1
|
Jeon D, Hill E, McNeel DG. Toll-like receptor agonists as cancer vaccine adjuvants. Hum Vaccin Immunother 2024; 20:2297453. [PMID: 38155525 PMCID: PMC10760790 DOI: 10.1080/21645515.2023.2297453] [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: 10/04/2023] [Accepted: 12/16/2023] [Indexed: 12/30/2023] Open
Abstract
Cancer immunotherapy has emerged as a promising strategy to treat cancer patients. Among the wide range of immunological approaches, cancer vaccines have been investigated to activate and expand tumor-reactive T cells. However, most cancer vaccines have not shown significant clinical benefit as monotherapies. This is likely due to the antigen targets of vaccines, "self" proteins to which there is tolerance, as well as to the immunosuppressive tumor microenvironment. To help circumvent immune tolerance and generate effective immune responses, adjuvants for cancer vaccines are necessary. One representative adjuvant family is Toll-Like receptor (TLR) agonists, synthetic molecules that stimulate TLRs. TLRs are the largest family of pattern recognition receptors (PRRs) that serve as the sensors of pathogens or cellular damage. They recognize conserved foreign molecules from pathogens or internal molecules from cellular damage and propel innate immune responses. When used with vaccines, activation of TLRs signals an innate damage response that can facilitate the development of a strong adaptive immune response against the target antigen. The ability of TLR agonists to modulate innate immune responses has positioned them to serve as adjuvants for vaccines targeting infectious diseases and cancers. This review provides a summary of various TLRs, including their expression patterns, their functions in the immune system, as well as their ligands and synthetic molecules developed as TLR agonists. In addition, it presents a comprehensive overview of recent strategies employing different TLR agonists as adjuvants in cancer vaccine development, both in pre-clinical models and ongoing clinical trials.
Collapse
Affiliation(s)
- Donghwan Jeon
- Department of Oncology, University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Ethan Hill
- Department of Medicine, University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Douglas G. McNeel
- Department of Medicine, University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| |
Collapse
|
2
|
Xu Y, Zhang E, Wei L, Dai Z, Chen S, Zhou S, Huang Y. NINJ1: A new player in multiple sclerosis pathogenesis and potential therapeutic target. Int Immunopharmacol 2024; 141:113021. [PMID: 39197295 DOI: 10.1016/j.intimp.2024.113021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/07/2024] [Accepted: 08/23/2024] [Indexed: 09/01/2024]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) characterized by demyelination. Current treatment options for MS focus on immunosuppression, but their efficacy can be limited. Recent studies suggest a potential role for nerve injury-induced protein 1 (NINJ1) in MS pathogenesis. NINJ1, a protein involved in cell death and inflammation, may contribute to the infiltration and activation of inflammatory cells in the CNS, potentially through enhanced blood-brain barrier crossing; enhancing plasma membrane rupture during cell death, leading to the release of inflammatory mediators and further tissue damage. This review explores the emerging evidence for NINJ1's involvement in MS. It discusses how NINJ1 might mediate the migration of immune cells across the blood-brain barrier, exacerbate neuroinflammation, and participate in plasma membrane rupture-related damage. Finally, the review examines potential therapeutic strategies targeting NINJ1 for improved MS management. Abbreviations: MS, Multiple sclerosis; CNS, Central nervous system; BBB, Blood-brain barrier; GSDMD, Gasdermin-D; EAE, Experimental autoimmune encephalitis; HMGB-1, High mobility group box-1 protein; LDH, Lactate dehydrogenase; PMR, Plasma membrane rupture; DMF, Dimethyl fumarate; DUSP1, Dual-specificity phosphatase 1; PAMPs, Pathogen-associated molecular patterns; DAMPs, Danger-associated molecular patterns; PRRs, Pattern recognition receptors; GM-CSF, Granulocyte-macrophage colony stimulating factor; IFN-γ, Interferon gamma; TNF, Tumor necrosis factor; APCs, Antigen-presenting cells; ECs, Endothelial cells; TGF-β, Transforming growth factor-β; PBMCs, Peripheral blood mononuclear cells; FACS, Fluorescence-activated cell sorting; MCP-1, Monocyte chemoattractant protein-1; NLRP3, Pyrin domain-containing 3; TCR, T cell receptor; ROS, Reactive oxygen species; AP-1, Activator protein-1; ANG1, Angiopoietin 1; BMDMs, Bone marrow-derived macrophages; Arp2/3, actin-related protein 2/3; EMT, epithelial-mesenchymal transition; FAK, focal adhesion kinase; LIMK1, LIM domain kinase 1; PAK1, p21-activated kinases 1; Rac1, Ras-related C3 botulinum toxin substrate 1; β-cat, β-caten; MyD88, myeloid differentiation primary response gene 88; TIRAP, Toll/interleukin-1 receptor domain-containing adapter protein; TLR4, Toll-like receptor 4; IRAKs, interleukin-1 receptor-associated kinases; TRAF6, TNF receptor associated factor 6; TAB2/3, TAK1 binding protein 2/3; TAK1, transforming growth factor-β-activated kinase 1; JNK, c-Jun N-terminal kinase; ERK1/2, Extracellular Signal Regulated Kinase 1/2; IKK, inhibitor of kappa B kinase; IκB, inhibitor of NF-κB; NF-κB, nuclear factor kappa-B; AP-1, activator protein-1; ASC, Apoptosis-associated Speck-like protein containing a CARD; NEK7, NIMA-related kinase 7; NLRP3, Pyrin domain-containing 3; CREB, cAMP response element-binding protein.
Collapse
Affiliation(s)
- Yinbin Xu
- Department of Neurosurgery, Ningbo Key Laboratory of Nervous System and Brain Function, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China
| | - Enhao Zhang
- Department of Neurosurgery, Ningbo Key Laboratory of Nervous System and Brain Function, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China
| | - Liangzhe Wei
- Department of Neurosurgery, Ningbo Key Laboratory of Nervous System and Brain Function, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China
| | - Zifeng Dai
- Department of Neurosurgery, Ningbo Key Laboratory of Nervous System and Brain Function, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China
| | - Siqi Chen
- Department of Neurosurgery, Ningbo Key Laboratory of Nervous System and Brain Function, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China
| | - Shengjun Zhou
- Department of Neurosurgery, Ningbo Key Laboratory of Nervous System and Brain Function, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China.
| | - Yi Huang
- Department of Neurosurgery, Ningbo Key Laboratory of Nervous System and Brain Function, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China; Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, Zhejiang 315010, China.
| |
Collapse
|
3
|
Ji H, Ma W, Zheng A, Tang D. The role and molecular mechanism of Trametes Robiniophila Murr(Huaier) in tumor therapy. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118578. [PMID: 39004194 DOI: 10.1016/j.jep.2024.118578] [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: 04/11/2024] [Revised: 06/12/2024] [Accepted: 07/11/2024] [Indexed: 07/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Trametes Robiniophila Murr, commonly known as Huaier, has been extensively documented in ethnopharmacology research in China. Huaier has a long history of clinical usage spanning over 1000 years in China. Traditional clinical application records demonstrate the wide utilization of Huaier for treating various cancers and enhancing the autoimmunity of tumor patients. AIM OF THE REVIEW The present study provides a comprehensive review of the traditional uses, phytochemical constituents, pharmacological activities, anti-tumor mechanism, and potential applications of Huaier, thereby offering valuable insights for the further development and utilization of this natural product. MATERIALS AND METHODS This study employed the keywords "Trametes Robiniophila Murr" and "Huaier" to retrieve relevant information on Huaier from various databases, including PubMed, Web of Science, Springer, Science Direct, ACS, Wiley, CNKI, Baidu Scholar, Google Scholar, and ancient materia medica. RESULTS Trametes Robiniophila Murr (Huaier), a traditional Chinese medicine, has demonstrated significant efficacy in the clinical treatment of various tumors. The primary bioactive constituents of Huaier consist of fungal-derived compounds, including polysaccharides, proteins, ketones, alkaloids, and minerals. The research findings demonstrate that Huaier serves as a reliable adjunctive therapeutic agent by effectively inhibiting cancer cell proliferation, inducing apoptosis in cancer cells, suppressing tumor metastasis, regulating tumor stem cells and immune function. Therefore, it exerts a potent anti-tumor effect when used in conjunction with conventional anti-cancer therapies. CONCLUSIONS The analysis of traditional uses, phytochemical composition, and pharmacological activity reveals that Huaier exhibits significant potential as a medicinal plant with diverse pharmacological effects. Owing to its numerous advantages, Huaier holds immense promise for application in the domains of tumor prevention and treatment, enhancing both survival time and quality of life among cancer patients.
Collapse
Affiliation(s)
- Hao Ji
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou University, Yangzhou, 225000, China.
| | - Wei Ma
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou University, Yangzhou, 225000, China.
| | - Aiyu Zheng
- Department of Geriatrics, Taixing People's Hospital, Taixing, 225400, China.
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital; Northern Jiangsu People's Hospital Affiliated to Yangzhou University; The Yangzhou Clinical Medical College of Xuzhou Medical University; The Yangzhou School of Clinical Medicine of Dalian Medical University; The Yangzhou School of Clinical Medicine of Nanjing Medical University; Northern Jiangsu People's Hospital, Clinical Teaching Hospital of Medical School, Nanjing University, Yangzhou, 225000, China.
| |
Collapse
|
4
|
Hisaoka-Nakashima K, Takeuchi Y, Saito Y, Shimoda T, Nakamura Y, Wang D, Liu K, Nishibori M, Morioka N. Glucocorticoids induce HMGB1 release in primary cultured rat cortical microglia. Neuroscience 2024; 560:56-66. [PMID: 39304023 DOI: 10.1016/j.neuroscience.2024.09.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 09/01/2024] [Accepted: 09/15/2024] [Indexed: 09/22/2024]
Abstract
Stress, a risk factor for major depressive disorder and Alzheimer disease, leads to the release of high-mobility group box-1 (HMGB1) protein, which in turn causes neuroinflammation. The mechanism underlying stress-induced HMGB1 release is unknown, but stress-associated glucocorticoids could be involved. Primary cultured rat cortical microglia and neurons were treated with corticosterone, a stress-associated glucocorticoid, and HMGB1 release was measured by ELISA and western blotting to test this hypothesis. With corticosterone treatment, significant HMGB1 was released in microglia but not in neuronal cell cultures. HMGB1 mRNA expression and HMGB1 protein expression in microglia were not affected by corticosterone treatment. Thus, the source of extracellular HMGB1 released into the medium is likely to be existing nuclear HMGB1 rather than newly synthesized HMGB1. Corticosterone-induced HMGB1 release in microglia culture was significantly attenuated by blocking glucocorticoid receptors but not mineralocorticoid receptors. Dexamethasone, a selective glucocorticoid receptor agonist, and dexamethasone-bovine serum albumin (BSA), a membrane-impermeable glucocorticoid receptor agonist used to confirm the membrane receptor-mediated effects of glucocorticoids, increased the release of HMGB1. Immunocytochemistry showed that HMGB1 translocated from the nucleus to the cytoplasm following dexamethasone or dexamethasone-BSA treatment through glucocorticoid receptors. The present findings suggest that glucocorticoids stimulate microglial membrane glucocorticoid receptors and trigger cytoplasmic translocation and extracellular release of nuclear HMGB1. Thus, under stress conditions, glucocorticoids induce microglial HMGB1 release, leading to a neuroinflammatory state that could mediate neurological disorders.
Collapse
Affiliation(s)
- Kazue Hisaoka-Nakashima
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Yuka Takeuchi
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Yukino Saito
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Takahisa Shimoda
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan; Laboratory for Pharmacology, Pharmaceuticals Research Center, Asahi Kasei Pharma Co. Ltd., 632-1 Mifuku, Izunokuni, Shizuoka 410-2321, Japan
| | - Yoki Nakamura
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Dengli Wang
- Department of Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Shikata, Okayama, Japan
| | - Keyue Liu
- Department of Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Shikata, Okayama, Japan
| | - Masahiro Nishibori
- Department of Translational Research & Drug Development, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Shikata, Okayama, Japan
| | - Norimitsu Morioka
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan.
| |
Collapse
|
5
|
Martinez PJ, Song JJ, Castillo JI, DeSisto J, Song KH, Green AL, Borden M. Effect of Microbubble Size, Composition, and Multiple Sonication Points on Sterile Inflammatory Response in Focused Ultrasound-Mediated Blood-Brain Barrier Opening. ACS Biomater Sci Eng 2024. [PMID: 39497639 DOI: 10.1021/acsbiomaterials.4c00777] [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: 11/13/2024]
Abstract
Blood-brain barrier opening (BBBO) using focused ultrasound (FUS) and microbubbles (MBs) has emerged as a promising technique for delivering therapeutics to the brain. However, the influence of various FUS and MB parameters on BBBO and subsequent sterile inflammatory response (SIR) remains unclear. In this study, we investigated the effects of MB size and composition, as well as the number of FUS sonication points, on BBBO and SIR in an immunocompetent mouse model. Using MRI-guided MB + FUS, we targeted the striatum and assessed extravasation of an MRI contrast agent to assess BBBO and RNaseq to assess SIR. Our results revealed distinct effects of these parameters on BBBO and SIR. Specifically, at a matched microbubble volume dose (MVD), MB size did not affect the extent of BBBO, but smaller (1 μm diameter) MBs exhibited a lower classification of SIR than larger (3 or 5 μm diameter) MBs. Lipid-shelled microbubbles exhibited greater BBBO and a more pronounced SIR compared to albumin-shelled microbubbles, likely owing to the latter's poor in vivo stability. As expected, increasing the number of sonication points resulted in greater BBBO and SIR. Furthermore, correlation analysis revealed strong associations between passive cavitation detection measurements of harmonic and inertial MB echoes, BBBO, and the expression of SIR gene sets. Our findings highlight the critical role of MB and FUS parameters in modulating BBBO and subsequent SIR in the brain. These insights inform the development of targeted drug delivery strategies and the mitigation of adverse inflammatory reactions in neurological disorders.
Collapse
Affiliation(s)
- Payton J Martinez
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, Colorado 80303, United States
| | - Jane J Song
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, Colorado 80303, United States
| | - Jair I Castillo
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, Colorado 80303, United States
| | - John DeSisto
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado 80045, United States
| | - Kang-Ho Song
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, Colorado 80303, United States
| | - Adam L Green
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado 80045, United States
| | - Mark Borden
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, Colorado 80303, United States
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, Colorado 80303, United States
| |
Collapse
|
6
|
Smith EE, Biessels GJ, Gao V, Gottesman RF, Liesz A, Parikh NS, Iadecola C. Systemic determinants of brain health in ageing. Nat Rev Neurol 2024; 20:647-659. [PMID: 39375564 DOI: 10.1038/s41582-024-01016-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2024] [Indexed: 10/09/2024]
Abstract
Preservation of brain health is a worldwide priority. The traditional view is that the major threats to the ageing brain lie within the brain itself. Consequently, therapeutic approaches have focused on protecting the brain from these presumably intrinsic pathogenic processes. However, an increasing body of evidence has unveiled a previously under-recognized contribution of peripheral organs to brain dysfunction and damage. Thus, in addition to the well-known impact of diseases of the heart and endocrine glands on the brain, accumulating data suggest that dysfunction of other organs, such as gut, liver, kidney and lung, substantially affects the development and clinical manifestation of age-related brain pathologies. In this Review, a framework is provided to indicate how organ dysfunction can alter brain homeostasis and promote neurodegeneration, with a focus on dementia. We delineate the associations of subclinical dysfunction in specific organs with dementia risk and provide suggestions for public health promotion and clinical management.
