1
|
Yang R, Gao Q, Lu X, Wu Y, Zhu C, Han Z, Li C, Wei M. Wash-free and ultra-low concentration monitor lysosomal viscosity in apoptosis with a noteworthy fluorescent probe. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 329:125481. [PMID: 39631200 DOI: 10.1016/j.saa.2024.125481] [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: 09/22/2024] [Revised: 11/03/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024]
Abstract
The visualization and subsequent monitoring of apoptosis holds paramount significance in the domains of physiology, pathology, and pharmacology. However, traditional probes require high staining concentrations and multiple washing steps, which would alter the specimen's micro-environment, potentially inducing harm to specimen. To overcome these challenging issues, we have rationally designed and prepared a pH-inert lysosomal probe (named IVTI) to wash-free visualize apoptosis with ultra-low concentration to alleviate the disturbance of probe concentration, washing procedure and pH variations. Compared with general lysosomal probes, IVTI showed a significant fluorescence boost in reflex to elevated viscosity, while its fluorescence intensity remained mostly still when altering pH values, which could achieve more accurate visualization of lysosomes. Moreover, the probe can detect minute viscosity fluctuations in lysosomes under extra-low concentration, greatly eliminating the effect of probe concentration and washing steps to live bio-samples. Furthermore, compared to LTR (Lyso-Tracker Red, a commercial lysosome probe), IVTI offered exceptional imaging capabilities, and the fluorescence images of IVTI was still clear when lysosomal pH increased, which attributed to the pH-inert properties of IVTI. In view of the excellent imaging abilities, the pH-inert probe was applied to in-situ and real time visualize viscosity changes of live cells under extra-low concentration without washing procedure, and the increase of lysosomal viscosity during apoptosis was also monitored by the probe, thereby minimizing the disturbance of probe concentration, washing procedure and pH variations during apoptosis. The probe possesses tremendous potential in the visualization of dynamic changes related to lysosomes in various physiological processes.
Collapse
Affiliation(s)
- Rui Yang
- School of Electronics and Information Engineering, Changshu Institute of Technology, Changshu 215500, PR China.
| | - Qinyi Gao
- School of Electronics and Information Engineering, Changshu Institute of Technology, Changshu 215500, PR China
| | - Xue Lu
- School of Electronics and Information Engineering, Changshu Institute of Technology, Changshu 215500, PR China
| | - Yukun Wu
- School of Electronics and Information Engineering, Changshu Institute of Technology, Changshu 215500, PR China
| | - Changxin Zhu
- School of Electronics and Information Engineering, Changshu Institute of Technology, Changshu 215500, PR China
| | - Zhida Han
- School of Electronics and Information Engineering, Changshu Institute of Technology, Changshu 215500, PR China.
| | - Chuanya Li
- State Key Laboratory of Solidification Processing, Atomic Control & Catalysis Engineering Laboratory (ACCEL), School of Materials Science and Engineering, Northwestern Polytechnical University, Xi'an 710072, PR China.
| | - Mengmeng Wei
- School of Electronics and Information Engineering, Changshu Institute of Technology, Changshu 215500, PR China.
| |
Collapse
|
2
|
Miao S, Sun J, Li Y, Zhang Q, Chen H, Gao F. Engineered DR/NIR dual-emission carbonized polymer dots for simultaneous tracking of lipid droplets and lysosomes. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 329:125598. [PMID: 39706069 DOI: 10.1016/j.saa.2024.125598] [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: 08/18/2024] [Revised: 11/25/2024] [Accepted: 12/12/2024] [Indexed: 12/23/2024]
Abstract
Developing near-infrared fluorescent probes for simultaneous tracking of lipid droplets (LDs) and lysosomes is highly desirable for studying cell metabolism. In this work, deep-red/near-infrared dual-emission carbonized polymer dots (DN-CPDs) were prepared for ratiometric monitoring of the intracellular polarity. Detailed structural analysis revealed that the deep-red emission and near-infrared peak of DN-CPDs originate from the molecular state and surface state, respectively. The surface-state emission was derived from the intraparticle charge-transfer (ICT) effect of the donor-bridge-acceptor (D-π-A) structure of DN-CPDs. The obtained DN-CPDs exhibited excellent dual-labeling ability, large Stokes shifts, ratiometric polarity sensitivity, high selectivity, and satisfactory photostability. Moreover, with the polarity distinction between LDs and lysosomes, DN-CPDs nanoprobes were successfully used to observe the dynamic changes of the two aforementioned organelles during starvation-induced lipophagy and drug-induced lipophagy inhibition processes. This work not only provides a useful tool for LD-lysosome related studies but is also valuable for the preparation of CPDs with long wavelength emission.
Collapse
Affiliation(s)
- Shan Miao
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Key Laboratory of Chemo/Biosensing, Laboratory of Optical Probes and Bioelectrocatalysis (LOPAB), Anhui Province Key Laboratory of Biomedical Materials and Chemical Measurement, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, PR China
| | - Junyong Sun
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Key Laboratory of Chemo/Biosensing, Laboratory of Optical Probes and Bioelectrocatalysis (LOPAB), Anhui Province Key Laboratory of Biomedical Materials and Chemical Measurement, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, PR China.
| | - Ying Li
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Key Laboratory of Chemo/Biosensing, Laboratory of Optical Probes and Bioelectrocatalysis (LOPAB), Anhui Province Key Laboratory of Biomedical Materials and Chemical Measurement, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, PR China
| | - Qiang Zhang
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs and Institute of Synthesis and Application of Medical Materials, Department of Pharmacy, Wannan Medical College, Wuhu 241002, PR China
| | - Hongqi Chen
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Key Laboratory of Chemo/Biosensing, Laboratory of Optical Probes and Bioelectrocatalysis (LOPAB), Anhui Province Key Laboratory of Biomedical Materials and Chemical Measurement, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, PR China
| | - Feng Gao
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Key Laboratory of Chemo/Biosensing, Laboratory of Optical Probes and Bioelectrocatalysis (LOPAB), Anhui Province Key Laboratory of Biomedical Materials and Chemical Measurement, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, PR China.
| |
Collapse
|
3
|
Harvey LD, Alotaibi M, Tai YY, Tang Y, Kim HJJ, Kelly NJ, Sun W, Woodcock CSC, Arshad S, Culley MK, El Khoury W, Xie R, Al Aaraj Y, Zhao J, Hafeez N, Rao RJ, Jiang S, Negi V, Kirillova A, Perk D, Watson AM, St Croix CM, Stolz DB, Lee JY, Cheng MH, Zhang M, Detmer S, Guzman E, Manan RS, Saggar R, Haley KJ, Waxman AB, Okawa S, Schwantes-An TH, Pauciulo MW, Wang B, Webb A, Chauvet C, Anderson DG, Nichols WC, Desai AA, Lafyatis R, Nouraie SM, Wu H, McDonald JG, Cheng S, Bahar I, Bertero T, Benza RL, Jain M, Chan SY. Lysosomal dysfunction and inflammatory sterol metabolism in pulmonary arterial hypertension. Science 2025; 387:eadn7277. [PMID: 39847635 DOI: 10.1126/science.adn7277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 11/21/2024] [Indexed: 01/25/2025]
Abstract
Vascular inflammation regulates endothelial pathophenotypes, particularly in pulmonary arterial hypertension (PAH). Dysregulated lysosomal activity and cholesterol metabolism activate pathogenic inflammation, but their relevance to PAH is unclear. Nuclear receptor coactivator 7 (NCOA7) deficiency in endothelium produced an oxysterol and bile acid signature through lysosomal dysregulation, promoting endothelial pathophenotypes. This oxysterol signature overlapped with a plasma metabolite signature associated with human PAH mortality. Mice deficient for endothelial Ncoa7 or exposed to an inflammatory bile acid developed worsened PAH. Genetic predisposition to NCOA7 deficiency was driven by single-nucleotide polymorphism rs11154337, which alters endothelial immunoactivation and is associated with human PAH mortality. An NCOA7-activating agent reversed endothelial immunoactivation and rodent PAH. Thus, we established a genetic and metabolic paradigm that links lysosomal biology and oxysterol processes to endothelial inflammation and PAH.
Collapse
Affiliation(s)
- Lloyd D Harvey
- Medical Scientist Training Program, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mona Alotaibi
- Division of Pulmonary and Critical Care Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
| | - Yi-Yin Tai
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ying Tang
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Hee-Jung J Kim
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Neil J Kelly
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Division of Cardiology, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Wei Sun
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Chen-Shan C Woodcock
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sanya Arshad
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Miranda K Culley
- Medical Scientist Training Program, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Wadih El Khoury
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rong Xie
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yassmin Al Aaraj
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jingsi Zhao
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Neha Hafeez
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rashmi J Rao
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Siyi Jiang
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Vinny Negi
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Anna Kirillova
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Dror Perk
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Annie M Watson
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Donna B Stolz
- Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ji Young Lee
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, USA
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
| | - Mary Hongying Cheng
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, USA
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
| | - Manling Zhang
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Division of Cardiology, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Samuel Detmer
- Department of Chemistry, Massachusetts Institute of Technology, Boston, MA, USA
| | - Edward Guzman
- Institute of Medical Engineering and Science, Massachusetts Institute of Technology, Boston, MA, USA
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Rajith S Manan
- Department of Chemical Engineering, Massachusetts Institute of Technology, Boston, MA, USA
| | - Rajan Saggar
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
- Department of Pathology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Kathleen J Haley
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Aaron B Waxman
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Satoshi Okawa
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tae-Hwi Schwantes-An
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN, USA
- Department of Medicine, Indiana University, Indianapolis, IN, USA
| | - Michael W Pauciulo
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Bing Wang
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Amy Webb
- Department of Biomedical Informatics, Ohio State University, Columbus, OH, USA
| | - Caroline Chauvet
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), Sophia-Antipolis, Valbonne, France
| | - Daniel G Anderson
- Institute of Medical Engineering and Science, Massachusetts Institute of Technology, Boston, MA, USA
| | - William C Nichols
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ankit A Desai
- Department of Medicine, Indiana University, Indianapolis, IN, USA
| | - Robert Lafyatis
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - S Mehdi Nouraie
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Haodi Wu
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jeffrey G McDonald
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Susan Cheng
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ivet Bahar
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, USA
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
| | - Thomas Bertero
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), Sophia-Antipolis, Valbonne, France
| | - Raymond L Benza
- Division of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mohit Jain
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
| | - Stephen Y Chan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
4
|
Xie Y, Luo Y, Li W, Zhou Y, Men Y, Liu F, Pan F, Cai L, Jiao Z, Zhang P. A Molecular Logic Gate Enables Unconventional Super-resolution Same-Day Imaging of Lysosome Membrane in Live Cells. Anal Chem 2025; 97:33-37. [PMID: 39722175 PMCID: PMC11740186 DOI: 10.1021/acs.analchem.4c05907] [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: 11/03/2024] [Revised: 12/12/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024]
Abstract
Lysosomes are acidic membrane-bound organelles that aid digestion, excretion, and cell renewal. The lysosomal membranes are essential for maintaining lysosomal functions and cellular homeostasis. In this work, we developed a molecular "NOR" logic gate, SIATFluor-580L, by introducing malachite green into the spirocyclic rhodamine. SIATFluor-580L requires restriction of molecular rotation of the malachite green motif (Input 1, tight membrane structure) and a large amount of H+ ions to convert the spirocyclic rhodamine into the zwitterionic form (Input 2, acidic environment) to produce a fluorescent product (Output), providing a fluorogenic probe to visualize the lysosomal membrane dynamics in living cells with subdiffraction resolution by using HyVolution (also known as Lightning), an unconventional and inexpensive super-resolution imaging approach based on a basic confocal optical system.
Collapse
Affiliation(s)
- Yangzi Xie
- School
of Environment and Civil Engineering, Dongguan
University of Technology, Dongguan 523808, P. R. China
- Guangdong
Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials,
CAS Key Laboratory of Biomedical Imaging Science and System, Institute
of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced
Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| | - Yuan Luo
- Guangdong
Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials,
CAS Key Laboratory of Biomedical Imaging Science and System, Institute
of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced
Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| | - Wen Li
- College
of Biotechnology, Tianjin University of
Science and Technology, Tianjin 300457, P. R. China
| | - Yi Zhou
- Department
of Bioengineering, Imperial College London, London SW7 2AZ, U.K.
| | - Yongfan Men
- Research
Center for Biomedical Optics and Molecular Imaging, Institute of Biomedical
and Health Engineering, Shenzhen Institute
of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| | - Fufeng Liu
- College
of Biotechnology, Tianjin University of
Science and Technology, Tianjin 300457, P. R. China
| | - Fan Pan
- Guangdong
Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials,
CAS Key Laboratory of Biomedical Imaging Science and System, Institute
of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced
Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| | - Lintao Cai
- Guangdong
Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials,
CAS Key Laboratory of Biomedical Imaging Science and System, Institute
of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced
Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, P. R. China
- Sino-Euro
Center of Biomedicine and Health, Luohu, Shenzhen 518024, P. R. China
| | - Zhe Jiao
- School
of Environment and Civil Engineering, Dongguan
University of Technology, Dongguan 523808, P. R. China
| | - Pengfei Zhang
- Guangdong
Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials,
CAS Key Laboratory of Biomedical Imaging Science and System, Institute
of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced
Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| |
Collapse
|
5
|
Lu PS, Sun SC. Mycotoxin toxicity and its alleviation strategy on female mammalian reproduction and fertility. J Adv Res 2025:S2090-1232(25)00041-4. [PMID: 39814223 DOI: 10.1016/j.jare.2025.01.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 12/23/2024] [Accepted: 01/12/2025] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Mycotoxin, a secondary metabolite of fungus, found worldwide and concerning in crops and food, causes multiple acute and chronic toxicities. Its toxic profile includes hepatotoxicity, carcinogenicity, teratogenicity, estrogenicity, immunotoxicity, and neurotoxicity, leading to deleterious impact on human and animal health. Emerging evidence suggests that it adversely affects perinatal health and progeny by its ability to cross placental barriers. AIM OF REVIEW Due to its wide occurrence and potential toxicity on reproductive health, it is essential to understand the mechanisms of mycotoxin-related reproductive toxicity. This review summarizes the toxicities and mechanisms of mycotoxin on maternal and offspring reproduction among mammalian species. Approaches for effective mycotoxin alleviation are also discussed, providing strategies against mycotoxin contamination. KEY SCIENTIFIC CONCEPTS OF REVIEW The profound mycotoxin toxicities in female mammalian reproduction affect follicle assembly, embryo development, and fetus growth, thereby decreasing offspring fertility. Factors from endocrine system such as hypothalamic-pituitary-gonadal axis and gut-ovarian axis, placenta ABC transporters, organelle and cytoskeleton dynamics, cell cycle control, genomic stability, and redox homeostasis are found to be closely related to mycotoxin toxicities. Approaches from physical, chemical, biological, and supplementation of natural antioxidants are discussed for the mycotoxin elimination, while their applications are not widespread. Available ways for mycotoxin and its toxicities alleviation need further study. Since a species-, time-, and dose-specific response might exist in mycotoxin toxicities, more consideration should be given to the protocols for mycotoxin toxicity studies, such as experimental animal models, exposure duration, and dosage. Specific mechanism for mycotoxin, especially form a molecular biology perspective, could be investigated with multi-omics technologies and advanced imaging techniques. Mass spectrometry with algorithms may provide more accurate exposure assessments, and it may be further helpful to identify the high-risk individuals in the future.
