1
|
Shi J, Ji Z, Yao X, Yao Y, Li C, Liang Q, Zhang X. HSP90 Enhances Mitophagy to Improve the Resistance of Car-Diomyocytes to Heat Stress in Wenchang Chickens. Int J Mol Sci 2024; 25:11695. [PMID: 39519247 PMCID: PMC11546521 DOI: 10.3390/ijms252111695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/22/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Heat shock protein 90 (HSP90) is recognized for its protective effects against heat stress damage; however, the specific functions and underlying molecular mechanisms of HSP90 in heat-stressed cardiomyocytes remain largely unexplored, particularly in tropical species. In our study, Wenchang chickens (WCCs) were classified into two groups: the heat stress survival (HSS) group and the heat stress death (HSD) group, based on their survival following exposure to heat stress. Heat stress resulted in significant cardiomyocyte damage, mitochondrial dysfunction, and apoptosis in the HSD group, while the damage was less pronounced in the HSS group. We further validated these findings in primary cardiomyocytes derived from Wenchang chickens (PCWs). Additionally, heat stress was found to upregulate Pink1/Parkin-mediated mitophagy, which was accompanied by an increase in HSP90 expression in both cardiomyocytes and PCWs. Our results demonstrated that HSP90 overexpression enhances PINK1/Parkin-mediated mitophagy, ultimately inhibiting apoptosis and oxidative stress in heat-stressed PCWs. However, the application of Geldanamycin (GA) reversed these effects. Notably, we discovered that HSP90 interacts with Beclin-1 through mitochondrial translocation and directly regulates mitophagy levels in PCWs. In summary, we have elucidated a novel role for HSP90 and mitophagy in regulating heat stress-induced acute cardiomyocyte injury.
Collapse
Affiliation(s)
- Jiachen Shi
- Key Laboratory of Tropical Animal Breeding and Epidemic Disease Research of Hainan Province, School of Tropical Agriculture and Forestry, Hainan University, Haikou 570100, China; (J.S.); (Z.J.); (X.Y.); (Y.Y.); (C.L.); (Q.L.)
- School of Life and Health Sciences, Hainan University, Haikou 570228, China
| | - Zeping Ji
- Key Laboratory of Tropical Animal Breeding and Epidemic Disease Research of Hainan Province, School of Tropical Agriculture and Forestry, Hainan University, Haikou 570100, China; (J.S.); (Z.J.); (X.Y.); (Y.Y.); (C.L.); (Q.L.)
| | - Xu Yao
- Key Laboratory of Tropical Animal Breeding and Epidemic Disease Research of Hainan Province, School of Tropical Agriculture and Forestry, Hainan University, Haikou 570100, China; (J.S.); (Z.J.); (X.Y.); (Y.Y.); (C.L.); (Q.L.)
| | - Yujie Yao
- Key Laboratory of Tropical Animal Breeding and Epidemic Disease Research of Hainan Province, School of Tropical Agriculture and Forestry, Hainan University, Haikou 570100, China; (J.S.); (Z.J.); (X.Y.); (Y.Y.); (C.L.); (Q.L.)
| | - Chengyun Li
- Key Laboratory of Tropical Animal Breeding and Epidemic Disease Research of Hainan Province, School of Tropical Agriculture and Forestry, Hainan University, Haikou 570100, China; (J.S.); (Z.J.); (X.Y.); (Y.Y.); (C.L.); (Q.L.)
| | - Qijun Liang
- Key Laboratory of Tropical Animal Breeding and Epidemic Disease Research of Hainan Province, School of Tropical Agriculture and Forestry, Hainan University, Haikou 570100, China; (J.S.); (Z.J.); (X.Y.); (Y.Y.); (C.L.); (Q.L.)
| | - Xiaohui Zhang
- Key Laboratory of Tropical Animal Breeding and Epidemic Disease Research of Hainan Province, School of Tropical Agriculture and Forestry, Hainan University, Haikou 570100, China; (J.S.); (Z.J.); (X.Y.); (Y.Y.); (C.L.); (Q.L.)
| |
Collapse
|
2
|
Shukla M, Narayan M. Proteostasis and Its Role in Disease Development. Cell Biochem Biophys 2024:10.1007/s12013-024-01581-6. [PMID: 39422790 DOI: 10.1007/s12013-024-01581-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2024] [Indexed: 10/19/2024]
Abstract
Proteostasis (protein homeostasis) refers to the general biological process that maintains the proper balance between the synthesis of proteins, their folding, trafficking, and degradation. It ensures proteins are functional, locally distributed, and appropriately folded inside cells. Genetic information enclosed in mRNA is translated into proteins. To ensure newly synthesized proteins take on the exact three-dimensional conformation, molecular chaperones assist in proper folding. Misfolded proteins can be refolded or targeted for elimination to stop aggregation. Cells utilize different degradation pathways, for instance, the ubiquitin-proteasome system, the autophagy-lysosome pathway, and the unfolded protein response, to degrade unwanted or damaged proteins. Quality control systems of the cell monitor the folding of proteins. These checkpoint mechanisms are aimed at degrading or refolding misfolded or damaged proteins. Under stress response pathways, such as heat shock response and unfolded protein response, which are triggered under conditions that perturb proteostasis, the capacity for folding is increased, and degradation pathways are activated to help cells handle stressful conditions. The deregulation of proteostasis is implicated in a variety of illnesses, comprising cancer, metabolic diseases, cardiovascular diseases, and neurological disorders. Therapeutic strategies with a deeper insight into the mechanism of proteostasis are crucial for the treatment of illnesses linked with proteostasis and to support cellular health. Thus, proteostasis is required not only for the maintenance of cellular homeostasis and function but also for proper protein function and prevention of injurious protein aggregation. In this review, we have covered the concept of proteostasis, its mechanism, and how disruptions to it can result in a number of disorders.