Collapse
Affiliation(s)
- Eric E Smith
- Department of Clinical Neurosciences and Hotchkiss Brain Institute, University of Calgary, Calgary, Canada.
| | - Geert Jan Biessels
- Department of Neurology, UMC Utrecht Brain Center, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Virginia Gao
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | | | - Arthur Liesz
- Institute for Stroke and Dementia Research, University Medical Center Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Neal S Parikh
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
7
|
Pan J, Qu J, Fang W, Zhao L, Zheng W, Zhai L, Tan M, Xu Q, Du Q, Lv W, Sun Y. SHP2-Triggered Endothelial Cell Activation Fuels Estradiol-Independent Endometrial Sterile Inflammation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403038. [PMID: 39234819 PMCID: PMC11538683 DOI: 10.1002/advs.202403038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 07/30/2024] [Indexed: 09/06/2024]
Abstract
Sterile inflammation occurs in various chronic diseases due to many nonmicrobe factors. Examples include endometrial hyperplasia (EH), endometriosis, endometrial cancer, and breast cancer, which are all sterile inflammation diseases induced by estrogen imbalances. However, how estrogen-induced sterile inflammation regulates EH remains unclear. Here, a single-cell RNA-Seq is used to show that SHP2 upregulation in endometrial endothelial cells promotes their inflammatory activation and subsequent transendothelial macrophage migration. Independent of the initial estrogen stimulation, IL1β and TNFα from macrophages then create a feedforward loop that enhances endothelial cell activation and IGF1 secretion. This endothelial cell-macrophage interaction sustains sterile endometrial inflammation and facilitates epithelial cell proliferation, even after estradiol withdrawal. The bulk RNA-Seq results and phosphoproteomic analysis show that endothelial SHP2 mechanistically enhances RIPK1 activity by dephosphorylating RIPK1Tyr380. This event activates downstream activator protein 1 (AP-1) and instigates the inflammation response. Furthermore, targeting SHP2 using SHP099 (an allosteric inhibitor) or endothelial-specific SHP2 deletion alleviates endothelial cell activation, macrophage infiltration, and EH progression in mice. Collectively, the findings demonstrate that SHP2 mediates the transition of endothelial activation from estradiol-driven acute inflammation to macrophage-amplified chronic inflammation. Targeting sterile inflammation mediated by endothelial cell activation is a promising strategy for nonhormonal intervention in estrogen-related diseases.
Collapse
Affiliation(s)
- Jie Pan
- State Key Laboratory of Pharmaceutical Biotechnology and Nanjing Drum Tower Hospital the Affiliated Hospital of Nanjing University Medical SchoolSchool of Life SciencesNanjing University163 Xianlin AvenueNanjing210023China
| | - Jiao Qu
- State Key Laboratory of Pharmaceutical Biotechnology and Nanjing Drum Tower Hospital the Affiliated Hospital of Nanjing University Medical SchoolSchool of Life SciencesNanjing University163 Xianlin AvenueNanjing210023China
- Jiangsu Key Laboratory of New Drug Research and Clinical PharmacyXuzhou Medical University209 Tongshan RoadXuzhouJiangsu221004China
| | - Wen Fang
- State Key Laboratory of Pharmaceutical Biotechnology and Nanjing Drum Tower Hospital the Affiliated Hospital of Nanjing University Medical SchoolSchool of Life SciencesNanjing University163 Xianlin AvenueNanjing210023China
| | - Lixin Zhao
- State Key Laboratory of Pharmaceutical Biotechnology and Nanjing Drum Tower Hospital the Affiliated Hospital of Nanjing University Medical SchoolSchool of Life SciencesNanjing University163 Xianlin AvenueNanjing210023China
| | - Wei Zheng
- State Key Laboratory of Pharmaceutical Biotechnology and Nanjing Drum Tower Hospital the Affiliated Hospital of Nanjing University Medical SchoolSchool of Life SciencesNanjing University163 Xianlin AvenueNanjing210023China
| | - Linhui Zhai
- Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - Minjia Tan
- Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology and Nanjing Drum Tower Hospital the Affiliated Hospital of Nanjing University Medical SchoolSchool of Life SciencesNanjing University163 Xianlin AvenueNanjing210023China
| | - Qianming Du
- General Clinical Research CenterNanjing First HospitalNanjing Medical UniversityNanjing210006China
- School of Basic Medicine & Clinical PharmacyChina Pharmaceutical UniversityNanjing210009China
| | - Wen Lv
- Department of GynecologyTongde Hospital of Zhejiang Province234 Gucui RoadHangzhouZhejiang310012China
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology and Nanjing Drum Tower Hospital the Affiliated Hospital of Nanjing University Medical SchoolSchool of Life SciencesNanjing University163 Xianlin AvenueNanjing210023China
- Jiangsu Key Laboratory of New Drug Research and Clinical PharmacyXuzhou Medical University209 Tongshan RoadXuzhouJiangsu221004China
| |
Collapse
|
8
|
Aboulaghras S, Bouyahya A, El Kadri K, Khalid A, Abdalla AN, Hassani R, Lee LH, Bakrim S. Protective and stochastic correlation between infectious diseases and autoimmune disorders. Microb Pathog 2024; 196:106919. [PMID: 39245422 DOI: 10.1016/j.micpath.2024.106919] [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: 08/10/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
A priori, early exposure to a wide range of bacteria, viruses, and parasites appears to fortify and regulate the immune system, potentially reducing the risk of autoimmune diseases. However, improving hygiene conditions in numerous societies has led to a reduction in these microbial exposures, which, according to certain theories, could contribute to an increase in autoimmune diseases. Indeed, molecular mimicry is a key factor triggering immune system reactions; while it seeks pathogens, it can bind to self-molecules, leading to autoimmune diseases associated with microbial infections. On the other hand, a hygiene-based approach aimed at reducing the load of infectious agents through better personal hygiene can be beneficial for such pathologies. This review sheds light on how the evolution of the innate immune system, following the evolution of molecular patterns associated with microbes, contributes to our protection but may also trigger autoimmune diseases linked to microbes. Furthermore, it addresses how hygiene conditions shield us against autoimmune diseases related to microbes but may lead to autoimmune pathologies not associated with microbes.
Collapse
Affiliation(s)
| | - Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, 10106, Morocco.
| | - Kawtar El Kadri
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, 10106, Morocco.
| | - Asaad Khalid
- Health Research Centre, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia.
| | - Ashraf N Abdalla
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah, 21955, Saudi Arabia.
| | - Rym Hassani
- Environment and Nature Research Centre, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia; Biology Department, University College AlDarb, Jazan University, Jazan 45142, Saudi Arabia.
| | - Learn-Han Lee
- Microbiome Research Group, Research Centre for Life Science and Healthcare, Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute (CBI), University of Nottingham Ningbo China, 315000, Ningbo, China; Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Selangor, 47500, Malaysia.
| | - Saad Bakrim
- Geo-Bio-Environment Engineering and Innovation Laboratory, Molecular Engineering, Biotechnology and Innovation Team, Polydisciplinary Faculty of Taroudant, Ibn Zohr University, Agadir, 80000, Morocco.
| |
Collapse
|
9
|
Castellanos-Molina A, Bretheau F, Boisvert A, Bélanger D, Lacroix S. Constitutive DAMPs in CNS injury: From preclinical insights to clinical perspectives. Brain Behav Immun 2024; 122:583-595. [PMID: 39222725 DOI: 10.1016/j.bbi.2024.07.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/28/2024] [Accepted: 07/04/2024] [Indexed: 09/04/2024] Open
Abstract
Damage-associated molecular patterns (DAMPs) are endogenous molecules released in tissues upon cellular damage and necrosis, acting to initiate sterile inflammation. Constitutive DAMPs (cDAMPs) have the particularity to be present within the intracellular compartments of healthy cells, where they exert diverse functions such as regulation of gene expression and cellular homeostasis. However, after injury to the central nervous system (CNS), cDAMPs are rapidly released by stressed, damaged or dying neuronal, glial and endothelial cells, and can trigger inflammation without undergoing structural modifications. Several cDAMPs have been described in the injured CNS, such as interleukin (IL)-1α, IL-33, nucleotides (e.g. ATP), and high-mobility group box protein 1. Once in the extracellular milieu, these molecules are recognized by the remaining surviving cells through specific DAMP-sensing receptors, thereby inducing a cascade of molecular events leading to the production and release of proinflammatory cytokines and chemokines, as well as cell adhesion molecules. The ensuing immune response is necessary to eliminate cellular debris caused by the injury, allowing for damage containment. However, seeing as some molecules associated with the inflammatory response are toxic to surviving resident CNS cells, secondary damage occurs, aggravating injury and exacerbating neurological and behavioral deficits. Thus, a better understanding of these cDAMPs, as well as their receptors and downstream signaling pathways, could lead to identification of novel therapeutic targets for treating CNS injuries such as SCI, TBI, and stroke. In this review, we summarize the recent literature on cDAMPs, their specific functions, and the therapeutic potential of interfering with cDAMPs or their signaling pathways.
Collapse
Affiliation(s)
- Adrian Castellanos-Molina
- Axe Neurosciences du Centre de recherche du Centre hospitalier universitaire (CHU) de Québec-Université Laval et Département de médecine moléculaire de l'Université Laval, Québec, QC G1V 4G2, Canada
| | - Floriane Bretheau
- Axe Neurosciences du Centre de recherche du Centre hospitalier universitaire (CHU) de Québec-Université Laval et Département de médecine moléculaire de l'Université Laval, Québec, QC G1V 4G2, Canada
| | - Ana Boisvert
- Axe Neurosciences du Centre de recherche du Centre hospitalier universitaire (CHU) de Québec-Université Laval et Département de médecine moléculaire de l'Université Laval, Québec, QC G1V 4G2, Canada
| | - Dominic Bélanger
- Axe Neurosciences du Centre de recherche du Centre hospitalier universitaire (CHU) de Québec-Université Laval et Département de médecine moléculaire de l'Université Laval, Québec, QC G1V 4G2, Canada
| | - Steve Lacroix
- Axe Neurosciences du Centre de recherche du Centre hospitalier universitaire (CHU) de Québec-Université Laval et Département de médecine moléculaire de l'Université Laval, Québec, QC G1V 4G2, Canada.
| |
Collapse
|
10
|
Wu X, Zhao X, Li F, Wang Y, Ou Y, Zhang H, Li X, Wu X, Wang L, Li M, Zhang Y, Liu J, Xing M, Liu H, Tan Y, Wang Y, Xie Y, Zhang H, Luo Y, Li H, Wang J, Sun L, Li Y, Zhang H. MLKL-mediated endothelial necroptosis drives vascular damage and mortality in systemic inflammatory response syndrome. Cell Mol Immunol 2024; 21:1309-1321. [PMID: 39349742 PMCID: PMC11527879 DOI: 10.1038/s41423-024-01217-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 09/11/2024] [Indexed: 11/02/2024] Open
Abstract
The hypersecretion of cytokines triggers life-threatening systemic inflammatory response syndrome (SIRS), leading to multiple organ dysfunction syndrome (MODS) and mortality. Although both coagulopathy and necroptosis have been identified as important factors in the pathogenesis of SIRS, the specific cell types that undergo necroptosis and the interrelationships between coagulopathy and necroptosis remain unclear. In this study, we utilized visualization analysis via intravital microscopy to demonstrate that both anticoagulant heparin and nonanticoagulant heparin (NAH) pretreatment protect mice against TNF-α-induced mortality in SIRS. Moreover, the deletion of Mlkl or Ripk3 resulted in decreased coagulation and reduced mortality in TNF-α-induced SIRS. These findings suggest that necroptosis plays a key role upstream of coagulation in SIRS-related mortality. Furthermore, using a genetic lineage tracing mouse model (Tie2-Cre;Rosa26-tdT), we tracked endothelial cells (ECs) and verified that EC necroptosis is responsible for the vascular damage observed in TNF-α-treated mice. Importantly, Mlkl deletion in vascular ECs in mice had a similar protective effect against lethal SIRS by blocking EC necroptosis to protect the integrity of the endothelium. Collectively, our findings demonstrated that RIPK3-MLKL-dependent necroptosis disrupted vascular integrity, resulting in coagulopathy and multiorgan failure, eventually leading to mortality in SIRS patients. These results highlight the importance of targeting vascular EC necroptosis for the development of effective treatments for SIRS patients.
Collapse
Affiliation(s)
- Xiaoxia Wu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Xiaoming Zhao
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Fang Li
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yang Wang
- Department of Microbiology and Immunology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yangjing Ou
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Haiwei Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Xiaoming Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Xuanhui Wu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Lingxia Wang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Ming Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Yue Zhang
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Jianling Liu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Mingyan Xing
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Han Liu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Yongchang Tan
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yangyang Wang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Yangyang Xie
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Hanwen Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Yan Luo
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Hong Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Jing Wang
- Department of Microbiology and Immunology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Liming Sun
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, PR China
| | - Yu Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Haibing Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China.
| |
Collapse
|
11
|
Jumabayi W, Reyimu A, Zheng R, Paerhati P, Rahman M, Zou X, Xu A. Ferroptosis: A new way to intervene in the game between Mycobacterium tuberculosis and macrophages. Microb Pathog 2024; 197:107014. [PMID: 39396689 DOI: 10.1016/j.micpath.2024.107014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/06/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
Mycobacterium tuberculosis (Mtb), the main pathogen responsible for the high mortality and morbidity of tuberculosis (TB) worldwide, primarily targets and invades macrophages. Infected macrophages activate a series of immune mechanisms to clear Mtb, however, Mtb evades host immune surveillance through subtle immune escape strategies to create a microenvironment conducive to its own proliferation, growth, and dissemination, while inducing immune cell death. The course of TB is strongly correlated with the form of cell death, including apoptosis, pyroptosis, and necrosis. Recent studies have revealed that ferroptosis, a novel type of programmed cell death characterized by iron-dependent lipid peroxidation, is closely linked to the regulatory mechanisms of TB. The central role of ferroptosis in the pathologic process of TB is increasingly becoming a focal point for exploring new therapeutic targets in this field. This paper will delve into the dynamic game between Mtb and host immune cells, especially the role of ferroptosis in the pathogenesis of TB. At the same time, this paper will analyze the regulatory pathways of ferroptosis and provide unique insights and innovative perspectives for TB therapeutic strategies based on the ferroptosis mechanism. This study not only expands the theoretical basis of TB treatment, but also points out the direction of future drug development, providing new possibilities for overcoming this global health problem.
