Affiliation(s)
- Yinfeng Xu
- Laboratory of Basic Biology, Hunan First Normal University, Changsha 410205, Hunan, China.
| | - Qian Wang
- Department of Thoracic Surgery of Sir Run Run Shaw Hospital, and Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Jun Wang
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Chuying Qian
- Department of Thoracic Surgery of Sir Run Run Shaw Hospital, and Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Yusha Wang
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Sheng Lu
- Department of Thoracic Surgery of Sir Run Run Shaw Hospital, and Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Lijiang Song
- Department of Thoracic Surgery of Sir Run Run Shaw Hospital, and Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Zhengfu He
- Department of Thoracic Surgery of Sir Run Run Shaw Hospital, and Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Wei Liu
- Department of Metabolic Medicine, International Institutes of Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, Zhejiang, China.
| | - Wei Wan
- Department of Thoracic Surgery of Sir Run Run Shaw Hospital, and Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China.
| |
Collapse
|
9
|
Conesa-Bakkali R, Morillo-Huesca M, Martínez-Fábregas J. Non-Canonical, Extralysosomal Activities of Lysosomal Peptidases in Physiological and Pathological Conditions: New Clinical Opportunities for Cancer Therapy. Cells 2025; 14:68. [PMID: 39851495 PMCID: PMC11763575 DOI: 10.3390/cells14020068] [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: 11/29/2024] [Revised: 12/20/2024] [Accepted: 12/31/2024] [Indexed: 01/26/2025] Open
Abstract
Lysosomes are subcellular compartments characterised by an acidic pH, containing an ample variety of acid hydrolases involved in the recycling of biopolymers. Among these hydrolases, lysosomal proteases have merely been considered as end-destination proteases responsible for the digestion of waste proteins, trafficked to the lysosomal compartment through autophagy and endocytosis. However, recent reports have started to unravel specific roles for these proteases in the regulation of initially unexpected biological processes, both under physiological and pathological conditions. Furthermore, some lysosomal proteases are no longer restricted to the lysosomal compartment, as more novel non-canonical, extralysosomal targets are being identified. Currently, lysosomal proteases are accepted to play key functions in the extracellular milieu, attached to the plasma membrane and even in the cytosolic and nuclear compartments of the cell. Under physiological conditions, lysosomal proteases, through non-canonical, extralysosomal activities, have been linked to cell differentiation, regulation of gene expression, and cell division. Under pathological conditions, these proteases have been linked to cancer, mostly through their extralysosomal activities in the cytosol and nuclei of cells. In this review, we aim to provide a comprehensive summary of our current knowledge about the extralysosomal, non-canonical functions of lysosomal proteases, both under physiological and pathological conditions, with a particular interest in cancer, that could potentially offer new opportunities for clinical intervention.
Collapse
Affiliation(s)
- Ryan Conesa-Bakkali
- Centro Andaluz de Biología Molecular y Medicina Regenerativa—CABIMER, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Pablo de Olavide, Américo Vespucio 24, 41092 Sevilla, Spain; (R.C.-B.); (M.M.-H.)
| | - Macarena Morillo-Huesca
- Centro Andaluz de Biología Molecular y Medicina Regenerativa—CABIMER, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Pablo de Olavide, Américo Vespucio 24, 41092 Sevilla, Spain; (R.C.-B.); (M.M.-H.)
| | - Jonathan Martínez-Fábregas
- Centro Andaluz de Biología Molecular y Medicina Regenerativa—CABIMER, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Pablo de Olavide, Américo Vespucio 24, 41092 Sevilla, Spain; (R.C.-B.); (M.M.-H.)
- Departamento de Bioquímica Vegetal y Biología Molecular, Facultad de Biología, Universidad de Sevilla, Avenida Reina Mercedes, 41012 Sevilla, Spain
| |
Collapse
|
10
|
Soni S, Kori SK, Nema P, Iyer AK, Soni V, Kashaw SK. Cell-penetrating Peptides as Keys to Endosomal Escape and Intracellular Trafficking in Nanomedicine Delivery. Curr Med Chem 2025; 32:1288-1312. [PMID: 38362688 DOI: 10.2174/0109298673278936240107121907] [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: 10/05/2023] [Revised: 12/18/2023] [Accepted: 12/27/2023] [Indexed: 02/17/2024]
Abstract
This review article discusses the challenges of delivering cargoes to the cytoplasm, for example, proteins, peptides, and nucleic acids, and the mechanisms involved in endosomal escape. Endocytosis, endosomal maturation, and exocytosis pose significant barriers to effective cytoplasmic delivery. The article explores various endosomal escape mechanisms, such as the proton sponge effect, osmotic lysis, membrane fusion, pore formation, membrane destabilization/ disruption, and vesicle budding and collapse. Additionally, it discusses the role of lysosomes, glycocalyx, and molecular crowding in the cytoplasmic delivery process. Despite the recent advances in nonviral delivery systems, there is still a need to improve cytoplasmic delivery. Strategies such as fusogenic peptides, endosomolytic polymers, and cell-penetrating peptides have shown promise in improving endosomal escape and cytoplasmic delivery. More research is needed to refine these strategies and make them safer and more effective. In conclusion, the article highlights the challenges associated with cytoplasmic delivery and the importance of understanding the mechanisms involved in endosomal escape. A better understanding of these processes could result in the creation of greater effectiveness and safe delivery systems for various cargoes, including proteins, peptides, and nucleic acids.
Collapse
Affiliation(s)
- Sakshi Soni
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| | - Shivam K Kori
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| | - Priyanshu Nema
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| | - Arun K Iyer
- Use-inspired Biomaterials and Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Wayne State University, Detroit, Michigan, USA
- Molecular Imaging Program, Karmanos Cancer Institute, Detroit, Michigan, USA
| | - Vandana Soni
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| | - Sushil K Kashaw
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| |
Collapse
|
11
|
Sun R, Li S, Ye W, Lu Y. Development of a prognostic model based on lysosome-related genes for ovarian cancer: insights into tumor microenvironment, mutation patterns, and personalized treatment strategies. Cancer Cell Int 2024; 24:419. [PMID: 39702158 DOI: 10.1186/s12935-024-03586-w] [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: 01/15/2024] [Accepted: 11/26/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Ovarian cancer (OC) is often associated with an unfavorable prognosis. Given the crucial involvement of lysosomes in tumor advancement, lysosome-related genes (LRGs) hold promise as potential therapeutic targets. METHODS To identify differentially expressed lysosome-related genes (DE-LRGs), we performed a matching analysis between differentially expressed genes (DEGs) in OC and the pool of LRGs. Genes with prognostic significance were analyzed using multiple regression analyses to construct a prognostic risk signature. The model's efficacy was validated through survival analysis in various cohorts. We further explored the model's correlation with clinical attributes, tumor microenvironment (TME), mutational patterns, and drug sensitivity. The quantitative real-time polymerase chain reaction (qRT-PCR) validated gene expression in OC cells. RESULTS A 10-gene prognostic risk signature was established. Survival analysis confirmed its predictive accuracy across cohorts. The signature served as an independent prognostic element for OC. The high-risk and low-risk groups demonstrated notable disparities in terms of immune infiltration patterns, mutational characteristics, and sensitivity to therapeutic agents. The qRT-PCR results corroborated and validated the findings obtained from the bioinformatic analyses. CONCLUSIONS We devised a 10-LRG prognostic model linked to TME, offering insights for tailored OC treatments.
Collapse
Affiliation(s)
- Ran Sun
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110022, China
| | - Siyi Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110022, China
| | - Wanlu Ye
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110022, China
| | - Yanming Lu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110022, China.
| |
Collapse
|
12
|
Hu L, Su L, Wang Z, Yang J, Wang Y, Wang J, Gu X, Wang H. Application of acid-activated near-infrared viscosity fluorescent probe targeting lysosomes in cancer visualization. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 323:124918. [PMID: 39096675 DOI: 10.1016/j.saa.2024.124918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/20/2024] [Accepted: 07/30/2024] [Indexed: 08/05/2024]
Abstract
The higher viscosity and lower pH in lysosomes of cancer cells highlight their potential as biomarkers for cancer. Therefore, the development of acid-activated viscosity fluorescent probes is significant for the early diagnosis and treatment of cancer. Based on this, we have designed and synthesized a near-infrared fluorescent probe based on the 2-(2-hydroxyphenyl)benzothiazole (HBT) group, namely HBTH, to monitor the viscosity changes within lysosomes. It has been demonstrated that HBTH was extremely sensitive to viscosity, with a strong linear relationship between fluorescence intensity and log(viscosity) within the range of (logη) = 0-3.06 (a correlation coefficient of 0.98), proving its capability for quantitative viscosity measurement. In particular, the most obvious fluorescence enhancement of HBTH was only efficiently triggered by the combined effect of low pH and high viscosity. Furthermore, HBTH can rapidly localize to lysosomes by wash-free procedure at a low concentration (100 nM) and achieve high-fidelity imaging within 20 s. It can also monitor the dynamic processes of lysosomes in cells, viscosity changes under drug stimuli, and lysosomal behavior during mitophagy. Importantly, HBTH is capable of identifying tumors in tumor-bearing nude mice through in vivo imaging. These features make HBTH a powerful tool for the early diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Lei Hu
- School of Pharmacy, Wannan Medical College, Wuhu 241002, China
| | - Liping Su
- Huaxi MR Research Centre (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610000, China
| | - Zhiyu Wang
- School of Pharmacy, Wannan Medical College, Wuhu 241002, China
| | - Jing Yang
- School of Pharmacy, Wannan Medical College, Wuhu 241002, China
| | - Yuqing Wang
- School of Pharmacy, Wannan Medical College, Wuhu 241002, China
| | - Jie Wang
- School of Pharmacy, Wannan Medical College, Wuhu 241002, China
| | - Xiaoxia Gu
- School of Pharmacy, Wannan Medical College, Wuhu 241002, China.
| | - Hui Wang
- School of Pharmacy, Wannan Medical College, Wuhu 241002, China.
| |
Collapse
|
13
|
Chavda J, Siwach A, Sabharwal S, Janaagal A, Bhatia D, Gupta I. BODIPYS Based Fluorescent Markers To Monitor Autophagic Lysosomes and Lipid Droplets in TNBC. ACS Med Chem Lett 2024; 15:2115-2120. [PMID: 39691523 PMCID: PMC11647722 DOI: 10.1021/acsmedchemlett.4c00275] [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: 06/12/2024] [Revised: 09/09/2024] [Accepted: 09/09/2024] [Indexed: 12/19/2024] Open
Abstract
Lysosomal enzymes and high accumulation of lipid droplets are associated with breast cancer. The lysosomes and lipid droplets were monitored by BODIPYs, acting as autophagy activators in cancer cells. BD-1 and BD-2 were synthesized and characterized by Mass, UV-visible, fluorescence, and NMR spectroscopies. In BODIPYs, the effect of carbazole groups was reflected by the large Stokes shifts (2143-1651 cm-1) and red fluorescence. BODIPYs generated ROS and induced autophagy in triple negative breast cancer cells (MDA-MB-231) under white light. Confocal experiments revealed that BD-1 and BD-2 preferentially colocalized in lysosomes and lipid droplets. Autophagic lysosomes and lipid droplets released Ca2+ ions in the cytoplasm, which was evident with blue fluorescence of Fura-2M dye. In combination with an autophagy inhibitor, BD-1 displayed excellent photocytotoxicity (5.57 μM) on triple negative breast cancer cells under white light. This work demonstrates the potential of BODIPYs as theranostic agents for the photodynamic therapy against TNBC.
