51
|
Martinez-Banaclocha MA. Targeting the Cysteine Redox Proteome in Parkinson's Disease: The Role of Glutathione Precursors and Beyond. Antioxidants (Basel) 2023; 12:1373. [PMID: 37507913 PMCID: PMC10376658 DOI: 10.3390/antiox12071373] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 06/22/2023] [Accepted: 06/28/2023] [Indexed: 07/30/2023] Open
Abstract
Encouraging recent data on the molecular pathways underlying aging have identified variants and expansions of genes associated with DNA replication and repair, telomere and stem cell maintenance, regulation of the redox microenvironment, and intercellular communication. In addition, cell rejuvenation requires silencing some transcription factors and the activation of pluripotency, indicating that hidden molecular networks must integrate and synchronize all these cellular mechanisms. Therefore, in addition to gene sequence expansions and variations associated with senescence, the optimization of transcriptional regulation and protein crosstalk is essential. The protein cysteinome is crucial in cellular regulation and plays unexpected roles in the aging of complex organisms, which show cumulative somatic mutations, telomere attrition, epigenetic modifications, and oxidative dysregulation, culminating in cellular senescence. The cysteine thiol groups are highly redox-active, allowing high functional versatility as structural disulfides, redox-active disulfides, active-site nucleophiles, proton donors, and metal ligands to participate in multiple regulatory sites in proteins. Also, antioxidant systems control diverse cellular functions, including the transcription machinery, which partially depends on the catalytically active cysteines that can reduce disulfide bonds in numerous target proteins, driving their biological integration. Since we have previously proposed a fundamental role of cysteine-mediated redox deregulation in neurodegeneration, we suggest that cellular rejuvenation of the cysteine redox proteome using GSH precursors, like N-acetyl-cysteine, is an underestimated multitarget therapeutic approach that would be particularly beneficial in Parkinson's disease.
Collapse
|
52
|
Lisi V, Moulton C, Fantini C, Grazioli E, Guidotti F, Sgrò P, Dimauro I, Capranica L, Parisi A, Di Luigi L, Caporossi D. Steady-state redox status in circulating extracellular vesicles: A proof-of-principle study on the role of fitness level and short-term aerobic training in healthy young males. Free Radic Biol Med 2023; 204:266-275. [PMID: 37182793 DOI: 10.1016/j.freeradbiomed.2023.05.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/24/2023] [Accepted: 05/07/2023] [Indexed: 05/16/2023]
Abstract
Considering the role of redox homeostasis in exercise-induced signaling and adaptation, this study focuses on the exercise training-related intercellular communication of redox status mediated by circulating extracellular vesicles (EVs). 19 healthy young males were divided into trained (TG, 7) and untrained (UG, 12) subjects based on their VO2MAX. The UG subjects were further randomly distributed in experimental (UGEX, N = 7) and control (UGCTRL, N = 5) groups. The steady state of plasma EVs in TG and UGEX have been characterized for total number and size, as well as cargo redox status (antioxidants, transcription factors, HSPs) before, 3 and 24 h after a single bout of aerobic exercise (30', 70% HRM). Plasma EVs from UGEX and UGCTRL have been further characterized after 24 h from the last session of a 5-day consecutive aerobic training or no training, respectively. No differences were detected in the EVs' size and distribution at baseline in TG and UGEX (p>0.05), while the EVs cargo of UGEX showed a significantly higher concentration of protein carbonyl, Catalase, SOD2, and HSF1 compared to TG (p<0.05). 5 days of consecutive aerobic training in UGEX did not determine major changes in the steady-state number and size of EVs. The post-training levels of protein carbonyl, HSF1, Catalase, and SOD2 in EVs cargo of UGEX resulted significantly lower compared with UGEX before training and UGCTRL, resembling the steady-state levels in circulating EVs of TG subjects. Altogether, these preliminary data indicate that individual aerobic capacity influences the redox status of circulating EVs, and that short-term aerobic training impacts the steady-state redox status of EVs. Taking this pilot study as a paradigm for physio-pathological stimuli impacting redox homeostasis, our results offer new insights into the utilization of circulating EVs as biomarkers of exercise efficacy and of early impairment of oxidative-stress related diseases.
Collapse
Affiliation(s)
- Veronica Lisi
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Italy
| | - Chantalle Moulton
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Italy
| | - Cristina Fantini
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Italy
| | - Elisa Grazioli
- Physical Exercise and Sport Sciences Unit, Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", Rome, Italy
| | - Flavia Guidotti
- Sport Performance Laboratory, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Italy
| | - Paolo Sgrò
- Endocrinology Unit, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Italy
| | - Ivan Dimauro
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Italy
| | - Laura Capranica
- Sport Performance Laboratory, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Italy
| | - Attilio Parisi
- Physical Exercise and Sport Sciences Unit, Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", Rome, Italy
| | - Luigi Di Luigi
- Endocrinology Unit, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Italy
| | - Daniela Caporossi
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Italy.
| |
Collapse
|
53
|
Travagli V, Iorio EL. The Biological and Molecular Action of Ozone and Its Derivatives: State-of-the-Art, Enhanced Scenarios, and Quality Insights. Int J Mol Sci 2023; 24:ijms24108465. [PMID: 37239818 DOI: 10.3390/ijms24108465] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/19/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023] Open
Abstract
The ultimate objective of this review is to encourage a multi-disciplinary and integrated methodological approach that, starting from the recognition of some current uncertainties, helps to deepen the molecular bases of ozone treatment effects on human and animal well-being and to optimize their performance in terms of reproducibility of results, quality, and safety. In fact, the common therapeutic treatments are normally documented by healthcare professionals' prescriptions. The same applies to medicinal gases (whose uses are based on their pharmacological effects) that are intended for patients for treatment, diagnostic, or preventive purposes and that have been produced and inspected in accordance with good manufacturing practices and pharmacopoeia monographs. On the contrary, it is the responsibility of healthcare professionals, who thoughtfully choose to use ozone as a medicinal product, to achieve the following objectives: (i) to understand the molecular basis of the mechanism of action; (ii) to adjust the treatment according to the clinical responses obtained in accordance with the principles of precision medicine and personalized therapy; (iii) to ensure all quality standards.
Collapse
Affiliation(s)
- Valter Travagli
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Viale Aldo Moro 2, 53100 Siena, Italy
| | - Eugenio Luigi Iorio
- International Observatory of Oxidative Stress, 84127 Salerno, Italy
- Campus Uberlândia, Universidade de Uberaba (UNIUBE), Uberlândia 38055-500, Brazil
| |
Collapse
|
54
|
Pillai SS, Pereira DG, Zhang J, Huang W, Beg MA, Knaack DA, de Souza Goncalves B, Sahoo D, Silverstein RL, Shapiro JI, Sodhi K, Chen Y. Contribution of adipocyte Na/K-ATPase α1/CD36 signaling induced exosome secretion in response to oxidized LDL. Front Cardiovasc Med 2023; 10:1046495. [PMID: 37180782 PMCID: PMC10174328 DOI: 10.3389/fcvm.2023.1046495] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 04/13/2023] [Indexed: 05/16/2023] Open
Abstract
Introduction Adipose tissue constantly secretes adipokines and extracellular vesicles including exosomes to crosstalk with distinct tissues and organs for whole-body homeostasis. However, dysfunctional adipose tissue under chronic inflammatory conditions such as obesity, atherosclerosis, and diabetes shows pro-inflammatory phenotypes accompanied by oxidative stress and abnormal secretion. Nevertheless, molecular mechanisms of how adipocytes are stimulated to secrete exosomes under those conditions remain poorly understood. Methods Mouse and human in vitro cell culture models were used for performing various cellular and molecular studies on adipocytes and macrophages. Statistical analysis was performed using Student's t-test (two-tailed, unpaired, and equal variance) for comparisons between two groups or ANOVA followed by Bonferroni's multiple comparison test for comparison among more than two groups. Results and discussion In this work, we report that CD36, a scavenger receptor for oxidized LDL, formed a signaling complex with another membrane signal transducer Na/K-ATPase in adipocytes. The atherogenic oxidized LDL induced a pro-inflammatory response in in vitro differentiated mouse and human adipocytes and also stimulated the cells to secrete more exosomes. This was largely blocked by either CD36 knockdown using siRNA or pNaKtide, a peptide inhibitor of Na/K-ATPase signaling. These results showed a critical role of the CD36/Na/K-ATPase signaling complex in oxidized LDL-induced adipocyte exosome secretion. Moreover, by co-incubation of adipocyte-derived exosomes with macrophages, we demonstrated that oxidized LDL-induced adipocyte-derived exosomes promoted pro-atherogenic phenotypes in macrophages, including CD36 upregulation, IL-6 secretion, metabolic switch to glycolysis, and mitochondrial ROS production. Altogether, we show here a novel mechanism through which adipocytes increase exosome secretion in response to oxidized LDL and that the secreted exosomes can crosstalk with macrophages, which may contribute to atherogenesis.
Collapse
Affiliation(s)
- Sneha S. Pillai
- Department of Surgery, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Duane G. Pereira
- Department of Surgery, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Jue Zhang
- Versiti Blood Research Institute, Milwaukee, WI, United States
| | - Wenxin Huang
- Versiti Blood Research Institute, Milwaukee, WI, United States
| | - Mirza Ahmar Beg
- Versiti Blood Research Institute, Milwaukee, WI, United States
| | - Darcy A. Knaack
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Bruno de Souza Goncalves
- Department of Surgery, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Daisy Sahoo
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Roy L. Silverstein
- Versiti Blood Research Institute, Milwaukee, WI, United States
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Joseph I. Shapiro
- Department of Surgery, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Komal Sodhi
- Department of Surgery, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Yiliang Chen
- Versiti Blood Research Institute, Milwaukee, WI, United States
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
55
|
Ma Z, Yu P, Li X, Dai F, Jiang H, Liu J. Anemonin reduces hydrogen peroxide-induced oxidative stress, inflammation and extracellular matrix degradation in nucleus pulposus cells by regulating NOX4/NF-κB signaling pathway. J Orthop Surg Res 2023; 18:189. [PMID: 36899420 PMCID: PMC10007850 DOI: 10.1186/s13018-023-03679-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 03/04/2023] [Indexed: 03/12/2023] Open
Abstract
BACKGROUND Excessive oxidative stress plays a critical role in the progression of various diseases, including intervertebral disk degeneration (IVDD). Recent studies have found that anemonin (ANE) possesses antioxidant and anti-inflammatory effects. However, the role of ANE in IVDD is still unclear. Therefore, this study investigated the effect and mechanism of ANE on H2O2 induced degeneration of nucleus pulposus cells (NPCs). METHODS NPCs were pretreated with ANE, and then treated with H2O2. NOX4 was upregulated by transfection of pcDNA-NOX4 into NPCs. Cytotoxicity was detected by MTT, oxidative stress-related indicators and inflammatory factors were measured by ELISA, mRNA expression was assessed by RT-PCR, and protein expression was tested by western blot. RESULTS ANE attenuated H2O2-induced inhibition of NPCs activity. H2O2 enhanced oxidative stress, namely, increased ROS and MDA levels and decreased SOD level. However, these were suppressed and pretreated by ANE. ANE treatment repressed the expression of inflammatory factors (IL-6, IL-1β and TNF-α) in H2O2-induced NPCs. ANE treatment also prevented the degradation of extracellular matrix induced by H2O2, showing the downregulation of MMP-3, 13 and ADAMTS-4, 5 and the upregulation of collagen II. NOX4 is a key factor regulating oxidative stress. Our study confirmed that ANE could restrain NOX4 and p-NF-κB. In addition, overexpression of NOX4 counteracted the antioxidant and anti-inflammatory activities of ANE in H2O2-induced NPCs, and the inhibition of the degradation of extracellular matrix induced by ANE was also reversed by overexpression of NOX4. CONCLUSION ANE repressed oxidative stress, inflammation and extracellular matrix degradation in H2O2-induced NPCs by inhibiting NOX4/NF-κB pathway. Our study indicated that ANE might be a candidate drug for the treatment of IVDD.
