1
|
Xuan W, Wu X, Zheng L, Jia H, Zhang X, Zhang X, Cao B. Gut microbiota-derived acetic acids promoted sepsis-induced acute respiratory distress syndrome by delaying neutrophil apoptosis through FABP4. Cell Mol Life Sci 2024; 81:438. [PMID: 39453486 PMCID: PMC11511807 DOI: 10.1007/s00018-024-05474-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 08/28/2024] [Accepted: 10/04/2024] [Indexed: 10/26/2024]
Abstract
In patients with sepsis, neutrophil apoptosis tends to be inversely proportional to the severity of sepsis, but its mechanism is not yet clear. This study aimed to explore the mechanism of fatty acid binding protein 4 (FABP4) regulating neutrophil apoptosis through combined analysis of gut microbiota and short-chain fatty acids (SCFAs) metabolism. First, neutrophils from bronchoalveolar lavage fluid (BALF) of patients with sepsis-induced acute respiratory distress syndrome (ARDS) were purified and isolated RNA was applied for sequencing. Then, the cecal ligation and puncture (CLP) method was applied to induce the mouse sepsis model. After intervention with differential SCFAs sodium acetate, neutrophil apoptosis and FABP4 expression were further analyzed. Then, FABP4 inhibitor BMS309403 was used to treat neutrophils. We found CLP group had increased lung injury score, lung tissue wet/dry ratio, lung vascular permeability, and inflammatory factors IL-1β, TNF-α, IL-6, IFN-γ, and CCL3 levels in both bronchoalveolar lavage fluid and lung tissue. Additionally, FABP4 was lower in neutrophils of ARDS patients and mice. Meanwhile, CLP-induced dysbiosis of gut microbiota and changes in SCFAs levels were observed. Further verification showed that acetic acids reduced neutrophil apoptosis and FABP4 expression via FFAR2. Besides, FABP4 affected neutrophil apoptosis through endoplasmic reticulum (ER) stress, and neutrophil depletion alleviated the promotion of ARDS development by BMS309403. Moreover, FABP4 in neutrophils regulated the injury of RLE-6TN through inflammatory factors. In conclusion, FABP4 affected by gut microbiota-derived SCFAs delayed neutrophil apoptosis through ER stress, leading to increased inflammatory factors mediating lung epithelial cell damage.
Collapse
Affiliation(s)
- Weixia Xuan
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital (People's Hospital of Zhengzhou University), Zhengzhou, China
| | - Xu Wu
- Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421000, Hunan, China.
| | - Longcheng Zheng
- People's Hospital of Henan University, Department of Respiratory and Critical Care Medicine, People's Hospital of Henan Province, Zhengzhou, 450003, China
| | - Huayun Jia
- Hunan Province Center for Disease Control and Prevention, Changsha, 410000, Hunan, China
| | - Xiaoju Zhang
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital (People's Hospital of Zhengzhou University), Zhengzhou, China
| | - Xulong Zhang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
- Department of Respiratory Medicine, Capital Medical University, Beijing, 100069, China.
| | - Bin Cao
- Department of Pulmonary and Critical Care Medicine, Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, 100029, China.
- National Clinical Research Center for Respiratory Diseases, Beijing, 100029, China.
- Department of Respiratory Medicine, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
2
|
Fontes NFDA, Fernandes M, González-Ballesteros N, Rodríguez-Argüelles MC, Gomes AC, Duarte ASG. Exploring the Therapeutic Potential of Green-Synthesized Gold Nanoparticles and Ericaria selaginoides Extract for Inflammatory Bowel Disease. Antioxidants (Basel) 2024; 13:884. [PMID: 39199130 PMCID: PMC11351725 DOI: 10.3390/antiox13080884] [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: 05/22/2024] [Revised: 07/13/2024] [Accepted: 07/15/2024] [Indexed: 09/01/2024] Open
Abstract
Addressing disease remission and treatment adherence in inflammatory bowel diseases (IBDs), such as Crohn's disease, poses significant challenges due to underlying oxidative and inflammatory processes. Nanotechnology emerges as a promising avenue for enhancing therapeutic outcomes in IBD by optimizing drug bioactivity, reducing toxicity, and extending circulation time. Gold nanoparticles, known for their resistance to gastrointestinal pH and possessing antioxidant and anti-inflammatory properties, offer particular promise. They can be produced by green synthesis with seaweed Ericaria selaginoides (ES), itself associated with gastroprotective and anti-inflammatory activities. In a murine model of Crohn's disease induced with 8% acetic acid, pretreatment with dexamethasone (0.2 mL/30 g) or Au@ES (25 and 50 mg/kg) effectively mitigated inflammatory features. Notably, ES (50 mg/kg) and Au@ES (50 mg/kg) administration resulted in significant reductions in both macroscopic and microscopic inflammation scores compared to the disease control group. Furthermore, these treatments normalized inflammatory cytokine expression while safeguarding myenteric plexus glial cells. They also impeded neutrophil activation, leading to reduced myeloperoxidase activity and lipid peroxidation, coupled with increased glutathione levels. In conclusion, ES and Au@ES exhibit potent efficacy in counteracting inflammation and oxidation processes in an experimental Crohn's disease model, suggesting their potential as alternative therapeutic strategies for IBD.
Collapse
Affiliation(s)
- Nayana Freire de Almeida Fontes
- Departamento de Morfologia, Faculdade de Medicina, Centro de Ciências da Saúde, Universidade Federal do Ceará, Fortaleza 60440-900, Brazil (A.S.G.D.)
| | - Mário Fernandes
- Centre of Molecular and Environmental Biology (CBMA)/Aquatic Research Network (ARNET) Associate Laboratory, Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- Institute of Science and Innovation for Sustainability (IB-S), Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | | | | | - Andreia Castro Gomes
- Centre of Molecular and Environmental Biology (CBMA)/Aquatic Research Network (ARNET) Associate Laboratory, Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- Institute of Science and Innovation for Sustainability (IB-S), Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Antoniella Souza Gomes Duarte
- Departamento de Morfologia, Faculdade de Medicina, Centro de Ciências da Saúde, Universidade Federal do Ceará, Fortaleza 60440-900, Brazil (A.S.G.D.)
| |
Collapse
|
3
|
Liu Y, Hu S, Shi B, Yu B, Luo W, Peng S, Du X. The Role of Iron Metabolism in Sepsis-associated Encephalopathy: a Potential Target. Mol Neurobiol 2024; 61:4677-4690. [PMID: 38110647 DOI: 10.1007/s12035-023-03870-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 11/30/2023] [Indexed: 12/20/2023]
Abstract
Sepsis-associated encephalopathy (SAE) is an acute cerebral dysfunction secondary to infection, and the severity can range from mild delirium to deep coma. Disorders of iron metabolism have been proven to play an important role in a variety of neurodegenerative diseases by inducing cell damage through iron accumulation in glial cells and neurons. Recent studies have found that iron accumulation is also a potential mechanism of SAE. Systemic inflammation can induce changes in the expression of transporters and receptors on cells, especially high expression of divalent metal transporter1 (DMT1) and low expression of ferroportin (Fpn) 1, which leads to iron accumulation in cells. Excessive free Fe2+ can participate in the Fenton reaction to produce reactive oxygen species (ROS) to directly damage cells or induce ferroptosis. As a result, it may be of great help to improve SAE by treatment of targeting disorders of iron metabolism. Therefore, it is important to review the current research progress on the mechanism of SAE based on iron metabolism disorders. In addition, we also briefly describe the current status of SAE and iron metabolism disorders and emphasize the therapeutic prospect of targeting iron accumulation as a treatment for SAE, especially iron chelator. Moreover, drug delivery and side effects can be improved with the development of nanotechnology. This work suggests that treating SAE based on disorders of iron metabolism will be a thriving field.