Collapse
Affiliation(s)
- Jaydeepsinh Chavda
- Department
of Chemistry, IIT Gandhinagar, Palaj, Gandhinagar, Gujarat 382055, India
| | - Arjun Siwach
- Department
of Chemistry, IIT Gandhinagar, Palaj, Gandhinagar, Gujarat 382055, India
| | - Sudhir Sabharwal
- Department
of Chemistry, IIT Gandhinagar, Palaj, Gandhinagar, Gujarat 382055, India
| | - Anu Janaagal
- Department
of Chemistry, IIT Gandhinagar, Palaj, Gandhinagar, Gujarat 382055, India
| | - Dhiraj Bhatia
- Department
of Biological Engineering, IIT Gandhinagar,
Palaj, Gandhinagar, Gujarat 382055, India
| | - Iti Gupta
- Department
of Chemistry, IIT Gandhinagar, Palaj, Gandhinagar, Gujarat 382055, India
| |
Collapse
|
14
|
Nam HY, Park SH, Lee GH, Kim EY, Lee S, Chang HW, Chang EJ, Choi KC, Kim SW. TIGAR coordinates senescence-associated secretory phenotype via lysosome repositioning and α-tubulin deacetylation. Exp Mol Med 2024; 56:2726-2738. [PMID: 39633033 DOI: 10.1038/s12276-024-01362-4] [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: 06/18/2024] [Revised: 09/06/2024] [Accepted: 09/25/2024] [Indexed: 12/07/2024] Open
Abstract
TP53-induced glycolysis and apoptosis regulator (TIGAR) regulates redox homeostasis and provides the intermediates necessary for cell growth by reducing the glycolytic rate. During cellular senescence, cells undergo metabolic rewiring towards the glycolytic pathway, along with the development of the senescence-associated secretory phenotype (SASP), also known as the secretome. We observed that TIGAR expression increased during replicative senescence following the in vitro expansion of human mesenchymal stromal cells (MSCs) and that TIGAR knockout (KO) decreased SASP factors and triggered premature senescence with decelerated progression. Additionally, TIGAR KO impaired flexible lysosomal movement to the perinuclear region and decreased the autophagic flux of MSCs. Research on the mechanism of lysosomal movement revealed that, while native senescent MSCs presented low levels of Ac-α-tubulin (lysine 40) and increased sirtuin 2 (SIRT2) activity compared with those in growing cells, TIGAR KO-MSCs maintained Ac-α-tubulin levels and exhibited decreased SIRT2 activity despite being in a senescent state. The overexpression of SIRT2 reduced Ac-α-tubulin as a protein target of SIRT2 and induced the positioning of lysosomes at the perinuclear region, restoring the cytokine secretion of TIGAR KO-MSCs. Furthermore, TIGAR expression was positively correlated with SIRT2 activity, indicating that TIGAR affects SIRT2 activity partly by modulating the NAD+ level. Thus, our study demonstrated that TIGAR provides a foundation that translates the regulation of energy metabolism into lysosome positioning, affecting the secretome for senescence development. Considering the functional value of the cell-secretome in aging-related diseases, these findings suggest the feasibility of TIGAR for the regulation of secretory phenotypes.
Collapse
Affiliation(s)
- Hae Yun Nam
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea.
| | - Seung-Ho Park
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Geun-Hee Lee
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Eun-Young Kim
- Department Hematology and Medical Oncology, Whinship Cancer Institute of Emory University, Atlanta, GA, 30322, USA
| | - SangEun Lee
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Hyo Won Chang
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Eun-Ju Chang
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Kyung-Chul Choi
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea.
| | - Seong Who Kim
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea.
| |
Collapse
|
15
|
Alesi N, Asrani K, Lotan TL, Henske EP. The Spectrum of Renal "TFEopathies": Flipping the mTOR Switch in Renal Tumorigenesis. Physiology (Bethesda) 2024; 39:0. [PMID: 39012319 DOI: 10.1152/physiol.00026.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/11/2024] [Accepted: 07/11/2024] [Indexed: 07/17/2024] Open
Abstract
The mammalian target of Rapamycin complex 1 (mTORC1) is a serine/threonine kinase that couples nutrient and growth factor signaling to the cellular control of metabolism and plays a fundamental role in aberrant proliferation in cancer. mTORC1 has previously been considered an "on/off" switch, capable of phosphorylating the entire pool of its substrates when activated. However, recent studies have indicated that mTORC1 may be active toward its canonical substrates, eukaryotic translation initiation factor 4E-binding protein 1 (4EBP1) and S6 kinase (S6K), involved in mRNA translation and protein synthesis, and inactive toward TFEB and TFE3, transcription factors involved in the regulation of lysosome biogenesis, in several pathological contexts. Among these conditions are Birt-Hogg-Dubé syndrome (BHD) and, recently, tuberous sclerosis complex (TSC). Furthermore, increased TFEB and TFE3 nuclear localization in these syndromes, and in translocation renal cell carcinomas (tRCC), drives mTORC1 activity toward the canonical substrates, through the transcriptional activation of the Rag GTPases, thereby positioning TFEB and TFE3 upstream of mTORC1 activity toward 4EBP1 and S6K. The expanding importance of TFEB and TFE3 in the pathogenesis of these renal diseases warrants a novel clinical grouping that we term "TFEopathies." Currently, there are no therapeutic options directly targeting TFEB and TFE3, which represents a challenging and critically required avenue for cancer research.
Collapse
Affiliation(s)
- Nicola Alesi
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Kaushal Asrani
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Elizabeth P Henske
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
16
|
Fernandes SA, Angelidaki DD, Nüchel J, Pan J, Gollwitzer P, Elkis Y, Artoni F, Wilhelm S, Kovacevic-Sarmiento M, Demetriades C. Spatial and functional separation of mTORC1 signalling in response to different amino acid sources. Nat Cell Biol 2024; 26:1918-1933. [PMID: 39385049 PMCID: PMC11567901 DOI: 10.1038/s41556-024-01523-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 09/09/2024] [Indexed: 10/11/2024]
Abstract
Amino acid (AA) availability is a robust determinant of cell growth through controlling mechanistic/mammalian target of rapamycin complex 1 (mTORC1) activity. According to the predominant model in the field, AA sufficiency drives the recruitment and activation of mTORC1 on the lysosomal surface by the heterodimeric Rag GTPases, from where it coordinates the majority of cellular processes. Importantly, however, the teleonomy of the proposed lysosomal regulation of mTORC1 and where mTORC1 acts on its effector proteins remain enigmatic. Here, by using multiple pharmacological and genetic means to perturb the lysosomal AA-sensing and protein recycling machineries, we describe the spatial separation of mTORC1 regulation and downstream functions in mammalian cells, with lysosomal and non-lysosomal mTORC1 phosphorylating distinct substrates in response to different AA sources. Moreover, we reveal that a fraction of mTOR localizes at lysosomes owing to basal lysosomal proteolysis that locally supplies new AAs, even in cells grown in the presence of extracellular nutrients, whereas cytoplasmic mTORC1 is regulated by exogenous AAs. Overall, our study substantially expands our knowledge about the topology of mTORC1 regulation by AAs and hints at the existence of distinct, Rag- and lysosome-independent mechanisms that control its activity at other subcellular locations. Given the importance of mTORC1 signalling and AA sensing for human ageing and disease, our findings will probably pave the way towards the identification of function-specific mTORC1 regulators and thus highlight more effective targets for drug discovery against conditions with dysregulated mTORC1 activity in the future.
Collapse
Affiliation(s)
- Stephanie A Fernandes
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Cologne Graduate School of Ageing Research, Cologne, Germany
| | | | - Julian Nüchel
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Jiyoung Pan
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Cologne Graduate School of Ageing Research, Cologne, Germany
| | | | - Yoav Elkis
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Filippo Artoni
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Cologne Graduate School of Ageing Research, Cologne, Germany
| | - Sabine Wilhelm
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | | | - Constantinos Demetriades
- Max Planck Institute for Biology of Ageing, Cologne, Germany.
- Cologne Graduate School of Ageing Research, Cologne, Germany.
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany.
| |
Collapse
|
17
|
Zou GY, Bi F, Yu YL, Liu MX, Chen S. Tetrahedral DNA-Based Ternary Recognition Ratiometric Fluorescent Probes for Real-Time In Situ Resolving Lysosome Subpopulations in Living Cells via Cl -, Ca 2+, and pH. Anal Chem 2024; 96:16639-16648. [PMID: 39382097 DOI: 10.1021/acs.analchem.4c02723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
Lysosomes are multifunctional organelles vital for cellular homeostasis with distinct subpopulations characterized by varying levels of Cl-, Ca2+, and H+. In situ visualization of these parameters is crucial for lysosomal research, yet developing probes that can simultaneously detect multiple ions remains challenging. Herein, we developed a lysosome-targeting ternary recognition ratiometric fluorescent probe based on tetrahedral DNA nanostructures (TDNs) to analyze lysosome subpopulations by Cl-, Ca2+, and pH. The TDN probe is assembled from four single-stranded DNAs, each end-modified with responsive fluorophores (Pr-Cl for Cl-, Pr-Ca for Ca2+, and Pr-pH for pH) or a reference fluorophore (Cy5). The fluorophores are integrated at the vertices of the rigid TDN to minimize mutual interference, and their fixed stoichiometry establishes a robust ternary recognition ratiometric fluorescence sensor for in situ resolution of lysosome subpopulations in living cells. Accordingly, a rise in lysosome subpopulations 2/6 characterized by low [Cl-], medium/high [Ca2+], and high pH was observed in the Niemann-Pick disease model cells but seldom observed in the control group. Conversely, there was a marked decline in the fraction of subpopulations 1/4/5 characterized by high [Cl-], medium to low [Ca2+], and pH. These changes were substantially reversed upon treatment. The probe holds great promise for studying lysosome subpopulations and the diagnosis and treatment of related diseases.
Collapse
Affiliation(s)
- Guang-Yue Zou
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, P.O. Box 332, Shenyang 110819, China
| | - Fan Bi
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, P.O. Box 332, Shenyang 110819, China
| | - Yong-Liang Yu
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, P.O. Box 332, Shenyang 110819, China
| | - Meng-Xian Liu
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai 980-8577, Miyagi, Japan
| | - Shuai Chen
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, P.O. Box 332, Shenyang 110819, China
- Foshan Graduate School of Innovation, Northeastern University, Foshan City, Guangdong 528311, China
| |
Collapse
|
18
|
Elblová P, Lunova M, Henry SJ, Tu X, Calé A, Dejneka A, Havelková J, Petrenko Y, Jirsa M, Stephanopoulos N, Lunov O. Peptide-coated DNA nanostructures as a platform for control of lysosomal function in cells. CHEMICAL ENGINEERING JOURNAL (LAUSANNE, SWITZERLAND : 1996) 2024; 498:155633. [PMID: 39372137 PMCID: PMC11448966 DOI: 10.1016/j.cej.2024.155633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
DNA nanotechnology is a rapidly growing field that provides exciting tools for biomedical applications. Targeting lysosomal functions with nanomaterials, such as DNA nanostructures (DNs), represents a rational and systematic way to control cell functionality. Here we present a versatile DNA nanostructure-based platform that can modulate a number of cellular functions depending on the concentration and surface decoration of the nanostructure. Utilizing different peptides for surface functionalization of DNs, we were able to rationally modulate lysosomal activity, which in turn translated into the control of cellular function, ranging from changes in cell morphology to modulation of immune signaling and cell death. Low concentrations of decalysine peptide-coated DNs induced lysosomal acidification, altering the metabolic activity of susceptible cells. In contrast, DNs coated with an aurein-bearing peptide promoted lysosomal alkalization, triggering STING activation. High concentrations of decalysine peptide-coated DNs caused lysosomal swelling, loss of cell-cell contacts, and morphological changes without inducing cell death. Conversely, high concentrations of aurein-coated DNs led to lysosomal rupture and mitochondrial damage, resulting in significant cytotoxicity. Our study holds promise for the rational design of a new generation of versatile DNA-based nanoplatforms that can be used in various biomedical applications, like the development of combinatorial anti-cancer platforms, efficient systems for endolysosomal escape, and nanoplatforms modulating lysosomal pH.
