1
|
Zou J, Cui W, Deng N, Li C, Yang W, Ye X, Yao F, Zhang T, Xiao J, Ma C, Wu L, Dong D, Chen J, Guo C, Liu A, Wu H. Fate reversal: Exosome-driven macrophage rejuvenation and bacterial-responsive drug release for infection immunotherapy in diabetes. J Control Release 2025; 382:113730. [PMID: 40250625 DOI: 10.1016/j.jconrel.2025.113730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 03/27/2025] [Accepted: 04/11/2025] [Indexed: 04/20/2025]
Abstract
Superficial surgical site infection (SSI) is a significant risk factor for the development of periprosthetic joint infection (PJI), particularly in diabetic patients. A high-glucose microenvironment is observed to compromise phagocytosis by inducing cellular senescence, which leads to impaired antibacterial immune function. Exosomes derived from umbilical cord stem cells (H-Exos) can reverse the immunosuppressive microenvironment by rejuvenating senescent cells, thereby terminating excessive, persistent, and ineffective inflammatory responses. Thus, a novel exosome-based immunotherapeutic antibacterial strategy to reverse fate is proposed. Vancomycin & lysostaphin-loaded exosomes are incorporated in a customizable microneedle patch (ExoV-ExoL@MN) for controlled release, enabling tailored treatments for diverse clinical scenarios. While rejuvenating macrophage senescent phenotype, the antibiotics encapsulated within exosomes can be responsively released by the hemolysin secreted by bacteria, triggering rapid bacterial killing. Post-infection clearance, they induce a shift from M1 to M2 macrophage polarization, thereby enhancing anti-inflammatory and reparative responses. Furthermore, the components can be mixed on demand and at any time, allowing for real-time customization and fabrication directly at the clinic (fabrication@clinic). This strategy reverses the immunosuppressive microenvironment by rejuvenating senescent macrophages and effectively combats bacterial invasion into deep tissues through bacteria-responsive antibiotic release, providing a promising approach for preventing and treating SSI-induced PJI.
Collapse
Affiliation(s)
- Jiaxuan Zou
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310002, PR China; Orthopedics Research Institute of Zhejiang University, Hangzhou 310002, PR China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University, Hangzhou 310002, PR China; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310002, PR China
| | - Wushi Cui
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310002, PR China; Orthopedics Research Institute of Zhejiang University, Hangzhou 310002, PR China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University, Hangzhou 310002, PR China; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310002, PR China
| | - Niping Deng
- School of Engineering, Westlake University, Hangzhou 310024, PR China
| | - Congsun Li
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310002, PR China; Orthopedics Research Institute of Zhejiang University, Hangzhou 310002, PR China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University, Hangzhou 310002, PR China; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310002, PR China
| | - Weinan Yang
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310002, PR China; Orthopedics Research Institute of Zhejiang University, Hangzhou 310002, PR China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University, Hangzhou 310002, PR China; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310002, PR China
| | - Xiaojun Ye
- Department of Ultrasound, Hangzhou Women's Hospital, Hangzhou 310008, PR China
| | - Feng Yao
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310002, PR China; Orthopedics Research Institute of Zhejiang University, Hangzhou 310002, PR China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University, Hangzhou 310002, PR China; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310002, PR China
| | - Tao Zhang
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310002, PR China; Orthopedics Research Institute of Zhejiang University, Hangzhou 310002, PR China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University, Hangzhou 310002, PR China; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310002, PR China; Department of Orthopedics, Lishui Hospital, Zhejiang University School of Medicine, Lishui 323000, PR China
| | - Jian Xiao
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310002, PR China; Orthopedics Research Institute of Zhejiang University, Hangzhou 310002, PR China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University, Hangzhou 310002, PR China; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310002, PR China; Department of Orthopedics, The First People's Hospital of Jiashan, Jiaxing 314100, PR China
| | - Chiyuan Ma
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310002, PR China; Orthopedics Research Institute of Zhejiang University, Hangzhou 310002, PR China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University, Hangzhou 310002, PR China; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310002, PR China
| | - Lingfeng Wu
- Department of Orthopedics, Lishui Hospital, Zhejiang University School of Medicine, Lishui 323000, PR China
| | - Dahai Dong
- Department of Orthopedics, Suichang County People's Hospital in Zhejiang Province, Lishui 323300, PR China
| | - Jing Chen
- Institute of Medical Sciences, The Second Hospital and Shandong University Center for Orthopaedics, Cheeloo College of Medicine, Shandong University, Jinan 250033, PR China.
| | - Chengchen Guo
- School of Engineering, Westlake University, Hangzhou 310024, PR China.
| | - An Liu
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310002, PR China; Orthopedics Research Institute of Zhejiang University, Hangzhou 310002, PR China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University, Hangzhou 310002, PR China; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310002, PR China.
| | - Haobo Wu
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310002, PR China; Orthopedics Research Institute of Zhejiang University, Hangzhou 310002, PR China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University, Hangzhou 310002, PR China; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310002, PR China.
| |
Collapse
|
2
|
Saadh MJ, Allela OQB, Kareem RA, Kyada A, Malathi H, Nathiya D, Bhanot D, Sameer HN, Hamad AK, Athab ZH, Adil M. Immune cell dysfunction: A critical player in development of diabetes complications. Curr Res Transl Med 2025; 73:103510. [PMID: 40339429 DOI: 10.1016/j.retram.2025.103510] [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: 11/18/2024] [Revised: 03/08/2025] [Accepted: 03/28/2025] [Indexed: 05/10/2025]
Abstract
Diabetes mellitus, a global health challenge, influences millions worldwide by leading to severe complications and premature death. A key factor in its pathogenesis is immune cell dysfunction, which aggravates both type 1 and type 2 diabetes. The important role that immune cell dysregulation plays in the emergence of diabetes complications is investigated in this research. It highlights the manner in which diabetes compromises the immune system's adaptive as well as innate responses. Key defects in innate immunity include impaired pathogen recognition, and dysfunctional behavior of macrophages, neutrophils, and natural killer (NK) cells. Additionally, the complement system is dysregulated, and cytokine production is altered, affecting overall immune signaling. The study investigates the dysfunction of several T and B cell subsets, such as CD4+ T cells, CD8+ T cells, regulatory T cells, and B cells, in relation to adaptive immunity. These dysfunctions collectively contribute to chronic inflammation, reduced pathogen clearance, and increased susceptibility to infections, ultimately exacerbating diabetes complications. Developing targeted therapies to reduce diabetes complications and enhance patient outcomes requires an understanding of these mechanisms.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | | | | | - Ashishkumar Kyada
- Marwadi University Research Center, Department of Pharmacy, Faculty of Health Sciences, Marwadi University, Rajkot 360003, Gujarat, India
| | - H Malathi
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Deepak Nathiya
- Department of Pharmacy Practice, Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Deepak Bhanot
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar, 64001, Iraq
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | | |
Collapse
|
3
|
He X, Peng L, Zhou L, Liu H, Hao Y, Li Y, Lv Z, Zeng B, Guo X, Guo R. A biphasic drug-releasing microneedle with ROS scavenging and angiogenesis for the treatment of diabetic ulcers. Acta Biomater 2024; 189:270-285. [PMID: 39362454 DOI: 10.1016/j.actbio.2024.09.045] [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: 07/03/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/05/2024]
Abstract
Diabetic ulcers are one of the common complications in diabetic patients. Delayed wound healing is associated with persistent pro-inflammatory M1 polarization, reduced angiogenesis and increased reactive oxygen species (ROS) in the microenvironment. Wound healing consists of multiple phases and therefore requires treatment tailored to each phase. In this study, a biphasic drug-releasing microneedle (MN) was fabricated to achieve early ROS scavenging and late accelerated angiogenesis to promote wound healing. Vascular endothelial growth factor (VEGF) was first encapsulated in methacryloylated sulfonated chitosan (SCSMA) microspheres (V@MP), and then V@MP was loaded into hyaluronic acid (HA) microneedles along with cerium dioxide nanoparticles (CONPs). Rapid dissolution of HA rapidly releases the CONPs to clear ROS, whereas the V@MP stays in the wound. SCSMA slow degradation prolongs the release of VEGF, thereby promoting angiogenesis. In vitro and in vivo studies have shown that this biphasic drug-releasing smart microneedle improves cell proliferation and migration, effectively scavenges ROS, promotes angiogenesis and tissue regeneration, and synergistically promotes M2 macrophage polarization. It provides a new delivery mode for nano-enzymes and growth factors that could be multifunctional and synergistic in the treatment of diabetic ulcers. STATEMENT OF SIGNIFICANCE: In our study, we present a microneedle (V@MP/C@MN) that can release drugs biphasically, which showed good repair ability in diabetic ulcer model. Large amounts of CONPs were rapidly released to alleviate oxidative stress during the inflammation of the wound, and V@MP stayed in the wound for a long period of time to release VEGF and promote angiogenesis in the late stage of wound healing. The results indicated that V@MP/C@MN could promote cell proliferation and migration, effectively scavenge ROS, promote angiogenesis and tissue regeneration, and synergistically promote M2 macrophage polarization, which could play a multifunctional and synergistic role in the treatment of diabetic ulcers.