Collapse
Affiliation(s)
- Yinuo Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- The Clinical Medical College of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Shengnan Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- The Clinical Medical College of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Bowen Shi
- The Clinical Medical College of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Bodong Yu
- The Clinical Medical College of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Wei Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Shengliang Peng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China.
| | - Xiaohong Du
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
4
|
Chiang MC, Yang YP, Nicol CJB, Wang CJ. Gold Nanoparticles in Neurological Diseases: A Review of Neuroprotection. Int J Mol Sci 2024; 25:2360. [PMID: 38397037 PMCID: PMC10888679 DOI: 10.3390/ijms25042360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/10/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
This review explores the diverse applications of gold nanoparticles (AuNPs) in neurological diseases, with a specific focus on Alzheimer's disease (AD), Parkinson's disease (PD), and stroke. The introduction highlights the pivotal role of neuroinflammation in these disorders and introduces the unique properties of AuNPs. The review's core examines the mechanisms by which AuNPs exert neuroprotection and anti-neuro-inflammatory effects, elucidating various pathways through which they manifest these properties. The potential therapeutic applications of AuNPs in AD are discussed, shedding light on promising avenues for therapy. This review also explores the prospects of utilizing AuNPs in PD interventions, presenting a hopeful outlook for future treatments. Additionally, the review delves into the potential of AuNPs in providing neuroprotection after strokes, emphasizing their significance in mitigating cerebrovascular accidents' aftermath. Experimental findings from cellular and animal models are consolidated to provide a comprehensive overview of AuNPs' effectiveness, offering insights into their impact at both the cellular and in vivo levels. This review enhances our understanding of AuNPs' applications in neurological diseases and lays the groundwork for innovative therapeutic strategies in neurology.
Collapse
Affiliation(s)
- Ming-Chang Chiang
- Department of Life Science, College of Science and Engineering, Fu Jen Catholic University, New Taipei City 242, Taiwan
| | - Yu-Ping Yang
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Christopher J. B. Nicol
- Departments of Pathology & Molecular Medicine and Biomedical & Molecular Sciences, Cancer Biology and Genetics Division, Cancer Research Institute, Queen’s University, Kingston, ON K7L 3N6, Canada;
| | - Chieh-Ju Wang
- Department of Life Science, College of Science and Engineering, Fu Jen Catholic University, New Taipei City 242, Taiwan
| |
Collapse
|
5
|
Jin X, Sun H, Yang L. How Extracellular Nano-Vesicles Can Play a Role in Sepsis? An Evidence-Based Review of the Literature. Int J Nanomedicine 2023; 18:5797-5814. [PMID: 37869065 PMCID: PMC10588718 DOI: 10.2147/ijn.s427116] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 10/08/2023] [Indexed: 10/24/2023] Open
Abstract
Sepsis is a systemic inflammatory reaction caused by infection. Severe sepsis can lead to multiple organ dysfunction, with a high incidence rate and mortality. The molecular pathogenesis of sepsis is complex and diverse. In recent years, with further study of the role of extracellular vesicles (EVs) in inflammatory diseases, it has been found that EVs play a dual role in the imbalance of inflammatory response in sepsis. Due to the great advantages such as lower toxicity, lower immunogenicity compared with stem cells and better circulation stability, EVs are increasingly used for the diagnosis and treatment of sepsis. The roles of EVs in the pathogenesis, diagnosis and treatment of sepsis were summarized to guide further clinical studies.
Collapse
Affiliation(s)
- Xiaolin Jin
- Department of International Physical Examination Center, The First Hospital of China Medical University, Shengyang, People’s Republic of China
| | - Haiyan Sun
- Department of Endodontics, School of Stomatology, China Medical University, Shenyang, People’s Republic of China
| | - Lina Yang
- Department of International Physical Examination Center, The First Hospital of China Medical University, Shengyang, People’s Republic of China
- Department of Geriatrics, The First Hospital of China Medical University, Shenyang, People’s Republic of China
| |
Collapse
|
6
|
Dumbuya JS, Chen X, Du J, Li S, Liang L, Xie H, Zeng Q. Hydrogen-rich saline regulates NLRP3 inflammasome activation in sepsis-associated encephalopathy rat model. Int Immunopharmacol 2023; 123:110758. [PMID: 37556997 DOI: 10.1016/j.intimp.2023.110758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/30/2023] [Accepted: 08/01/2023] [Indexed: 08/11/2023]
Abstract
Sepsis-associated encephalopathy (SAE) is characterised by long-term cognitive impairment and psychiatric illness in sepsis survivors, associated with increased morbidity and mortality. There is a lack of effective therapeutics for SAE. Molecular hydrogen (H2) plays multiple roles in septic diseases by regulating neuroinflammation, reducing oxidative stress parameters, regulating signalling pathways, improving mitochondrial dysfunction, and regulating astrocyte and microglia activation. Here we report the protective effect of hydrogen-rich saline in the juvenile SAE rat model and its possible underlying mechanisms. Rats were injected intraperitoneally with lipopolysaccharide at a dose of 5 mg/kg to induce sepsis; Hydrogen-rich saline (HRS) was administered 1 h after LPS induction at a dose of 5 ml/kg and nigericin at 1 mg/kg 1 h before LPS injection. H&E staining for neuronal damage, TUNEL assay for detection of apoptotic cells, immunofluorescence, ELISA protocol for inflammatory cytokines and 8-OHdG determination and western blot analysis to determine the effect of HRS in LPS-induced septic rats. Rats treated with HRS showed decreased TNF-α and IL-1β expression levels. HRS treatment enhanced the activities of antioxidant enzymes (SOD, CAT and GPX) and decreased MDA and MPO activities. The number of MMP-9 and NLRP3 positive immunoreactivity cells decreased in the HRS-treated group. Subsequently, GFAP, IBA-1 and CD86 immunoreactivity were reduced, and CD206 increased after HRS treatment. 8-OHdG expression was decreased in the HRS-treated rats. Western blot analysis showed decreased NLRP3, ASC, caspase-1, MMP-2/9, TLR4 and Bax protein levels after HRS treatment, while Bcl-2 expression increased after HRS treatment. These data demonstrated that HRS attenuated neuroinflammation, NLRP3 inflammasome activation, neuronal injury, and mitochondrial damage via NLRP3/Caspase-1/TLR4 signalling in the juvenile rat model, making it a potential therapeutic agent in the treatment of paediatric SAE.