Collapse
Affiliation(s)
- Petra Elblová
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague, 18221, Czech Republic
- Faculty of Mathematics and Physics, Charles University, Ke Karlovu 3, CZ-121 16 Prague 2, Czech Republic
| | - Mariia Lunova
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague, 18221, Czech Republic
- Institute for Clinical & Experimental Medicine (IKEM), Prague, 14021, Czech Republic
| | - Skylar J.W. Henry
- School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
- Biodesign Center for Molecular Design and Biomimetics, Arizona State University, Tempe, United States
| | - Xinyi Tu
- School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
- Biodesign Center for Molecular Design and Biomimetics, Arizona State University, Tempe, United States
| | - Alicia Calé
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague, 18221, Czech Republic
- Faculty of Mathematics and Physics, Charles University, Ke Karlovu 3, CZ-121 16 Prague 2, Czech Republic
| | - Alexandr Dejneka
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague, 18221, Czech Republic
| | - Jarmila Havelková
- Department of Neuroregeneration, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, 14220, Czech Republic
- Laboratory of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Prague, 14220, Czech Republic
| | - Yuriy Petrenko
- Department of Neuroregeneration, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, 14220, Czech Republic
| | - Milan Jirsa
- Institute for Clinical & Experimental Medicine (IKEM), Prague, 14021, Czech Republic
| | - Nicholas Stephanopoulos
- School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
- Biodesign Center for Molecular Design and Biomimetics, Arizona State University, Tempe, United States
| | - Oleg Lunov
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague, 18221, Czech Republic
| |
Collapse
|
19
|
Zhu C, Zhou H, Bao M, Tang S, Gu X, Han M, Li P, Jiang Q. Polystyrene microplastics induce molecular toxicity in Simocephalus vetulus: A transcriptome and intestinal microorganism analysis. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2024; 275:107046. [PMID: 39197247 DOI: 10.1016/j.aquatox.2024.107046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/01/2024]
Abstract
The global prevalence and accumulation of plastic waste is leading to pollution levels that cause significant damage to ecosystems and ecological security. Exposure to two concentrations (1 and 5 mg/L) of 500 nm polystyrene (PS)-nanoplastics (NPs) for 14 d was evaluated in Simocephalus vetulus using transcriptome and 16 s rRNA sequencing analyses. PS-NP exposure resulted in stress-induced antioxidant defense, disturbed energy metabolism, and affected the FoxO signaling pathway, causing neurotoxicity. The expression of Cyclin D1 (CCND), glucose-6-phosphatase (G6PC) and phosphoenolpyruvate carboxykinase (PCK) genes was decreased compared to the control, whereas the expression of caspase3 (CASP3), caspase7 (CASP7), Superoxide dismutase (SOD), Heat shock protein 70 (HSP70), MPV17, and Glutathione S-transferase (GST) genes was increased, thus, suggesting that NP ingestion triggered oxidative stress and disrupted energy metabolism.. PS-NPs were present in the digestive tract of S. vetulus after 14 days of exposure. In addition, the abundance of the Proteobacteria and opportunistic pathogens was elevated after PS-NPs exposure. The diversity and homeostasis of the S. vetulus gut microbiota were disrupted and the stability of intestinal barrier function was impaired. Multiomic analyses highlighted the molecular toxicity and microbial changes in S. vetulus after exposure to NPs, providing an overview of how plastic pollution affects freshwater organisms and ecosystems.
Collapse
Affiliation(s)
- Chenxi Zhu
- Freshwater Fisheries Research Institute of Jiangsu Province. 79 Chating East Street, Nanjing 210017, China; Geography, School of Humanities, Universiti Sains Malaysia, 11800 Minden, Penang, Malaysia
| | - Hui Zhou
- Herpetological Research Center, College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu 210023, PR China
| | - Mengyu Bao
- Herpetological Research Center, College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu 210023, PR China; Freshwater Fisheries Research Institute of Jiangsu Province. 79 Chating East Street, Nanjing 210017, China
| | - Shengkai Tang
- Freshwater Fisheries Research Institute of Jiangsu Province. 79 Chating East Street, Nanjing 210017, China
| | - Xiankun Gu
- Freshwater Fisheries Research Institute of Jiangsu Province. 79 Chating East Street, Nanjing 210017, China
| | - Mingming Han
- Freshwater Fisheries Research Institute of Jiangsu Province. 79 Chating East Street, Nanjing 210017, China
| | - Peng Li
- Herpetological Research Center, College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu 210023, PR China.
| | - Qichen Jiang
- Freshwater Fisheries Research Institute of Jiangsu Province. 79 Chating East Street, Nanjing 210017, China.
| |
Collapse
|
20
|
Joby P, Ramasamy R, Solomon RV, Wilson P. Molecular engineering of BODIPY-bridged fluorescent probes for lysosome imaging - a computational study. Phys Chem Chem Phys 2024; 26:22912-22930. [PMID: 39171363 DOI: 10.1039/d4cp02570a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Lysosome imaging plays an important role in diagnosing many diseases and understanding various intracellular processes. Recently, B0 was reported as a fluorescent probe capable of detecting lysosomal viscosity changes. BODIPY is fused into the molecule as a bridge between the acceptor and donor components of B0, yielding nine new B molecules. Computational design and analysis of their optoelectronic properties were conducted to evaluate their effectiveness as fluorescent probes for lysosome imaging, with a specific target of HSA inside lysosomes. Optimized geometries reveal excellent π electron delocalization, resulting in nearly planar molecular structures. Frontier molecular orbital analysis suggests intramolecular charge transfer, along with π-π* transitions, from donor to bridge. TD-DFT calculations were performed to study absorption properties in the solvent phase, with B3PW91 showing good agreement with experiments. Molecular docking studies indicate that B derivatives can bind with HSA, and molecular dynamics simulations confirm their HSA targeting ability. This investigation highlights the introduction of BODIPY as a bridge for developing new probes capable of producing NIR fluorescence for bio-imaging, aiding in disease diagnosis.
Collapse
Affiliation(s)
- Prince Joby
- Department of Chemistry, Madras Christian College (Autonomous) (Affiliated to the University of Madras), Chennai, Tamil Nadu 600 059, India.
| | - Rohith Ramasamy
- Department of Chemistry, Madras Christian College (Autonomous) (Affiliated to the University of Madras), Chennai, Tamil Nadu 600 059, India.
| | - Rajadurai Vijay Solomon
- Department of Chemistry, Madras Christian College (Autonomous) (Affiliated to the University of Madras), Chennai, Tamil Nadu 600 059, India.
| | - Paul Wilson
- Department of Chemistry, Madras Christian College (Autonomous) (Affiliated to the University of Madras), Chennai, Tamil Nadu 600 059, India.
| |
Collapse
|
21
|
Ferrari V, Tedesco B, Cozzi M, Chierichetti M, Casarotto E, Pramaggiore P, Cornaggia L, Mohamed A, Patelli G, Piccolella M, Cristofani R, Crippa V, Galbiati M, Poletti A, Rusmini P. Lysosome quality control in health and neurodegenerative diseases. Cell Mol Biol Lett 2024; 29:116. [PMID: 39237893 PMCID: PMC11378602 DOI: 10.1186/s11658-024-00633-2] [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: 04/12/2024] [Accepted: 08/13/2024] [Indexed: 09/07/2024] Open
Abstract
Lysosomes are acidic organelles involved in crucial intracellular functions, including the degradation of organelles and protein, membrane repair, phagocytosis, endocytosis, and nutrient sensing. Given these key roles of lysosomes, maintaining their homeostasis is essential for cell viability. Thus, to preserve lysosome integrity and functionality, cells have developed a complex intracellular system, called lysosome quality control (LQC). Several stressors may affect the integrity of lysosomes, causing Lysosomal membrane permeabilization (LMP), in which membrane rupture results in the leakage of luminal hydrolase enzymes into the cytosol. After sensing the damage, LQC either activates lysosome repair, or induces the degradation of the ruptured lysosomes through autophagy. In addition, LQC stimulates the de novo biogenesis of functional lysosomes and lysosome exocytosis. Alterations in LQC give rise to deleterious consequences for cellular homeostasis. Specifically, the persistence of impaired lysosomes or the malfunctioning of lysosomal processes leads to cellular toxicity and death, thereby contributing to the pathogenesis of different disorders, including neurodegenerative diseases (NDs). Recently, several pieces of evidence have underlined the importance of the role of lysosomes in NDs. In this review, we describe the elements of the LQC system, how they cooperate to maintain lysosome homeostasis, and their implication in the pathogenesis of different NDs.
Collapse
Affiliation(s)
- Veronica Ferrari
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Barbara Tedesco
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Marta Cozzi
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Marta Chierichetti
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Elena Casarotto
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Paola Pramaggiore
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Laura Cornaggia
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Ali Mohamed
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Guglielmo Patelli
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Margherita Piccolella
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Riccardo Cristofani
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Valeria Crippa
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Mariarita Galbiati
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Angelo Poletti
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy.
| | - Paola Rusmini
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| |
Collapse
|
22
|
Hou J, Feng Y, Yang Z, Ding Y, Cheng D, Shi Z, Li R, Xue L. Primary Sjögren's syndrome: new perspectives on salivary gland epithelial cells. Eur J Med Res 2024; 29:371. [PMID: 39014509 PMCID: PMC11253495 DOI: 10.1186/s40001-024-01967-5] [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: 08/13/2023] [Accepted: 07/09/2024] [Indexed: 07/18/2024] Open
Abstract
Primary Sjögren's syndrome (pSS) is a chronic autoimmune disease primarily affecting exocrine glands such as the salivary glands, leading to impaired secretion and sicca symptoms. As the mainstay of salivation, salivary gland epithelial cells (SGECs) have an important role in the pathology of pSS. Emerging evidence suggests that the interplay between immunological factors and SGECs may not be the initial trigger or the sole mechanism responsible for xerostomia in pSS, challenging conventional perceptions. To deepen our understanding, current research regarding SGECs in pSS was reviewed. Among the extensive aberrations in cellular architecture and function, this review highlighted certain alterations of SGECs that were identified to occur independently of or in absence of lymphocytic infiltration. In particular, some of these alterations may serve as upstream factors of immuno-inflammatory responses. These findings underscore the significance of introspecting the pathogenesis of pSS and developing interventions targeting SGECs in the early stages of the disease.
Collapse
Affiliation(s)
- Jiaqi Hou
- Rheumatology Department, Yueyang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Hongkou District, Shanghai, 200437, China
| | - Yiyi Feng
- Rheumatology Department, Yueyang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Hongkou District, Shanghai, 200437, China
| | - Zhixia Yang
- Rheumatology Department, Yueyang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Hongkou District, Shanghai, 200437, China
| | - Yimei Ding
- Rheumatology Department, Yueyang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Hongkou District, Shanghai, 200437, China
| | - Dandan Cheng
- Shanghai Skin Diseases Hospital, 200 Wuyi Road, Changning District, Shanghai, 200050, China
| | - Zhonghao Shi
- Rheumatology Department, Yueyang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Hongkou District, Shanghai, 200437, China
| | - Rouxin Li
- Rheumatology Department, Yueyang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Hongkou District, Shanghai, 200437, China
| | - Luan Xue
- Rheumatology Department, Yueyang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Hongkou District, Shanghai, 200437, China.
| |
Collapse
|
23
|
Do H, Meena NK, Raben N. Failure of Autophagy in Pompe Disease. Biomolecules 2024; 14:573. [PMID: 38785980 PMCID: PMC11118179 DOI: 10.3390/biom14050573] [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: 04/08/2024] [Revised: 05/10/2024] [Accepted: 05/11/2024] [Indexed: 05/25/2024] Open
Abstract
Autophagy is an evolutionarily conserved lysosome-dependent degradation of cytoplasmic constituents. The system operates as a critical cellular pro-survival mechanism in response to nutrient deprivation and a variety of stress conditions. On top of that, autophagy is involved in maintaining cellular homeostasis through selective elimination of worn-out or damaged proteins and organelles. The autophagic pathway is largely responsible for the delivery of cytosolic glycogen to the lysosome where it is degraded to glucose via acid α-glucosidase. Although the physiological role of lysosomal glycogenolysis is not fully understood, its significance is highlighted by the manifestations of Pompe disease, which is caused by a deficiency of this lysosomal enzyme. Pompe disease is a severe lysosomal glycogen storage disorder that affects skeletal and cardiac muscles most. In this review, we discuss the basics of autophagy and describe its involvement in the pathogenesis of muscle damage in Pompe disease. Finally, we outline how autophagic pathology in the diseased muscles can be used as a tool to fast track the efficacy of therapeutic interventions.