Collapse
Affiliation(s)
- Xinyue He
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Key Laboratory of Regenerative Medicine of Ministry of Education, Guangdong Provincial Engineering and Technological Research Centre for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Lianghong Peng
- Department of Ophthalmology, General Hospital of Southern Theater Command, PLA, Guangzhou 510010, China
| | - Liming Zhou
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Key Laboratory of Regenerative Medicine of Ministry of Education, Guangdong Provincial Engineering and Technological Research Centre for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Huiling Liu
- Head Department of Oral and Maxillofacial Surgery, Leiden University Medical Centre, Amsterdam, De Boelelaan 1117, the Netherlands
| | - Yifan Hao
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Key Laboratory of Regenerative Medicine of Ministry of Education, Guangdong Provincial Engineering and Technological Research Centre for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Yuhan Li
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Key Laboratory of Regenerative Medicine of Ministry of Education, Guangdong Provincial Engineering and Technological Research Centre for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Zijin Lv
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Key Laboratory of Regenerative Medicine of Ministry of Education, Guangdong Provincial Engineering and Technological Research Centre for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Baohui Zeng
- Department of Ultrasound, Guangzhou Red Cross Hospital of Jinan University, Guangzhou 510220, China
| | - Xinmin Guo
- Department of Ultrasound, Guangzhou Red Cross Hospital of Jinan University, Guangzhou 510220, China.
| | - Rui Guo
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Key Laboratory of Regenerative Medicine of Ministry of Education, Guangdong Provincial Engineering and Technological Research Centre for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
4
|
Zendehdel R, Hahad O, Panjali Z. Human epithelial lung cell toxicity assessment of collected graphite particles from an iron casting industry (in vitro study). INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2024; 34:3126-3135. [PMID: 38146704 DOI: 10.1080/09603123.2023.2298248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/18/2023] [Indexed: 12/27/2023]
Abstract
Workers in the iron casting industries are exposed to various chemicals, especially graphite in furnace process. This study aims to investigate the toxic effects of graphite particles on human lung cells. Particle characteristics were confirmed by electron microscope and light scattering. Cell viability and oxidative stress markers were measured. The expression of oxidative repair genes, namely OGG1, MTH1, and ITPA, was evaluated. The average particle size was determined to be 172.1 ± 11.96 nm. The median inhibition concentration (IC50) of graphite particles was 46.75 µg/mL. Notably, 25 and 50 µg/mL concentrations resulted in significant GSH depletion and MDA production. The high concentration of graphite particles (200 µg/mL) led to OGG1 suppression and increased MTH1 expression. Based on these findings, graphite exposure may induce toxicity in human lung cells by increasing oxidative stress. Further research is necessary to fully understand the mechanisms underlying graphite toxicity.
Collapse
Affiliation(s)
- Rezvan Zendehdel
- Department of Occupational Health and Safety, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Omar Hahad
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), partner site Rhine-Main, Mainz, Germany
| | - Zahra Panjali
- Department of Occupational Health and Safety, Faculty of Health Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
5
|
Khan H, Khanam A, Khan AA, Ahmad R, Husain A, Habib S, Ahmad S, Moinuddin. The complex landscape of intracellular signalling in protein modification under hyperglycaemic stress leading to metabolic disorders. Protein J 2024; 43:425-436. [PMID: 38491250 DOI: 10.1007/s10930-024-10191-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2024] [Indexed: 03/18/2024]
Abstract
Hyperglycaemia is a life-threatening risk factor that occurs in both chronic and acute phases and has been linked to causing injury to many organs. Protein modification was triggered by hyperglycaemic stress, which resulted in pathogenic alterations such as impaired cellular function and tissue damage. Dysregulation in cellular function increases the condition associated with metabolic disorders, including cardiovascular diseases, nephropathy, retinopathy, and neuropathy. Hyperglycaemic stress also increases the proliferation of cancer cells. The major areas of experimental biomedical research have focused on the underlying mechanisms involved in the cellular signalling systems involved in diabetes-associated chronic hyperglycaemia. Reactive oxygen species and oxidative stress generated by hyperglycaemia modify many intracellular signalling pathways that result in insulin resistance and β-cell function degradation. The dysregulation of post translational modification in β cells is clinically associated with the development of diabetes mellitus and its associated diseases. This review will discuss the effect of hyperglycaemic stress on protein modification and the cellular signalling involved in it. The focus will be on the significant molecular changes associated with severe metabolic disorders.
Collapse
Affiliation(s)
- Hamda Khan
- Department of Biochemistry, Faculty of Medicine, Jawahar Lal Nehru Medical College, Aligarh Muslim University, 202002, Aligarh, India.
| | - Afreen Khanam
- Department of Biotechnology and Life Sciences, Mangalayatan University, Aligarh, India
| | - Adnan Ahmad Khan
- Faculty of Pharmacy, Integral University, Lucknow, 226026, India
| | - Rizwan Ahmad
- Department of Biochemistry, Faculty of Medicine, Jawahar Lal Nehru Medical College, Aligarh Muslim University, 202002, Aligarh, India
| | - Arbab Husain
- Department of Biotechnology and Life Sciences, Mangalayatan University, Aligarh, India
| | - Safia Habib
- Department of Biochemistry, Faculty of Medicine, Jawahar Lal Nehru Medical College, Aligarh Muslim University, 202002, Aligarh, India
| | - Saheem Ahmad
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail, Saudi Arabia
| | - Moinuddin
- Department of Biochemistry, Faculty of Medicine, Jawahar Lal Nehru Medical College, Aligarh Muslim University, 202002, Aligarh, India
| |
Collapse
|
6
|
Liu Q, Yan X, Yuan Y, Li R, Zhao Y, Fu J, Wang J, Su J. HTRA2/OMI-Mediated Mitochondrial Quality Control Alters Macrophage Polarization Affecting Systemic Chronic Inflammation. Int J Mol Sci 2024; 25:1577. [PMID: 38338855 PMCID: PMC10855076 DOI: 10.3390/ijms25031577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
Systemic chronic inflammation (SCI) due to intrinsic immune over-activation is an important factor in the development of many noninfectious chronic diseases, such as neurodegenerative diseases and diabetes mellitus. Among these immune responses, macrophages are extensively involved in the regulation of inflammatory responses by virtue of their polarization plasticity; thus, dysregulation of macrophage polarization direction is one of the potential causes of the generation and maintenance of SCI. High-temperature demand protein A2 (HtrA2/Omi) is an important regulator of mitochondrial quality control, not only participating in the degradation of mis-accumulated proteins in the mitochondrial unfolded protein response (UPRmt) to maintain normal mitochondrial function through its enzymatic activity, but also participating in the regulation of mitochondrial dynamics-related protein interactions to maintain mitochondrial morphology. Recent studies have also reported the involvement of HtrA2/Omi as a novel inflammatory mediator in the regulation of the inflammatory response. HtrA2/Omi regulates the inflammatory response in BMDM by controlling TRAF2 stabilization in a collagen-induced arthritis mouse model; the lack of HtrA2 ameliorates pro-inflammatory cytokine expression in macrophages. In this review, we summarize the mechanisms by which HtrA2/Omi proteins are involved in macrophage polarization remodeling by influencing macrophage energy metabolism reprogramming through the regulation of inflammatory signaling pathways and mitochondrial quality control, elucidating the roles played by HtrA2/Omi proteins in inflammatory responses. In conclusion, interfering with HtrA2/Omi may become an important entry point for regulating macrophage polarization, providing new research space for developing HtrA2/Omi-based therapies for SCI.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jing Su
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basical Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130012, China
| |
Collapse
|
7
|
Rizwan H, Satapathy SS, Si S, Kumar S, Kumari G, Pal A. Effect of Au@SiO 2 core shell nanoparticles on HG-induced oxidative stress triggered apoptosis in keratinocytes. Life Sci 2023; 328:121893. [PMID: 37392778 DOI: 10.1016/j.lfs.2023.121893] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/17/2023] [Accepted: 06/25/2023] [Indexed: 07/03/2023]
Abstract
Growing evidences suggest that excess generation of highly reactive free oxygen/nitrogen radicals (ROS/RNS) are largely due to hyperglycemia causes oxidative stress. Further, excess accumulation of ROS/RNS in cellular compartments aggravates the development and progression of diabetes and its associated complications. Impaired wound healing in diabetic condition is a known vital complication all around the world. Thus, an antioxidant agent having the potential for hindering the oxidative/nitrosative stress triggered diabetic skin complication is required. The present investigation was carried out to understand the impact of silica coated gold nanoparticle (Au@SiO2 NPs) on high glucose (HG)-induced keratinocyte complications. We demonstrated that HG environment enhanced the ROS and RNS accumulations and reduced in cellular antioxidant capacities in keratinocte cells, however, Au@SiO2 NPs treatment restored the HG effect. Furthermore, excess production of ROS/RNS was associated with mitochondrial dysfunction, characterized by loss of mitochondrial membrane potential (ΔΨm), and increased in mitochondrial mass, which was restored by Au@SiO2 NPs treatment in keratinocyte cells. In addition, HG-induced excess production of ROS/RNA caused an increased in the biomolecules damage including lipid peroxidation (LPO), and protein carbonylation (PC), 8-oxoguanine DNA glycosylase-1 (OGG1) expression and increased 8-hydroxydeoxyguanosine (8-OHdG) accumulations in DNA, leading to activation of ERK1/2MAPK, AKT and tuberin pathway, inflammatory reaction, and finally apoptotic cell death. In conclusion, our findings showed that Au@SiO2 NPs treatment improved the HG-induced keratinocytes injury by suppressing the oxidative/nitrosative stress, elevating the antioxidant defence system, thereby inhibiting the inflammatory mediators and apoptosis, which may be a therapeutic cure for the diabetic keratinocyte problems.
