1
|
Kaempfer R. Subduing the Inflammatory Cytokine Storm. Int J Mol Sci 2024; 25:11194. [PMID: 39456976 PMCID: PMC11508503 DOI: 10.3390/ijms252011194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/10/2024] [Accepted: 10/13/2024] [Indexed: 10/28/2024] Open
Abstract
The inflammatory cytokine response is essential for protective immunity, yet bacterial and viral pathogens often elicit an exaggerated response ("cytokine storm") harmful to the host that can cause multi-organ damage and lethality. Much has been published recently on the cytokine storm within the context of the coronavirus pandemic, yet bacterial sepsis, severe wound infections and toxic shock provide other prominent examples. The problem of the cytokine storm is compounded by the increasing incidence of multidrug-resistant bacterial strains. We created an incisive molecular tool for analyzing the role of the B7/CD28 costimulatory axis in the human inflammatory response. To attenuate the cytokine storm underlying infection pathology, yet preserve host defenses, we uniquely targeted the engagement of CD28 with its B7 co-ligands by means of short peptide mimetics of the human CD28 and B7 receptor homodimer interfaces. These peptides are not only effective tools for dissecting mechanism but also serve to attenuate the inflammatory response as a broad host-oriented therapeutic strategy against the cytokine storm. Indeed, such peptides protect mice from lethal Gram-positive bacterial superantigen-induced toxic shock even when dosed in molar amounts well below that of the superantigen and show promise in protecting humans from the severe inflammatory disease necrotizing soft tissue infections ('flesh-eating' bacterial sepsis) following traumatic wound injuries.
Collapse
Affiliation(s)
- Raymond Kaempfer
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| |
Collapse
|
2
|
Smith A, Dobinda K, Chen S, Zieba P, Paunesku T, Sun Z, Woloschak GE. X-ray Fluorescence Microscopy to Develop Elemental Classifiers and Investigate Elemental Signatures in BALB/c Mouse Intestine a Week after Exposure to 8 Gy of Gamma Rays. Int J Mol Sci 2024; 25:10256. [PMID: 39408586 PMCID: PMC11477073 DOI: 10.3390/ijms251910256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/17/2024] [Accepted: 09/17/2024] [Indexed: 10/20/2024] Open
Abstract
Iron redistribution in the intestine after total body irradiation is an established phenomenon. However, in the literature, there are no reports about the use of X-ray fluorescence microscopy or equivalent techniques to generate semi-quantitative 2D maps of iron in sectioned intestine samples from irradiated mice. In this work, we used X-ray fluorescence microscopy (XFM) to map the elemental content of iron as well as phosphorus, sulfur, calcium, copper and zinc in tissue sections of the small intestine from eight-week-old BALB/c male mice that developed gastrointestinal acute radiation syndrome (GI-ARS) in response to exposure to 8 Gray of gamma rays. Seven days after irradiation, we found that the majority of the iron is localized as hot spots in the intercellular regions of the area surrounding crypts and stretching between the outer perimeter of the intestine and the surface cell layer of villi. In addition, this study represents our current efforts to develop elemental cell classifiers that could be used for the automated generation of regions of interest for analyses of X-ray fluorescence maps. Once developed, such a tool will be instrumental for studies of effects of radiation and other toxicants on the elemental content in cells and tissues. While XFM studies cannot be conducted on living organisms, it is possible to envision future scenarios where XFM imaging of single cells sloughed from the human (or rodent) intestine could be used to follow up on the progression of GI-ARS.
