1
|
Wu M, Yang J, Tian Z, Sun H, Zhang Z, Luo J, Guan G, Yin H, Niu Q, Hao R. Transcriptome profiling reveals that the host BRD4 protein facilitates African swine fever virus infection and suppresses inflammatory cytokine expression by downregulating transcriptional regulatory signaling pathways. Vet Microbiol 2025; 305:110498. [PMID: 40215802 DOI: 10.1016/j.vetmic.2025.110498] [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: 11/16/2024] [Revised: 03/10/2025] [Accepted: 03/25/2025] [Indexed: 05/17/2025]
Abstract
The African swine fever virus (ASFV), a complex DNA virus belonging to the Asfarviridae family, is a significant threat to the global swine industry because of its high mortality rates and impact on international trade. The establishment of a stable and efficient cell culture model of ASFV in vitro is helpful for the development of effective vaccines. Several passaged cell lines supporting ASFV replication have been reported to meet the scientific purpose of serial passage of ASFV to a certain extent, but it remains to be determined whether gene expression is lost or whether immunogenicity changes after serial passage of the virus. It is also unclear these edited cell lines how to affect ASFV replication. In our previous study, 3D4/21 cells were transduced with a lentivirus packaging system to express the BD1/2 domain of bromodomain-containing protein 4 (BRD4-BD1/2) and establish a 3D4/21-BD1/2 cell line, which efficiently increased ASFV replication. In this study, the role of bromodomain-containing protein 4 (BRD4), particularly its BD1/2 domains,in enhancing ASFV replication was investigated using an engineered 3D4/21 cell line. Through RNA-Seq transcriptomic analysis, we revealed that the host BRD4 protein facilitates ASFV infection and suppresses key transcription factors (CDK9 and p-CDK9) and inflammatory cytokine expression by downregulating transcriptional regulatory signaling pathways and suppressing innate immune responses. This dual mechanism of BRD4-BD1/2 in promoting ASFV immune evasion and adaptation underscores the virus's strategic exploitation of host epigenetic factors. These findings provide valuable insights into viral pathogenesis and identify potential therapeutic targets, paving the way for future antiviral strategies.
Collapse
Affiliation(s)
- Mengli Wu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730000, China; African Swine Fever Regional Laboratory of China (Lanzhou), Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, Gansu 730046, China.
| | - Jifei Yang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730000, China; African Swine Fever Regional Laboratory of China (Lanzhou), Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, Gansu 730046, China.
| | - Zhancheng Tian
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730000, China; African Swine Fever Regional Laboratory of China (Lanzhou), Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, Gansu 730046, China.
| | - Hualin Sun
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730000, China; African Swine Fever Regional Laboratory of China (Lanzhou), Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, Gansu 730046, China.
| | - Zhonghui Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730000, China; African Swine Fever Regional Laboratory of China (Lanzhou), Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, Gansu 730046, China.
| | - Jianxun Luo
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730000, China; African Swine Fever Regional Laboratory of China (Lanzhou), Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, Gansu 730046, China.
| | - Guiquan Guan
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730000, China; African Swine Fever Regional Laboratory of China (Lanzhou), Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, Gansu 730046, China.
| | - Hong Yin
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730000, China; African Swine Fever Regional Laboratory of China (Lanzhou), Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, Gansu 730046, China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, Jiangsu 225009, China.
| | - Qingli Niu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730000, China; African Swine Fever Regional Laboratory of China (Lanzhou), Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, Gansu 730046, China.
| | - Rongzeng Hao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730000, China; African Swine Fever Regional Laboratory of China (Lanzhou), Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, Gansu 730046, China.
| |
Collapse
|
2
|
Huang J, Feng L, Jiang WD, Liu Y, Jiang J, Ren HM, Wu CM, Zhou XQ, Wu P. Dietary AiiO-AIO6 mitigated Aeromonas hydrophila-induced intestinal inflammation in juvenile grass carp (Ctenopharyngodon idella) involving in NF-κB signaling and pyroptosis. FISH & SHELLFISH IMMUNOLOGY 2025; 161:110297. [PMID: 40139288 DOI: 10.1016/j.fsi.2025.110297] [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: 11/25/2024] [Revised: 03/05/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
AiiO-AIO6 is a quorum-sensing quenching enzyme that could decrease the virulence of pathogenic bacteria, and improve animal immunity. However, the precise regulatory mechanism of AiiO-AIO6 on intestinal immunity remains unknown. Thus, this study aimed to reduce this knowledge gap. After feeding with graded levels of AIO-AIO6 (0.0 (un-supplemented group), 2.5, 5.0, 7.5, 10.0, and 12.5 U/g) for 70 days, juvenile grass carp was selected from each group for a 6-day Aeromonas hydrophila challenge test. Meanwhile, some other fish selected from un-supplemented control group were injected with saline as un-challenged control. Results showed that A. hydrophila infection increased enteritis morbidity, and caused intestinal inflammation in grass carp compared with saline group, while AiiO-AIO6 supplementation decreased enteritis morbidity, mitigated inflammatory cell infiltration, pyroptosis, and apoptosis in the intestine after A. hydrophila infection. Furthermore, both 5.0 and 7.5 U/g AiiO-AIO6 decreased gene expressions of tumor necrosis factor-α and interleukin-1β, and elevated gene expressions of transforming growth factor-β1 and IL-10, potentially associating with decreased NF-κB p65 and increased PPARγ signaling in the intestine. Supplementing 5.0 or 7.5 U/g AiiO-AIO6 also mitigated intestinal pyroptosis, as indicated by reduced mRNA levels of NLRP3, PYCARD, caspase-1, GSDME a, and GSDME b, and decreased protein levels of N-GSDME and IL-1β. Additionally, AiiO-AIO6 alleviated intestinal apoptosis, demonstrated by reduced gene expressions of pro-apoptotic genes apoptotic protease activating factor-1, Bcl-2 associated X protein, Fas ligand, and caspase-3, as well as c-Jun N-terminal kinase and mitogen-activated protein kinase p38, and elevated gene expressions of anti-apoptotic factors, B-cell lymphoma-2, myeloid cell leukemia 1, and inhibitor of apoptosis protein. Altogether, optimal levels of AiiO-AIO6 attenuated intestinal inflammation probably relating to down-regulated NF-κB signaling, and reduced NLRP3 and GSDME-mediated pyroptosis, and finally reduced apoptosis in the intestine of grass carp.
Collapse
Affiliation(s)
- Jie Huang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China
| | - Wei-Dan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China
| | - Jun Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
| | - Hong-Mei Ren
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
| | - Chai-Mei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
| | - Xiao-Qiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China.