Collapse
Affiliation(s)
- Huma Rizwan
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar 751024, India
| | - Smith Sagar Satapathy
- School of Chemical Technology, KIIT Deemed to be University, Bhubaneswar 751024, India
| | - Satyabrata Si
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar 751024, India; School of Chemical Technology, KIIT Deemed to be University, Bhubaneswar 751024, India
| | - Sonu Kumar
- Department of Zoology, School of Life Sciences, Mahatma Gandhi Central University, Motihari, Bihar 845401, India
| | - Golden Kumari
- Department of Zoology, School of Life Sciences, Mahatma Gandhi Central University, Motihari, Bihar 845401, India
| | - Arttatrana Pal
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar 751024, India; Department of Zoology, School of Life Sciences, Mahatma Gandhi Central University, Motihari, Bihar 845401, India.
| |
Collapse
|
8
|
Zhang W, Zhong R, Qu X, Xiang Y, Ji M. Effect of 8-Hydroxyguanine DNA Glycosylase 1 on the Function of Immune Cells. Antioxidants (Basel) 2023; 12:1300. [PMID: 37372030 DOI: 10.3390/antiox12061300] [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: 05/20/2023] [Revised: 06/07/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
Excess reactive oxygen species (ROS) can cause an imbalance between oxidation and anti-oxidation, leading to the occurrence of oxidative stress in the body. The most common product of ROS-induced base damage is 8-hydroxyguanine (8-oxoG). Failure to promptly remove 8-oxoG often causes mutations during DNA replication. 8-oxoG is cleared from cells by the 8-oxoG DNA glycosylase 1 (OGG1)-mediated oxidative damage base excision repair pathway so as to prevent cells from suffering dysfunction due to oxidative stress. Physiological immune homeostasis and, in particular, immune cell function are vulnerable to oxidative stress. Evidence suggests that inflammation, aging, cancer, and other diseases are related to an imbalance in immune homeostasis caused by oxidative stress. However, the role of the OGG1-mediated oxidative damage repair pathway in the activation and maintenance of immune cell function is unknown. This review summarizes the current understanding of the effect of OGG1 on immune cell function.
Collapse
Affiliation(s)
- Weiran Zhang
- Department of Physiology, School of Basic Medicine, Central South University, Changsha 410078, China
| | - Ranwei Zhong
- Department of Physiology, School of Basic Medicine, Central South University, Changsha 410078, China
| | - Xiangping Qu
- Department of Physiology, School of Basic Medicine, Central South University, Changsha 410078, China
| | - Yang Xiang
- Department of Physiology, School of Basic Medicine, Central South University, Changsha 410078, China
| | - Ming Ji
- Department of Physiology, School of Basic Medicine, Central South University, Changsha 410078, China
| |
Collapse
|
9
|
Rizwan H, Kumar S, Kumari G, Pal A. High glucose-induced increasing reactive nitrogen species accumulation triggered mitochondrial dysfunction, inflammation, and apoptosis in keratinocytes. Life Sci 2022; 312:121208. [PMID: 36427546 DOI: 10.1016/j.lfs.2022.121208] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/12/2022] [Accepted: 11/15/2022] [Indexed: 11/24/2022]
Abstract
Growing evidence indicates that skin injuries are a common complication of diabetes. However, the cellular and molecular mechanisms of high glucose (HG) environment trigger nitrosative stress-mediated inflammation and apoptosis in keratinocytes remains unknown. Here we investigated whether reactive nitrogen species (RNS) induced by HG environment restrain antioxidant activity, and mitochondrial dysfunction leading to inflammation, and apoptosis via stress signaling pathways in keratinocytes. Our results established that the HG environment enhanced the production of nitric oxide (NO) and peroxynitrite anion (ONOO-) by inducible NO synthase (iNOS) in keratinocytes. Overproduction of RNS in HG environment suppress the antioxidants activity leading to mitochondrial dysfunction, characterized by loss of mitochondrial membrane potential (ΔΨm), increase in mitochondrial mass, decrease in mitochondrial transcription factor A(TFAM), increase in mitochondrial DNA (mtDNA) displacement loop (D-loop) and decrease in glycolytic flux concentration, which was attenuated by pharmacological inhibitors of NO/ONOO-, Nω-Nitro-l-argininemethyl ester hydrochloride (NAME)/hydralazine hydrochloride (Hyd.HCl). Excess production of RNS in HG environment restrained 8-oxoguanine DNA glycosylase-1 (OGG1) expression and increased 8-hydroxydeoxyguanosine (8-OHdG) accumulations in DNA were regulated by NO or ONOO-. Further, HG-induced RNA production caused an increase in the production of inflammatory mediators accompanied by activation of ERK1/2MAPK/Akt/tuberin-mTOR/IRF3 signaling cascade, lipid peroxidation (LPO), and protein carbonylation (PC) reactions followed by breakdown the cell-cell communication and apoptosis. Pre-treatment of cell with NAME/Hyd.HCl, diminished the expression of ERK1/2MAPK/Akt/tuberin-mTOR/IRF3, inflammatory mediators, and attenuated apoptosis in keratinocytes. Together, our results indicated that excess production of RNS in HG environment triggered inflammation and apoptosis, mediated by activation of ERK1/2MAPK/Akt/tuberin-mTOR/IRF3 signaling cascades in keratinocytes.
Collapse
Affiliation(s)
- Huma Rizwan
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar 751024, India
| | - Sonu Kumar
- Department of Zoology, School of Life Sciences, Mahatma Gandhi Central University, Motihari, Bihar 845401, India
| | - Golden Kumari
- Department of Zoology, School of Life Sciences, Mahatma Gandhi Central University, Motihari, Bihar 845401, India
| | - Arttatrana Pal
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar 751024, India; Department of Zoology, School of Life Sciences, Mahatma Gandhi Central University, Motihari, Bihar 845401, India.
| |
Collapse
|
10
|
Tan F, Cao Y, Zheng L, Wang T, Zhao S, Chen J, Pang C, Xia W, Xia Z, Li N, Chi X. Diabetes exacerbated sepsis-induced intestinal injury by promoting M1 macrophage polarization via miR-3061/Snail1 signaling. Front Immunol 2022; 13:922614. [PMID: 36159784 PMCID: PMC9503829 DOI: 10.3389/fimmu.2022.922614] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 08/15/2022] [Indexed: 11/29/2022] Open
Abstract
Background Macrophages play important roles in diabetes and sepsis-related intestinal injury. Accumulating evidence suggests that microRNAs (miRNAs) act as the fundamental link between macrophage polarization and tissue injury. However, the underlying mechanisms of miRNAs in regulating macrophage polarization–related intestinal injury under diabetes and sepsis conditions remain unclear. Methods The cecal ligation and puncture (CLP)–induced sepsis models were established in male wild-type (WT) and diabetic mice. Clodronate liposome was used to deplete macrophage. H&E staining, inflammatory cytokines [tumor necrosis factor–α (TNF-α), interleukin-1β (IL-1β), and IL-6], and intestinal mucosal barrier function markers [occludin, ZO-1, lipopolysaccharide (LPS), and intestinal fatty acid binding protein (iFABP)] were used to assess elevated intestinal damage. miRNA array, RNA-seq, and bioinformatic analysis were performed to detect the miRNA and messenger RNA (mRNA) expression and the potential regulation mechanism. In vitro, RAW264.7 cells were cultured in the absence or presence of high glucose and LPS, miR-3061 mimics, and Snail small interfering RNA stimulation, respectively, for further mechanism studies. Luciferase reporter assay was used to confirm the interplay between miRNA and its target genes. Results Compared with WT CLP mice, the diabetic CLP mice showed severe intestinal damage characterized by significant increases in Chui’s scores, expression of inflammatory cytokines (TNF-α, IL-1β, and IL-6), serum LPS and iFABP concentration, and significant reductions in tight junction protein occludin and ZO-1 levels. Macrophage depletion reversed the intestinal damage caused by CLP. The bioinformatic analysis revealed that miR-3061/Snail1 might be a potential regulation axis of macrophage polarization. Furthermore, high glucose and LPS stimulation increased M1 macrophage and reduced the levels of miR-3061, which was negatively associated with Snail1 in RAW264.7 cells. Mechanistic studies demonstrated that miR-3061 regulated macrophage polarization by targeting the Snail1 mRNA 3′‐untranslated region. Moreover, miR-3061 overexpression suppressed Snail1 expression and inhibited M1 macrophage and inflammatory cytokines. Conclusion This study elucidated that diabetes exacerbated sepsis-induced intestinal injury by promoting M1 macrophage polarization and further demonstrated that the miR-3061/Sani1 axis may be the potential target of macrophage polarization.