Collapse
Affiliation(s)
- Ping-Shuang Lu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Shao-Chen Sun
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China; Key Laboratory of Research On Clinical Molecular Diagnosis for High Incidence Diseases in Western Guangxi of Guangxi Higher Education Institutions, Reproductive Medicine of Guangxi Medical and Health Key Discipline Construction Project, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China.
| |
Collapse
|
6
|
Yaeger JDW, Sengupta S, Walz AL, Morita M, Morgan TK, Vermeer PD, Francis KR. Cholesterol deficiency directs autophagy-dependent secretion of extracellular vesicles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.11.632510. [PMID: 39829772 PMCID: PMC11741461 DOI: 10.1101/2025.01.11.632510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Extracellular vesicle (EV) secretion is an important, though not fully understood, intercellular communication process. Lipid metabolism has been shown to regulate EV activity, though the impact of specific lipid classes is unclear. Through analysis of small EVs (sEVs), we observe aberrant increases in sEV release within genetic models of cholesterol biosynthesis disorders, where cellular cholesterol is diminished. Inhibition of cholesterol synthesis at multiple synthetic steps mimics genetic models in terms of cholesterol reduction and sEVs secreted. Further analyses of sEVs from cholesterol-depleted cells revealed structural deficits and altered surface marker expression, though these sEVs were also more easily internalized by recipient cells. Transmission electron microscopy of cells with impaired cholesterol biosynthesis demonstrated multivesicular and multilamellar structures potentially associated with autophagic defects. We further found autophagic vesicles being redirected toward late endosomes at the expense of autophagolysosomes. Through CRISPR-mediated inhibition of autophagosome formation, we mechanistically determined that release of sEVs after cholesterol depletion is autophagy dependent. We conclude that cholesterol imbalance initiates autophagosome-dependent secretion of sEVs, which may have pathological relevance in diseases of cholesterol disequilibrium.
Collapse
Affiliation(s)
- Jazmine D. W. Yaeger
- Cellular Therapies and Stem Cell Biology Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Sonali Sengupta
- Cellular Therapies and Stem Cell Biology Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Austin L. Walz
- Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Mayu Morita
- Department of Pathology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Terry K. Morgan
- Department of Pathology, Oregon Health & Science University, Portland, OR 97239, USA
- Center for Developmental Health, Oregon Health and Science University, Portland, OR 97239, USA
| | - Paola D. Vermeer
- Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, SD 57104, USA
- Department of Surgery, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD 57105, USA
| | - Kevin R. Francis
- Cellular Therapies and Stem Cell Biology Group, Sanford Research, Sioux Falls, SD 57104, USA
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD 57105, USA
| |
Collapse
|
7
|
Chen Y, Tang S, Hameed MS, Xu X, Wang Q, Chen Q, Yan J, Zhang K, Han X. A bifunctional naphthalimide-based fluorescent probe for imaging lysosomal peroxynitrite and viscosity in living cells and zebrafish. Bioorg Chem 2025; 154:108061. [PMID: 39709734 DOI: 10.1016/j.bioorg.2024.108061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/01/2024] [Accepted: 12/10/2024] [Indexed: 12/24/2024]
Abstract
Peroxynitrite (ONOO-) and viscosity are critical indicators of lysosome functionality, intimately linked to numerous diseases' pathophysiological processes. Hence, creating reliable analytical techniques to observe fluctuations in lysosomal ONOO- and viscosity is highly important. This study presents the development of a novel naphthalimide-based fluorescent probe, Nap-Cy, specifically designed to target lysosomes and simultaneously detect both ONOO- and viscosity. Nap-Cy displayed a near-infrared fluorescence "turn-on" response to viscosity (ranging from 1.0 to 1410 cp) with an approximately 400-fold increase in intensity. At the same time, it functioned as a ratiometric probe with high sensitivity for detecting ONOO-, featuring a quick response time of approximately 10 min, a low detection limit of 42 nM, a broad pH range (5-11), and excellent selectivity for ONOO- over other chemical and biological species. Additionally, Nap-Cy was successfully applied for fluorescence imaging to monitor ONOO- and viscosity variations in SH-SY5Y cells and zebrafish across multiple channels. This research introduces a valuable molecular probe for investigating the biological functions and interactions of ONOO- and viscosity within lysosomes.
Collapse
Affiliation(s)
- Yiliang Chen
- School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan 243032, PR China
| | - Siyuan Tang
- School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan 243032, PR China
| | - Muhammad Salman Hameed
- State Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, Central China Normal University, Wuhan 430079, PR China
| | - Xiaolong Xu
- School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan 243032, PR China
| | - Qi Wang
- School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan 243032, PR China
| | - Qian Chen
- School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan 243032, PR China
| | - Jufen Yan
- School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan 243032, PR China; Maanshan People's Hospital, Ma'anshan 243099, PR China.
| | - Kui Zhang
- School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan 243032, PR China
| | - Xinya Han
- School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan 243032, PR China.
| |
Collapse
|
8
|
Chan SJW, Zhu JY, Yip BRP, Shyamasundar S, Bazan GC. Molecular Design of a Diketopyrrolopyrrole Conjugated Oligoelectrolyte Capable of Imaging Intracellular Vesicle Membrane Dynamics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2411329. [PMID: 39604323 DOI: 10.1002/adma.202411329] [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: 08/01/2024] [Revised: 11/05/2024] [Indexed: 11/29/2024]
Abstract
Conjugated oligoelectrolytes (COEs) are lipid bilayer spanning optical reporters that hold promise for delineating spatiotemporal changes in subcellular compartments. However, their ability to probe a broader range of biological processes remains restricted due to the lack of environmentally-responsive chemical functionalities. Herein, the study reports a novel COE, namely COE-KP, for monitoring spatiotemporal changes in the endolysosomal vesicles. COE-KP features a central diketopyrrolopyrrole functional group in the optically active conjugated core that confers photophysical properties suitable for bioimaging, in particular responding to the presence of hydrogen bonding functionalities within the hydrophobic domain of the lipid bilayer. COE-KP can thus discriminate cells in different growth states through two-photon fluorescence lifetime imaging microscopy (FLIM) with excitation in the NIR-II range. These changes in lifetime most reasonably reflect the degree of water permeability through the membrane and are not linked to notable differences in membrane tension or solvent polarity. Furthermore, using stimulated emission depletion (STED) nanoscopy, it is possible to directly visualize membrane-bound COEs within cellular vesicles. These results illustrate further opportunities for applying COE-based reporters for super-resolution microscopy of organelle membranes, visualization of subcellular membrane morphologies, and imaging long-term changes in membrane properties.
Collapse
Affiliation(s)
- Samuel J W Chan
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
- Institute for Functional Intelligent Materials, National University of Singapore, Singapore, 117544, Singapore
| | - Ji-Yu Zhu
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
- Institute for Functional Intelligent Materials, National University of Singapore, Singapore, 117544, Singapore
| | - Benjamin Rui Peng Yip
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
- Institute for Functional Intelligent Materials, National University of Singapore, Singapore, 117544, Singapore
| | - Sukanya Shyamasundar
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117594, Singapore
| | - Guillermo C Bazan
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
- Institute for Functional Intelligent Materials, National University of Singapore, Singapore, 117544, Singapore
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore, 117585, Singapore
- Institute for Digital Molecular Analytics and Science, Nanyang Technological University, Singapore, 636921, Singapore
| |
Collapse
|
9
|
Xiang L, Lou J, Zhao J, Geng Y, Zhang J, Wu Y, Zhao Y, Tao Z, Li Y, Qi J, Chen J, Yang L, Zhou K. Underlying Mechanism of Lysosomal Membrane Permeabilization in CNS Injury: A Literature Review. Mol Neurobiol 2025; 62:626-642. [PMID: 38888836 DOI: 10.1007/s12035-024-04290-6] [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: 01/27/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024]
Abstract
Lysosomes play a crucial role in various intracellular pathways as their final destination. Various stressors, whether mild or severe, can induce lysosomal membrane permeabilization (LMP), resulting in the release of lysosomal enzymes into the cytoplasm. LMP not only plays a pivotal role in various cellular events but also significantly contributes to programmed cell death (PCD). Previous research has demonstrated the participation of LMP in central nervous system (CNS) injuries, including traumatic brain injury (TBI), spinal cord injury (SCI), subarachnoid hemorrhage (SAH), and hypoxic-ischemic encephalopathy (HIE). However, the mechanisms underlying LMP in CNS injuries are poorly understood. The occurrence of LMP leads to the activation of inflammatory pathways, increased levels of oxidative stress, and PCD. Herein, we present a comprehensive overview of the latest findings regarding LMP and highlight its functions in cellular events and PCDs (lysosome-dependent cell death, apoptosis, pyroptosis, ferroptosis, and autophagy). In addition, we consolidate the most recent insights into LMP in CNS injury by summarizing and exploring the latest advances. We also review potential therapeutic strategies that aim to preserve LMP or inhibit the release of enzymes from lysosomes to alleviate the consequences of LMP in CNS injury. A better understanding of the role that LMP plays in CNS injury may facilitate the development of strategic treatment options for CNS injury.
Collapse
Affiliation(s)
- Linyi Xiang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, 325027, China
| | - Junsheng Lou
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jiayi Zhao
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, 325027, China
| | - Yibo Geng
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, 325027, China
| | - Jiacheng Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, 325027, China
| | - Yuzhe Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, 325027, China
| | - Yinuo Zhao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310000, China
| | - Zhichao Tao
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, 325027, China
| | - Yao Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, 325027, China
| | - Jianjun Qi
- Department of Clinical Laboratory, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, China.
| | - Jiaoxiang Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, 325027, China.
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, 325027, China.
| | - Liangliang Yang
- School of Pharmaceutical Sciences, Wenzhou Medical University, WenzhouZhejiang, 325035, China.
| | - Kailiang Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, 325027, China.
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
10
|
Cao B, Gu S, Shen Z, Zhang Y, Shen Y. Construction of lysosome-related prognostic signature to predict the survival outcomes and selecting suitable drugs for patients with HNSCC. Biofactors 2025; 51:e2140. [PMID: 39495139 DOI: 10.1002/biof.2140] [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: 07/16/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024]
Abstract
Lysosomes are digestive organelles responsible for endocytosis and autophagy. Recently, the malignancy and invasiveness head and neck squamous cell carcinoma (HNSCC) has been increasingly studied with the role of lysosomes. A list of lysosome-related genes were obtained from MSigDB. A Spearman correlation and univariate Cox regression analyses combined with differential expression analysis were conducted to detect differentially expressed lysosome-related genes related to prognosis. The prediction of prognostic signature was evaluated by plotting survival curve, ROC, and by developing a nomogram. Immune subtypes, infiltration of immune cells, GSVA, TIDE, IC50 of common chemotherapy and targeted therapy, GO, and KEGG function enrichment analyses were carried out to explore the immune microenvironment of the signature. We constructed a lysosome-related prognostic signature that could function as an independent prognostic indicator for patients with HNSCC. High-risk patients were better suited to receive Doxorubicin, Mitomycin C, Pyrimethamine, anti-PD-L1 and anti-CTLA-4 immunotherapy, whereas low-risk patients had sensitivity to Lapatinib. GO functional enrichment analysis showed that prognostic features were strongly associated with epidermis-related functions (e.g., epidermal cell differentiation, epidermal development, and keratinization). In addition, a KEGG function enrichment analysis revealed a potential relationship between the risk assessment model and cardiomyopathy. We constructed a prognostic signature based on lysosome-related genes and successfully predicted the survival outcome of HNSCC patients, which not only provides potential guidance for personalized treatment but also provides a new idea for precision treatment of HNSCC.
Collapse
Affiliation(s)
- Bing Cao
- Department of Otorhinolaryngology Head and Neck Surgery, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
| | - Shanshan Gu
- Department of Otorhinolaryngology Head and Neck Surgery, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
| | - Zhisen Shen
- Department of Otorhinolaryngology Head and Neck Surgery, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
| | - Yuna Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
| | - Yiming Shen
- Department of Otology and Skull Base Surgery, National Health Commission Key Laboratory of Hearing Medicine (Fudan University), Shanghai, China
| |
Collapse
|
11
|
Datta D, Navalkar A, Sakunthala A, Paul A, Patel K, Masurkar S, Gadhe L, Manna S, Bhattacharyya A, Sengupta S, Poudyal M, Devi J, Sawner AS, Kadu P, Shaw R, Pandey S, Mukherjee S, Gahlot N, Sengupta K, Maji SK. Nucleo-cytoplasmic environment modulates spatiotemporal p53 phase separation. SCIENCE ADVANCES 2024; 10:eads0427. [PMID: 39661689 PMCID: PMC11633762 DOI: 10.1126/sciadv.ads0427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 11/07/2024] [Indexed: 12/13/2024]
Abstract
Liquid-liquid phase separation of various transcription factors into biomolecular condensates plays an essential role in gene regulation. Here, using cellular models and in vitro studies, we show the spatiotemporal formation and material properties of p53 condensates that might dictate its function. In particular, p53 forms liquid-like condensates in the nucleus of cells, which can bind to DNA and perform transcriptional activity. However, cancer-associated mutations promote misfolding and partially rigidify the p53 condensates with impaired DNA binding ability. Irrespective of wild-type and mutant forms, the partitioning of p53 into cytoplasm leads to the condensate formation, which subsequently undergoes rapid solidification. In vitro studies show that abundant nuclear components such as RNA and nonspecific DNA promote multicomponent phase separation of the p53 core domain and maintain their liquid-like property, whereas specific DNA promotes its dissolution into tetrameric functional p53. This work provides mechanistic insights into how the life cycle and DNA binding properties of p53 might be regulated by phase separation.