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China.
| |
Collapse
|
3
|
Toumpanakis D, Bartziokas K, Bakakos A, Fouka E, Bakakos P, Loukides S, Steiropoulos P, Papaioannou AI. Monoclonal Antibodies for the Treatment of Chronic Obstructive Pulmonary Disease. Pulm Ther 2025; 11:177-193. [PMID: 40123030 PMCID: PMC12102449 DOI: 10.1007/s41030-025-00291-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 02/26/2025] [Indexed: 03/25/2025] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a common and complex disease characterized by persistent airflow limitation and the presence of exacerbations, resulting in significant morbidity and mortality. Although the pathogenesis of COPD is multifactorial, airway inflammation plays a significant role in disease progression. Despite the advantages of non-pharmaceutical and pharmaceutical interventions that have significantly improved the symptom burden and exacerbation frequency in COPD, there is a lack of disease-modifying therapies that target the underlying disease mechanisms. Monoclonal antibodies (mAbs), a drug class that has improved treatment in severe asthma by blocking mediators of the type 2 (Th2) and allergic inflammatory cascades, are currently under investigation for their efficacy in COPD. Our review summarizes the evidence for the use of monoclonal antibodies in COPD and discusses current limitations and promising advances. Although targeting Th1 inflammation has failed to improve COPD outcomes, recent clinical trials have shown beneficial effects of monoclonal antibodies targeting Th2 inflammation, providing evidence for a personalized approach in COPD treatment.
Collapse
Affiliation(s)
- Dimitrios Toumpanakis
- 2Nd Department of Critical Care, Medical School, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - Konstantinos Bartziokas
- 2Nd Respiratory Medicine Department, Medical School, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - Agamemnon Bakakos
- 1St Respiratory Medicine Department, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Diseases Hospital, Athens, Greece
| | - Evangelia Fouka
- Respiratory Medicine Department, Aristotle University of Thessaloniki, G Papanikolaou Hospital, Thessaloniki, Greece
| | - Petros Bakakos
- 1St Respiratory Medicine Department, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Diseases Hospital, Athens, Greece
| | - Stelios Loukides
- 2Nd Respiratory Medicine Department, Medical School, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - Paschalis Steiropoulos
- Department of Pneumonology, Medical School, Democritus University of Thrace, 68100, Alexandroupolis, Greece.
| | - Andriana I Papaioannou
- 1St Respiratory Medicine Department, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Diseases Hospital, Athens, Greece
| |
Collapse
|
4
|
Lindkvist M, Göthlin Eremo A, Paramel GV, Anisul Haque S, Rydberg Millrud C, Rattik S, Grönberg C, Liberg D, Sirsjö A, Fransén K. IL1RAP Expression in Human Atherosclerosis: A Target of Novel Antibodies to Reduce Vascular Inflammation and Adhesion. J Am Heart Assoc 2025; 14:e039557. [PMID: 40371594 DOI: 10.1161/jaha.124.039557] [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: 10/21/2024] [Accepted: 04/09/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Blockade of IL1RAP (interleukin 1 receptor associated protein) was recently shown to reduce atherosclerosis in mice, but the effect on human vascular cells is largely unknown. Targeting the IL1RAP coreceptor represents a novel strategy to block the IL1RAP-dependent cytokines IL (interleukin)-1, IL-33, and IL-36. In the present study, we aimed to evaluate the role of novel antibodies targeting IL1RAP to reduce the effects of IL-1β, IL-33, or IL-36γ in human vascular cells. METHODS Expression of IL1RAP was observed in human atherosclerotic plaques by immunohistochemistry and microarray and in endothelial cells by flow cytometry. Endothelial cells were cultured with IL-1β, IL-33, or IL-36γ cytokines with or without IL1RAP antibodies and analyzed with Olink proteomics, ELISA, Western blot, and real-time quantitative polymerase chain reaction. The functional effect of IL1RAP antibodies on endothelial cells were analyzed with adhesion and permeability assays. RESULTS Olink proteomics showed inhibition of the inflammatory proteins LIF (leukemia inhibitory factor), OPG (osteoprotegerin), CCL4 (C-C motif chemokine ligand 4), and MCP-3 (monocyte chemoattractant protein 3) by IL1RAP-blockade in endothelial cells after IL-1β stimulation. In addition, the IL1RAP antibodies inhibited IL-1β, and IL-33 induced IL-6 and IL-8 secretion. Secretion of MCP-1 (monocyte chemoattractant protein 1) was induced by IL-1β, IL-33, and IL-36γ, and subsequently was inhibited by IL1RAP antibodies. Similar effects were found on mRNA expression level. Endothelial expression of the adhesion markers ICAM1, VCAM1, and SELE were significantly reduced by IL1RAP antibodies, and neutrophil adhesion to endothelial cells induced by IL-1β and IL-33 was reduced by IL1RAP blockade. In human atherosclerotic lesions, IL1RAP expression correlated with markers of inflammation like IL6, IL8, and MCP1. CONCLUSIONS IL1RAP-targeting antibodies can reduce the expression of inflammatory cytokines and markers of adhesion in endothelial cells, which may be of importance for future putative targeted treatments against cardiovascular disease.
Collapse
Affiliation(s)
- Madelene Lindkvist
- Cardiovascular Research Centre, Faculty of Medicine and Health Örebro University Örebro Sweden
- School of Medical Sciences, Faculty of Medicine and Health Örebro University Örebro Sweden
| | - Anna Göthlin Eremo
- Cardiovascular Research Centre, Faculty of Medicine and Health Örebro University Örebro Sweden
- Department of Clinical Research Laboratory, School of Medical Sciences, Faculty of Medicine and Health Örebro University Örebro Sweden
| | - Geena Varghese Paramel
- Cardiovascular Research Centre, Faculty of Medicine and Health Örebro University Örebro Sweden
- School of Medical Sciences, Faculty of Medicine and Health Örebro University Örebro Sweden
| | - Sheikh Anisul Haque
- Cardiovascular Research Centre, Faculty of Medicine and Health Örebro University Örebro Sweden
- School of Medical Sciences, Faculty of Medicine and Health Örebro University Örebro Sweden
| | | | | | | | | | - Allan Sirsjö
- Cardiovascular Research Centre, Faculty of Medicine and Health Örebro University Örebro Sweden
- School of Medical Sciences, Faculty of Medicine and Health Örebro University Örebro Sweden
| | - Karin Fransén
- Cardiovascular Research Centre, Faculty of Medicine and Health Örebro University Örebro Sweden
- School of Medical Sciences, Faculty of Medicine and Health Örebro University Örebro Sweden
| |
Collapse
|
5
|
Leventoğlu E, Bakkaloğlu SA. A new era in the treatment of kidney diseases: NLRP3 inflammasome and cytokine-targeted therapies. Pediatr Nephrol 2025; 40:1515-1521. [PMID: 39485496 DOI: 10.1007/s00467-024-06578-0] [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: 08/13/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 11/03/2024]
Abstract
The kidneys are crucial for filtering blood, managing overall body water, electrolyte, and acid-base balance, and regulating blood pressure. They remove metabolic waste products, toxins, and drugs. In addition, they limit inflammation by clearing cytokines and reduce immune cell activation by removing bacterial components. Dendritic cells (DCs) in the kidney maintain peripheral tolerance. About 85% of filtered water is reabsorbed by the proximal tubule, exposing distal nephron cells to high concentrations of low molecular weight antigens. These antigens are captured by DCs, helping to inactivate potentially autoreactive T cells and maintain tolerance to circulating antigens. In kidney failure, immune function is severely compromised due to the retention of toxins and cytokines, which activate immune cells and increase systemic inflammation. The kidneys are also vulnerable to immune-mediated diseases. Loss of immune homeostasis, characterized by over- or under-activity of the immune response, can adversely affect kidney function. With advances in immunology and cellular biology, biologic therapies targeting various pathways involved in the pathophysiology of kidney diseases are being developed. In this review, the immunologic aspects of kidney diseases and focus on cytokine-based therapies that may hold promise for the treatment of kidney diseases in the future will be presented.