Collapse
Affiliation(s)
- Fang Tan
- Department of Anaesthesiology, the Seventh Affiliated Hospital of Sun Yat-Sen University (SYSU), Shenzhen, China
| | - Yuling Cao
- Department of Anaesthesiology, the Seventh Affiliated Hospital of Sun Yat-Sen University (SYSU), Shenzhen, China
| | - Lei Zheng
- Department of Anaesthesiology, the Seventh Affiliated Hospital of Sun Yat-Sen University (SYSU), Shenzhen, China
| | - Tao Wang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-Sen University (SYSU), Shenzhen, China
| | - Shuhua Zhao
- Department of Anaesthesiology, the Seventh Affiliated Hospital of Sun Yat-Sen University (SYSU), Shenzhen, China
| | - Jiong Chen
- Department of Anaesthesiology, the Seventh Affiliated Hospital of Sun Yat-Sen University (SYSU), Shenzhen, China
| | - Changji Pang
- Department of Anaesthesiology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Weiyi Xia
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Zhengyuan Xia
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
- The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Ningning Li
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-Sen University (SYSU), Shenzhen, China
| | - Xinjin Chi
- Department of Anaesthesiology, the Seventh Affiliated Hospital of Sun Yat-Sen University (SYSU), Shenzhen, China
- *Correspondence: Xinjin Chi,
| |
Collapse
|
11
|
Panjali Z, Abdolmaleki P, Hajipour-Verdom B, Hahad O, Zendehdel R. Lung cell toxicity of co-exposure to airborne particulate matter and extremely low-frequency magnetic field. Xenobiotica 2022; 52:370-379. [PMID: 35608272 DOI: 10.1080/00498254.2022.2082342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Although the toxic effects of urban airborne particulate matter (PM) have been known on lung cells, there is less attention to co-exposure to PM and extremely low frequency magnetic (ELF-MF) in occupational settings. The present study investigated the influences of PM and ELF-MF co-exposure on toxicity in human lung cells (A549).In this case, total PM (TPM) was evaluated according to NIOSH-0500. The TPM SiO2 and metal contents were determined based on NIOSH-7602 and 7302, respectively. Besides, 900 mG ELF-MF exposure was simulated based on field measurements. The toxicity mechanisms were assessed by examining malondialdehyde, glutathione ratio, gene expression, and DNA strand breaks. Also, the toxicity indicators of the TPM samples were MDA generation, glutathione depletion, and DNA damage, and their impacts were analysed at doses below the LD50 (4 µg).In addition, gene expression of OGG1 and MTH1 was upregulated after TPM exposure at the lowest dose (2 µg). But ITPA was upregulated in the presence of ELF-MF. The co-exposure to TPM and ELF-MF decreased oxidative stress and DNA damage levels compared to a single exposure to TPM.Although the ELF-MF reduced toxicity in response to TPM, this reduction was not lower than the unexposed cells.
Collapse
Affiliation(s)
- Zahra Panjali
- Department of Occupational Health Engineering, Faculty of Health and Medical Engineering, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Parviz Abdolmaleki
- Department of Biophysics, Faculty of Biological Science, Tarbiat Modarres University, Tehran, Iran
| | - Behnam Hajipour-Verdom
- Department of Biophysics, Faculty of Biological Science, Tarbiat Modarres University, Tehran, Iran
| | - Omar Hahad
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Rezvan Zendehdel
- Department of Occupational Health and Safety, School of Public Health and Safety, Shahid Beheshti University of Medical Science, Tehran, Iran
| |
Collapse
|
12
|
Lima-Silva LF, Lee J, Moraes-Vieira PM. Soluble Carrier Transporters and Mitochondria in the Immunometabolic Regulation of Macrophages. Antioxid Redox Signal 2022; 36:906-919. [PMID: 34555943 PMCID: PMC9271333 DOI: 10.1089/ars.2021.0181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Significance: Immunometabolic regulation of macrophages is a growing area of research across many fields. Here, we review the contribution of solute carriers (SLCs) in regulating macrophage metabolism. We also highlight key mechanisms that regulate SLC function, their effects on mitochondrial activity, and how these intracellular activities contribute to macrophage fitness in health and disease. Recent Advances: SLCs serve as a major drug absorption pathway and represent a novel category of therapeutic drug targets. SLC dynamics affect cellular nutritional sensors, such as AMP-activated protein kinase and mammalian target of rapamycin, and consequently alter the cellular metabolism and mitochondrial dynamics within macrophages to adapt to a new functional phenotype. Critical Issues: SLC function affects macrophage phenotype, but their mechanisms of action and how their functions contribute to host health remain incompletely defined. Future Directions: Few studies focus on the impact of solute transporters on macrophage function. Identifying which SLCs are present in macrophages and determining their functional roles may reveal novel therapeutic targets with which to treat metabolic and inflammatory diseases. Antioxid. Redox Signal. 36, 906-919.
Collapse
Affiliation(s)
- Lincon Felipe Lima-Silva
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil.,Post Graduate Program in Immunology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Jennifer Lee
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Pedro M Moraes-Vieira
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil.,Experimental Medicine Research Cluster (EMRC), University of Campinas, Campinas, Brazil.,Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, Brazil
| |
Collapse
|
13
|
Tian H, Zhang Z, Han X, Pan T, Tao G, Jiao P, Zhai L, Yang L, Wang X, Yao Y, Qin S, Yao S. D4F alleviates the C/EBP homologous protein-mediated apoptosis in glycated high-density lipoprotein-treated macrophages by facilitating autophagy. Exp Biol Med (Maywood) 2021; 246:2595-2609. [PMID: 34525858 DOI: 10.1177/15353702211045323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The present study aimed to investigate the role of D4F, an apolipoprotein A-I mimetic peptide, in macrophage apoptosis induced by the glycated high-density lipoprotein (gly-HDL)-induced endoplasmic reticulum (ER) stress C/EBP homologous protein (CHOP) pathway, and unravel the regulatory role of autophagy in this process. Our results revealed that except for suppressing the accumulation of lipids within RAW264.7 macrophages caused by gly-HDL, D4F inhibited gly-HDL-induced decrease in the cell viability and increase in lactate dehydrogenase leakage and cell apoptosis, which were similar to 4-phenylbutyric acid (PBA, an ER stress inhibitor). Besides, similar to PBA, D4F inhibited gly-HDL-induced ER stress response activation evaluated through the decreased PERK and eIF2α phosphorylation, together with reduced ATF6 nuclear translocation as well as the downregulation of GRP78 and CHOP. Interestingly, D4F facilitated gly-HDL-triggered activation of autophagy, measured as elevated levels of beclin-1, LC3-II, and ATG5 expressions in macrophages. Furthermore, the inhibition effect of D4F on gly-HDL-induced ER stress-CHOP-induced apoptosis of macrophages was restrained after beclin-1 siRNA and 3-methyladenine (3-MA, an inhibitor of autophagy) treatments, while this effect was further reinforced after rapamycin (Rapa, an inducer of autophagy) treatment. Furthermore, administering D4F or Rapa to T2DM mice upregulated LC3-II and attenuated CHOP expression, cell apoptosis, and atherosclerotic lesions. However, the opposite results were obtained when 3-MA was administered to these mice. These results support that D4F effectively protects macrophages against gly-HDL-induced ER stress-CHOP-mediated apoptosis by promoting autophagy.