Collapse
Affiliation(s)
- Debalina Datta
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Ambuja Navalkar
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Arunima Sakunthala
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
- Sunita Sanghi Centre of Aging and Neurodegenerative Diseases, IIT Bombay, Powai, Mumbai 400076, India
| | - Ajoy Paul
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Komal Patel
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
- Sunita Sanghi Centre of Aging and Neurodegenerative Diseases, IIT Bombay, Powai, Mumbai 400076, India
| | - Shalaka Masurkar
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Laxmikant Gadhe
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
- Sunita Sanghi Centre of Aging and Neurodegenerative Diseases, IIT Bombay, Powai, Mumbai 400076, India
| | - Shouvik Manna
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Arpita Bhattacharyya
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Shinjinee Sengupta
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Manisha Poudyal
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Jyoti Devi
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Ajay Singh Sawner
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Pradeep Kadu
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Ranjit Shaw
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Satyaprakash Pandey
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Semanti Mukherjee
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Nitisha Gahlot
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Kundan Sengupta
- Chromosome Biology Lab, Indian Institute of Science Education and Research, Pune, India
| | - Samir K. Maji
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
- Sunita Sanghi Centre of Aging and Neurodegenerative Diseases, IIT Bombay, Powai, Mumbai 400076, India
| |
Collapse
|
12
|
Wei J, Wu Y, Cheng C, Tan H, Li L, Jiang J. Responses of the mud snail Cipangopaludina cathayensis to thermal stress: Insights from metabolism, oxidative stress damage, and hepatopancreas transcriptional modulation. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2024; 54:101398. [PMID: 39674102 DOI: 10.1016/j.cbd.2024.101398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/08/2024] [Accepted: 12/09/2024] [Indexed: 12/16/2024]
Abstract
Global warming linked to climate change poses a significant risk to aquatic animals. Invertebrates, such as Cipangopaludina cathayensis are especially susceptible to elevated temperature. Understanding how C. cathayensis responds to high-temperature stress is crucial for predicting the putative effects of climate change on its cultivation. In this study, we exposed C. cathayensis to various temperature conditions (26 °C, 28 °C, 30 °C, and 32 °C) for 3 h, revealing that both oxygen consumption and ammonia excretion rates increased gradually with increasing temperature, reaching maximum values of 77.711 ± 2.364 μg·(g·h)-1 and 4.701 ± 0.036 μg·(g·h)-1 at 30 °C and 28 °C, respectively. However, values of these parameters decreased when the culture temperature increased to 32 °C. High-temperature stress also resulted in a reduced O:N ratio and decreased energy metabolism rate. To investigate how high temperature impacts antioxidant activities, immune function, and transcriptional regulation in the hepatopancreas, C. cathayensis were exposed to temperatures of 26 °C or 32 °C for 3 and 7 days, respectively. Our results indicated that high temperature disrupted the antioxidant defense system and led to immunosuppression in the hepatopancreas. Comparative transcriptome analysis identified 6638 genes with significantly altered expression between these two temperature groups. Functional enrichment analysis of differentially expressed genes demonstrated that high temperature affected protein homeostasis, energy metabolism, and immune function of C. cathayensis. Together, these findings offer valuable information for evaluating the impacts of global warming on the culture of mud snail.
Collapse
Affiliation(s)
- Jinyou Wei
- Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection, Guangxi Normal University, Ministry of Education, Guilin 541006, China; College of Life Science, Guangxi Normal University, Guilin 541006, China
| | - Yangyang Wu
- Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection, Guangxi Normal University, Ministry of Education, Guilin 541006, China; College of Life Science, Guangxi Normal University, Guilin 541006, China
| | - Chunxing Cheng
- Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection, Guangxi Normal University, Ministry of Education, Guilin 541006, China; College of Life Science, Guangxi Normal University, Guilin 541006, China
| | - Haizhen Tan
- Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection, Guangxi Normal University, Ministry of Education, Guilin 541006, China; College of Life Science, Guangxi Normal University, Guilin 541006, China
| | - Linli Li
- Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection, Guangxi Normal University, Ministry of Education, Guilin 541006, China; College of Life Science, Guangxi Normal University, Guilin 541006, China
| | - Jiaoyun Jiang
- Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection, Guangxi Normal University, Ministry of Education, Guilin 541006, China; College of Life Science, Guangxi Normal University, Guilin 541006, China; Guangxi Key Laboratory of Rare and Endangered Animal Ecology, Guangxi Normal University, Guilin 541006, China.
| |
Collapse
|
13
|
Sho T, Li Y, Jiao H, Yu L. Migratory autolysosome disposal mitigates lysosome damage. J Cell Biol 2024; 223:e202403195. [PMID: 39347717 PMCID: PMC11457477 DOI: 10.1083/jcb.202403195] [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: 03/30/2024] [Revised: 06/26/2024] [Accepted: 08/14/2024] [Indexed: 10/01/2024] Open
Abstract
Lysosomes, essential for intracellular degradation and recycling, employ damage-control strategies such as lysophagy and membrane repair mechanisms to maintain functionality and cellular homeostasis. Our study unveils migratory autolysosome disposal (MAD), a response to lysosomal damage where cells expel LAMP1-LC3 positive structures via autolysosome exocytosis, requiring autophagy machinery, SNARE proteins, and cell migration. This mechanism, crucial for mitigating lysosomal damage, underscores the role of cell migration in lysosome damage control and facilitates the release of small extracellular vesicles, highlighting the intricate relationship between cell migration, organelle quality control, and extracellular vesicle release.
Collapse
Affiliation(s)
- Takami Sho
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Ying Li
- Cryo-EM Facility of Tsinghua University, Branch of National Protein Science Facility, Tsinghua University, Beijing, China
| | - Haifeng Jiao
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Li Yu
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
14
|
Gao S, Niu YD, Chen L, Chen MF, Bing XL, Hong XY. Transcriptomic landscapes reveal development-related physiological processes in the two-spotted spider mite, Tetranychus urticae. EXPERIMENTAL & APPLIED ACAROLOGY 2024; 93:743-759. [PMID: 39150623 DOI: 10.1007/s10493-024-00956-x] [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: 05/27/2024] [Accepted: 08/06/2024] [Indexed: 08/17/2024]
Abstract
The two-spotted spider mite (Tetranychus urticae Koch, TSSM) is recognized as one of the most problematic spider mite pests. However, the precise gene expression patterns across its key developmental stages remain elusive. Here, we performed a comprehensive transcriptome analysis of TSSM eggs, nymphs and adult females using publicly available RNA sequencing (RNA-seq) data to elucidate the overarching transcriptomic differences between these developmental stages. Principal component analysis and hierarchical clustering analysis unveiled distinct separations among samples across different developmental stages, regardless of their Wolbachia infection status. Differential expression analysis revealed 4,089,2,762, and 1,282 core genes specifically enriched in eggs, nymphs, and adults, respectively. KEGG and GO enrichment analyses showed upregulation of genes in eggs are associated with proteolysis, Wnt signaling pathway, DNA transcription, RNA biosynthetic and metabolic processes, as well as protein folding, sorting, and degradation pathways. Meanwhile, nymphs exhibited increased abundance of genes related to chitin/amino sugar metabolic processes, G protein-coupled receptor signaling pathways, monoatomic ion transport, and neurotransmitter transport pathways. Pathways involving sphingolipid and carbohydrate metabolic processes, proteolysis, lipid transport, and localization were particularly enriched in older females. Altogether, our findings suggest that the egg stage exhibits higher activity in cell differentiation processes, the nymph stage is more involved in chitin development, and the adult stage shows increased metabolic and reproductive activity. This study enhances our understanding of the molecular mechanisms underlying TSSM development and paves the way for further research into the intricate physiological processes of TSSM.
Collapse
Affiliation(s)
- Shuo Gao
- Department of Entomology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yue-Di Niu
- Department of Entomology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Lei Chen
- Department of Entomology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Meng-Fei Chen
- Department of Entomology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xiao-Li Bing
- Department of Entomology, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Xiao-Yue Hong
- Department of Entomology, Nanjing Agricultural University, Nanjing, 210095, China
| |
Collapse
|
15
|
Zhang H, Wang Y, Wang R, Zhang X, Chen H. TRPML1 agonist ML-SA5 mitigates uranium-induced nephrotoxicity via promoting lysosomal exocytosis. Biomed Pharmacother 2024; 181:117728. [PMID: 39647321 DOI: 10.1016/j.biopha.2024.117728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 11/24/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024] Open
Abstract
Uranium (U) released from U mining and spent nuclear fuel reprocessing in the nuclear industry, nuclear accidents and military activities as a primary environmental pollutant (e.g., drinking water pollution) is a threat to human health. Kidney is one of the main target organs for U accumulation, leading to nephrotoxicity mainly associated with the injuries in proximal tubular epithelial cells (PTECs). Transient receptor potential mucolipin 1 (TRPML1) is a novel therapeutic target for nephrotoxicity caused by acute or chronic U poisoning. We herein investigate the therapeutic efficacy of ML-SA5, a small molecule agonist of TRPML1, in U-induced nephrotoxicity in acute U intoxicated mice. We demonstrate that delayed treatment with ML-SA5 enhances U clearance from the kidneys via urine excretion by activating lysosomal exocytosis, and thereby attenuates U-induced kidney dysfunction and cell death/apoptosis of renal PTECs in acute U intoxicated mice. In addition, ML-SA5 promotes the nuclear translocation of transcription factor EB (TFEB) in renal PTECs in acute U intoxicated mice. Mechanistically, ML-SA5 triggers the TRPML1-mediated lysosomal calcium release and consequently induces TFEB activation in U-loaded renal PTECs-derived HK-2 cells. Moreover, knockdown of TRPML1 or TFEB abolishes the effects of ML-SA5 on the removal of intracellular U and reduction of the cellular injury/death in U-loaded HK-2 cells. Our findings indicate that pharmacological activation of TRPML1 is a promising therapeutic approach for the delayed treatment of U-induced nephrotoxicity via the activation of the positive feedback loop of TRPML1 and TFEB and consequent the induction of lysosomal exocytosis.
Collapse
Affiliation(s)
- Hongjing Zhang
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, No. 2094, Xie-Tu Road, Shanghai 200032, PR China
| | - Yifei Wang
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, No. 2094, Xie-Tu Road, Shanghai 200032, PR China
| | - Ruiyun Wang
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, No. 2094, Xie-Tu Road, Shanghai 200032, PR China
| | - Xuxia Zhang
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, No. 2094, Xie-Tu Road, Shanghai 200032, PR China
| | - Honghong Chen
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, No. 2094, Xie-Tu Road, Shanghai 200032, PR China.
| |
Collapse
|
16
|
Živanović M, Selaković M, Pavić A, Selaković Ž, Šolaja B, Santibanez JF, Srdić-Rajić T. Unveiling the 4-aminoquinoline derivatives as potent agents against pancreatic ductal adenocarcinoma (PDAC) cell lines. Chem Biol Interact 2024; 404:111281. [PMID: 39428053 DOI: 10.1016/j.cbi.2024.111281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/04/2024] [Accepted: 10/16/2024] [Indexed: 10/22/2024]
Abstract
Common antimalarials such as artemisinins, chloroquine and their derivatives also possess potent anti-inflamantory, antiviral and anticancer properties. In the search for new therapeutics to combat difficult-to-treat pancreatic carcinomas, we unveiled that 4-aminoquinoline derivatives, with significant antiplasmodial properties and a great safety profile in vivo, have remarkable anticancer activity against pancreatic ductal adenocarcinoma (PDAC) and considerable efficacy in the xenograft model in vivo. The aim of the present study was to further investigate anticancer properties of these compounds in a drug-repurposing manner. The compounds showed profound cytotoxic effects at nanomolar to low micromolar concentration in 2D cultured cells (in vitro) and in the zebrafish PDAC xenograft model (in vivo). A deeper insight into their mechanisms of cytotoxic action showed these compounds induce apoptosis while increasing reactive oxygen species levels along with autophagy inhibition. Additional investigation of the autophagy modulation proved that tested quinoline derivatives cause P62 and LC3-II accumulation in PDAC cells alongside lysosomal alkalinization. Further, in vivo toxicity studies in the zebrafish model showed low toxicity without developmental side effects of the investigated 4-aminoquinolines, while the applied compounds effectively inhibited tumor growth and prevented the metastasis of xenografted pancreatic cells. Taken together, these results highlight the 4-aminoquinolines as privileged structures that ought to be investigated further for potential application in pancreatic carcinoma treatment.
Collapse
Affiliation(s)
- Marija Živanović
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000, Belgrade, Serbia; Department of Molecular Oncology, Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, Dr. Subotića 4, 11129 Belgrade, Serbia
| | - Milica Selaković
- Innovative Centre of the Faculty of Chemistry in Belgrade, ltd., Studentski Trg 12-16, 11158, Belgrade, Serbia.
| | - Aleksandar Pavić
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042, Belgrade, Serbia
| | - Života Selaković
- University of Belgrade - Faculty of Chemistry, Studentski Trg 12-16, 11158, Belgrade, Serbia
| | - Bogdan Šolaja
- Serbian Academy of Sciences and Arts, Knez Mihailova 35, 11158, Belgrade, Serbia
| | - Juan F Santibanez
- Department of Molecular Oncology, Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, Dr. Subotića 4, 11129 Belgrade, Serbia
| | - Tatjana Srdić-Rajić
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000, Belgrade, Serbia.
| |
Collapse
|
17
|
Chen XQ, Yang Q, Chen WM, Chen ZW, Guo GH, Zhang X, Sun XM, Shen T, Xiao FH, Li YF. Dual Role of Lysosome in Cancer Development and Progression. FRONT BIOSCI-LANDMRK 2024; 29:393. [PMID: 39614447 DOI: 10.31083/j.fbl2911393] [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: 07/09/2024] [Revised: 09/22/2024] [Accepted: 09/29/2024] [Indexed: 12/01/2024]
Abstract
Lysosomes are essential intracellular catabolic organelles that contain digestive enzymes involved in the degradation and recycle of damaged proteins, organelles, etc. Thus, they play an important role in various biological processes, including autophagy regulation, ion homeostasis, cell death, cell senescence. A myriad of studies has shown that the dysfunction of lysosome is implicated in human aging and various age-related diseases, including cancer. However, what is noteworthy is that the modulation of lysosome-based signaling and degradation has both the cancer-suppressive and cancer-promotive functions in diverse cancers depending on stage, biology, or tumor microenvironment. This dual role limits their application as targets in cancer therapy. In this review, we provide an overview of lysosome and autophagy-lysosomal pathway and outline their critical roles in many cellular processes, including cell death. We highlight the different functions of autophagy-lysosomal pathway in cancer development and progression, underscoring its potential as a target for effective cancer therapies.