Collapse
Affiliation(s)
- Emre Leventoğlu
- Department of Pediatric Nephrology, Konya City Hospital, Konya, Turkey.
| | - Sevcan A Bakkaloğlu
- Faculty of Medicine, Department of Pediatric Nephrology, Gazi University, Ankara, Turkey
| |
Collapse
|
6
|
Zhu Y, Guo Y, Guo P, Zhang J, He Y, Xia Y, Wei Z, Dai Y. Estrogen receptor β activation alleviates inflammatory bowel disease by suppressing NLRP3-dependent IL-1β production in macrophages via downregulation of intracellular calcium level. J Adv Res 2025; 71:571-584. [PMID: 38844124 DOI: 10.1016/j.jare.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 05/30/2024] [Accepted: 06/02/2024] [Indexed: 06/11/2024] Open
Abstract
INTRODUCTION Although several estrogen receptor β (ERβ) agonists have been reported to alleviate IBD, the pivotal mechanism remains obscure. OBJECTIVES To examine the effects and mechanisms of ERβ activation on cytokine/chemokine networks in colitis mice. METHODS Dextran sulfate sodium salt (DSS) and trinitro-benzene-sulfonic acid (TNBS) were used to induce mouse colitis model. Multiple molecular biological methods were employed to evaluate the severity of mouse colitis and the level of cytokine and/or chemokine. RESULTS Bioinformatics analysis, ELISA and immunofluorescence results showed that the targeted cytokines and/or chemokines associated with ERβ expression and activation is IL-1β, and the anti-colitis effect of ERβ activation was significantly attenuated by the overexpression of AAV9-IL-1β. Immunofluorescence analysis indicated that ERβ activation led to most evident downregulation of IL-1β expression in colonic macrophages as compared to monocytes and neutrophils. Given the pivotal roles of NLRP3, NLRC4, and AIM2 inflammasome activation in the production of IL-1β, we examined the influence of ERβ activation on inflammasome activity. ELISA and WB results showed that ERβ activation selectively blocked the NLRP3 inflammasome assembly-mediated IL-1β secretion. The calcium-sensing receptor (CaSR) and calcium signaling play crucial roles in the assembly of the NLRP3 inflammasome. WB and immunofluorescence results showed that ERβ activation reduced intracellular CaSR expression and calcium signaling in colonic macrophages. Combination with CaSR overexpression plasmid reversed the suppressive effect of ERβ activation on NLRP3 inflammasome assembly, and counteracting the downregulation of IL-1β secretion. CONCLUSION Our research uncovers that the anti-colitis effect of ERβ activation is accomplished through the reduction of IL-1β levels in colonic tissue, achieved by specifically decreasing CaSR expression in macrophages to lower intracellular calcium levels and inhibit NLRP3 inflammasome assembly-mediated IL-1β production.
Collapse
Affiliation(s)
- Yanrong Zhu
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China
| | - Yilei Guo
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China
| | - Pengxiang Guo
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China
| | - Jing Zhang
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China
| | - Yue He
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China
| | - Yufeng Xia
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China
| | - Zhifeng Wei
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China.
| | - Yue Dai
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China.
| |
Collapse
|
7
|
Shen Y, Lin P. The Role of Cytokines in Postherpetic Neuralgia. J Integr Neurosci 2025; 24:25829. [PMID: 40302252 DOI: 10.31083/jin25829] [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: 07/24/2024] [Revised: 10/02/2024] [Accepted: 10/23/2024] [Indexed: 05/02/2025] Open
Abstract
Nerve injury is a significant cause of postherpetic neuralgia (PHN). It is marked by upregulated expression of cytokines secreted by immune cells such as tumor necrosis factor alpha, interleukin 1 beta (IL-1β), IL-6, IL-18, and IL-10. In neuropathic pain (NP) due to nerve injury, cytokines are important for the induction of neuroinflammation, activation of glial cells, and expression of cation channels. The release of chemokines due to nerve injury promotes immune cell infiltration, recruiting inflammatory cytokines and further amplifying the inflammatory response. The resulting disequilibrium in neuroimmune response and neuroinflammation leads to a reduction of nerve fibers, altered nerve excitability, and neuralgia. PHN is a typical NP and cytokines may induce PHN by promoting central and peripheral sensitization. Currently, treating PHN is challenging and research on the role of cytokine signaling pathways in PHN is lacking. This review summarizes the potential mechanisms of cytokine-mediated PHN and discusses the cytokine signaling pathways associated with the central and peripheral sensitization of PHN. By elucidating the mechanisms of cytokines, the cells and molecules that regulate cytokines, and their signaling systems in PHN, this review reveals important research developments regarding cytokines and their signaling pathways mediating PHN, highlighting new targets of action for the development of analgesic drugs.
Collapse
Affiliation(s)
- Yunyan Shen
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, 310053 Hangzhou, Zhejiang, China
| | - Ping Lin
- Department of Geriatrics, Hangzhou Third People's Hospital, 310009 Hangzhou, Zhejiang, China
| |
Collapse
|
8
|
Zhou Y, Wang K, Ren M, Wang J, Wang F, Zhuang B, Chen L, Zhang Z, Wang C. Identification and functional validation of ACSL1 as a biomarker regulating ferroptosis in nucleus pulposus cell. Biosci Rep 2025; 45:BSR20241414. [PMID: 40042449 DOI: 10.1042/bsr20241414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 02/18/2025] [Accepted: 03/04/2025] [Indexed: 04/05/2025] Open
Abstract
Intervertebral disc degeneration (IVDD) is a prevalent musculoskeletal disorder characterized by the deterioration of nucleus pulposus (NP) cells, leading to significant impairments in patients' quality of life. Elucidating the molecular mechanisms underlying IVDD is essential for developing effective therapeutic strategies. In this study, we utilized weighted gene co-expression network analysis to identify key module eigengenes (MEs) from the GSE124272 dataset, combined with differential gene expression analysis to pinpoint differentially expressed genes (DEGs). Functional enrichment analysis revealed that MEs were primarily associated with lipid metabolism and immune response, while DEGs were enriched in immune response and cell proliferation pathways. By integrating MEs, DEGs, and ferroptosis-related genes, we identified six hub genes (acyl-CoA synthetase long-chain family member 1 [ACSL1], BACH1, CBS, CP, AKR1C1, and AKR1C3). Consensus clustering analysis classified samples into two immune-related subgroups, C1 and C2, with single-sample gene set enrichment analysis demonstrating distinct immune scores between the subgroups. Notably, ACSL1 showed the strongest correlation with immune cell infiltration and was significantly up-regulated in the C1 subgroup, which exhibited higher immune scores. In vitro experiments confirmed elevated ACSL1 expression in an IL-1β-induced degenerative NP cell model. Silencing ACSL1 improved cell viability, reduced apoptosis, and restored mitochondrial membrane potential, alongside significant changes in intracellular Fe2+, malondialdehyde, and glutathione levels. In vivo experiments further validated increased ACSL1 expression in intervertebral disc tissues of IVDD rats. Collectively, these findings highlight ACSL1 as a potential biomarker for the early diagnosis of IVDD and a promising therapeutic target.