Collapse
Affiliation(s)
- Hua Tian
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China.,College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Zhaoqiang Zhang
- College of Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| | - Xiaoyan Han
- College of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| | - Tianqi Pan
- College of Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| | - Geru Tao
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| | - Peng Jiao
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| | - Lei Zhai
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| | - Libo Yang
- Department of Endocrinology, Central Hospital of Taian, Taian 271000, China
| | - Xiaoxu Wang
- College of Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| | - Yilin Yao
- College of Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| | - Shucun Qin
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| | - Shutong Yao
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China.,College of Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| |
Collapse
|
14
|
Pal S, Rao GN, Pal A. Inflammation and apoptosis, two key events induced by hyperglycemia mediated reactive nitrogen species in RGC-5 cells. Life Sci 2021; 279:119693. [PMID: 34111464 DOI: 10.1016/j.lfs.2021.119693] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 05/23/2021] [Accepted: 05/31/2021] [Indexed: 12/21/2022]
Abstract
Nitrosative stress plays a critical role in retinal injury in high glucose (HG) environment of eye, but the mechanisms remain poorly understood. Here we tested the hypothesis that HG induced reactive nitrogen species (RNS) production acts as a key functional mediator of antioxidant depletion, mitochondrial dysfunction, biomolecule damage, inflammation and apoptosis. Our findings illustrated that exposure of cultured RGC-5 cells to HG significantly disrupts the antioxidant defense mechanism and mitochondrial machineries by increasing the loss of mitochondrial membrane potential (ΔѰM) and elevating mitochondrial mass. Furthermore, we used biochemical tools to analyze the changes in metabolites, sulfur amino acids (SAAs) such as L-glutathione (GSH) and L-cysteine (Cys), in the presence of HG environment. These metabolic changes were followed by an increase in glycolytic flux that is phosphofructokinase-2 (PFK-2) activity. Moreover, HG exposure results in a significant disruption of protein carbonylation (PC) and lipid peroxidation (LPO), downregulation of OGG1 and increase in 8-OHdG accumulations in RGC-5 cells. In addition, our results demonstrated that HG environment coinciding with increased expression of inflammatory mediators, cell cycle deregulation, decreased in cell viability and expression of FoxOs, increased lysosomal content leading to apoptosis. Pre-treatment of selective inhibitors of RNS significantly reduced the HG-induced cell cycle deregulation and apoptosis in RGC-5 cells. Collectively, these results illustrated that accumulated RNS exacerbates the antioxidant depletion, mitochondrial dysfunction, biomolecule damage, inflammation and apoptosis induced by HG exposure in RGC-5 cells. Treatment of pharmacological inhibitors attenuated the HG induced in retinal cells.
Collapse
Affiliation(s)
- Sweta Pal
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar 751024, India
| | - G Nageswar Rao
- Department of Ophthalmology, Kalinga Institute of Medical Sciences, Kalinga Institute of Industrial Technology, Bhubaneswar 751024, India
| | - Arttatrana Pal
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar 751024, India; Department of Zoology, School of Life Sciences, Mahatma Gandhi Central University, Motihari 845401, India.
| |
Collapse
|
15
|
Wei Y, Gao X, Zhao F, Baimanov D, Cong Y, Jiang Y, Hameed S, Ouyang Y, Gao X, Lin X, Wang L. Induced Autophagy of Macrophages and the Regulation of Inflammatory Effects by Perovskite Nanomaterial LaNiO 3. Front Immunol 2021; 12:676773. [PMID: 33968087 PMCID: PMC8100511 DOI: 10.3389/fimmu.2021.676773] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 03/25/2021] [Indexed: 11/25/2022] Open
Abstract
Perovskite nanomaterials (NMs) possess excellent physicochemical properties and have promising applications in light-emitting diodes (LEDs), lasers, photodetectors, and artificial synapse electronics. Potential exposure to these NMs happens in the manufacture and application of the perovskite-based products, however, the biological safety of these NMs is still unknown. Here, we used the LaNiO3 NM (LNO), a typical kind of perovskite nanostructures to study the interaction with macrophages (J774A.1) and to explore its biological effects at the cellular level. Firstly, we characterized the properties of LNO including the size, shape, and crystal structure using Transmission electronic microscope (TEM), Dynamic lighting scattering (DLS), and X-ray diffraction (XRD). Secondly, to gain a better understanding of the biological effect, we evaluated the effect of LNO on cell viability and found that LNO induced cell autophagy at a concentration of 5 μg/ml and influenced the inflammatory response based on RT-PCR result. Finally, we demonstrated the mechanism that LNO causes cell autophagy and immune response is probably due to the metal ions released from LNO in acidic lysosomes, which triggered ROS and increased lysosomal membrane permeation. This study indicates the safety aspect of perovskite NMs and may guide the rational design of perovskite NMs with more biocompatibility during their manufacture and application.
Collapse
Affiliation(s)
- Yang Wei
- College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, China.,CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety & CAS-HKU Joint Laboratory of Metallomics on Health and Environment, and Beijing Metallomics Facility, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
| | - Xuejiao Gao
- College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, China
| | - Feng Zhao
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety & CAS-HKU Joint Laboratory of Metallomics on Health and Environment, and Beijing Metallomics Facility, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
| | - Didar Baimanov
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety & CAS-HKU Joint Laboratory of Metallomics on Health and Environment, and Beijing Metallomics Facility, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yalin Cong
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety & CAS-HKU Joint Laboratory of Metallomics on Health and Environment, and Beijing Metallomics Facility, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, China
| | - Yingying Jiang
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety & CAS-HKU Joint Laboratory of Metallomics on Health and Environment, and Beijing Metallomics Facility, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China.,School of Public Health, Qingdao University, Qingdao, China
| | - Saima Hameed
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety & CAS-HKU Joint Laboratory of Metallomics on Health and Environment, and Beijing Metallomics Facility, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
| | - Yixin Ouyang
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety & CAS-HKU Joint Laboratory of Metallomics on Health and Environment, and Beijing Metallomics Facility, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China.,School of Public Health, Capital Medical University, Beijing, China
| | - Xingfa Gao
- College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, China
| | - Xiaoying Lin
- College of Pharmacy, Jilin Medical University, Jilin, China
| | - Liming Wang
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety & CAS-HKU Joint Laboratory of Metallomics on Health and Environment, and Beijing Metallomics Facility, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
16
|
Pal S, Rao GN, Pal A. High glucose-induced ROS accumulation is a critical regulator of ERK1/2-Akt-tuberin-mTOR signalling in RGC-5 cells. Life Sci 2020; 256:117914. [DOI: 10.1016/j.lfs.2020.117914] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/25/2020] [Accepted: 06/01/2020] [Indexed: 02/07/2023]
|
17
|
Shi C, Wang Q, Rao Z, Shi Y, Wei S, Wang H, Lu X, Wang P, Lu L, Zhou H, Cheng F. Diabetes induces hepatocyte pyroptosis by promoting oxidative stress-mediated NLRP3 inflammasome activation during liver ischaemia and reperfusion injury. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:739. [PMID: 32647664 PMCID: PMC7333130 DOI: 10.21037/atm-20-1839] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background Although diabetes mellitus has been reported to aggravate liver ischaemia and reperfusion (IR) injury, the basic mechanism remains largely unknown. The object of the present study was to determine the role of oxidative stress and hepatocellular pyroptosis in liver IR injury in diabetic mice. Methods Db/db and C57BL/6 mice at 8 weeks of age were subjected to liver IR injury. Liver injury and hepatocyte cell death were analyzed. A NOD-like receptor family pyrin domain-containing 3 protein (NLRP3) inflammasome antagonist (CY09) and a reactive oxygen species (ROS) antagonist (N-Acetyl-L-cysteine, NAC) were used to determine the role of ROS-mediated hepatocellular pyroptosis in diabetic mice post-IR. Results Aggravated liver IR injury was found in db/db mice compared to C57BL/6 control mice, as demonstrated by increased serum alanine aminotransaminase (ALT) and aspartate aminotransaminase (AST) levels, liver architecture damage and Suzuki scores. Interestingly, IR induces the pyroptosis of hepatocytes in db/db mice, as evidenced by enhanced NLRP3 inflammasome activation, increased numbers of terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL)-positive hepatocytes and increased gene expression of interleukin-1β (IL-1β) and IL-18 in livers post-IR. The inhibitory effect of CY09, an NLRP3 antagonist, efficiently abrogated the exacerbation effects of diabetes on liver IR injury in db/db mice. Furthermore, increased ROS expression was detected in db/db mice compared to control mice after IR. ROS scavenging by NAC pretreatment markedly inhibited hepatocellular NLRP3 inflammasome activation and pyroptosis in the db/db mice post-IR, indicating that ROS play an essential role in mediating hepatocyte pyroptosis in the setting of diabetes mellitus. Conclusions Our results demonstrate that diabetes induces hepatocyte pyroptosis by promoting oxidative stress-mediated NLRP3 inflammasome activation during liver IR injury. Strategies targeting ROS and NLRP3 inflammasome activation would be beneficial for preventing liver IR injury in diabetic patients.