Collapse
Affiliation(s)
- Manisha Shukla
- Department of Biotechnology, Pandit S.N. Shukla University, Shahdol, Madhya Pradesh, India
| | - Mahesh Narayan
- Department of Chemistry and Biochemistry, University of Texas, El Paso, TX, USA.
| |
Collapse
|
3
|
Liu Z, Bian X, Li L, Liu L, Feng C, Wang Y, Ni J, Li S, Lu D, Li Y, Ma C, Yu T, Xiao X, Xue N, Wang Y, Zhang C, Ma X, Gao X, Fan X, Liu X, Fan G. SENP1-Mediated HSP90ab1 DeSUMOylation in Cardiomyocytes Prevents Myocardial Fibrosis by Paracrine Signaling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400741. [PMID: 38992961 PMCID: PMC11425837 DOI: 10.1002/advs.202400741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 05/06/2024] [Indexed: 07/13/2024]
Abstract
Myocardial infarction (MI) triggers a poor ventricular remodeling response, but the underlying mechanisms remain unclear. Here, the authors show that sentrin-specific protease 1 (SENP1) is downregulated in post-MI mice and in patients with severe heart failure. By generating cardiomyocyte-specific SENP1 knockout and overexpression mice to assess cardiac function and ventricular remodeling responses under physiological and pathological conditions. Increased cardiac fibrosis in the cardiomyocyte-specific SENP1 deletion mice, associated with increased fibronectin (Fn) expression and secretion in cardiomyocytes, promotes fibroblast activation in response to myocardial injury. Mechanistically, SENP1 deletion in mouse cardiomyocytes increases heat shock protein 90 alpha family class B member 1 (HSP90ab1) SUMOylation with (STAT3) activation and Fn secretion after ventricular remodeling initiated. Overexpression of SENP1 or mutation of the HSP90ab1 Lys72 ameliorates adverse ventricular remodeling and dysfunction after MI. Taken together, this study identifies SENP1 as a positive regulator of cardiac repair and a potential drug target for the treatment of MI. Inhibition of HSP90ab1 SUMOylation stabilizes STAT3 to inhibit the adverse ventricular remodeling response.
Collapse
Affiliation(s)
- Zhihao Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China
| | - Xiyun Bian
- Tianjin Key Laboratory of Epigenetics for Organ Development of Preterm Infants, Tianjin fifth Central Hospital, Tianjin, 300450, China
- Central Laboratory, Tianjin Fifth Central Hospital, Tianjin, 300450, China
| | - Lan Li
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China
| | - Li Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China
| | - Chao Feng
- Department of Cardiology, Tianjin Chest Hospital, Tianjin, 300051, China
| | - Ying Wang
- Tianjin Key Laboratory of Epigenetics for Organ Development of Preterm Infants, Tianjin fifth Central Hospital, Tianjin, 300450, China
- Central Laboratory, Tianjin Fifth Central Hospital, Tianjin, 300450, China
| | - Jingyu Ni
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
| | - Sheng Li
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China
| | - Dading Lu
- Tianjin Key Laboratory of Epigenetics for Organ Development of Preterm Infants, Tianjin fifth Central Hospital, Tianjin, 300450, China
- Central Laboratory, Tianjin Fifth Central Hospital, Tianjin, 300450, China
| | - Yanxia Li
- Tianjin Key Laboratory of Epigenetics for Organ Development of Preterm Infants, Tianjin fifth Central Hospital, Tianjin, 300450, China
- Central Laboratory, Tianjin Fifth Central Hospital, Tianjin, 300450, China
| | - Chuanrui Ma
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
| | - Tian Yu
- Tianjin Key Laboratory of Epigenetics for Organ Development of Preterm Infants, Tianjin fifth Central Hospital, Tianjin, 300450, China
- Central Laboratory, Tianjin Fifth Central Hospital, Tianjin, 300450, China
| | - Xiaolin Xiao
- Tianjin Key Laboratory of Epigenetics for Organ Development of Preterm Infants, Tianjin fifth Central Hospital, Tianjin, 300450, China
- Central Laboratory, Tianjin Fifth Central Hospital, Tianjin, 300450, China
| | - Na Xue
- Tianjin Key Laboratory of Epigenetics for Organ Development of Preterm Infants, Tianjin fifth Central Hospital, Tianjin, 300450, China
- Central Laboratory, Tianjin Fifth Central Hospital, Tianjin, 300450, China
| | - Yuxiang Wang
- Tianjin Key Laboratory of Epigenetics for Organ Development of Preterm Infants, Tianjin fifth Central Hospital, Tianjin, 300450, China
- Central Laboratory, Tianjin Fifth Central Hospital, Tianjin, 300450, China
| | - Chunyan Zhang
- Tianjin Key Laboratory of Epigenetics for Organ Development of Preterm Infants, Tianjin fifth Central Hospital, Tianjin, 300450, China
- Central Laboratory, Tianjin Fifth Central Hospital, Tianjin, 300450, China
| | - Xiaofang Ma
- Tianjin Key Laboratory of Epigenetics for Organ Development of Preterm Infants, Tianjin fifth Central Hospital, Tianjin, 300450, China
- Central Laboratory, Tianjin Fifth Central Hospital, Tianjin, 300450, China
| | - Xiumei Gao
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
| | - Xiaohui Fan
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314100, China
| | - Xiaozhi Liu
- Tianjin Key Laboratory of Epigenetics for Organ Development of Preterm Infants, Tianjin fifth Central Hospital, Tianjin, 300450, China