Collapse
Affiliation(s)
- Zhijia Ma
- Department of Orthopaedic Surgery, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, No. 889, Wuzhong West Road, Gusu District, Suzhou, 215009, Jiangsu, China
| | - Pengfei Yu
- Department of Orthopaedic Surgery, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, No. 889, Wuzhong West Road, Gusu District, Suzhou, 215009, Jiangsu, China
| | - Xiaochun Li
- Department of Orthopaedic Surgery, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, No. 889, Wuzhong West Road, Gusu District, Suzhou, 215009, Jiangsu, China
| | - Feng Dai
- Department of Orthopaedic Surgery, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, No. 889, Wuzhong West Road, Gusu District, Suzhou, 215009, Jiangsu, China
| | - Hong Jiang
- Department of Orthopaedic Surgery, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, No. 889, Wuzhong West Road, Gusu District, Suzhou, 215009, Jiangsu, China.
| | - Jintao Liu
- Department of Orthopaedic Surgery, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, No. 889, Wuzhong West Road, Gusu District, Suzhou, 215009, Jiangsu, China.
| |
Collapse
|
56
|
Mas-Bargues C, Alique M. Extracellular Vesicles as "Very Important Particles" (VIPs) in Aging. Int J Mol Sci 2023; 24:ijms24044250. [PMID: 36835661 PMCID: PMC9964932 DOI: 10.3390/ijms24044250] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/15/2023] [Accepted: 02/18/2023] [Indexed: 02/25/2023] Open
Abstract
In recent decades, extracellular vesicles have been recognized as "very important particles" (VIPs) associated with aging and age-related disease. During the 1980s, researchers discovered that these vesicle particles released by cells were not debris but signaling molecules carrying cargoes that play key roles in physiological processes and physiopathological modulation. Following the International Society for Extracellular Vesicles (ISEV) recommendation, different vesicle particles (e.g., exosomes, microvesicles, oncosomes) have been named globally extracellular vesicles. These vesicles are essential to maintain body homeostasis owing to their essential and evolutionarily conserved role in cellular communication and interaction with different tissues. Furthermore, recent studies have shown the role of extracellular vesicles in aging and age-associated diseases. This review summarizes the advances in the study of extracellular vesicles, mainly focusing on recently refined methods for their isolation and characterization. In addition, the role of extracellular vesicles in cell signaling and maintenance of homeostasis, as well as their usefulness as new biomarkers and therapeutic agents in aging and age-associated diseases, has also been highlighted.
Collapse
Affiliation(s)
- Cristina Mas-Bargues
- Grupo de Investigación Freshage, Departamento de Fisiología, Facultad de Medicina, Universidad de Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), Instituto Sanitario de Investigación INCLIVA, 46010 Valencia, Spain
- Correspondence: (C.M.-B.); (M.A.)
| | - Matilde Alique
- Departamento de Biología de Sistemas, Universidad de Alcalá, Alcalá de Henares, 28871 Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain
- Correspondence: (C.M.-B.); (M.A.)
| |
Collapse
|
57
|
Barranco I, Sanchez-López CM, Bucci D, Alvarez-Barrientos A, Rodriguez-Martinez H, Marcilla A, Roca J. The Proteome of Large or Small Extracellular Vesicles in Pig Seminal Plasma Differs, Defining Sources and Biological Functions. Mol Cell Proteomics 2023; 22:100514. [PMID: 36796643 PMCID: PMC10017305 DOI: 10.1016/j.mcpro.2023.100514] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 11/10/2022] [Accepted: 01/11/2023] [Indexed: 02/16/2023] Open
Abstract
Seminal plasma contains many morphologically heterogeneous extracellular vesicles (sEVs). These are sequentially released by cells of the testis, epididymis, and accessory sex glands and involved in male and female reproductive processes. This study aimed to define in depth sEV subsets isolated by ultrafiltration and size exclusion chromatography, decode their proteomic profiles using liquid chromatography-tandem mass spectrometry, and quantify identified proteins using sequential window acquisition of all theoretical mass spectra. The sEV subsets were defined as large (L-EVs) or small (S-EVs) by their protein concentration, morphology, size distribution, and EV-specific protein markers and purity. Liquid chromatography-tandem mass spectrometry identified a total of 1034 proteins, 737 of them quantified by SWATH in S-EVs, L-EVs, and non-EVs-enriched samples (18-20 size exclusion chromatography-eluted fractions). The differential expression analysis revealed 197 differentially abundant proteins between both EV subsets, S-EVs and L-EVs, and 37 and 199 between S-EVs and L-EVs versus non-EVs-enriched samples, respectively. The gene ontology enrichment analysis of differentially abundant proteins suggested, based on the type of protein detected, that S-EVs could be mainly released through an apocrine blebbing pathway and be involved in modulating the immune environment of the female reproductive tract as well as during sperm-oocyte interaction. In contrast, L-EVs could be released by fusion of multivesicular bodies with the plasma membrane becoming involved in sperm physiological processes, such as capacitation and avoidance of oxidative stress. In conclusion, this study provides a procedure capable of isolating subsets of EVs from pig seminal plasma with a high degree of purity and shows differences in the proteomic profile between EV subsets, indicating different sources and biological functions for the sEVs.
Collapse
Affiliation(s)
- Isabel Barranco
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, Bologna, Italy
| | - Christian M Sanchez-López
- Àrea de Parasitologia, Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Universitat de València, Burjassot, Valencia, Spain; Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, Health Research Institute La Fe-Universitat de València, Valencia, Spain
| | - Diego Bucci
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, Bologna, Italy
| | | | | | - Antonio Marcilla
- Àrea de Parasitologia, Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Universitat de València, Burjassot, Valencia, Spain; Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, Health Research Institute La Fe-Universitat de València, Valencia, Spain
| | - Jordi Roca
- Department of Medicine and Animal Surgery, Faculty of Veterinary Science, University of Murcia, Murcia, Spain.
| |
Collapse
|
58
|
Liu Y, Yuang G, Chen X, Liu J. Near-infrared band responsive ROS regulator selectively inhibits breast cancer cells by programming combination phototherapy. J Mater Chem B 2023; 11:1356-1364. [PMID: 36655692 DOI: 10.1039/d2tb02508f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Catalytic therapy can effectively kill tumor cells and inhibit tumor growth by producing highly toxic reactive oxygen species (ROS). However, the long-term catalysis of nanozymes easily lead to ROS breaking through the boundary in tumor tissues, resulting in spillover and injuring normal cells. Therefore, how to control the threshold of ROS production from nanozymes in tumor tissues is an unsolved problem. In this work, to prevent the boundary effect of the photosensitizer ([Ru(bpy)2(tip)]2+, RBT) during ROS generation, we used the sensitivity of RBT and PdH0.2-Ir with different wavelengths of near-infrared light (NIR) to generate ROS and H2, respectively. Therefore, an intelligent nanosystem PdH0.2-Ir@RBT(PIH@R) was constructed to precisely control ROS generation by adjusting the NIR laser wavelength. The palladium-iridium alloy (Pd-Ir) nanoparticles as the core can co-load hydrogen (H2) and RBT and show NIR-responsive behaviors. Under 808 nm laser irradiation, PIH@R produces ROS with the photocatalysis of RBT, while under 1064 nm laser irradiation PIH@R will quickly activate and release H2 to eliminate ROS. Interestingly, in vitro and in vivo experiments showed that PIH@R acted like a "Trojan horse": PIH@R can destroy the mitochondria of 4T1 cells to destroy their redox homeostasis system, resulting in cancer cells relying on exogenous PIH@R to change their reactive oxygen species levels. Subsequently, when PIH@R is activated into a harmful oxidation state, it can easily crush the redox homeostasis system of cancer cells and induce cancer cell apoptosis.
Collapse
Affiliation(s)
- Yanan Liu
- Shenzhen Longhua Maternity and Child Healthcare Hospital, Shenzhen, 518110, China.
| | - Guanglong Yuang
- Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou 511436, China.
| | - Xu Chen
- Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou 511436, China. .,Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, Guangzhou 510317, P. R. China
| | - Jie Liu
- Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou 511436, China.
| |
Collapse
|
59
|
Yadav P, Beura SK, Panigrahi AR, Bhardwaj T, Giri R, Singh SK. Platelet-derived microvesicles activate human platelets via intracellular calcium mediated reactive oxygen species release. Blood Cells Mol Dis 2023; 98:102701. [DOI: 10.1016/j.bcmd.2022.102701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/18/2022] [Accepted: 08/23/2022] [Indexed: 11/30/2022]
|
60
|
Yilmaz G, Tavsan Z, Cagatay E, Kursunluoglu G, Kayali HA. Exosomes released from cisplatin-resistant ovarian cancer cells modulate the reprogramming of cells in tumor microenvironments toward the cancerous cells. Biomed Pharmacother 2023; 157:113973. [PMID: 36413836 DOI: 10.1016/j.biopha.2022.113973] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/28/2022] [Accepted: 11/02/2022] [Indexed: 11/21/2022] Open
Abstract
Exosomes released from cancer cells are involved in the reorganization of the tumor microenvironment which is the essential aspect of cancer pathogenesis. The intercommunications between cancer cells and diverse cell types in the microenvironment are accomplished by exosomes in ovarian cancer. Internalization pathway, intracellular fate, and biological functions in recipient cells mediated by exosomes released from cisplatin-resistant A2780cis have been studied. Also, histopathological evaluation of tumor, ovary, liver tissues and lymph nodes in vivo studies have been performed. The recipient cells internalized the exosomes via active uptake mechanisms, as shown by confocal microscopy. However, inhibitor studies and flow cytometry analysis showed that each recipient cell line used different uptake pathways. Also, confocal microscopy imaging indicated that the internalized exosomes trapped in the endosomes or phagosomes were distributed to the different cellular compartments including ER, Golgi, and lysosome. The transfer of exosomal oncogenic cargo into the cells modified the intracellular signaling of recipient cells including invasion and metastasis by Boyden-Chamber assay, proliferation by ATP analysis, epithelial-mesenchymal transition (EMT) markers at protein and mRNA levels by western blotting and real-time PCR, and protein kinases in the phospho-kinase array. This remodeling contributed to the initiation of carcinogenesis in ovarian epithelial and peritoneal mesothelial cells, and the progression of carcinogenesis in ovarian cancer cells. In addition, intraperitoneal tumor model studies show that exosomes released from cisplatin-resistant A2780cis cells may play role in the enlargement of lymph nodes, and tumor formations integrated with the liver, attached to the stomach and in the ovarian tissues.
Collapse
Affiliation(s)
- Gizem Yilmaz
- Izmir Biomedicine and Genome Center, 35340 İzmir, Turkey; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 İzmir, Turkey
| | - Zehra Tavsan
- Izmir Biomedicine and Genome Center, 35340 İzmir, Turkey
| | - Elcin Cagatay
- Izmir Biomedicine and Genome Center, 35340 İzmir, Turkey; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 İzmir, Turkey
| | - Gizem Kursunluoglu
- Izmir Biomedicine and Genome Center, 35340 İzmir, Turkey; ERFARMA-Drug Application and Research Center, Erciyes University, Kayseri 38280 Turkey
| | - Hulya Ayar Kayali
- Izmir Biomedicine and Genome Center, 35340 İzmir, Turkey; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 İzmir, Turkey; Department of Chemistry, Division of Biochemistry, Faculty of Science, Dokuz Eylul University, 35390 İzmir, Turkey.
| |
Collapse
|
61
|
Wang S, Li C, Yuan Y, Xiong Y, Xu H, Pan W, Pan H, Zhu Z. Microvesicles as drug delivery systems: A new frontier for bionic therapeutics in cancer. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2022.104088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
62
|
Xiong Y, Zhang Y, Zhou C, Yu T. ROS scavenging Manganese-loaded mesoporous silica nanozymes for catalytic anti-inflammatory therapy. ADV POWDER TECHNOL 2023. [DOI: 10.1016/j.apt.2022.103886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
63
|
Liu C, Wang M, Yao H, Cui M, Gong X, Wang L, Sui C, Zhang H. Inhibition of oocyte maturation by follicular extracellular vesicles of non-hyperandrogenic PCOS patients requiring IVF. J Clin Endocrinol Metab 2022; 108:1394-1404. [PMID: 36527699 DOI: 10.1210/clinem/dgac733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 12/14/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
CONTEXT Polycystic ovarian syndrome (PCOS) is one of the most common diseases that contribute to subfertility. Recent evidence showed that oocytes of women with PCOS matured in vitro away from the follicular fluid presented better potentials, whereas the reason remained unclear. OBJECTIVE To investigate whether follicular extracellular vesicles (EVs) of PCOS patients interfere with the quality of oocytes. METHODS Follicular EVs of women with PCOS (PCOS-EVs) and control women (CTRL-EVs) were isolated and determined using western blotting, nanoparticle tracking analysis, and transmission electron microscopy. The two types of EVs were co-cultured with murine germinal vesicle oocytes, respectively. Fluorescence labeled EVs were used to visualize internalization by oocytes. After co-culture, oocyte maturation rates were calculated. Mitochondria distribution and reactive oxygen species (ROS) level were detected in the different groups. Spindle morphology was evaluated using immunofluorescence. Moreover, the expression of catalase (CAT), glutathione synthetase (GSS), and superoxide dismutase (SOD) was determined in the oocytes. RESULTS Both PCOS-EVs and CTRL-EVs are bilayered vesicles, approximately 100-150 nm in size, and enriched in EV-associating protein markers. EVs were internalized by oocytes within one hour. Oocyte maturation rate decreased significantly in the PCOS-EV group compared with the CTRL-EV group; whereas the abnormal mitochondria distribution rate and abnormal spindle rate were significantly increased in the PCOS-EV group. Moreover, PCOS-EVs increased the ROS level and the expression of CAT, GSS, and SOD in the oocytes. CONCLUSIONS PCOS-EVs interfered with oocyte mitochondria and spindles and inhibited oocyte maturation. Moreover, oxidative stress induced by PCOS-EVs might be a potential cause.