Collapse
Affiliation(s)
| | | | - Nina Raben
- M6P Therapeutics, 20 S. Sarah Street, St. Louis, MO 63108, USA; (H.D.); (N.K.M.)
| |
Collapse
|
24
|
Martins TS, Correia M, Pinheiro D, Lemos C, Mendes MV, Pereira C, Costa V. Sit4 Genetically Interacts with Vps27 to Regulate Mitochondrial Function and Lifespan in Saccharomyces cerevisiae. Cells 2024; 13:655. [PMID: 38667270 PMCID: PMC11049076 DOI: 10.3390/cells13080655] [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: 01/06/2024] [Revised: 03/27/2024] [Accepted: 04/06/2024] [Indexed: 04/28/2024] Open
Abstract
The Sit4 protein phosphatase plays a key role in orchestrating various cellular processes essential for maintaining cell viability during aging. We have previously shown that SIT4 deletion promotes vacuolar acidification, mitochondrial derepression, and oxidative stress resistance, increasing yeast chronological lifespan. In this study, we performed a proteomic analysis of isolated vacuoles and yeast genetic interaction analysis to unravel how Sit4 influences vacuolar and mitochondrial function. By employing high-resolution mass spectrometry, we show that sit4Δ vacuolar membranes were enriched in Vps27 and Hse1, two proteins that are part of the endosomal sorting complex required for transport-0. In addition, SIT4 exhibited a negative genetic interaction with VPS27, as sit4∆vps27∆ double mutants had a shortened lifespan compared to sit4∆ and vps27∆ single mutants. Our results also show that Vps27 did not increase sit4∆ lifespan by improving protein trafficking or vacuolar sorting pathways. However, Vps27 was critical for iron homeostasis and mitochondrial function in sit4∆ cells, as sit4∆vps27∆ double mutants exhibited high iron levels and impaired mitochondrial respiration. These findings show, for the first time, cross-talk between Sit4 and Vps27, providing new insights into the mechanisms governing chronological lifespan.
Collapse
Affiliation(s)
- Telma S. Martins
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (T.S.M.); (M.C.); (D.P.); (C.L.); (M.V.M.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Miguel Correia
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (T.S.M.); (M.C.); (D.P.); (C.L.); (M.V.M.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Denise Pinheiro
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (T.S.M.); (M.C.); (D.P.); (C.L.); (M.V.M.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Carolina Lemos
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (T.S.M.); (M.C.); (D.P.); (C.L.); (M.V.M.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Marta Vaz Mendes
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (T.S.M.); (M.C.); (D.P.); (C.L.); (M.V.M.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Clara Pereira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (T.S.M.); (M.C.); (D.P.); (C.L.); (M.V.M.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Vítor Costa
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (T.S.M.); (M.C.); (D.P.); (C.L.); (M.V.M.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| |
Collapse
|
25
|
Morse J, Wang D, Mei S, Whitham D, Hladun C, Darie CC, Sintim HO, Wang M, Leung K. Chloride Homeostasis Regulates cGAS-STING Signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.08.588475. [PMID: 38645072 PMCID: PMC11030317 DOI: 10.1101/2024.04.08.588475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The cGAS-STING signaling pathway has emerged as a key mediator of inflammation. However, the roles of chloride homeostasis on this pathway are unclear. Here, we uncovered a correlation between chloride homeostasis and cGAS-STING signaling. We found that dysregulation of chloride homeostasis attenuates cGAS-STING signaling in a lysosome-independent manner. Treating immune cells with chloride channel inhibitors attenuated 2'3'-cGAMP production by cGAS and also suppressed STING polymerization, leading to reduced cytokine production. We also demonstrate that non-selective chloride channel blockers can suppress the NPC1 deficiency-induced, hyper-activated STING signaling in skin fibroblasts derived from Niemann Pick disease type C (NPC) patients. Our findings reveal that chloride homeostasis majorly affects cGAS-STING pathway and suggest a provocative strategy to dampen STING-mediated inflammation via targeting chloride channels.
Collapse
Affiliation(s)
- Jared Morse
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| | - Danna Wang
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| | - Serena Mei
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| | - Danielle Whitham
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| | - Colby Hladun
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| | - Costel C. Darie
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| | - Herman O. Sintim
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Modi Wang
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| | - KaHo Leung
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| |
Collapse
|
26
|
Yang YL, Zeng WH, Peng Y, Zuo SY, Fu YQ, Xiao YM, Huang WL, Wen ZY, Hu W, Yang YY, Huang XF. Characterization of three lamp genes from largemouth bass ( Micropterus salmoides): molecular cloning, expression patterns, and their transcriptional levels in response to fast and refeeding strategy. Front Physiol 2024; 15:1386413. [PMID: 38645688 PMCID: PMC11026864 DOI: 10.3389/fphys.2024.1386413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 03/14/2024] [Indexed: 04/23/2024] Open
Abstract
Lysosomes-associated membrane proteins (LAMPs), a family of glycosylated proteins and major constituents of the lysosomal membranes, play a dominant role in various cellular processes, including phagocytosis, autophagy and immunity in mammals. However, their roles in aquatic species remain poorly known. In the present study, three lamp genes were cloned and characterized from Micropterus salmoides. Subsequently, their transcriptional levels in response to different nutritional status were investigated. The full-length coding sequences of lamp1, lamp2 and lamp3 were 1251bp, 1224bp and 771bp, encoding 416, 407 and 256 amino acids, respectively. Multiple sequence alignment showed that LAMP1-3 were highly conserved among the different fish species, respectively. 3-D structure prediction, genomic survey, and phylogenetic analysis were further confirmed that these genes are widely existed in vertebrates. The mRNA expression of the three genes was ubiquitously expressed in all selected tissues, including liver, brain, gill, heart, muscle, spleen, kidney, stomach, adipose and intestine, lamp1 shows highly transcript levels in brain and muscle, lamp2 displays highly expression level in heart, muscle and spleen, but lamp3 shows highly transcript level in spleen, liver and kidney. To analyze the function of the three genes under starvation stress in largemouth bass, three experimental treatment groups (fasted group and refeeding group, control group) were established in the current study. The results indicated that the expression of lamp1 was significant induced after starvation, and then returned to normal levels after refeeding in the liver. The expression of lamp2 and lamp3 exhibited the same trend in the liver. In addition, in the spleen and the kidney, the transcript level of lamp1 and lamp2 was remarkably increased in the fasted treatment group and slightly decreased in the refed treatment group, respectively. Collectively, our findings suggest that three lamp genes may have differential function in the immune and energetic organism in largemouth bass, which is helpful in understanding roles of lamps in aquatic species.
Collapse
Affiliation(s)
- Yan-Lin Yang
- Hubei Key Laboratory of Waterlogging Disaster and Agricultural Use of Wetland, Yangtze University, Jingzhou, China
- School of Animal Science, Yangtze University, Jingzhou, China
| | - Wan-Hong Zeng
- Hubei Key Laboratory of Waterlogging Disaster and Agricultural Use of Wetland, Yangtze University, Jingzhou, China
- School of Animal Science, Yangtze University, Jingzhou, China
| | - Yong Peng
- Hubei Key Laboratory of Waterlogging Disaster and Agricultural Use of Wetland, Yangtze University, Jingzhou, China
- School of Animal Science, Yangtze University, Jingzhou, China
| | - Shi-Yu Zuo
- Hubei Key Laboratory of Waterlogging Disaster and Agricultural Use of Wetland, Yangtze University, Jingzhou, China
- School of Animal Science, Yangtze University, Jingzhou, China
| | - Yuan-Qi Fu
- Hubei Key Laboratory of Waterlogging Disaster and Agricultural Use of Wetland, Yangtze University, Jingzhou, China
- School of Animal Science, Yangtze University, Jingzhou, China
| | - Yi-Ming Xiao
- Hubei Key Laboratory of Waterlogging Disaster and Agricultural Use of Wetland, Yangtze University, Jingzhou, China
- School of Animal Science, Yangtze University, Jingzhou, China
| | - Wen-Li Huang
- Hubei Key Laboratory of Waterlogging Disaster and Agricultural Use of Wetland, Yangtze University, Jingzhou, China
- School of Animal Science, Yangtze University, Jingzhou, China
| | - Zheng-Yong Wen
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang, China
| | - Wei Hu
- Hubei Key Laboratory of Waterlogging Disaster and Agricultural Use of Wetland, Yangtze University, Jingzhou, China
- School of Animal Science, Yangtze University, Jingzhou, China
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang, China
| | - Yu-Ying Yang
- Hubei Key Laboratory of Waterlogging Disaster and Agricultural Use of Wetland, Yangtze University, Jingzhou, China
- School of Animal Science, Yangtze University, Jingzhou, China
| | - Xiao-Feng Huang
- Hubei Key Laboratory of Waterlogging Disaster and Agricultural Use of Wetland, Yangtze University, Jingzhou, China
- School of Animal Science, Yangtze University, Jingzhou, China
| |
Collapse
|
27
|
Arines FM, Wielenga A, Henn D, Burata OE, Garcia FN, Stockbridge RB, Li M. Lysosomal membrane transporter purification and reconstitution for functional studies. Mol Biol Cell 2024; 35:ar28. [PMID: 38117592 PMCID: PMC10916862 DOI: 10.1091/mbc.e23-06-0259] [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: 07/03/2023] [Revised: 11/20/2023] [Accepted: 12/13/2023] [Indexed: 12/22/2023] Open
Abstract
Lysosomes achieve their function through numerous transporters that import or export nutrients across their membrane. However, technical challenges in membrane protein overexpression, purification, and reconstitution hinder our understanding of lysosome transporter function. Here, we developed a platform to overexpress and purify the putative lysine transporter Ypq1 using a constitutive overexpression system in protease- and ubiquitination-deficient yeast vacuoles. Using this method, we purified and reconstituted Ypq1 into proteoliposomes and showed lysine transport function, supporting its role as a basic amino acid transporter on the vacuole membrane. We also found that the absence of lysine destabilizes purified Ypq1 and causes it to aggregate, consistent with its propensity to be downregulated in vivo upon lysine starvation. Our approach may be useful for the biochemical characterization of many transporters and membrane proteins to understand organellar transport and regulation.
Collapse
Affiliation(s)
- Felichi Mae Arines
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Aleksander Wielenga
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Danielle Henn
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Olive E. Burata
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Francisco Narro Garcia
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Randy B. Stockbridge
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Ming Li
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
28
|
Kang JS, Joo MD, Lee SH, Kang SM, Haider Z, Perera CD, Idrees M, Jin Y, Kong IK. Effect of additional cytoplasm injection on the cloned bovine embryo organelle distribution and stress mitigation. Theriogenology 2024; 216:12-19. [PMID: 38147714 DOI: 10.1016/j.theriogenology.2023.11.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 11/29/2023] [Accepted: 11/29/2023] [Indexed: 12/28/2023]
Abstract
Although somatic cell nuclear transfer (SCNT) is a critical component of animal cloning, this approach has several issues. We previously introduced the cytoplasm injection cloning technology (CICT), which significantly improves the quality and quantity of cloned embryos. This study examined the residual status of fused cytoplasmic organelles, such as the endoplasmic reticulum (ER) and lysosomes, in the CICT group during early embryo development. We found that extra-cytoplasmic organelles stained using the ER-Tracker™ Green dye and LysoTracker™ Deep Red probe were fused and dispersed throughout the recipient oocyte and were still visible in day 8 blastocysts. We screened for ER stress, autophagy, and apoptosis-related genes to elucidate the association between the added organelles and improved embryo quality in CICT-cloned embryos. We found that CHOP, ATF4, ATG5, ATG7, and LC3 genes showed non-significantly up- or downregulated expression between CICT- and in vitro fertilization (IVF)-derived embryos but showed significantly (p < 0.05) upregulated expression in SCNT-cloned embryos. Surprisingly, a non-significant difference in the expression of some genes, such as ATF6 and caspase-3, was observed between the CICT- and SCNT-cloned embryos. Our findings imply that compared to conventional SCNT cloning, CICT-derived cloned embryos with additional cytoplasm have much higher organelle activity, lower autophagy, lower rates of apoptosis, and higher embryo development rates.