Collapse
Affiliation(s)
- John Sieh Dumbuya
- Department of Paediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282 PR China
| | - Xinxin Chen
- Department of Paediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282 PR China
| | - Jiang Du
- Department of Paediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282 PR China
| | - Siqi Li
- Department of Paediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282 PR China
| | - Lili Liang
- Department of Paediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282 PR China
| | - Hairui Xie
- Department of Paediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282 PR China.
| | - Qiyi Zeng
- Department of Paediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282 PR China.
| |
Collapse
|
7
|
Smołka S, Skorupa M, Fołta K, Banaś A, Balcerzak K, Krok D, Shyntum DY, Skonieczna M, Turczyn R, Krukiewicz K. Antibacterial coatings for electroceutical devices based on PEDOT decorated with gold and silver particles. Bioelectrochemistry 2023; 153:108484. [PMID: 37302335 DOI: 10.1016/j.bioelechem.2023.108484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 06/13/2023]
Abstract
The continuous progression in the field of electrotherapies implies the development of multifunctional materials exhibiting excellent electrochemical performance and biocompatibility, promoting cell adhesion, and possessing antibacterial properties. Since the conditions favouring the adhesion of mammalian cells are similar to conditions favouring the adhesion of bacterial cells, it is necessary to engineer the surface to exhibit selective toxicity, i.e., to kill or inhibit the growth of bacteria without damaging mammalian tissues. The aim of this paper is to introduce a surface modification approach based on a subsequent deposition of silver and gold particles on the surface of a conducting polymer, poly(3,4-ethylenedioxythiophene) (PEDOT). The resulting PEDOT-Au/Ag surface is found to possess optimal wettability, roughness, and surface features making it an excellent platform for cell adhesion. By depositing Ag particles on PEDOT surface decorated with Au particles, it is possible to reduce toxic effects of Ag particles, while maintaining their antibacterial activity. Besides, electroactive and capacitive properties of PEDOT-Au/Ag account for its applicability in various electroceutical therapies.
Collapse
Affiliation(s)
- Szymon Smołka
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, M. Strzody 9, 44-100 Gliwice, Poland
| | - Małgorzata Skorupa
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, M. Strzody 9, 44-100 Gliwice, Poland; Joint Doctoral School, Silesian University of Technology, Akademicka 2A, Gliwice, Poland
| | - Kaja Fołta
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, M. Strzody 9, 44-100 Gliwice, Poland
| | - Angelika Banaś
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, M. Strzody 9, 44-100 Gliwice, Poland
| | - Kinga Balcerzak
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, M. Strzody 9, 44-100 Gliwice, Poland
| | - Dawid Krok
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, M. Strzody 9, 44-100 Gliwice, Poland
| | - Divine Yufetar Shyntum
- Biotechnology Centre, Silesian University of Technology, B. Krzywoustego 8, 44-100 Gliwice, Poland
| | - Magdalena Skonieczna
- Biotechnology Centre, Silesian University of Technology, B. Krzywoustego 8, 44-100 Gliwice, Poland; Department of Systems Biology and Engineering, Faculty of Automatic Control, Electronics and Computer Science, Silesian University of Technology, Akademicka 16, 44-100 Gliwice, Poland
| | - Roman Turczyn
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, M. Strzody 9, 44-100 Gliwice, Poland; Centre for Organic and Nanohybrid Electronics, Silesian University of Technology, S. Konarskiego 22B, 44-100 Gliwice, Poland
| | - Katarzyna Krukiewicz
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, M. Strzody 9, 44-100 Gliwice, Poland; Centre for Organic and Nanohybrid Electronics, Silesian University of Technology, S. Konarskiego 22B, 44-100 Gliwice, Poland.
| |
Collapse
|
8
|
Aili M, Zhou K, Zhan J, Zheng H, Luo F. Anti-inflammatory role of gold nanoparticles in the prevention and treatment of Alzheimer's disease. J Mater Chem B 2023; 11:8605-8621. [PMID: 37615596 DOI: 10.1039/d3tb01023f] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that causes memory and cognitive dysfunction and reduces a person's decision-making and reasoning functions. AD is the leading cause of dementia in the elderly. Patients with AD have increased expression of pro-inflammatory cytokines in the nervous system, and the sustained inflammatory response impairs neuronal function. Meanwhile, long-term use of anti-inflammatory drugs can reduce the incidence of AD to some extent. This confirms that anti-neuroinflammation may be an effective treatment for AD. Gold nanoparticles (AuNPs) are an emerging nanomaterial with promising physicochemical properties, anti-inflammatory and antioxidant. AuNPs reduce neuroinflammation by inducing macrophage polarization toward the M2 phenotype, reducing pro-inflammatory cytokine expression, blocking leukocyte adhesion, and decreasing oxidative stress. Therefore, AuNPs are gradually attracting the interest of scholars and are used for treating inflammatory diseases and drug delivery. Herein, we explored the role and mechanism of AuNPs in treating neuroinflammation in AD. The use of AuNPs for treating AD is a topic worth exploring in the future, not only to help solve a global public health problem but also to provide a reference for treating other neuroinflammatory diseases.
Collapse
Affiliation(s)
- Munire Aili
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Kebing Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Jun Zhan
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Huaping Zheng
- Department of Dermatology, Rare Diseases Center, Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Feng Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China.
- Department of Prosthodontics, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renmin Nanlu, Chengdu 610041, China
| |
Collapse
|
9
|
Hunt RD, Sedighi O, Clark WM, Doiron AL, Cipolla M. Differential effect of gold nanoparticles on cerebrovascular function and biomechanical properties. Physiol Rep 2023; 11:e15789. [PMID: 37604668 PMCID: PMC10442527 DOI: 10.14814/phy2.15789] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/24/2023] [Accepted: 07/24/2023] [Indexed: 08/23/2023] Open
Abstract
Human stroke serum (HSS) has been shown to impair cerebrovascular function, likely by factors released into the circulation after ischemia. 20 nm gold nanoparticles (GNPs) have demonstrated anti-inflammatory properties, with evidence that they decrease pathologic markers of ischemic severity. Whether GNPs affect cerebrovascular function, and potentially protect against the damaging effects of HSS on the cerebral circulation remains unclear. HSS obtained 24 h poststroke was perfused through the lumen of isolated and pressurized third-order posterior cerebral arteries (PCAs) from male Wistar rats with and without GNPs (~2 × 109 GNP/ml), or GNPs in vehicle, in an arteriograph chamber (n = 8/group). All vessels were myogenically reactive ≥60 mmHg intravascular pressure; however, vessels containing GNPs had significantly less myogenic tone. GNPs increased vasoreactivity to small and intermediate conductance calcium activated potassium channel activation via NS309; however, reduced vasoconstriction to nitric oxide synthase inhibition. Hydraulic conductivity and transvascular filtration, were decreased by GNPs, suggesting a protective effect on the blood-brain barrier. The stress-strain curves of PCAs exposed to GNPs were shifted leftward, indicating increased vessel stiffness. This study provides the first evidence that GNPs affect the structure and function of the cerebrovasculature, which may be important for their development and use in biomedical applications.