Collapse
Affiliation(s)
- Yichi Zhou
- Department of Spine Surgery, The First Affiliated Hospital of Fujian Medical University, 20 Cha Zhong Road, Taijiang District, Fuzhou, Fujian Province, 350000, China
- Spine Center, Wuhan Fourth Hospital, 473 Hanzheng Road, Qiaokou District, Wuhan, Hubei Province, 430000, China
| | - Ke Wang
- Department of Spine Surgery, The First Affiliated Hospital of Fujian Medical University, 20 Cha Zhong Road, Taijiang District, Fuzhou, Fujian Province, 350000, China
| | - Min Ren
- Department of Spine Surgery, The First Affiliated Hospital of Fujian Medical University, 20 Cha Zhong Road, Taijiang District, Fuzhou, Fujian Province, 350000, China
| | - Jiebin Wang
- Department of Spine Surgery, The First Affiliated Hospital of Fujian Medical University, 20 Cha Zhong Road, Taijiang District, Fuzhou, Fujian Province, 350000, China
| | - Fanglin Wang
- Department of Spine Surgery, The First Affiliated Hospital of Fujian Medical University, 20 Cha Zhong Road, Taijiang District, Fuzhou, Fujian Province, 350000, China
| | - Bingbing Zhuang
- Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Lin Chen
- Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhiqiang Zhang
- Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Changsheng Wang
- Department of Spine Surgery, The First Affiliated Hospital of Fujian Medical University, 20 Cha Zhong Road, Taijiang District, Fuzhou, Fujian Province, 350000, China
| |
Collapse
|
9
|
Luo J, Yang Q, Jiang W, Liu Y, Hu Q, Peng X. The interaction between Angelica sinensis polysaccharide ASP-2pb and specific gut bacteria alleviates rheumatoid arthritis in rats. Int J Biol Macromol 2025; 301:140473. [PMID: 39889994 DOI: 10.1016/j.ijbiomac.2025.140473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/07/2025] [Accepted: 01/27/2025] [Indexed: 02/03/2025]
Abstract
Angelica sinensis polysaccharide (ASP) alleviated Rheumatoid arthritis (RA), but whether the relief was attributed to ASP itself or its microbial metabolites remained unclear. We characterized the main fraction of ASP (ASP-2pb) as a polysaccharide with molecular weight of 92.02 kDa. It contained approximately 48 repeating units of →6)-β-D-Galp-(1 → 3)-4-OMe-β-D-Galp-(1 → 4)-α-D-GalpA-(1 → 6)-β-D-Galp-(1 → 3)-4-OMe-β-D-Galp-(1→3)-β-D-Galp-(1 → 3)-β-D-Galp-(1 → 3)-β-D-Galp-(1 → with branches of Araf and Galp. Using ASP-2pb as intervention, the symptoms of RA in rats including joint swelling and inflammation were alleviated. Pseudo-germ-free animal test confirmed the necessity of specific gut bacteria during this alleviation. Bacteria such as Candidatus_Saccharimonas, Lactobacillus, Bifidobacterium, Faecalibaculum, Parvibacter, Ruminococcus_torques_group, Fournierella and Alloprevotella ought to be the key bacteria. Metabolites generated by these gut bacteria such as myristoleic acid, cuminaldehyde, 4-deoxypyridoxine and galactosylhydroxylysine, should be the key to RA remission. Therefore, specific metabolites were the consequence of the interaction between ASP-2pb and specific intestinal bacteria, and were responsible for the RA improvement.
Collapse
Affiliation(s)
- Jianming Luo
- Department of Food Science and Engineering, Jinan University, 601 Huangpu Avenue, Guangzhou, Guangdong 510632, China.
| | - Qianyi Yang
- Department of Food Science and Engineering, Jinan University, 601 Huangpu Avenue, Guangzhou, Guangdong 510632, China
| | - Wenwen Jiang
- Department of Food Science and Engineering, Jinan University, 601 Huangpu Avenue, Guangzhou, Guangdong 510632, China
| | - Yanghanxiu Liu
- Department of Food Science and Engineering, Jinan University, 601 Huangpu Avenue, Guangzhou, Guangdong 510632, China
| | - Qing Hu
- Department of Food Science and Engineering, Jinan University, 601 Huangpu Avenue, Guangzhou, Guangdong 510632, China
| | - Xichun Peng
- Department of Food Science and Engineering, Jinan University, 601 Huangpu Avenue, Guangzhou, Guangdong 510632, China.
| |
Collapse
|
10
|
He X, He Y, Deng X, Lu N, Li A, Gao S, He S, Wang Y, Fu N, Wang Z, Nie Y, Xu L. Rv2741 Promotes Mycobacterium Survival by Modulating Macrophage Function via the IL-1α-MAPK Axis. ACS Infect Dis 2025; 11:676-688. [PMID: 40009799 DOI: 10.1021/acsinfecdis.4c00790] [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] [Indexed: 02/28/2025]
Abstract
One of the primary healthcare problems in the world today is tuberculosis (TB), a chronic infectious illness brought on by Mycobacterium tuberculosis (M. tuberculosis). A distinct family of PE_PGRS proteins, encoded by the M. tuberculosis genome, has attracted more attention because of their involvement in immune evasion and bacterial pathogenicity. Nevertheless, the specific functions and mechanisms of action for the majority of PE_PGRS proteins remain largely unexplored. This study focuses on the Rv2741 (PE_PGRS47) gene, which is exclusively present in pathogenic mycobacteria. To examine the function of Rv2741 in host-pathogen interactions, we created recombinant strains of Mycobacterium smegmatis (M. smegmatis) that expressed the M. tuberculosis Rv2741 gene. IL-1α was found to be a key mediator of host response modulation by Rv2741. Rv2741 downregulates the secretion of IL-1α and inhibits the MAPK signaling pathway, particularly the p38 and ERK1/2 pathways, thereby cooperatively inhibiting macrophage autophagy and apoptosis. Meanwhile, the decrease in IL-1α secretion directly leads to changes in the cytokine secretion pattern and a reduction in nitric oxide (NO) production. This multifaceted regulatory mechanism ultimately favors the survival of M. smegmatis in macrophages. This research significantly expands our understanding of Rv2741 function, revealing its crucial role as a multifunctional virulence factor in the immune evasion of M. tuberculosis.