Collapse
Affiliation(s)
- Ji-Su Kang
- Division of Applied Life Science (BK21 Four), Graduate School of Applied Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea.
| | - Myeong-Don Joo
- Division of Applied Life Science (BK21 Four), Graduate School of Applied Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea.
| | - Seo-Hyeon Lee
- Division of Applied Life Science (BK21 Four), Graduate School of Applied Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Seon-Min Kang
- Division of Applied Life Science (BK21 Four), Graduate School of Applied Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Zaheer Haider
- Division of Applied Life Science (BK21 Four), Graduate School of Applied Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Chalani Dilshani Perera
- Division of Applied Life Science (BK21 Four), Graduate School of Applied Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Muhammad Idrees
- Division of Applied Life Science (BK21 Four), Graduate School of Applied Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea; Institute of Agriculture and Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Yongxun Jin
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, 130062, Jilin, PR China.
| | - Il-Keun Kong
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, 130062, Jilin, PR China; Division of Applied Life Science (BK21 Four), Graduate School of Applied Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea; Institute of Agriculture and Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea; Thekingkong Co. Ltd., Gyeongsang National University, Jinju, 52828, Republic of Korea.
| |
Collapse
|
29
|
Hüsnügil HH, Güleç Taşkıran AE, Güderer I, Nehri LN, Oral G, Menemenli NŞ, Özcan Ö, Noghreh A, Akyol A, Banerjee S. Lysosomal alkalinization in nutrient restricted cancer cells activates cytoskeletal rearrangement to enhance partial epithelial to mesenchymal transition. Transl Oncol 2024; 41:101860. [PMID: 38262111 PMCID: PMC10832471 DOI: 10.1016/j.tranon.2023.101860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/22/2023] [Accepted: 12/06/2023] [Indexed: 01/25/2024] Open
Abstract
INTRODUCTION Nutrient restriction in cancer cells can activate a number of stress response pathways for cell survival. We aimed to determine mechanistically how nutrient depletion in colorectal cancer (CRC) cells leads to cellular adaptation. MATERIALS AND METHODS Cell survival under nutrient depletion (ND) was evaluated by colony formation and in vivo tumor formation assays. Lysosomes are activated with ND; therefore, we incubated the ND cells with the V-ATPase inhibitor Bafilomycin A1 (ND+Baf). The expression of epithelial and mesenchymal markers with ND+Baf was determined by RNA sequencing and RT-qPCR while motility was determined with an in vivo Chorioallantoic membrane (CAM) assay. Reorganization of cytoskeletal network and lysosomal positioning was determined by immunocytochemistry. RESULTS 4 different colorectal cancer (CRC) cell lines under ND showed high viability, tumor forming ability and increased expression of one or more epithelial and mesenchymal markers, suggesting the activation of partial (p)-EMT. We observed a further increase in p-EMT markers, numerous membrane protrusions, decreased cell-cell adhesion in 3D, and increased motility in ND+Baf cells. The protrusions in the ND+Baf cells were primarily mediated by microtubules and enabled the relocalization of lysosomes from the perinuclear region to the periphery. CONCLUSIONS ND activated p-EMT in CRC cells, which was exacerbated by lysosomal alkalinization. The ND+Baf cells also showed numerous protrusions containing lysosomes, which may lead to lysosomal exocytosis and enhanced motility.
Collapse
Affiliation(s)
- H Hazal Hüsnügil
- Department of Biological Sciences, Orta Dogu Teknik Universitesi, Ankara, Turkey
| | - Aliye Ezgi Güleç Taşkıran
- Department of Biological Sciences, Orta Dogu Teknik Universitesi, Ankara, Turkey; Department of Molecular Biology and Genetics, Başkent University, Ankara, Turkey
| | - Ismail Güderer
- Department of Biological Sciences, Orta Dogu Teknik Universitesi, Ankara, Turkey
| | - Leman Nur Nehri
- Department of Biological Sciences, Orta Dogu Teknik Universitesi, Ankara, Turkey
| | - Göksu Oral
- Department of Biological Sciences, Orta Dogu Teknik Universitesi, Ankara, Turkey
| | | | - Özün Özcan
- Department of Biological Sciences, Orta Dogu Teknik Universitesi, Ankara, Turkey
| | - Ariana Noghreh
- Department of Biological Sciences, Orta Dogu Teknik Universitesi, Ankara, Turkey
| | - Aytekin Akyol
- Hacettepe University Faculty of Medicine, Department of Medical Pathology, Ankara, Turkey
| | - Sreeparna Banerjee
- Department of Biological Sciences, Orta Dogu Teknik Universitesi, Ankara, Turkey; Cancer Systems Biology Laboratory CanSyL, Orta Dogu Teknik Universitesi, Ankara, Turkey.
| |
Collapse
|
30
|
Mohapatra G, Dachet F, Coleman LJ, Gillis B, Behm FG. Identification of unique genomic signatures in patients with fibromyalgia and chronic pain. Sci Rep 2024; 14:3949. [PMID: 38366049 PMCID: PMC10873305 DOI: 10.1038/s41598-024-53874-8] [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: 12/02/2022] [Accepted: 02/06/2024] [Indexed: 02/18/2024] Open
Abstract
Fibromyalgia (FM) is a chronic pain syndrome characterized by widespread pain. The pathophysiology of fibromyalgia is not clearly understood and there are no specific biomarkers available for accurate diagnosis. Here we define genomic signatures using high throughput RNA sequencing on 96 fibromyalgia and 93 control cases. Our findings revealed three major fibromyalgia-associated expression signatures. The first group included 43 patients with a signature enriched for gene expression associated with extracellular matrix and downregulation of RhoGDI signaling pathway. The second group included 30 patients and showed a profound reduction in the expression of inflammatory mediators with an increased expression of genes involved in the CLEAR signaling pathway. These results suggest defective tissue homeostasis associated with the extra-cellular matrix and cellular program that regulates lysosomal biogenesis and participates in macromolecule clearance in fibromyalgia. The third group of 17 FM patients showed overexpression of pathways that control acute inflammation and dysfunction of the global transcriptional process. The result of this study indicates that FM is a heterogeneous and complex disease. Further elucidation of these pathways will lead to the development of accurate diagnostic markers, and effective therapeutic options for fibromyalgia.
Collapse
Affiliation(s)
- Gayatry Mohapatra
- Laboratory of Genomic Medicine, Department of Pathology, University of Illinois at Chicago (UIC) College of Medicine, 840 S. Wood St., Chicago, IL, 60612, USA.
| | - Fabien Dachet
- Laboratory of Genomic Medicine, Department of Pathology, University of Illinois at Chicago (UIC) College of Medicine, 840 S. Wood St., Chicago, IL, 60612, USA
| | - Louis J Coleman
- Laboratory of Genomic Medicine, Department of Pathology, University of Illinois at Chicago (UIC) College of Medicine, 840 S. Wood St., Chicago, IL, 60612, USA
| | - Bruce Gillis
- Department of Medicine, University of Illinois at Chicago (UIC) College of Medicine, Chicago, USA
| | - Frederick G Behm
- Laboratory of Genomic Medicine, Department of Pathology, University of Illinois at Chicago (UIC) College of Medicine, 840 S. Wood St., Chicago, IL, 60612, USA
| |
Collapse
|
31
|
Jerabkova-Roda K, Marwaha R, Das T, Goetz JG. Organelle morphology and positioning orchestrate physiological and disease-associated processes. Curr Opin Cell Biol 2024; 86:102293. [PMID: 38096602 PMCID: PMC7616369 DOI: 10.1016/j.ceb.2023.102293] [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: 08/28/2023] [Revised: 10/24/2023] [Accepted: 11/19/2023] [Indexed: 02/15/2024]
Abstract
In cells, organelles are distributed nonrandomly to regulate cells' physiological and disease-associated processes. Based on their morphology, position within the cell, and contacts with other organelles, they exert different biological functions. Endo-lysosomes are critical cell metabolism and nutrient-sensing regulators modulating cell growth and cellular adaptation in response to nutrient availability. Their spatial distribution is intimately linked to their function. In this review, we will discuss the role of endolysosomes under physiological conditions and in the context of cancer progression, with a special focus on their morphology, the molecular mechanisms determining their subcellular position, and the contacts they form with other organelles. We aim to highlight the relationship between cell architecture and cell function and its impact on maintaining organismal homeostasis.
Collapse
Affiliation(s)
- Katerina Jerabkova-Roda
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France; Université de Strasbourg, Strasbourg, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France; Equipe Labellisée Ligue Contre le Cancer, France.
| | - Rituraj Marwaha
- Tata Institute of Fundamental Research Hyderabad (TIFR-H), Hyderabad, 500 046, India
| | - Tamal Das
- Tata Institute of Fundamental Research Hyderabad (TIFR-H), Hyderabad, 500 046, India
| | - Jacky G Goetz
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France; Université de Strasbourg, Strasbourg, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France; Equipe Labellisée Ligue Contre le Cancer, France
| |
Collapse
|
32
|
Pozzi M, Vantaggiato C, Brivio F, Orso G, Bassi MT. Olanzapine, risperidone and ziprasidone differently affect lysosomal function and autophagy, reflecting their different metabolic risk in patients. Transl Psychiatry 2024; 14:13. [PMID: 38191558 PMCID: PMC10774340 DOI: 10.1038/s41398-023-02686-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 11/16/2023] [Accepted: 11/27/2023] [Indexed: 01/10/2024] Open
Abstract
The metabolic effects induced by antipsychotics in vitro depend on their action on the trafficking and biosynthesis of sterols and lipids. Previous research showed that antipsychotics with different adverse effects in patients cause similar alterations in vitro, suggesting the low clinical usefulness of cellular studies. Moreover, the inhibition of peripheral AMPK was suggested as potential aetiopathogenic mechanisms of olanzapine, and different effects on autophagy were reported for several antipsychotics. We thus assessed, in clinically-relevant culture conditions, the aetiopathogenic mechanisms of olanzapine, risperidone and ziprasidone, antipsychotics with respectively high, medium, low metabolic risk in patients, finding relevant differences among them. We highlighted that: olanzapine impairs lysosomal function affecting autophagy and autophagosome clearance, and increasing intracellular lipids and sterols; ziprasidone activates AMPK increasing the autophagic flux and reducing intracellular lipids; risperidone increases lipid accumulation, while it does not affect lysosomal function. These in vitro differences align with their different impact on patients. We also provided evidence that metformin add-on improved autophagy in olanzapine-treated cells and reduced lipid accumulation induced by both risperidone and olanzapine in an AMPK-dependent way; metformin also increased the production of bile acids to eliminate cholesterol accumulations caused by olanzapine. These results have different clinical implications. We demonstrated that antipsychotics with different metabolic impacts on patients actually have different mechanisms of action, thus supporting the possibility of a personalised antipsychotic treatment. Moreover, we found that metformin can fully revert the phenotype caused by risperidone but not the one caused by olanzapine, that still activates SREBP2.
Collapse
Affiliation(s)
- Marco Pozzi
- Scientific Institute IRCCS Eugenio Medea, Laboratory of Molecular Biology, Via D. L. Monza 20, 23842, Bosisio Parini, Lecco, Italy.
| | - Chiara Vantaggiato
- Scientific Institute IRCCS Eugenio Medea, Laboratory of Molecular Biology, Via D. L. Monza 20, 23842, Bosisio Parini, Lecco, Italy
| | - Francesca Brivio
- Scientific Institute IRCCS Eugenio Medea, Laboratory of Molecular Biology, Via D. L. Monza 20, 23842, Bosisio Parini, Lecco, Italy
| | - Genny Orso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Largo E. Meneghetti 2, Padova, Italy
| | - Maria Teresa Bassi
- Scientific Institute IRCCS Eugenio Medea, Laboratory of Molecular Biology, Via D. L. Monza 20, 23842, Bosisio Parini, Lecco, Italy
| |
Collapse
|
33
|
Vallucci M, Boutin JA, Janda E, Blandel F, Musgrove R, Di Monte D, Ferry G, Michel PP, Hirsch EC. The specific NQO2 inhibitor, S29434, only marginally improves the survival of dopamine neurons in MPTP-intoxicated mice. J Neural Transm (Vienna) 2024; 131:1-11. [PMID: 37851107 DOI: 10.1007/s00702-023-02709-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 10/02/2023] [Indexed: 10/19/2023]
Abstract
Over the years, evidence has accumulated on a possible contributive role of the cytosolic quinone reductase NQO2 in models of dopamine neuron degeneration induced by parkinsonian toxin, but most of the data have been obtained in vitro. For this reason, we asked the question whether NQO2 is involved in the in vivo toxicity of MPTP, a neurotoxin classically used to model Parkinson disease-induced neurodegeneration. First, we show that NQO2 is expressed in mouse substantia nigra dopaminergic cell bodies and in human dopaminergic SH-SY5Y cells as well. A highly specific NQO2 inhibitor, S29434, was able to reduce MPTP-induced cell death in a co-culture system of SH-SY5Y cells with astrocytoma U373 cells but was inactive in SH-SY5Y monocultures. We found that S29434 only marginally prevents substantia nigra tyrosine hydroxylase+ cell loss after MPTP intoxication in vivo. The compound produced a slight increase of dopaminergic cell survival at day 7 and 21 following MPTP treatment, especially with 1.5 and 3 mg/kg dosage regimen. The rescue effect did not reach statistical significance (except for one experiment at day 7) and tended to decrease with the 4.5 mg/kg dose, at the latest time point. Despite the lack of robust protective activity of the inhibitor of NQO2 in the mouse MPTP model, we cannot rule out a possible role of the enzyme in parkinsonian degeneration, particularly because it is substantially expressed in dopaminergic neurons.