Collapse
Affiliation(s)
- Chang Liu
- Reproductive Medicine Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People's Republic of China
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Meng Wang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Haixia Yao
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Shangrao People's Hospital, Shangrao, China
| | - Mengge Cui
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Xueqi Gong
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Lan Wang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Cong Sui
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Hanwang Zhang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
| |
Collapse
|
64
|
Serafini FL, Delli Pizzi A, Simeone P, Giammarino A, Mannetta C, Villani M, Izzi J, Buca D, Catitti G, Chiacchiaretta P, Trebeschi S, Miscia S, Caulo M, Lanuti P. Circulating Extracellular Vesicles: Their Role in Patients with Abdominal Aortic Aneurysm (AAA) Undergoing EndoVascular Aortic Repair (EVAR). Int J Mol Sci 2022; 23:ijms232416015. [PMID: 36555653 PMCID: PMC9782915 DOI: 10.3390/ijms232416015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/03/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a frequent aortic disease. If the diameter of the aorta is larger than 5 cm, an open surgical repair (OSR) or an endovascular aortic repair (EVAR) are recommended. To prevent possible complications (i.e., endoleaks), EVAR-treated patients need to be monitored for 5 years following the intervention, using computed tomography angiography (CTA). However, this radiological method involves high radiation exposure in terms of CTA/year. In such a context, the study of peripheral-blood-circulating extracellular vesicles (pbcEVs) has great potential to identify biomarkers for EVAR complications. We analyzed several phenotypes of pbcEVs using polychromatic flow cytometry in 22 patients with AAA eligible for EVAR. From each enrolled patient, peripheral blood samples were collected at AAA diagnosis, and after 1, 6, and 12 months following EVAR implantation, i.e. during the diagnostic follow-up protocol. Patients developing an endoleak displayed a significant decrease in activated-platelet-derived EVs between the baseline condition and 6 months after EVAR intervention. Furthermore, we also observed, that 1 month after EVAR implantation, patients developing an endoleak showed higher concentrations of activated-endothelial-derived EVs than patients who did not develop one, suggesting their great potential as a noninvasive and specific biomarker for early identification of EVAR complications.
Collapse
Affiliation(s)
- Francesco Lorenzo Serafini
- Unit of Radiology, “SS. Annunziata” Hospital, 66100 Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy
| | - Andrea Delli Pizzi
- Unit of Radiology, “SS. Annunziata” Hospital, 66100 Chieti, Italy
- Department of Innovative Technologies in Medicine & Dentistry, University “G. d’Annunzio”, 66100 Chieti, Italy
- Institute of Advanced Biomedical Technologies (ITAB), University “G. d’Annunzio”, 66100 Chieti, Italy
- Correspondence: (A.D.P.); (P.S.)
| | - Pasquale Simeone
- Department of Medicine and Aging Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy
- Center for Advanced Studies and Technologies (CAST), University “G. d’Annunzio”, 66100 Chieti, Italy
- Correspondence: (A.D.P.); (P.S.)
| | | | - Cristian Mannetta
- Unit of Vascular Surgery, “SS. Annunziata” Hospital, 66100 Chieti, Italy
| | - Michela Villani
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy
| | - Jacopo Izzi
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy
| | - Davide Buca
- Department of Medicine and Aging Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy
- Center for Advanced Studies and Technologies (CAST), University “G. d’Annunzio”, 66100 Chieti, Italy
| | - Giulia Catitti
- Department of Medicine and Aging Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy
- Center for Advanced Studies and Technologies (CAST), University “G. d’Annunzio”, 66100 Chieti, Italy
| | - Piero Chiacchiaretta
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy
- Institute of Advanced Biomedical Technologies (ITAB), University “G. d’Annunzio”, 66100 Chieti, Italy
| | - Stefano Trebeschi
- Department of Radiology, Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Sebastiano Miscia
- Department of Medicine and Aging Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy
- Center for Advanced Studies and Technologies (CAST), University “G. d’Annunzio”, 66100 Chieti, Italy
| | - Massimo Caulo
- Unit of Radiology, “SS. Annunziata” Hospital, 66100 Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy
- Institute of Advanced Biomedical Technologies (ITAB), University “G. d’Annunzio”, 66100 Chieti, Italy
| | - Paola Lanuti
- Department of Medicine and Aging Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy
- Center for Advanced Studies and Technologies (CAST), University “G. d’Annunzio”, 66100 Chieti, Italy
| |
Collapse
|
65
|
Lian MQ, Chng WH, Liang J, Yeo HQ, Lee CK, Belaid M, Tollemeto M, Wacker MG, Czarny B, Pastorin G. Plant-derived extracellular vesicles: Recent advancements and current challenges on their use for biomedical applications. J Extracell Vesicles 2022; 11:e12283. [PMID: 36519808 PMCID: PMC9753580 DOI: 10.1002/jev2.12283] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/27/2022] [Accepted: 11/02/2022] [Indexed: 12/23/2022] Open
Abstract
Extracellular vesicles (EVs) represent a diverse class of lipid bilayer membrane vesicles released by both animal and plant cells. These ubiquitous vesicles are involved in intercellular communication and transport of various biological cargos, including proteins, lipids, and nucleic acids. In recent years, interest in plant-derived EVs has increased tremendously, as they serve as a scalable and sustainable alternative to EVs derived from mammalian sources. In vitro and in vivo findings have demonstrated that these plant-derived vesicles (PDVs) possess intrinsic therapeutic activities that can potentially treat diseases and improve human health. In addition, PDVs can also act as efficient and biocompatible drug carriers. While preclinical studies have shown promising results, there are still several challenges and knowledge gaps that have to be addressed for the successful translation of PDVs into clinical applications, especially in view of the lack of standardised protocols for material handling and PDV isolation from various plant sources. This review provides the readers with a quick overview of the current understanding and research on PDVs, critically analysing the current challenges and highlighting the immense potential of PDVs as a novel class of therapeutics to treat human diseases. It is expected that this work will guide scientists to address the knowledge gaps currently associated with PDVs and promote new advances in plant-based therapeutic solutions.
Collapse
Affiliation(s)
| | - Wei Heng Chng
- Department of PharmacyNational University of SingaporeSingaporeSingapore
- Integrative Sciences and Engineering Programme, NUS Graduate SchoolNational University of SingaporeSingaporeSingapore
| | - Jeremy Liang
- Department of ChemistryNational University of SingaporeSingaporeSingapore
| | - Hui Qing Yeo
- Department of PharmacyNational University of SingaporeSingaporeSingapore
- Cancer Science Institute of SingaporeNational University of SingaporeSingaporeSingapore
| | - Choon Keong Lee
- Department of PharmacyNational University of SingaporeSingaporeSingapore
| | - Mona Belaid
- Institute of Pharmaceutical ScienceKing's College LondonLondonUnited Kingdom
| | - Matteo Tollemeto
- Department of Health TechnologyTechnical University of DenmarkKongens LyngbyDenmark
| | | | - Bertrand Czarny
- School of Materials Science & EngineeringNanyang Technological UniversitySingaporeSingapore
| | - Giorgia Pastorin
- Department of PharmacyNational University of SingaporeSingaporeSingapore
| |
Collapse
|
66
|
You Y, Liu C, Liu T, Tian M, Wu N, Yu Z, Zhao F, Qi J, Zhu Q. FNDC3B protects steatosis and ferroptosis via the AMPK pathway in alcoholic fatty liver disease. Free Radic Biol Med 2022; 193:808-819. [PMID: 36336231 DOI: 10.1016/j.freeradbiomed.2022.10.322] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/27/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Alcoholic liver disease (ALD) is a leading cause of chronic liver disease worldwide with limited therapeutic options. The role of fibronectin type III domain-containing protein 3B (FNDC3B), an important regulator of metabolism, in ALD, and the underlying mechanism as well as its potential implication in ALD therapeutic strategies remain unknown. METHODS Hepatocyte-specific FNDC3B knockdown or control C57BL/6 N mice received a Lieber-DeCarli diet for four weeks, followed by oral gavage (chronic-binge). Primary mouse hepatocytes and cell lines were used for in vitro studies. Liver injury, hepatic steatosis, and lipid peroxidation were assessed. RESULTS In cultured cells and mouse livers, alcohol exposure increased FNDC3B expression. Hepatocyte-specific FNDC3B deletion aggravated alcohol-induced liver steatosis via AMP-activated protein kinase (AMPK) inhibition. In vitro, FNDC3B expression was negatively regulated by miR-192-5p. Furthermore, FNDC3B deletion significantly exacerbated ethanol-mediated lipid peroxidation. The RNA sequence assay revealed a connection between FNDC3B and ferroptosis, which was verified by the administration of the ferroptosis inhibitor ferrostatin-1 (Fer-1). Additionally, FNDC3B inhibition-mediated AMPK inactivation downregulated transferrin expression, which was associated with marked iron overload and ferroptosis. CONCLUSIONS This study elucidated the critical role of FNDC3B in preventing hepatic steatosis and ferroptosis in response to chronic alcohol consumption. Our findings indicate that FNDC3B is a potential therapeutic target for ALD.
Collapse
Affiliation(s)
- Yajing You
- Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China
| | - Chenxi Liu
- Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China; Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Tiantian Liu
- Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China
| | - Miaomiao Tian
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Nijin Wu
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Zhen Yu
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Fenglin Zhao
- Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China
| | - Jianni Qi
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| | - Qiang Zhu
- Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China; Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China; The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830000, Xinjiang, China.
| |
Collapse
|
67
|
Rodríguez-Giraldo M, González-Reyes RE, Ramírez-Guerrero S, Bonilla-Trilleras CE, Guardo-Maya S, Nava-Mesa MO. Astrocytes as a Therapeutic Target in Alzheimer's Disease-Comprehensive Review and Recent Developments. Int J Mol Sci 2022; 23:13630. [PMID: 36362415 PMCID: PMC9654484 DOI: 10.3390/ijms232113630] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 09/20/2023] Open
Abstract
Alzheimer's disease (AD) is a frequent and disabling neurodegenerative disorder, in which astrocytes participate in several pathophysiological processes including neuroinflammation, excitotoxicity, oxidative stress and lipid metabolism (along with a critical role in apolipoprotein E function). Current evidence shows that astrocytes have both neuroprotective and neurotoxic effects depending on the disease stage and microenvironmental factors. Furthermore, astrocytes appear to be affected by the presence of amyloid-beta (Aβ), with alterations in calcium levels, gliotransmission and proinflammatory activity via RAGE-NF-κB pathway. In addition, astrocytes play an important role in the metabolism of tau and clearance of Aβ through the glymphatic system. In this review, we will discuss novel pharmacological and non-pharmacological treatments focused on astrocytes as therapeutic targets for AD. These interventions include effects on anti-inflammatory/antioxidant systems, glutamate activity, lipid metabolism, neurovascular coupling and glymphatic system, calcium dysregulation, and in the release of peptides which affects glial and neuronal function. According to the AD stage, these therapies may be of benefit in either preventing or delaying the progression of the disease.
Collapse
Affiliation(s)
| | | | | | | | | | - Mauricio O. Nava-Mesa
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111711, Colombia
| |
Collapse
|
68
|
Cao S, Shi L, Shen Y, He L, Meng X. Ecological roles of secondary metabolites of Saposhnikovia divaricata in adaptation to drought stress. PeerJ 2022; 10:e14336. [PMID: 36353606 PMCID: PMC9639429 DOI: 10.7717/peerj.14336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 10/13/2022] [Indexed: 11/06/2022] Open
Abstract
Saposhnikovia divaricata is a traditional Chinese herb that mainly grows in arid grasslands and strongly adapts to various stresses. Drought is not only a major abiotic stress factor but also a typical feature conducive to producing high-quality medicinal material. The present study investigated by treating S. divaricata plants with polyethylene glycol (PEG-6000). Ultra-high performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF-MS) identified 146 compounds from the roots of S. divaricata, among which seven primary metabolites and 28 secondary metabolites showed significant changes after drought treatment. UV-Vis spectrophotometer detected the activity of antioxidant enzymes and the content of superoxide anion (O2 -.) and malondialdehyde (MDA). The differential primary metabolites revealed that drought promotes glycolysis, reducing primary metabolism and enhancing secondary metabolism. Meanwhile, the differential secondary metabolites showed an increase in the content of compounds upstream of the secondary metabolic pathway, and other glycosides and increased that of the corresponding aglycones. The activities of antioxidant enzymes and the content of O2 -. and MDA shown different changes duing the drought treatment. These observations indicate that drought promotes the biosynthesis and transformation of the secondary metabolites and activity of antioxidant enzymes, improving plant adaptability. The present study also analyzed a few primary and secondary metabolites of S. divaricata under different degrees and durations of drought and speculated on the metabolic pathways in an arid environment. The findings indicate the biological nature, diversity, and complexity of secondary metabolites and the mechanisms of plant adaptation to ecological stress.