Collapse
Affiliation(s)
- Anthony Smith
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Katrina Dobinda
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Si Chen
- X-ray Imaging Division, Advanced Photon Source, Argonne National Laboratory, Argonne, IL 60439, USA
| | - Peter Zieba
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Tatjana Paunesku
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Zequn Sun
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Gayle E. Woloschak
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
3
|
Bucataru C, Ciobanasu C. Antimicrobial peptides: Opportunities and challenges in overcoming resistance. Microbiol Res 2024; 286:127822. [PMID: 38986182 DOI: 10.1016/j.micres.2024.127822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/12/2024]
Abstract
Antibiotic resistance represents a global health threat, challenging the efficacy of traditional antimicrobial agents and necessitating innovative approaches to combat infectious diseases. Among these alternatives, antimicrobial peptides have emerged as promising candidates against resistant pathogens. Unlike traditional antibiotics with only one target, these peptides can use different mechanisms to destroy bacteria, with low toxicity to mammalian cells compared to many conventional antibiotics. Antimicrobial peptides (AMPs) have encouraging antibacterial properties and are currently employed in the clinical treatment of pathogen infection, cancer, wound healing, cosmetics, or biotechnology. This review summarizes the mechanisms of antimicrobial peptides against bacteria, discusses the mechanisms of drug resistance, the limitations and challenges of AMPs in peptide drug applications for combating drug-resistant bacterial infections, and strategies to enhance their capabilities.
Collapse
Affiliation(s)
- Cezara Bucataru
- Alexandru I. Cuza University, Institute of Interdisciplinary Research, Department of Exact and Natural Sciences, Bulevardul Carol I, Nr.11, Iasi 700506, Romania
| | - Corina Ciobanasu
- Alexandru I. Cuza University, Institute of Interdisciplinary Research, Department of Exact and Natural Sciences, Bulevardul Carol I, Nr.11, Iasi 700506, Romania.
| |
Collapse
|
4
|
Liu GY, Yu D, Fan MM, Zhang X, Jin ZY, Tang C, Liu XF. Antimicrobial resistance crisis: could artificial intelligence be the solution? Mil Med Res 2024; 11:7. [PMID: 38254241 PMCID: PMC10804841 DOI: 10.1186/s40779-024-00510-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Antimicrobial resistance is a global public health threat, and the World Health Organization (WHO) has announced a priority list of the most threatening pathogens against which novel antibiotics need to be developed. The discovery and introduction of novel antibiotics are time-consuming and expensive. According to WHO's report of antibacterial agents in clinical development, only 18 novel antibiotics have been approved since 2014. Therefore, novel antibiotics are critically needed. Artificial intelligence (AI) has been rapidly applied to drug development since its recent technical breakthrough and has dramatically improved the efficiency of the discovery of novel antibiotics. Here, we first summarized recently marketed novel antibiotics, and antibiotic candidates in clinical development. In addition, we systematically reviewed the involvement of AI in antibacterial drug development and utilization, including small molecules, antimicrobial peptides, phage therapy, essential oils, as well as resistance mechanism prediction, and antibiotic stewardship.
Collapse
Affiliation(s)
- Guang-Yu Liu
- Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Hangzhou Normal University, Key Laboratory of Aging and Cancer Biology of Zhejiang Province, Key Laboratory of Inflammation and Immunoregulation of Hangzhou, Hangzhou Normal University, Hangzhou, 311121, China
| | - Dan Yu
- National Key Discipline of Pediatrics Key Laboratory of Major Diseases in Children Ministry of Education, Laboratory of Dermatology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Mei-Mei Fan
- Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Hangzhou Normal University, Key Laboratory of Aging and Cancer Biology of Zhejiang Province, Key Laboratory of Inflammation and Immunoregulation of Hangzhou, Hangzhou Normal University, Hangzhou, 311121, China
| | - Xu Zhang
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Ze-Yu Jin
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Christoph Tang
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK.