Collapse
Affiliation(s)
- Xiao-Qiong Chen
- Colorectal Surgery, Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, 650000 Kunming, Yunnan, China
| | - Quan Yang
- Colorectal Surgery, Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, 650000 Kunming, Yunnan, China
| | - Wei-Min Chen
- Colorectal Surgery, Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, 650000 Kunming, Yunnan, China
| | - Zi-Wei Chen
- Colorectal Surgery, Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, 650000 Kunming, Yunnan, China
| | - Guang-Hui Guo
- Colorectal Surgery, Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, 650000 Kunming, Yunnan, China
| | - Xuan Zhang
- Colorectal Surgery, Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, 650000 Kunming, Yunnan, China
| | - Xiao-Ming Sun
- Colorectal Surgery, Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, 650000 Kunming, Yunnan, China
| | - Tao Shen
- Colorectal Surgery, Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, 650000 Kunming, Yunnan, China
| | - Fu-Hui Xiao
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 650000 Kunming, Yunnan, China
| | - Yun-Feng Li
- Colorectal Surgery, Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, 650000 Kunming, Yunnan, China
| |
Collapse
|
18
|
Riedel JA, Smolina I, Donat C, Svendheim LH, Farkas J, Hansen BH, Olsvik PA. Into the deep: Exploring the molecular mechanisms of hyperactive behaviour induced by three rare earth elements in early life-stages of the deep-sea scavenging amphipod Tmetonyx cicada (Lysianassidae). THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 952:175968. [PMID: 39226952 DOI: 10.1016/j.scitotenv.2024.175968] [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: 06/29/2024] [Revised: 08/13/2024] [Accepted: 08/30/2024] [Indexed: 09/05/2024]
Abstract
With increasing socio-economic importance of the rare earth elements and yttrium (REY), Norway has laid out plans for REY mining, from land-based to deep-sea mining, thereby enhancing REY mobility in the marine ecosystem. Little is known about associated environmental consequences, especially in the deep ocean. We explored the toxicity and modes of action of a light (Nd), medium (Gd) and heavy (Yb) REY-Cl3 at four concentrations (3, 30, 300, and 3000 μg L-1) in the Arcto-boreal deep-sea amphipod Tmetonyx cicada. At the highest concentration, REY solubility was limited and increased with atomic weight (Nd < Gd < Yb). Lethal effects were practically restricted to this treatment, with the lighter elements being more acutely toxic than Yb (from ∼50 % mortality in the Gd-group at dissolved 689-504 μg L-1 to <20 % in the Yb-group at ca. 2000 μg L-1), which could be a function of bioavailability. All three REY induced hyperactivity at the low-medium concentrations. Delving into the transcriptome of T. cicada allowed us to determine a whole array of potential (neurotoxic) mechanisms underlying this behaviour. Gd induced the vastest response, affecting serotonin-synthesis; sphingolipid-synthesis; the renin-angiotensin system; mitochondrial and endoplasmic reticulum functioning (Gd, Nd); and lysosome integrity (Gd, Yb); as well as the expression of hemocyanin, potentially governing REY-uptake (Gd, Yb). While Nd and Yb shared only few pathways, suggesting a link between mode of action and atomic weight/radius, almost all discussed mechanisms imply the disruption of organismal Ca-homeostasis. Despite only fragmental genomic information available for crustaceans to date, our results provide novel insight into the toxicophysiology of REY in marine biota. The neurotoxic/behavioural effects in T. cicada at concentrations with potential environmental relevance warn about the possibility of bottom-up ecological consequences in mining exposed fjords and deep-sea ecosystems, calling for follow-up studies and regulatory measures prior to the onset of REY mining in Norway.
Collapse
Affiliation(s)
- Juliane Annemieke Riedel
- Faculty of Biosciences and Aquaculture, Nord University, Universitetsalléen 11, 8026 Bodø, Norway.
| | - Irina Smolina
- Faculty of Biosciences and Aquaculture, Nord University, Universitetsalléen 11, 8026 Bodø, Norway
| | - Coline Donat
- IUT de Saint Étienne, Université Jean Monnet, 28 Av. Léon Jouhaux, 42100 Saint-Étienne, France
| | | | - Julia Farkas
- Department of Climate and Environment, SINTEF Ocean, Brattørkaia 17C, 7010 Trondheim, Norway
| | - Bjørn Henrik Hansen
- Department of Climate and Environment, SINTEF Ocean, Brattørkaia 17C, 7010 Trondheim, Norway
| | - Pål Asgeir Olsvik
- Faculty of Biosciences and Aquaculture, Nord University, Universitetsalléen 11, 8026 Bodø, Norway
| |
Collapse
|
19
|
Khuu A, Verreault M, Colin P, Tran H, Idbaih A. Clinical Applications of Antisense Oligonucleotides in Cancer: A Focus on Glioblastoma. Cells 2024; 13:1869. [PMID: 39594617 PMCID: PMC11592788 DOI: 10.3390/cells13221869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
Antisense oligonucleotides (ASOs) are promising drugs capable of modulating the protein expression of virtually any target with high specificity and high affinity through complementary base pairing. However, this requires a deep understanding of the target sequence and significant effort in designing the correct complementary drug. In addition, ASOs have been demonstrated to be well tolerated during their clinical use. Indeed, they are already used in many diseases due to pathogenic RNAs of known sequences and in several neurodegenerative diseases and metabolic diseases, for which they were given marketing authorizations (MAs) in Europe and the United States. Their use in oncology is gaining momentum with several identified targets, promising preclinical and clinical results, and recent market authorizations in the US. However, many challenges remain for their clinical use in cancer. It seems necessary to take a step back and review our knowledge of ASOs and their therapeutic uses in oncology. The objectives of this review are (i) to summarize the current state of the art of ASOs; (ii) to discuss the therapeutic use of ASOs in cancer; and (iii) to focus on ASO usage in glioblastoma, the challenges, and the perspective ahead.
Collapse
Affiliation(s)
- Alexandre Khuu
- AP-HP, Institut du Cerveau, Paris Brain Institute, ICM, Inserm U 1127, CNRS UMR 7225, Hôpitaux Universitaires La Pitié Salpêtrière, Charles Foix, DMU Neurosciences, Service de Neuro-Oncologie-Institut de Neurologie, Sorbonne Université, 75013 Paris, France; (A.K.); (M.V.)
- Institut de Recherche Servier, Rue Francis Perrin, 91190 Gif-sur-Yvette, France;
| | - Maïté Verreault
- AP-HP, Institut du Cerveau, Paris Brain Institute, ICM, Inserm U 1127, CNRS UMR 7225, Hôpitaux Universitaires La Pitié Salpêtrière, Charles Foix, DMU Neurosciences, Service de Neuro-Oncologie-Institut de Neurologie, Sorbonne Université, 75013 Paris, France; (A.K.); (M.V.)
| | - Philippe Colin
- Institut de Recherche Servier, Rue Francis Perrin, 91190 Gif-sur-Yvette, France;
| | - Helene Tran
- Institut de Recherche Servier, Rue Francis Perrin, 91190 Gif-sur-Yvette, France;
| | - Ahmed Idbaih
- AP-HP, Institut du Cerveau, Paris Brain Institute, ICM, Inserm U 1127, CNRS UMR 7225, Hôpitaux Universitaires La Pitié Salpêtrière, Charles Foix, DMU Neurosciences, Service de Neuro-Oncologie-Institut de Neurologie, Sorbonne Université, 75013 Paris, France; (A.K.); (M.V.)
| |
Collapse
|
20
|
Liu Y, Liu Q, Shang H, Li J, Chai H, Wang K, Guo Z, Luo T, Liu S, Liu Y, Wang X, Zhang H, Wu C, Song SJ, Yang J. Potential application of natural compounds in ischaemic stroke: Focusing on the mechanisms underlying "lysosomocentric" dysfunction of the autophagy-lysosomal pathway. Pharmacol Ther 2024; 263:108721. [PMID: 39284368 DOI: 10.1016/j.pharmthera.2024.108721] [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: 03/27/2024] [Revised: 07/06/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
Ischaemic stroke (IS) is the second leading cause of death and a major cause of disability worldwide. Currently, the clinical management of IS still depends on restoring blood flow via pharmacological thrombolysis or mechanical thrombectomy, with accompanying disadvantages of narrow therapeutic time window and risk of haemorrhagic transformation. Thus, novel pathophysiological mechanisms and targeted therapeutic candidates are urgently needed. The autophagy-lysosomal pathway (ALP), as a dynamic cellular lysosome-based degradative process, has been comprehensively studied in recent decades, including its upstream regulatory mechanisms and its role in mediating neuronal fate after IS. Importantly, increasing evidence has shown that IS can lead to lysosomal dysfunction, such as lysosomal membrane permeabilization, impaired lysosomal acidity, lysosomal storage disorder, and dysfunctional lysosomal ion homeostasis, which are involved in the IS-mediated defects in ALP function. There is tightly regulated crosstalk between transcription factor EB (TFEB), mammalian target of rapamycin (mTOR) and lysosomal function, but their relationship remains to be systematically summarized. Notably, a growing body of evidence emphasizes the benefits of naturally derived compounds in the treatment of IS via modulation of ALP function. However, little is known about the roles of natural compounds as modulators of lysosomes in the treatment of IS. Therefore, in this context, we provide an overview of the current understanding of the mechanisms underlying IS-mediated ALP dysfunction, from a lysosomal perspective. We also provide an update on the effect of natural compounds on IS, according to their chemical structural types, in different experimental stroke models, cerebral regions and cell types, with a primary focus on lysosomes and autophagy initiation. This review aims to highlight the therapeutic potential of natural compounds that target lysosomal and ALP function for IS treatment.
Collapse
Affiliation(s)
- Yueyang Liu
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Qingbo Liu
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Hanxiao Shang
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Jichong Li
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - He Chai
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Kaixuan Wang
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Zhenkun Guo
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Tianyu Luo
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Shiqi Liu
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Yan Liu
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Xuemei Wang
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Hangyi Zhang
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Chunfu Wu
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Shao-Jiang Song
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China.
| | - Jingyu Yang
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China.
| |
Collapse
|
21
|
He Y, Fan Y, Ahmadpoor X, Wang Y, Li ZA, Zhu W, Lin H. Targeting lysosomal quality control as a therapeutic strategy against aging and diseases. Med Res Rev 2024; 44:2472-2509. [PMID: 38711187 DOI: 10.1002/med.22047] [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: 08/19/2023] [Revised: 04/04/2024] [Accepted: 04/21/2024] [Indexed: 05/08/2024]
Abstract
Previously, lysosomes were primarily referred to as the digestive organelles and recycling centers within cells. Recent discoveries have expanded the lysosomal functional scope and revealed their critical roles in nutrient sensing, epigenetic regulation, plasma membrane repair, lipid transport, ion homeostasis, and cellular stress response. Lysosomal dysfunction is also found to be associated with aging and several diseases. Therefore, function of macroautophagy, a lysosome-dependent intracellular degradation system, has been identified as one of the updated twelve hallmarks of aging. In this review, we begin by introducing the concept of lysosomal quality control (LQC), which is a cellular machinery that maintains the number, morphology, and function of lysosomes through different processes such as lysosomal biogenesis, reformation, fission, fusion, turnover, lysophagy, exocytosis, and membrane permeabilization and repair. Next, we summarize the results from studies reporting the association between LQC dysregulation and aging/various disorders. Subsequently, we explore the emerging therapeutic strategies that target distinct aspects of LQC for treating diseases and combatting aging. Lastly, we underscore the existing knowledge gap and propose potential avenues for future research.
Collapse
Affiliation(s)
- Yuchen He
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yishu Fan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xenab Ahmadpoor
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yumin Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhong Alan Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, China
| | - Weihong Zhu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Orthopaedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Hang Lin
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
22
|
Venkatesan J, Murugan D, Lakshminarayanan K, Smith AR, Vasanthakumari Thirumalaiswamy H, Kandhasamy H, Zender B, Zheng G, Rangasamy L. Powering up targeted protein degradation through active and passive tumour-targeting strategies: Current and future scopes. Pharmacol Ther 2024; 263:108725. [PMID: 39322067 DOI: 10.1016/j.pharmthera.2024.108725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 08/31/2024] [Accepted: 09/16/2024] [Indexed: 09/27/2024]
Abstract
Targeted protein degradation (TPD) has emerged as a prominent and vital strategy for therapeutic intervention of cancers and other diseases. One such approach involves the exploration of proteolysis targeting chimeras (PROTACs) for the selective elimination of disease-causing proteins through the innate ubiquitin-proteasome pathway. Due to the unprecedented achievements of various PROTAC molecules in clinical trials, researchers have moved towards other physiological protein degradation approaches for the targeted degradation of abnormal proteins, including lysosome-targeting chimeras (LYTACs), autophagy-targeting chimeras (AUTACs), autophagosome-tethering compounds (ATTECs), molecular glue degraders, and other derivatives for their precise mode of action. Despite numerous advantages, these molecules face challenges in solubility, permeability, bioavailability, and potential off-target or on-target off-tissue effects. Thus, an urgent need arises to direct the action of these degrader molecules specifically against cancer cells, leaving the proteins of non-cancerous cells intact. Recent advancements in TPD have led to innovative delivery methods that ensure the degraders are delivered in a cell- or tissue-specific manner to achieve cell/tissue-selective degradation of target proteins. Such receptor-specific active delivery or nano-based passive delivery of the PROTACs could be achieved by conjugating them with targeting ligands (antibodies, aptamers, peptides, or small molecule ligands) or nano-based carriers. These techniques help to achieve precise delivery of PROTAC payloads to the target sites. Notably, the successful entry of a Degrader Antibody Conjugate (DAC), ORM-5029, into a phase 1 clinical trial underscores the therapeutic potential of these conjugates, including LYTAC-antibody conjugates (LACs) and aptamer-based targeted protein degraders. Further, using bispecific antibody-based degraders (AbTACs) and delivering the PROTAC pre-fused with E3 ligases provides a solution for cell type-specific protein degradation. Here, we highlighted the current advancements and challenges associated with developing new tumour-specific protein degrader approaches and summarized their potential as single agents or combination therapeutics for cancer.
Collapse
Affiliation(s)
- Janarthanan Venkatesan
- Department of Chemistry, School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore 632014, India; Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Dhanashree Murugan
- Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, India; School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Kalaiarasu Lakshminarayanan
- Department of Chemistry, School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore 632014, India; Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Alexis R Smith
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA; University of Florida Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
| | - Harashkumar Vasanthakumari Thirumalaiswamy
- Department of Chemistry, School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore 632014, India; Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Hariprasath Kandhasamy
- Department of Chemistry, School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Boutheina Zender
- Department of Biomedical Engineering, Bahçeşehir University, Istanbul 34353, Turkey
| | - Guangrong Zheng
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA; University of Florida Health Cancer Center, University of Florida, Gainesville, FL 32610, USA.
| | - Loganathan Rangasamy
- Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, India.
| |
Collapse
|
23
|
Balkrishna A, Gohel V, Pathak N, Bhattacharya K, Dev R, Varshney A. Livogrit prevents Amiodarone-induced toxicity in experimental model of human liver (HepG2) cells and Caenorhabditis elegans by regulating redox homeostasis. Drug Chem Toxicol 2024; 47:987-1003. [PMID: 38425274 DOI: 10.1080/01480545.2024.2320189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/09/2024] [Accepted: 02/13/2024] [Indexed: 03/02/2024]
Abstract
Treatment with cationic amphiphilic drugs like Amiodarone leads to development of phospholipidosis, a type of lysosomal storage disorder characterized by excessive deposition of phospholipids. Such disorder in liver enhances accumulation of drugs and its metabolites, and dysregulates lipid profiles, which subsequently leads to hepatotoxicity. In the present study, we assessed pharmacological effects of herbal medicine, Livogrit, against hepatic phospholipidosis-induced toxicity. Human liver (HepG2) cells and in vivo model of Caenorhabditis elegans (N2 and CF1553 strains) were used to study effect of Livogrit on Amiodarone-induced phospholipidosis. In HepG2 cells, Livogrit treatment displayed enhanced uptake of acidic pH-based stains and reduced phospholipid accumulation, oxidative stress, AST, ALT, cholesterol levels, and gene expression of SCD-1 and LSS. Protein levels of LPLA2 were also normalized. Livogrit treatment restored Pgp functionality which led to decreased cellular accumulation of Amiodarone as observed by UHPLC analysis. In C. elegans, Livogrit prevented ROS generation, fat-6/7 gene overexpression, and lysosomal trapping of Amiodarone in N2 strain. SOD-3::GFP expression in CF1553 strain normalized by Livogrit treatment. Livogrit regulates phospholipidosis by regulation of redox homeostasis, phospholipid anabolism, and Pgp functionality hindered by lysosomal trapping of Amiodarone. Livogrit could be a potential therapeutic intervention for amelioration of drug-induced phospholipidosis and prevent hepatotoxicity.