Collapse
Affiliation(s)
- Ryan D. Hunt
- Department of Neurological SciencesUniversity of Vermont Larner College of MedicineBurlingtonVermontUSA
| | - Omid Sedighi
- Department of Electrical and Biomedical EngineeringUniversity of Vermont College of Engineering and Mathematical SciencesBurlingtonVermontUSA
| | - Wayne M. Clark
- Oregon Stroke Center, Department of NeurologyOregon Health, and Science UniversityPortlandUSA
| | - Amber L. Doiron
- Department of Electrical and Biomedical EngineeringUniversity of Vermont College of Engineering and Mathematical SciencesBurlingtonVermontUSA
| | - Marilyn J. Cipolla
- Department of Neurological SciencesUniversity of Vermont Larner College of MedicineBurlingtonVermontUSA
- Department of Electrical and Biomedical EngineeringUniversity of Vermont College of Engineering and Mathematical SciencesBurlingtonVermontUSA
- Department of Obstetrics, Gynecology and Reproductive SciencesUniversity of Vermont Larner College of MedicineBurlingtonVermontUSA
- Department of PharmacologyUniversity of Vermont Larner College of MedicineBurlingtonVermontUSA
| |
Collapse
|
10
|
El Hanafi K, Gomez-Gomez B, Pedrero Z, Bustamante P, Cherel Y, Amouroux D, Madrid Y. Simple and rapid formic acid sample treatment for the isolation of HgSe nanoparticles from animal tissues. Anal Chim Acta 2023; 1250:340952. [PMID: 36898809 DOI: 10.1016/j.aca.2023.340952] [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: 12/02/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023]
Abstract
The present work explores for the first time the potential of formic acid on the extraction of tiemannite (HgSe) nanoparticles from seabird tissues, in particular giant petrels. Mercury (Hg) is considered one of the top ten chemicals of major public health concern. However, the fate and metabolic pathways of Hg in living organisms remain unknown. Methylmercury (MeHg), largely produced by microbial activity in the aquatic ecosystems is biomagnified in the trophic web. HgSe is considered the end-product of MeHg demethylation in biota and an increasing number of studies focuses on the characterization of this solid compound to understand its biomineralization. In this study, a conventional enzymatic treatment is compared with a simpler and environmentally friendly extraction by using formic acid (5 mL of = 50 % formic acid) as exclusive reagent. The analyses by spICP-MS of the resulting extracts from a variety of seabird biological tissues (liver, kidneys, brain, muscle) reveal comparable results by both extraction approaches in terms of nanoparticles stability and extraction efficiency. Therefore, the results included in this work demonstrate the good performance of employing organic acid as simple, cost effective and green procedure to extract HgSe nanoparticles from animal tissues. Moreover, an alternative consisting of a classical enzymatic procedure but with ultrasonic assistance reducing the extraction time from 12 h to 2 min is also described for the first time. The sample processing methodologies developed, combined with spICP-MS, have emerged as powerful tools for the rapid screening and quantification of HgSe nanoparticles in animal tissues. Finally, this combination allowed us to identify the possible occurrence of Cd particles and As particles associated with HgSe NPs in seabirds.
Collapse
Affiliation(s)
- K El Hanafi
- Université de Pau et des Pays de l'Adour, E2S UPPA, CNRS, IPREM, Institut des Sciences Analytiques et de Physico-chimie pour l'Environnement et les Matériaux, Pau, France
| | - B Gomez-Gomez
- Departamento de Química Analítica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Madrid, 28040, Spain
| | - Z Pedrero
- Université de Pau et des Pays de l'Adour, E2S UPPA, CNRS, IPREM, Institut des Sciences Analytiques et de Physico-chimie pour l'Environnement et les Matériaux, Pau, France.
| | - P Bustamante
- Littoral Environnement et Sociétés (LIENSs), UMR 7266 CNRS- La Rochelle Université, 2 Rue Olympe de Gouges, 17000, La Rochelle, France; Institut Universitaire de France (IUF), 1 Rue Descartes, 75005, Paris, France
| | - Y Cherel
- Centre d'Etudes Biologiques de Chizé, UMR 7372 CNRS-La Rochelle Université, 79360, Villiers-en-Bois, France
| | - D Amouroux
- Université de Pau et des Pays de l'Adour, E2S UPPA, CNRS, IPREM, Institut des Sciences Analytiques et de Physico-chimie pour l'Environnement et les Matériaux, Pau, France
| | - Y Madrid
- Departamento de Química Analítica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Madrid, 28040, Spain
| |
Collapse
|
11
|
Peres RAS, Silva-Aguiar RP, Teixeira DE, Peruchetti DB, Alves SAS, Leal ABC, Castro GF, Ribeiro NBS, Guimarães FV, Pinheiro AAS, Silva PMRE, Martins MA, Caruso-Neves C. Gold nanoparticles reduce tubule-interstitial injury and proteinuria in a murine model of subclinical acute kidney injury. Biochim Biophys Acta Gen Subj 2023; 1867:130314. [PMID: 36693453 DOI: 10.1016/j.bbagen.2023.130314] [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: 08/26/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 01/22/2023]
Abstract
Subclinical acute kidney injury (subAKI) is characterized by tubule-interstitial injury without significant changes in glomerular function. SubAKI is associated with the pathogenesis and progression of acute and chronic kidney diseases. Currently, therapeutic strategies to treat subAKI are limited. The use of gold nanoparticles (AuNPs) has shown promising benefits in different models of diseases. However, their possible effects on subAKI are still unknown. Here, we investigated the effects of AuNPs on a mouse model of subAKI. Animals with subAKI showed increased functional and histopathologic markers of tubular injury. There were no changes in glomerular function and structure. The animals with subAKI also presented an inflammatory profile demonstrated by activation of Th1 and Th17 cells in the renal cortex. This phenotype was associated with decreased megalin-mediated albumin endocytosis and expression of proximal tubular megalin. AuNP treatment prevented tubule-interstitial injury induced by subAKI. This effect was associated with a shift to an anti-inflammatory Th2 response. Furthermore, AuNP treatment preserved megalin-mediated albumin endocytosis in vivo and in vitro. AuNPs were not nephrotoxic in healthy mice. These results suggest that AuNPs have a protective effect in the tubule-interstitial injury observed in subAKI, highlighting a promising strategy as a future antiproteinuric treatment.