Collapse
Affiliation(s)
- Wuerken Jumabayi
- The Third Clinical Medical College (Affiliated Cancer Hospital) of Xinjiang Medical University, Urumqi, China
| | | | | | | | | | | | - Aimin Xu
- The First People's Hospital of Kashi, Kashi, China.
| |
Collapse
|
12
|
Jiang YY, Yan ST, Zhang SZ, Wang M, Diao WM, Li J, Fang XM, Yin H. Discovery of pyrazolo[1,5-a]pyrimidine derivatives targeting TLR4-TLR4∗ homodimerization via AI-powered next-generation screening. Eur J Med Chem 2024; 280:116945. [PMID: 39388907 DOI: 10.1016/j.ejmech.2024.116945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/28/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024]
Abstract
TLR4 signaling is instrumental in orchestrating multiple aspects of innate immunity. Developing small molecule inhibitors targeting the TLR4 pathway holds potential therapeutic promise for TLR4-related disorders. Herein, an artificial intelligence (AI)-powered next-generation screening approach, employing HelixVS and HelixDock, was utilized to focus on the TLR4-TLR4∗ (a second copy of TLR4) homodimerization surface, leading to the identification of a potent pyrazolo[1,5-a]pyrimidine derivative, designated as compound 1. An extensive structure-activity relationship (SAR) exploration culminated in the discovery of the lead compound TH023, which effectively blocked the LPS-stimulated NF-κB activation and nitric oxide overproduction in HEK-Blue hTLR4 and RAW264.7 cells, with IC50 values of 0.354 and 1.61 μM, respectively. Molecular dynamic (MD) simulations indicated that TH023 stabilized TLR4-MD-2 and disrupted its association with TLR4∗. Moreover, TH023 alleviated the lung injury and decreased pro-inflammatory cytokine levels in LPS-induced septic mice. These findings not only illuminated the strategic advantage of HelixDock in advancing the frontiers of AI-driven drug discovery, but also provided valuable structural insights for the rational design of TLR4-TLR4∗ protein-protein interaction (PPI) inhibitors based on the pyrazolo[1,5-a]pyrimidine scaffold. Overall, this study validated a new strategy for TLR4 signaling regulation by targeting its dimerization, thereby underscoring the therapeutic promise of TH023 in treating TLR4-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Yao-Yao Jiang
- State Key Laboratory of Membrane Biology, School of Pharmaceutical Sciences, Tsinghua-Peking Center for Life Sciences, Key Laboratory of Bioorganic Phosphorous Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing, 100084, China
| | - Shuai-Ting Yan
- State Key Laboratory of Membrane Biology, School of Pharmaceutical Sciences, Tsinghua-Peking Center for Life Sciences, Key Laboratory of Bioorganic Phosphorous Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing, 100084, China
| | | | - Meng Wang
- Toll Biotech Co., Ltd. (Beijing), Beijing, 102209, China
| | - Wei-Ming Diao
- Toll Biotech Co., Ltd. (Beijing), Beijing, 102209, China
| | - Jun Li
- PaddleHelix Team, Baidu Inc., Shenzhen, 518000, China
| | - Xiao-Min Fang
- PaddleHelix Team, Baidu Inc., Shenzhen, 518000, China
| | - Hang Yin
- State Key Laboratory of Membrane Biology, School of Pharmaceutical Sciences, Tsinghua-Peking Center for Life Sciences, Key Laboratory of Bioorganic Phosphorous Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
13
|
Antonio L, Visalli G, Facciolà A, Saija C, Bertuccio MP, Baluce B, Celesti C, Iannazzo D, Di Pietro A. Sterile inflammation induced by respirable micro and nano polystyrene particles in the pathogenesis of pulmonary diseases. Toxicol Res (Camb) 2024; 13:tfae138. [PMID: 39233846 PMCID: PMC11368663 DOI: 10.1093/toxres/tfae138] [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: 05/07/2024] [Revised: 07/24/2024] [Accepted: 08/26/2024] [Indexed: 09/06/2024] Open
Abstract
Sterile inflammation is involved in the lung pathogenesis induced by respirable particles, including micro- and nanoplastics. Their increasing amounts in the ambient and in indoor air pose a risk to human health. In two human cell lines (A549 and THP-1) we assessed the proinflammatory behavior of polystyrene nanoplastics (nPS) and microplastics (mPS) (Ø 0.1 and 1 μm). Reproducing environmental aging, in addition to virgin, the cells were exposed to oxidized nPS/mPS. To study the response of the monocytes to the inflammatory signal transmitted by the A549 through the release of soluble factors (e.g. alarmins and cytokines), THP-1 cells were also exposed to the supernatants of previously nPS/mPS-treated A549. After dynamic-light-scattering (DLS) analysis and protein measurements for the assessment of protein corona in nPS/mPS, real-time PCR and enzyme-linked-immunosorbent (ELISA) assays were performed in exposed cells. The pro-inflammatory effects of v- and ox-nPS/mPS were attested by the imbalance of the Bax/Bcl-2 ratio in A549, which was able to trigger the inflammatory cascade, inhibiting the immunologically silent apoptosis. The involvement of NFkB was confirmed by the overexpression of p65 after exposure to ox-nPS and v- and ox-mPS. The fast and higher levels of IL-1β, only in THP-1 cells, underlined the NLPR3 inflammasome activation.
Collapse
Affiliation(s)
- Laganà Antonio
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
- Istituto Clinico Polispecialistico C.O.T. Cure Ortopediche Traumatologiche s.p.a., Viale Italia, 98124 Messina, Italy
| | - Giuseppa Visalli
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Alessio Facciolà
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Caterina Saija
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Maria Paola Bertuccio
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Barbara Baluce
- Department of Transfusion Medicine and Hematology and Lombardy Regional Rare Blood Bank, IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Via Francesco Sforza, 35, 20122 Milan, Italy
| | - Consuelo Celesti
- Department of Electronic Engineering, Industrial Chemistry and Engineering, University of Messina, Via Stagno d'Alcontres, 98125 Messina, Italy
| | - Daniela Iannazzo
- Department of Electronic Engineering, Industrial Chemistry and Engineering, University of Messina, Via Stagno d'Alcontres, 98125 Messina, Italy
| | - Angela Di Pietro
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| |
Collapse
|
14
|
Ji Y, Hua H, Jia Z, Zhang A, Ding G. Therapy Targeted to the NLRP3 Inflammasome in Chronic Kidney Disease. KIDNEY DISEASES (BASEL, SWITZERLAND) 2024; 10:369-383. [PMID: 39430292 PMCID: PMC11488838 DOI: 10.1159/000539496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 05/07/2024] [Indexed: 10/22/2024]
Abstract
Background The NLRP3 inflammasome is a cytoplasmic polymeric protein complex composed of the cytoplasmic sensor NLRP3, the apoptosis-related spot-like protein ASC, and the inflammatory protease caspase-1. NLRP3 activates and releases IL-1β through classical pathways, and IL-18 mediates inflammation and activates gasdermin-D protein to induce cellular pyroptosis. Numerous studies have also emphasized the non-classical pathway activated by the NLRP3 inflammasome in chronic kidney disease (CKD) and the inflammasome-independent function of NLRP3. Summary The NLRP3-targeting inflammasome and its associated pathways have thus been widely studied in models of CKD treatment, but no drug that targets NLRP3 has thus far been approved for the treatment of CKD. Key Messages We herein reviewed the current interventional methods for targeting the NLRP3 inflammasome in various CKD models, analyzed their underlying mechanisms of action, classified and compared them, and discussed the advantages and follow-up directions of various interventional methods. This review therefore provides novel ideas and a reference for the development of targeted NLRP3-inflammasome therapy in CKD.
Collapse
Affiliation(s)
- Yong Ji
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Hu Hua
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Zhanjun Jia
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Aihua Zhang
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Guixia Ding
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
15
|
Tong J, Yao G, Chen Y, Xie H, Zheng X, Sun L, Huang Z, Xie Z. Mesenchymal Stem Cells Regulate Microglial Polarization via Inhibition of the HMGB1/TLR4 Signaling Pathway in Diabetic Retinopathy. Inflammation 2024; 47:1728-1743. [PMID: 38625640 DOI: 10.1007/s10753-024-02005-6] [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: 01/05/2024] [Revised: 02/29/2024] [Accepted: 03/05/2024] [Indexed: 04/17/2024]
Abstract
Diabetic retinopathy (DR) is recognized as the most prevalent retinal degenerative disorder. Inflammatory response usually precedes microvascular alteration and is the primary factor of diabetic retinopathy. Activated microglia express many pro-inflammatory cytokines that exacerbate retina inflammation and disruption. In the present study, we found that MSCs alleviated blood-retina barrier (BRB) breakdown in diabetic rats, as evidenced by reduced retinal edema, decreased vascular leakage, and increased occludin expression. The MSC-treated retinal microglia exhibited reduced expression of M1-phenotype markers in the diabetic rats, including inducible nitric oxide synthase (iNOS), CD16, and pro-inflammatory cytokines. On the other hand, MSCs increased the expression of M2-phenotype markers, such as arginase-1 (Arg-1), CD206, and anti-inflammatory cytokines. HMGB1/TLR4 signaling pathway is activated in DR and inhibited after MSC treatment. Consistent with in vivo evidence, MSCs drove BV2 microglia toward M2 phenotype in vitro. Overexpression of HMGB1 in microglia reversed the effects of MSC treatment, suggesting HMGB1/TLR4 pathway is necessary for MSCs' regulatory effects on microglia polarization. Collectively, MSCs exert beneficial effects on DR by polarizing microglia from M1 toward M2 phenotype via inhibiting the HMGB1/TLR4 signaling pathway.
Collapse
Affiliation(s)
- Jun Tong
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of Ophthalmology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Genhong Yao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yueqin Chen
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Hairong Xie
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xinyu Zheng
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Zhenping Huang
- Department of Ophthalmology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Zhenggao Xie
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
16
|
Ji P, Zhou Z, Zhang J, Bai T, Li C, Zhou B, Wang M, Tan Y, Wang S. Non-apoptotic cell death in osteoarthritis: Recent advances and future. Biomed Pharmacother 2024; 179:117344. [PMID: 39191021 DOI: 10.1016/j.biopha.2024.117344] [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: 04/12/2024] [Revised: 06/23/2024] [Accepted: 08/22/2024] [Indexed: 08/29/2024] Open
Abstract
Osteoarthritis (OA) is the most common degenerative joint disease. Multiple tissues are altered during the development of OA, resulting in joint pain and permanent damage to the osteoarticular joints. Current research has demonstrated that non-apoptotic cell death plays a crucial role in OA. With the continuous development of targeted therapies, non-apoptotic cell death has shown great potential in the prevention and treatment of OA. We systematically reviewed research progress on the role of non-apoptotic cell death in the pathogenesis, development, and outcome of OA, including autophagy, pyroptosis, ferroptosis, necroptosis, immunogenic cell death, and parthanatos. This article reviews the mechanism of non-apoptotic cell death in OA and provides a theoretical basis for the identification of new targets for OA treatment.
Collapse
Affiliation(s)
- Pengfei Ji
- The Second Clinical Medical College, Lanzhou University, No. 199 DongGang West Road, Lanzhou, Gansu 730000, China
| | - Ziyu Zhou
- The Second Clinical Medical College, Lanzhou University, No. 199 DongGang West Road, Lanzhou, Gansu 730000, China
| | - Jinyuan Zhang
- The Second Clinical Medical College, Lanzhou University, No. 199 DongGang West Road, Lanzhou, Gansu 730000, China
| | - Tianding Bai
- People's Hospital of Guazhou County, Guazhou, Gansu 736100, China
| | - Chao Li
- The Second Clinical Medical College, Lanzhou University, No. 199 DongGang West Road, Lanzhou, Gansu 730000, China
| | - Binghao Zhou
- The Second Clinical Medical College, Lanzhou University, No. 199 DongGang West Road, Lanzhou, Gansu 730000, China
| | - Mengjie Wang
- The Second Clinical Medical College, Lanzhou University, No. 199 DongGang West Road, Lanzhou, Gansu 730000, China
| | - Yingdong Tan
- People's Hospital of Guazhou County, Guazhou, Gansu 736100, China.
| | - Shengwang Wang
- People's Hospital of Guazhou County, Guazhou, Gansu 736100, China.
| |
Collapse
|
17
|
Lee HY, Park YM, Shin DY, Hwang HM, Jeong HN, Park HY, Yang HJ, Ha GS, Ryu MS, Seo JW, Jeong DY, Bae JS, Kim BS, Kim JG. Immune-enhancing effect of fermented soybean food, Cheonggukjang on cyclophosphamide-treated immunosuppressed rat. Heliyon 2024; 10:e37845. [PMID: 39328544 PMCID: PMC11425096 DOI: 10.1016/j.heliyon.2024.e37845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/02/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024] Open
Abstract
Cheonggukjang (CGJ) is a traditional food, made by the fermentation of beans, and it has different recipes for different regions in Korea. However, it has anti-inflammatory, anti-cancer, and anti-obesity effects, and is known to affect changes in the intestinal microbiota. In this study, we investigated the immune-enhancing effects of four type CGJs (one commercial and three transitional CGJs). In the cyclophosphamide (CP)-treated immunosuppressed rat, oral administration of CGJs for 4 weeks was used to investigate weight of body and immune organ, change of microbiota, blood and serum parameters, inflammation pathways (MAPKs and NFκB) and histology of spleen. It showed an immunity-enhancing effect through increase Bacteroidetes in gut, the recovery of complete blood count, levels of cytokines and IgG, activation of inflammatory pathways, and histology of spleen. In conclusion, these results show that the intake of a commercial brand CGJ, and traditional CGJs can maintain or promote the body's immunity.
Collapse
Affiliation(s)
- Hak Yong Lee
- INVIVO Co. Ltd., 121, Deahak-ro, Nonsan, Chungnam, 32992, Republic of Korea
| | - Young Mi Park
- INVIVO Co. Ltd., 121, Deahak-ro, Nonsan, Chungnam, 32992, Republic of Korea
- Department of Pathology, College of Korean Medicine, Wonkwang University, Iksan, Republic of Korea
| | - Dong Yeop Shin
- INVIVO Co. Ltd., 121, Deahak-ro, Nonsan, Chungnam, 32992, Republic of Korea
- Department of Integrated Life Science and Technology, Kongju National University, 32439, Republic of Korea
| | - Hai Min Hwang
- INVIVO Co. Ltd., 121, Deahak-ro, Nonsan, Chungnam, 32992, Republic of Korea
| | - Han Na Jeong
- INVIVO Co. Ltd., 121, Deahak-ro, Nonsan, Chungnam, 32992, Republic of Korea
| | - Hyo Yeon Park
- INVIVO Co. Ltd., 121, Deahak-ro, Nonsan, Chungnam, 32992, Republic of Korea
| | - Hee-Jong Yang
- Microbial Institute for Fermentation Industry (MIFI), Sunchang, 56048, Republic of Korea
| | - Gwang Su Ha
- Microbial Institute for Fermentation Industry (MIFI), Sunchang, 56048, Republic of Korea
| | - Myeong Seon Ryu
- Microbial Institute for Fermentation Industry (MIFI), Sunchang, 56048, Republic of Korea
| | - Ji Won Seo
- Microbial Institute for Fermentation Industry (MIFI), Sunchang, 56048, Republic of Korea
| | - Do-Youn Jeong
- Microbial Institute for Fermentation Industry (MIFI), Sunchang, 56048, Republic of Korea
| | - Jun Sang Bae
- Department of Pathology, College of Korean Medicine, Wonkwang University, Iksan, Republic of Korea
| | - Byeong Soo Kim
- Department of Integrated Life Science and Technology, Kongju National University, 32439, Republic of Korea
| | - Jae Gon Kim
- INVIVO Co. Ltd., 121, Deahak-ro, Nonsan, Chungnam, 32992, Republic of Korea
| |
Collapse
|
18
|
Anand PK. From fat to fire: The lipid-inflammasome connection. Immunol Rev 2024. [PMID: 39327931 DOI: 10.1111/imr.13403] [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] [Indexed: 09/28/2024]
Abstract
Inflammasomes are multiprotein complexes that play a crucial role in regulating immune responses by governing the activation of Caspase-1, the secretion of pro-inflammatory cytokines, and the induction of inflammatory cell death, pyroptosis. The inflammasomes are pivotal in effective host defense against a range of pathogens. Yet, overt activation of inflammasome signaling can be detrimental. The most well-studied NLRP3 inflammasome has the ability to detect a variety of stimuli including pathogen-associated molecular patterns, environmental irritants, and endogenous stimuli released from dying cells. Additionally, NLRP3 acts as a key sensor of cellular homeostasis and can be activated by disturbances in diverse metabolic pathways. Consequently, NLRP3 is considered a key player linking metabolic dysregulation to numerous inflammatory disorders such as gout, diabetes, and atherosclerosis. Recently, compelling studies have highlighted a connection between lipids and the regulation of NLRP3 inflammasome. Lipids are integral to cellular processes that serve not only in maintaining the structural integrity and subcellular compartmentalization, but also in contributing to physiological equilibrium. Certain lipid species are known to define NLRP3 subcellular localization, therefore directly influencing the site of inflammasome assembly and activation. For instance, phosphatidylinositol 4-phosphate plays a crucial role in NLRP3 localization to the trans Golgi network. Moreover, new evidence has demonstrated the roles of lipid biosynthesis and trafficking in activation of the NLRP3 inflammasome. This review summarizes and discusses these emerging and varied roles of lipid metabolism in inflammasome activation. A deeper understanding of lipid-inflammasome interactions may open new avenues for therapeutic interventions to prevent or treat chronic inflammatory and autoimmune conditions.