Collapse
Affiliation(s)
- Chengyu Shi
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, China
| | - Qi Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, China.,School of Medical, Southeast University, Nanjing, China
| | - Zhuqing Rao
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yong Shi
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, China
| | - Song Wei
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, China.,School of Medical, Southeast University, Nanjing, China
| | - Hao Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, China
| | - Xu Lu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, China
| | - Ping Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, China
| | - Ling Lu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, China.,School of Medical, Southeast University, Nanjing, China.,Department of General Surgery, People's Hospital of Qinghai Province, Xining, Qinghai, China
| | - Haoming Zhou
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, China
| | - Feng Cheng
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, China
| |
Collapse
|
18
|
Rizwan H, Pal S, Sabnam S, Pal A. High glucose augments ROS generation regulates mitochondrial dysfunction and apoptosis via stress signalling cascades in keratinocytes. Life Sci 2019; 241:117148. [PMID: 31830478 DOI: 10.1016/j.lfs.2019.117148] [Citation(s) in RCA: 189] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 01/12/2023]
Abstract
Mitochondria are fascinating structures of the cellular compartments that generate energy to run the cells. However, inherent disorders of mitochondria due to diabetes can cause major disruption of metabolism that produces huge amount of reactive oxygen species (ROS). Here we study the elevated level of ROS provoked by high glucose (HG) environment triggered mitochondrial dysfunction, inflammatory response and apoptosis via stress signalling pathway in keratinocytes. Our results demonstrated that elevated glucose level in keratinoctes, increase the accumulations of ROS and decrease in cellular antioxidant capacities. Moreover, excess production of ROS was associated with mitochondrial dysfunction, characterized by loss of mitochondrial membrane potential (ΔΨm), increase in mitochondrial mass, alteration of mitochondrial respiratory complexes, cytochrome c (Cyt c) release, decrease in mitochondrial transcription factor A (TFAM) and increase in mitochondrial DNA (mtDNA) fragmentation. Damaged mtDNA escaped into the cytosol, where it engaged the activation of ERK1/2, PI3K/Akt, tuberin and mTOR via cGAS-STING leading to IRF3 activation. Pre-treatment of pharmacological inhibitors, ERK1/2 or PI3K/Akt suppressed the IRF3 activation. Furthermore, our results demonstrated that activation of IRF3 in HG environment coinciding with increased expression of inflammatory mediators. Excess production of ROS interfered with decreased in cell viability, increased lysosomal content and expression of FoxOs, leading to cell cycle deregulation and apoptosis. Pre-treatment of N-acetyl-l-cysteine (NAC) significantly reduced the HG-induced cell cycle deregulation and apoptosis in keratinocytes. In conclusion, increased oxidative stress underlies the decrease in antioxidant capacities and mitochondrial dysfunction in HG environment correlate with inflammation response and apoptosis via ERK1/2-PI3K/Akt-IRF3 pathway in keratinoctes.
Collapse
Affiliation(s)
- Huma Rizwan
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar 751024, India
| | - Sweta Pal
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar 751024, India
| | - Silpa Sabnam
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar 751024, India
| | - Arttatrana Pal
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar 751024, India; Department of Zoology, School of Life Sciences, Mahatma Gandhi Central University, Motihari, Bihar 845401, India.
| |
Collapse
|
19
|
Mitochondrial ROS-induced lysosomal dysfunction impairs autophagic flux and contributes to M1 macrophage polarization in a diabetic condition. Clin Sci (Lond) 2019; 133:1759-1777. [PMID: 31383716 DOI: 10.1042/cs20190672] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 06/30/2019] [Accepted: 08/02/2019] [Indexed: 02/05/2023]
Abstract
Macrophage polarization toward the M1 phenotype and its subsequent inflammatory response have been implicated in the progression of diabetic complications. Despite adverse consequences of autophagy impairment on macrophage inflammation, the regulation of macrophage autophagy under hyperglycemic conditions is incompletely understood. Here, we report that the autophagy-lysosome system and mitochondrial function are impaired in streptozotocin (STZ)-induced diabetic mice and high glucose (HG)-stimulated RAW 264.7 cells. Mitochondrial dysfunction promotes reactive oxygen species (ROS) production and blocks autophagic flux by impairing lysosome function in macrophages under hyperglycemic conditions. Conversely, inhibition of mitochondrial ROS by Mito-TEMPO prevents HG-induced M1 macrophage polarization, and its effect is offset by blocking autophagic flux. The role of mitochondrial ROS in lysosome dysfunction and M1 macrophage polarization is also demonstrated in mitochondrial complex I defective RAW 264.7 cells induced by silencing NADH:ubiquinone oxidoreductase subunit-S4 (Ndufs4). These findings prove that mitochondrial ROS plays a key role in promoting macrophage polarization to inflammatory phenotype by impairing autophagy-lysosome system, which might provide clue to a novel treatment for diabetic complications.
Collapse
|
20
|
Hyperglycemia exacerbates acetaminophen-induced acute liver injury by promoting liver-resident macrophage proinflammatory response via AMPK/PI3K/AKT-mediated oxidative stress. Cell Death Discov 2019; 5:119. [PMID: 31341645 PMCID: PMC6642179 DOI: 10.1038/s41420-019-0198-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/04/2019] [Accepted: 06/11/2019] [Indexed: 02/08/2023] Open
Abstract
Although diabetes mellitus/hyperglycemia is a risk factor for acute liver injury, the underlying mechanism remains largely unknown. Liver-resident macrophages (Kupffer cells, KCs) and oxidative stress play critical roles in the pathogenesis of toxin-induced liver injury. Here, we evaluated the role of oxidative stress in regulating KC polarization against acetaminophen (APAP)-mediated acute liver injury in a streptozotocin-induced hyperglycemic murine model. Compared to the controls, hyperglycemic mice exhibited a significant increase in liver injury and intrahepatic inflammation. KCs obtained from hyperglycemic mice secreted higher levels of the proinflammatory factors, such as TNF-α and IL-6, lower levels of the anti-inflammatory factor IL-10. Furthermore, enhanced oxidative stress was revealed by increased levels of reactive oxygen species (ROS) in KCs from hyperglycemic mice post APAP treatment. In addition, ROS inhibitor NAC resulted in a significant decrease of ROS production in hyperglycemic KCs from mice posttreated with APAP. We also analyzed the role of hyperglycemia in macrophage M1/M2 polarization. Interestingly, we found that hyperglycemia promoted M1 polarization, but inhibited M2 polarization of KCs obtained from APAP-exposed livers, as evidenced by increased MCP-1 and inducible NO synthase (iNOS) gene induction but decreased Arg-1 and CD206 gene induction accompanied by increased STAT1 activation and decreased STAT6 activation. NAC restored Arg-1, CD206 gene induction, and STAT6 activation. To explore the mechanism how hyperglycemia regulates KCs polarization against APAP-induced acute liver injury, we examined the AMPK/PI3K/AKT signaling pathway and found decreased AMPK activation and increased AKT activation in liver and KCs from hyperglycemic mice post APAP treatment. AMPK activation by its agonist AICAR or PI3K inhibition by its antagonist LY294002 inhibited ROS production in KCs from hyperglycemic mice post APAP treatment and significantly attenuated APAP-induced liver injury in the hyperglycemic mice, compared to the control mice. Our results demonstrated that hyperglycemia exacerbated APAP-induced acute liver injury by promoting liver-resident macrophage proinflammatory response via AMPK/PI3K/AKT-mediated oxidative stress.
Collapse
|
21
|
Thaane T, Motala AA, Mckune AJ. Lifestyle modification in the management of insulin resistance states in overweight/obesity: the role of exercise training. JOURNAL OF ENDOCRINOLOGY, METABOLISM AND DIABETES OF SOUTH AFRICA 2019. [DOI: 10.1080/16089677.2019.1608054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Tshidi Thaane
- Discipline of Biokinetics, Exercise and Leisure Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Ayesha A Motala
- Department of Diabetes and Endocrinology, University of KwaZulu-Natal, Durban, South Africa
| | - Andrew J Mckune
- Discipline of Biokinetics, Exercise and Leisure Sciences, University of KwaZulu-Natal, Durban, South Africa
- University of Canberra Research Institute for Sport and Exercise Science, University of Canberra, Canberra, Australia
- Collaborative Research in Bioactives and Biomarkers (CRIBB) Group, University of Canberra, Canberra, Australia
| |
Collapse
|
22
|
Relevance of Erk1/2-PI3K/Akt signaling pathway in CEES-induced oxidative stress regulates inflammation and apoptosis in keratinocytes. Cell Biol Toxicol 2019; 35:541-564. [PMID: 30805762 DOI: 10.1007/s10565-019-09467-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 02/13/2019] [Indexed: 12/24/2022]
|
23
|
Tian H, Li Y, Kang P, Wang Z, Yue F, Jiao P, Yang N, Qin S, Yao S. Endoplasmic reticulum stress-dependent autophagy inhibits glycated high-density lipoprotein-induced macrophage apoptosis by inhibiting CHOP pathway. J Cell Mol Med 2019; 23:2954-2969. [PMID: 30746902 PMCID: PMC6433656 DOI: 10.1111/jcmm.14203] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/07/2018] [Accepted: 01/13/2019] [Indexed: 12/15/2022] Open
Abstract
This study was designed to explore the inductive effect of glycated high‐density lipoprotein (gly‐HDL) on endoplasmic reticulum (ER) stress‐C/EBP homologous protein (CHOP)‐mediated macrophage apoptosis and its relationship with autophagy. Our results showed that gly‐HDL caused macrophage apoptosis with concomitant activation of ER stress pathway, including nuclear translocation of activating transcription factor 6, phosphorylation of protein kinase‐like ER kinase (PERK) and eukaryotic translation initiation factor 2α, and CHOP up‐regulation, which were inhibited by 4‐phenylbutyric acid (PBA, an ER stress inhibitor) and the gene silencing of PERK and CHOP. Similar data were obtained from macrophages treated by HDL isolated from diabetic patients. Gly‐HDL induced macrophage autophagy as assessed by up‐regulation of beclin‐1, autophagy‐related gene 5 and microtubule‐associated protein one light chain 3‐II, which were depressed by PBA and PERK siRNA. Gly‐HDL‐induced apoptosis, PERK phosphorylation and CHOP up‐regulation were suppressed by rapamycin (an autophagy inducer), whereas aggravated by 3‐methyladenine (an autophagy inhibitor) and beclin‐1 siRNA. Administration of diabetic apoE−/− mice with rapamycin attenuated MOMA‐2 and CHOP up‐regulation and apoptosis in atherosclerotic lesions. These data indicate that gly‐HDL may induce macrophage apoptosis through activating ER stress‐CHOP pathway and ER stress mediates gly‐HDL‐induced autophagy, which in turn protects macrophages against apoptosis by alleviating CHOP pathway.