Collapse
Affiliation(s)
- Maeva Vallucci
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute -ICM, INSERM, CNRS, Paris, France
| | - Jean A Boutin
- Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication (NorDiC), Univ Rouen Normandie, Inserm, NorDiC, UMR 1239, 76000, Rouen, France.
| | - Elzbieta Janda
- Department of Health Sciences, Campus Germaneto, Magna Graecia University, 88100, Catanzaro, Italy
| | - Florence Blandel
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute -ICM, INSERM, CNRS, Paris, France
| | - Ruth Musgrove
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Donato Di Monte
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Gilles Ferry
- Institut de R&D, Servier Paris-Saclay, 91190, Gif-Sur-Yvette, France
- Gilles Ferry Consulting, Les Issambres, France
| | - Patrick P Michel
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute -ICM, INSERM, CNRS, Paris, France
| | - Etienne C Hirsch
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute -ICM, INSERM, CNRS, Paris, France
| |
Collapse
|
34
|
Srivastav S, van der Graaf K, Singh P, Utama AB, Meyer MD, McNew JA, Stern M. Atl (atlastin) regulates mTor signaling and autophagy in Drosophila muscle through alteration of the lysosomal network. Autophagy 2024; 20:131-150. [PMID: 37649246 PMCID: PMC10761077 DOI: 10.1080/15548627.2023.2249794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 08/08/2023] [Accepted: 08/11/2023] [Indexed: 09/01/2023] Open
Abstract
ABBREVIATIONS atl atlastin; ALR autophagic lysosome reformation; ER endoplasmic reticulum; GFP green fluorescent protein; HSP hereditary spastic paraplegia; Lamp1 lysosomal associated membrane protein 1 PolyUB polyubiquitin; RFP red fluorescent protein; spin spinster; mTor mechanistic Target of rapamycin; VCP valosin containing protein.
Collapse
Affiliation(s)
| | | | - Pratibha Singh
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, USA
| | | | - Matthew D. Meyer
- Shared Equipment Authority, Rice University, Houston, Texas, USA
| | - James A. McNew
- Department of BioSciences, Rice University, Houston, Texas, USA
| | - Michael Stern
- Department of BioSciences, Rice University, Houston, Texas, USA
| |
Collapse
|
35
|
Shirbhate E, Singh V, Mishra A, Jahoriya V, Veerasamy R, Tiwari AK, Rajak H. Targeting Lysosomes: A Strategy Against Chemoresistance in Cancer. Mini Rev Med Chem 2024; 24:1449-1468. [PMID: 38343053 DOI: 10.2174/0113895575287242240129120002] [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: 10/13/2023] [Revised: 01/11/2024] [Accepted: 01/19/2024] [Indexed: 07/23/2024]
Abstract
Chemotherapy is still the major method of treatment for many types of cancer. Curative cancer therapy is hampered significantly by medication resistance. Acidic organelles like lysosomes serve as protagonists in cellular digestion. Lysosomes, however, are gaining popularity due to their speeding involvement in cancer progression and resistance. For instance, weak chemotherapeutic drugs of basic nature permeate through the lysosomal membrane and are retained in lysosomes in their cationic state, while extracellular release of lysosomal enzymes induces cancer, cytosolic escape of lysosomal hydrolases causes apoptosis, and so on. Drug availability at the sites of action is decreased due to lysosomal drug sequestration, which also enhances cancer resistance. This review looks at lysosomal drug sequestration mechanisms and how they affect cancer treatment resistance. Using lysosomes as subcellular targets to combat drug resistance and reverse drug sequestration is another method for overcoming drug resistance that is covered in this article. The present review has identified lysosomal drug sequestration as one of the reasons behind chemoresistance. The article delves deeper into specific aspects of lysosomal sequestration, providing nuanced insights, critical evaluations, or novel interpretations of different approaches that target lysosomes to defect cancer.
Collapse
Affiliation(s)
- Ekta Shirbhate
- Department of Pharmacy, Guru Ghasidas University, Bilaspur-495 009, (C.G.), India
| | - Vaibhav Singh
- Department of Pharmacy, Guru Ghasidas University, Bilaspur-495 009, (C.G.), India
| | - Aditya Mishra
- Department of Pharmacy, Guru Ghasidas University, Bilaspur-495 009, (C.G.), India
| | - Varsha Jahoriya
- Department of Pharmacy, Guru Ghasidas University, Bilaspur-495 009, (C.G.), India
| | - Ravichandran Veerasamy
- Faculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Kedah Darul Aman, Malaysia
| | - Amit K Tiwari
- UAMS College of Pharmacy; UAMS - University of Arkansas for Medical Sciences, (AR) USA
| | - Harish Rajak
- Department of Pharmacy, Guru Ghasidas University, Bilaspur-495 009, (C.G.), India
| |
Collapse
|
36
|
Maji S, Pirozzi M, Ruturaj, Pandey R, Ghosh T, Das S, Gupta A. Copper-independent lysosomal localisation of the Wilson disease protein ATP7B. Traffic 2023; 24:587-609. [PMID: 37846526 DOI: 10.1111/tra.12919] [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: 10/20/2022] [Revised: 09/10/2023] [Accepted: 09/23/2023] [Indexed: 10/18/2023]
Abstract
In hepatocytes, the Wilson disease protein ATP7B resides on the trans-Golgi network (TGN) and traffics to peripheral lysosomes to export excess intracellular copper through lysosomal exocytosis. We found that in basal copper or even upon copper chelation, a significant amount of ATP7B persists in the endolysosomal compartment of hepatocytes but not in non-hepatic cells. These ATP7B-harbouring lysosomes lie in close proximity of ~10 nm to the TGN. ATP7B constitutively distributes itself between the sub-domain of the TGN with a lower pH and the TGN-proximal lysosomal compartments. The presence of ATP7B on TGN-lysosome colocalising sites upon Golgi disruption suggested a possible exchange of ATP7B directly between the TGN and its proximal lysosomes. Manipulating lysosomal positioning significantly alters the localisation of ATP7B in the cell. Contrary to previous understanding, we found that upon copper chelation in a copper-replete hepatocyte, ATP7B is not retrieved back to TGN from peripheral lysosomes; rather, ATP7B recycles to these TGN-proximal lysosomes to initiate the next cycle of copper transport. We report a hitherto unknown copper-independent lysosomal localisation of ATP7B and the importance of TGN-proximal lysosomes but not TGN as the terminal acceptor organelle of ATP7B in its retrograde pathway.
Collapse
Affiliation(s)
- Saptarshi Maji
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| | | | - Ruturaj
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| | - Raviranjan Pandey
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| | - Tamal Ghosh
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| | - Santanu Das
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| | - Arnab Gupta
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| |
Collapse
|
37
|
Zhao S, Hu Q, Jiang H, Zhao Y, Wang Y, Feng C, Li X. Multi-omics analysis of oxidative stress and apoptosis in hepatopancreas cells induced by Polyascus gregaria parasitizing the Eriocheir sinensis. FISH & SHELLFISH IMMUNOLOGY 2023; 143:109180. [PMID: 37863124 DOI: 10.1016/j.fsi.2023.109180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/16/2023] [Accepted: 10/18/2023] [Indexed: 10/22/2023]
Abstract
Polyascus gregaria, a parasitic barnacle, poses a significant threat to Eriocheir sinensis farms by inhibiting crab growth. However, the molecular and pathological mechanisms behind P. gregaria infection in the hepatopancreas of E. sinensis remain unclear. In this study, we investigated the impact and underlying mechanisms of P. gregaria infection on E. sinensis through analyzing the infected hepatopancreatic tissues by tandem mass tag technology and RNA-Seq high-throughput sequencing. Among the identified 10,693 differentially expressed genes, 294 genes were significantly altered following P. gregaria infection, including 92 upregulated and 202 downregulated genes. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses further revealed the involvement of these genes in oxidative decomposition, lipid metabolism, inflammation, and hepatopancreas metabolism. Meanwhile, the identified 253 differentially expressed proteins, including 143 upregulated and 110 downregulated proteins, are mainly related to cellular and metabolic processes, catalytic activity, and cell components. The pathway analysis indicated their enrichment in glycolysis/gluconeogenesis, oxidative phosphorylation, endoplasmic reticulum protein processing, and actin cytoskeleton regulation. The involvement of these differentially expressed genes and proteins in the peroxisome proliferator-activated receptors pathway during host immune responses against P. gregaria infection has been highlighted. Furthermore, pathological examinations and biochemical indicators jointly demonstrated the hepatopancreatic damage and increased oxidative stress and apoptosis in the infected E. sinensis. Collectively, our study provides crucial insights into the mechanisms underlying the E. sinensis-P. gregaria interactions, and may contribute to the development of novel strategies for parasite control and reducing economic losses in aquaculture.
Collapse
Affiliation(s)
- Shiwei Zhao
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
| | - Qingbiao Hu
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
| | - Hongbo Jiang
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
| | - Yingying Zhao
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
| | - Yanping Wang
- Linong Testing (Binzhou) Co., Ltd., Binzhou Bohai Advanced Technology Research Institute, Binzhou, 256600, China
| | - Chengcheng Feng
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China.
| | - Xiaodong Li
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China.
| |
Collapse
|
38
|
Oh KW, Kim DK, Hsu AL, Lee SJ. Distinct sets of lysosomal genes define synucleinopathy and tauopathy. BMB Rep 2023; 56:657-662. [PMID: 37817435 PMCID: PMC10761752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/23/2023] [Accepted: 09/11/2023] [Indexed: 10/12/2023] Open
Abstract
Neurodegenerative diseases are characterized by distinct protein aggregates, such as those of α-synuclein and tau. Lysosomal defect is a key contributor to the accumulation and propagation of aberrant protein aggregates in these diseases. The discoveries of common proteinopathies in multiple forms of lysosomal storage diseases (LSDs) and the identification of some LSD genes as susceptible genes for those proteinopathies suggest causative links between LSDs and the proteinopathies. The present study hypothesized that defects in lysosomal genes will differentially affect the propagation of α-synuclein and tau proteins, thereby determining the progression of a specific proteinopathy. We established an imaging-based high-contents screening (HCS) system in Caenorhabditis elegans (C. elegans) model, by which the propagation of α-synuclein or tau is measured by fluorescence intensity. Using this system, we performed RNA interference (RNAi) screening to induce a wide range of lysosomal malfunction through knock down of 79 LSD genes, and to obtain the candidate genes with significant change in protein propagation. While some LSD genes commonly affected both α-synuclein and tau propagation, our study identified the distinct sets of LSD genes that differentially regulate the propagation of either α-synuclein or tau. The specificity and efficacy of these LSD genes were retained in the disease-related phenotypes, such as pharyngeal pumping behavior and life span. This study suggests that distinct lysosomal genes differentially regulate the propagation of α-synuclein and tau, and offer a steppingstone to understanding disease specificity. [BMB Reports 2023; 56(12): 657-662].