Collapse
Affiliation(s)
- Paras K Anand
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
19
|
Pajonczyk D, Sternschulte MF, Soehnlein O, Bermudez M, Raabe CA, Rescher U. Comparative analysis of formyl peptide receptor 1 and formyl peptide receptor 2 reveals shared and preserved signalling profiles. Br J Pharmacol 2024. [PMID: 39294930 DOI: 10.1111/bph.17334] [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: 02/19/2024] [Revised: 07/03/2024] [Accepted: 08/06/2024] [Indexed: 09/21/2024] Open
Abstract
BACKGROUND AND PURPOSE The pattern recognition receptors, formyl peptide receptors, FPR1 and FPR2, are G protein-coupled receptors that recognize many different pathogen- and host-derived ligands. While FPR1 conveys pro-inflammatory signals, FPR2 is linked with pro-resolving outcomes. To analyse how the two very similar FPRs exert opposite effects in modulating inflammatory responses despite their high homology, a shared expression profile on immune cells and an overlapping ligand repertoire, we questioned whether the signalling profile differs between these two receptors. EXPERIMENTAL APPROACH We deduced EC50 and Emax values for synthetic, pathogen-derived and host-derived peptide agonists for both FPR1 and FPR2 and analysed them within the framework of biased signalling. We furthermore investigated whether FPR isoform-specific agonists affect the ex vivo lifespan of human neutrophils. KEY RESULTS The FPRs share a core signature across signalling pathways. Whereas the synthetic WKYMVm and formylated peptides acted as potent agonists at FPR1, and at FPR2, only WKYMVm was a full agonist. Natural FPR2 agonists, irrespective of N-terminal formylation, displayed lower activity ratios, suggesting an underutilized signalling potential of this receptor. FPR2 agonism did not counteract LPS-induced neutrophil survival, indicating that FPR2 activation per se is not linked with a pro-resolving function. CONCLUSION AND IMPLICATIONS Activation of FPR1 and FPR2 by a representative agonist panel revealed a lack of a receptor-specific signalling texture, challenging assumptions about distinct inflammatory profiles linked to specific receptor isoforms, signalling patterns or agonist classes. These conclusions are restricted to the specific agonists and signalling pathways examined.
Collapse
Affiliation(s)
- Denise Pajonczyk
- Research Group Cellular Biochemistry - Regulatory Mechanisms of Inflammation, Institute of Molecular Virology, Center of Molecular Biology of Inflammation and "Cells in Motion" Interfaculty Centre, University of Muenster, Muenster, Germany
| | - Merle F Sternschulte
- Research Group Cellular Biochemistry - Regulatory Mechanisms of Inflammation, Institute of Molecular Virology, Center of Molecular Biology of Inflammation and "Cells in Motion" Interfaculty Centre, University of Muenster, Muenster, Germany
- Institute of Experimental Pathology, Center of Molecular Biology of Inflammation, University of Muenster, Muenster, Germany
| | - Oliver Soehnlein
- Institute of Experimental Pathology, Center of Molecular Biology of Inflammation, University of Muenster, Muenster, Germany
| | - Marcel Bermudez
- Institute of Pharmaceutical and Medicinal Chemistry, University of Muenster, Muenster, Germany
| | - Carsten A Raabe
- Research Group Cellular Biochemistry - Regulatory Mechanisms of Inflammation, Institute of Molecular Virology, Center of Molecular Biology of Inflammation and "Cells in Motion" Interfaculty Centre, University of Muenster, Muenster, Germany
| | - Ursula Rescher
- Research Group Cellular Biochemistry - Regulatory Mechanisms of Inflammation, Institute of Molecular Virology, Center of Molecular Biology of Inflammation and "Cells in Motion" Interfaculty Centre, University of Muenster, Muenster, Germany
| |
Collapse
|
20
|
Henlon Y, Panir K, McIntyre I, Hogg C, Dhami P, Cuff AO, Senior A, Moolchandani-Adwani N, Courtois ET, Horne AW, Rosser M, Ott S, Greaves E. Single-cell analysis identifies distinct macrophage phenotypes associated with prodisease and proresolving functions in the endometriotic niche. Proc Natl Acad Sci U S A 2024; 121:e2405474121. [PMID: 39255000 PMCID: PMC11420174 DOI: 10.1073/pnas.2405474121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 07/24/2024] [Indexed: 09/11/2024] Open
Abstract
Endometriosis negatively impacts the health-related quality of life of 190 million women worldwide. Novel advances in nonhormonal treatments for this debilitating condition are desperately needed. Macrophages play a vital role in the pathophysiology of endometriosis and represent a promising therapeutic target. In the current study, we revealed the full transcriptomic complexity of endometriosis-associated macrophage subpopulations using single-cell analyses in a preclinical mouse model of experimental endometriosis. We have identified two key lesion-resident populations that resemble i) tumor-associated macrophages (characterized by expression of Folr2, Mrc1, Gas6, and Ccl8+) that promoted expression of Col1a1 and Tgfb1 in human endometrial stromal cells and increased angiogenic meshes in human umbilical vein endothelial cells, and ii) scar-associated macrophages (Mmp12, Cd9, Spp1, Trem2+) that exhibited a phenotype associated with fibrosis and matrix remodeling. We also described a population of proresolving large peritoneal macrophages that align with a lipid-associated macrophage phenotype (Apoe, Saa3, Pid1) concomitant with altered lipid metabolism and cholesterol efflux. Gain of function experiments using an Apoe mimetic resulted in decreased lesion size and fibrosis, and modification of peritoneal macrophage populations in the preclinical model. Using cross-species analysis of mouse and human single-cell datasets, we determined the concordance of peritoneal and lesion-resident macrophage subpopulations, identifying key similarities and differences in transcriptomic phenotypes. Ultimately, we envisage that these findings will inform the design and use of specific macrophage-targeted therapies and open broad avenues for the treatment of endometriosis.
Collapse
Affiliation(s)
- Yasmin Henlon
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, CoventryCV4 7AL, United Kingdom
- Centre for Early Life, University of Warwick, CoventryCV4 7AL, United Kingdom
| | - Kavita Panir
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, CoventryCV4 7AL, United Kingdom
- Centre for Early Life, University of Warwick, CoventryCV4 7AL, United Kingdom
| | - Iona McIntyre
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, CoventryCV4 7AL, United Kingdom
- Centre for Early Life, University of Warwick, CoventryCV4 7AL, United Kingdom
| | - Chloe Hogg
- Centre for Reproductive Health, Institute of Regeneration and Repair, The University of Edinburgh, EdinburghEH16 4UU, United Kingdom
| | - Priya Dhami
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, CoventryCV4 7AL, United Kingdom
- Centre for Early Life, University of Warwick, CoventryCV4 7AL, United Kingdom
| | - Antonia O. Cuff
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, CoventryCV4 7AL, United Kingdom
- Centre for Early Life, University of Warwick, CoventryCV4 7AL, United Kingdom
| | - Anna Senior
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, CoventryCV4 7AL, United Kingdom
| | - Niky Moolchandani-Adwani
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, CoventryCV4 7AL, United Kingdom
- Centre for Early Life, University of Warwick, CoventryCV4 7AL, United Kingdom
| | - Elise T. Courtois
- Single Cell Biology Lab, The Jackson Laboratory for Genomic Medicine, Farmington, CT06032
| | - Andrew W. Horne
- Centre for Reproductive Health, Institute of Regeneration and Repair, The University of Edinburgh, EdinburghEH16 4UU, United Kingdom
| | - Matthew Rosser
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, CoventryCV4 7AL, United Kingdom
- Centre for Early Life, University of Warwick, CoventryCV4 7AL, United Kingdom
| | - Sascha Ott
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, CoventryCV4 7AL, United Kingdom
- Centre for Early Life, University of Warwick, CoventryCV4 7AL, United Kingdom
| | - Erin Greaves
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, CoventryCV4 7AL, United Kingdom
- Centre for Early Life, University of Warwick, CoventryCV4 7AL, United Kingdom
| |
Collapse
|
21
|
Lesouhaitier M, Belicard F, Tadié JM. Cardiopulmonary bypass and VA-ECMO induced immune dysfunction: common features and differences, a narrative review. Crit Care 2024; 28:300. [PMID: 39256830 PMCID: PMC11389086 DOI: 10.1186/s13054-024-05058-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 08/06/2024] [Indexed: 09/12/2024] Open
Abstract
Cardiopulmonary bypass (CPB) and veno-arterial extracorporeal membrane oxygenation are critical tools in contemporary cardiac surgery and intensive care, respectively. While these techniques share similar components, their application contexts differ, leading to distinct immune dysfunctions which could explain the higher incidence of nosocomial infections among ECMO patients compared to those undergoing CPB. This review explores the immune modifications induced by these techniques, comparing their similarities and differences, and discussing potential treatments to restore immune function and prevent infections. The immune response to CPB and ECMO involves both humoral and cellular components. The kinin system, complement system, and coagulation cascade are rapidly activated upon blood contact with the circuit surfaces, leading to the release of pro-inflammatory mediators. Ischemia-reperfusion injury and the release of damage-associated molecular patterns further exacerbate the inflammatory response. Cellular responses involve platelets, neutrophils, monocytes, dendritic cells, B and T lymphocytes, and myeloid-derived suppressor cells, all of which undergo phenotypic and functional alterations, contributing to immunoparesis. Strategies to mitigate immune dysfunctions include reducing the inflammatory response during CPB/ECMO and enhancing immune functions. Approaches such as off-pump surgery, corticosteroids, complement inhibitors, leukocyte-depleting filters, and mechanical ventilation during CPB have shown varying degrees of success in clinical trials. Immunonutrition, particularly arginine supplementation, has also been explored with mixed results. These strategies aim to balance the inflammatory response and support immune function, potentially reducing infection rates and improving outcomes. In conclusion, both CPB and ECMO trigger significant immune alterations that increase susceptibility to nosocomial infections. Addressing these immune dysfunctions through targeted interventions is essential to improving patient outcomes in cardiac surgery and critical care settings. Future research should focus on refining these strategies and developing new approaches to better manage the immune response in patients undergoing CPB and ECMO.
Collapse
Affiliation(s)
- Mathieu Lesouhaitier
- Infectious Diseases and Intensive Care Unit, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France.
- SITI, Pole de Biologie, Pontchaillou University Hospital, Etablissement Français du Sang Bretagne, 2 rue Henri Le Guilloux, 35033, Rennes, France.
- UMR 1236, Univ Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France.
- CIC-1414, INSERM, Rennes, France.
| | - Félicie Belicard
- Infectious Diseases and Intensive Care Unit, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France
- SITI, Pole de Biologie, Pontchaillou University Hospital, Etablissement Français du Sang Bretagne, 2 rue Henri Le Guilloux, 35033, Rennes, France
- UMR 1236, Univ Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Jean-Marc Tadié
- Infectious Diseases and Intensive Care Unit, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France.
- SITI, Pole de Biologie, Pontchaillou University Hospital, Etablissement Français du Sang Bretagne, 2 rue Henri Le Guilloux, 35033, Rennes, France.
- UMR 1236, Univ Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France.
- CIC-1414, INSERM, Rennes, France.
| |
Collapse
|
22
|
Kircheis R, Planz O. Special Issue "The Role of Toll-Like Receptors (TLRs) in Infection and Inflammation 2.0". Int J Mol Sci 2024; 25:9709. [PMID: 39273656 PMCID: PMC11396464 DOI: 10.3390/ijms25179709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
Toll-like receptors (TLRs) are key players in the innate immune system, in host' first-line defense against pathogens [...].
Collapse
Affiliation(s)
| | - Oliver Planz
- Institute of Cell Biology and Immunology, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany
| |
Collapse
|
23
|
Liao Y, Zhang W, Zhou M, Zhu C, Zou Z. Ubiquitination in pyroptosis pathway: A potential therapeutic target for sepsis. Cytokine Growth Factor Rev 2024:S1359-6101(24)00068-6. [PMID: 39294049 DOI: 10.1016/j.cytogfr.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 09/20/2024]
Abstract
Sepsis remains a significant clinical challenge, causing numerous deaths annually and representing a major global health burden. Pyroptosis, a unique form of programmed cell death characterized by cell lysis and the release of inflammatory mediators, is a crucial factor in the pathogenesis and progression of sepsis, septic shock, and organ dysfunction. Ubiquitination, a key post-translational modification influencing protein fate, has emerged as a promising target for managing various inflammatory conditions, including sepsis. This review integrates the current knowledge on sepsis, pyroptosis, and the ubiquitin system, focusing on the molecular mechanisms of ubiquitination within pyroptotic pathways activated during sepsis. By exploring how modulating ubiquitination can regulate pyroptosis and its associated inflammatory signaling pathways, this review provides insights into potential therapeutic strategies for sepsis, highlighting the need for further research into these complex molecular networks.