Collapse
Affiliation(s)
- Xintong He
- Department of Pathogenic Biology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yonglin He
- Department of Pathogenic Biology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xichuan Deng
- Pathogen Biology and Immunology Laboratory, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing 400016, China
| | - Nan Lu
- Department of Pathogenic Biology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Anlong Li
- Department of Pathogenic Biology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Sijia Gao
- Department of Pathogenic Biology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Shiyan He
- Department of Pathogenic Biology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yuran Wang
- Department of Pathogenic Biology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Nanzhe Fu
- International Medical School, Chongqing Medical University, Chongqing 400016, China
| | - Zijie Wang
- International Medical School, Chongqing Medical University, Chongqing 400016, China
| | - Yuxin Nie
- The Second Clinical College, Chongqing Medical University, Chongqing 400016, China
| | - Lei Xu
- Department of Pathogenic Biology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
11
|
Yu XJ, Chen LL, Ren ZJ, Li YP, Chen JY, Zhao YX, Jiang JB. Aspirin-based PROTACs as COX-2 degraders for anti-inflammation. Bioorg Med Chem 2025; 119:118061. [PMID: 39793401 DOI: 10.1016/j.bmc.2025.118061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 12/30/2024] [Accepted: 01/01/2025] [Indexed: 01/13/2025]
Abstract
Cyclooxygenase-2 (COX-2) is a key enzyme in the biosynthesis of prostaglandins and plays a special role in the process of inflammatory response. COX-2 is a target of non-steroidal anti-inflammatory drugs (NSAIDs), which can effectively relieve inflammation, pain and fever responses by inhibiting COX-2. Despite the significant study progress of inhibitors targeting COX-2, the development of COX-2 degraders remains insufficient. Proteolysis targeting chimaeras (PROTACs) have recently emerged as a fascinating technology for targeted protein degradation and drug discovery. In this report, we present the design, synthesis and detection of aspirin-based PROTACs that demonstrate effective ubiquitin-proteasome pathway degradation of COX-2 in lipopolysaccharide-stimulated RAW264.7 cells, and the aspirin-based negative PROTACs does not promote the degradation of COX-2. Moreover, we show AspPROTACs could significantly affect proteasome degradation and inflammatory signaling pathways through quantitative proteomic data analysis. These COX-2 degraders offer valuable chemical tools and novel insights for research in anti-inflammatory drugs.
Collapse
Affiliation(s)
- Xuan-Jie Yu
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China; Institute for Inheritance-Based Innovation of Chinese Medicine, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Li-Li Chen
- Institute for Inheritance-Based Innovation of Chinese Medicine, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Zhi-Jie Ren
- Institute for Inheritance-Based Innovation of Chinese Medicine, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Yan-Peng Li
- Institute for Inheritance-Based Innovation of Chinese Medicine, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Jia-Yu Chen
- Institute for Inheritance-Based Innovation of Chinese Medicine, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Yu-Xi Zhao
- Shenzhen Wininnovate Bio-Tech Co., Ltd, 410034 Shenzhen, China
| | - Jian-Bing Jiang
- Institute for Inheritance-Based Innovation of Chinese Medicine, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China.
| |
Collapse
|
12
|
Chen G, Wang A, Wang K, Deng J, Yang J, Chen Q, Ye M, Yan S, Shi G, Lin D. Tyrosol promotes skin flap survival by downregulating the p38/NF-κB signaling pathway. Burns 2025; 51:107334. [PMID: 39721233 DOI: 10.1016/j.burns.2024.107334] [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: 08/12/2024] [Revised: 10/30/2024] [Accepted: 11/23/2024] [Indexed: 12/28/2024]
Abstract
Skin flaps are often used to repair wounds and improve a patient's appearance, particularly after severe burns. Although skin flaps are widely used, they are prone to necrosis, which poses a major clinical challenge. Tyrosol is a natural phenolic antioxidant found in olive oil that has anti-inflammatory, anti-apoptotic, antioxidant, and pro-angiogenic properties. Despite the recognition of these properties, no studies have investigated the effects of tyrosol on flap survival, although tyrosol significantly reduced flap edema and the necrotic area. In a rat study using the McFarland random flap model, gelatin lead oxide angiography showed that angiogenesis was increased significantly in high- and low-dose tyrosol groups. These immunohistochemistry findings highlight the impact of tyrosol on the inflammatory response and angiogenesis, underscoring its potential significance in flap survival. Western blot analysis showed that the expression of p38, NF-κB, and BAX was significantly down-regulated, while that of Bcl-2 was significantly up-regulated, in the high- and low-dose tyrosol groups. This modulation of key signaling pathways and apoptotic proteins not only validates the impact of tyrosol but also reinforces its role in providing protection, thus promoting flap survival. In summary, tyrosol may inhibit inflammation, oxidative stress, and apoptosis by downregulating the p38-NF-κB signaling pathway and promoting angiogenesis, thereby enhancing skin flap survival.
Collapse
Affiliation(s)
- Guodong Chen
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - An Wang
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Kaitao Wang
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Jiapeng Deng
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Jialong Yang
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Qingyu Chen
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Minle Ye
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Shuxu Yan
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Genghe Shi
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Dingsheng Lin
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
13
|
Shen J, Lin A, Jiang A, Xie Z, Cheng Q, Zhang J, Zhang J, Luo P. Dietary inflammatory index predicts cancer mortality in male patients but not female patients: Results from NHANES 1999 to 2014. Nutr Res 2025; 135:52-66. [PMID: 39946775 DOI: 10.1016/j.nutres.2025.01.007] [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: 11/05/2024] [Revised: 01/19/2025] [Accepted: 01/19/2025] [Indexed: 03/14/2025]
Abstract
This study explored sex differences between dietary inflammatory index (DII) and cancer prognosis and their mechanisms. We hypothesized that association between dietary inflammatory index and cancer prognosis differs by sex. The study included 2874 adults with cancer from the National Health and Nutrition Examination Survey covering 1999 to 2014. Mortality status was linked to National Death Index mortality data through 31 December 2019. Cox proportional hazards regression models were applied to calculate hazard risk and 95% confidence intervals (Cis) in male patients and female patients. Sex-specific cancer and nonsex-specific cancer subgroup analyses were performed, and the role of C-reactive protein in sex differences was analyzed. The Cancer Genome Atlas pan-cancer transcriptome data were combined to explore the biological mechanisms of the sex differences. Multivariate Cox regression showed higher DII in male patients correlated with increased all-cause mortality (hazard risk highest vs lowest quartile = 1.57 [95% confidence intervals 1.24-1.98]; P for trend <.01), but not in female patients (P = .44). For sex-specific cancers, higher DII potentially correlated with increased mortality in prostate cancer (unadjusted P for trend = .04), but not in breast (P = .83), ovarian (P = .49), or cervical cancers (P = .91). In melanoma and colon cancer, higher DII correlated with increased mortality in male patients but not female patients. Serum C-reactive protein, interleukin-1 binding, interleukin-35 pathway, and programmed cell death protein 1 pathway may contribute to these sex differences. In conclusion, sex differences exist between DII and mortality risk in cancer patients.