- Central Laboratory, Tianjin Fifth Central Hospital, Tianjin, 300450, China
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
| |
Collapse
|
4
|
Wang Y, Yang H, Zhao L, Yin X, Ai S, Hu M, Pan X, Zheng Y, Shi S, Li G, Pan Y, Yang T, Zhang J. Human Plasma‐Derived Extracellular Vesicles Protect Against Cerebral Ischemia‐Reperfusion Injury via HSP27 Phosphorylation. ADVANCED THERAPEUTICS 2024; 7. [DOI: 10.1002/adtp.202400006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Indexed: 01/03/2025]
Abstract
AbstractIschemic stroke (IS) has become a serious public health problem, with patients undergoing endovascular treatment experiencing ischemia‐reperfusion (I/R) injury, which exacerbates cerebrovascular diseases. Circulating extracellular vesicles (EVs) have shown potential for treating cerebral I/R injury. In this study, the therapeutic effect of human plasma‐derived EVs and protective mechanisms in cerebral I/R injury is explored. An oxygen‐glucose deprivation/reperfusion (OGD/R) model is used to treat SH‐SY5Y cells in vitro, and an I/R injury model is constructed by transient middle cerebral artery occlusion (tMCAO) in mice. Human plasma‐derived EVs are extracted by size exclusion chromatography. Western blot, Immunofluorescence Staining, and 2,3,5‐Triphenyltetrazolium Chloride (TTC) Staining are employed to observe the effects of EVs on the neuroinflammatory response and infarct volumes in tMCAO mice, while TUNEL Staining, Flow Cytometry, and Western Blot are employed to assess cell apoptosis. Human plasma‐derived EVs alleviated apoptosis in SH‐SY5Y cells under OGD/R stress and exerted a protective effect against brain I/R injury in tMCAO mice. Mechanistically, EVs protected against cerebral I/R injury via HSP27 phosphorylation, and the HSP27 phosphorylation inhibitor KRIBB3 attenuated the anti‐apoptotic effects of EVs. Human plasma‐derived EVs activated the phosphorylation of HSP27, thereby inhibiting cell apoptosis and protecting against cerebral I/R injury.
Collapse
Affiliation(s)
- Yunan Wang
- Department of Neurology Fourth Affiliated Hospital of Harbin Medical University Harbin Heilongjiang 150001 China
| | - Hongxia Yang
- Department of Neurology Fourth Affiliated Hospital of Harbin Medical University Harbin Heilongjiang 150001 China
| | - Linlin Zhao
- Department of Neurology Fourth Affiliated Hospital of Harbin Medical University Harbin Heilongjiang 150001 China
| | - Xiaohang Yin
- Institute of Cardiovascular Sciences Shanghai Engineering Research Center of Organ Repair School of Life Science Shanghai University Shanghai 200444 China
| | - Songwei Ai
- Institute of Cardiovascular Sciences Shanghai Engineering Research Center of Organ Repair School of Life Science Shanghai University Shanghai 200444 China
| | - Meiyu Hu
- Institute of Cardiovascular Sciences Shanghai Engineering Research Center of Organ Repair School of Life Science Shanghai University Shanghai 200444 China
| | - Xue Pan
- Institute of Cardiovascular Sciences Shanghai Engineering Research Center of Organ Repair School of Life Science Shanghai University Shanghai 200444 China
| | - Yonghui Zheng
- Department of Neurology Fourth Affiliated Hospital of Harbin Medical University Harbin Heilongjiang 150001 China
| | - Shuxian Shi
- Department of Neurology Fourth Affiliated Hospital of Harbin Medical University Harbin Heilongjiang 150001 China
| | - Genjie Li
- Department of Neurology Fourth Affiliated Hospital of Harbin Medical University Harbin Heilongjiang 150001 China
| | - Yonghui Pan
- Fourth Ward of Neurology Department First Affiliated Hospital of Harbin Medical University Harbin Heilongjiang 150001 China
| | - Tingting Yang
- Institute of Cardiovascular Sciences Shanghai Engineering Research Center of Organ Repair School of Life Science Shanghai University Shanghai 200444 China
| | - Jingyu Zhang
- Department of Neurology Fourth Affiliated Hospital of Harbin Medical University Harbin Heilongjiang 150001 China
| |
Collapse
|
5
|
Li Y, Jiang Z, Xu Y, Yan J, Wu Q, Huang S, Wang L, Xie Y, Wu X, Wang Y, Li Y, Fan X, Li F, Yuan W. Pygo-F773W Mutation Reveals Novel Functions beyond Wnt Signaling in Drosophila. Int J Mol Sci 2024; 25:5998. [PMID: 38892188 PMCID: PMC11172468 DOI: 10.3390/ijms25115998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/21/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
Pygopus (Pygo) has been identified as a specific nuclear co-activator of the canonical Wingless (Wg)/Wnt signaling pathway in Drosophila melanogaster. Pygo proteins consist of two conserved domains: an N-terminal homologous domain (NHD) and a C-terminal plant homologous domain (PHD). The PHD's ability to bind to di- and trimethylated lysine 4 of histone H3 (H3K4me2/3) appears to be independent of Wnt signaling. There is ongoing debate regarding the significance of Pygo's histone-binding capacity. Drosophila Pygo orthologs have a tryptophan (W) > phenylalanine (F) substitution in their histone pocket-divider compared to vertebrates, leading to reduced histone affinity. In this research, we utilized CRISPR/Cas9 technology to introduce the Pygo-F773W point mutation in Drosophila, successfully establishing a viable homozygous Pygo mutant line for the first time. Adult mutant flies displayed noticeable abnormalities in reproduction, locomotion, heart function, and lifespan. RNA-seq and cluster analysis indicated that the mutation primarily affected pathways related to immunity, metabolism, and posttranslational modification in adult flies rather than the Wnt signaling pathway. Additionally, a reduction in H3K9 acetylation levels during the embryonic stage was observed in the mutant strains. These findings support the notion that Pygo plays a wider role in chromatin remodeling, with its involvement in Wnt signaling representing only a specific aspect of its chromatin-related functions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Fang Li