Collapse
Affiliation(s)
- Acharya Balkrishna
- Drug Discovery and Development Division, Patanjali Research Foundation, Governed by Patanjali Research Foundation Trust, Haridwar, Uttarakhand, India
- Department of Allied and Applied Sciences, University of Patanjali, Haridwar, Uttarakhand, India
- Patanjali Yog Peeth (UK) Trust, Glasgow, UK
| | - Vivek Gohel
- Drug Discovery and Development Division, Patanjali Research Foundation, Governed by Patanjali Research Foundation Trust, Haridwar, Uttarakhand, India
| | - Nishit Pathak
- Drug Discovery and Development Division, Patanjali Research Foundation, Governed by Patanjali Research Foundation Trust, Haridwar, Uttarakhand, India
| | - Kunal Bhattacharya
- Drug Discovery and Development Division, Patanjali Research Foundation, Governed by Patanjali Research Foundation Trust, Haridwar, Uttarakhand, India
| | - Rishabh Dev
- Drug Discovery and Development Division, Patanjali Research Foundation, Governed by Patanjali Research Foundation Trust, Haridwar, Uttarakhand, India
| | - Anurag Varshney
- Drug Discovery and Development Division, Patanjali Research Foundation, Governed by Patanjali Research Foundation Trust, Haridwar, Uttarakhand, India
- Department of Allied and Applied Sciences, University of Patanjali, Haridwar, Uttarakhand, India
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
24
|
Ye L, Zhao J, Xiao Z, Gu W, Liu X, Ajuyo NMC, Min Y, Pei Y, Wang D. Integrative Human Genetic and Cellular Analysis of the Pathophysiological Roles of AnxA2 in Alzheimer's Disease. Antioxidants (Basel) 2024; 13:1274. [PMID: 39456526 PMCID: PMC11504888 DOI: 10.3390/antiox13101274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/14/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Alzheimer's disease (AD) is an intractable and progressive neurodegenerative disease. Amyloid beta (Aβ) aggregation is the hallmark of AD. Aβ induces neurotoxicity through a variety of mechanisms, including interacting with membrane receptors to alter downstream signaling, damaging cellular or organelle membranes, interfering with protein degradation and synthesis, and inducing an excessive immune-inflammatory response, all of which lead to neuronal death and other pathological changes associated with AD. In this study, we extracted gene expression profiles from the GSE5281 and GSE97760 microarray datasets in the GEO (Gene Expression Omnibus) database, as well as from the Human Gene Database. We identified differentially expressed genes in the brain tissues of AD patients and healthy persons. Through GO, KEGG, and ROC analyses, annexin A2 (AnxA2) was identified as a putative target gene. Notably, accumulating evidence suggests that intracellular AnxA2 is a key regulator in various biological processes, including endocytosis, transmembrane transport, neuroinflammation, and apoptosis. Thus, we conducted a series of cell biology experiments to explore the biological function of AnxA2 in AD. The results indicate that AnxA2 gene knockdown primarily affects oxidative phosphorylation, cell cycle, AD, protein processing in the endoplasmic reticulum, SNARE interactions in vesicular transport, and autophagy. In SH-SY5Y cells secreting Aβ42, AnxA2 gene knockdown exacerbated Aβ42-induced cytotoxicity, including cell death, intracellular ROS levels, and neuronal senescence, altered cell cycle, and reduced ATP levels, suggesting its critical role in mitochondrial function maintenance. AnxA2 gene knockdown also exacerbated the inhibitory effect of Aβ42 on cell migration. AnxA2 overexpression reduced the inflammatory response induced by Aβ42, while its absence increased pro-inflammatory and decreased anti-inflammatory responses. Furthermore, AnxA2 gene knockdown facilitated apoptosis and decreased autophagy. These results indicated potential pathophysiological roles of AnxA2 in AD.
Collapse
Affiliation(s)
- Lianmeng Ye
- Laboratory of Biopharmaceuticals and Molecular Pharmacology, Key Laboratory of Tropical Biological Resources of the Ministry of Education of China, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
- One Health Cooperative Innovation Center, Hainan University, Haikou 570228, China
- Department of Biotechnology, School of Life and Health Sciences, Hainan University, Haikou 570228, China
| | - Jiazheng Zhao
- Laboratory of Biopharmaceuticals and Molecular Pharmacology, Key Laboratory of Tropical Biological Resources of the Ministry of Education of China, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
- One Health Cooperative Innovation Center, Hainan University, Haikou 570228, China
- Department of Biotechnology, School of Life and Health Sciences, Hainan University, Haikou 570228, China
| | - Zhengpan Xiao
- Laboratory of Biopharmaceuticals and Molecular Pharmacology, Key Laboratory of Tropical Biological Resources of the Ministry of Education of China, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
- One Health Cooperative Innovation Center, Hainan University, Haikou 570228, China
- Department of Biotechnology, School of Life and Health Sciences, Hainan University, Haikou 570228, China
| | - Wenyu Gu
- Laboratory of Biopharmaceuticals and Molecular Pharmacology, Key Laboratory of Tropical Biological Resources of the Ministry of Education of China, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
- One Health Cooperative Innovation Center, Hainan University, Haikou 570228, China
- Department of Biotechnology, School of Life and Health Sciences, Hainan University, Haikou 570228, China
| | - Xiaoxuan Liu
- Laboratory of Biopharmaceuticals and Molecular Pharmacology, Key Laboratory of Tropical Biological Resources of the Ministry of Education of China, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
- One Health Cooperative Innovation Center, Hainan University, Haikou 570228, China
- Department of Biotechnology, School of Life and Health Sciences, Hainan University, Haikou 570228, China
| | - Nuela Manka’a Che Ajuyo
- Laboratory of Biopharmaceuticals and Molecular Pharmacology, Key Laboratory of Tropical Biological Resources of the Ministry of Education of China, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
- One Health Cooperative Innovation Center, Hainan University, Haikou 570228, China
| | - Yi Min
- One Health Cooperative Innovation Center, Hainan University, Haikou 570228, China
- Department of Biotechnology, School of Life and Health Sciences, Hainan University, Haikou 570228, China
| | - Yechun Pei
- One Health Cooperative Innovation Center, Hainan University, Haikou 570228, China
- Department of Biotechnology, School of Life and Health Sciences, Hainan University, Haikou 570228, China
| | - Dayong Wang
- Laboratory of Biopharmaceuticals and Molecular Pharmacology, Key Laboratory of Tropical Biological Resources of the Ministry of Education of China, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
- One Health Cooperative Innovation Center, Hainan University, Haikou 570228, China
| |
Collapse
|
25
|
Tantipanjaporn A, Kung KYK, Deng JR, Wong MK. Modular synthesis of pentacyclic-fused pyranoquinoliziniums as organelle-selective fluorescent probes. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 319:124524. [PMID: 38824759 DOI: 10.1016/j.saa.2024.124524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/30/2024] [Accepted: 05/23/2024] [Indexed: 06/04/2024]
Abstract
On basis of their unique chemical and photophysical properties, and excellent biological activities, quinoliziniums have been widely used in various research fields. Herein, modular synthetic strategies for efficient synthesis of novel fluorescent quinoliziniums by using one-pot and stepwise rhodium(III)-catalyzed C-H annulations were developed. In the one-pot synthesis, the reaction between 2-aryl-4-quinolones (1) and 1,2-diarylalkynes (2) proceeded in a chemo- and regioselective manner to give quinolinone-fused isoquinolines (3) and pentacyclic-fused pyranoquinoliziniums (4). The structural diversity of pentacyclic-fused pyranoquinoliziniums (4) was expanded by the stepwise synthesis from 3 and 2, allowing the strategic incorporation of electron-donating (OMe and OH) and electron-withdrawing (Cl) substituents on the top and bottom parts of the pyranoquinoliziniums (4). These newly synthesized pyranoquinoliziniums (4) exhibited tunable absorptions (455-532 nm), emissions (520-610 nm), fluorescence lifetime (0.3-5.6 ns), large Stokes shifts (up to 120 nm), and excellent fluorescence quantum yields (up to 0.73) upon adjusting the different substituents. The the unique arrangement of N and O atoms and extended π-conjugation of 4 could cause the relocation of HOMO comparing with our previous quinoliziniums. Importantly, pyranoquinoliziniums (4a-4g and 4i) targeted the mitochondria, while 4h was localized in lysosome. Due to the remarkable photophysical properties and the potential for organelle targeting of the novel class of quinoliziniums, they could be further applied for biological, chemical and material applications.
Collapse
Affiliation(s)
- Ajcharapan Tantipanjaporn
- State Key Laboratory of Chemical Biology and Drug Discovery, Research Institute for Future Food, Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Ka-Yan Karen Kung
- State Key Laboratory of Chemical Biology and Drug Discovery, Research Institute for Future Food, Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Jie-Ren Deng
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Man-Kin Wong
- State Key Laboratory of Chemical Biology and Drug Discovery, Research Institute for Future Food, Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China.
| |
Collapse
|
26
|
Hao M, Sebag SC, Qian Q, Yang L. Lysosomal physiology and pancreatic lysosomal stress in diabetes mellitus. EGASTROENTEROLOGY 2024; 2:e100096. [PMID: 39512752 PMCID: PMC11542681 DOI: 10.1136/egastro-2024-100096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Endocrine and exocrine functions of the pancreas control nutritional absorption, utilisation and systemic metabolic homeostasis. Under basal conditions, the lysosome is pivotal in regulating intracellular organelles and metabolite turnover. In response to acute or chronic stress, the lysosome senses metabolic flux and inflammatory challenges, thereby initiating the adaptive programme to re-establish cellular homeostasis. A growing body of evidence has demonstrated the pathophysiological relevance of the lysosomal stress response in metabolic diseases in diverse sets of tissues/organs, such as the liver and the heart. In this review, we discuss the pathological relevance of pancreatic lysosome stress in diabetes mellitus. We begin by summarising lysosomal biology, followed by exploring the immune and metabolic functions of lysosomes and finally discussing the interplay between lysosomal stress and the pathogenesis of pancreatic diseases. Ultimately, our review aims to enhance our understanding of lysosomal stress in disease pathogenesis, which could potentially lead to the discovery of innovative treatment methods for these conditions.
Collapse
Affiliation(s)
- Meihua Hao
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Sara C Sebag
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Qingwen Qian
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Ling Yang
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| |
Collapse
|
27
|
Wang P, Ai S, Deng M, Liu Y, Liu Y, He L, Li S. A lysosomal-targeted and viscosity-ultrasensitive near-infrared fluorescent probe for sensing viscosity in cells and a diabetic mice model. Talanta 2024; 278:126506. [PMID: 38968659 DOI: 10.1016/j.talanta.2024.126506] [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: 04/15/2024] [Revised: 05/26/2024] [Accepted: 07/01/2024] [Indexed: 07/07/2024]
Abstract
Diabetes, as a metabolic disorder, has been implicated in organ dysfunction, often correlated with aberrant changes in viscosity. Lysosomal viscosity serves as an indicator of the lysosome's condition and activity, as it always varies synchronously with the change of lysosome's positioning, structure, and internal constituents. Diabetes, a condition within the metabolic disease category, has the potential to disrupt organ function due to irregular changes in viscosity. Therefore, early and precise diagnosis of diabetes is crucial for the prevention and management of diabetic conditions. Understanding the correlation between viscosity variations and lysosomal changes in vivo is vitally important for researching associated diseases. In this study, we developed Lyso-V, a near-infrared (NIR) fluorescent probe targeting lysosomes, with ultrasensitivity to viscosity changes. This probe, designed with a donor-π-bridge-acceptor (D-π-A) structure, exhibits a significant increase in NIR fluorescence intensity (approximately 690 times) when responding to viscosity, due to a twisted intramolecular charge transfer (TICT) mechanism. Furthermore, the probe designed specifically for lysosomes, enables the detection of changes in lysosomal viscosity as well as autophagy processes. Notably, through the application of this probe, we have detected an increased viscosity within the pathological model of the diabetic mouse. Moreover, Lyso-V was employed to measure the viscosity in diabetic mice. Owing to the multifaceted nature of the Lyso-V probe, it is anticipated to act as a practical and potent resource for deepening our understanding of the pathophysiological aspects of diabetes and aiding in its early detection.
Collapse
Affiliation(s)
- Peipei Wang
- Cancer Research Institute, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Siwei Ai
- Cancer Research Institute, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Min Deng
- Cancer Research Institute, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Ying Liu
- Cancer Research Institute, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yin Liu
- Cancer Research Institute, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Longwei He
- Cancer Research Institute, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Songjiao Li
- Cancer Research Institute, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| |
Collapse
|
28
|
Fei S, Xia J, Mehmood N, Wang Y, Feng M, Sun J. Autophagy promotes replication of Bombyx mori Nucleopolyhedrovirus in insect cells. Int J Biol Macromol 2024; 277:134325. [PMID: 39089561 DOI: 10.1016/j.ijbiomac.2024.134325] [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: 03/27/2024] [Revised: 06/21/2024] [Accepted: 07/25/2024] [Indexed: 08/04/2024]
Abstract
BmNPV is a pathogen that infects silkworms exclusively. Although the interaction between BmNPV and the silkworm has been widely noticed and studied, its specific mechanism has still not been elucidated. In this study, we investigated whether BmNPV infection induces the onset of host cell autophagy to enhance viral replication. We observed a significant increase in double- or single-membrane vesicles and an accumulation of enhanced green fluorescent protein eGFP-ATG8 spots in virus-infected cells 72 h after BmNPV infection, accompanied by a conversion of ATG8 to ATG8-PE. In addition, we observed changes in the mitochondrial morphology of BmN cells after BmNPV infection by transmission electron microscopy. By detecting the mitochondrial membrane potential, we found that BmNPV infection resulted in the decrease of mitochondrial membrane potential, and that eGFP-ATG8 was able to co-localise with mitochondria after virus infection of the cells. Moreover, the use of drugs to regulate the occurrence of autophagy affects the replication of cellular BmNPV. Our data demonstrates that BmNPV infection induces host cell autophagy and leads to cellular mitochondrial damage, which in turn may lead to mitochondrial autophagy, and that BmNPV-induced host autophagy promotes its replication in cells. These findings will provide clues for further understanding of host-virus interactions.
Collapse
Affiliation(s)
- Shigang Fei
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Junming Xia
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Nasir Mehmood
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yeyuan Wang
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China.
| | - Min Feng
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China.
| | - Jingchen Sun
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China.
| |
Collapse
|
29
|
You Y, Lin S, Tang C, Li Y, Yan D, Wang D, Chen X. Dual-/multi-organelle-targeted AIE probes associated with oxidative stress for biomedical applications. J Mater Chem B 2024; 12:8812-8824. [PMID: 39150370 DOI: 10.1039/d4tb01440e] [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: 08/17/2024]
Abstract
In situ monitoring of biological processes between different organelles upon oxidative stress is one of the most important research hotspots. Fluorescence imaging is especially suitable for biomedical applications due to its distinct advantages of high spatiotemporal resolution, high sensitivity, non-invasiveness, and in situ monitoring capabilities. However, most fluorescent probes can only achieve light-up imaging of single organelles, thus the combined use of two or more probes is usually required for monitoring biological processes between organelles, which can suffer from tedious staining and washing procedures, increased cytotoxicity and poor photostability. Exogenetic oxidants can affect broad-spectrum subcellular organelles, which are not conducive to in situ monitoring of biological processes between specific organelles. To tackle these challenges, a series of dual-/multi-organelle-targeted aggregation-induced emission (AIE) probes associated with oxidative stress have been designed and developed in the past few years. Herein, the recent progress of these AIE probes is summarized in biomedical applications, such as apoptosis monitoring, interplay between organelles, microenvironmental changes of organelles, organelle morphology tracking, precise cancer therapy, and so forth. Moreover, the further outlook for dual-/multi-organelle-targeted AIE probes is discussed, aiming to promote innovative research in biomedical applications.