| | - Xiao-Fen Liu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission of the People's Republic of China, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
5
|
Popugailo A, Rotfogel Z, Levy M, Turgeman O, Hillman D, Levy R, Arad G, Shpilka T, Kaempfer R. The homodimer interfaces of costimulatory receptors B7 and CD28 control their engagement and pro-inflammatory signaling. J Biomed Sci 2023; 30:49. [PMID: 37381064 DOI: 10.1186/s12929-023-00941-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 06/19/2023] [Indexed: 06/30/2023] Open
Abstract
BACKGROUND The inflammatory response is indispensable for protective immunity, yet microbial pathogens often trigger an excessive response, 'cytokine storm', harmful to the host. Full T-cell activation requires interaction of costimulatory receptors B7-1(CD80) and B7-2(CD86) expressed on antigen-presenting cells with CD28 expressed on the T cells. We created short peptide mimetics of the homodimer interfaces of the B7 and CD28 receptors and examined their ability to attenuate B7/CD28 coligand engagement and signaling through CD28 for inflammatory cytokine induction in human immune cells, and to protect from lethal toxic shock in vivo. METHODS Short B7 and CD28 receptor dimer interface mimetic peptides were synthesized and tested for their ability to attenuate the inflammatory cytokine response of human peripheral blood mononuclear cells, as well as for their ability to attenuate B7/CD28 intercellular receptor engagement. Mice were used to test the ability of such peptides to protect from lethal superantigen toxin challenge when administered in molar doses far below the toxin dose. RESULTS B7 and CD28 homodimer interfaces are remote from the coligand binding sites, yet our finding is that by binding back into the receptor dimer interfaces, short dimer interface mimetic peptides inhibit intercellular B7-2/CD28 as well as the tighter B7-1/CD28 engagement, attenuating thereby pro-inflammatory signaling. B7 mimetic peptides exhibit tight selectivity for the cognate receptor in inhibiting intercellular receptor engagement with CD28, yet each diminishes signaling through CD28. In a prominent example of inflammatory cytokine storm, by attenuating formation of the B7/CD28 costimulatory axis, B7-1 and CD28 dimer interface mimetic peptides protect mice from lethal toxic shock induced by a bacterial superantigen even when administered in doses far submolar to the superantigen. CONCLUSIONS Our results reveal that the B7 and CD28 homodimer interfaces each control B7/CD28 costimulatory receptor engagement and highlight the protective potential against cytokine storm of attenuating, yet not ablating, pro-inflammatory signaling via these receptor domains.
Collapse
Affiliation(s)
- Andrey Popugailo
- Department of Biochemistry and Molecular Biology, Institute of Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, 9112102, Jerusalem, Israel
| | - Ziv Rotfogel
- Department of Biochemistry and Molecular Biology, Institute of Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, 9112102, Jerusalem, Israel
| | - Michal Levy
- Department of Biochemistry and Molecular Biology, Institute of Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, 9112102, Jerusalem, Israel
| | - Orli Turgeman
- Department of Biochemistry and Molecular Biology, Institute of Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, 9112102, Jerusalem, Israel
| | - Dalia Hillman
- Department of Biochemistry and Molecular Biology, Institute of Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, 9112102, Jerusalem, Israel
| | - Revital Levy
- Department of Biochemistry and Molecular Biology, Institute of Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, 9112102, Jerusalem, Israel
| | - Gila Arad
- Department of Biochemistry and Molecular Biology, Institute of Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, 9112102, Jerusalem, Israel
| | - Tomer Shpilka
- Department of Biochemistry and Molecular Biology, Institute of Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, 9112102, Jerusalem, Israel
| | - Raymond Kaempfer
- Department of Biochemistry and Molecular Biology, Institute of Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, 9112102, Jerusalem, Israel.
| |
Collapse
|
6
|
Abstract
INTRODUCTION Acute kidney injury (AKI) is a clinically critical disease exhibiting an acute decline in renal function. The lack of an effective prevention and treatment method equates to a high morbidity and mortality rate. Consequently, over the past few decades, many therapeutic drugs with different mechanisms of action have been proposed and gradually applied to the clinic. The involved drug mechanisms evaluated have included hemodynamic modulation, anti-inflammatory, antioxidant, repair agents, metabolic derangement and mitochondrial function. AREAS COVERED The authors of this review provide the reader with a reference point for the latest advances in pharmacotherapy in acute kidney injury. This is achieved by the evaluation of the latest data collected on potential therapeutic drugs with different mechanisms of action, as well as their preclinical and clinical impact on AKI. EXPERT OPINION Presently, the vast majority of drugs are still in clinical development, which is a huge challenge. Nevertheless, in addition to current chemical drugs and gene therapy strategies, the advent of mesenchymal stem cell treatments and other emerging pharmaceutical strategies could enable clinicians to better treat AKI. Due to the nonselective distribution and low bioavailability of some of the latest pharmaceutical strategies, there is hope that these treatment options may provide more efficacious avenues.