- The Laboratory of Heart Development Research, College of Life Science, Hunan Normal University, Changsha 410081, China; (Y.L.); (Z.J.); (X.F.)
| | - Wuzhou Yuan
- The Laboratory of Heart Development Research, College of Life Science, Hunan Normal University, Changsha 410081, China; (Y.L.); (Z.J.); (X.F.)
| |
Collapse
|
6
|
Zhang X, Zhen D, Yi F, Zhang T, Li X, Wang Y, Li X, Sheng Y, Liu X, Jin T, He Y. Identification of Six Pathogenic Genes for Tibetan Familial Ventricular Septal Defect by Whole Exome Sequencing. J Surg Res 2024; 296:18-28. [PMID: 38215673 DOI: 10.1016/j.jss.2023.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 12/01/2023] [Accepted: 12/14/2023] [Indexed: 01/14/2024]
Abstract
INTRODUCTION Ventricular septal defect (VSD) is the most common congenital heart malformation in children. This study aimed to investigate potential pathogenic genes associated with Tibetan familial VSD. METHODS Whole genomic DNA was extracted from eight Tibetan children with VSD and their healthy parents (a total of 16 individuals). Whole-exome sequencing was performed using the Illumina HiSeq platform. After filtration, detection, and annotation, single nucleotide variations and insertion-deletion markers were examined. Comparative evaluations using the Sorting Intolerant from Tolerant, PolyPhen V2, Mutation Taster, and Combined Annotation Dependent Depletion databases were conducted to predict harmful mutant genes associated with the etiology of Tibetan familial VSD. RESULTS A total of six missense mutations in genetic disease-causing genes associated with the development of Tibetan familial VSD were identified: activin A receptor type II-like 1 (c.652 C > T: p.R218 W), ATPase cation transporting 13A2 (c.1363 C > T: p.R455 W), endoplasmic reticulum aminopeptidase 1 (c.481 G > A: p.G161 R), MRI1 (c.629 G > A: p.R210Q), tumor necrosis factor receptor-associated protein 1 (c.224 G > A: p.R75H), and FBN2 (c.2260 G > A: p.G754S). The Human Gene Mutation Database confirmed activin A receptor type II-like 1, MRI1, and tumor necrosis factor receptor-associated protein 1 as pathogenic mutations, while FBN2 was classified as a probable pathogenic mutation. CONCLUSIONS This novel study directly screens genetic variations associated with Tibetan familial VSD using whole-exome sequencing, providing new insights into the pathogenesis of VSD.
Collapse
Affiliation(s)
- Xiaohui Zhang
- Key Laboratory of High Altitude Hypoxia Environment and Life Health, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, China; Department of Ultrasound, the Affiliated Hospital of Xizang Minzu University, Xianyang, Shaanxi, China
| | - Da Zhen
- Department of Medical, Tibet Autonomous Region Maternity and Children's Hospital, Lhasa, Tibet, China
| | - Faling Yi
- Key Laboratory of High Altitude Hypoxia Environment and Life Health, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, China; School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, China
| | - Tianyi Zhang
- Key Laboratory of High Altitude Hypoxia Environment and Life Health, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, China; School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, China
| | - Xuemei Li
- Key Laboratory of High Altitude Hypoxia Environment and Life Health, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, China; School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, China
| | - Yuhe Wang
- Key Laboratory of High Altitude Hypoxia Environment and Life Health, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, China; Department of Clinical Laboratory, the Affiliated Hospital of Xizang Minzu University, Xianyang, Shaanxi, China
| | - Xuguang Li
- Key Laboratory of High Altitude Hypoxia Environment and Life Health, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, China; School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, China
| | - Yemeng Sheng
- Key Laboratory of High Altitude Hypoxia Environment and Life Health, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, China; School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, China
| | - Xiaoli Liu
- Key Laboratory of High Altitude Hypoxia Environment and Life Health, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, China; School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, China
| | - Tianbo Jin
- School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, China.