Collapse
Affiliation(s)
- Sisi Cao
- Department of Pharmacognosy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
- Medical College, Harbin Vocational & Technical College, Harbin, Heilongjiang, China
| | - Lei Shi
- Department of Pharmacognosy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Ying Shen
- Department of Pharmacognosy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Luwen He
- Department of Pharmacognosy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Xiangcai Meng
- Department of Pharmacognosy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| |
Collapse
|
69
|
Inhibitory effect of protocatechualdehyde on Yersinia enterocolitica and its critical virulence factors. Microb Pathog 2022; 173:105877. [DOI: 10.1016/j.micpath.2022.105877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/23/2022] [Accepted: 11/06/2022] [Indexed: 11/11/2022]
|
70
|
Li F, Tang Y, Wei L, Yang M, Lu Z, Shi F, Zhan F, Li Y, Liao W, Lin L, Qin Z. Alginate oligosaccharide modulates immune response, fat metabolism, and the gut bacterial community in grass carp (Ctenopharyngodon idellus). FISH & SHELLFISH IMMUNOLOGY 2022; 130:103-113. [PMID: 36044935 DOI: 10.1016/j.fsi.2022.08.067] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/23/2022] [Accepted: 08/23/2022] [Indexed: 06/15/2023]
Abstract
Alginate oligosaccharide (AOS) is widely used in agriculture because of its many excellent biological properties. However, the possible beneficial effects of AOS and their underlying mechanisms are currently not well known in grass carp (Ctenopharyngodon idellus). Here, grass carp were fed diets supplemented with 5, 10, or 20 g/kg AOS for six weeks. HE and PAS staining showed that the diets of AOS significantly increased the number of goblet cells in the intestinal. According to transcriptome and quantitative real-time PCR (qRT-PCR) data, AOS-supplemented diets activated the expression of fat metabolism-related pathways and genes. The 16S rRNA sequencing results showed that supplementation with AOS affected the distribution and abundance of the gut bacterial assembly. qRT-PCR and activity assays revealed that the AOS diets significantly increased the antioxidant resistance in gut of grass carp, and down-regulated the expression of inflammatory and up-regulated anti-inflammatory cytokines. Finally, the Aeromonas hydrophila infection assay suggested that the mortality in the groups fed dietary AOS was slightly lower than that in the control. Therefore, supplementing the diet of grass carp with an appropriate amount of AOS can improve fat metabolism and immune responses and alter the intestinal bacterial community, which may help to fight bacterial infection.
Collapse
Affiliation(s)
- Fenglin Li
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Ying Tang
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Lixiang Wei
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Minxuan Yang
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Zhijie Lu
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Fei Shi
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Fanbin Zhan
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Yanan Li
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Wenchong Liao
- South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301, China
| | - Li Lin
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China; Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, 524025, China.
| | - Zhendong Qin
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China.
| |
Collapse
|
71
|
Song T, Zhou M, Li W, Lv M, Zheng L, Zhao M. The anti-inflammatory effect of vasoactive peptides from soybean protein hydrolysates by mediating serum extracellular vesicles-derived miRNA-19b/CYLD/TRAF6 axis in the vascular microenvironment of SHRs. Food Res Int 2022; 160:111742. [DOI: 10.1016/j.foodres.2022.111742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 07/21/2022] [Accepted: 07/24/2022] [Indexed: 11/28/2022]
|
72
|
Muscone suppresses myocardial ischemia damage by regulating PI3K/Akt signaling pathway. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166539. [PMID: 36100155 DOI: 10.1016/j.bbadis.2022.166539] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/26/2022] [Accepted: 09/07/2022] [Indexed: 11/21/2022]
Abstract
Muscone is the main active compound of Moschus. In this paper, the cardioprotective effect of Muscone on acute myocardial ischemia (AMI) rats and its potential mechanisms were investigated. AMI rat models were established to evaluate the protective effect and antioxidative function of Muscone on the hearts. Moreover, Western blot analysis was conducted to quantify the phosphorylated PI3K and AKT levels in PI3K/Akt pathway for further investigating the mechanism of Muscone. Results showed that Muscone could markedly lessen the infarct size and myocardial injury, improve cardiac function, inhibit cardiomyocyte apoptosis and down-regulate serum reactive oxygen species level as indicated by the decreased MDA, BNP and c-TnI activities and the increased SOD, GSH-px, CAT activities and the expression of Bax protein. In addition, it was revealed that Muscone notably promoted the phosphorylation of PI3K and AKT. These findings denote that Muscone exerts a protective effect in heart via inhibition of oxidative stress and apoptosis, offering new insights into the treatment of CHD and the clinical application of Muscone.
Collapse
|
73
|
Wang X, Sun B, Ye Z, Zhang W, Xu W, Gao S, Zhou N, Wu F, Shen J. Enzyme-Responsive COF-Based Thiol-Targeting Nanoinhibitor for Curing Bacterial Infections. ACS APPLIED MATERIALS & INTERFACES 2022; 14:38483-38496. [PMID: 35989491 DOI: 10.1021/acsami.2c08845] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Pathogen infections impose severe challenges in clinical practice, especially for patients infected with antibiotic-resistant microbes. The thioredoxin (Trx) system in Gram-positive bacteria serves as an ideal antimicrobial target for novel medicine design due to the structural differences from corresponding system in mammals. However, a backup thiol-dependent antioxidant glutathione (GSH) system limits the effectiveness of drugs in many Gram-negative bacteria. Herein, we synthesize a thiol-targeting nanoinhibitor based on an enzyme-responsive covalent organic framework (COF) coloaded with silver nanoparticles (AgNPs) and ebselen (EBS) (Ag-TA-CON@EBS@PEG) to exert synergistic antibacterial effects. Since azoreductase can dissociate the enzyme-responsive COF, we adopt this strategy to achieve the accurate release of EBS and Ag+ at infection sites. Our research identifies that the functionalized nanoinhibitor shows excellent bactericidal performance for Gram-positive and Gram-negative bacteria in vitro and exhibits low toxicity to normal cells. Besides, the nanoinhibitor presents favorable biocompatibility, anti-inflammatory property, and effective wound healing ability in mice. This paper provides a promising clinical strategy for synergistic antibacterial therapy and enhanced wound healing properties via an optimized combination of the targeted nanomedicines with an intelligent drug conveying platform.
Collapse
Affiliation(s)
- Xinye Wang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Baohong Sun
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Ziqiu Ye
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Wenjia Zhang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Wang Xu
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Shurui Gao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Ninglin Zhou
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Fan Wu
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Jian Shen
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| |
Collapse
|
74
|
Tang X, Li J, Liu L, Jing H, Zuo W, Zeng Y. Transcriptome Analysis Provides Insights into Potentilla bifurca Adaptation to High Altitude. Life (Basel) 2022; 12:life12091337. [PMID: 36143374 PMCID: PMC9503701 DOI: 10.3390/life12091337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 08/25/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022] Open
Abstract
Potentilla bifurca is widely distributed in Eurasia, including the Tibetan Plateau. It is a valuable medicinal plant in the Tibetan traditional medicine system, especially for the treatment of diabetes. This study investigated the functional gene profile of Potentilla bifurca at different altitudes by RNA-sequencing technology, including de novo assembly of 222,619 unigenes from 405 million clean reads, 57.64% of which were annotated in Nr, GO, KEGG, Pfam, and Swiss-Prot databases. The most significantly differentially expressed top 50 genes in the high-altitude samples were derived from plants that responded to abiotic stress, such as peroxidase, superoxide dismutase protein, and the ubiquitin-conjugating enzyme. Pathway analysis revealed that a large number of DEGs encode key enzymes involved in secondary metabolites, including phenylpropane and flavonoids. In addition, a total of 298 potential genomic SSRs were identified in this study, which provides information on the development of functional molecular markers for genetic diversity assessment. In conclusion, this study provides the first comprehensive assessment of the Potentilla bifurca transcriptome. This provides new insights into coping mechanisms for non-model organisms surviving in harsh environments at high altitudes, as well as molecular evidence for the selection of superior medicinal plants.
Collapse
Affiliation(s)
- Xun Tang
- College of Life Sciences, Qinghai Normal University, Xining 810008, China
- Academy of Plateau Science and Sustainability, Qinghai Normal University, Xining 810008, China
- College of Life Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Jinping Li
- College of Life Sciences, Qinghai Normal University, Xining 810008, China
- Academy of Plateau Science and Sustainability, Qinghai Normal University, Xining 810008, China
| | - Likuan Liu
- College of Life Sciences, Qinghai Normal University, Xining 810008, China
- Academy of Plateau Science and Sustainability, Qinghai Normal University, Xining 810008, China
| | - Hui Jing
- Qinghai Agricultural Technology Extension Station, Xining 810007, China
| | - Wenming Zuo
- College of Life Sciences, Qinghai Normal University, Xining 810008, China
| | - Yang Zeng
- College of Life Sciences, Qinghai Normal University, Xining 810008, China
- Academy of Plateau Science and Sustainability, Qinghai Normal University, Xining 810008, China
- Correspondence:
| |
Collapse
|
75
|
Bordin A, Chirivì M, Pagano F, Milan M, Iuliano M, Scaccia E, Fortunato O, Mangino G, Dhori X, De Marinis E, D'Amico A, Miglietta S, Picchio V, Rizzi R, Romeo G, Pulcinelli F, Chimenti I, Frati G, De Falco E. Human platelet lysate-derived extracellular vesicles enhance angiogenesis through miR-126. Cell Prolif 2022; 55:e13312. [PMID: 35946052 DOI: 10.1111/cpr.13312] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 06/08/2022] [Accepted: 06/23/2022] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES Extracellular vesicles (EVs) are key biological mediators of several physiological functions within the cell microenvironment. Platelets are the most abundant source of EVs in the blood. Similarly, platelet lysate (PL), the best platelet derivative and angiogenic performer for regenerative purposes, is enriched of EVs, but their role is still too poorly discovered to be suitably exploited. Here, we explored the contribution of the EVs in PL, by investigating the angiogenic features extrapolated from that possessed by PL. METHODS We tested angiogenic ability and molecular cargo in 3D bioprinted models and by RNA sequencing analysis of PL-derived EVs. RESULTS A subset of small vesicles is highly represented in PL. The EVs do not retain aggregation ability, preserving a low redox state in human umbilical vein endothelial cells (HUVECs) and increasing the angiogenic tubularly-like structures in 3D endothelial bioprinted constructs. EVs resembled the miRNome profile of PL, mainly enriched with small RNAs and a high amount of miR-126, the most abundant angiogenic miRNA in platelets. The transfer of miR-126 by EVs in HUVEC after the in vitro inhibition of the endogenous form, restored angiogenesis, without involving VEGF as a downstream target in this system. CONCLUSION PL is a biological source of available EVs with angiogenic effects involving a miRNAs-based cargo. These properties can be exploited for targeted molecular/biological manipulation of PL, by potentially developing a product exclusively manufactured of EVs.
Collapse
Affiliation(s)
- Antonella Bordin
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Maila Chirivì
- Department of Pathophysiology and Transplantation, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesca Pagano
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), Monterotondo, Rome, Italy
| | - Marika Milan
- UOC Neurologia, Fondazione Ca'Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Marco Iuliano
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Eleonora Scaccia
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- Institute of Transfusion Medicine and Immunology, Mannheim Institute of Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Orazio Fortunato
- Tumor Genomics Unit, Department of Research, IRCCS Fondazione Istituto Nazionale dei Tumori, Milan, Italy
| | - Giorgio Mangino
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Xhulio Dhori
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Elisabetta De Marinis
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Alessandra D'Amico
- Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", Rome, Italy
| | - Selenia Miglietta
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, La Sapienza University of Rome, Rome, Italy
| | - Vittorio Picchio
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Roberto Rizzi
- Istituto Nazionale Genetica Molecolare INGM 'Romeo ed Enrica Invernizzi', Milan, Italy
- Institute of Biomedical Technologies, National Research Council of Italy (ITB-CNR), Milan, Italy
| | - Giovanna Romeo
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Fabio Pulcinelli
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Isotta Chimenti
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- Mediterranea Cardiocentro, Naples, Italy
| | - Giacomo Frati
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- Department of AngioCardioNeurology, IRCCS Neuromed, Pozzili, Italy
| | - Elena De Falco
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- Mediterranea Cardiocentro, Naples, Italy
| |
Collapse
|
76
|
Liu Z, Guo S, Fang X, Shao X, Zhao Z. Antibacterial and plant growth-promoting properties of novel Fe 3O 4/Cu/CuO magnetic nanoparticles. RSC Adv 2022; 12:19856-19867. [PMID: 35865197 PMCID: PMC9260745 DOI: 10.1039/d2ra03114k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 06/29/2022] [Indexed: 11/21/2022] Open
Abstract
In this work, an Fe3O4/Cu/CuO (FC) antibacterial nano-agent was synthesized in a "one-pot" approach using copper sulfate and ferric chloride as raw materials, and it was studied using TEM, XRD, XPS, UV-vis, and VSM methods. The antibacterial activity and mechanism of FC were studied, using a commercially available Bordeaux mixture as a control. The effects of an FC on mung bean development and its toxicity to human mammary epithelial cells were also investigated. The results revealed that FC could break the cell walls of E. coli and S. aureus, quadrupling the antibacterial activity of the Bordeaux combination. Furthermore, it was shown that FC might improve the germination, root development, and chlorophyll content of mung bean seeds while being 1/8 as hazardous to human mammary epithelial cells as the Bordeaux combination. The as-prepared FC can replace the Bordeaux combination in the management of agroforestry pathogens.