Collapse
Affiliation(s)
- Rodrigo A S Peres
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rodrigo P Silva-Aguiar
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Douglas E Teixeira
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Diogo B Peruchetti
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sarah A S Alves
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Anna Beatriz C Leal
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Guilherme F Castro
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Natalia B S Ribeiro
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Fernanda V Guimarães
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Ana Acacia S Pinheiro
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil
| | - Patrícia M R E Silva
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil
| | - Marco A Martins
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil
| | - Celso Caruso-Neves
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil.
| |
Collapse
|
12
|
Yin XY, Tang XH, Wang SX, Zhao YC, Jia M, Yang JJ, Ji MH, Shen JC. HMGB1 mediates synaptic loss and cognitive impairment in an animal model of sepsis-associated encephalopathy. J Neuroinflammation 2023; 20:69. [PMID: 36906561 PMCID: PMC10007818 DOI: 10.1186/s12974-023-02756-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 03/02/2023] [Indexed: 03/13/2023] Open
Abstract
BACKGROUND Microglial activation-mediated neuroinflammation is one of the essential pathogenic mechanisms of sepsis-associated encephalopathy (SAE). Mounting evidence suggests that high mobility group box-1 protein (HMGB1) plays a pivotal role in neuroinflammation and SAE, yet the mechanism by which HMGB1 induces cognitive impairment in SAE remains unclear. Therefore, this study aimed to investigate the mechanism of HMGB1 underlying cognitive impairment in SAE. METHODS An SAE model was established by cecal ligation and puncture (CLP); animals in the sham group underwent cecum exposure alone without ligation and perforation. Mice in the inflachromene (ICM) group were continuously injected with ICM intraperitoneally at a daily dose of 10 mg/kg for 9 days starting 1 h before the CLP operation. The open field, novel object recognition, and Y maze tests were performed on days 14-18 after surgery to assess locomotor activity and cognitive function. HMGB1 secretion, the state of microglia, and neuronal activity were measured by immunofluorescence. Golgi staining was performed to detect changes in neuronal morphology and dendritic spine density. In vitro electrophysiology was performed to detect changes in long-term potentiation (LTP) in the CA1 of the hippocampus. In vivo electrophysiology was performed to detect the changes in neural oscillation of the hippocampus. RESULTS CLP-induced cognitive impairment was accompanied by increased HMGB1 secretion and microglial activation. The phagocytic capacity of microglia was enhanced, resulting in aberrant pruning of excitatory synapses in the hippocampus. The loss of excitatory synapses reduced neuronal activity, impaired LTP, and decreased theta oscillation in the hippocampus. Inhibiting HMGB1 secretion by ICM treatment reversed these changes. CONCLUSIONS HMGB1 induces microglial activation, aberrant synaptic pruning, and neuron dysfunction in an animal model of SAE, leading to cognitive impairment. These results suggest that HMGB1 might be a target for SAE treatment.
Collapse
Affiliation(s)
- Xiao-Yu Yin
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Xiao-Hui Tang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Shi-Xu Wang
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Yong-Chang Zhao
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Min Jia
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Jian-Jun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China.
| | - Mu-Huo Ji
- Department of Anesthesiology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210011, China.
| | - Jin-Chun Shen
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China.
| |
Collapse
|
13
|
Zhu L, Luo M, Zhang Y, Fang F, Li M, An F, Zhao D, Zhang J. Free radical as a double-edged sword in disease: Deriving strategic opportunities for nanotherapeutics. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.214875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
14
|
Dorovskikh SI, Vikulova ES, Sergeevichev DS, Guselnikova TY, Zheravin AA, Nasimov DA, Vasilieva MB, Chepeleva EV, Saprykin AI, Basova TV, Morozova NB. Biological Studies of New Implant Materials Based on Carbon and Polymer Carriers with Film Heterostructures Containing Noble Metals. Biomedicines 2022; 10:biomedicines10092230. [PMID: 36140329 PMCID: PMC9496383 DOI: 10.3390/biomedicines10092230] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 11/18/2022] Open
Abstract
This paper presents pioneering results on the evaluation of noble metal film hetero-structures to improve some functional characteristics of carbon-based implant materials: carbon-composite material (CCM) and carbon-fiber-reinforced polyetheretherketone (CFR-PEEK). Metal-organic chemical vapor deposition (MOCVD) was successfully applied to the deposition of Ir, Pt, and PtIr films on these carriers. A noble metal layer as thin as 1 µm provided clear X-ray imaging of 1−2.5 mm thick CFR-PEEK samples. The coated and pristine CCM and CFR-PEEK samples were further surface-modified with Au and Ag nanoparticles (NPs) through MOCVD and physical vapor deposition (PVD) processes, respectively. The composition and microstructural features, the NPs sizes, and surface concentrations were determined. In vitro biological studies included tests for cytotoxicity and antibacterial properties. A series of samples were selected for subcutaneous implantation in rats (up to 3 months) and histological studies. The bimetallic PtIr-based heterostructures showed no cytotoxicity in vitro, but were less biocompatible due to a dense two-layered fibrous capsule. AuNP heterostructures on CFR-PEEK promoted cell proliferation in vitro and exhibited a strong inhibition of bacterial growth (p < 0.05) and high in vitro biocompatibility, especially Au/Ir structures. AgNP heterostructures showed a more pronounced antibacterial effect, while their in vivo biocompatibility was better than that of the pristine CFR-PEEK, but worse than that of AuNP heterostructures.