| | - Yongjun He
- Key Laboratory of High Altitude Hypoxia Environment and Life Health, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, China; School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, China.
| |
Collapse
|
7
|
Tsare EPG, Klapa MI, Moschonas NK. Protein-protein interaction network-based integration of GWAS and functional data for blood pressure regulation analysis. Hum Genomics 2024; 18:15. [PMID: 38326862 PMCID: PMC11465932 DOI: 10.1186/s40246-023-00565-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 11/12/2023] [Indexed: 02/09/2024] Open
Abstract
BACKGROUND It is valuable to analyze the genome-wide association studies (GWAS) data for a complex disease phenotype in the context of the protein-protein interaction (PPI) network, as the related pathophysiology results from the function of interacting polyprotein pathways. The analysis may include the design and curation of a phenotype-specific GWAS meta-database incorporating genotypic and eQTL data linking to PPI and other biological datasets, and the development of systematic workflows for PPI network-based data integration toward protein and pathway prioritization. Here, we pursued this analysis for blood pressure (BP) regulation. METHODS The relational scheme of the implemented in Microsoft SQL Server BP-GWAS meta-database enabled the combined storage of: GWAS data and attributes mined from GWAS Catalog and the literature, Ensembl-defined SNP-transcript associations, and GTEx eQTL data. The BP-protein interactome was reconstructed from the PICKLE PPI meta-database, extending the GWAS-deduced network with the shortest paths connecting all GWAS-proteins into one component. The shortest-path intermediates were considered as BP-related. For protein prioritization, we combined a new integrated GWAS-based scoring scheme with two network-based criteria: one considering the protein role in the reconstructed by shortest-path (RbSP) interactome and one novel promoting the common neighbors of GWAS-prioritized proteins. Prioritized proteins were ranked by the number of satisfied criteria. RESULTS The meta-database includes 6687 variants linked with 1167 BP-associated protein-coding genes. The GWAS-deduced PPI network includes 1065 proteins, with 672 forming a connected component. The RbSP interactome contains 1443 additional, network-deduced proteins and indicated that essentially all BP-GWAS proteins are at most second neighbors. The prioritized BP-protein set was derived from the union of the most BP-significant by any of the GWAS-based or the network-based criteria. It included 335 proteins, with ~ 2/3 deduced from the BP PPI network extension and 126 prioritized by at least two criteria. ESR1 was the only protein satisfying all three criteria, followed in the top-10 by INSR, PTN11, CDK6, CSK, NOS3, SH2B3, ATP2B1, FES and FINC, satisfying two. Pathway analysis of the RbSP interactome revealed numerous bioprocesses, which are indeed functionally supported as BP-associated, extending our understanding about BP regulation. CONCLUSIONS The implemented workflow could be used for other multifactorial diseases.
Collapse
Affiliation(s)
- Evridiki-Pandora G Tsare
- Department of General Biology, School of Medicine, University of Patras, Patras, Greece
- Metabolic Engineering and Systems Biology Laboratory, Institute of Chemical Engineering Sciences, Foundation for Research and Technology-Hellas (FORTH/ICE-HT), Patras, Greece
| | - Maria I Klapa
- Metabolic Engineering and Systems Biology Laboratory, Institute of Chemical Engineering Sciences, Foundation for Research and Technology-Hellas (FORTH/ICE-HT), Patras, Greece.
| | - Nicholas K Moschonas
- Department of General Biology, School of Medicine, University of Patras, Patras, Greece.
- Metabolic Engineering and Systems Biology Laboratory, Institute of Chemical Engineering Sciences, Foundation for Research and Technology-Hellas (FORTH/ICE-HT), Patras, Greece.