Collapse
Affiliation(s)
- Junyi Shen
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Aimin Jiang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Zhenyu Xie
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jing Zhang
- The Second Department of Infectious Disease, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China; Center of Community-Based Health Research, Fudan University, Shanghai, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
14
|
Zhai Z, Yang C, Yin W, Liu Y, Li S, Ye Z, Xie M, Song X. Engineered Strategies to Interfere with Macrophage Fate in Myocardial Infarction. ACS Biomater Sci Eng 2025; 11:784-805. [PMID: 39884780 DOI: 10.1021/acsbiomaterials.4c02061] [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] [Indexed: 02/01/2025]
Abstract
Myocardial infarction (MI), a severe cardiovascular condition, is typically triggered by coronary artery disease, resulting in ischemic damage and the subsequent necrosis of the myocardium. Macrophages, known for their remarkable plasticity, are capable of exhibiting a range of phenotypes and functions as they react to diverse stimuli within their local microenvironment. In recent years, there has been an increasing number of studies on the regulation of macrophage behavior based on tissue engineering strategies, and its regulatory mechanisms deserve further investigation. This review first summarizes the effects of key regulatory factors of engineered biomaterials (including bioactive molecules, conductivity, and some microenvironmental factors) on macrophage behavior, then explores specific methods for inducing macrophage behavior through tissue engineering materials to promote myocardial repair, and summarizes the role of macrophage-host cell crosstalk in regulating inflammation, vascularization, and tissue remodeling. Finally, we propose some future challenges in regulating macrophage-material interactions and tailoring personalized biomaterials to guide macrophage phenotypes.
Collapse
Affiliation(s)
- Zitong Zhai
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Chang Yang
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Wenming Yin
- Department of Neurology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Yali Liu
- Department of Neurology, Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong 528000, China
| | - Shimin Li
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Ziyi Ye
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Mingxiang Xie
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Xiaoping Song
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
- Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong 510515, China
| |
Collapse
|
15
|
Oliveira ACDR, Figueiredo CS, Raony Í, Von-Held-Ventura JS, Granja MG, Mázala-de-Oliveira T, Pedrosa-Soares VH, dos Santos AA, Giestal-de-Araujo E. Ouabain Counteracts Retinal Ganglion Cell Death Through Modulation of BDNF and IL-1 Signaling Pathways. Brain Sci 2025; 15:123. [PMID: 40002456 PMCID: PMC11853102 DOI: 10.3390/brainsci15020123] [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: 12/19/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Ouabain is a steroid hormone that binds to the sodium pump (Na+, K+-ATPase) at physiological (nanomolar) concentrations, activating different signaling pathways. This interaction has been shown to prevent the axotomy-induced death of retinal ganglion cells (RGCs), although the underlying mechanisms remain unclear. Objective: In this study, we investigated potential mechanisms by which ouabain promotes RGC survival using primary cultures of rat neural retina. Results: Our findings indicate that ouabain regulates brain-derived neurotrophic factor (BDNF) signaling in retinal cells via matrix metalloproteinase-9-mediated processing of proBDNF to mature BDNF (mBDNF) and by increasing the phosphorylation of the mBDNF receptor, tropomyosin-related receptor kinase B. Ouabain also enhances the maturation of interleukin (IL)-1β through the increased activation of caspase-1, which mediates the processing of proIL-1β into IL-1β, and transiently upregulates both IL-1 receptor and IL-1 receptor antagonist (IL-1Ra). Treatment using either IL-1β or IL-1Ra alone is sufficient to enhance RGC survival similarly to that achieved with ouabain. Finally, we further show that ouabain prevents RGC death through a complex signaling mechanism shared by BDNF and IL-1β, which includes the activation of the Src and protein kinase C pathways. Conclusions: Collectively, these results suggest that ouabain stimulates the maturation and signaling of both BDNF and IL-1β, which act as key mediators of RGC survival.
Collapse
Affiliation(s)
- Amanda Candida da Rocha Oliveira
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niteroi 24020-141, Brazil; (A.C.d.R.O.); (C.S.F.); (J.S.V.-H.-V.); (T.M.-d.-O.); (V.H.P.-S.)
- National Institute of Science and Technology on Neuroimmunomodulation—INCT-NIM, Oswaldo Cruz Foundation, Rio de Janeiro 21041-250, Brazil;
| | - Camila Saggioro Figueiredo
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niteroi 24020-141, Brazil; (A.C.d.R.O.); (C.S.F.); (J.S.V.-H.-V.); (T.M.-d.-O.); (V.H.P.-S.)
- National Institute of Science and Technology on Neuroimmunomodulation—INCT-NIM, Oswaldo Cruz Foundation, Rio de Janeiro 21041-250, Brazil;
| | - Ícaro Raony
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Juliana Salles Von-Held-Ventura
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niteroi 24020-141, Brazil; (A.C.d.R.O.); (C.S.F.); (J.S.V.-H.-V.); (T.M.-d.-O.); (V.H.P.-S.)
| | - Marcelo Gomes Granja
- Research, Innovation, and Surveillance Center for COVID-19 and Health Emergencies, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil;
| | - Thalita Mázala-de-Oliveira
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niteroi 24020-141, Brazil; (A.C.d.R.O.); (C.S.F.); (J.S.V.-H.-V.); (T.M.-d.-O.); (V.H.P.-S.)
- National Institute of Science and Technology on Neuroimmunomodulation—INCT-NIM, Oswaldo Cruz Foundation, Rio de Janeiro 21041-250, Brazil;
| | - Vinícius Henrique Pedrosa-Soares
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niteroi 24020-141, Brazil; (A.C.d.R.O.); (C.S.F.); (J.S.V.-H.-V.); (T.M.-d.-O.); (V.H.P.-S.)
| | - Aline Araujo dos Santos
- National Institute of Science and Technology on Neuroimmunomodulation—INCT-NIM, Oswaldo Cruz Foundation, Rio de Janeiro 21041-250, Brazil;
- Department of Physiology and Pharmacology, Biomedical Institute, Federal Fluminense University, Niteroi 24210-130, Brazil
| | - Elizabeth Giestal-de-Araujo
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niteroi 24020-141, Brazil; (A.C.d.R.O.); (C.S.F.); (J.S.V.-H.-V.); (T.M.-d.-O.); (V.H.P.-S.)