Collapse
Affiliation(s)
- Svetlana I. Dorovskikh
- Nikolaev Institute of Inorganic Chemistry Siberian Branch of the Russian Academy of Sciences SB RAS, 3 Lavrentiev Ave., 630090 Novosibirsk, Russia
| | - Evgeniia S. Vikulova
- Nikolaev Institute of Inorganic Chemistry Siberian Branch of the Russian Academy of Sciences SB RAS, 3 Lavrentiev Ave., 630090 Novosibirsk, Russia
| | - David S. Sergeevichev
- «E. Meshalkin National Medical Research Center» of the Ministry of Health of the Russian Federation, 15 Rechkunovskaya Str., 630055 Novosibirsk, Russia
| | - Tatiana Ya. Guselnikova
- Nikolaev Institute of Inorganic Chemistry Siberian Branch of the Russian Academy of Sciences SB RAS, 3 Lavrentiev Ave., 630090 Novosibirsk, Russia
| | - Alexander A. Zheravin
- «E. Meshalkin National Medical Research Center» of the Ministry of Health of the Russian Federation, 15 Rechkunovskaya Str., 630055 Novosibirsk, Russia
| | - Dmitriy A. Nasimov
- Rzhanov Institute of Semiconductor Physics SB RAS, 13 Lavrentiev Ave., 630090 Novosibirsk, Russia
| | - Maria B. Vasilieva
- «E. Meshalkin National Medical Research Center» of the Ministry of Health of the Russian Federation, 15 Rechkunovskaya Str., 630055 Novosibirsk, Russia
- Zelman Institute for the Medicine and Psychology, Novosibirsk State University, 1, Pirogov Str., 630090 Novosibirsk, Russia
| | - Elena V. Chepeleva
- «E. Meshalkin National Medical Research Center» of the Ministry of Health of the Russian Federation, 15 Rechkunovskaya Str., 630055 Novosibirsk, Russia
| | - Anatoly I. Saprykin
- Nikolaev Institute of Inorganic Chemistry Siberian Branch of the Russian Academy of Sciences SB RAS, 3 Lavrentiev Ave., 630090 Novosibirsk, Russia
| | - Tamara V. Basova
- Nikolaev Institute of Inorganic Chemistry Siberian Branch of the Russian Academy of Sciences SB RAS, 3 Lavrentiev Ave., 630090 Novosibirsk, Russia
| | - Natalya B. Morozova
- Nikolaev Institute of Inorganic Chemistry Siberian Branch of the Russian Academy of Sciences SB RAS, 3 Lavrentiev Ave., 630090 Novosibirsk, Russia
- Correspondence: ; Tel.: +7-3833309556
| |
Collapse
|
15
|
Serra MF, Cotias AC, Pimentel AS, Arantes ACSD, Pires ALA, Lanzetti M, Hickmann JM, Barreto E, Carvalho VF, Silva PMRE, Cordeiro RSB, Martins MA. Gold Nanoparticles Inhibit Steroid-Insensitive Asthma in Mice Preserving Histone Deacetylase 2 and NRF2 Pathways. Antioxidants (Basel) 2022; 11:antiox11091659. [PMID: 36139733 PMCID: PMC9495660 DOI: 10.3390/antiox11091659] [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: 07/01/2022] [Revised: 08/19/2022] [Accepted: 08/21/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Gold nanoparticles (AuNPs) can inhibit pivotal pathological changes in experimental asthma, but their effect on steroid-insensitive asthma is unclear. The current study assessed the effectiveness of nebulized AuNPs in a murine model of glucocorticoid (GC)-resistant asthma. Methods: A/J mice were sensitized and subjected to intranasal instillations of ovalbumin (OVA) once a week for nine weeks. Two weeks after starting allergen stimulations, mice were subjected to Budesonide or AuNP nebulization 1 h before stimuli. Analyses were carried out 24 h after the last provocation. Results: We found that mice challenged with OVA had airway hyperreactivity, eosinophil, and neutrophil infiltrates in the lung, concomitantly with peribronchiolar fibrosis, mucus production, and pro-inflammatory cytokine generation compared to sham-challenged mice. These changes were inhibited in mice treated with AuNPs, but not Budesonide. In the GC-resistant asthmatic mice, oxidative stress was established, marked by a reduction in nuclear factor erythroid 2-related factor 2 (NRF2) levels and catalase activity, accompanied by elevated values of thiobarbituric acid reactive substances (TBARS), phosphoinositide 3-kinases δ (PI3Kδ) expression, as well as a reduction in the nuclear expression of histone deacetylase 2 (HDAC2) in the lung tissue, all of which sensitive to AuNPs but not Budesonide treatment. Conclusion: These findings suggest that AuNPs can improve GC-insensitive asthma by preserving HDAC2 and NRF2.
Collapse
Affiliation(s)
- Magda F Serra
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, RJ, Brazil
| | - Amanda C Cotias
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, RJ, Brazil
| | - Andreza S Pimentel
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, RJ, Brazil
| | - Ana Carolina S de Arantes
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, RJ, Brazil
| | - Ana Lucia A Pires
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, RJ, Brazil
| | - Manuella Lanzetti
- Institute of Biomedical Science, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Jandir M Hickmann
- Institute of Physics, Federal University of Rio Grande do Sul, Porto Alegre 91509-900, RS, Brazil
| | - Emiliano Barreto
- Laboratory of Cell Biology, Federal University of Alagoas, Maceió 50072-900, AL, Brazil
| | - Vinicius F Carvalho
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, RJ, Brazil
| | - Patrícia M R E Silva
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, RJ, Brazil
| | - Renato S B Cordeiro
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, RJ, Brazil
| | - Marco Aurélio Martins
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, RJ, Brazil
| |
Collapse
|
16
|
Gao YL, Liu YC, Zhang X, Shou ST, Chai YF. Insight Into Regulatory T Cells in Sepsis-Associated Encephalopathy. Front Neurol 2022; 13:830784. [PMID: 35370925 PMCID: PMC8965708 DOI: 10.3389/fneur.2022.830784] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 02/18/2022] [Indexed: 01/09/2023] Open
Abstract
Sepsis-associated encephalopathy (SAE) is a diffuse central nervous system (CNS) dysfunction during sepsis, and is associated with increased mortality and poor outcomes in septic patients. Despite the high incidence and clinical relevance, the exact mechanisms driving SAE pathogenesis are not yet fully understood, and no specific therapeutic strategies are available. Regulatory T cells (Tregs) have a role in SAE pathogenesis, thought to be related with alleviation of sepsis-induced hyper-inflammation and immune responses, promotion of T helper (Th) 2 cells functional shift, neuroinflammation resolution, improvement of the blood-brain barrier (BBB) function, among others. Moreover, in a clinical point of view, these cells have the potential value of improving neurological and psychiatric/mental symptoms in SAE patients. This review aims to provide a general overview of SAE from its initial clinical presentation to long-term cognitive impairment and summarizes the main features of its pathogenesis. Additionally, a detailed overview on the main mechanisms by which Tregs may impact SAE pathogenesis is given. Finally, and considering that Tregs may be a novel target for immunomodulatory intervention in SAE, different therapeutic options, aiming to boost peripheral and brain infiltration of Tregs, are discussed.
Collapse
Affiliation(s)
- Yu-lei Gao
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
- Yu-lei Gao
| | - Yan-cun Liu
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiang Zhang
- Department of Emergency Medicine, Rizhao People's Hospital of Shandong Province, Rizhao, China
| | - Song-tao Shou
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan-fen Chai
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
- *Correspondence: Yan-fen Chai
| |
Collapse
|
17
|
Zare I, Yaraki MT, Speranza G, Najafabadi AH, Haghighi AS, Nik AB, Manshian BB, Saraiva C, Soenen SJ, Kogan MJ, Lee JW, Apollo NV, Bernardino L, Araya E, Mayer D, Mao G, Hamblin MR. Gold nanostructures: synthesis, properties, and neurological applications. Chem Soc Rev 2022; 51:2601-2680. [PMID: 35234776 DOI: 10.1039/d1cs01111a] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recent advances in technology are expected to increase our current understanding of neuroscience. Nanotechnology and nanomaterials can alter and control neural functionality in both in vitro and in vivo experimental setups. The intersection between neuroscience and nanoscience may generate long-term neural interfaces adapted at the molecular level. Owing to their intrinsic physicochemical characteristics, gold nanostructures (GNSs) have received much attention in neuroscience, especially for combined diagnostic and therapeutic (theragnostic) purposes. GNSs have been successfully employed to stimulate and monitor neurophysiological signals. Hence, GNSs could provide a promising solution for the regeneration and recovery of neural tissue, novel neuroprotective strategies, and integrated implantable materials. This review covers the broad range of neurological applications of GNS-based materials to improve clinical diagnosis and therapy. Sub-topics include neurotoxicity, targeted delivery of therapeutics to the central nervous system (CNS), neurochemical sensing, neuromodulation, neuroimaging, neurotherapy, tissue engineering, and neural regeneration. It focuses on core concepts of GNSs in neurology, to circumvent the limitations and significant obstacles of innovative approaches in neurobiology and neurochemistry, including theragnostics. We will discuss recent advances in the use of GNSs to overcome current bottlenecks and tackle technical and conceptual challenges.