| |
Collapse
|
8
|
Tang CM, Zhang Z, Sun Y, Ding WJ, Yang XC, Song YP, Ling MY, Li XH, Yan R, Zheng YJ, Yu N, Zhang WH, Wang Y, Wang SP, Gao HQ, Zhao CL, Xing YQ. Multi-omics reveals aging-related pathway in natural aging mouse liver. Heliyon 2023; 9:e21011. [PMID: 37920504 PMCID: PMC10618800 DOI: 10.1016/j.heliyon.2023.e21011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 10/01/2023] [Accepted: 10/12/2023] [Indexed: 11/04/2023] Open
Abstract
Aging is associated with gradual changes in liver structure, altered metabolites and other physiological/pathological functions in hepatic cells. However, its characterized phenotypes based on altered metabolites and the underlying biological mechanism are unclear. Advancements in high-throughput omics technology provide new opportunities to understand the pathological process of aging. Here, in our present study, both metabolomics and phosphoproteomics were applied to identify the altered metabolites and phosphorylated proteins in liver of young (the WTY group) and naturally aged (the WTA group) mice, to find novel biomarkers and pathways, and uncover the biological mechanism. Analysis showed that the body weights, alanine aminotransferase (ALT) and aspartate aminotransferase (AST) increased in the WTA group. The grips decreased with age, while the triglyceride (TG) and cholesterol (TC) did not change significantly. The increase of fibrosis, accumulation of inflammatory cells, hepatocytes degeneration, the deposition of lipid droplets and glycogen, the damaged mitochondria, and deduction of endoplasmic reticulum were observed in the aging liver under optical and electron microscopes. In addition, a network of metabolites and phosphorylated proteomes of the aging liver was established. Metabolomics detected 970 metabolites in the positive ion mode and 778 metabolites in the negative ion mode. A total of 150 pathways were pooled. Phosphoproteomics identified 2618 proteins which contained 16621 phosphosites. A total of 164 pathways were detected. 65 common pathways were detected in two omics. Phosphorylated protein heat shock protein HSP 90-alpha (HSP90A) and v-raf murine viral oncogene homolog B1(BRAF), related to cancer pathway, were significantly upregulated in aged mice liver. Western blot verified that protein expression of MEK and ERK, downstream of BRAF pathway were elevated in the liver of aging mice. However, the protein expression of BRAF was not a significant difference. Overall, these findings revealed a close link between aging and cancer and contributed to our understanding of the multi-omics changes in natural aging.
Collapse
Affiliation(s)
- Cong-min Tang
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Institute of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan 250012, Shandong Province, China
- Department of Ultrasound, Shandong Provincial Third Hospital, Jinan 250031, Shandong Province, China
| | - Zhen Zhang
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Yan Sun
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Wen-jing Ding
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Institute of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan 250012, Shandong Province, China
| | - Xue-chun Yang
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Institute of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan 250012, Shandong Province, China
| | - Yi-ping Song
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Ming-ying Ling
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Institute of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan 250012, Shandong Province, China
| | - Xue-hui Li
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Rong Yan
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Yu-jing Zheng
- Shandong Precision Medicine Engineering Laboratory of Bacterial Anti-tumor Drugs, Jinan 250101, Shandong Province, China
| | - Na Yu
- Shandong Precision Medicine Engineering Laboratory of Bacterial Anti-tumor Drugs, Jinan 250101, Shandong Province, China
| | - Wen-hua Zhang
- Shandong Precision Medicine Engineering Laboratory of Bacterial Anti-tumor Drugs, Jinan 250101, Shandong Province, China
| | - Yong Wang
- Shandong Precision Medicine Engineering Laboratory of Bacterial Anti-tumor Drugs, Jinan 250101, Shandong Province, China
| | - Shao-peng Wang
- Shandong Precision Medicine Engineering Laboratory of Bacterial Anti-tumor Drugs, Jinan 250101, Shandong Province, China
| | - Hai-qing Gao
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Chuan-li Zhao
- Dept of Hematology, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Yan-qiu Xing
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| |
Collapse
|
9
|
Jafari N, Gheitasi R, Khorasani HR, Golpour M, Mehri M, Nayeri K, Pourbagher R, Mostafazadeh M, Kalali B, Mostafazadeh A. Proteome analysis, bioinformatic prediction and experimental evidence revealed immune response down-regulation function for serum-starved human fibroblasts. Heliyon 2023; 9:e19238. [PMID: 37674821 PMCID: PMC10477462 DOI: 10.1016/j.heliyon.2023.e19238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 06/15/2023] [Accepted: 08/16/2023] [Indexed: 09/08/2023] Open
Abstract
Emerging evidence indicates that fibroblasts play pivotal roles in immunoregulation by producing various proteins under health and disease states. In the present study, for the first time, we compared the proteomes of serum-starved human skin fibroblasts and peripheral blood mononuclear cells (PBMCs) using Nano-LC-ESI-tandem mass spectrometry. This analysis contributes to a better understanding of the underlying molecular mechanisms of chronic inflammation and cancer, which are intrinsically accompanied by growth factor deficiency.The proteomes of starved fibroblasts and PBMCs consisted of 307 and 294 proteins, respectively, which are involved in lymphocyte migration, complement activation, inflammation, acute phase response, and immune regulation. Starved fibroblasts predominantly produced extracellular matrix-related proteins such as collagen/collagenase, while PBMCs produced focal adhesion-related proteins like beta-parvin and vinculin which are involved in lymphocyte migration. PBMCs produced a more diverse set of inflammatory molecules like heat shock proteins, while fibroblasts produced human leukocytes antigen-G and -E that are known as main immunomodulatory molecules. Fifty-four proteins were commonly found in both proteomes, including serum albumin, amyloid-beta, heat shock cognate 71 kDa, and complement C3. GeneMANIA bioinformatic tool predicted 418 functions for PBMCs, including reactive oxygen species metabolic processes and 241 functions for starved fibroblasts such as antigen processing and presentation including non-classical MHC -Ib pathway, and negative regulation of the immune response. Protein-protein interactions network analysis indicated the immunosuppressive function for starved fibroblasts-derived human leucocytes antigen-G and -E. Moreover, in an in vitro model of allogeneic transplantation, the immunosuppressive activity of starved fibroblasts was experimentally documented. Conclusion Under serum starvation-induced metabolic stress, both PBMCs and fibroblasts produced molecules like heat shock proteins and amyloid-beta, which can have pathogenic roles in auto-inflammatory diseases such as rheumatoid arthritis, type 1 diabetes mellitus, systemic lupus erythematosus, aging, and cancer. However, starved fibroblasts showed immunosuppressive activity in an in vitro model of allogeneic transplantation, suggesting their potential to modify such adverse reactions by down-regulating the immune system.