Collapse
Affiliation(s)
- Yali Xu
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ping Zou
- Department of Pharmacy, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaojing Cao
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
7
|
Chen G, Han Y, Zhang H, Tu W, Zhang S. Radiotherapy-Induced Digestive Injury: Diagnosis, Treatment and Mechanisms. Front Oncol 2021; 11:757973. [PMID: 34804953 PMCID: PMC8604098 DOI: 10.3389/fonc.2021.757973] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 10/20/2021] [Indexed: 12/12/2022] Open
Abstract
Radiotherapy is one of the main therapeutic methods for treating cancer. The digestive system consists of the gastrointestinal tract and the accessory organs of digestion (the tongue, salivary glands, pancreas, liver and gallbladder). The digestive system is easily impaired during radiotherapy, especially in thoracic and abdominal radiotherapy. In this review, we introduce the physical classification, basic pathogenesis, clinical characteristics, predictive/diagnostic factors, and possible treatment targets of radiotherapy-induced digestive injury. Radiotherapy-induced digestive injury complies with the dose-volume effect and has a radiation-based organ correlation. Computed tomography (CT), MRI (magnetic resonance imaging), ultrasound (US) and endoscopy can help diagnose and evaluate the radiation-induced lesion level. The latest treatment approaches include improvement in radiotherapy (such as shielding, hydrogel spacers and dose distribution), stem cell transplantation and drug administration. Gut microbiota modulation may become a novel approach to relieving radiogenic gastrointestinal syndrome. Finally, we summarized the possible mechanisms involved in treatment, but they remain varied. Radionuclide-labeled targeting molecules (RLTMs) are promising for more precise radiotherapy. These advances contribute to our understanding of the assessment and treatment of radiation-induced digestive injury.
Collapse
Affiliation(s)
- Guangxia Chen
- Department of Gastroenterology, The First People's Hospital of Xuzhou, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, China
| | - Yi Han
- Department of Gastroenterology, The First People's Hospital of Xuzhou, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, China
| | - Haihan Zhang
- Department of Gastroenterology, The First People's Hospital of Xuzhou, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, China
| | - Wenling Tu
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
| | - Shuyu Zhang
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China.,West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
8
|
Nakamura N. A hypothesis: radiation carcinogenesis may result from tissue injuries and subsequent recovery processes which can act as tumor promoters and lead to an earlier onset of cancer. Br J Radiol 2020; 93:20190843. [PMID: 31860335 PMCID: PMC8519633 DOI: 10.1259/bjr.20190843] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Cancer risks from radiation can be observed as an increase in mortality when compared to a control group. However, it is unknown if this increased risk results from the induction of cancer or from an earlier onset of cancer. In mouse studies, it has been repeatedly shown that after an irradiation, the survival curve is shifted toward lower ages, but remains parallel to the control curve, and the extent of the shift in time to lower ages is dose-dependent. This shift is not satisfactorily explained by the induction model which assumes that cancers in the exposed group consist of spontaneous and induced events. Consequently, it seems that this shift could be interpreted to mean that all animals in the exposed group had suffered from life shortening. Under this scenario, however, it turns out that the radiation effects can no longer be interpreted as the result of oncogenic mutations, because these effects would have to involve all tumors, and the effectiveness of radiation changes with the dose. This leads to the speculation that radiation exposures induce a broad range of tissue injuries, and that these injuries are subsequently subjected to longlasting systemic recovery processes which act as promoters for tumor cells. In other words, potential cancer stem cells which were located in the irradiated field can escape oncogenic damage but undergo stimulation later in life toward the development of malignancy from radiation-induced activated microenvironment. This is an unusual form of the non-targeted or bystander effects of radiation. It is worth noting that this model suggests that there could be a path or paths which could be used to intervene in the process of post-exposure carcinogenesis, and that cancer risks at low doses could be described as days or weeks of life lost.