Collapse
Affiliation(s)
- Yuanyuan You
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China.
| | - Songling Lin
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China.
| | - Chengwei Tang
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, 523808, China.
| | - Yuchao Li
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, 523808, China.
| | - Dingyuan Yan
- Center for AIE Research, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China
| | - Dong Wang
- Center for AIE Research, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China
| | - Xiaohui Chen
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, 523808, China.
| |
Collapse
|
30
|
Ghorpade M, Soppina V, Kanvah S. Substitution Effects on Subcellular Organelle Localization in Live-cell Imaging. Chembiochem 2024; 25:e202400273. [PMID: 38924297 DOI: 10.1002/cbic.202400273] [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: 03/25/2024] [Revised: 06/24/2024] [Accepted: 06/24/2024] [Indexed: 06/28/2024]
Abstract
A series of D-π-A indole-containing fluorescent probes were developed, followed by an investigation of their photophysical properties and compounds' suitability for subcellular imaging in living cells. We demonstrate that the preference for mitochondrial localization was lost when morpholine was substituted, resulting in the accumulation of the molecule in the lysosomes. However, interestingly, the presence of a nitro group led to their localization within the lipid droplets despite the presence of the morpholine pendant. We also showcase the probes' sensitivity to pH, the influence of added chloroquine, and the temperature response on the changes in fluorescence intensity within lysosomes. The design of the probes with strong intramolecular charge transfer and substantial Stokes shift could facilitate extensive application in various cellular lysosomal models and contribute to a better understanding of the mechanisms involved in stimuli-responsive diseases.
Collapse
Affiliation(s)
- Mohini Ghorpade
- Department of Chemistry, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, 382055
| | - Virupakshi Soppina
- Department of Biological Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, 382055
| | - Sriram Kanvah
- Department of Chemistry, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, 382055
| |
Collapse
|
31
|
Xu J, Gu J, Pei W, Zhang Y, Wang L, Gao J. The role of lysosomal membrane proteins in autophagy and related diseases. FEBS J 2024; 291:3762-3785. [PMID: 37221945 DOI: 10.1111/febs.16820] [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: 02/14/2023] [Revised: 04/12/2023] [Accepted: 05/15/2023] [Indexed: 05/25/2023]
Abstract
As a self-degrading and highly conserved survival mechanism, autophagy plays an important role in maintaining cell survival and recycling. The discovery of autophagy-related (ATG) genes has revolutionized our understanding of autophagy. Lysosomal membrane proteins (LMPs) are important executors of lysosomal function, and increasing evidence has demonstrated their role in the induction and regulation of autophagy. In addition, the functional dysregulation of the process mediated by LMPs at all stages of autophagy is closely related to neurodegenerative diseases and cancer. Here, we review the role of LMPs in autophagy, focusing on their roles in vesicle nucleation, vesicle elongation and completion, the fusion of autophagosomes and lysosomes, and degradation, as well as their broad association with related diseases.
Collapse
Affiliation(s)
- Jiahao Xu
- Department of Endocrinology and Genetic Metabolism, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- Department of Endocrinology and Genetic Metabolism, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- Anhui Province Key Laboratory of Biological Macro-Molecules Research, Wannan Medical College, Wuhu, China
- School of Clinical Medicine, Wannan Medical College, Wuhu, China
| | - Jing Gu
- Department of Endocrinology and Genetic Metabolism, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- Department of Endocrinology and Genetic Metabolism, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- Anhui Province Key Laboratory of Biological Macro-Molecules Research, Wannan Medical College, Wuhu, China
| | - Wenjun Pei
- Anhui Province Key Laboratory of Biological Macro-Molecules Research, Wannan Medical College, Wuhu, China
- Department of Biochemistry and Molecular Biology, Wannan Medical College, Wuhu, China
| | - Yao Zhang
- Anhui Province Key Laboratory of Biological Macro-Molecules Research, Wannan Medical College, Wuhu, China
- Department of Biochemistry and Molecular Biology, Wannan Medical College, Wuhu, China
| | - Lizhuo Wang
- Anhui Province Key Laboratory of Biological Macro-Molecules Research, Wannan Medical College, Wuhu, China
- Department of Biochemistry and Molecular Biology, Wannan Medical College, Wuhu, China
| | - Jialin Gao
- Department of Endocrinology and Genetic Metabolism, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- Department of Endocrinology and Genetic Metabolism, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- Anhui Province Key Laboratory of Biological Macro-Molecules Research, Wannan Medical College, Wuhu, China
- Anhui Provincial College Key Laboratory of Non-coding RNA Transformation Research on Critical Diseases, Wannan Medical College, Wuhu, China
| |
Collapse
|
32
|
Su J, Wang Y, Yao J, Sun L, Zhao C, Liu L, Zhang L. Systemic knockout of Tmem175 results in aberrant differentiation but no effect on hematopoietic reconstitution. Stem Cell Res 2024; 79:103469. [PMID: 38878670 DOI: 10.1016/j.scr.2024.103469] [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: 12/21/2023] [Revised: 06/06/2024] [Accepted: 06/09/2024] [Indexed: 08/04/2024] Open
Abstract
Lysosomes play crucial roles in regulating cell metabolism, and K+ channels are critical for controlling various aspects of lysosomal function. Additionally, lysosomal activity is essential for maintaining the quiescence of hematopoietic stem cells (HSCs) under both steady-state and stress conditions. Tmem175 is a lysosomal potassium channel protein. To further investigate the role of K+ channels in HSCs, our study employed knockout mice to examine the function of Tmem175. Our research findings demonstrate that the deletion of Tmem175 does not disrupt the functionality of HSCs in both stable and stressed conditions, including irradiation and intraperitoneal 5-FU injections. However, we did observe that the absence of Tmem175 impairs the long-term differentiation capacity of HSCs into myeloid differentiated subpopulation cells(In this paper, it is referred to simply as M cells)in HSC transplantation test, while promoting their differentiation into T cells. This suggests that Tmem175 plays a role in the lineage differentiation of HSCs without being essential for their self-renewal or long-term regenerative capabilities.
Collapse
Affiliation(s)
- Jingjing Su
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Shandong Second Medical University, Weifang , 261053, China
| | - Yue Wang
- Center for Metabolic Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Jiyuan Yao
- Center for Metabolic Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Leimin Sun
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
| | - Chunzhen Zhao
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Shandong Second Medical University, Weifang , 261053, China.
| | - Leiming Liu
- Key Laboratory of Artificial Organs and Computational Medicine, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, Zhejiang, 310015, China.
| | - Lingling Zhang
- Center for Metabolic Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China.
| |
Collapse
|
33
|
Domingues N, Catarino S, Cristóvão B, Rodrigues L, Carvalho FA, Sarmento MJ, Zuzarte M, Almeida J, Ribeiro-Rodrigues T, Correia-Rodrigues Â, Fernandes F, Rodrigues-Santos P, Aasen T, Santos NC, Korolchuk VI, Gonçalves T, Milosevic I, Raimundo N, Girão H. Connexin43 promotes exocytosis of damaged lysosomes through actin remodelling. EMBO J 2024; 43:3627-3649. [PMID: 39044100 PMCID: PMC11377567 DOI: 10.1038/s44318-024-00177-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/25/2024] Open
Abstract
A robust and efficient cellular response to lysosomal membrane damage prevents leakage from the lysosome lumen into the cytoplasm. This response is understood to happen through either lysosomal membrane repair or lysophagy. Here we report exocytosis as a third response mechanism to lysosomal damage, which is further potentiated when membrane repair or lysosomal degradation mechanisms are impaired. We show that Connexin43 (Cx43), a protein canonically associated with gap junctions, is recruited from the plasma membrane to damaged lysosomes, promoting their secretion and accelerating cell recovery. The effects of Cx43 on lysosome exocytosis are mediated by a reorganization of the actin cytoskeleton that increases plasma membrane fluidity and decreases cell stiffness. Furthermore, we demonstrate that Cx43 interacts with the actin nucleator Arp2, the activity of which was shown to be necessary for Cx43-mediated actin rearrangement and lysosomal exocytosis following damage. These results define a novel mechanism of lysosomal quality control whereby Cx43-mediated actin remodelling potentiates the secretion of damaged lysosomes.
Collapse
Affiliation(s)
- Neuza Domingues
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
- Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal
| | - Steve Catarino
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Beatriz Cristóvão
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Lisa Rodrigues
- Univ Coimbra, Center for Neurosciences and Cell Biology (CNC), Coimbra, Portugal
| | - Filomena A Carvalho
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Maria João Sarmento
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Mónica Zuzarte
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Jani Almeida
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
- Univ Coimbra, Center for Neurosciences and Cell Biology (CNC), Coimbra, Portugal
| | - Teresa Ribeiro-Rodrigues
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Ânia Correia-Rodrigues
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Fábio Fernandes
- Institute for Bioengineering and Biosciences (IBB) and Associate Laboratory i4HB-Institute for Health and Bioeconomy, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Paulo Rodrigues-Santos
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
- Univ Coimbra, Center for Neurosciences and Cell Biology (CNC), Coimbra, Portugal
| | - Trond Aasen
- Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | - Nuno C Santos
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Viktor I Korolchuk
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Teresa Gonçalves
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Univ Coimbra, Center for Neurosciences and Cell Biology (CNC), Coimbra, Portugal
| | - Ira Milosevic
- Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal
- University of Oxford, Centre for Human Genetics, Nuffield Department of Medicine, Oxford, UK
| | - Nuno Raimundo
- Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, USA
| | - Henrique Girão
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal.
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal.
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal.
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal.
| |
Collapse
|
34
|
Guo L, Li P, Jing Z, Gong Y, Lai K, Fu H, Dong H, Yang Z, Liu Z. Iminoamido chelated iridium(III) and ruthenium(II) anticancer complexes with mitochondria-targeting ability and potential to overcome cisplatin resistance. J Inorg Biochem 2024; 258:112631. [PMID: 38843774 DOI: 10.1016/j.jinorgbio.2024.112631] [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: 03/03/2024] [Revised: 05/27/2024] [Accepted: 05/31/2024] [Indexed: 07/01/2024]
Abstract
A diverse set of neutral half-sandwich iminoamido iridium and ruthenium organometallic complexes is synthesized through the utilization of Schiff base pro-ligands with N˄N donors. Notably, these metal complexes with varying leaving groups (Cl- or OAc-) are formed by employing different quantities of the deprotonating agent NaOAc, and exhibit promising cytotoxicity against various cancer cell lines such as A549 and cisplatin-resistant A549/DDP lung cancer cells, as well as HeLa cells, with IC50 values spanning from 9.26 to 15.98 μM. Cytotoxicity and anticancer selectivity (SI: 1.9-2.4) of these metal complexes remain unaffected by variations in the metal center, leaving group, and ligand substitution. Further investigations reveal that these metal complexes specifically target mitochondria, leading to the depolarization of the mitochondrial membrane and instigating the production of intracellular reactive oxygen species. Furthermore, the metal complexes are found to induce late apoptosis and disrupt the cell cycle, leading to G2/M cell cycle arrest specifically in A549 cancer cells. In light of these findings, it is evident that the primary mechanism contributing to the anticancer effectiveness of these metal complexes is the redox pathway.
Collapse
Affiliation(s)
- Lihua Guo
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, PR China.
| | - Pengwei Li
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, PR China
| | - Zhihong Jing
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, PR China
| | - Yuwen Gong
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, PR China
| | - Kangning Lai
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, PR China
| | - Hanxiu Fu
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, PR China
| | - Heqian Dong
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, PR China
| | - Zhihao Yang
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, PR China
| | - Zhe Liu
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, PR China.
| |
Collapse
|
35
|
Shim G, Youn YS. Precise subcellular targeting approaches for organelle-related disorders. Adv Drug Deliv Rev 2024; 212:115411. [PMID: 39032657 DOI: 10.1016/j.addr.2024.115411] [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: 04/23/2024] [Revised: 06/14/2024] [Accepted: 07/14/2024] [Indexed: 07/23/2024]
Abstract
Pharmacological research has expanded to the nanoscale level with advanced imaging technologies, enabling the analysis of drug distribution at the cellular organelle level. These advances in research techniques have contributed to the targeting of cellular organelles to address the fundamental causes of diseases. Beyond navigating the hurdles of reaching lesion tissues upon administration and identifying target cells within these tissues, controlling drug accumulation at the organelle level is the most refined method of disease management. This approach opens new avenues for the development of more potent therapeutic strategies by delving into the intricate roles and interplay of cellular organelles. Thus, organelle-targeted approaches help overcome the limitations of conventional therapies by precisely regulating functionally compartmentalized spaces based on their environment. This review discusses the basic concepts of organelle targeting research and proposes strategies to target diseases arising from organelle dysfunction. We also address the current challenges faced by organelle targeting and explore future research directions.
Collapse
Affiliation(s)
- Gayong Shim
- School of Systems Biomedical Science and Integrative Institute of Basic Sciences, Soongsil University, Seoul 06978, Republic of Korea
| | - Yu Seok Youn
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea.
| |
Collapse
|
36
|
Mai LD, Wimberley SC, Champion JA. Intracellular delivery strategies using membrane-interacting peptides and proteins. NANOSCALE 2024; 16:15465-15480. [PMID: 39091235 PMCID: PMC11340348 DOI: 10.1039/d4nr02093f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024]
Abstract
While the cellular cytosol and organelles contain attractive targets for disease treatments, it remains a challenge to deliver therapeutic biomacromolecules to these sites. This is due to the selective permeability of the plasma and endosomal membranes, especially for large and hydrophilic therapeutic cargos such as proteins and nucleic acids. In response, many different delivery systems and molecules have been devised to help therapeutics cross these barriers to reach cytosolic targets. Among them are peptide and protein-based systems, which have several advantages over other natural and synthetic materials including their ability to interact with cell membranes. In this review, we will describe recent advances and current challenges of peptide and protein strategies that leverage cell membrane association and modulation to enable cytosolic delivery of biomacromolecule cargo. The approaches covered here include peptides and proteins derived from or inspired by natural sequences as well as those designed de novo for delivery function.
Collapse
Affiliation(s)
- Linh D Mai
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Dr NW, Atlanta, GA, 30332-2000, USA.
| | - Sydney C Wimberley
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Dr NW, Atlanta, GA, 30332-2000, USA.
- BioEngineering Program, Georgia Institute of Technology, USA
| | - Julie A Champion
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Dr NW, Atlanta, GA, 30332-2000, USA.