Collapse
Affiliation(s)
- Iman Zare
- Research and Development Department, Sina Medical Biochemistry Technologies Co. Ltd., Shiraz 7178795844, Iran
| | | | - Giorgio Speranza
- CMM - FBK, v. Sommarive 18, 38123 Trento, Italy.,IFN - CNR, CSMFO Lab., via alla Cascata 56/C Povo, 38123 Trento, Italy.,Department of Industrial Engineering, University of Trento, v. Sommarive 9, 38123 Trento, Italy
| | - Alireza Hassani Najafabadi
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA.,Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Alireza Shourangiz Haghighi
- Department of Mechanical Engineering, Shiraz University of Technology, Modarres Boulevard, 13876-71557, Shiraz, Iran
| | - Amirala Bakhshian Nik
- Department of Biomedical Engineering, Florida International University, Miami, FL 33174, USA
| | - Bella B Manshian
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Cláudia Saraiva
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 Avenue des Hauts-Fourneaux, 4362 Esch-sur-Alzette, Luxembourg.,Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Rua Marques d'Avila e Bolama, 6201-001 Covilha, Portugal
| | - Stefaan J Soenen
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Marcelo J Kogan
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas, Departamento de Química Farmacológica y Toxicológica, Universidad de Chile, 8380492 Santiago, Chile
| | - Jee Woong Lee
- Department of Medical Sciences, Clinical Neurophysiology, Uppsala University, Uppsala, SE-751 23, Sweden
| | - Nicholas V Apollo
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,School of Physics, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Liliana Bernardino
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Rua Marques d'Avila e Bolama, 6201-001 Covilha, Portugal
| | - Eyleen Araya
- Departamento de Ciencias Quimicas, Facultad de Ciencias Exactas, Universidad Andres Bello, Av. Republica 275, Santiago, Chile
| | - Dirk Mayer
- Institute of Biological Information Processing, Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, Germany
| | - Guangzhao Mao
- School of Chemical Engineering, University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia
| | - Michael R Hamblin
- Laser Research Center, University of Johannesburg, Doorfontein 2028, South Africa.
| |
Collapse
|
18
|
Zhang Y, Chen S, Tian W, Zhu H, Li W, Dai W, Zhang X, Gu X, Su D. Emerging Trends and Hot Spots in Sepsis-Associated Encephalopathy Research From 2001 to 2021: A Bibliometric Analysis. Front Med (Lausanne) 2022; 9:817351. [PMID: 35295600 PMCID: PMC8918530 DOI: 10.3389/fmed.2022.817351] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/27/2022] [Indexed: 12/30/2022] Open
Abstract
Study Objectives To evaluate sepsis-associated encephalopathy (SAE) research and to quantitatively and qualitatively predict research hot spots using bibliometric analysis. Methods We extracted relevant publications from the Web of Science Core Collection on July 28, 2021. We investigated the retrieved data by bibliometric analysis (e.g. co-cited and cluster analysis, keyword co-occurrence) using the software CiteSpace and VOSviewer, the Online Analysis Platform of Literature Metrology (http://bibliometric.com/) and Bibliometrix to analyse and predict the trends and hot spots in this field. Main Results We identified 1,582 published articles and reviews on SAE from 2001 to 2021. During this period, the number of manuscripts on SAE increased steadily and peaked in 2021. The USA and China were the leading countries that had a critical impact on SAE research. Among all institutions, Vanderbilt University and Pittsburgh University held leading positions and became central in the collaboration network. Among all the journals, Critical Care Medicine published the maximum number of manuscripts in the field of SAE within 20 years. Dal-Pizzol Felipe was the most productive author (61 papers) and received the largest number of citations (930 citations). Co-citation cluster analysis revealed that the most popular terms on SAE in the manner of cluster labels were critical illness, sepsis-associated encephalopathy, polymicrobial sepsis, posterior reversible encephalopathy syndrome, rat brain, intensive care unit, prior sepsis, molecular hydrogen, inflammation drive, metabolic encephalopathies, delirium pathophysiology, and clinical neuroscience. Keyword burst detection indicated that neuroinflammation, blood-brain barrier (BBB) and mitochondria dysfunction were the current research hot spots. Conclusions Our study revealed that neuroinflammation, blood-brain barrier, and mitochondria dysfunction had been the research foci of SAE over the past 20 years. These have emerged as the basis for transformation from basic research to clinical application in finding effective methods for the prevention and treatment of SAE.
Collapse
|
19
|
Ko WC, Wang SJ, Hsiao CY, Hung CT, Hsu YJ, Chang DC, Hung CF. Pharmacological Role of Functionalized Gold Nanoparticles in Disease Applications. Molecules 2022; 27:1551. [PMID: 35268651 PMCID: PMC8911979 DOI: 10.3390/molecules27051551] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/19/2022] [Accepted: 02/24/2022] [Indexed: 12/12/2022] Open
Abstract
Gold has always been regarded as a symbol of nobility, and its shiny golden appearance has always attracted the attention of many people. Gold has good ductility, molecular recognition properties, and good biocompatibility. At present, gold is being used in many fields. When gold particles are as small as several nanometers, their physical and chemical properties vary with their size in nanometers. The surface area of a nano-sized gold surface has a special effect. Therefore, gold nanoparticles can, directly and indirectly, give rise to different biological activities. For example, if the surface of the gold is sulfided. Various substances have a strong chemical reactivity and are easy to combine with sulfhydryl groups; hence, nanogold is often used in biomedical testing, disease diagnosis, and gene detection. Nanogold is easy to bind to proteins, such as antibodies, enzymes, or cytokines. In fact, scientists use nanogold to bind special antibodies, as a tool for targeting cancer cells. Gold nanoparticles are also directly cytotoxic to cancer cells. For diseases caused by inflammation and oxidative damage, gold nanoparticles also have antioxidant and anti-inflammatory effects. Based on these unique properties, gold nanoparticles have become the most widely studied metal nanomaterials. Many recent studies have further demonstrated that gold nanoparticles are beneficial for humans, due to their functional pharmacological properties in a variety of diseases. The content of this review will be the application of gold nanoparticles in treating or diagnosing pressing diseases, such as cancers, retinopathy, neurological diseases, skin disorders, bowel diseases, bone cartilage disorders, cardiovascular diseases, infections, and metabolic syndrome. Gold nanoparticles have shown very obvious therapeutic and application potential.