Collapse
Affiliation(s)
- Negar Jafari
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Reza Gheitasi
- Institutes for Infectious Diseases and Infection Control, Jena University Hospital, Jena, Germany
| | - Hamid Reza Khorasani
- Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Babol, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Monireh Golpour
- Department of Immunology, Molecular and Cell Biology Research Center, Student Research Committee, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Maryam Mehri
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Kosar Nayeri
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Roghayeh Pourbagher
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | | | - Behnam Kalali
- Department of Medicine II, Klinikum Grosshadern, LMU University, 81377, Munich, Germany
| | - Amrollah Mostafazadeh
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
10
|
Poznyak AV, Orekhova VA, Sukhorukov VN, Khotina VA, Popov MA, Orekhov AN. Atheroprotective Aspects of Heat Shock Proteins. Int J Mol Sci 2023; 24:11750. [PMID: 37511509 PMCID: PMC10380699 DOI: 10.3390/ijms241411750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Atherosclerosis is a major global health problem. Being a harbinger of a large number of cardiovascular diseases, it ultimately leads to morbidity and mortality. At the same time, effective measures for the prevention and treatment of atherosclerosis have not been developed, to date. All available therapeutic options have a number of limitations. To understand the mechanisms behind the triggering and development of atherosclerosis, a deeper understanding of molecular interactions is needed. Heat shock proteins are important for the normal functioning of cells, actively helping cells adapt to gradual changes in the environment and survive in deadly conditions. Moreover, multiple HSP families play various roles in the progression of cardiovascular disorders. Some heat shock proteins have been shown to have antiatherosclerotic effects, while the role of others remains unclear. In this review, we considered certain aspects of the antiatherosclerotic activity of a number of heat shock proteins.
Collapse
Affiliation(s)
- Anastasia V Poznyak
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, 121609 Moscow, Russia
| | - Varvara A Orekhova
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, 121609 Moscow, Russia
| | - Vasily N Sukhorukov
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, 121609 Moscow, Russia
| | - Victoria A Khotina
- Institute of General Pathology and Pathophysiology, 8, Baltiyskaya St., 125315 Moscow, Russia
| | - Mikhail A Popov
- Department of Cardiac Surgery, Moscow Regional Research and Clinical Institute (MONIKI), 61/2, Shchepkin St., 129110 Moscow, Russia
| | - Alexander N Orekhov
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, 121609 Moscow, Russia
| |
Collapse
|
11
|
Zou Y, Shi H, Liu N, Wang H, Song X, Liu B. Mechanistic insights into heat shock protein 27, a potential therapeutic target for cardiovascular diseases. Front Cardiovasc Med 2023; 10:1195464. [PMID: 37252119 PMCID: PMC10219228 DOI: 10.3389/fcvm.2023.1195464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 04/25/2023] [Indexed: 05/31/2023] Open
Abstract
Heat shock protein 27 (HSP27) is a small chaperone protein that is overexpressed in a variety of cellular stress states. It is involved in regulating proteostasis and protecting cells from multiple sources of stress injury by stabilizing protein conformation and promoting the refolding of misfolded proteins. Previous studies have confirmed that HSP27 is involved in the development of cardiovascular diseases and plays an important regulatory role in this process. Herein, we comprehensively and systematically summarize the involvement of HSP27 and its phosphorylated form in pathophysiological processes, including oxidative stress, inflammatory responses, and apoptosis, and further explore the potential mechanisms and possible roles of HSP27 in the diagnosis and treatment of cardiovascular diseases. Targeting HSP27 is a promising future strategy for the treatment of cardiovascular diseases.
Collapse
|
12
|
Luo S, Zhang X, Xiao X, Luo W, Yang Z, Tang S, Huang W. Exploring Potential Biomarkers and Molecular Mechanisms of Ischemic Cardiomyopathy and COVID-19 Comorbidity Based on Bioinformatics and Systems Biology. Int J Mol Sci 2023; 24:ijms24076511. [PMID: 37047484 PMCID: PMC10094917 DOI: 10.3390/ijms24076511] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
Cardiovascular complications combined with COVID-19 (SARS-CoV-2) lead to a poor prognosis in patients. The common pathogenesis of ischemic cardiomyopathy (ICM) and COVID-19 is still unclear. Here, we explored potential molecular mechanisms and biomarkers for ICM and COVID-19. Common differentially expressed genes (DEGs) of ICM (GSE5406) and COVID-19 (GSE164805) were identified using GEO2R. We performed enrichment and protein–protein interaction analyses and screened key genes. To confirm the diagnostic performance for these hub genes, we used external datasets (GSE116250 and GSE211979) and plotted ROC curves. Transcription factor and microRNA regulatory networks were constructed for the validated hub genes. Finally, drug prediction and molecular docking validation were performed using cMAP. We identified 81 common DEGs, many of which were enriched in terms of their relation to angiogenesis. Three DEGs were identified as key hub genes (HSP90AA1, HSPA9, and SRSF1) in the protein–protein interaction analysis. These hub genes had high diagnostic performance in the four datasets (AUC > 0.7). Mir-16-5p and KLF9 transcription factor co-regulated these hub genes. The drugs vindesine and ON-01910 showed good binding performance to the hub genes. We identified HSP90AA1, HSPA9, and SRSF1 as markers for the co-pathogenesis of ICM and COVID-19, and showed that co-pathogenesis of ICM and COVID-19 may be related to angiogenesis. Vindesine and ON-01910 were predicted as potential therapeutic agents. Our findings will contribute to a deeper understanding of the comorbidity of ICM with COVID-19.