Collapse
Affiliation(s)
- Zhifeng Liu
- Shaanxi Key Laboratory of Catalysis, School of Chemical & Environment Science, Shaanxi University of Technology Hanzhong Shaanxi 723001 China +86-0916-2641660 +86-0916-2641660
- State Key Laboratory of Qinba Bio-Resource and Ecological Environment, Shaanxi University of Technology Hanzhong Shaanxi 723001 China
| | - Shaobo Guo
- Shaanxi Key Laboratory of Catalysis, School of Chemical & Environment Science, Shaanxi University of Technology Hanzhong Shaanxi 723001 China +86-0916-2641660 +86-0916-2641660
- State Key Laboratory of Qinba Bio-Resource and Ecological Environment, Shaanxi University of Technology Hanzhong Shaanxi 723001 China
| | - Xun Fang
- Shaanxi Key Laboratory of Catalysis, School of Chemical & Environment Science, Shaanxi University of Technology Hanzhong Shaanxi 723001 China +86-0916-2641660 +86-0916-2641660
| | - Xianzhao Shao
- Shaanxi Key Laboratory of Catalysis, School of Chemical & Environment Science, Shaanxi University of Technology Hanzhong Shaanxi 723001 China +86-0916-2641660 +86-0916-2641660
- State Key Laboratory of Qinba Bio-Resource and Ecological Environment, Shaanxi University of Technology Hanzhong Shaanxi 723001 China
| | - Zuoping Zhao
- Shaanxi Key Laboratory of Catalysis, School of Chemical & Environment Science, Shaanxi University of Technology Hanzhong Shaanxi 723001 China +86-0916-2641660 +86-0916-2641660
- State Key Laboratory of Qinba Bio-Resource and Ecological Environment, Shaanxi University of Technology Hanzhong Shaanxi 723001 China
| |
Collapse
|
77
|
Ellistasari EY, Kariosentono H, Purwanto B, Wasita B, Riswiyant RCA, Pamungkasari EP, Soetrisno S. Role of Exosomes Derived from Secretome Human Umbilical Vein Endothelial Cells (Exo-HUVEC) as Anti-Apoptotic, Anti-Oxidant, and Increasing Fibroblast Migration in Photoaging Skin Models. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.9969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background: Prolonged skin exposure to ultraviolet light rays leads to photoaging, which is characterized molecularly by an increase in reactive oxygen species (ROS), cell apoptosis, and a decrease in collagen. Photoaging therapy has been a challenge until recently. Fibroblasts exposed to ultraviolet B (UVB) light proved to be a good model for photoaging skin. They are also the primary dermal cells that stimulate collagen production and extracellular matrix (ECM), which contribute to skin aging. Exo-HUVEC is rich in growth factors, cytokines, and miRNAs, and they all play a vital role in cell-to-cell communication. The migration of fibroblasts is crucial for the development, repair, and regeneration of skin tissue during the repair of skin aging.
Objective: An in vitro experimental study was conducted to analyze the effect of Exo-HUVEC on oxidative stress levels, cell apoptosis, and fibroblast migration rate after UVB ray exposure on fibroblasts.
Methods: The fibroblast cultures were divided into five groups, including one without UVB exposure, one with UVB exposure, and one with UVB+Exo-HUVEC exposure at 0.1%, 0.5%, and 1%, respectively. Oxidative stress levels were measured using the ELISA test for malondialdehyde (MDA). Furthermore, flow cytometry was used to measure apoptosis using PI/Annexin markers, while a scratch assay examination was used to measure fibroblast migration rate using imaging readings.
Results: There were significant differences in the levels of MDA, PI/Annexin, and the rate of fibroblast migration between the UVB-irradiated control group and the Exo-HUVEC treatment group (p<0.001).
Conclusion: Exo-HUVEC is a marker of photoaging improvement, which has anti-apoptotic effects and reduces oxidative stress, as well as increases fibroblast migration rate.
Collapse
|
78
|
Li M, Fang F, Sun M, Zhang Y, Hu M, Zhang J. Extracellular vesicles as bioactive nanotherapeutics: An emerging paradigm for regenerative medicine. Theranostics 2022; 12:4879-4903. [PMID: 35836815 PMCID: PMC9274746 DOI: 10.7150/thno.72812] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 06/05/2022] [Indexed: 01/12/2023] Open
Abstract
In recent decades, extracellular vesicles (EVs), as bioactive cell-secreted nanoparticles which are involved in various physiological and pathological processes including cell proliferation, immune regulation, angiogenesis and tissue repair, have emerged as one of the most attractive nanotherapeutics for regenerative medicine. Herein we provide a systematic review of the latest progress of EVs for regenerative applications. Firstly, we will briefly introduce the biogenesis, function and isolation technology of EVs. Then, the underlying therapeutic mechanisms of the native unmodified EVs and engineering strategies of the modified EVs as regenerative entities will be discussed. Subsequently, the main focus will be placed on the tissue repair and regeneration applications of EVs on various organs including brain, heart, bone and cartilage, liver and kidney, as well as skin. More importantly, current clinical trials of EVs for regenerative medicine will also be briefly highlighted. Finally, the future challenges and insightful perspectives of the currently developed EV-based nanotherapeutics in biomedicine will be discussed. In short, the bioactive EV-based nanotherapeutics have opened new horizons for biologists, chemists, nanoscientists, pharmacists, as well as clinicians, making possible powerful tools and therapies for regenerative medicine.
Collapse
Affiliation(s)
- Min Li
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Sciences, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Fang Fang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Sciences, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Meng Sun
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Sciences, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Yinfeng Zhang
- International Medical Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, P. R. China
| | - Min Hu
- Department of Hepatobiliary Surgery, Jinan University First Affiliated Hospital, Guangzhou, 510630, P. R. China
| | - Jinfeng Zhang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Sciences, Beijing Institute of Technology, Beijing 100081, P. R. China
| |
Collapse
|
79
|
Qin Y, Fan Y, Chen R, Yin H, Zou H, Qu X, Tan J, Xu Y, Zhu C. Harnessing Oxidative Microenvironment for In Vivo Synthesis of Subcellular Conductive Polymer Microesicles Enhances Nerve Reconstruction. NANO LETTERS 2022; 22:3825-3831. [PMID: 35499361 DOI: 10.1021/acs.nanolett.2c01123] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Conductive polymers (CPs) are promising biomaterials to address signal connection at biointerfaces for tissue regeneration. However, regulating material microstructure at the subcellular scale to provide a more seamless interface between conductive substrates and cells remains a great challenge. Here, we demonstrate that chemical factors and enzyme-carried subcellular structures at lesion site provide a natural bioreactor to self-assemble conductive microvesicles (CMVs) for improving bioelectrical signal reconstruction. The synthesized CMVs contribute to the electrical conduction of the injured nerve in the early stage. Moreover, CMVs are eventually expelled via lymphatic capillary to minimize space-occupying and chronic inflammation. Therefore, we provide a prototype to integrate specific physiological microenvironments and polymer chemistry to manufacture subcellular functional materials with self-adaptive interface in vivo for biomedical applications.
Collapse
Affiliation(s)
- Yinhua Qin
- Department of Anatomy, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yonghong Fan
- Department of Anatomy, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Ruyue Chen
- Department of Anatomy, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Haiyan Yin
- Department of Anatomy, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Hao Zou
- Department of Anatomy, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Xiaohang Qu
- Department of Anatomy, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Ju Tan
- Department of Anatomy, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Youqian Xu
- Department of Anatomy, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Chuhong Zhu
- Department of Anatomy, Army Medical University (Third Military Medical University), Chongqing 400038, China
| |
Collapse
|
80
|
Zhao P, Liu D, Hu H, Qiu Z, Liang Y, Chen Z. Anticancer activity of four trinuclear cobalt complexes bearing bis(salicylidene)-1,3-propanediamine derivatives. J Inorg Biochem 2022; 233:111860. [DOI: 10.1016/j.jinorgbio.2022.111860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/17/2022] [Accepted: 05/08/2022] [Indexed: 10/18/2022]
|
81
|
Qu P, Zhao J, Hu H, Cao W, Zhang Y, Qi J, Meng B, Zhao J, Liu S, Ding C, Wu Y, Liu E. Loss of Renewal of Extracellular Vesicles: Harmful Effects on Embryo Development in vitro. Int J Nanomedicine 2022; 17:2301-2318. [PMID: 35615541 PMCID: PMC9126234 DOI: 10.2147/ijn.s354003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 05/09/2022] [Indexed: 12/12/2022] Open
Abstract
Background Methods Results Conclusion
Collapse
Affiliation(s)
- Pengxiang Qu
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Centre, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an, Shaanxi, People's Republic of China
| | - Jinpeng Zhao
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Centre, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an, Shaanxi, People's Republic of China
| | - Huizhong Hu
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Centre, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an, Shaanxi, People's Republic of China
| | - Wenbin Cao
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Centre, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an, Shaanxi, People's Republic of China
| | - Yanru Zhang
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Centre, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an, Shaanxi, People's Republic of China
| | - Jia Qi
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Centre, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an, Shaanxi, People's Republic of China
| | - Bin Meng
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Centre, Xi’an, Shaanxi, People’s Republic of China
- The Assisted Reproduction Center, Northwest Women’s and Children’s Hospital, Xi’an, People’s Republic of China
| | - Juan Zhao
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an, Shaanxi, People's Republic of China
- Department of Hematology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Shuangqing Liu
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Centre, Xi’an, Shaanxi, People’s Republic of China
| | - Chong Ding
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Centre, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an, Shaanxi, People's Republic of China
| | - Yuqi Wu
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Centre, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an, Shaanxi, People's Republic of China
| | - Enqi Liu
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Centre, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an, Shaanxi, People's Republic of China
- Correspondence: Enqi Liu, Email
| |
Collapse
|
82
|
Sanmartin MC, Borzone FR, Giorello MB, Yannarelli G, Chasseing NA. Mesenchymal Stromal Cell-Derived Extracellular Vesicles as Biological Carriers for Drug Delivery in Cancer Therapy. Front Bioeng Biotechnol 2022; 10:882545. [PMID: 35497332 PMCID: PMC9046597 DOI: 10.3389/fbioe.2022.882545] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 03/25/2022] [Indexed: 12/11/2022] Open
Abstract
Cancer is the second leading cause of death worldwide, with 10.0 million cancer deaths in 2020. Despite advances in targeted therapies, some pharmacological drawbacks associated with anticancer chemo and immunotherapeutic agents include high toxicities, low bioavailability, and drug resistance. In recent years, extracellular vesicles emerged as a new promising platform for drug delivery, with the advantage of their inherent biocompatibility and specific targeting compared to artificial nanocarriers, such as liposomes. Particularly, mesenchymal stem/stromal cells were proposed as a source of extracellular vesicles for cancer therapy because of their intrinsic properties: high in vitro self-renewal and proliferation, regenerative and immunomodulatory capacities, and secretion of extracellular vesicles that mediate most of their paracrine functions. Moreover, extracellular vesicles are static and safer in comparison with mesenchymal stem/stromal cells, which can undergo genetic/epigenetic or phenotypic changes after their administration to patients. In this review, we summarize currently reported information regarding mesenchymal stem/stromal cell-derived extracellular vesicles, their proper isolation and purification techniques - from either naive or engineered mesenchymal stem/stromal cells - for their application in cancer therapy, as well as available downstream modification methods to improve their therapeutic properties. Additionally, we discuss the challenges associated with extracellular vesicles for cancer therapy, and we review some preclinical and clinical data available in the literature.
Collapse
Affiliation(s)
- María Cecilia Sanmartin
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Laboratorio de Regulación Génica y Células Madre, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMeTTyB), Universidad Favaloro - CONICET, Buenos Aires, Argentina
| | - Francisco Raúl Borzone
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - María Belén Giorello
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Gustavo Yannarelli
- Laboratorio de Regulación Génica y Células Madre, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMeTTyB), Universidad Favaloro - CONICET, Buenos Aires, Argentina
| | - Norma Alejandra Chasseing
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
83
|
Las Heras K, Royo F, Garcia-Vallicrosa C, Igartua M, Santos-Vizcaino E, Falcon-Perez JM, Hernandez RM. Extracellular vesicles from hair follicle-derived mesenchymal stromal cells: isolation, characterization and therapeutic potential for chronic wound healing. Stem Cell Res Ther 2022; 13:147. [PMID: 35395929 PMCID: PMC8994406 DOI: 10.1186/s13287-022-02824-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/23/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs) and their extracellular vesicles (MSC-EVs) have demonstrated to elicit immunomodulatory and pro-regenerative properties that are beneficial for the treatment of chronic wounds. Thanks to different mediators, MSC-EVs have shown to play an important role in the proliferation, migration and cell survival of different skin cell populations. However, there is still a big bid to achieve the most effective, suitable and available source of MSC-EVs. METHODS We isolated, characterized and compared medium-large EVs (m-lEVs) and small EVs (sEVs) obtained from hair follicle-derived MSCs (HF-MSCs) against the gold standard in regenerative medicine, EVs isolated from adipose tissue-derived MSCs (AT-MSCs). RESULTS We demonstrated that HF-EVs, as well as AT-EVs, expressed typical MSC-EVs markers (CD9, CD44, CD63, CD81 and CD105) among other different functional markers. We showed that both cell types were able to increase human dermal fibroblasts (HDFs) proliferation and migration. Moreover, both MSC-EVs were able to increase angiogenesis in human umbilical vein endothelial cells (HUVECs) and protect HDFs exposed to a hyperglycemic environment from oxidative stress and cytotoxicity. CONCLUSIONS Taken together, HF-EVs demonstrated to exhibit comparable potential to that of AT-EVs as promising candidates in the treatment of chronic wounds.