Collapse
Affiliation(s)
- Nori Nakamura
- Dept. of Molecular Biosciences, Radiation Effects Research Foundation, Hiroshima city, Japan
| |
Collapse
|
9
|
Edgar R, Tarrio ML, Maislin G, Chiguang F, Kaempfer R, Cross A, Opal SM, Shirvan A. Treatment with One Dose of Reltecimod is Superior to Two Doses in Mouse Models of Lethal Infection. Int J Pept Res Ther 2019. [DOI: 10.1007/s10989-019-09974-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
10
|
CD28 Deficiency Ameliorates Blast Exposure-Induced Lung Inflammation, Oxidative Stress, Apoptosis, and T Cell Accumulation in the Lungs via the PI3K/Akt/FoxO1 Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4848560. [PMID: 31565151 PMCID: PMC6745179 DOI: 10.1155/2019/4848560] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 05/14/2019] [Accepted: 05/22/2019] [Indexed: 02/07/2023]
Abstract
Although CD28 is associated with the expression of inflammatory mediators, apoptosis-related protein, immunosuppression, and tumorigenesis, the effects of CD28 deficiency on blast exposure-induced lung injury have not been investigated. In this study, we have explored the effects of CD28 on blast exposure-induced lung injury and studied its potential molecular mechanisms. A mouse model of blast exposure-induced acute lung injury was established. Sixty C57BL/6 wild-type (WT) and CD28 knockout (CD28−/−) mice were randomly divided into control or model groups. Lung tissue samples were collected 24 h and 48 h after blast injury. Histopathological changes and the expressions of inflammatory-related proteins were detected by hematoxylin-eosin, immunohistochemistry, and immunofluorescence staining. Apoptosis and oxidative stress were evaluated by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining and reactive oxygen species (ROS). Inflammation, apoptosis, oxidative stress, and related pathway protein expression were studied by western blotting. In addition, the levels of CD3 and CD28 proteins were measured by flow cytometry. In the current study, we found that CD28 deficiency significantly inhibited blast exposure-induced increases in the lung weight/body weight ratio and wet weight/dry weight ratio; decreased the infiltration of CD44+ leukocytes, CD163+ macrophages, and CD3+ T cells into the lungs; reduced the expressions of proinflammatory cytokines including IL-1β, TNF-α, and IL-6; and markedly increased IL-10 expression. CD28 deficiency also significantly attenuated blast exposure-induced ROS, MDA5, and IREα expressions; increased SOD-1 expression; lowered the number of apoptotic cells and Bax, Caspase-3, and active Caspase-8 expressions; and increased Bcl-2 expression. Additionally, CD28 deficiency significantly ameliorated blast exposure-induced increases of p-PI3K and p-Akt and ameliorated the decrease in the p-FoxO1 expression. Our results suggest that CD28 deficiency has a protective effect on blast exposure-induced lung injury, which might be associated with the PI3K/Akt/FoxO1 signaling pathway.
Collapse
|
11
|
Affiliation(s)
- Hye Been Koo
- Department of ChemistrySchool of Physics and Chemistry, Gwangju Institute of Science and Technology Gwangju Republic of Korea
| | - Jiwon Seo
- Department of ChemistrySchool of Physics and Chemistry, Gwangju Institute of Science and Technology Gwangju Republic of Korea
| |
Collapse
|
12
|
Cong P, Tong C, Liu Y, Shi L, Shi X, Zhao Y, Xiao K, Jin H, Liu Y, Hou M. CD28 Deficiency Ameliorates Thoracic Blast Exposure-Induced Oxidative Stress and Apoptosis in the Brain through the PI3K/Nrf2/Keap1 Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8460290. [PMID: 31885821 PMCID: PMC6915017 DOI: 10.1155/2019/8460290] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/25/2019] [Accepted: 10/24/2019] [Indexed: 12/22/2022]
Abstract
Blast exposure is a worldwide public health concern, but most related research has been focused on direct injury. Thoracic blast exposure-induced neurotrauma is a type of indirect injuries where research is lacking. As CD28 stimulates T cell activation and survival and contributes to inflammation initiation, it may play a role in thoracic blast exposure-induced neurotrauma. However, it has not been investigated. To explore the effects of CD28 on thoracic blast exposure-induced brain injury and its potential molecular mechanisms, a mouse model of thoracic blast exposure-induced brain injury was established. Fifty C57BL/6 wild-type (WT) and fifty CD28 knockout (CD28-/-) mice were randomly divided into five groups (one control group and four model groups), with ten mice (from each of the two models) for each group. Lung and brain tissue and serum samples were collected at 12 h, 24 h, 48 h, and 1 week after thoracic blast exposure. Histopathological changes were detected by hematoxylin-eosin staining. The expressions of inflammatory-related factors were detected by ELISA. Oxidative stress in the brain tissue was evaluated by determining the generation of reactive