Collapse
Affiliation(s)
- Yan Liao
- School of Anesthesiology, Naval Medical University, Shanghai 200433, China
| | - Wangzheqi Zhang
- School of Anesthesiology, Naval Medical University, Shanghai 200433, China
| | - Miao Zhou
- Department of Anesthesiology, the Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University, Nanjing, Jiangsu 210009, China
| | - Chenglong Zhu
- School of Anesthesiology, Naval Medical University, Shanghai 200433, China.
| | - Zui Zou
- School of Anesthesiology, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
24
|
Al-Madhagi H, Muhammed MT. Targeting COVID-19 and varicocele by blocking inflammasome: Ligand-based virtual screening. Arch Biochem Biophys 2024; 759:110107. [PMID: 39074718 DOI: 10.1016/j.abb.2024.110107] [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: 05/12/2024] [Revised: 07/22/2024] [Accepted: 07/26/2024] [Indexed: 07/31/2024]
Abstract
COVID-19 is a new generation of outbreaks that invade not only local emerging region, continental but also the whole globe. Varicocele on the other hand, is a testicular vascular disease that underlies 40 % of male infertility cases. Fortunately, the two diseases can be blocked through targeting one common target, NLRP3 inflammasome. Upon searching for similar drugs that gained FDA-approval in ChEMBL library along with examining their potential blockade of the receptor through docking using CB-DOCK-2, three potential approved drugs can be repurposed, ChEMBL 4297185, ChEMBL 1201749, ChEMBL 1200545 which had binding energy of -9.8 and -9.7 kcal/mol (stronger than the reference inhibitor, -9.3 kcal/mol). Also, ADME profile of the top 3 drugs showed better attributes. Also, the simulated proteins exhibited stable pattern with strong free binding energies. Among the potential inhibitor drugs ChEMBL 4297185 was found to remain inside the binding site of the protein during the 200 ns simulation time. Hence, it is anticipated to have the highest binding and thus inhibition potential against the protein. The suggested drugs, especially ChEMBL 4297185, are potentially repurposable toward treating COVID-19 and varicocele which deserve further experimental validation.
Collapse
Affiliation(s)
| | - Muhammed Tilahun Muhammed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Suleyman Demirel University, Isparta, Turkiye.
| |
Collapse
|
25
|
Galli F, Bartolini D, Ronco C. Oxidative stress, defective proteostasis and immunometabolic complications in critically ill patients. Eur J Clin Invest 2024; 54:e14229. [PMID: 38676423 DOI: 10.1111/eci.14229] [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: 02/22/2024] [Revised: 03/31/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024]
Abstract
Oxidative stress (OS) develops in critically ill patients as a metabolic consequence of the immunoinflammatory and degenerative processes of the tissues. These induce increased and/or dysregulated fluxes of reactive species enhancing their pro-oxidant activity and toxicity. At the same time, OS sustains its own inflammatory and immunometabolic pathogenesis, leading to a pervasive and vitious cycle of events that contribute to defective immunity, organ dysfunction and poor prognosis. Protein damage is a key player of these OS effects; it generates increased levels of protein oxidation products and misfolded proteins in both the cellular and extracellular environment, and contributes to forms DAMPs and other proteinaceous material to be removed by endocytosis and proteostasis processes of different cell types, as endothelial cells, tissue resident monocytes-macrophages and peripheral immune cells. An excess of OS and protein damage in critical illness can overwhelm such cellular processes ultimately interfering with systemic proteostasis, and consequently with innate immunity and cell death pathways of the tissues thus sustaining organ dysfunction mechanisms. Extracorporeal therapies based on biocompatible/bioactive membranes and new adsorption techniques may hold some potential in reducing the impact of OS on the defective proteostasis of patients with critical illness. These can help neutralizing reactive and toxic species, also removing solutes in a wide spectrum of molecular weights thus improving proteostasis and its immunometabolic corelates. Pharmacological therapy is also moving steps forward which could help to enhance the efficacy of extracorporeal treatments. This narrative review article explores the aspects behind the origin and pathogenic role of OS in intensive care and critically ill patients, with a focus on protein damage as a cause of impaired systemic proteostasis and immune dysfunction in critical illness.
Collapse
Affiliation(s)
- Francesco Galli
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Desirée Bartolini
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Claudio Ronco
- Department of Medicine, International Renal Research Institute of Vicenza, University of Padova, San Bortolo Hospital Vicenza, Vicenza, Italy
| |
Collapse
|
26
|
Zhi L, Wang X, Pan X, Han C. The asynchronous dynamic changes and interrelationships between leukocyte composition and inflammatory markers and potential clinical significance in the early stage and sepsis stage in severe burns. Burns 2024; 50:1752-1761. [PMID: 38724345 DOI: 10.1016/j.burns.2024.04.006] [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: 11/24/2023] [Revised: 02/25/2024] [Accepted: 04/10/2024] [Indexed: 08/30/2024]
Abstract
OBJECTIVE The aim of this study is to investigate the dynamic changes and interrelationships between leukocyte components and inflammatory markers in the early stages and sepsis stage in severe burns, and explore their potential clinical significance. METHODS This is a 5-year retrospective cohort study involving 107 patients with severe burns (the total body surface area of burn (TBSA) > 50%), in which, neutrophil count, lymphocyte count, monocyte count, the ratio of the product of monocyte and lymphocyte count to neutrophil count (MLPN), procalcitonin (PCT), C-reactive protein (CRP), capillary leakage index (CLI) and creatinine (Scr) were investigated. RESULTS Within one week after injury, the leukocyte components and MLPN showed a V-shaped change, with a peak immediately after injury and a trough on the 4th or 5th day after injury, while CRP showed a continuous upward trend, and the leukocyte compositions of all patients were negatively correlated with CRP values. The counts of leukocytes components and CRP values in deceased patients were higher than those in surviving patients within 2 days after injury. In the resorption stage, although no significant difference in lymphocyte and monocyte counts between surviving and deceased patients was found, the monocyte and lymphocyte counts in deceased patients were lower than those in surviving patients on the 5th to 7th day after injury, while neutrophils counts and CRP values remained higher than those in surviving patients. And the dynamic changes of MLPN were consistent with those of leukocyte compositions and opposite to those of PCT values. Moreover, MLPN were negatively correlated with CRP, CLI, and Scr values in the early stage of severe burns. In sepsis stage, as the condition worsened, the values of CRP, PCT and neutrophil counts continuously increased with varying degree, while lymphocyte and monocyte counts, and MLPN showed continuously decrease, but rebounded to increase before death. And the occurrence of the trough of monocyte counts was earlier than that of lymphocyte counts, a negative correlation between neutrophil counts and PCT values was found. CONCLUSION The results of this study revealed the dynamic interrelationships between leucocyte components and inflammatory indicators in the early stages and sepsis stage in severe burns, reflecting the different weightings of inflammatory responses and immune dysfunction in different disease stages and its correlation with outcomes, which providing useful clinical information for dynamic immunomodulatory therapy. Moreover, dynamic monitoring of MLPN value can provide timely information for clinical evaluation.
Collapse
Affiliation(s)
- Lizhu Zhi
- Department of Burn Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.
| | - Xingang Wang
- Department of Burn Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xuanliang Pan
- Department of Burn Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Chunmao Han
- Department of Burn Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
27
|
Lee HT, Lin CS, Liu CY, Chen P, Tsai CY, Wei YH. Mitochondrial Plasticity and Glucose Metabolic Alterations in Human Cancer under Oxidative Stress-From Viewpoints of Chronic Inflammation and Neutrophil Extracellular Traps (NETs). Int J Mol Sci 2024; 25:9458. [PMID: 39273403 PMCID: PMC11395599 DOI: 10.3390/ijms25179458] [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: 07/24/2024] [Revised: 08/20/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Oxidative stress elicited by reactive oxygen species (ROS) and chronic inflammation are involved both in deterring and the generation/progression of human cancers. Exogenous ROS can injure mitochondria and induce them to generate more endogenous mitochondrial ROS to further perpetuate the deteriorating condition in the affected cells. Dysfunction of these cancer mitochondria may possibly be offset by the Warburg effect, which is characterized by amplified glycolysis and metabolic reprogramming. ROS from neutrophil extracellular traps (NETs) are an essential element for neutrophils to defend against invading pathogens or to kill cancer cells. A chronic inflammation typically includes consecutive NET activation and tissue damage, as well as tissue repair, and together with NETs, ROS would participate in both the destruction and progression of cancers. This review discusses human mitochondrial plasticity and the glucose metabolic reprogramming of cancer cells confronting oxidative stress by the means of chronic inflammation and neutrophil extracellular traps (NETs).
Collapse
Affiliation(s)
- Hui-Ting Lee
- Division of Allergy, Immunology & Rheumatology, Department of Internal Medicine, Mackay Memorial Hospital, Taipei 104, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei City 252, Taiwan
| | - Chen-Sung Lin
- Division of Thoracic Surgery, Department of Surgery, Taipei Hospital, Ministry of Health and Welfare, New Taipei City 242, Taiwan
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Center for General Education, Kainan University, Taoyuan City 338, Taiwan
| | - Chao-Yu Liu
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Division of Thoracic Surgery, Department of Surgery, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan
| | - Po Chen
- Cancer Free Biotech, Taipei 114, Taiwan
| | - Chang-Youh Tsai
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Clinical Trial Center, Division of Immunology & Rheumatology, Fu Jen Catholic University Hospital, New Taipei City 243, Taiwan
- Faculty of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
| | - Yau-Huei Wei
- Department of Medicine, Mackay Medical College, New Taipei City 252, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Center for Mitochondrial Medicine and Free Radical Research, Changhua Christian Hospital, Changhua City 500, Taiwan
| |
Collapse
|
28
|
Rajasekaran JJ, Krishnamurthy HK, Bosco J, Jayaraman V, Krishna K, Wang T, Bei K. Oral Microbiome: A Review of Its Impact on Oral and Systemic Health. Microorganisms 2024; 12:1797. [PMID: 39338471 PMCID: PMC11434369 DOI: 10.3390/microorganisms12091797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/07/2024] [Accepted: 08/16/2024] [Indexed: 09/30/2024] Open
Abstract
PURPOSE OF REVIEW This review investigates the oral microbiome's composition, functions, influencing factors, connections to oral and systemic diseases, and personalized oral care strategies. RECENT FINDINGS The oral microbiome is a complex ecosystem consisting of bacteria, fungi, archaea, and viruses that contribute to oral health. Various factors, such as diet, smoking, alcohol consumption, lifestyle choices, and medical conditions, can affect the balance of the oral microbiome and lead to dysbiosis, which can result in oral health issues like dental caries, gingivitis, periodontitis, oral candidiasis, and halitosis. Importantly, our review explores novel associations between the oral microbiome and systemic diseases including gastrointestinal, cardiovascular, endocrinal, and neurological conditions, autoimmune diseases, and cancer. We comprehensively review the efficacy of interventions like dental probiotics, xylitol, oral rinses, fluoride, essential oils, oil pulling, and peptides in promoting oral health by modulating the oral microbiome. SUMMARY This review emphasizes the critical functions of the oral microbiota in dental and overall health, providing insights into the effects of microbial imbalances on various diseases. It underlines the significant connection between the oral microbiota and general health. Furthermore, it explores the advantages of probiotics and other dental care ingredients in promoting oral health and addressing common oral issues, offering a comprehensive strategy for personalized oral care.
Collapse
Affiliation(s)
- John J. Rajasekaran
- Vibrant Sciences LLC, Santa Clara, CA 95054, USA; (H.K.K.); (V.J.); (K.K.); (T.W.); (K.B.)
| | | | - Jophi Bosco
- Vibrant America LLC, Santa Clara, CA 95054, USA;
| | - Vasanth Jayaraman
- Vibrant Sciences LLC, Santa Clara, CA 95054, USA; (H.K.K.); (V.J.); (K.K.); (T.W.); (K.B.)
| | - Karthik Krishna
- Vibrant Sciences LLC, Santa Clara, CA 95054, USA; (H.K.K.); (V.J.); (K.K.); (T.W.); (K.B.)
| | - Tianhao Wang
- Vibrant Sciences LLC, Santa Clara, CA 95054, USA; (H.K.K.); (V.J.); (K.K.); (T.W.); (K.B.)
| | - Kang Bei
- Vibrant Sciences LLC, Santa Clara, CA 95054, USA; (H.K.K.); (V.J.); (K.K.); (T.W.); (K.B.)
| |
Collapse
|
29
|
Botella Lucena P, Heneka MT. Inflammatory aspects of Alzheimer's disease. Acta Neuropathol 2024; 148:31. [PMID: 39196440 DOI: 10.1007/s00401-024-02790-2] [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: 04/26/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 08/29/2024]
Abstract
Alzheimer´s disease (AD) stands out as the most common chronic neurodegenerative disorder. AD is characterized by progressive cognitive decline and memory loss, with neurodegeneration as its primary pathological feature. The role of neuroinflammation in the disease course has become a focus of intense research. While microglia, the brain's resident macrophages, have been pivotal to study central immune inflammation, recent evidence underscores the contributions of other cellular entities to the neuroinflammatory process. In this article, we review the inflammatory role of microglia and astrocytes, focusing on their interactions with AD's core pathologies, amyloid beta deposition, and tau tangle formation. Additionally, we also discuss how different modes of regulated cell death in AD may impact the chronic neuroinflammatory environment. This review aims to highlight the evolving landscape of neuroinflammatory research in AD and underscores the importance of considering multiple cellular contributors when developing new therapeutic strategies.
Collapse
Affiliation(s)
- Pablo Botella Lucena
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6, Avenue du Swing, Belvaux, L-4367, Esch-Belval, Luxembourg
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6, Avenue du Swing, Belvaux, L-4367, Esch-Belval, Luxembourg.
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
30
|
Prud’homme GJ, Wang Q. Anti-Inflammatory Role of the Klotho Protein and Relevance to Aging. Cells 2024; 13:1413. [PMID: 39272986 PMCID: PMC11394293 DOI: 10.3390/cells13171413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/17/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
The α-Klotho protein (hereafter Klotho) is an obligate coreceptor for fibroblast growth factor 23 (FGF23). It is produced in the kidneys, brain and other sites. Klotho insufficiency causes hyperphosphatemia and other anomalies. Importantly, it is associated with chronic pathologies (often age-related) that have an inflammatory component. This includes atherosclerosis, diabetes and Alzheimer's disease. Its mode of action in these diseases is not well understood, but it inhibits or regulates multiple major pathways. Klotho has a membrane form and a soluble form (s-Klotho). Cytosolic Klotho is postulated but not well characterized. s-Klotho has endocrine properties that are incompletely elucidated. It binds to the FGF receptor 1c (FGFR1c) that is widely expressed (including endothelial cells). It also attaches to soluble FGF23, and FGF23/Klotho binds to FGFRs. Thus, s-Klotho might be a roaming FGF23 coreceptor, but it has other functions. Notably, Klotho (cell-bound or soluble) counteracts inflammation and appears to mitigate related aging (inflammaging). It inhibits NF-κB and the NLRP3 inflammasome. This inflammasome requires priming by NF-κB and produces active IL-1β, membrane pores and cell death (pyroptosis). In accord, Klotho countered inflammation and cell injury induced by toxins, damage-associated molecular patterns (DAMPs), cytokines, and reactive oxygen species (ROS). s-Klotho also blocks the TGF-β receptor and Wnt ligands, which lessens fibrotic disease. Low Klotho is associated with loss of muscle mass (sarcopenia), as occurs in aging and chronic diseases. s-Klotho counters the inhibitory effects of myostatin and TGF-β on muscle, reduces inflammation, and improves muscle repair following injury. The inhibition of TGF-β and other factors may also be protective in diabetic retinopathy and age-related macular degeneration (AMD). This review examines Klotho functions especially as related to inflammation and potential applications.