Collapse
|
13
|
Akram W, Rihan M, Ahmed S, Arora S, Ahmad S, Vashishth R. Marine-Derived Compounds Applied in Cardiovascular Diseases: Submerged Medicinal Industry. Mar Drugs 2023; 21:md21030193. [PMID: 36976242 PMCID: PMC10052127 DOI: 10.3390/md21030193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/20/2023] [Accepted: 02/20/2023] [Indexed: 03/29/2023] Open
Abstract
Cardiovascular diseases (CVDs) are among the most impactful illnesses globally. Currently, the available therapeutic option has several side effects, including hypotension, bradycardia, arrhythmia, and alteration in different ion concentrations. Recently, bioactive compounds from natural sources, including plants, microorganisms, and marine creatures, have gained a lot of interest. Marine sources serve as reservoirs for new bioactive metabolites with various pharmacological activities. The marine-derived compound such as omega-3 acid ethyl esters, xyloketal B, asperlin, and saringosterol showed promising results in several CVDs. The present review focuses on marine-derived compounds' cardioprotective potential for hypertension, ischemic heart disease, myocardial infarction, and atherosclerosis. In addition to therapeutic alternatives, the current use of marine-derived components, the future trajectory, and restrictions are also reviewed.
Collapse
Affiliation(s)
- Wasim Akram
- Department of Pharmacology, SPER, Jamia Hamdard, New Delhi 110062, India
| | - Mohd Rihan
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Mohali 160062, India
| | - Sakeel Ahmed
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Mohali 160062, India
| | - Swamita Arora
- Department of Pharmacology, R. V. Northland Institute of Pharmacy, Dadri 203207, India
| | - Sameer Ahmad
- Department of Food Technology Jamia Hamdard, New Delhi 110062, India
| | - Rahul Vashishth
- School of BioSciences and Technology-Food Technology, Vellore Institute of Technology, Vellore 632014, India
| |
Collapse
|
14
|
Selenium Nanoparticles Can Influence the Immune Response Due to Interactions with Antibodies and Modulation of the Physiological State of Granulocytes. Pharmaceutics 2022; 14:pharmaceutics14122772. [PMID: 36559266 PMCID: PMC9783826 DOI: 10.3390/pharmaceutics14122772] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/02/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
Currently, selenium nanoparticles (SeNPs) are considered potential immunomodulatory agents and as targets for activity modulation are granulocytes, which have the most abundant population of immune blood cells. The present study aims to evaluate the cytotoxic effect and its effect on the functional responses of granulocytes. In addition to the intrinsic activity of SeNPs, we studied the activity of the combination of SeNPs and IgG antibodies. Using laser ablation and fragmentation, we obtained nanoparticles with an average size of 100 nm and a rather narrow size evolution. The resulting nanoparticles do not show acute toxicity to primary cultures of fibroblasts and hepatocytes, epithelial-like cell line L-929 and granulocyte-like culture of HL-60 at a concentration of 109 NPs/mL. SeNPs at a concentration of 1010 NPs/mL reduced the viability of HL-60 cells by no more than 10% and did not affect the viability of the primary culture of mouse granulocytes, and did not have a genotoxic effect on progenitor cells. The addition of SeNPs can affect the production of reactive oxygen species (ROS) by mouse bone marrow granulocytes, modulate the proportion of granulocytes with calcium spikes and enhance fMLF-induced granulocytes degranulation. SeNPs can modulate the effect of IgG on the physiological responses of granulocytes. We studied the expression level of genes associated with inflammation and cell stress. SeNPs increase the expression of catalase, NF-κB, Xrcc5 and some others; antibodies enhance the effect of SeNPs, but IgG without SeNPs decreases the expression level of these genes. This fact can be explained by the interaction between SeNPs and IgG. It has been established that antibodies interact with SeNPs. We showed that antibodies bind to the surface of selenium nanoparticles and are present in aqueous solutions in a bound form from DLS methods, ultraviolet-visible spectroscopy, vibrational-rotational spectrometry, fluorescence spectrometry, and refractometry. At the same time, in a significant part of the antibodies, a partial change in the tertiary and secondary structure is observed. The data obtained will allow a better understanding of the principles of the interaction of immune cells with antibodies and SeNPs and, in the future, may serve to create a new generation of immunomodulators.
Collapse
|