- National Institute of Science and Technology on Neuroimmunomodulation—INCT-NIM, Oswaldo Cruz Foundation, Rio de Janeiro 21041-250, Brazil;
| |
Collapse
|
16
|
Zhang FS, Li HJ, Yu X, Song YP, Ren YF, Qian XZ, Liu JL, Li WX, Huang YR, Gao K. Global trends and hotspots of type 2 diabetes in children and adolescents: A bibliometric study and visualization analysis. World J Diabetes 2025; 16:96032. [PMID: 39817223 PMCID: PMC11718446 DOI: 10.4239/wjd.v16.i1.96032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 09/30/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Epidemiological surveys indicate an increasing incidence of type 2 diabetes mellitus (T2DM) among children and adolescents worldwide. Due to rapid disease progression, severe long-term cardiorenal complications, a lack of effective treatment strategies, and substantial socioeconomic burdens, it has become an urgent public health issue that requires management and resolution. Adolescent T2DM differs from adult T2DM. Despite a significant increase in our understanding of youth-onset T2DM over the past two decades, the related review and evidence-based content remain limited. AIM To visualize the hotspots and trends in pediatric and adolescent T2DM research and to forecast their future research themes. METHODS This study utilized the terms "children", "adolescents", and "type 2 diabetes", retrieving relevant articles published between 1983 and 2023 from three citation databases within the Web of Science Core Collection (SCI, SSCI, ESCI). Utilizing CiteSpace and VoSviewer software, we analyze and visually represent the annual output of literature, countries involved, and participating institutions. This allows us to predict trends in this research field. Our analysis encompasses co-cited authors, journal overlays, citation overlays, time-zone views, keyword analysis, and reference analysis, etc. RESULTS A total of 9210 articles were included, and the annual publication volume in this field showed a steady growth trend. The United States had the highest number of publications and the highest H-index. The United States also had the most research institutions and the strongest research capacity. The global hot journals were primarily diabetes professional journals but also included journals related to nutrition, endocrinology, and metabolism. Keyword analysis showed that research related to endothelial dysfunction, exposure risk, cardiac metabolic risk, changes in gut microbiota, the impact on comorbidities and outcomes, etc., were emerging keywords. They have maintained their popularity in this field, suggesting that these areas have garnered significant research interest in recent years. CONCLUSION Pediatric and adolescent T2DM is increasingly drawing global attention, with genes, behaviors, environmental factors, and multisystemic interventions potentially emerging as future research hot spots.
Collapse
Affiliation(s)
- Fang-Shuo Zhang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Hai-Jing Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xue Yu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yi-Ping Song
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yan-Feng Ren
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xuan-Zhu Qian
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jia-Li Liu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Wen-Xun Li
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yi-Ran Huang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Kuo Gao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| |
Collapse
|
17
|
Ni M, Wang Y, Yang J, Ma Q, Pan W, Li Y, Xu Q, Lv H, Wang Y. IL-33 aggravates extranodal NK/T cell lymphoma aggressiveness and angiogenesis by activating the Wnt/β-catenin signaling pathway. Mol Cell Biochem 2025; 480:265-278. [PMID: 38443748 DOI: 10.1007/s11010-024-04944-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 01/16/2024] [Indexed: 03/07/2024]
Abstract
Extranodal NK/T cell lymphoma (ENKTCL) is an extremely aggressive form of lymphoma and lacks of specific diagnostic markers. The study intended to unearth the role of interleukin-33 (IL-33) in ENKTCL. RT-qPCR was conducted to assess mRNA levels of ENKTCL tissues and cells, while western blot assay was performed for evaluating protein levels. Plate cloning experiment and transwell assay were employed to measure aggressiveness of ENKTCL. Tube formation assay was executed to determine the angiogenesis ability. Mice ENKTCL xenograft model was designed to probe the impacts of IL-33 in vivo. IL-33 and suppression of tumorigenicity 2 receptor (ST2, receptor of IL-33) were enhanced in ENKTCL. IL-33 inhibition suppressed viability, migration, and invasion of ENKTCL cells. Moreover, IL-33 knockdown restricted angiogenesis in human umbilical vein endothelial cells (HUVECs). Furthermore, Wnt/β-catenin pathway associated proteins (β-catenin, c-myc, and cyclin D1) were downregulated by loss of IL-33. However, these impacts were overturned by Wnt/β-catenin signaling agonist lithium chloride (LiCl). Additionally, IL-33 silencing exerted anti-tumor effect via Wnt/β-catenin pathway in vivo. Silencing of IL-33 inhibited ENKTCL tumorigenesis and angiogenesis by inactivating Wnt/β-catenin signaling pathway. As such, IL-33 might be a prospective treatment target for ENKTCL.
Collapse
Affiliation(s)
- Mingli Ni
- Department of Oncology, The First Affiliated Hospital of Henan University of CM, Zhengzhou, 450099, Henan, China
- Medical Oncology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, 471099, Henan, China
| | - Yuhui Wang
- Day Operating Room, Luoyang Central Hospital, Luoyang, 471099, Henan, China
| | - Jiezhi Yang
- Medical Oncology, Luoyang Central Hospital, Luoyang, 471099, Henan, China
| | - Qianwen Ma
- Medical Oncology, Luoyang Central Hospital, Luoyang, 471099, Henan, China
| | - Wei Pan
- Medical Oncology, Luoyang Central Hospital, Luoyang, 471099, Henan, China
| | - Yulin Li
- Medical Oncology, Luoyang Central Hospital, Luoyang, 471099, Henan, China
| | - Qian Xu
- Medical Oncology, Luoyang Central Hospital, Luoyang, 471099, Henan, China
| | - Hongqiong Lv
- Medical Oncology, Luoyang Central Hospital, Luoyang, 471099, Henan, China
| | - Yunlong Wang
- Department of Oncology, The First Affiliated Hospital of Henan University of CM, Zhengzhou, 450099, Henan, China.
- Henan Bioengineering Research Center, No. 81, Zhengshang Road, Zhengzhou, 450066, Henan, China.
| |
Collapse
|
18
|
Mgwenya TN, Abrahamse H, Houreld NN. Photobiomodulation studies on diabetic wound healing: An insight into the inflammatory pathway in diabetic wound healing. Wound Repair Regen 2025; 33:e13239. [PMID: 39610015 PMCID: PMC11628774 DOI: 10.1111/wrr.13239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 08/06/2024] [Accepted: 09/02/2024] [Indexed: 11/30/2024]
Abstract
Diabetes mellitus remains a global challenge to public health as it results in non-healing chronic ulcers of the lower limb. These wounds are challenging to heal, and despite the different treatments available to improve healing, there is still a high rate of failure and relapse, often necessitating amputation. Chronic diabetic ulcers do not follow an orderly progression through the wound healing process and are associated with a persistent inflammatory state characterised by the accumulation of pro-inflammatory macrophages, cytokines and proteases. Photobiomodulation has been successfully utilised in diabetic wound healing and involves illuminating wounds at specific wavelengths using predominantly light-emitting diodes or lasers. Photobiomodulation induces wound healing through diminishing inflammation and oxidative stress, among others. Research into the application of photobiomodulation for wound healing is current and ongoing and has drawn the attention of many researchers in the healthcare sector. This review focuses on the inflammatory pathway in diabetic wound healing and the influence photobiomodulation has on this pathway using different wavelengths.