oxygen species (ROS) and the expressions of thioredoxin (TRX), malondialdehyde (MDA), SOD-1, and SOD-2. Apoptosis in the brain tissue was evaluated by TUNEL staining and the levels of Bax, Bcl-xL, Bad, Cytochrome C, and caspase-3. In addition, proteins of related pathways were also studied by western blotting and immunofluorescence. We found that CD28 deficiency significantly reduced thoracic blast exposure-induced histopathological changes and decreased the levels of inflammatory-related factors, including IL-1β, TNF-α, and S100β. In the brain tissue, CD28 deficiency also significantly attenuated thoracic blast exposure-induced generation of ROS and expressions of MDA, TRX, SOD-1, and SOD-2; lowered the number of apoptotic cells and the expression of Bax, cleaved caspase-3, Cytochrome C, and Bad; and maintained Bcl-xL expression. Additionally, CD28 deficiency significantly ameliorated thoracic blast exposure-induced increases of p-PI3K and Keap1 and the decrease of Nrf2 expression in the brain. Our results indicate that CD28 deficiency has a protective effect on thoracic blast exposure-induced brain injury that might be associated with the PI3K/Nrf2/Keap1 signaling pathway.
Collapse
Affiliation(s)
- Peifang Cong
- 1College of Medicine and Biological Information Engineering, Northeastern University, No. 195, Chuangxin Road, Hunnan District, Shenyang l10016, China
- 2Emergency Medicine Department of General Hospital of Northern theater command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, No. 83, Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Changci Tong
- 2Emergency Medicine Department of General Hospital of Northern theater command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, No. 83, Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Ying Liu
- 2Emergency Medicine Department of General Hospital of Northern theater command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, No. 83, Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Lin Shi
- 2Emergency Medicine Department of General Hospital of Northern theater command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, No. 83, Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Xiuyun Shi
- 2Emergency Medicine Department of General Hospital of Northern theater command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, No. 83, Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Yan Zhao
- 3Institute of Metal Research, Chinese Academy of Sciences, No. 72, Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Keshen Xiao
- 3Institute of Metal Research, Chinese Academy of Sciences, No. 72, Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Hongxu Jin
- 2Emergency Medicine Department of General Hospital of Northern theater command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, No. 83, Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Yunen Liu
- 2Emergency Medicine Department of General Hospital of Northern theater command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, No. 83, Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Mingxiao Hou
- 1College of Medicine and Biological Information Engineering, Northeastern University, No. 195, Chuangxin Road, Hunnan District, Shenyang l10016, China
| |
Collapse
|
13
|
Esensten JH, Helou YA, Chopra G, Weiss A, Bluestone JA. CD28 Costimulation: From Mechanism to Therapy. Immunity 2016; 44:973-88. [PMID: 27192564 PMCID: PMC4932896 DOI: 10.1016/j.immuni.2016.04.020] [Citation(s) in RCA: 627] [Impact Index Per Article: 69.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Indexed: 02/07/2023]
Abstract
Ligation of the CD28 receptor on T cells provides a critical second signal alongside T cell receptor (TCR) ligation for naive T cell activation. Here, we discuss the expression, structure, and biochemistry of CD28 and its ligands. CD28 signals play a key role in many T cell processes, including cytoskeletal remodeling, production of cytokines, survival, and differentiation. CD28 ligation leads to unique epigenetic, transcriptional, and post-translational changes in T cells that cannot be recapitulated by TCR ligation alone. We discuss the function of CD28 and its ligands in both effector and regulatory T cells. CD28 is critical for regulatory T cell survival and the maintenance of immune homeostasis. We outline the roles that CD28 and its family members play in human disease and we review the clinical efficacy of drugs that block CD28 ligands. Despite the centrality of CD28 and its family members and ligands to immune function, many aspects of CD28 biology remain unclear. Translation of a basic understanding of CD28 function into immunomodulatory therapeutics has been uneven, with both successes and failures. Such real-world results might stem from multiple factors, including complex receptor-ligand interactions among CD28 family members, differences between the mouse and human CD28 families, and cell-type specific roles of CD28 family members.