Collapse
Affiliation(s)
- Wen-Chin Ko
- School of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan; (W.-C.K.); (S.-J.W.)
- Division of Cardiac Electrophysiology, Department of Cardiovascular Center, Cathay General Hospital, Taipei 10630, Taiwan
| | - Su-Jane Wang
- School of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan; (W.-C.K.); (S.-J.W.)
| | - Chien-Yu Hsiao
- Department of Nutrition and Health Science, Chang Guang University of Science and Technology, Taoyuan 33303, Taiwan;
- Research Center for Food and Cosmetic Safety and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan
| | - Chen-Ting Hung
- Graduate Institute and Department of Pharmacology, National Taiwan University College of Medicine, Taipei 10051, Taiwan;
| | - Yu-Jou Hsu
- PhD Program in Pharmaceutical Biotechnology, Fu Jen Catholic University, New Taipei City 24205, Taiwan;
| | - Der-Chen Chang
- Department of Mathematics and Statistics and Department of Computer Science, Georgetown University, Washington, DC 20057, USA;
| | - Chi-Feng Hung
- School of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan; (W.-C.K.); (S.-J.W.)
- PhD Program in Pharmaceutical Biotechnology, Fu Jen Catholic University, New Taipei City 24205, Taiwan;
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| |
Collapse
|
20
|
Ding H, Li Y, Chen S, Wen Y, Zhang S, Luo E, Li X, Zhong W, Zeng H. Fisetin ameliorates cognitive impairment by activating mitophagy and suppressing neuroinflammation in rats with sepsis-associated encephalopathy. CNS Neurosci Ther 2021; 28:247-258. [PMID: 34837343 PMCID: PMC8739041 DOI: 10.1111/cns.13765] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 11/06/2021] [Accepted: 11/09/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Fisetin, the effective ingredient of the traditional Chinese medicine named Cotinus coggygria, is recommended to be active therapeutic in many disorders. However, its role in sepsis-associated encephalopathy (SAE) remains unclarified. METHODS Cecal ligation and puncture (CLP) operation was performed to establish a rat model of SAE. Rats were grouped according to the surgery operation and fisetin administration. Cognitive impairment was assessed by Morris water maze test. Disruption of blood-brain barrier (BBB) integrity was detected by Evan's blue staining. The mitophagy, reactive oxygen species (ROS) generation, NLRP3 inflammasome activation, and pro-inflammatory cytokines levels were measured through western blot and double immunofluorescence labeling. A transmission electron microscope was applied for the observation of mitochondrial autophagosomes. RESULTS Rats in the CLP group presented increased expression of IL-1R1, pNF-κB, TNF-α, and iNOS in microglial cells, indicating severe inflammation in the central nervous system (CNS). Nevertheless, there was no increase in BBB permeability. Meanwhile, NLRP3 inflammasome was activated in cerebral microvascular endothelial cells (CMECs), presented with an elevation of caspase-1 expression and IL-1β secretion into CNS. In addition, we found fisetin significantly improved cognitive dysfunction in rats with SAE. Neuroprotective effects of fisetin might be associated with inhibition of neuroinflammation, represented with decreased expression of IL-1R1, pNF-κB, TNF-α, and iNOS in microglia. Furthermore, fisetin induced mitophagy, scavenged ROS, blocked NLRP3 inflammasome activation of CMECs, as evidenced by decreased expression of caspase-1 and reduced release of IL-1β into CNS. CONCLUSION Collectively, fisetin-blocked NLRP3 inflammasome activation via promoting mitophagy in CMECs may suppress the secretion of IL-1β into CNS, reduce neuroinflammation, and contribute to the amelioration of cognitive impairment.
Collapse
Affiliation(s)
- Hongguang Ding
- Department of Emergency Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ya Li
- Department of Emergency Medicine, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Shenglong Chen
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yin Wen
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Shiying Zhang
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ensi Luo
- Department of Endocrinology, Binhaiwan Central Hospital of Dongguan, Dongguan Hospital Affiliated to Medical College of Jinan University, Dongguan, China
| | - Xusheng Li
- Department of Emergency Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Wenhong Zhong
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Hongke Zeng
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
21
|
Peng X, Luo Z, He S, Zhang L, Li Y. Blood-Brain Barrier Disruption by Lipopolysaccharide and Sepsis-Associated Encephalopathy. Front Cell Infect Microbiol 2021; 11:768108. [PMID: 34804998 PMCID: PMC8599158 DOI: 10.3389/fcimb.2021.768108] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/14/2021] [Indexed: 12/29/2022] Open
Abstract
As a complex multicellular structure of the vascular system at the central nervous system (CNS), the blood-brain barrier (BBB) separates the CNS from the system circulation and regulates the influx and efflux of substances to maintain the steady-state environment of the CNS. Lipopolysaccharide (LPS), the cell wall component of Gram-negative bacteria, can damage the barrier function of BBB and further promote the occurrence and development of sepsis-associated encephalopathy (SAE). Here, we conduct a literature review of the direct and indirect damage mechanisms of LPS to BBB and the relationship between these processes and SAE. We believe that after LPS destroys BBB, a large number of inflammatory factors and neurotoxins will enter and damage the brain tissue, which will activate brain immune cells to mediate inflammatory response and in turn further destroys BBB. This vicious circle will ultimately lead to the progression of SAE. Finally, we present a succinct overview of the treatment of SAE by restoring the BBB barrier function and summarize novel opportunities in controlling the progression of SAE by targeting the BBB.
Collapse
Affiliation(s)
- Xiaoyao Peng
- Department of Clinical Medicine, School of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Zhixuan Luo
- Department of Clinical Medicine, School of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Shuang He
- Department of Clinical Medicine, School of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Luhua Zhang
- Department of Pathogenic Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Ying Li
- Department of Immunology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| |
Collapse
|
22
|
Anik MI, Mahmud N, Al Masud A, Hasan M. Gold nanoparticles (GNPs) in biomedical and clinical applications: A review. NANO SELECT 2021. [DOI: 10.1002/nano.202100255] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Muzahidul I. Anik
- Department of Chemical Engineering University of Rhode Island South Kingstown Rhode Island USA
| | - Niaz Mahmud
- Department of Biomedical Engineering Military Institute of Science and Technology Dhaka Bangladesh
| | - Abdullah Al Masud
- Department of Chemical Engineering Bangladesh University of Engineering and Technology Dhaka Bangladesh
| | - Maruf Hasan
- Department of Biomedical Engineering Military Institute of Science and Technology Dhaka Bangladesh
| |
Collapse
|