Collapse
Affiliation(s)
- Kevin Las Heras
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy (UPV/EHU), 01006, Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Félix Royo
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Exosomes Laboratory, 48160, Derio, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas Y Digestivas (CIBERehd), 28029, Madrid, Spain
| | - Clara Garcia-Vallicrosa
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Exosomes Laboratory, 48160, Derio, Spain
| | - Manoli Igartua
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy (UPV/EHU), 01006, Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029, Madrid, Spain
| | - Edorta Santos-Vizcaino
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy (UPV/EHU), 01006, Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029, Madrid, Spain
| | - Juan M Falcon-Perez
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Exosomes Laboratory, 48160, Derio, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas Y Digestivas (CIBERehd), 28029, Madrid, Spain.
- IKERBASQUE, Basque Foundation for Science, 48013, Bilbao, Spain.
| | - Rosa Maria Hernandez
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy (UPV/EHU), 01006, Vitoria-Gasteiz, Spain.
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain.
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029, Madrid, Spain.
| |
Collapse
|
84
|
Xia X, Zhang L, Wu H, Chen F, Liu X, Xu H, Cui Y, Zhu Q, Wang M, Hao H, Li DP, Fay WP, Martinez-Lemus LA, Hill MA, Xu C, Liu Z. CagA+Helicobacter pylori, Not CagA–Helicobacter pylori, Infection Impairs Endothelial Function Through Exosomes-Mediated ROS Formation. Front Cardiovasc Med 2022; 9:881372. [PMID: 35433874 PMCID: PMC9008404 DOI: 10.3389/fcvm.2022.881372] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/08/2022] [Indexed: 12/12/2022] Open
Abstract
BackgroundHelicobacter pylori (H. pylori) infection increases the risk for atherosclerosis, and ROS are critical to endothelial dysfunction and atherosclerosis. CagA is a major H. pylori virulence factor associated with atherosclerosis. The present study aimed to test the hypothesis that CagA+H. pylori effectively colonizes gastric mucosa, and CagA+H. pylori, but not CagA–H. pylori, infection impairs endothelial function through exosomes-mediated ROS formation.MethodsC57BL/6 were used to determine the colonization ability of CagA+H. pylori and CagA–H. pylori. ROS production, endothelial function of thoracic aorta and atherosclerosis were measured in CagA+H. pylori and CagA–H. pylori infected mice. Exosomes from CagA+H. pylori and CagA–H. pylori or without H. pylori infected mouse serum or GES-1 were isolated and co-cultured with bEND.3 and HUVECs to determine how CagA+H. pylori infection impairs endothelial function. Further, GW4869 was used to determine if CagA+H. pylori infection could lead to endothelial dysfunction and atherosclerosis through an exosomes-mediated mechanism.ResultsCagA+H. pylori colonized gastric mucosa more effectively than CagA–H. pylori in mice. CagA+H. pylori, not CagA–H. pylori, infection significantly increased aortic ROS production, decreased ACh-induced aortic relaxation, and enhanced early atherosclerosis formation, which were prevented with N-acetylcysteine treatment. Treatment with CagA-containing exosomes significantly increased intracellular ROS production in endothelial cells and impaired their function. Inhibition of exosomes secretion with GW4869 effectively prevented excessive aortic ROS production, endothelial dysfunction, and atherosclerosis in mice with CagA+H. pylori infection.ConclusionThese data suggest that CagA+H. pylori effectively colonizes gastric mucosa, impairs endothelial function, and enhances atherosclerosis via exosomes-mediated ROS formation in mice.
Collapse
Affiliation(s)
- Xiujuan Xia
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, United States
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Linfang Zhang
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, United States
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Hao Wu
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, United States
| | - Feng Chen
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, United States
| | - Xuanyou Liu
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, United States
| | - Huifang Xu
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, United States
| | - Yuqi Cui
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, United States
| | - Qiang Zhu
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, United States
| | - Meifang Wang
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, United States
| | - Hong Hao
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, United States
| | - De-Pei Li
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, United States
| | - William P. Fay
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, United States
| | - Luis A. Martinez-Lemus
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, United States
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
- Department of Medical Pharmacology and Physiology, Columbia, MO, United States
| | - Michael A. Hill
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
- Department of Medical Pharmacology and Physiology, Columbia, MO, United States
| | - Canxia Xu
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhenguo Liu
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, United States
- *Correspondence: Zhenguo Liu,
| |
Collapse
|
85
|
Reymond S, Vujić T, Schvartz D, Sanchez JC. Morphine-induced modulation of Nrf2-antioxidant response element signaling pathway in primary human brain microvascular endothelial cells. Sci Rep 2022; 12:4588. [PMID: 35301408 PMCID: PMC8931063 DOI: 10.1038/s41598-022-08712-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 03/07/2022] [Indexed: 11/09/2022] Open
Abstract
Morphine is one of the most potent opioid analgesic used for pain treatment. Morphine action in the central nervous system requires crossing the blood-brain barrier. Due to the controversial relationship between morphine and oxidative stress, the potential pro- or antioxidant effects of morphine in the blood-brain barrier is important to be understood, as oxidative stress could cause its disruption and predispose to neurodegenerative diseases. However, investigation is scarce in human brain endothelial cells. Therefore, the present study evaluated the impact of morphine exposure at three different concentrations (1, 10 and 100 µM) for 24 h and 48 h on primary human brain microvascular endothelial cells. A quantitative data-independent acquisition mass spectrometry strategy was used to analyze proteome modulations. Almost 3000 proteins were quantified of which 217 were reported to be significantly regulated in at least one condition versus untreated control. Pathway enrichment analysis unveiled dysregulation of the Nrf2 pathway involved in oxidative stress response. Seahorse assay underlined mitochondria dysfunctions, which were supported by significant expression modulations of relevant mitochondrial proteins. In conclusion, our study revealed the dysregulation of the Nrf2 pathway and mitochondria dysfunctions after morphine exposure, highlighting a potential redox imbalance in human brain endothelial cells.
Collapse
Affiliation(s)
- Sandrine Reymond
- Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Swiss Center for Applied Human Toxicology, Geneva, Switzerland
| | - Tatjana Vujić
- Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Swiss Center for Applied Human Toxicology, Geneva, Switzerland
| | - Domitille Schvartz
- Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Swiss Center for Applied Human Toxicology, Geneva, Switzerland
| | - Jean-Charles Sanchez
- Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland. .,Swiss Center for Applied Human Toxicology, Geneva, Switzerland.
| |
Collapse
|
86
|
Yaker L, Tebani A, Lesueur C, Dias C, Jung V, Bekri S, Guerrera IC, Kamel S, Ausseil J, Boullier A. Extracellular Vesicles From LPS-Treated Macrophages Aggravate Smooth Muscle Cell Calcification by Propagating Inflammation and Oxidative Stress. Front Cell Dev Biol 2022; 10:823450. [PMID: 35356285 PMCID: PMC8959646 DOI: 10.3389/fcell.2022.823450] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 02/09/2022] [Indexed: 12/29/2022] Open
Abstract
Background: Vascular calcification (VC) is a cardiovascular complication associated with a high mortality rate among patients with diseases such as atherosclerosis and chronic kidney disease. During VC, vascular smooth muscle cells (VSMCs) undergo an osteogenic switch and secrete a heterogeneous population of extracellular vesicles (EVs). Recent studies have shown involvement of EVs in the inflammation and oxidative stress observed in VC. We aimed to decipher the role and mechanism of action of macrophage-derived EVs in the propagation of inflammation and oxidative stress on VSMCs during VC. Methods: The macrophage murine cell line RAW 264.7 treated with lipopolysaccharide (LPS-EK) was used as a cellular model for inflammatory and oxidative stress. EVs secreted by these macrophages were collected by ultracentrifugation and characterized by transmission electron microscopy, cryo-electron microscopy, nanoparticle tracking analysis, and the analysis of acetylcholinesterase activity, as well as that of CD9 and CD81 protein expression by western blotting. These EVs were added to a murine VSMC cell line (MOVAS-1) under calcifying conditions (4 mM Pi—7 or 14 days) and calcification assessed by the o-cresolphthalein calcium assay. EV protein content was analyzed in a proteomic study and EV cytokine content assessed using an MSD multiplex immunoassay. Results: LPS-EK significantly decreased macrophage EV biogenesis. A 24-h treatment of VSMCs with these EVs induced both inflammatory and oxidative responses. LPS-EK-treated macrophage-derived EVs were enriched for pro-inflammatory cytokines and CAD, PAI-1, and Saa3 proteins, three molecules involved in inflammation, oxidative stress, and VC. Under calcifying conditions, these EVs significantly increase the calcification of VSMCs by increasing osteogenic markers and decreasing contractile marker expression. Conclusion: Our results show that EVs derived from LPS-EK–treated-macrophages are able to induce pro-inflammatory and pro-oxidative responses in surrounding cells, such as VSMCs, thus aggravating the VC process.
Collapse
Affiliation(s)
- Linda Yaker
- MP3CV-UR7517, CURS-University of Picardie Jules Verne, Amiens, France
| | - Abdellah Tebani
- INSERM U1245, CHU Rouen, Normandie University, UNIROUEN, Rouen, France
- Department of Metabolic Biochemistry, Rouen University Hospital, Rouen, France
| | - Céline Lesueur
- INSERM U1245, CHU Rouen, Normandie University, UNIROUEN, Rouen, France
| | - Chloé Dias
- Infinity, INSERM UMR1291, CNRS UMR5051, University of Toulouse III, Toulouse, France
| | - Vincent Jung
- INSERM US24/CNRS UAR3633, Proteomic Platform Necker, University of Paris—Federative Research Structure Necker, Paris, France
| | - Soumeya Bekri
- INSERM U1245, CHU Rouen, Normandie University, UNIROUEN, Rouen, France
- Department of Metabolic Biochemistry, Rouen University Hospital, Rouen, France
| | - Ida Chiara Guerrera
- INSERM US24/CNRS UAR3633, Proteomic Platform Necker, University of Paris—Federative Research Structure Necker, Paris, France
| | - Saïd Kamel
- MP3CV-UR7517, CURS-University of Picardie Jules Verne, Amiens, France
- Laboratory of Biochemistry, CHU Amiens-Picardie, Amiens, France
| | - Jérôme Ausseil
- Infinity, INSERM UMR1291, CNRS UMR5051, University of Toulouse III, Toulouse, France
- Service de Biochimie, Institut Fédératif de Biologie, CHU Toulouse, Toulouse, France
| | - Agnès Boullier
- MP3CV-UR7517, CURS-University of Picardie Jules Verne, Amiens, France
- Laboratory of Biochemistry, CHU Amiens-Picardie, Amiens, France
- *Correspondence: Agnès Boullier,
| |
Collapse
|
87
|
Martinez-Banaclocha M. N-Acetyl-Cysteine: Modulating the Cysteine Redox Proteome in Neurodegenerative Diseases. Antioxidants (Basel) 2022; 11:antiox11020416. [PMID: 35204298 PMCID: PMC8869501 DOI: 10.3390/antiox11020416] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/13/2022] [Accepted: 02/16/2022] [Indexed: 12/14/2022] Open
Abstract
In the last twenty years, significant progress in understanding the pathophysiology of age-associated neurodegenerative diseases has been made. However, the prevention and treatment of these diseases remain without clinically significant therapeutic advancement. While we still hope for some potential genetic therapeutic approaches, the current reality is far from substantial progress. With this state of the issue, emphasis should be placed on early diagnosis and prompt intervention in patients with increased risk of neurodegenerative diseases to slow down their progression, poor prognosis, and decreasing quality of life. Accordingly, it is urgent to implement interventions addressing the psychosocial and biochemical disturbances we know are central in managing the evolution of these disorders. Genomic and proteomic studies have shown the high molecular intricacy in neurodegenerative diseases, involving a broad spectrum of cellular pathways underlying disease progression. Recent investigations indicate that the dysregulation of the sensitive-cysteine proteome may be a concurrent pathogenic mechanism contributing to the pathophysiology of major neurodegenerative diseases, opening new therapeutic opportunities. Considering the incidence and prevalence of these disorders and their already significant burden in Western societies, they will become a real pandemic in the following decades. Therefore, we propose large-scale investigations, in selected groups of people over 40 years of age with decreased blood glutathione levels, comorbidities, and/or mild cognitive impairment, to evaluate supplementation of the diet with low doses of N-acetyl-cysteine, a promising and well-tolerated therapeutic agent suitable for long-term use.