Collapse
Affiliation(s)
- Kyu Won Oh
- Department of Biomedical Sciences, Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea, Seoul 04796, Korea
| | - Dong-Kyu Kim
- Department of Biomedical Sciences, Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea, Seoul 04796, Korea
| | - Ao-Lin Hsu
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 112-304, Taiwan, Seoul 04796, Korea
| | - Seung-Jae Lee
- Department of Biomedical Sciences, Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea, Seoul 04796, Korea
- Convergence Research Center for Dementia, Seoul National University College of Medicine, Seoul 03081, Korea
- Neuramedy Co. Ltd., Seoul 04796, Korea
| |
Collapse
|
39
|
MacLeod CM, Yousufzai FAK, Spencer LT, Kim S, Rivera-Rosario LA, Barrera ZD, Walsh L, Krummenacher C, Carone B, Soto I. Trehalose enhances mitochondria deficits in human NPC1 mutant fibroblasts but disrupts mouse Purkinje cell dendritic growth ex vivo. PLoS One 2023; 18:e0294312. [PMID: 38033125 PMCID: PMC10688965 DOI: 10.1371/journal.pone.0294312] [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: 06/25/2023] [Accepted: 10/29/2023] [Indexed: 12/02/2023] Open
Abstract
Lysosomes play important roles in catabolism, nutrient sensing, metabolic signaling, and homeostasis. NPC1 deficiency disrupts lysosomal function by inducing cholesterol accumulation that leads to early neurodegeneration in Niemann-Pick type C (NPC) disease. Mitochondria pathology and deficits in NPC1 deficient cells are associated with impaired lysosomal proteolysis and metabolic signaling. It is thought that activation of the transcription factor TFEB, an inducer of lysosome biogenesis, restores lysosomal-autophagy activity in lysosomal storage disorders. Here, we investigated the effect of trehalose, a TFEB activator, in the mitochondria pathology of NPC1 mutant fibroblasts in vitro and in mouse developmental Purkinje cells ex vivo. We found that in NPC1 mutant fibroblasts, serum starvation or/and trehalose treatment, both activators of TFEB, reversed mitochondria fragmentation to a more tubular mitochondrion. Trehalose treatment also decreased the accumulation of Filipin+ cholesterol in NPC1 mutant fibroblasts. However, trehalose treatment in cerebellar organotypic slices (COSCs) from wild-type and Npc1nmf164 mice caused mitochondria fragmentation and lack of dendritic growth and degeneration in developmental Purkinje cells. Our data suggest, that although trehalose successfully restores mitochondria length and decreases cholesterol accumulation in NPC1 mutant fibroblasts, in COSCs, Purkinje cells mitochondria and dendritic growth are negatively affected possibly through the overactivation of the TFEB-lysosomal-autophagy pathway.
Collapse
Affiliation(s)
- Collin M. MacLeod
- Department of Biology, Providence College, Providence, RI, United States of America
| | - Fawad A. K. Yousufzai
- Department of Biological & Biomedical Sciences, Rowan University, Glassboro, NJ, United States of America
| | - Liam T. Spencer
- Department of Biology, Providence College, Providence, RI, United States of America
| | - Sarah Kim
- Department of Biological & Biomedical Sciences, Rowan University, Glassboro, NJ, United States of America
| | | | - Zerian D. Barrera
- Department of Biological & Biomedical Sciences, Rowan University, Glassboro, NJ, United States of America
| | - Lindsay Walsh
- Department of Biology, Providence College, Providence, RI, United States of America
| | - Claude Krummenacher
- Department of Biological & Biomedical Sciences, Rowan University, Glassboro, NJ, United States of America
| | - Benjamin Carone
- Department of Biological & Biomedical Sciences, Rowan University, Glassboro, NJ, United States of America
| | - Ileana Soto
- Department of Biology, Providence College, Providence, RI, United States of America
| |
Collapse
|
40
|
He L, Chen J, Deng P, Huang S, Liu P, Wang C, Huang X, Li Y, Chen B, Shi D, Xiao Y, Chen X, Ouyang Y, Song L, Lin C. Lysosomal cyst(e)ine storage potentiates tolerance to oxidative stress in cancer cells. Mol Cell 2023; 83:3502-3519.e11. [PMID: 37751742 DOI: 10.1016/j.molcel.2023.08.032] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 07/17/2023] [Accepted: 08/30/2023] [Indexed: 09/28/2023]
Abstract
Cyst(e)ine is a key precursor for the synthesis of glutathione (GSH), which protects cancer cells from oxidative stress. Cyst(e)ine is stored in lysosomes, but its role in redox regulation is unclear. Here, we show that breast cancer cells upregulate major facilitator superfamily domain containing 12 (MFSD12) to increase lysosomal cyst(e)ine storage, which is released by cystinosin (CTNS) to maintain GSH levels and buffer oxidative stress. We find that mTORC1 regulates MFSD12 by directly phosphorylating residue T254, while mTORC1 inhibition enhances lysosome acidification that activates CTNS. This switch modulates lysosomal cyst(e)ine levels in response to oxidative stress, fine-tuning redox homeostasis to enhance cell fitness. MFSD12-T254A mutant inhibits MFSD12 function and suppresses tumor progression. Moreover, MFSD12 overexpression correlates with poor neoadjuvant chemotherapy response and prognosis in breast cancer patients. Our findings reveal the critical role of lysosomal cyst(e)ine storage in adaptive redox homeostasis and suggest that MFSD12 is a potential therapeutic target.
Collapse
Affiliation(s)
- Lixin He
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Jinxin Chen
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Pinwei Deng
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Shumei Huang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Pian Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chanjuan Wang
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xinjian Huang
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yue Li
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Boyu Chen
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Dongni Shi
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yunyun Xiao
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xiangfu Chen
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Ying Ouyang
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Libing Song
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Institute of Oncology, Tumor Hospital, Guangzhou Medical University, Guangzhou 510080, China
| | - Chuyong Lin
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; Guangdong Esophageal Cancer Institute, Guangzhou 510060, China.
| |
Collapse
|
41
|
Mahé M, Rios-Fuller TJ, Karolin A, Schneider RJ. Genetics of enzymatic dysfunctions in metabolic disorders and cancer. Front Oncol 2023; 13:1230934. [PMID: 37601653 PMCID: PMC10433910 DOI: 10.3389/fonc.2023.1230934] [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: 05/29/2023] [Accepted: 07/19/2023] [Indexed: 08/22/2023] Open
Abstract
Inherited metabolic disorders arise from mutations in genes involved in the biogenesis, assembly, or activity of metabolic enzymes, leading to enzymatic deficiency and severe metabolic impairments. Metabolic enzymes are essential for the normal functioning of cells and are involved in the production of amino acids, fatty acids and nucleotides, which are essential for cell growth, division and survival. When the activity of metabolic enzymes is disrupted due to mutations or changes in expression levels, it can result in various metabolic disorders that have also been linked to cancer development. However, there remains much to learn regarding the relationship between the dysregulation of metabolic enzymes and metabolic adaptations in cancer cells. In this review, we explore how dysregulated metabolism due to the alteration or change of metabolic enzymes in cancer cells plays a crucial role in tumor development, progression, metastasis and drug resistance. In addition, these changes in metabolism provide cancer cells with a number of advantages, including increased proliferation, resistance to apoptosis and the ability to evade the immune system. The tumor microenvironment, genetic context, and different signaling pathways further influence this interplay between cancer and metabolism. This review aims to explore how the dysregulation of metabolic enzymes in specific pathways, including the urea cycle, glycogen storage, lysosome storage, fatty acid oxidation, and mitochondrial respiration, contributes to the development of metabolic disorders and cancer. Additionally, the review seeks to shed light on why these enzymes represent crucial potential therapeutic targets and biomarkers in various cancer types.
Collapse
Affiliation(s)
| | | | | | - Robert J. Schneider
- Department of Microbiology, Grossman NYU School of Medicine, New York, NY, United States
| |
Collapse
|
42
|
Wang T, Qin Y, Ye Z, Jing DS, Fan GX, Liu MQ, Zhuo QF, Ji SR, Chen XM, Yu XJ, Xu XW, Li Z. A new glance at autophagolysosomal-dependent or -independent function of transcriptional factor EB in human cancer. Acta Pharmacol Sin 2023; 44:1536-1548. [PMID: 37012494 PMCID: PMC10374590 DOI: 10.1038/s41401-023-01078-7] [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: 10/27/2022] [Accepted: 03/14/2023] [Indexed: 04/05/2023]
Abstract
Autophagy-lysosome system plays a variety of roles in human cancers. In addition to being implicated in metabolism, it is also involved in tumor immunity, remodeling the tumor microenvironment, vascular proliferation, and promoting tumor progression and metastasis. Transcriptional factor EB (TFEB) is a major regulator of the autophagy-lysosomal system. With the in-depth studies on TFEB, researchers have found that it promotes various cancer phenotypes by regulating the autophagolysosomal system, and even in an autophagy-independent way. In this review, we summarize the recent findings about TFEB in various types of cancer (melanoma, pancreatic ductal adenocarcinoma, renal cell carcinoma, colorectal cancer, breast cancer, prostate cancer, ovarian cancer and lung cancer), and shed some light on the mechanisms by which it may serve as a potential target for cancer treatment.
Collapse
Affiliation(s)
- Ting Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, China
| | - Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, China
| | - Zeng Ye
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - De-Sheng Jing
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Gui-Xiong Fan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Meng-Qi Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Qi-Feng Zhuo
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Shun-Rong Ji
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Xue-Min Chen
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, China
| | - Xian-Jun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Xiao-Wu Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Zheng Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
43
|
P K, Unniram Parambil AR, Silswal A, Pramanik A, Koner AL. Trivalent metal ion sensor enabled bioimaging and quantification of vaccine-deposited Al 3+ in lysosomes. Analyst 2023; 148:2425-2437. [PMID: 37194365 DOI: 10.1039/d3an00562c] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Extracellular metallic debris is deposited into the well-known 'recycle bins' of the cells named lysosomes. The accumulation of unwanted metal ions can cause dysfunction of hydrolyzing enzymes and membrane rupturing. Thus, herein, we synthesized rhodamine-acetophenone/benzaldehyde derivatives for the detection of trivalent metal ions in aqueous media. In solution, the synthesized probes exhibited a 'turn-on' colorimetric and fluorometric response upon complexation with trivalent metal ions (M3+). Mechanistically, M3+ chelation enables the appearance of a new emission band at approximately 550 nm, which verifies the disruption of the closed ring and the restoration of conjugation on the xanthene core in rhodamine 6G derivatives. Exclusive localization of the biocompatible probes at the lysosomal compartment favored the quantification of deposited Al3+. Moreover, the novelty of the work lies in the detection of Al3+ deposited in the lysosome that originated from hepatitis B vaccines, which shows their efficiency for near future in vivo applications.
Collapse
Affiliation(s)
- Kavyashree P
- Bionanotechnology Lab, Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal-462066, Madhya Pradesh, India.
| | - Ajmal Roshan Unniram Parambil
- Bionanotechnology Lab, Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal-462066, Madhya Pradesh, India.
| | - Akshay Silswal
- Bionanotechnology Lab, Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal-462066, Madhya Pradesh, India.
| | - Anup Pramanik
- Department of Chemistry, Sidho-Kanho-Birsha University, Purulia, 723104, West Bengal, India
| | - Apurba Lal Koner
- Bionanotechnology Lab, Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal-462066, Madhya Pradesh, India.
| |
Collapse
|
44
|
Tsujimoto K, Takamatsu H, Kumanogoh A. The Ragulator complex: delving its multifunctional impact on metabolism and beyond. Inflamm Regen 2023; 43:28. [PMID: 37173755 PMCID: PMC10175929 DOI: 10.1186/s41232-023-00278-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 05/01/2023] [Indexed: 05/15/2023] Open
Abstract
Our understanding of lysosomes has undergone a significant transformation in recent years, from the view that they are static organelles primarily responsible for the disposal and recycling of cellular waste to their recognition as highly dynamic structures. Current research posits that lysosomes function as a signaling hub that integrates both extracellular and intracellular stimuli, thereby regulating cellular homeostasis. The dysregulation of lysosomal function has been linked to a wide range of diseases. Of note, lysosomes contribute to the activation of mammalian target of rapamycin complex 1 (mTORC1), a key regulator of cellular metabolism. The Ragulator complex, a protein complex anchored on the lysosomal membrane, was initially shown to tether the mTORC1 complex to lysosomes. Recent research has substantially expanded our understanding of the roles of the Ragulator complex in lysosomes, including roles in the regulation of metabolism, inflammation, cell death, cell migration, and the maintenance of homeostasis, via interactions with various proteins. This review summarizes our current knowledge on the diverse functions of the Ragulator complex, highlighting important protein interactions.
Collapse
Affiliation(s)
- Kohei Tsujimoto
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Department of Immunopathology, Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka, Japan
| | - Hyota Takamatsu
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.
- Department of Immunopathology, Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka, Japan.