- BioEngineering Program, Georgia Institute of Technology, USA
| |
Collapse
|
37
|
Shang J, Ma Y, Liu X, Sun S, Pang X, Zhou R, Huan S, He Y, Xiong B, Zhang XB. Single-particle rotational microrheology enables pathological staging of macrophage foaming and antiatherosclerotic studies. Proc Natl Acad Sci U S A 2024; 121:e2403740121. [PMID: 39102540 PMCID: PMC11331104 DOI: 10.1073/pnas.2403740121] [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: 02/22/2024] [Accepted: 07/01/2024] [Indexed: 08/07/2024] Open
Abstract
The formation of macrophage-derived foam cells has been recognized as the pathological hallmark of atherosclerotic diseases. However, the pathological evolution dynamics and underlying regulatory mechanisms remain largely unknown. Herein, we introduce a single-particle rotational microrheology method for pathological staging of macrophage foaming and antiatherosclerotic explorations by probing the dynamic changes of lysosomal viscous feature over the pathological evolution progression. The principle of this method involves continuous monitoring of out-of-plane rotation-caused scattering brightness fluctuations of the gold nanorod (AuNR) probe-based microrheometer and subsequent determination of rotational relaxation time to analyze the viscous feature in macrophage lysosomes. With this method, we demonstrated the lysosomal viscous feature as a robust pathological reporter and uncovered three distinct pathological stages underlying the evolution dynamics, which are highly correlated with a pathological stage-dependent activation of the NLRP3 inflammasome-involved positive feedback loop. We also validated the potential of this positive feedback loop as a promising therapeutic target and revealed the time window-dependent efficacy of NLRP3 inflammasome-targeted drugs against atherosclerotic diseases. To our knowledge, the pathological staging of macrophage foaming and the pathological stage-dependent activation of the NLRP3 inflammasome-involved positive feedback mechanism have not yet been reported. These findings provide insights into in-depth understanding of evolutionary features and regulatory mechanisms of macrophage foaming, which can benefit the analysis of effective therapeutical drugs as well as the time window of drug treatment against atherosclerotic diseases in preclinical studies.
Collapse
Affiliation(s)
- Jinhui Shang
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha410082, China
| | - Yuan Ma
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha410082, China
| | - Xixuan Liu
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha410082, China
| | - Shijie Sun
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha410082, China
| | - Xiayun Pang
- State Key Laboratory of Research and Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi University of Chinese Medicine, Xianyang712083, China
| | - Rui Zhou
- State Key Laboratory of Research and Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi University of Chinese Medicine, Xianyang712083, China
| | - Shuangyan Huan
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha410082, China
| | - Yan He
- Department of Chemistry, Tsinghua University, Beijing100084, China
| | - Bin Xiong
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha410082, China
| | - Xiao-Bing Zhang
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha410082, China
| |
Collapse
|
38
|
Wang PX, Wu SL, Ju JQ, Jiao L, Zou YJ, Zhang KH, Sun SC, Hu LL, Zheng XB. Benzo[a]pyrene exposure disrupts the organelle distribution and function of mouse oocytes. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 281:116630. [PMID: 38917590 DOI: 10.1016/j.ecoenv.2024.116630] [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: 02/29/2024] [Revised: 04/22/2024] [Accepted: 06/19/2024] [Indexed: 06/27/2024]
Abstract
Benzo[a]pyrene (BaP) is a polycyclic aromatic hydrocarbon compound that is generated during combustion processes, and is present in various substances such as foods, tobacco smoke, and burning emissions. BaP is extensively acknowledged as a highly carcinogenic substance to induce multiple forms of cancer, such as lung cancer, skin cancer, and stomach cancer. Recently it is shown to adversely affect the reproductive system. Nevertheless, the potential toxicity of BaP on oocyte quality remains unclear. In this study, we established a BaP exposure model via mouse oral gavage and found that BaP exposure resulted in a notable decrease in the ovarian weight, number of GV oocytes in ovarian, and oocyte maturation competence. BaP exposure caused ribosomal dysfunction, characterized by a decrease in the expression of RPS3 and HPG in oocytes. BaP exposure also caused abnormal distribution of the endoplasmic reticulum (ER) and induced ER stress, as indicated by increased expression of GRP78. Besides, the Golgi apparatus exhibited an abnormal localization pattern, which was confirmed by the GM130 localization. Disruption of vesicle transport processes was observed by the abnormal expression and localization of Rab10. Additionally, an enhanced lysosome and LC3 fluorescence intensity indicated the occurrence of protein degradation in oocytes. In summary, our results suggested that BaP exposure disrupted the distribution and functioning of organelles, consequently affecting the developmental competence of mouse oocytes.
Collapse
Affiliation(s)
- Peng-Xia Wang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Guangxi University, Nanning, 530004, China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Guangxi University, Nanning 530004, China
| | - Si-Le Wu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jia-Qian Ju
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Le Jiao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yuan-Jing Zou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Kun-Huan Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Shao-Chen Sun
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Lin-Lin Hu
- Key Laboratory of Research on Clinical Molecular Diagnosis for High Incidence Diseases in Western Guangxi, Reproductive Medicine of Guangxi Medical and Health Key Discipline Construction Project, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China.
| | - Xi-Bang Zheng
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Guangxi University, Nanning, 530004, China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Guangxi University, Nanning 530004, China.
| |
Collapse
|
39
|
Zou GY, Bi F, Chen S, Liu MX, Yu YL. Ternary recognition fluorescent probe for lysosome acidification counter-ion studies via Cl -, K +, and pH. Chem Commun (Camb) 2024; 60:8087-8090. [PMID: 38990605 DOI: 10.1039/d4cc02800g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
Lysosomal acidity relies on H+ inflow, which requires counter-ion flows (Cl- and K+) to balance charge. A lysosome targeting ternary recognition fluorescent probe for Cl-, K+, and pH was developed for lysosome acidification counter-ion research. The probe was used to study counter-ion changes when the Cl- channel was blocked and under oxidative pressure.
Collapse
Affiliation(s)
- Guang-Yue Zou
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China.
| | - Fan Bi
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China.
| | - Shuai Chen
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China.
- Foshan Graduate School of Innovation, Northeastern University, Foshan 528311, China
| | - Meng-Xian Liu
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai 980-8577, Miyagi, Japan.
| | - Yong-Liang Yu
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China.
| |
Collapse
|
40
|
Park J, Kim J, Park H, Kim T, Lee S. ESCRT-III: a versatile membrane remodeling machinery and its implications in cellular processes and diseases. Anim Cells Syst (Seoul) 2024; 28:367-380. [PMID: 39070887 PMCID: PMC11275535 DOI: 10.1080/19768354.2024.2380294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 07/03/2024] [Indexed: 07/30/2024] Open
Abstract
The endosomal sorting complexes required for transport (ESCRT) machinery is an evolutionarily conserved cytosolic protein complex that plays a crucial role in membrane remodeling and scission events across eukaryotes. Initially discovered for its function in multivesicular body (MVB) formation, the ESCRT complex has since been implicated in a wide range of membrane-associated processes, including endocytosis, exocytosis, cytokinesis, and autophagy. Recent advances have elucidated the ESCRT assembly pathway and highlighted the distinct functions of the various ESCRT complexes and their associated partners. Among the ESCRT complexes, ESCRT-III stands out as a critical player in membrane remodeling, with its subunits assembled into higher-order multimers capable of bending and severing membranes. This review focuses on the ESCRT-III complex, exploring its diverse functions in cellular processes beyond MVB biogenesis. We delve into the molecular mechanisms underlying ESCRT-III-mediated membrane remodeling and highlight its emerging roles in processes such as viral budding, autophagosome closure, and cytokinetic abscission. We also discuss the implications of ESCRT-III dysregulation in neurodegenerative diseases. The versatile membrane remodeling capabilities of ESCRT-III across diverse cellular processes underscore its importance in maintaining proper cellular function. Furthermore, we highlight the promising potential of ESCRT-III as a therapeutic target for neurodegenerative diseases, offering insights into the treatments of the diseases and the technical applications in related research fields.
Collapse
Affiliation(s)
- Jisoo Park
- Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea
| | - Jongyoon Kim
- Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea
| | - Hyungsun Park
- Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea
| | - Taewan Kim
- Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea
| | - Seongju Lee
- Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea
- Department of Anatomy, College of Medicine, Inha University, Incheon, Republic of Korea
| |
Collapse
|
41
|
Kofuji S, Wolfe K, Sumita K, Kageyama S, Yoshino H, Hirota Y, Ogawa-Iio A, Kanoh H, Sasaki M, Kofuji K, Davis MI, Pragani R, Shen M, Boxer MB, Nakatsu F, Nigorikawa K, Sasaki T, Takeuchi K, Senda T, Kim SM, Edinger AL, Simeonov A, Sasaki AT. A high dose KRP203 induces cytoplasmic vacuoles associated with altered phosphoinositide segregation and endosome expansion. Biochem Biophys Res Commun 2024; 718:149981. [PMID: 38735134 PMCID: PMC11416131 DOI: 10.1016/j.bbrc.2024.149981] [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: 04/09/2024] [Accepted: 04/22/2024] [Indexed: 05/14/2024]
Abstract
In animal cells, vacuoles are absent, but can be induced by diseases and drugs. While phosphoinositides are critical for membrane trafficking, their role in the formation of these vacuoles remains unclear. The immunosuppressive KRP203/Mocravimod, which antagonizes sphingosine-1-phosphate receptors, has been identified as having novel multimodal activity against phosphoinositide kinases. However, the impact of this novel KRP203 activity is unknown. Here, we show that KRP203 disrupts the spatial organization of phosphoinositides and induces extensive vacuolization in tumor cells and immortalized fibroblasts. The KRP203-induced vacuoles are primarily from endosomes, and augmented by inhibition of PIKFYVE and VPS34. Conversely, overexpression of PTEN decreased KRP203-induced vacuole formation. Furthermore, V-ATPase inhibition completely blunted KRP203-induced vacuolization, pointing to a critical requirement of the endosomal maturation process. Importantly, nearly a half of KRP203-induced vacuoles are significantly decorated with PI4P, a phosphoinositide typically enriched at the plasma membrane and Golgi. These results suggest a model that noncanonical spatial reorganization of phosphoinositides by KRP203 alters the endosomal maturation process, leading to vacuolization. Taken together, this study reveals a previously unrecognized bioactivity of KRP203 as a vacuole-inducing agent and its unique mechanism of phosphoinositide modulation, providing a new insight of phosphoinositide regulation into vacuolization-associated diseases and their molecular pathologies.
Collapse
Affiliation(s)
- Satoshi Kofuji
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA; Department of Developmental and Regenerative Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan
| | - Kara Wolfe
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Kazutaka Sumita
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA; Department of Endovascular Surgery, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan
| | - Shun Kageyama
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, 997-0052, Japan
| | - Hirofumi Yoshino
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Yoshihisa Hirota
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA; Department of Bioscience and Engineering, College of Systems Engineering and Science, Shibaura Institute of Technology, Minuma-ku, Saitama, 337-8570, Japan
| | - Aki Ogawa-Iio
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA; Department of Bioscience and Engineering, College of Systems Engineering and Science, Shibaura Institute of Technology, Minuma-ku, Saitama, 337-8570, Japan
| | - Hirotaka Kanoh
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, 997-0052, Japan
| | - Mika Sasaki
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Kaori Kofuji
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Mindy I Davis
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Rajan Pragani
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Min Shen
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Matthew B Boxer
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Fubito Nakatsu
- Department of Neurochemistry and Molecular Cell Biology, Niigata University School of Medicine and Graduate School of Medical/Dental Sciences, Niigata, Japan
| | - Kiyomi Nigorikawa
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takehiko Sasaki
- Department of Biochemical Pathophysiology, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan
| | - Koh Takeuchi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Toshiya Senda
- Structural Biology Research Center, High Energy Accelerator Research Organization (KEK), Tsukuba, Ibaraki, 305-0801, Japan; Department of Accelerator Science, SOKENDAI, Japan; Faculty of Pure and Applied Sciences, University of Tsukuba, Ibaraki, 305-8572, Japan
| | - Seong M Kim
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California Irvine, California, 92697, USA
| | - Aimee L Edinger
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California Irvine, California, 92697, USA
| | - Anton Simeonov
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Atsuo T Sasaki
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA; Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, 997-0052, Japan; Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA; Department of Neurosurgery, Brain Tumor Center at UC Gardner Neuroscience Institute, Cincinnati, OH, 45267, USA; Department of Clinical and Molecular Genetics, Hiroshima University Hospital, Hiroshima, 734-8551, Japan.
| |
Collapse
|
42
|
Chakraborty S, Bindra AK, Thomas A, Zhao Y, Ajayaghosh A. pH-Assisted multichannel heat shock monitoring in the endoplasmic reticulum with a pyridinium fluorophore. Chem Sci 2024; 15:10851-10857. [PMID: 39027278 PMCID: PMC11253182 DOI: 10.1039/d4sc01977f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/27/2024] [Indexed: 07/20/2024] Open
Abstract
Heat shock is a global health concern as it causes permanent damage to living cells and has a relatively high mortality rate. Therefore, diagnostic tools that facilitate a better understanding of heat shock damage and the defense mechanism at the sub-cellular level are of great importance. In this report, we have demonstrated the use of a pyridinium-based fluorescent molecule, PM-ER-OH, as a 'multichannel' imaging probe to monitor the pH change associated with a heat shock in the endoplasmic reticulum. Among the three pyridinium derivatives synthesized, PM-ER-OH was chosen for study due to its excellent biocompatibility, good localization in the endoplasmic reticulum, and intracellular pH response signaled by a yellow fluorescence (λ max = 556 nm) at acidic pH and a far red fluorescence (λ max = 660 nm) at basic pH. By changing the excitation wavelength, we could modulate the fluorescence signal in 'turn-ON', single excitation ratiometric and 'turn-OFF' modes, making the fluorophore a 'multichannel' probe for both ex vitro and in vitro pH monitoring in the endoplasmic reticulum. The probe could efficiently monitor the pH change when heat shock was applied to cells either directly or in a pre-heated manner, which gives insight on cellular acidification caused by heat stress.
Collapse
Affiliation(s)
- Sandip Chakraborty
- CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST) Thiruvananthapuram 695 019 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Anivind Kaur Bindra
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University 21 Nanyang Link 637371 Singapore
| | - Anagha Thomas
- CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST) Thiruvananthapuram 695 019 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Yanli Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University 21 Nanyang Link 637371 Singapore
| | - Ayyappanpillai Ajayaghosh
- CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST) Thiruvananthapuram 695 019 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
- Department of Chemistry, SRM Institute of Science and Technology Chennai 603203 India
| |
Collapse
|
43
|
Li Z, Lu J, Li X. Recent Progress in Thermally Activated Delayed Fluorescence Photosensitizers for Photodynamic Therapy. Chemistry 2024; 30:e202401001. [PMID: 38742479 DOI: 10.1002/chem.202401001] [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: 03/11/2024] [Revised: 05/04/2024] [Accepted: 05/13/2024] [Indexed: 05/16/2024]
Abstract
Photodynamic therapy (PDT) is a rapidly growing discipline that is expected to become an encouraging noninvasive therapeutic strategy for cancer treatment. In the PDT process, an efficient intersystem crossing (ISC) process for photosensitizers from the singlet excited state (S1) to the triplet excited state (T1) is critical for the formation of cytotoxic reactive oxygen species and improvement of PDT performance. Thermally activated delayed fluorescence (TADF) molecules featuring an extremely small singlet-triplet energy gap and an efficient ISC process represent an enormous breakthrough for the PDT process. Consequently, the development of advanced TADF photosensitizers has become increasingly crucial and pressing. The most recent developments in TADF photosensitizers aimed at enhancing PDT efficiency for bio-applications are presented in this review. TADF photosensitizers with water dispersibility, targeting ability, activatable ability, and two-photon excitation properties are highlighted. Furthermore, the future challenges and perspectives of TADF photosensitizers in PDT are proposed.