Collapse
|
88
|
Buffolo F, Monticone S, Camussi G, Aikawa E. Role of Extracellular Vesicles in the Pathogenesis of Vascular Damage. Hypertension 2022; 79:863-873. [PMID: 35144490 PMCID: PMC9010370 DOI: 10.1161/hypertensionaha.121.17957] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Extracellular vesicles (EVs) are nanosized membrane-bound structures released by cells that are able to transfer nucleic acids, protein cargos, and metabolites to specific recipient cells, allowing cell-to-cell communications in an endocrine and paracrine manner. Endothelial, leukocyte, and platelet-derived EVs have emerged both as biomarkers and key effectors in the development and progression of different stages of vascular damage, from earliest alteration of endothelial function, to advanced atherosclerotic lesions and cardiovascular calcification. Under pathological conditions, circulating EVs promote endothelial dysfunction by impairing vasorelaxation and instigate vascular inflammation by increasing levels of adhesion molecules, reactive oxygen species, and proinflammatory cytokines. Platelets, endothelial cells, macrophages, and foam cells secrete EVs that regulate macrophage polarization and contribute to atherosclerotic plaque progression. Finally, under pathological stimuli, smooth muscle cells and macrophages secrete EVs that aggregate between collagen fibers and serve as nucleation sites for ectopic mineralization in the vessel wall, leading to formation of micro- and macrocalcification. In this review, we summarize the emerging evidence of the pathological role of EVs in vascular damage, highlighting the major findings from the most recent studies and discussing future perspectives in this research field.
Collapse
Affiliation(s)
- Fabrizio Buffolo
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Italy. (F.B., S.M.).,Center for Interdisciplinary Cardiovascular Sciences, Department of Cardiovascular Medicine (F.B, E.A.)
| | - Silvia Monticone
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Italy. (F.B., S.M.)
| | - Giovanni Camussi
- Department of Medical Sciences, Molecular Biotechnology Center, University of Torino, Italy. (G.C.)
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Department of Cardiovascular Medicine (F.B, E.A.).,Center for Excellence in Vascular Biology, Department of Cardiovascular Medicine (E.A.)
| |
Collapse
|
89
|
Aparicio-Trejo OE, Aranda-Rivera AK, Osorio-Alonso H, Martínez-Klimova E, Sánchez-Lozada LG, Pedraza-Chaverri J, Tapia E. Extracellular Vesicles in Redox Signaling and Metabolic Regulation in Chronic Kidney Disease. Antioxidants (Basel) 2022; 11:antiox11020356. [PMID: 35204238 PMCID: PMC8868440 DOI: 10.3390/antiox11020356] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 01/26/2022] [Accepted: 02/03/2022] [Indexed: 12/17/2022] Open
Abstract
Chronic kidney disease (CKD) is a world health problem increasing dramatically. The onset of CKD is driven by several mechanisms; among them, metabolic reprogramming and changes in redox signaling play critical roles in the advancement of inflammation and the subsequent fibrosis, common pathologies observed in all forms of CKD. Extracellular vesicles (EVs) are cell-derived membrane packages strongly associated with cell-cell communication since they transfer several biomolecules that serve as mediators in redox signaling and metabolic reprogramming in the recipient cells. Recent studies suggest that EVs, especially exosomes, the smallest subtype of EVs, play a fundamental role in spreading renal injury in CKD. Therefore, this review summarizes the current information about EVs and their cargos’ participation in metabolic reprogramming and mitochondrial impairment in CKD and their role in redox signaling changes. Finally, we analyze the effects of these EV-induced changes in the amplification of inflammatory and fibrotic processes in the progression of CKD. Furthermore, the data suggest that the identification of the signaling pathways involved in the release of EVs and their cargo under pathological renal conditions can allow the identification of new possible targets of injury spread, with the goal of preventing CKD progression.
Collapse
Affiliation(s)
- Omar Emiliano Aparicio-Trejo
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología "Ignacio Chávez", Mexico City 14080, Mexico
| | - Ana Karina Aranda-Rivera
- Laboratorio F-315, Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Horacio Osorio-Alonso
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología "Ignacio Chávez", Mexico City 14080, Mexico
| | - Elena Martínez-Klimova
- Laboratorio F-315, Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Laura Gabriela Sánchez-Lozada
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología "Ignacio Chávez", Mexico City 14080, Mexico
| | - José Pedraza-Chaverri
- Laboratorio F-315, Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Edilia Tapia
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología "Ignacio Chávez", Mexico City 14080, Mexico
| |
Collapse
|
90
|
Zhang DM, Deng JJ, Wu YG, Tang T, Xiong L, Zheng YF, Xu XM. MicroRNA-223-3p Protect Against Radiation-Induced Cardiac Toxicity by Alleviating Myocardial Oxidative Stress and Programmed Cell Death via Targeting the AMPK Pathway. Front Cell Dev Biol 2022; 9:801661. [PMID: 35111759 PMCID: PMC8801819 DOI: 10.3389/fcell.2021.801661] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
Objectives: Radiotherapy improves the survival rate of cancer patients, yet it also involves some inevitable complications. Radiation-induced heart disease (RIHD) is one of the most serious complications, especially the radiotherapy of thoracic tumors, which is characterized by cardiac oxidative stress disorder and programmed cell death. At present, there is no effective treatment strategy for RIHD; in addition, it cannot be reversed when it progresses. This study aims to explore the role and potential mechanism of microRNA-223-3p (miR-223-3p) in RIHD.Methods: Mice were injected with miR-223-3p mimic, inhibitor, or their respective controls in the tail vein and received a single dose of 20 Gy whole-heart irradiation (WHI) for 16 weeks after 3 days to construct a RIHD mouse model. To inhibit adenosine monophosphate activated protein kinase (AMPK) or phosphodiesterase 4D (PDE4D), compound C (CompC) and AAV9-shPDE4D were used.Results: WHI treatment significantly inhibited the expression of miR-223-3p in the hearts; furthermore, the levels of miR-223-3p decreased in a radiation time-dependent manner. miR-223-3p mimic significantly relieved, while miR-223-3p inhibitor aggravated apoptosis, oxidative damage, and cardiac dysfunction in RIHD mice. In addition, we found that miR-223-3p mimic improves WHI-induced myocardial injury by activating AMPK and that the inhibition of AMPK by CompC completely blocks these protective effects of miR-223-3p mimic. Further studies found that miR-223-3p lowers the protein levels of PDE4D and inhibiting PDE4D by AAV9-shPDE4D blocks the WHI-induced myocardial injury mediated by miR-223-3p inhibitor.Conclusion: miR-223-3p ameliorates WHI-induced RIHD through anti-oxidant and anti-programmed cell death mechanisms via activating AMPK by PDE4D regulation. miR-223-3p mimic exhibits potential value in the treatment of RIHD.
Collapse
Affiliation(s)
- Dao-ming Zhang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jun-jian Deng
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yao-gui Wu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tian Tang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lin Xiong
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yong-fa Zheng
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
- *Correspondence: Yong-fa Zheng, ; Xi-ming Xu,
| | - Xi-ming Xu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
- *Correspondence: Yong-fa Zheng, ; Xi-ming Xu,
| |
Collapse
|
91
|
Tsamesidis I, Egwu CO, Samara D, Vogiatzi D, Lettas A, Lymperaki E. Effects of Greek Honey and Propolis on Oxidative Stress and Biochemical Parameters in Regular Blood Donors. J Xenobiot 2022; 12:13-20. [PMID: 35076563 PMCID: PMC8788442 DOI: 10.3390/jox12010002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/17/2021] [Accepted: 12/28/2021] [Indexed: 01/03/2023] Open
Abstract
Background and objectives: Honey products contain a lot of compounds, such as vitamins, enzymes, and minerals, which make honey and its products a great antioxidant with a critical role in health status. It is well accepted that honey and propolis can improve a lot of health problems when they are consumed in certain quantities. The objective of this study is to help regular blood donors improve their health status after donation. Material and methods: Eighty regular blood donor volunteers—30 males aged 19–61 and 30 females aged 21–64—were divided into 4 groups: group A (n = 20) consumed 2 spoons of Greek honey and 1 drop of propolis per day for 1 month, group B (n = 20) consumed 2 spoons of honey per day for 1 month, group C (n = 20) consumed 1 drop of propolis per day, and group D (n = 20) did not consume any Greek honey products. Blood samples were collected from all participants just before the consumption of the products, one month after the consumption, and six months after honey product consumption had ceased. All samples were analyzed for reactive oxygen species (ROS), lipid profiles, and ferritin levels. Results: The ROS were significantly (p < 0.05) lower in groups A, B, and C after the honey product consumption and increased significantly again after six months. No significant differences in lipid profiles were observed. Only triglyceride levels were increased after six months in all groups. On the other hand, ferritin levels were not statistically significantly decreased after six months in groups A and B, while they were increased in group C. Conclusions: In the present study, statistically significant decreases in ROS status was found after a small dose of honey product consumption, indicating a diet with an extra small dose of honey products after blood donation.
Collapse
Affiliation(s)
- Ioannis Tsamesidis
- Department of Biomedical Sciences, International Hellenic University, 570 01 Thessaloniki, Greece;
- School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
- Correspondence: ; Tel.: +30-6996311260
| | - Chinedu O. Egwu
- Laboratory Medical Biochemistry, College of Medicine, Alex-Ekwueme Federal University Ndufu-Alike, Abakaliki 1010, Nigeria;
| | - Diana Samara
- Blood Bank Section, Naoussa General Hospital, 592 00 Naousa, Greece; (D.S.); (A.L.)
| | - Dimitra Vogiatzi
- Department of Food Science and Nutrition, University of Aegean, 811 00 Mitilini, Greece;
| | - Athanasios Lettas
- Blood Bank Section, Naoussa General Hospital, 592 00 Naousa, Greece; (D.S.); (A.L.)
| | - Evgenia Lymperaki
- Department of Biomedical Sciences, International Hellenic University, 570 01 Thessaloniki, Greece;
| |
Collapse
|
92
|
Mas-Bargues C, Alique M, Barrús-Ortiz MT, Borrás C, Rodrigues-Díez R. Exploring New Kingdoms: The Role of Extracellular Vesicles in Oxi-Inflamm-Aging Related to Cardiorenal Syndrome. Antioxidants (Basel) 2021; 11:78. [PMID: 35052582 PMCID: PMC8773353 DOI: 10.3390/antiox11010078] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/20/2021] [Accepted: 12/23/2021] [Indexed: 12/12/2022] Open
Abstract
The incidence of age associated chronic diseases has increased in recent years. Although several diverse causes produce these phenomena, abundant evidence shows that oxidative stress plays a central role. In recent years, numerous studies have focused on elucidating the role of oxidative stress in the development and progression of both aging and chronic diseases, opening the door to the discovery of new underlying mechanisms and signaling pathways. Among them, senolytics and senomorphics, and extracellular vesicles offer new therapeutic strategies to slow the development of aging and its associated chronic diseases by decreasing oxidative stress. In this review, we aim to discuss the role of extracellular vesicles in human cardiorenal syndrome development and their possible role as biomarkers, targets, or vehicles of drugs to treat this syndrome.
Collapse
Affiliation(s)
- Cristina Mas-Bargues
- Grupo de Investigación Freshage, Departmento de Fisiología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (C.M.-B.); (C.B.)
- Instituto Sanitario de Investigación INCLIVA, 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable, Instituto de Salud Carlos III (CIBERFES, ISCIII), 28029 Madrid, Spain
| | - Matilde Alique
- Departamento de Biología de Sistemas, Universidad de Alcalá, 28871 Madrid, Spain;
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain
| | - María Teresa Barrús-Ortiz
- Área de Fisiología, Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Univesidad Rey Juan Carlos, Avenida de Atenas s/n, 28922 Madrid, Spain
| | - Consuelo Borrás
- Grupo de Investigación Freshage, Departmento de Fisiología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (C.M.-B.); (C.B.)
- Instituto Sanitario de Investigación INCLIVA, 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable, Instituto de Salud Carlos III (CIBERFES, ISCIII), 28029 Madrid, Spain
| | - Raquel Rodrigues-Díez
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain;
- Instituto de Investigación Hospital La Paz (IdiPAZ), 28046 Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 08036 Barcelona, Spain
| |
Collapse
|
93
|
Li SR, Man QW, Gao X, Lin H, Wang J, Su FC, Wang HQ, Bu LL, Liu B, Chen G. Tissue-derived extracellular vesicles in cancers and non-cancer diseases: Present and future. J Extracell Vesicles 2021; 10:e12175. [PMID: 34918479 PMCID: PMC8678102 DOI: 10.1002/jev2.12175] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 11/02/2021] [Accepted: 11/24/2021] [Indexed: 12/24/2022] Open
Abstract
Extracellular vesicles (EVs) are lipid‐bilayer membrane structures secreted by most cell types. EVs act as messengers via the horizontal transfer of lipids, proteins, and nucleic acids, and influence various pathophysiological processes in both parent and recipient cells. Compared to EVs obtained from body fluids or cell culture supernatants, EVs isolated directly from tissues possess a number of advantages, including tissue specificity, accurate reflection of tissue microenvironment, etc., thus, attention should be paid to tissue‐derived EVs (Ti‐EVs). Ti‐EVs are present in the interstitium of tissues and play pivotal roles in intercellular communication. Moreover, Ti‐EVs provide an excellent snapshot of interactions among various cell types with a common histological background. Thus, Ti‐EVs may be used to gain insights into the development and progression of diseases. To date, extensive investigations have focused on the role of body fluid‐derived EVs or cell culture‐derived EVs; however, the number of studies on Ti‐EVs remains insufficient. Herein, we summarize the latest advances in Ti‐EVs for cancers and non‐cancer diseases. We propose the future application of Ti‐EVs in basic research and clinical practice. Workflows for Ti‐EV isolation and characterization between cancers and non‐cancer diseases are reviewed and compared. Moreover, we discuss current issues associated with Ti‐EVs and provide potential directions.