Collapse
Affiliation(s)
- Hua Tian
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Taishan Medical University, Taian, China
| | - Yanyan Li
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Taishan Medical University, Taian, China
| | - Panpan Kang
- Affiliated hospital of Chengde Medical University, Chengde Medical University, Chengde, China
| | - Zhichao Wang
- College of Nursing, Taishan Medical University, Taian, China
| | - Feng Yue
- Department of Endocrinology, Central Hospital of Taian, Taian, China
| | - Peng Jiao
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Taishan Medical University, Taian, China
| | - Nana Yang
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Taishan Medical University, Taian, China
| | - Shucun Qin
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Taishan Medical University, Taian, China
| | - Shutong Yao
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Taishan Medical University, Taian, China.,College of Basic Medical Sciences, Taishan Medical University, Taian, China
| |
Collapse
|
24
|
Reactive oxygen species (ROS) in macrophage activation and function in diabetes. Immunobiology 2018; 224:242-253. [PMID: 30739804 DOI: 10.1016/j.imbio.2018.11.010] [Citation(s) in RCA: 372] [Impact Index Per Article: 53.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 11/27/2018] [Accepted: 11/27/2018] [Indexed: 12/14/2022]
Abstract
In a diabetic milieu high levels of reactive oxygen species (ROS) are induced. This contributes to the vascular complications of diabetes. Recent studies have shown that ROS formation is exacerbated in diabetic monocytes and macrophages due to a glycolytic metabolic shift. Macrophages are important players in the progression of diabetes and promote inflammation through the release of pro-inflammatory cytokines and proteases. Because ROS is an important mediator for the activation of pro-inflammatory signaling pathways, obesity and hyperglycemia-induced ROS production may favor induction of M1-like pro-inflammatory macrophages during diabetes onset and progression. ROS induces MAPK, STAT1, STAT6 and NFκB signaling, and interferes with macrophage differentiation via epigenetic (re)programming. Therefore, a comprehensive understanding of the impact of ROS on macrophage phenotype and function is needed in order to improve treatment of diabetes and its vascular complications. In the current comprehensive review, we dissect the role of ROS in macrophage polarization, and analyze how ROS production links metabolism and inflammation in diabetes and its complications. Finally, we discuss the contribution of ROS to the crosstalk between macrophages and endothelial cells in diabetic complications.
Collapse
|
25
|
Yaribeygi H, Atkin SL, Pirro M, Sahebkar A. A review of the anti-inflammatory properties of antidiabetic agents providing protective effects against vascular complications in diabetes. J Cell Physiol 2018; 234:8286-8294. [PMID: 30417367 DOI: 10.1002/jcp.27699] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 10/15/2018] [Indexed: 12/18/2022]
Abstract
The global prevalence of Type 2 diabetes mellitus and its associated complications are growing rapidly. Although the role of hyperglycemia is well recognized in the pathophysiology of diabetic complications, its exact underlying mechanisms are not fully understood. In this regard, accumulating evidence suggests that the role of inflammation appears pivotal, with studies showing that most diabetic complications are associated with an inflammatory response. Several classes of antidiabetic agents have been introduced for controlling glycemia, with evidence that these pharmacological agents may have modulatory effects on inflammation beyond their glucose-lowering activity. Here we review the latest evidence on the anti-inflammatory effects of commonly used antidiabetic medications and discuss the relevance of these effects on preventing diabetic complications.
Collapse
Affiliation(s)
- Habib Yaribeygi
- Chronic Kidney Disease Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Matteo Pirro
- Department of Medicine, Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, University of Perugia, Perugia, Italy
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
26
|
Intermittent High Glucose Exacerbates A-FABP Activation and Inflammatory Response through TLR4-JNK Signaling in THP-1 Cells. J Immunol Res 2018; 2018:1319272. [PMID: 29850615 PMCID: PMC5925160 DOI: 10.1155/2018/1319272] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 02/15/2018] [Accepted: 03/19/2018] [Indexed: 01/01/2023] Open
Abstract
Background Glucose fluctuation confers additional risks on diabetes-related vascular diseases, but the underlying mechanisms are unknown. Macrophage activation mediated by TLR4-JNK signaling plays an important role during the progress of diabetes. In the present study, we hypothesize that glucose fluctuation results in macrophage inflammation through TLR4-JNK signaling pathways. Methods THP-1 cells were treated with normal glucose (5 mM), constant high glucose (25 mM), and intermittent high glucose (rotation per 6 h in 5 mM or 25 mM) for 24 h. The mRNA and protein expression levels of TLR4, p-JNK, and adipocyte fatty acid-binding protein (A-FABP) were determined, and the proinflammatory cytokines TNF-α and IL-1β were quantified. Results In constant high glucose, TLR4 expression and JNK phosphorylation levels increased, and this effect was more pronounced in intermittent high glucose. Accordingly, the expression of A-FABP and the release of the proinflammatory cytokines TNF-α and IL-1β also increased in response to constant high glucose, an effect that also was more evident in intermittent high glucose. The inhibition of p-JNK by SP600125 did not attenuate TLR4 expression, but totally inhibited both A-FABP expression and the production of the proinflammatory cytokines TNF-α and IL-1β in both constant and intermittent high glucose. Conclusions Intermittent high glucose potentiates A-FABP activation and inflammatory responses via TLR4/p-JNK signaling in THP-1 cells. These findings suggest a more detrimental impact of glucose fluctuation on macrophage inflammation in diabetes-related vascular diseases than thus far generally assumed.
Collapse
|
27
|
Rizwan H, Mohanta J, Si S, Pal A. Gold nanoparticles reduce high glucose-induced oxidative-nitrosative stress regulated inflammation and apoptosis via tuberin-mTOR/NF-κB pathways in macrophages. Int J Nanomedicine 2017; 12:5841-5862. [PMID: 28860752 PMCID: PMC5566318 DOI: 10.2147/ijn.s141839] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Hyperglycemia is a risk factor for cardiovascular mortality and morbidity, and directly responsible for exacerbating macrophage activation and atherosclerosis. We showed that gold nanoparticles (AuNPs) reduce the high glucose (HG)-induced atherosclerosis-related complications in macrophages via oxidative-nitrosative stress-regulated inflammation and apoptosis. The effects of AuNPs on oxidative-nitrosative stress markers such as cellular antioxidants were attenuated by HG exposure, leading to reduction in the accumulation of reactive oxygen/nitrogen species in cellular compartments. Further, these abnormalities of antioxidants level and reactive oxygen/nitrogen species accumulations initiate cellular stress, resulting in the activation of nuclear factor κB (NF-κB) via ERK1/2mitogen-activated protein kinase (MAPK)/Akt/tuberin-mammalian target of rapamycin (mTOR) pathways. The activated NF-κB stimulates inflammatory mediators, which subsequently subdue biomolecules damage, leading to aggravation of the inflammatory infiltration and immune responses. Treatment of AuNPs inhibits the intracellular redox-sensitive signaling pathways, inflammation, and apoptosis in macrophages. Together, our results indicate that AuNPs may modulate HG-induced oxidative-nitrosative stress. These effects may be sealed tight due to the fact that AuNPs treatment reduces the activation of NF-κB by ERK1/2MAPK/Akt/tuberin-mTOR pathways-mediated inflammatory genes expression and cellular stress responses, which may be beneficial for minimizing the atherosclerosis.