Collapse
Affiliation(s)
- Ziqi Li
- State Key Laboratory of Clean and Efficient Coal Utilization, Tai Yuan, Taiyuan University of Technology, Taiyuan, 030024, P.R. China
| | - Jianjun Lu
- State Key Laboratory of Clean and Efficient Coal Utilization, Tai Yuan, Taiyuan University of Technology, Taiyuan, 030024, P.R. China
| | - Xuping Li
- State Key Laboratory of Clean and Efficient Coal Utilization, Tai Yuan, Taiyuan University of Technology, Taiyuan, 030024, P.R. China
- College of Energy Materials and Chemistry, Inner Mongolia University, Hohhot, 010020, P.R. China
| |
Collapse
|
44
|
Liu Y, Gong XT, Wang KN, He S, Wang Y, Lin Q, Liu Z, Yu X, Liu B. Dual-targeted fluorescent probe for tracking polarity and phase transition processes during lipophagy. MATERIALS HORIZONS 2024; 11:3287-3297. [PMID: 38842407 DOI: 10.1039/d4mh00190g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Eukaryotic cells regulate various cellular processes through membrane-bound and membrane-less organelles, enabling active signal communication and material exchange. Lysosomes and lipid droplets are representative organelles, contributing to cell lipophagy when their interaction and metabolism are disrupted. Our limited understanding of the interacting behaviours and physicochemical properties of different organelles during lipophagy hinders accurate diagnosis and treatment of related diseases. In this contribution, we report a fluorescent probe, PTZ, engineered for dual-targeting of lipid droplets and lysosomes. PTZ can track liquid-liquid phase separation and respond to polarity shifts through ratiometric fluorescence emission, elucidating the lipophagy process from the perspective of organelle behavior and physicochemical properties. Leveraging on the multifunctionality of PTZ, we have successfully tracked the polarity and dynamic changes of lysosomes and lipid droplets during lipophagy. Furthermore, an unknown homogeneous transition of lipid droplets and lysosomes was discovered, which provided a new perspective for understanding lipophagy processes. And this work is expected to serve as a reference for diagnosis and treatment of lipophagy-related diseases.
Collapse
Affiliation(s)
- Yang Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan 250100, China.
| | - Xiao-Ting Gong
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore.
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Binhai New City, Fuzhou 350207, China
| | - Kang-Nan Wang
- State Key Laboratory of Crystal Materials, Shandong University, Jinan 250100, China.
| | - Simeng He
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Yumeng Wang
- State Key Laboratory of Crystal Materials, Shandong University, Jinan 250100, China.
| | - Qiaowen Lin
- State Key Laboratory of Crystal Materials, Shandong University, Jinan 250100, China.
| | - Zhiqiang Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan 250100, China.
| | - Xiaoqiang Yu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan 250100, China.
| | - Bin Liu
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore.
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Binhai New City, Fuzhou 350207, China
| |
Collapse
|
45
|
Li T, Dai C, Lu Q, Tian M. A polarity-responsive lysosomes-nucleus translocation probe for the dual-emissive visualization of cell apoptosis. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 315:124272. [PMID: 38603960 DOI: 10.1016/j.saa.2024.124272] [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: 02/20/2024] [Revised: 04/01/2024] [Accepted: 04/08/2024] [Indexed: 04/13/2024]
Abstract
Visualization of cell apoptosis is a critical task playing central roles in the fundamental studies in biology, pathology, and biomedicine. Dual-emissive fluorescent probes are desired molecular tools for study on apoptosis, which however were rarely reported. Herein, utilizing the polarity differences between lysosomes and nucleus, a translocation type of fluorescent probe (NA-S) was developed for the dual-color visualization of cell apoptosis. NA-S was designed to be polarity sensitive, bearing alkalescence group, and with DNA affinity. In living cells, NA-S targeted the lysosomes to give blue fluorescence, which translocated into the nucleus during cell apoptosis to give green emission. Thereby, the cell apoptosis could be visualized with NA-S in dual-emissive manner. With the unique probe, the cell apoptosis induced by oxidative stress, UV irradiation, rotenone, colchicine, and paclitaxel have been successfully visualized.
Collapse
Affiliation(s)
- Tianyu Li
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong 250022, China
| | - Chun Dai
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong 250022, China
| | - Qingqing Lu
- Engineering & Technology Center of Electrochemistry, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
| | - Minggang Tian
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong 250022, China.
| |
Collapse
|
46
|
Jiang T, Ma C, Chen H. Unraveling the ultrastructure and dynamics of autophagic vesicles: Insights from advanced imaging techniques. FASEB Bioadv 2024; 6:189-199. [PMID: 38974114 PMCID: PMC11226998 DOI: 10.1096/fba.2024-00035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/18/2024] [Accepted: 04/22/2024] [Indexed: 07/09/2024] Open
Abstract
Autophagy, an intracellular self-degradation process, is governed by a complex interplay of signaling pathways and interactions between proteins and organelles. Its fundamental purpose is to efficiently clear and recycle cellular components that are damaged or redundant. Central to this process are autophagic vesicles, specialized structures that encapsulate targeted cellular elements, playing a pivotal role in autophagy. Despite growing interest in the molecular components of autophagic machinery and their regulatory mechanisms, capturing the detailed ultrastructural dynamics of autophagosome formation continues to present significant challenges. However, recent advancements in microscopy, particularly in electron microscopy, have begun to illuminate the dynamic regulatory processes underpinning autophagy. This review endeavors to provide an exhaustive overview of contemporary research on the ultrastructure of autophagic processes. By synthesizing observations from diverse technological methodologies, this review seeks to deepen our understanding of the genesis of autophagic vesicles, their membrane origins, and the dynamic alterations that transpire during the autophagy process. The aim is to bridge gaps in current knowledge and foster a more comprehensive comprehension of this crucial cellular mechanism.
Collapse
Affiliation(s)
- Ting Jiang
- Institute of Reproductive MedicineMedical School of Nantong UniversityNantongPR China
| | - Chaoye Ma
- Institute of Reproductive MedicineMedical School of Nantong UniversityNantongPR China
| | - Hao Chen
- Institute of Reproductive MedicineMedical School of Nantong UniversityNantongPR China
- Guangzhou Women and Children’s Medical Center, GMU‐GIBH Joint School of Life ScienceGuangzhou Medical UniversityGuangzhouChina
| |
Collapse
|
47
|
Hu M, Feng X, Liu Q, Liu S, Huang F, Xu H. The ion channels of endomembranes. Physiol Rev 2024; 104:1335-1385. [PMID: 38451235 PMCID: PMC11381013 DOI: 10.1152/physrev.00025.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 02/20/2024] [Accepted: 02/25/2024] [Indexed: 03/08/2024] Open
Abstract
The endomembrane system consists of organellar membranes in the biosynthetic pathway [endoplasmic reticulum (ER), Golgi apparatus, and secretory vesicles] as well as those in the degradative pathway (early endosomes, macropinosomes, phagosomes, autophagosomes, late endosomes, and lysosomes). These endomembrane organelles/vesicles work together to synthesize, modify, package, transport, and degrade proteins, carbohydrates, and lipids, regulating the balance between cellular anabolism and catabolism. Large ion concentration gradients exist across endomembranes: Ca2+ gradients for most endomembrane organelles and H+ gradients for the acidic compartments. Ion (Na+, K+, H+, Ca2+, and Cl-) channels on the organellar membranes control ion flux in response to cellular cues, allowing rapid informational exchange between the cytosol and organelle lumen. Recent advances in organelle proteomics, organellar electrophysiology, and luminal and juxtaorganellar ion imaging have led to molecular identification and functional characterization of about two dozen endomembrane ion channels. For example, whereas IP3R1-3 channels mediate Ca2+ release from the ER in response to neurotransmitter and hormone stimulation, TRPML1-3 and TMEM175 channels mediate lysosomal Ca2+ and H+ release, respectively, in response to nutritional and trafficking cues. This review aims to summarize the current understanding of these endomembrane channels, with a focus on their subcellular localizations, ion permeation properties, gating mechanisms, cell biological functions, and disease relevance.
Collapse
Affiliation(s)
- Meiqin Hu
- Department of Neurology and Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- New Cornerstone Science Laboratory, Liangzhu Laboratory and School of Basic Medical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Xinghua Feng
- Department of Neurology and Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- New Cornerstone Science Laboratory, Liangzhu Laboratory and School of Basic Medical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Qiang Liu
- New Cornerstone Science Laboratory, Liangzhu Laboratory and School of Basic Medical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Siyu Liu
- New Cornerstone Science Laboratory, Liangzhu Laboratory and School of Basic Medical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Fangqian Huang
- New Cornerstone Science Laboratory, Liangzhu Laboratory and School of Basic Medical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Haoxing Xu
- Department of Neurology and Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- New Cornerstone Science Laboratory, Liangzhu Laboratory and School of Basic Medical Sciences, Zhejiang University, Hangzhou, People's Republic of China
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States
| |
Collapse
|
48
|
Duan J, Huang Z, Qin S, Li B, Zhang Z, Liu R, Wang K, Nice EC, Jiang J, Huang C. Oxidative stress induces extracellular vesicle release by upregulation of HEXB to facilitate tumour growth in experimental hepatocellular carcinoma. J Extracell Vesicles 2024; 13:e12468. [PMID: 38944674 PMCID: PMC11214608 DOI: 10.1002/jev2.12468] [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: 02/15/2024] [Revised: 06/01/2024] [Accepted: 06/07/2024] [Indexed: 07/01/2024] Open
Abstract
Extracellular vesicles (EVs) play a crucial role in triggering tumour-aggressive behaviours. However, the energetic process by which tumour cells produce EVs remains poorly understood. Here, we demonstrate the involvement of β-hexosaminidase B (HEXB) in mediating EV release in response to oxidative stress, thereby promoting the development of hepatocellular carcinoma (HCC). Mechanistically, reactive oxygen species (ROS) stimulate the nuclear translocation of transcription factor EB (TFEB), leading to the upregulation of both HEXB and its antisense lncRNA HEXB-AS. HEXB-AS can bind HEXB to form a protein/RNA complex, which elevates the protein stability of HEXB. The stabilized HEXB interacts with lysosome-associated membrane glycoprotein 1 (LAMP1), disrupting lysosome-multivesicular body (MVB) fusion, which protects EVs from degradation. Knockdown of HEXB efficiently inhibits EV release and curbs HCC growth both in vitro and in vivo. Moreover, targeting HEXB by M-31850 significantly inhibits HCC growth, especially when combined with GW4869, an inhibitor of exosome release. Our results underscore the critical role of HEXB as a modulator that promotes EV release during HCC development.
Collapse
Affiliation(s)
- Jiufei Duan
- Department of Biotherapy, Cancer Center and State Key Laboratory of BiotherapyWest China Hospital, and West China School of Basic Medical Sciences & Forensic MedicineSichuan UniversityChengduP.R. China
| | - Zhao Huang
- West China School of Public Health and West China Fourth HospitalSichuan UniversityChengduP.R. China
| | - Siyuan Qin
- Department of Biotherapy, Cancer Center and State Key Laboratory of BiotherapyWest China Hospital, and West China School of Basic Medical Sciences & Forensic MedicineSichuan UniversityChengduP.R. China
| | - Bowen Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of BiotherapyWest China Hospital, and West China School of Basic Medical Sciences & Forensic MedicineSichuan UniversityChengduP.R. China
| | - Zhe Zhang
- MOE Joint International Research Laboratory of Pancreatic Diseases, the First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouZhejiangChina
| | - Rui Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of StomatologySichuan UniversityChengduSichuanP.R. China
| | - Kui Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of BiotherapyWest China Hospital, and West China School of Basic Medical Sciences & Forensic MedicineSichuan UniversityChengduP.R. China
| | - Edouard C. Nice
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVictoriaAustralia
| | - Jingwen Jiang
- West China School of Public Health and West China Fourth HospitalSichuan UniversityChengduP.R. China
| | - Canhua Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of BiotherapyWest China Hospital, and West China School of Basic Medical Sciences & Forensic MedicineSichuan UniversityChengduP.R. China
| |
Collapse
|
49
|
Wang X, Chen H, Jiang J, Ma J. Hesperidin Alleviates Hepatic Injury Caused by Deoxynivalenol Exposure through Activation of mTOR and AKT/GSK3β/TFEB Pathways. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:14349-14363. [PMID: 38869217 DOI: 10.1021/acs.jafc.4c02039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2024]
Abstract
Deoxynivalenol (DON) is a common agricultural mycotoxin that is chemically stable and not easily removed from cereal foods. When organisms consume food made from contaminated crops, it can be hazardous to their health. Numerous studies in recent years have found that hesperidin (HDN) has hepatoprotective effects on a wide range of toxins. However, few scholars have explored the potential of HDN in attenuating DON-induced liver injury. In this study, we established a low-dose DON exposure model and intervened with three doses of HDN, acting on male C57 BL/6 mice and AML12 cells, which served as in vivo and in vitro models, respectively, to investigate the protective mechanism of HDN against DON exposure-induced liver injury. The results suggested that DON disrupted hepatic autophagic fluxes, thereby impairing liver structure and function, and HDN significantly attenuated these changes. Further studies revealed that HDN alleviated DON-induced excessive autophagy through the mTOR pathway and DON-induced lysosomal dysfunction through the AKT/GSK3β/TFEB pathway. Overall, our study suggested that HDN could ameliorate DON-induced autophagy flux disorders via the mTOR pathway and the AKT/GSK3β/TFEB pathway, thereby reducing liver injury.
Collapse
Affiliation(s)
- Xin Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Hao Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Junze Jiang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Jun Ma
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Harbin 150030, P. R. China
| |
Collapse
|
50
|
Chang X, Wang WX. Differential cellular uptake and trafficking of nanoplastics in two hemocyte subpopulations of mussels Perna viridis. JOURNAL OF HAZARDOUS MATERIALS 2024; 471:134388. [PMID: 38669925 DOI: 10.1016/j.jhazmat.2024.134388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/27/2024] [Accepted: 04/21/2024] [Indexed: 04/28/2024]
Affiliation(s)
- Xinyi Chang
- School of Energy and Environment and State Key Laboratory of Marine Pollution, City University of Hong Kong, Kowloon, Hong Kong, China; Research Centre for the Oceans and Human Health, City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, China
| | - Wen-Xiong Wang
- School of Energy and Environment and State Key Laboratory of Marine Pollution, City University of Hong Kong, Kowloon, Hong Kong, China; Research Centre for the Oceans and Human Health, City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, China.
| |
Collapse
|