Collapse
Affiliation(s)
- Gérald J. Prud’homme
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 220 Walmer Rd, Toronto, ON M5R 3R7, Canada
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
| | - Qinghua Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai 200030, China
- Shanghai Innogen Pharmaceutical Co., Ltd., Shanghai 201318, China
| |
Collapse
|
31
|
Javed MJ, Howard RM, Li H, Carrasco L, Dirain MLS, Su G, Cai G, Upchurch GR, Jiang Z. Gasdermin D deficiency attenuates development of ascending aortic dissections in a novel mouse model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.22.609270. [PMID: 39229014 PMCID: PMC11370574 DOI: 10.1101/2024.08.22.609270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Background Thoracic aortic dissection (TAD) is a silent killer. Approximately two-thirds of the cases occur in the ascending aorta (i.e. type A dissection) and majority of them are unrelated to genetic mutations. However, animal models of spontaneous type A dissection are not widely available. In the present study, a novel mouse TAD model was created. Further, the role of gasdermin D (GSDMD) in TAD development was evaluated. Methods TADs were created by treating ascending aorta of adult mice (C57BL/6J) with active elastase (40.0 U/ml) and β-aminopropionitrile (Act E+BAPN). The temporal progress of the TAD pathology was rigorously characterized by histological evaluation and scanning electron microscopy, while potential mechanisms explored with bulk RNA sequencing of specimens collected at multiple timepoints. With this novel TAD model, further experiments were performed with Gsdmd -/- mice to evaluate its impact on TAD formation. Results The ascending aorta challenged with Act E+BAPN developed pathology characterized by an early onset of intimomedial tears (complete penetration) and intramural hematoma, followed by progressive medial loss and aortic dilation. Ingenuity Pathway Analysis and functional annotation of differentially expressed genes suggested that a unique inflammatory micro-environment, rather than general inflammation, promoted the onset of TADs by specifically recruiting neutrophils to the aortic wall, while the pathology at the advanced stage was driven by T-cell mediated immune injury. Gsdmd -/- attenuated medial loss, adventitial fibrosis, and dilation of TADs. This protective effect was associated with a reduced number of TUNEL (terminal deoxynucleotidyl transferase dUTP nick end labeling) positive cells and T-cells in TADs. Conclusions A novel mouse TAD model was created in the ascending aorta. It produces a unique microenvironment to activate different immune cell subsets, promoting onset and subsequent remodeling of TADs. Consistently, Gsdmd -/- attenuates TAD development, with modulation of cell death and T-cell response likely acting as the underlying mechanism.
Collapse
|
32
|
Yesitayi G, Wang Q, Wang M, Ainiwan M, Kadier K, Aizitiaili A, Ma Y, Ma X. LPS-LBP complex induced endothelial cell pyroptosis in aortic dissection is associated with gut dysbiosis. Microbes Infect 2024:105406. [PMID: 39168178 DOI: 10.1016/j.micinf.2024.105406] [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: 01/22/2024] [Revised: 08/04/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024]
Abstract
Acute aortic dissection (AAD) is the most severe traumatic disease affecting the aorta. Pyroptosis-mediated vascular wall inflammation is a crucial trigger for AAD, and the exact mechanism requires further investigation. In this study, our proteomic analysis showed that Lipopolysaccharide (LPS)-binding protein (LBP) was significantly upregulated in the plasma and aortic tissue of patients with AAD. Further, 16S rRNA sequencing of stool samples suggested that patients with AAD exhibit gut dysbiosis, which may lead to an impaired intestinal barrier and LPS leakage. By comparing with control mice, we found that LBP, including Pyrin Domain Containing Protein3 (NLRP3), the CARD-containing adapter apoptosis-associated speck-like protein (ASC), and Cleaved caspase-1, were upregulated in the AAD aorta, whereas gut intestinal barrier-related proteins were downregulated. Moreover, treated with LBPK95A (an LBP inhibitor) attenuated the incidence of AAD, the expression levels of pyroptosis-related factors, and the extent of vascular pathological changes compared to those in AAD mice. In addition, LPS and LBP treatment of human umbilical vein endothelial cells (HUVECs) activated TLR4 signaling and intracellular reactive oxygen species (ROS) production, which stimulated NLRP3 inflammasome formation and mediated pyroptosis in endothelial cells. Our findings showed that gut dysbiosis mediates pyroptosis by the LPS-LBP complex, thus providing new insights into developing AAD.
Collapse
Affiliation(s)
- Gulinazi Yesitayi
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Medical University, Ürümqi, China.
| | - Qi Wang
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| | - Mengmeng Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China.
| | - Mierxiati Ainiwan
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Medical University, Ürümqi, China.
| | - Kaisaierjiang Kadier
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Medical University, Ürümqi, China.
| | - Aliya Aizitiaili
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Medical University, Ürümqi, China.
| | - Yitong Ma
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Medical University, Ürümqi, China.
| | - Xiang Ma
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Medical University, Ürümqi, China.
| |
Collapse
|
33
|
Kong C, Yang M, Yue N, Zhang Y, Tian C, Wei D, Shi R, Yao J, Wang L, Li D. Restore Intestinal Barrier Integrity: An Approach for Inflammatory Bowel Disease Therapy. J Inflamm Res 2024; 17:5389-5413. [PMID: 39161679 PMCID: PMC11330754 DOI: 10.2147/jir.s470520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 07/02/2024] [Indexed: 08/21/2024] Open
Abstract
The intestinal barrier maintained by various types of columnar epithelial cells, plays a crucial role in regulating the interactions between the intestinal contents (such as the intestinal microbiota), the immune system, and other components. Dysfunction of the intestinal mucosa is a significant pathophysiological mechanism and clinical manifestation of inflammatory bowel disease (IBD). However, current therapies for IBD primarily focus on suppressing inflammation, and no disease-modifying treatments specifically target the epithelial barrier. Given the side effects associated with chronic immunotherapy, effective alternative therapies that promote mucosal healing are highly attractive. In this review, we examined the function of intestinal epithelial barrier function and the mechanisms of behind its disruption in IBD. We illustrated the complex process of intestinal mucosal healing and proposed therapeutic approaches to promote mucosal healing strategies in IBD. These included the application of stem cell transplantation and organ-like tissue engineering approaches to generate new intestinal tissue. Finally, we discussed potential strategies to restore the function of the intestinal barrier as a treatment for IBD.
Collapse
Affiliation(s)
- Chen Kong
- The Second Clinical Medical College, Jinan University; Shenzhen, Guangdong, People’s Republic of China
| | - Meifeng Yang
- Department of Hematology, Yantian District People’s Hospital, Shenzhen, Guangdong, People’s Republic of China
| | - Ningning Yue
- Department of Gastroenterology, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, People’s Republic of China
| | - Yuan Zhang
- Department of Medical Administration, Huizhou Institute of Occupational Diseases Control and Prevention, Huizhou, Guangdong, People’s Republic of China
| | - Chengmei Tian
- Department of Emergency, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, People’s Republic of China
| | - Daoru Wei
- Department of Rehabilitation, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, People’s Republic of China
| | - Ruiyue Shi
- The Second Clinical Medical College, Jinan University; Shenzhen, Guangdong, People’s Republic of China
| | - Jun Yao
- The Second Clinical Medical College, Jinan University; Shenzhen, Guangdong, People’s Republic of China
| | - Lisheng Wang
- The Second Clinical Medical College, Jinan University; Shenzhen, Guangdong, People’s Republic of China
| | - Defeng Li
- The Second Clinical Medical College, Jinan University; Shenzhen, Guangdong, People’s Republic of China
| |
Collapse
|
34
|
Vana F, Szabo Z, Masarik M, Kratochvilova M. The interplay of transition metals in ferroptosis and pyroptosis. Cell Div 2024; 19:24. [PMID: 39097717 PMCID: PMC11297737 DOI: 10.1186/s13008-024-00127-9] [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: 03/26/2024] [Accepted: 07/08/2024] [Indexed: 08/05/2024] Open
Abstract
Cell death is one of the most important mechanisms of maintaining homeostasis in our body. Ferroptosis and pyroptosis are forms of necrosis-like cell death. These cell death modalities play key roles in the pathophysiology of cancer, cardiovascular, neurological diseases, and other pathologies. Transition metals are abundant group of elements in all living organisms. This paper presents a summary of ferroptosis and pyroptosis pathways and their connection to significant transition metals, namely zinc (Zn), copper (Cu), molybdenum (Mo), lead (Pb), cobalt (Co), iron (Fe), cadmium (Cd), nickel (Ni), mercury (Hg), uranium (U), platinum (Pt), and one crucial element, selenium (Se). Authors aim to summarize the up-to-date knowledge of this topic.In this review, there are categorized and highlighted the most common patterns in the alterations of ferroptosis and pyroptosis by transition metals. Special attention is given to zinc since collected data support its dual nature of action in both ferroptosis and pyroptosis. All findings are presented together with a brief description of major biochemical pathways involving mentioned metals and are visualized in attached comprehensive figures.This work concludes that the majority of disruptions in the studied metals' homeostasis impacts cell fate, influencing both death and survival of cells in the complex system of altered pathways. Therefore, this summary opens up the space for further research.
Collapse
Affiliation(s)
- Frantisek Vana
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, CZ-625 00, Czech Republic
| | - Zoltan Szabo
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, CZ-625 00, Czech Republic
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty kopec 7, Brno, 656 53, Czech Republic
| | - Michal Masarik
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, CZ-625 00, Czech Republic
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, CZ-625 00, Czech Republic
- First Faculty of Medicine, BIOCEV, Charles University, Prumyslova 595, Vestec, CZ-252 50, Czech Republic
| | - Monika Kratochvilova
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, CZ-625 00, Czech Republic.
| |
Collapse
|
35
|
Liang W, Liang B, Yan K, Zhang G, Zhuo J, Cai Y. Low-Intensity Pulsed Ultrasound: A Physical Stimulus with Immunomodulatory and Anti-inflammatory Potential. Ann Biomed Eng 2024; 52:1955-1981. [PMID: 38683473 DOI: 10.1007/s10439-024-03523-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 04/20/2024] [Indexed: 05/01/2024]
Abstract
Ultrasound has expanded into the therapeutic field as a medical imaging and diagnostic technique. Low-intensity pulsed ultrasound (LIPUS) is a kind of therapeutic ultrasound that plays a vital role in promoting fracture healing, wound repair, immunomodulation, and reducing inflammation. Its anti-inflammatory effects are manifested by decreased pro-inflammatory cytokines and chemokines, accelerated regression of immune cell invasion, and accelerated damage repair. Although the anti-inflammatory mechanism of LIPUS is not very clear, many in vitro and in vivo studies have shown that LIPUS may play its anti-inflammatory role by activating signaling pathways such as integrin/Focal adhesion kinase (FAK)/Phosphatidylinositol 3-kinase (PI3K)/Serine threonine kinase (Akt), Vascular endothelial growth factor (VEGF)/endothelial nitric oxide synthase (eNOS), or inhibiting signaling pathways such as Toll-like receptors (TLRs)/Nuclear factor kappa-B (NF-κB) and p38-Mitogen-activated protein kinase (MAPK). As a non-invasive physical therapy, the anti-inflammatory and immunomodulatory effects of LIPUS deserve further exploration.
Collapse
Affiliation(s)
- Wenxin Liang
- Center of Medicine Clinical Research, Department of Pharmacy, Medical Supplies Center of Chinese PLA General Hospital, 28 Fu Xing Road, Beijing, 100853, People's Republic of China
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Beibei Liang
- Center of Medicine Clinical Research, Department of Pharmacy, Medical Supplies Center of Chinese PLA General Hospital, 28 Fu Xing Road, Beijing, 100853, People's Republic of China
| | - Kaicheng Yan
- Center of Medicine Clinical Research, Department of Pharmacy, Medical Supplies Center of Chinese PLA General Hospital, 28 Fu Xing Road, Beijing, 100853, People's Republic of China
| | - Guanxuanzi Zhang
- Center of Medicine Clinical Research, Department of Pharmacy, Medical Supplies Center of Chinese PLA General Hospital, 28 Fu Xing Road, Beijing, 100853, People's Republic of China
| | - Jiaju Zhuo
- Center of Medicine Clinical Research, Department of Pharmacy, Medical Supplies Center of Chinese PLA General Hospital, 28 Fu Xing Road, Beijing, 100853, People's Republic of China
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Yun Cai
- Center of Medicine Clinical Research, Department of Pharmacy, Medical Supplies Center of Chinese PLA General Hospital, 28 Fu Xing Road, Beijing, 100853, People's Republic of China.
| |
Collapse
|
36
|
Chen Y, Huang J, Li Y, Chen Y, Gong Z, Xu M, Ma Y, Hu D, Peng X, Xu G, Cai S, Liu L, Zhao W, Zhao H. Bongkrekic acid alleviates airway inflammation via breaking the mPTP/mtDAMPs/RAGE feedback loop in a steroid-insensitive asthma model. Biomed Pharmacother 2024; 177:117111. [PMID: 39013220 DOI: 10.1016/j.biopha.2024.117111] [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: 04/11/2024] [Revised: 07/04/2024] [Accepted: 07/07/2024] [Indexed: 07/18/2024] Open
Abstract
Mitochondrial dysfunction is critical in the pathogenesis of asthma. Mitochondrial permeability transition pore (mPTP) regulates the release of mitochondrial damage-associated molecular patterns (mtDAMPs) to maintain mitochondrial homeostasis. Bongkrekic acid (BKA) is a highly selective inhibitor of mPTP opening, participates the progression of various diseases. This research investigated the exact roles of BKA and mPTP in the pathogenesis of asthma and elucidated its underlying mechanisms. In the present study, cytochrome c, one of the mtDAMPs, levels were elevated in asthmatic patients, and associated to airway inflammation and airway obstruction. BKA, the inhibitor of mPTP markedly reversed TDI-induced airway hyperresponsiveness, airway inflammation, and mitochondrial dysfunction. Pretreatment with mitochondrial precipitation, to simulate the release of mtDAMPs, further increased TDI-induced airway inflammation and the expression of RAGE in mice. Administration of the inhibitor of RAGE, FPS-ZM1, alleviated the airway inflammation, the abnormal open of mPTP and mitochondrial dysfunction induced by mtDAMPs and TDI. Furthermore, stimulation with different mtDAMPs activated RAGE signaling in human bronchial epithelial cells. Accordingly, our study indicated that mPTP was important and BKA was efficient in alleviating inflammation in TDI-induced asthma. A positive feedback loop involving mPTP, mtDAMPs and RAGE was present in TDI-induced asthma, indicating that mPTP might serve as a potential therapeutic target for asthma.