Collapse
Affiliation(s)
- Jonathan H Esensten
- Department of Laboratory Medicine, University of California, San Francisco, CA 94143, USA.
| | - Ynes A Helou
- Division of Rheumatology, Department of Medicine, Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center, University of California, San Francisco, CA 94143, USA
| | - Gaurav Chopra
- Department of Chemistry, Purdue Center for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA
| | - Arthur Weiss
- Division of Rheumatology, Department of Medicine, Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center, University of California, San Francisco, CA 94143, USA; Howard Hughes Medical Institute, University of California, San Francisco, CA 94143, USA
| | - Jeffrey A Bluestone
- Diabetes Center and Department of Medicine, University of California, San Francisco, CA 94143, USA.
| |
Collapse
|
14
|
Wang H, Sethi G, Loke WK, Sim MK. Des-Aspartate-Angiotensin I Attenuates Mortality of Mice Exposed to Gamma Radiation via a Novel Mechanism of Action. PLoS One 2015; 10:e0138009. [PMID: 26378927 PMCID: PMC4574738 DOI: 10.1371/journal.pone.0138009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 08/24/2015] [Indexed: 01/09/2023] Open
Abstract
ACE inhibitors and ARBs (angiotensin receptor blockers) have been shown to attenuate radiation injuries in animal models of lethal gamma irradiation. These two classes of drug act by curtailing the actions of angiotensin II-linked inflammatory pathways that are up-regulated during gamma radiation in organ systems such as the brain, lung, kidney, and bone marrow. ACE inhibitors inhibit ACE and attenuate the formation of angiotensin II from angiotensin I; ARBs block the angiotensin AT1 receptor and attenuate the actions of angiotensin II that are elicited through the receptor. DAA-I (des-aspartate-angiotensin I), an orally active angiotensin peptide, also attenuates the deleterious actions of angiotensin II. It acts as an agonist on the angiotensin AT1 receptor and elicits responses that oppose those of angiotensn II. Thus, DAA-I was investigated for its anticipated radioprotection in gamma irradiated mice. DAA-I administered orally at 800 nmole/kg/day for 30 days post exposure (6.4 Gy) attenuated the death of mice during the 30-day period. The attenuation was blocked by losartan (50 nmole/kg/day, i.p.) that was administered sequential to DAA-I administration. This shows that the radioprotection was mediated via the angiotensin AT1 receptor. Furthermore, the radioprotection correlated to an increase in circulating PGE2 of surviving animals, and this suggests that PGE2 is involved in the radioprotection in DAA-I-treated mice. At the hematopoietic level, DAA-I significantly improved two syndromes of myelosuppression (leucopenia and lymphocytopenia), and mice pre-treated with DAA-I prior to gamma irradiation showed significant improvement in the four myelodysplastic syndromes that were investigated, namely leucopenia, lymphocytopenia, monocytopenia and thrombocytopenia. Based on the known ability of PGE2 to attenuate the loss of functional hematopoietic stem and progenitor cells in radiation injury, we hypothesize that PGE2 mediated the action of DAA-I. DAA-I completely attenuated the increase in circulating level of two inflammatory cytokines, TNFα and IL-6, in irradiated mice; and this shows that DAA-I exerted additional anti-inflammatory actions, which could also have contributed to its radioprotection. These findings show that DAA-I acts via a novel mechanism of action on the angiotensin AT1 receptor to specifically release PGE2, which mediates radioprotection in the gamma irradiated mice.
Collapse
Affiliation(s)
- Hong Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Block MD11, 10 Medical Drive, Singapore, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Block MD11, 10 Medical Drive, Singapore, Singapore
| | - Weng-Keong Loke
- Agent Diagnostic and Therapeutics Laboratory, Defence & Environmental Research Institute, DSO National Laboratories, 11 Stockport Road, Singapore, Singapore
| | - Meng-Kwoon Sim
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Block MD11, 10 Medical Drive, Singapore, Singapore
| |
Collapse
|