Collapse
Affiliation(s)
- Su-Ran Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Qi-Wen Man
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Oral Maxillofacial Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Xin Gao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Hao Lin
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jing Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Fu-Chuan Su
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Han-Qi Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Lin-Lin Bu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Oral Maxillofacial Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Bing Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Oral Maxillofacial Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Gang Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Oral Maxillofacial Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China.,Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| |
Collapse
|
94
|
Folic Acid Protects Melanocytes from Oxidative Stress via Activation of Nrf2 and Inhibition of HMGB1. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:1608586. [PMID: 34917229 PMCID: PMC8670940 DOI: 10.1155/2021/1608586] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/13/2021] [Accepted: 11/13/2021] [Indexed: 12/21/2022]
Abstract
Vitiligo is a cutaneous depigmentation disease due to loss of epidermal melanocytes. Accumulating evidence has indicated that oxidative stress plays a vital role in vitiligo via directly destructing melanocytes and triggering inflammatory response that ultimately undermines melanocytes. Folic acid (FA), an oxidized form of folate with high bioavailability, exhibits potent antioxidant properties and shows therapeutic potential in multiple oxidative stress-related diseases. However, whether FA safeguards melanocytes from oxidative damages remains unknown. In this study, we first found that FA relieved melanocytes from H2O2-induced abnormal growth and apoptosis. Furthermore, FA enhanced the activity of antioxidative enzymes and remarkably reduced intracellular ROS levels in melanocytes. Subsequently, FA effectively activated nuclear factor E2-related factor 2 (Nrf2) pathway, and Nrf2 knockdown blocked the protective effects of FA on H2O2-treated melanocytes. Additionally, FA inhibited the production of proinflammatory HMGB1 in melanocytes under oxidative stress. Taken together, our findings support the protective effects of FA on human melanocytes against oxidative injury via the activation of Nrf2 and the inhibition of HMGB1, thus indicating FA as a potential therapeutic agent for the treatment of vitiligo.
Collapse
|
95
|
Franck T, Ceusters J, Graide H, Mouithys-Mickalad A, Serteyn D. Muscle Derived Mesenchymal Stem Cells Inhibit the Activity of the Free and the Neutrophil Extracellular Trap (NET)-Bond Myeloperoxidase. Cells 2021; 10:3486. [PMID: 34943996 PMCID: PMC8700239 DOI: 10.3390/cells10123486] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/21/2021] [Accepted: 12/07/2021] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are known to migrate to tissue injury sites to participate in immune modulation, tissue remodelling and wound healing, reducing tissue damage. Upon neutrophil activation, there is a release of myeloperoxidase (MPO), an oxidant enzyme. But little is known about the direct role of MSCs on MPO activity. The aim of this study was to investigate the effect of equine mesenchymal stem cells derived from muscle microinvasive biopsy (mdMSC) on the oxidant response of neutrophils and particularly on the activity of the myeloperoxidase released by stimulated equine neutrophils. After specific treatment (trypsin and washings in phosphate buffer saline), the mdMSCs were exposed to isolated neutrophils. The effect of the suspended mdMSCs was studied on the ROS production and the release of total and active MPO by stimulated neutrophils and specifically on the activity of MPO in a neutrophil-free model. Additionally, we developed a model combining adherent mdMSCs with neutrophils to study total and active MPO from the neutrophil extracellular trap (NET). Our results show that mdMSCs inhibited the ROS production, the activity of MPO released by stimulated neutrophils and the activity of MPO bound to the NET. Moreover, the co-incubation of mdMSCs directly with MPO results in a strong inhibition of the peroxidase activity of MPO, probably by affecting the active site of the enzyme. We confirm the strong potential of mdMSCs to lower the oxidant response of neutrophils. The novelty of our study is an evident inhibition of the activity of MPO by MSCs. The results indicated a new potential therapeutic approach of mdMSCs in the inhibition of MPO, which is considered as a pro-oxidant actor in numerous chronic and acute inflammatory pathologies.
Collapse
Affiliation(s)
- Thierry Franck
- Centre of Oxygen Research and Development (CORD), University of Liege, 4000 Liege, Belgium; (J.C.); (H.G.); (A.M.-M.); (D.S.)
- Research Unit FARAH, Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Liege, 4000 Liege, Belgium
| | - Justine Ceusters
- Centre of Oxygen Research and Development (CORD), University of Liege, 4000 Liege, Belgium; (J.C.); (H.G.); (A.M.-M.); (D.S.)
- Research Unit FARAH, Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Liege, 4000 Liege, Belgium
| | - Hélène Graide
- Centre of Oxygen Research and Development (CORD), University of Liege, 4000 Liege, Belgium; (J.C.); (H.G.); (A.M.-M.); (D.S.)
| | - Ange Mouithys-Mickalad
- Centre of Oxygen Research and Development (CORD), University of Liege, 4000 Liege, Belgium; (J.C.); (H.G.); (A.M.-M.); (D.S.)
| | - Didier Serteyn
- Centre of Oxygen Research and Development (CORD), University of Liege, 4000 Liege, Belgium; (J.C.); (H.G.); (A.M.-M.); (D.S.)
- Research Unit FARAH, Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Liege, 4000 Liege, Belgium
| |
Collapse
|
96
|
Xu Z, Yang Y, Sarath Babu V, Chen J, Li F, Yang M, Li N, Li J, Lin L, Qin Z. The antibacterial activity of erythrocytes from Clarias fuscus associated with phagocytosis and respiratory burst generation. FISH & SHELLFISH IMMUNOLOGY 2021; 119:96-104. [PMID: 34614395 DOI: 10.1016/j.fsi.2021.10.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 06/13/2023]
Abstract
It is widely known that red blood cells (RBCs) are responsible for respiration and the transport of gas. However, recent reports have also described the immune properties of RBCs, therefore creating new understanding for the functionality of RBCs. However, little is known about the immunological role of RBCs in bony fish. In this study, we used RBCs from Clarias fuscus as a model and demonstrate that these cells exhibited phagocytic ability with both latex beads and bacteria. Scanning electron microscopy and transmission electron microscopy provided visual confirmation of the phagocytotic process in RBCs. In addition, we used flow cytometry and fluorescence microscopy to analyse the rate of phagocytosis in RBCs. We found that RBCs exhibited stable phagocytotic ability with latex beads ranging from 0.5 to 1.0 μm in size. In response to bacterial stimulation, RBCs produced reactive oxygen species (ROS) and nitric oxide synthase (NOS), which are harmful to bacteria. RBCs also have an antioxidant system. Under conditions of oxidative stress, the expression levels of antioxidant enzymes, and particularly those of superoxide dismutase(SOD) increased significantly. Our results show that the erythrocytes of bony fish are phagocytic and also produce ROS which are toxic to bacteria. In addition, RBCs have an antioxidant system that removes excess ROS production to protect cells from oxidative damage.
Collapse
Affiliation(s)
- Zizheng Xu
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Youcheng Yang
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - V Sarath Babu
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Jiajun Chen
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Feng Li
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Minxuan Yang
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Ningqiu Li
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China; Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, China
| | - Jun Li
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China; School of Science and Medicine, Lake Superior State University, Sault Ste. Marie, MI, 49783, USA
| | - Li Lin
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China.
| | - Zhendong Qin
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China.
| |
Collapse
|
97
|
Beck-Joseph J, Tabrizian M, Lehoux S. Molecular Interactions Between Vascular Smooth Muscle Cells and Macrophages in Atherosclerosis. Front Cardiovasc Med 2021; 8:737934. [PMID: 34722670 PMCID: PMC8554018 DOI: 10.3389/fcvm.2021.737934] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 09/16/2021] [Indexed: 01/10/2023] Open
Abstract
Atherosclerosis is the largest contributor toward life-threatening cardiovascular events. Cellular activity and cholesterol accumulation lead to vascular remodeling and the formation of fatty plaques. Complications arise from blood clots, forming at sites of plaque development, which may detach and result in thrombotic occlusions. Vascular smooth muscle cells and macrophages play dominant roles in atherosclerosis. A firm understanding of how these cells influence and modulate each other is pivotal for a better understanding of the disease and the development of novel therapeutics. Recent studies have investigated molecular interactions between both cell types and their impact on disease progression. Here we aim to review the current knowledge. Intercellular communications through soluble factors, physical contact, and extracellular vesicles are discussed. We also present relevant background on scientific methods used to study the disease, the general pathophysiology and intracellular factors involved in phenotypic modulation of vascular smooth muscle cells. We conclude this review with a discussion of the current state, shortcomings and potential future directions of the field.
Collapse
Affiliation(s)
- Jahnic Beck-Joseph
- Biomat'X Research Laboratories, Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Maryam Tabrizian
- Biomat'X Research Laboratories, Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Stephanie Lehoux
- Department of Medicine, Lady Davis Institute, McGill University, Montreal, QC, Canada
| |
Collapse
|
98
|
Yuan K, Ye X, Liu W, Liu K, Wu D, Zhao W, Qian Z, Li S, Huang C, Yu Z, Chen Z. Preparation, characterization and antibacterial activity of a novel Zn(II) coordination polymer derived from carboxylic acid. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.130624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
99
|
Mei L, Zheng Y, Ma T, Xia B, Gao X, Hao Y, Luo Z, Huang J. The Novel Antioxidant Compound JSH-23 Prevents Osteolysis by Scavenging ROS During Both Osteoclastogenesis and Osteoblastogenesis. Front Pharmacol 2021; 12:734774. [PMID: 34566656 PMCID: PMC8458573 DOI: 10.3389/fphar.2021.734774] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/27/2021] [Indexed: 12/13/2022] Open
Abstract
Inflammatory osteolysis is a pathological skeletal disease associated with not only the production of inflammatory cytokines but also local oxidative status. Excessive reactive oxygen species (ROS) promote bone resorption by osteoclasts and induce the apoptosis of osteoblasts. In consideration of the lack of effective preventive or treatments options against osteolysis, the exploitation of novel pharmacological compounds/agents is critically required. In our study, we found that a novel antioxidant compound, JSH-23, plays a role in restoring bone homeostasis by scavenging intracellular ROS during both osteoclastogenesis and osteoblastogenesis. Mechanically, JSH-23 suppressed RANKL-induced osteoclastogenesis, bone resorption and the expression of specific genes (including NFATc1, c-Fos, TRAP, CTSK and DC-STAMP) via inhibition of the NF-κB signaling pathway. Meanwhile, JSH-23 suppressed RANKL-induced ROS generation via the TRAF6/Rac1/NOX1 pathway and the enhanced expression of Nrf2/HO-1. In addition, JSH-23 attenuated H2O2-induced apoptosis and mineralization reduction in osteoblasts by reducing ROS production and enhancing Nrf2/HO-1 expression. Our in vivo results further revealed that JSH-23 exerts its protective effects on bone mass through its antioxidant activity. In conclusion, our results show that the application of JSH-23 might be a novel and plausible strategy for the treatment of osteolysis-related disease.
Collapse
Affiliation(s)
- Liangwei Mei
- Department of Orthopaedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Yi Zheng
- Department of Orthopaedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Teng Ma
- Department of Orthopaedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Bing Xia
- Department of Orthopaedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Xue Gao
- Faculty of Life Sciences, Northwest University, Xi'an, China
| | - Yiming Hao
- Department of Orthopaedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Zhuojing Luo
- Department of Orthopaedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Jinghui Huang
- Department of Orthopaedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| |
Collapse
|
100
|
Qi H, Wang Y, Fa S, Yuan C, Yang L. Extracellular Vesicles as Natural Delivery Carriers Regulate Oxidative Stress Under Pathological Conditions. Front Bioeng Biotechnol 2021; 9:752019. [PMID: 34557480 PMCID: PMC8452933 DOI: 10.3389/fbioe.2021.752019] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 08/26/2021] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles are cellular secretory particles that can be used as natural drug delivery carriers. They have successfully delivered drugs including chemotherapeutics, proteins, and genes to treat various diseases. Oxidative stress is an abnormal physiological phenomenon, and it is associated with nearly all diseases. In this short review, we summarize the regulation of EVs on oxidative stress. There are direct effects and indirect effects on the regulation of oxidative stress through EVs. On the one hand, they can deliver antioxidant substances or oxides to recipient cells, directly relieving or aggravating oxidative stress. On the other hand, regulate factors of oxidative stress-related signaling pathways can be delivered to recipient cells by the mediation of EVs, realizing the indirect regulation of oxidative stress. To the best of our knowledge, however, only endogenous drugs have been delivered by EVs to regulate oxidative stress till now. And the heterogeneity of EVs may complicate the regulation of oxidative stress. Therefore, this short review aims to draw more attention to the EVs-based regulation of oxidative stress, and we hope excellent EVs-based delivery carriers that can deliver exogenous drugs to regulate oxidative stress can be exploited.
Collapse
Affiliation(s)
- Hongzhao Qi
- Department of Aging Research, Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Yingruo Wang
- Shandong University of Science and Technology, Qingdao, China
| | - Shunxin Fa
- School of Stomatology, Qingdao University, Qingdao, China.,York School, Monterey, CA, United States
| | - Changqing Yuan
- School of Stomatology, Qingdao University, Qingdao, China
| | - Lijun Yang
- Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
| |
Collapse
|