Collapse
Affiliation(s)
- Ying Chen
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Junwen Huang
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yuemao Li
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yaoxin Chen
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhaoqian Gong
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Maosheng Xu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yanyan Ma
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Dapeng Hu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xianru Peng
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Guilin Xu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Shaoxi Cai
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Laiyu Liu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wenqu Zhao
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Haijin Zhao
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
37
|
Liu Z, Zhang X, Xiong S, Huang S, Ding X, Xu M, Yao J, Liu S, Zhao F. Endothelial dysfunction of syphilis: Pathogenesis. J Eur Acad Dermatol Venereol 2024; 38:1478-1490. [PMID: 38376088 DOI: 10.1111/jdv.19899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 01/19/2024] [Indexed: 02/21/2024]
Abstract
Treponema pallidum is the causative factor of syphilis, a sexually transmitted disease (STD) characterized by perivascular infiltration of inflammatory cells, vascular leakage, swelling and proliferation of endothelial cells (ECs). The endothelium lining blood and lymphatic vessels is a key barrier separating body fluids from host tissues and is a major target of T. pallidum. In this review, we focus on how T. pallidum establish intimate interactions with ECs, triggering endothelial dysfunction such as endothelial inflammation, abnormal repairment and damage of ECs. In addition, we summarize that migration and invasion of T. pallidum across vascular ECs may occur through two pathways. These two mechanisms of transendothelial migration are paracellular and cholesterol-dependent, respectively. Herein, clarifying the relationship between T. pallidum and endothelial dysfunction is of great significance to provide novel strategies for diagnosis and prevention of syphilis, and has a great potential prospect of clinical application.
Collapse
Affiliation(s)
- Zhaoping Liu
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Xiaohong Zhang
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Shun Xiong
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Shaobin Huang
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Xuan Ding
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Man Xu
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Jiangchen Yao
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Shuangquan Liu
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Feijun Zhao
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| |
Collapse
|
38
|
Tian H, Liu Q, Yu X, Cao Y, Huang X. Damage-associated molecular patterns in viral infection: potential therapeutic targets. Crit Rev Microbiol 2024:1-18. [PMID: 39091137 DOI: 10.1080/1040841x.2024.2384885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 05/25/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024]
Abstract
Frequent viral infections leading to infectious disease outbreaks have become a significant global health concern. Fully elucidating the molecular mechanisms of the immune response against viral infections is crucial for epidemic prevention and control. The innate immune response, the host's primary defense against viral infection, plays a pivotal role and has become a breakthrough in research mechanisms. A component of the innate immune system, damage-associated molecular patterns (DAMPs) are involved in inducing inflammatory responses to viral infections. Numerous DAMPs are released from virally infected cells, activating downstream signaling pathways via internal and external receptors on immune cells. This activation triggers immune responses and helps regulate viral host invasion. This review examines the immune regulatory mechanisms of various DAMPs, such as the S100 protein family, high mobility group box 1 (HMGB1), and heat shock proteins, in various viral infections to provide a theoretical basis for designing novel antiviral drugs.
Collapse
Affiliation(s)
- Huizhen Tian
- School of Basic Medical Sciences, Jiangxi medical College, Nanchang University, Nanchang, China
| | - Qiong Liu
- School of Basic Medical Sciences, Jiangxi medical College, Nanchang University, Nanchang, China
| | - Xiaomin Yu
- School of Basic Medical Sciences, Jiangxi medical College, Nanchang University, Nanchang, China
- Medical Experimental Teaching Center, School of Basic Medical Sciences, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yanli Cao
- School of Basic Medical Sciences, Jiangxi medical College, Nanchang University, Nanchang, China
| | - Xiaotian Huang
- School of Basic Medical Sciences, Jiangxi medical College, Nanchang University, Nanchang, China
| |
Collapse
|
39
|
Yang Q, Sun X, Ding Q, Qi M, Liu C, Li T, Shi F, Wang L, Li C, Kim JS. An ATP-responsive metal-organic framework against periodontitis via synergistic ion-interference-mediated pyroptosis. Natl Sci Rev 2024; 11:nwae225. [PMID: 39071842 PMCID: PMC11275458 DOI: 10.1093/nsr/nwae225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/08/2024] [Accepted: 06/11/2024] [Indexed: 07/30/2024] Open
Abstract
Periodontitis involves hyperactivated stromal cells that recruit immune cells, exacerbating inflammation. This study presents an ATP-responsive metal-organic framework (Mg/Zn-MOF) designed for periodontitis treatment, utilizing ion interference to modulate immune responses and prevent tissue destruction. Addressing the challenges of synergistic ion effects and targeted delivery faced by traditional immunomodulatory nanomaterials, the Mg/Zn-MOF system is activated by extracellular ATP-a pivotal molecule in periodontitis pathology-ensuring targeted ion release. Magnesium and zinc ions released from the framework synergistically inhibit membrane pore formation by attenuating Gasdermin D (GSDMD) expression and activation. This action curtails pyroptosis, lactate dehydrogenase and IL-1β release, thwarting the onset of inflammatory cascades. Mechanistically, Mg/Zn-MOF intervenes in both the NLRP3/Caspase-1/GSDMD and Caspase-11/GSDMD pathways to mitigate pyroptosis. In vivo assessments confirm its effectiveness in diminishing inflammatory cell infiltration and preserving collagen integrity, thereby safeguarding against periodontal tissue damage and bone loss. This investigation highlights the promise of ion-interference strategies in periodontitis immunotherapy, representing a significant stride in developing targeted therapeutic approaches.
Collapse
Affiliation(s)
- Qijing Yang
- Department of Prosthodontics, Jilin Provincial Engineering Laboratory of Intelligent Oral Treatment Technology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Xiaolin Sun
- Department of Prosthodontics, Jilin Provincial Engineering Laboratory of Intelligent Oral Treatment Technology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Qihang Ding
- Department of Prosthodontics, Jilin Provincial Engineering Laboratory of Intelligent Oral Treatment Technology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
- Department of Chemistry, Korea University, Seoul 02841, South Korea
| | - Manlin Qi
- Department of Prosthodontics, Jilin Provincial Engineering Laboratory of Intelligent Oral Treatment Technology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Chengyu Liu
- Department of Prosthodontics, Jilin Provincial Engineering Laboratory of Intelligent Oral Treatment Technology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Tingxuan Li
- Department of Prosthodontics, Jilin Provincial Engineering Laboratory of Intelligent Oral Treatment Technology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Fangyu Shi
- Department of Prosthodontics, Jilin Provincial Engineering Laboratory of Intelligent Oral Treatment Technology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Lin Wang
- Department of Prosthodontics, Jilin Provincial Engineering Laboratory of Intelligent Oral Treatment Technology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Chunyan Li
- Department of Prosthodontics, Jilin Provincial Engineering Laboratory of Intelligent Oral Treatment Technology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul 02841, South Korea
| |
Collapse
|
40
|
Yan L, Wang J, Cai X, Liou Y, Shen H, Hao J, Huang C, Luo G, He W. Macrophage plasticity: signaling pathways, tissue repair, and regeneration. MedComm (Beijing) 2024; 5:e658. [PMID: 39092292 PMCID: PMC11292402 DOI: 10.1002/mco2.658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 08/04/2024] Open
Abstract
Macrophages are versatile immune cells with remarkable plasticity, enabling them to adapt to diverse tissue microenvironments and perform various functions. Traditionally categorized into classically activated (M1) and alternatively activated (M2) phenotypes, recent advances have revealed a spectrum of macrophage activation states that extend beyond this dichotomy. The complex interplay of signaling pathways, transcriptional regulators, and epigenetic modifications orchestrates macrophage polarization, allowing them to respond to various stimuli dynamically. Here, we provide a comprehensive overview of the signaling cascades governing macrophage plasticity, focusing on the roles of Toll-like receptors, signal transducer and activator of transcription proteins, nuclear receptors, and microRNAs. We also discuss the emerging concepts of macrophage metabolic reprogramming and trained immunity, contributing to their functional adaptability. Macrophage plasticity plays a pivotal role in tissue repair and regeneration, with macrophages coordinating inflammation, angiogenesis, and matrix remodeling to restore tissue homeostasis. By harnessing the potential of macrophage plasticity, novel therapeutic strategies targeting macrophage polarization could be developed for various diseases, including chronic wounds, fibrotic disorders, and inflammatory conditions. Ultimately, a deeper understanding of the molecular mechanisms underpinning macrophage plasticity will pave the way for innovative regenerative medicine and tissue engineering approaches.
Collapse
Affiliation(s)
- Lingfeng Yan
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| | - Jue Wang
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| | - Xin Cai
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| | - Yih‐Cherng Liou
- Department of Biological SciencesFaculty of ScienceNational University of SingaporeSingaporeSingapore
- National University of Singapore (NUS) Graduate School for Integrative Sciences and EngineeringNational University of SingaporeSingaporeSingapore
| | - Han‐Ming Shen
- Faculty of Health SciencesUniversity of MacauMacauChina
| | - Jianlei Hao
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and TreatmentZhuhai Institute of Translational MedicineZhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University)Jinan UniversityZhuhaiGuangdongChina
- The Biomedical Translational Research InstituteFaculty of Medical ScienceJinan UniversityGuangzhouGuangdongChina
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospitaland West China School of Basic Medical Sciences and Forensic MedicineSichuan University, and Collaborative Innovation Center for BiotherapyChengduChina
| | - Gaoxing Luo
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| | - Weifeng He
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| |
Collapse
|
41
|
Shen P, Jiang X, Kuang Y, Wang W, Raj R, Wang W, Zhu Y, Zhang X, Yu B, Zhang J. Natural triterpenoid-aided identification of the druggable interface of HMGB1 occupied by TLR4. RSC Chem Biol 2024; 5:751-762. [PMID: 39092445 PMCID: PMC11289874 DOI: 10.1039/d4cb00062e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 06/04/2024] [Indexed: 08/04/2024] Open
Abstract
HMGB1 interacts with TLR4 to activate the inflammatory cascade response, contributing to the pathogenesis of endogenous tissue damage and infection. The immense importance of HMGB1-TLR4 interaction in the immune system has made its binding interface an area of significant interest. To map the binding interface of HMGB1 occupied by TLR4, triterpenoids that disrupt the HMGB1-TLR4 interaction and interfere with HMGB1-induced inflammation were developed. Using the unique triterpenoid PT-22 as a probe along with photoaffinity labeling and site-directed mutagenesis, we found that the binding interface of HMGB1 was responsible for the recognition of TLR4 located on the "L" shaped B-box with K114 as a crucial hot-spot residue. Amazingly, this highly conserved interaction surface overlapped with the antigen-recognition epitope of an anti-HMGB1 antibody. Our findings propose a novel strategy for better understanding the druggable interface of HMGB1 that interacts with TLR4 and provide insights for the rational design of HMGB1-TLR4 PPI inhibitors to fine tune immune responses.
Collapse
Affiliation(s)
- Pingping Shen
- Department of Resources Science of Traditional Chinese Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University Nanjing 210009 P. R. China +86-25-86185158 +86-25-86185157
| | - Xuewa Jiang
- Department of Resources Science of Traditional Chinese Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University Nanjing 210009 P. R. China +86-25-86185158 +86-25-86185157
| | - Yi Kuang
- Department of Resources Science of Traditional Chinese Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University Nanjing 210009 P. R. China +86-25-86185158 +86-25-86185157
| | - Weiwei Wang
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine Nanjing 210046 P. R. China
| | - Richa Raj
- Department of Resources Science of Traditional Chinese Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University Nanjing 210009 P. R. China +86-25-86185158 +86-25-86185157
| | - Wei Wang
- Department of Chemistry, College of Liberal Arts and Sciences, University of Illinois Chicago Chicago IL USA
| | - Yuyuan Zhu
- The Center for Chemical Biology, Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 P. R. China
| | - Xiaochun Zhang
- School of Pharmaceutical Sciences, Tsinghua University Beijing 100084 P. R. China
| | - Boyang Yu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University Nanjing 211198 P. R. China
| | - Jian Zhang
- Department of Resources Science of Traditional Chinese Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University Nanjing 210009 P. R. China +86-25-86185158 +86-25-86185157
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University Nanjing 211198 P. R. China
| |
Collapse
|
42
|
Waltimo E, Eray M, Mäkitie A, Haglund C, Atula T, Hagström J. Toll-like receptors 2 and 4, and bacterial proteins in IgG4-related sialadenitis, other types of chronic sialadenitis and sialolithiasis. J Oral Microbiol 2024; 16:2382633. [PMID: 39055281 PMCID: PMC11271128 DOI: 10.1080/20002297.2024.2382633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 06/14/2024] [Accepted: 07/16/2024] [Indexed: 07/27/2024] Open
Abstract
Background The association of chronic sclerosing sialadenitis and IgG4-related disease (IgG4-RD) has resulted in the more frequent identification of IgG4-positivity in submandibular gland inflammations, also uncovering IgG4 overexpression in nonspecific inflammations. These findings lead us to hypothesise that IgG4-positive sialadenitis represents a continuous inflammatory process overlapping histologically with IgG4-RD, possibly differing in aetiology. However, the antigen underlying IgG4 overexpression in IgG4-positive sialadenitis and IgG4-RD remains unknown. Materials and methods Here, we investigated toll-like receptor (TLR) - mediated bacterial inflammation in submandibular gland tissues of patients with IgG4-positive and IgG4-negative chronic inflammatory lesions of the submandibular gland (n = 61), with noninflamed submandibular glands serving as controls (n = 4). Utilising immunohistochemistry, we assessed the expression of TLR2 and TLR4, lipopolysaccharide (LPS) and the P. gingivalis-specific antigen gingipain R1. Results We observed TLR2- and TLR4-immunopositivity in 64 (98%) samples. However, TLR2 and TLR4 staining intensity was significantly stronger in the IgG4-positive group. LPS- and gingipain R1 immunopositivity were observed in 56 (86%) and 58 (89%) samples, respectively. LPS-positivity localised exclusively in mast cell-like cells, while gingipain R1-positivity remained scarce. Conclusions A stronger TLR2 or TLR4 expression in IgG4-positive sialadenitis may indicate a tissue-related factor underlying this form of chronic sialadenitis. LPS- and P. gingivalis immunopositivity remained weak throughout this series. Thus, gram-negative bacteria may not represent pathogens underlying these forms of chronic sialadenitis.
Collapse
Affiliation(s)
- Elin Waltimo
- Department of Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - Mine Eray
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Antti Mäkitie
- Department of Otorhinolaryngology – Head and Neck Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program in Systems Oncology, Faculty of Medicine, Helsinki, Helsinki, Finland
| | - Caj Haglund
- Research Programmes Unit, Translational Cancer Medicine, University of Helsinki, Helsinki, Finland
- Department of Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Timo Atula
- Department of Otorhinolaryngology – Head and Neck Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Jaana Hagström
- Department of Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Programmes Unit, Translational Cancer Medicine, University of Helsinki, Helsinki, Finland
- Department of Oral Pathology and Radiology, University of Turku, Turku, Finland
| |
Collapse
|
43
|
Sun K, Lu F, Hou L, Zhang X, Pan C, Liu H, Zheng Z, Guo Z, Ruan Z, Hou Y, Zhang J, Guo F, Zhu W. IRF1 regulation of ZBP1 links mitochondrial DNA and chondrocyte damage in osteoarthritis. Cell Commun Signal 2024; 22:366. [PMID: 39026271 PMCID: PMC11256489 DOI: 10.1186/s12964-024-01744-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 07/10/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Z-DNA binding protein 1 (ZBP1) is a nucleic acid sensor that is involved in multiple inflammatory diseases, but whether and how it contributes to osteoarthritis (OA) are unclear. METHODS Cartilage tissues were harvested from patients with OA and a murine model of OA to evaluate ZBP1 expression. Subsequently, the functional role and mechanism of ZBP1 were examined in primary chondrocytes, and the role of ZBP1 in OA was explored in mouse models. RESULTS We showed the upregulation of ZBP1 in articular cartilage originating from OA patients and mice with OA after destabilization of the medial meniscus (DMM) surgery. Specifically, knockdown of ZBP1 alleviated chondrocyte damage and protected mice from DMM-induced OA. Mechanistically, tumor necrosis factor alpha induced ZBP1 overexpression in an interferon regulatory factor 1 (IRF1)-dependent manner and elicited the activation of ZBP1 via mitochondrial DNA (mtDNA) release and ZBP1 binding. The upregulated and activated ZBP1 could interact with receptor-interacting protein kinase 1 and activate the transforming growth factor-beta-activated kinase 1-NF-κB signaling pathway, which led to chondrocyte inflammation and extracellular matrix degradation. Moreover, inhibition of the mtDNA-IRF1-ZBP1 axis with Cyclosporine A, a blocker of mtDNA release, could delay the progression of DMM-induced OA. CONCLUSIONS Our data revealed the pathological role of the mtDNA-IRF1-ZBP1 axis in OA chondrocytes, suggesting that inhibition of this axis could be a viable therapeutic approach for OA.