Collapse
Affiliation(s)
- Huma Rizwan
- School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Jagdeep Mohanta
- School of Applied Sciences, KIIT University, Bhubaneswar, India
| | - Satyabrata Si
- School of Biotechnology, KIIT University, Bhubaneswar, India.,School of Applied Sciences, KIIT University, Bhubaneswar, India
| | - Arttatrana Pal
- Department of Zoology, School of Life Sciences, Mahatma Gandhi Central University, Bihar, India
| |
Collapse
|
28
|
Huang X, Xie H, Xue G, Ye M, Zhang L. MiR-3202 - Promoted H5V Cell Apoptosis by Directly Targeting Fas Apoptotic Inhibitory Molecule 2 (FAIM2) in High Glucose Condition. Med Sci Monit 2017; 23:975-983. [PMID: 28228635 PMCID: PMC5335645 DOI: 10.12659/msm.899443] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Background Vascular complications are a major concern for patients with diabetes. Endothelial cells (ECs) play a key role in vascular function. MicroRNAs (miRNAs) have been shown to play an important role in mediating EC function; miRNAs are vulnerable to hyperglycemic conditions. Previous reports verified that Fas apoptotic inhibitory molecule 2 (FAIM2) can inhibit cell apoptosis through repressing the FAS-associated death domain protein (FADD) pathway. This current study was designed to explore the potential involvement of miR-3202 in the pathogenesis of ECs in high-glucose conditions. Material/Methods The aim of this study was to investigate the role of miR-3202 in regulating hyperglycemia-induced ECs by targeting FAIM2. The endothelial cell line H5V was cultured in a high-glucose condition to induce damage to FAIM2 expression in ECs; mimic and inhibition of miR-3202 were used to enhance and depress miR-3202’s function to explore its function on FAIM2. Results Our study showed that FAIM2 was inhibited by high-glucose conditions, and miRNA-3202 was induced by high-glucose conditions. FAIM2 was identified as the target gene of miRNA-3202; luciferase reporter assays confirmed that FAIM2 was downregulated by miR-3202 directly, that is, miR-3202 can upregulate Fas/FADD through inhibiting FAIM2. Conclusions MiR-3202 can promote EC apoptosis in hyperglycemic conditions, which demonstrated that EC apoptosis induced by high-glucose conditions partly depends on miR-3202 targeting FAIM2.
Collapse
Affiliation(s)
- Xiaozhong Huang
- Department of Vascular Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (mainland)
| | - Hui Xie
- Department of Vascular Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (mainland)
| | - Guanhua Xue
- Department of Vascular Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (mainland)
| | - Meng Ye
- Department of Vascular Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (mainland)
| | - Lan Zhang
- Department of Vascular Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (mainland)
| |
Collapse
|
29
|
Sagar S, Parida SR, Sabnam S, Rizwan H, Pal S, Swain MM, Pal A. Increasing NO level regulates apoptosis and inflammation in macrophages after 2-chloroethyl ethyl sulphide challenge. Int J Biochem Cell Biol 2017; 83:1-14. [DOI: 10.1016/j.biocel.2016.12.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 11/23/2016] [Accepted: 12/08/2016] [Indexed: 12/19/2022]
|
30
|
Bulavintseva TS, Danilova IG, Briliant SA, Smirnyh SE, Abidov MT. The response of the mononuclear phagocyte system to chronic hyperglycemia. Biophysics (Nagoya-shi) 2016. [DOI: 10.1134/s0006350916050031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
31
|
Sałat K, Gdula-Argasińska J, Malikowska N, Podkowa A, Lipkowska A, Librowski T. Effect of pregabalin on contextual memory deficits and inflammatory state-related protein expression in streptozotocin-induced diabetic mice. Naunyn Schmiedebergs Arch Pharmacol 2016; 389:613-23. [PMID: 26984821 PMCID: PMC4866991 DOI: 10.1007/s00210-016-1230-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 03/07/2016] [Indexed: 01/02/2023]
Abstract
Diabetes mellitus is a metabolic disease characterized by hyperglycemia due to defects in insulin secretion or its action. Complications from long-term diabetes consist of numerous biochemical, molecular, and functional tissue alterations, including inflammation, oxidative stress, and neuropathic pain. There is also a link between diabetes mellitus and vascular dementia or Alzheimer’s disease. Hence, it is important to treat diabetic complications using drugs which do not aggravate symptoms induced by the disease itself. Pregabalin is widely used for the treatment of diabetic neuropathic pain, but little is known about its impact on cognition or inflammation-related proteins in diabetic patients. Thus, this study aimed to evaluate the effect of intraperitoneal (ip) pregabalin on contextual memory and the expression of inflammatory state-related proteins in the brains of diabetic, streptozotocin (STZ)-treated mice. STZ (200 mg/kg, ip) was used to induce diabetes mellitus. To assess the impact of pregabalin (10 mg/kg) on contextual memory, a passive avoidance task was applied. Locomotor and exploratory activities in pregabalin-treated diabetic mice were assessed by using activity cages. Using Western blot analysis, the expression of cyclooxygenase-2 (COX-2), cytosolic prostaglandin E synthase (cPGES), nuclear factor (erythroid-derived 2)-like 2 (Nrf2), nuclear factor-ĸB (NF-ĸB) p50 and p65, aryl hydrocarbon receptor (AhR), as well as glucose transporter type-4 (GLUT4) was assessed in mouse brains after pregabalin treatment. Pregabalin did not aggravate STZ-induced learning deficits in vivo or influence animals’ locomotor activity. We observed significantly lower expression of COX-2, cPGES, and NF-κB p50 subunit, and higher expression of AhR and Nrf2 in the brains of pregabalin-treated mice in comparison to STZ-treated controls, which suggested immunomodulatory and anti-inflammatory effects of pregabalin. Antioxidant properties of pregabalin in the brains of diabetic animals were also demonstrated. Pregabalin does not potentiate STZ-induced cognitive decline, and it has antioxidant, immunomodulatory, and anti-inflammatory properties in mice. These results confirm the validity of its use in diabetic patients. Effect of pregabalin on fear-motivated memory and markers of brain tissue inflammation in diabetic mice ![]()
Collapse
Affiliation(s)
- Kinga Sałat
- Faculty of Pharmacy, Department of Pharmacodynamics, Jagiellonian University Medical College, 9 Medyczna St, 30-688, Krakow, Poland.
| | - Joanna Gdula-Argasińska
- Faculty of Pharmacy, Department of Radioligands, Jagiellonian University Medical College, 9 Medyczna St, 30-688, Krakow, Poland
| | - Natalia Malikowska
- Faculty of Pharmacy, Department of Pharmacodynamics, Jagiellonian University Medical College, 9 Medyczna St, 30-688, Krakow, Poland
| | - Adrian Podkowa
- Faculty of Pharmacy, Department of Pharmacodynamics, Jagiellonian University Medical College, 9 Medyczna St, 30-688, Krakow, Poland
| | - Anna Lipkowska
- Faculty of Pharmacy, Department of Radioligands, Jagiellonian University Medical College, 9 Medyczna St, 30-688, Krakow, Poland
| | - Tadeusz Librowski
- Faculty of Pharmacy, Department of Radioligands, Jagiellonian University Medical College, 9 Medyczna St, 30-688, Krakow, Poland
| |
Collapse
|
32
|
Maiese K. Erythropoietin and mTOR: A "One-Two Punch" for Aging-Related Disorders Accompanied by Enhanced Life Expectancy. Curr Neurovasc Res 2016; 13:329-340. [PMID: 27488211 PMCID: PMC5079807 DOI: 10.2174/1567202613666160729164900] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 07/12/2016] [Accepted: 07/14/2016] [Indexed: 12/16/2022]
Abstract
Life expectancy continues to increase throughout the world, but is accompanied by a rise in the incidence of non-communicable diseases. As a result, the benefits of an increased lifespan can be limited by aging-related disorders that necessitate new directives for the development of effective and safe treatment modalities. With this objective, the mechanistic target of rapamycin (mTOR), a 289-kDa serine/threonine protein, and its related pathways of mTOR Complex 1 (mTORC1), mTOR Complex 2 (mTORC2), proline rich Akt substrate 40 kDa (PRAS40), AMP activated protein kinase (AMPK), Wnt signaling, and silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), have generated significant excitement for furthering novel therapies applicable to multiple systems of the body. Yet, the biological and clinical outcome of these pathways can be complex especially with oversight of cell death mechanisms that involve apoptosis and autophagy. Growth factors, and in particular erythropoietin (EPO), are one avenue under consideration to implement control over cell death pathways since EPO can offer potential treatment for multiple disease entities and is intimately dependent upon mTOR signaling. In experimental and clinical studies, EPO appears to have significant efficacy in treating several disorders including those involving the developing brain. However, in mature populations that are affected by aging-related disorders, the direction for the use of EPO to treat clinical disease is less clear that may be dependent upon a number of factors including the understanding of mTOR signaling. Continued focus upon the regulatory elements that control EPO and mTOR signaling could generate critical insights for targeting a broad range of clinical maladies.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, Newark, New Jersey 07101, USA.
| |
Collapse
|