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Department of Immunopathology, Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka, Japan
- Center for Infectious Diseases Education and Research (CiDER), Osaka University, Suita, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, Japan
- Japan Agency for Medical Research and Development - Core Research for Evolutional Science and Technology (AMED-CREST), Osaka University, Osaka, Japan
- Center for Advanced Modalities and DDS (CAMaD), Osaka University, Osaka, Japan
| |
Collapse
|
45
|
Li Z, Zou J, Chen X. In Response to Precision Medicine: Current Subcellular Targeting Strategies for Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2209529. [PMID: 36445169 DOI: 10.1002/adma.202209529] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 11/08/2022] [Indexed: 05/26/2023]
Abstract
Emerging as a potent anticancer treatment, subcellular targeted cancer therapy has drawn increasing attention, bringing great opportunities for clinical application. Here, two targeting strategies for four main subcellular organelles (mitochondria, lysosome, endoplasmic reticulum, and nucleus), including molecule- and nanomaterial (inorganic nanoparticles, micelles, organic polymers, and others)-based targeted delivery or therapeutic strategies, are summarized. Phototherapy, chemotherapy, radiotherapy, immunotherapy, and "all-in-one" combination therapy are among the strategies covered in detail. Such materials are constructed based on the specific properties and relevant mechanisms of organelles, enabling the elimination of tumors by inducing dysfunction in the corresponding organelles or destroying specific structures. The challenges faced by organelle-targeting cancer therapies are also summarized. Looking forward, a paradigm for organelle-targeting therapy with enhanced therapeutic efficacy compared to current clinical approaches is envisioned.
Collapse
Affiliation(s)
- Zheng Li
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Jianhua Zou
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| |
Collapse
|
46
|
Uzhytchak M, Smolková B, Lunova M, Frtús A, Jirsa M, Dejneka A, Lunov O. Lysosomal nanotoxicity: Impact of nanomedicines on lysosomal function. Adv Drug Deliv Rev 2023; 197:114828. [PMID: 37075952 DOI: 10.1016/j.addr.2023.114828] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 03/28/2023] [Accepted: 04/12/2023] [Indexed: 04/21/2023]
Abstract
Although several nanomedicines got clinical approval over the past two decades, the clinical translation rate is relatively small so far. There are many post-surveillance withdrawals of nanomedicines caused by various safety issues. For successful clinical advancement of nanotechnology, it is of unmet need to realize cellular and molecular foundation of nanotoxicity. Current data suggest that lysosomal dysfunction caused by nanoparticles is emerging as the most common intracellular trigger of nanotoxicity. This review analyzes prospect mechanisms of lysosomal dysfunction-mediated toxicity induced by nanoparticles. We summarized and critically assessed adverse drug reactions of current clinically approved nanomedicines. Importantly, we show that physicochemical properties have great impact on nanoparticles interaction with cells, excretion route and kinetics, and subsequently on toxicity. We analyzed literature on adverse reactions of current nanomedicines and hypothesized that adverse reactions might be linked with lysosomal dysfunction caused by nanomedicines. Finally, from our analysis it becomes clear that it is unjustifiable to generalize safety and toxicity of nanoparticles, since different particles possess distinct toxicological properties. We propose that the biological mechanism of the disease progression and treatment should be central in the optimization of nanoparticle design.
Collapse
Affiliation(s)
- Mariia Uzhytchak
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Barbora Smolková
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Mariia Lunova
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic; Institute for Clinical & Experimental Medicine (IKEM), 14021 Prague, Czech Republic
| | - Adam Frtús
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Milan Jirsa
- Institute for Clinical & Experimental Medicine (IKEM), 14021 Prague, Czech Republic
| | - Alexandr Dejneka
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Oleg Lunov
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic.
| |
Collapse
|
47
|
Makhoul C, Houghton FJ, Hinde E, Gleeson PA. Arf5-mediated regulation of mTORC1 at the plasma membrane. Mol Biol Cell 2023; 34:ar23. [PMID: 36735494 PMCID: PMC10092653 DOI: 10.1091/mbc.e22-07-0302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The mechanistic target of rapamycin (mTOR) kinase regulates a major signaling pathway in eukaryotic cells. In addition to regulation of mTORC1 at lysosomes, mTORC1 is also localized at other locations. However, little is known about the recruitment and activation of mTORC1 at nonlysosomal sites. To identify regulators of mTORC1 recruitment to nonlysosomal compartments, novel interacting partners with the mTORC1 subunit, Raptor, were identified using immunoprecipitation and mass spectrometry. We show that one of the interacting partners, Arf5, is a novel regulator of mTORC1 signaling at plasma membrane ruffles. Arf5-GFP localizes with endogenous mTOR at PI3,4P2-enriched membrane ruffles together with the GTPase required for mTORC1 activation, Rheb. Knockdown of Arf5 reduced the recruitment of mTOR to membrane ruffles. The activation of mTORC1 at membrane ruffles was directly demonstrated using a plasma membrane-targeted mTORC1 biosensor, and Arf5 was shown to enhance the phosphorylation of the mTORC1 biosensor substrate. In addition, endogenous Arf5 was shown to be required for rapid activation of mTORC1-mediated S6 phosphorylation following nutrient starvation and refeeding. Our findings reveal a novel Arf5-dependent pathway for recruitment and activation of mTORC1 at plasma membrane ruffles, a process relevant for spatial and temporal regulation of mTORC1 by receptor and nutrient stimuli.
Collapse
Affiliation(s)
- Christian Makhoul
- Department of Biochemistry and Pharmacology and Bio21 Molecular Science and Biotechnology Institute and
| | - Fiona J Houghton
- Department of Biochemistry and Pharmacology and Bio21 Molecular Science and Biotechnology Institute and
| | - Elizabeth Hinde
- Department of Biochemistry and Pharmacology and Bio21 Molecular Science and Biotechnology Institute and.,School of Physics, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Paul A Gleeson
- Department of Biochemistry and Pharmacology and Bio21 Molecular Science and Biotechnology Institute and
| |
Collapse
|
48
|
Zhong Y, Tang K, Nattel S, Zhai M, Gong S, Yu Q, Zeng Y, E G, Maimaitiaili N, Wang J, Xu Y, Peng W, Li H. Myosin light-chain 4 gene-transfer attenuates atrial fibrosis while correcting autophagic flux dysregulation. Redox Biol 2023; 60:102606. [PMID: 36645977 PMCID: PMC9860351 DOI: 10.1016/j.redox.2023.102606] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 01/12/2023] Open
Abstract
OBJECTIVES To determine the role of MYL4 regulation of lysosomal function and its disturbance in fibrotic atrial cardiomyopathy. BACKGROUND We have previously demonstrated that the atrial-specific essential light chain protein MYL4 is required for atrial contractile, electrical, and structural integrity. MYL4 mutation/dysfunction leads to atrial fibrosis, standstill, and dysrhythmia. However, the underlying pathogenic mechanisms remain unclear. METHODS AND RESULTS Rats subjected to knock-in of a pathogenic MYL4 mutant (p.E11K) developed fibrotic atrial cardiomyopathy. Proteome analysis and single-cell RNA sequencing indicate enrichment of autophagy pathways in mutant-MYL4 atrial dysfunction. Immunofluorescence and electron microscopy revealed undegraded autophagic vesicles accumulated in MYL4p.E11K rat atrium. Next, we identified that dysfunctional MYL4 protein impairs autophagy flux in vitro and in vivo. Cardiac lysosome positioning and mobility were regulated by MYL4 in cardiomyocytes, which affected lysosomal acidification and maturation of lysosomal cathepsins. We then examined the effects of MYL4 overexpression via adenoviral gene-transfer on atrial cardiomyopathy induced by MYL4 mutation: MYL4 protein overexpression attenuated atrial structural remodeling and autophagy dysfunction. CONCLUSIONS MYL4 regulates autophagic flux in atrial cardiomyocytes via lysosomal mobility. MYL4 overexpression attenuates MYL4 p.E11K induced fibrotic atrial cardiomyopathy, while correcting autophagy and lysosomal function. These results provide a molecular basis for MYL4-mutant induced fibrotic atrial cardiomyopathy and identify a potential biological-therapy approach for the treatment of atrial fibrosis.
Collapse
Affiliation(s)
- Yuan Zhong
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Kai Tang
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Stanley Nattel
- Department of Medicine, Montreal Heart Institute, Montreal, Quebec, Canada; Université de Montréal, Quebec, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada; Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany; HIU LYRIC and Fondation Bordeaux Université de Bordeaux, France
| | - Ming Zhai
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shiyu Gong
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qing Yu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yanxi Zeng
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guangxi E
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Nuerbiyemu Maimaitiaili
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yawei Xu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Wenhui Peng
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Hailing Li
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
49
|
Arines FM, Wielenga A, Burata OE, Garcia FN, Stockbridge RB, Li M. Lysosome transporter purification and reconstitution identifies Ypq1 pH-gated lysine transport and regulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.31.535002. [PMID: 37034749 PMCID: PMC10081341 DOI: 10.1101/2023.03.31.535002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
Lysosomes achieve their function through numerous transporters that import or export nutrients across their membrane. However, technical challenges in membrane protein overexpression, purification, and reconstitution hinder our understanding of lysosome transporter function. Here, we developed a platform to overexpress and purify the putative lysine transporter Ypq1 using a constitutive overexpression system in protease- and ubiquitination-deficient yeast vacuoles. Using this method, we purified and reconstituted Ypq1 into proteoliposomes and showed lysine transport function, supporting its role as a basic amino acid transporter on the vacuole membrane. We also found that the absence of lysine destabilizes purified Ypq1 and causes it to aggregate, consistent with its propensity to be downregulated in vivo upon lysine starvation. Our approach may be useful for the biochemical characterization of many transporters and membrane proteins to understand organellar transport and regulation.
Collapse
Affiliation(s)
- Felichi Mae Arines
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Aleksander Wielenga
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Olive E. Burata
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Francisco Narro Garcia
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Randy B. Stockbridge
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ming Li
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
50
|
Ekstrand J, Abrahamsson A, Lundberg P, Dabrosin C. Breast density and estradiol are associated with distinct different expression patterns of metabolic proteins in normal human breast tissue in vivo. Front Oncol 2023; 13:1128318. [PMID: 37064098 PMCID: PMC10090464 DOI: 10.3389/fonc.2023.1128318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/17/2023] [Indexed: 03/31/2023] Open
Abstract
BackgroundBreast density and exposure to sex steroids are major risk factors for breast cancer. The local microenvironment plays an essential role in progression of breast cancer. Metabolic adaption is a major hallmark of cancer. Whether proteins from the extracellular space regulating metabolism are affected in breast cancer, dense breasts or by estrogen exposure are not yet fully elucidated.MethodsWomen with breast cancer, postmenopausal women with normal breast tissue with varying breast density or premenopausal women with breasts exposed to high levels of estradiol were included in the study. Microdialysis was used to collect proteins from the extracellular space in vivo in 73 women; 12 with breast cancer, 42 healthy postmenopausal women with different breast densities, and 19 healthy premenopausal women. Breast density was determined as lean tissue fraction (LTF) using magnetic resonance imaging. Data were evaluated in a murine breast cancer model. We quantified a panel of 92 key proteins regulating metabolism using proximity extension assay.ResultsWe report that 29 proteins were upregulated in human breast cancer. In dense breasts 37 proteins were upregulated and 17 of these were similarly regulated as in breast cancer. 32 proteins correlated with LTF. In premenopausal breasts 19 proteins were up-regulated and 9 down-regulated. Of these, 27 correlated to estradiol, a result that was confirmed for most proteins in experimental breast cancer. Only two proteins, pro-cathepsin H and galanin peptide, were similarly regulated in breast cancer, dense- and estrogen exposed breasts.ConclusionsMetabolic proteins may be targetable for breast cancer prevention. Depending on risk factor, this may, however, require different approaches as breast density and estradiol induce distinct different expression patterns in the breast. Additionally, metabolic proteins from the extracellular space may indeed be further explored as therapeutic targets for breast cancer treatment.
Collapse
Affiliation(s)
- Jimmy Ekstrand
- Department of Oncology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Annelie Abrahamsson
- Department of Oncology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Peter Lundberg
- Department of Radiation Physics and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Charlotta Dabrosin
- Department of Oncology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- *Correspondence: Charlotta Dabrosin,
| |
Collapse
|