Collapse
Affiliation(s)
- Kai Sun
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Fan Lu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Liangcai Hou
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Xiong Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Chunran Pan
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Haigang Liu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zehang Zheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zhou Guo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zhaoxuan Ruan
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Yanjun Hou
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Jinming Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Fengjing Guo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Wentao Zhu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
44
|
Karthäuser JF, Gruhn D, Martínez Guajardo A, Kopecz R, Babel N, Stervbo U, Laschewsky A, Viebahn R, Salber J, Rosenhahn A. In vitro biocompatibility analysis of protein-resistant amphiphilic polysulfobetaines as coatings for surgical implants in contact with complex body fluids. Front Bioeng Biotechnol 2024; 12:1403654. [PMID: 39086500 PMCID: PMC11288920 DOI: 10.3389/fbioe.2024.1403654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 06/19/2024] [Indexed: 08/02/2024] Open
Abstract
The fouling resistance of zwitterionic coatings is conventionally explained by the strong hydrophilicity of such polymers. Here, the in vitro biocompatibility of a set of systematically varied amphiphilic, zwitterionic copolymers is investigated. Photocrosslinkable, amphiphilic copolymers containing hydrophilic sulfobetaine methacrylate (SPe) and butyl methacrylate (BMA) were systematically synthesized in different ratios (50:50, 70:30, and 90:10) with a fixed content of photo-crosslinker by free radical copolymerization. The copolymers were spin-coated onto substrates and subsequently photocured by UV irradiation. Pure pBMA and pSPe as well as the prepared amphiphilic copolymers showed BMA content-dependent wettability in the dry state, but overall hydrophilic properties a fortiori in aqueous conditions. All polysulfobetaine-containing copolymers showed high resistance against non-specific adsorption (NSA) of proteins, platelet adhesion, thrombocyte activation, and bacterial accumulation. In some cases, the amphiphilic coatings even outperformed the purely hydrophilic pSPe coatings.
Collapse
Affiliation(s)
- Jana F. Karthäuser
- Analytical Chemistry—Biointerfaces, Ruhr University Bochum, Bochum, Germany
| | - Dierk Gruhn
- Experimental Surgery, Ruhr University Bochum, Bochum, Germany
- Department of Surgery, Knappschaftskrankenhaus Bochum, University Hospital of the Ruhr University Bochum, Bochum, Germany
| | | | - Regina Kopecz
- Analytical Chemistry—Biointerfaces, Ruhr University Bochum, Bochum, Germany
| | - Nina Babel
- Centre for Translational Medicine, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr University Bochum, Herne, Germany
| | - Ulrik Stervbo
- Centre for Translational Medicine, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr University Bochum, Herne, Germany
| | - André Laschewsky
- Institute of Chemistry, Universität Potsdam, Potsdam, Germany
- Fraunhofer Institute of Applied Polymer Research IAP, Potsdam, Germany
| | - Richard Viebahn
- Department of Surgery, Knappschaftskrankenhaus Bochum, University Hospital of the Ruhr University Bochum, Bochum, Germany
| | - Jochen Salber
- Experimental Surgery, Ruhr University Bochum, Bochum, Germany
- Department of Surgery, Knappschaftskrankenhaus Bochum, University Hospital of the Ruhr University Bochum, Bochum, Germany
| | - Axel Rosenhahn
- Analytical Chemistry—Biointerfaces, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
45
|
Banderwal R, Kadian M, Garg S, Kumar A. 'Comprehensive review of emerging drug targets in traumatic brain injury (TBI): challenges and future scope. Inflammopharmacology 2024:10.1007/s10787-024-01524-w. [PMID: 39023681 DOI: 10.1007/s10787-024-01524-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 02/12/2024] [Indexed: 07/20/2024]
Abstract
Traumatic brain injury (TBI) is a complex brain problem that causes significant morbidity and mortality among people of all age groups. The complex pathophysiology, varied symptoms, and inadequate treatment further precipitate the problem. Further, TBI produces several psychiatric problems and other related complications in post-TBI survival patients, which are often treated symptomatically or inadequately. Several approaches, including neuroprotective agents targeting several pathways of oxidative stress, neuroinflammation, cytokines, immune system GABA, glutamatergic, microglia, and astrocytes, are being tried by researchers to develop effective treatments or magic bullets to manage the condition effectively. The problem of TBI is therefore treated as a challenge among pharmaceutical scientists or researchers to develop drugs for the effective management of this problem. The goal of the present comprehensive review is to provide an overview of the several pharmacological targets, processes, and cellular pathways that researchers are focusing on, along with an update on their current state.
Collapse
Affiliation(s)
- Rittu Banderwal
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), UGC- Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Monika Kadian
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), UGC- Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Sukant Garg
- Department of General Pathology, Dr HS Judge Institute of Dental Sciences and Hospital, Panjab University, Chandigarh, 160014, India
| | - Anil Kumar
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), UGC- Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
46
|
Kamal R, Awasthi A, Pundir M, Thakur S. Healing the diabetic wound: Unlocking the secrets of genes and pathways. Eur J Pharmacol 2024; 975:176645. [PMID: 38759707 DOI: 10.1016/j.ejphar.2024.176645] [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: 03/04/2024] [Revised: 05/03/2024] [Accepted: 05/13/2024] [Indexed: 05/19/2024]
Abstract
Diabetic wounds (DWs) are open sores that can occur anywhere on a diabetic patient's body. They are often complicated by infections, hypoxia, oxidative stress, hyperglycemia, and reduced growth factors and nucleic acids. The healing process involves four phases: homeostasis, inflammation, proliferation, and remodeling, regulated by various cellular and molecular events. Numerous genes and signaling pathways such as VEGF, TGF-β, NF-κB, PPAR-γ, MMPs, IGF, FGF, PDGF, EGF, NOX, TLR, JAK-STAT, PI3K-Akt, MAPK, ERK, JNK, p38, Wnt/β-catenin, Hedgehog, Notch, Hippo, FAK, Integrin, and Src pathways are involved in these events. These pathways and genes are often dysregulated in DWs leading to impaired healing. The present review sheds light on the pathogenesis, healing process, signaling pathways, and genes involved in DW. Further, various therapeutic strategies that target these pathways and genes via nanotechnology are also discussed. Additionally, clinical trials on DW related to gene therapy are also covered in the present review.
Collapse
Affiliation(s)
- Raj Kamal
- Department of Quality Assurance, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Ankit Awasthi
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| | - Mandeep Pundir
- School of Pharmaceutical Sciences, RIMT University, Punjab, 142001, India; Chitkara College of Pharmacy, Chitkara University, Punjab, 142001, India
| | - Shubham Thakur
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, 142001, India
| |
Collapse
|
47
|
Valencia R, Kranrod JW, Fang L, Soliman AM, Azer B, Clemente-Casares X, Seubert JM. Linoleic acid-derived diol 12,13-DiHOME enhances NLRP3 inflammasome activation in macrophages. FASEB J 2024; 38:e23748. [PMID: 38940767 DOI: 10.1096/fj.202301640rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 05/30/2024] [Accepted: 06/05/2024] [Indexed: 06/29/2024]
Abstract
12,13-dihydroxy-9z-octadecenoic acid (12,13-DiHOME) is a linoleic acid diol derived from cytochrome P-450 (CYP) epoxygenase and epoxide hydrolase (EH) metabolism. 12,13-DiHOME is associated with inflammation and mitochondrial damage in the innate immune response, but how 12,13-DiHOME contributes to these effects is unclear. We hypothesized that 12,13-DiHOME enhances macrophage inflammation through effects on NOD-like receptor protein 3 (NLRP3) inflammasome activation. To test this hypothesis, we utilized human monocytic THP1 cells differentiated into macrophage-like cells with phorbol myristate acetate (PMA). 12,13-DiHOME present during lipopolysaccharide (LPS)-priming of THP1 macrophages exacerbated nigericin-induced NLRP3 inflammasome activation. Using high-resolution respirometry, we observed that priming with LPS+12,13-DiHOME altered mitochondrial respiratory function. Mitophagy, measured using mito-Keima, was also modulated by 12,13-DiHOME present during priming. These mitochondrial effects were associated with increased sensitivity to nigericin-induced mitochondrial depolarization and reactive oxygen species production in LPS+12,13-DiHOME-primed macrophages. Nigericin-induced mitochondrial damage and NLRP3 inflammasome activation in LPS+12,13-DiHOME-primed macrophages were ablated by the mitochondrial calcium uniporter (MCU) inhibitor, Ru265. 12,13-DiHOME present during LPS-priming also enhanced nigericin-induced NLRP3 inflammasome activation in primary murine bone marrow-derived macrophages. In summary, these data demonstrate a pro-inflammatory role for 12,13-DiHOME by enhancing NLRP3 inflammasome activation in macrophages.
Collapse
Affiliation(s)
- Robert Valencia
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Cardiovascular Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Joshua W Kranrod
- Cardiovascular Research Institute, University of Alberta, Edmonton, Alberta, Canada
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Liye Fang
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Cardiovascular Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Amro M Soliman
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Brandon Azer
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Xavier Clemente-Casares
- Cardiovascular Research Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, Alberta, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - John M Seubert
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Cardiovascular Research Institute, University of Alberta, Edmonton, Alberta, Canada
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
48
|
Liu X, Yao Y, Zhu Y, Lu F, Chen X. Inhibition of Adipocyte Necroptosis Alleviates Fat Necrosis and Fibrosis After Grafting in a Murine Model. Aesthet Surg J 2024; 44:NP585-NP605. [PMID: 38796831 DOI: 10.1093/asj/sjae108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/24/2024] [Accepted: 04/24/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Because of the delicate structure of the adipose tissue, fat necrosis accounts for 43.7% of all complications after autologous fat grafting; however, its regulation remains unclear. OBJECTIVES The purpose of this study was to examine the role of necroptosis in fat graft remodeling after grafting. METHODS Clinical fat graft necrosis samples were collected, and the expression levels of the necroptosis marker phosphorylated(p)-MLKL were analyzed. Transcriptome analysis was performed on fat grafts before and 1 week after transplantation in C57BL/6 mouse fat grafting models. Additionally, the in vivo effects of RIPK1 inhibitor Nec-1s or RIPK3 inhibitor GSK'872 on the fat grafting complications, including fat necrosis and fibrosis, were investigated. RESULTS Necroptosis markers were observed and associated with higher occurrence of fibrosis in clinical fat graft necrosis samples compared to normal fat tissue. Amplification and RNA-Seq were conducted on RNA isolated from fat grafts before and after grafting. MLKL, RIPK1, and RIPK3's expression levels were significantly upregulated in comparison to controls. Higher expression levels of necroptotic RNAs were associated with higher levels of DAMPs, including Cxcl2, HMGB1, S100a8, S100a9, Nlrp3, and IL33, and activated proinflammatory signaling pathways, including the TNF, NF-kappa B, and chemokine signaling pathways. Necroptotic inhibitor Nec-1s and GSK'872 robustly suppressed the p-MLKL expression level and significantly inhibited necroptotic cell death, especially in adipocytes. Moreover, administration of Nec-1s and GSK'872 significantly alleviated fat necrosis and subsequent fibrosis in fat grafts. CONCLUSIONS Collectively, our study findings highlight the potential therapeutic applications of necroptosis inhibitors in preventing fat necrosis and fibrosis after grafting. LEVEL OF EVIDENCE: 4
Collapse
|
49
|
Ni H, Yu S, Qian S, Lu J, Feng J, Zhang J. Photothermal Particle-Loaded Panax Notoginseng Polysaccharide Cryogels As Personalized Tumor Vaccines. Biomacromolecules 2024; 25:4394-4405. [PMID: 38859583 DOI: 10.1021/acs.biomac.4c00442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
Combination immunotherapy is being increasingly explored for cancer treatment, leading to various vector materials for the codelivery of immune agents and drugs. However, current tumor vaccines exhibit poor immunogenicity, severely compromising their therapeutic efficacy. Herein, an injectable hydrogel was developed based on dopamine (DA) and Panax notoginseng polysaccharide (PNPS) loaded with hair microparticles (HMPs) to enhance the immunogenicity of tumor vaccines. Photothermal effects of incorporated HMPs can trigger immunogenic cancer cell death and the release of abundant autologous tumor antigens, which are captured by catechol groups. Concomitant breakdown of PNPS recruits and activates dendritic cells (DCs). The macroporous structure of cryogels allows immune cell infiltration and interaction with antigens adsorbed on PNPS and DA cryogels (PD cryogels), thereby provoking potent cytotoxic T-cell responses. Hence, PD cryogels enabling cell infiltration and accelerated DC maturation may serve as a therapeutic vaccination platform against cancer.
Collapse
Affiliation(s)
- Haifeng Ni
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, Zhejiang, P. R. China
| | - Shijie Yu
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, Zhejiang, P. R. China
| | - Sunxiang Qian
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, Zhejiang, P. R. China
| | - Jie Lu
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, Zhejiang, P. R. China
| | - Jie Feng
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, Zhejiang, P. R. China
| | - Jing Zhang
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, Zhejiang, P. R. China
| |
Collapse
|
50
|
Chen J, Qi D, Hu H, Wang X, Lin W. Unconventional posttranslational modification in innate immunity. Cell Mol Life Sci 2024; 81:290. [PMID: 38970666 PMCID: PMC11335215 DOI: 10.1007/s00018-024-05319-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 07/08/2024]
Abstract
Pattern recognition receptors (PRRs) play a crucial role in innate immunity, and a complex network tightly controls their signaling cascades to maintain immune homeostasis. Within the modification network, posttranslational modifications (PTMs) are at the core of signaling cascades. Conventional PTMs, which include phosphorylation and ubiquitination, have been extensively studied. The regulatory role of unconventional PTMs, involving unanchored ubiquitination, ISGylation, SUMOylation, NEDDylation, methylation, acetylation, palmitoylation, glycosylation, and myristylation, in the modulation of innate immune signaling pathways has been increasingly investigated. This comprehensive review delves into the emerging field of unconventional PTMs and highlights their pivotal role in innate immunity.
Collapse
Affiliation(s)
- Jiaxi Chen
- The Second Affiliated Hospital and Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Dejun Qi
- The Second Affiliated Hospital and Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Haorui Hu
- The Second Affiliated Hospital and Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Xiaojian Wang
- Institute of Immunology and Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China.
| | - Wenlong Lin
- The Second Affiliated Hospital and Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China.
| |
Collapse
|