Collapse
Affiliation(s)
- Tintswalo N. Mgwenya
- Laser Research Centre, Faculty of Health SciencesUniversity of JohannesburgJohannesburgGautengSouth Africa
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health SciencesUniversity of JohannesburgJohannesburgGautengSouth Africa
| | - Nicolette N. Houreld
- Laser Research Centre, Faculty of Health SciencesUniversity of JohannesburgJohannesburgGautengSouth Africa
| |
Collapse
|
19
|
Hansman DS, Du J, Casson RJ, Peet DJ. Eye on the horizon: The metabolic landscape of the RPE in aging and disease. Prog Retin Eye Res 2025; 104:101306. [PMID: 39433211 PMCID: PMC11833275 DOI: 10.1016/j.preteyeres.2024.101306] [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: 08/29/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 10/23/2024]
Abstract
To meet the prodigious bioenergetic demands of the photoreceptors, glucose and other nutrients must traverse the retinal pigment epithelium (RPE), a polarised monolayer of cells that lie at the interface between the outer retina and the choroid, the principal vascular layer of the eye. Recent investigations have revealed a metabolic ecosystem in the outer retina where the photoreceptors and RPE engage in a complex exchange of sugars, amino acids, and other metabolites. Perturbation of this delicate metabolic balance has been identified in the aging retina, as well as in age-related macular degeneration (AMD), the leading cause of blindness in the Western world. Also common in the aging and diseased retina are elevated levels of cytokines, oxidative stress, advanced glycation end-products, increased growth factor signalling, and biomechanical stress - all of which have been associated with metabolic dysregulation in non-retinal cell types and tissues. Herein, we outline the role of these factors in retinal homeostasis, aging, and disease. We discuss their effects on glucose, mitochondrial, lipid, and amino acid metabolism in tissues and cell types outside the retina, highlighting the signalling pathways through which they induce these changes. Lastly, we discuss promising avenues for future research investigating the roles of these pathological conditions on retinal metabolism, potentially offering novel therapeutic approaches to combat age-related retinal disease.
Collapse
Affiliation(s)
- David S Hansman
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia.
| | - Jianhai Du
- Department of Ophthalmology and Visual Sciences, Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV 26506, USA
| | - Robert J Casson
- Discipline of Ophthalmology and Visual Science, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Daniel J Peet
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
20
|
Berdeville CHDSF, Silva-Amaral D, Dalgalarrondo P, Banzato CEM, Martins-de-Souza D. A scoping review of protein biomarkers for schizophrenia: State of progress, underlying biology, and methodological considerations. Neurosci Biobehav Rev 2025; 168:105949. [PMID: 39577820 DOI: 10.1016/j.neubiorev.2024.105949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/07/2024] [Accepted: 11/17/2024] [Indexed: 11/24/2024]
Abstract
Schizophrenia is characterized by symptoms such as delusions, hallucinations, and avolition. The diagnosis is clinical, based on interviews and the main treatment involves antipsychotics. Currently, given the lack of clinically applicable biomarkers for schizophrenia, there is no molecular test based on its biological mechanisms to assist psychiatrists either in the prediction or diagnosis of the disorder, nor to measure medication efficacy. This scoping review assessed original articles in English about protein biomarkers for schizophrenia with samples that could be used in a clinical context, classifying them into diagnosis, prognosis, therapeutics, risk for psychosis, and side-effects. The search was conducted on PubMed and key findings were inserted on a summary table. We discussed the methodologies used in these papers, suggested protein panels for validation in longitudinal research, and proposed a hypothesis to explain the observed variability in results. This heterogeneity is explored in light of the debated validity of this construct, applying recent discussions and the disorder's history. Our data suggest that there is insufficient evidence to integrate protein biomarkers into clinical psychiatry for schizophrenia, not due to study quality, but possibly due to flaws in the current diagnostic system. We propose exploring alternative categorization systems.
Collapse
Affiliation(s)
| | - Danyelle Silva-Amaral
- Laboratory of Neuroproteomics, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Paulo Dalgalarrondo
- Department of Psychiatry, School of Medical Sciences, University of Campinas, Campinas, Brazil; Postgraduate Program in Child and Adolescent Health, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Claudio E M Banzato
- Department of Psychiatry, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Institute of Biology, University of Campinas, Campinas, Brazil; D'or Institute for Research and Education, São Paulo, Brazil; Experimental Medicine Research Cluster (EMRC), University of Campinas, Campinas, Brazil; National Institute of Biomarkers in Neuropsychiatry, National Council for Scientific and Technological Development, São Paulo, Brazil
| |
Collapse
|
21
|
Zhao L, Tang H, Cheng Z. Pharmacotherapy of Liver Fibrosis and Hepatitis: Recent Advances. Pharmaceuticals (Basel) 2024; 17:1724. [PMID: 39770566 PMCID: PMC11677259 DOI: 10.3390/ph17121724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/05/2024] [Accepted: 12/17/2024] [Indexed: 01/03/2025] Open
Abstract
Liver fibrosis is a progressive scarring process primarily caused by chronic inflammation and injury, often closely associated with viral hepatitis, alcoholic liver disease, metabolic dysfunction-associated steatotic liver disease (MASLD), drug-induced liver injury, and autoimmune liver disease (AILD). Currently, there are very few clinical antifibrotic drugs available, and effective targeted therapy is lacking. Recently, emerging antifibrotic drugs and immunomodulators have shown promising results in animal studies, and some have entered clinical research phases. This review aims to systematically review the molecular mechanisms underlying liver fibrosis, focusing on advancements in drug treatments for hepatic fibrosis. Furthermore, since liver fibrosis is a progression or endpoint of many diseases, it is crucial to address the etiological treatment and secondary prevention for liver fibrosis. We will also review the pharmacological treatments available for common hepatitis leading to liver fibrosis.
Collapse
Affiliation(s)
- Liangtao Zhao
- Hepato-Pancreato-Biliary Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China;
| | - Haolan Tang
- School of Medicine, Southeast University, Nanjing 210009, China;
| | - Zhangjun Cheng
- Hepato-Pancreato-Biliary Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China;
| |
Collapse
|
22
|
Butler JJ, Dankert JF, Keller LE, Azam MT, Dahmen J, Kerkhoffs GMMJ, Kennedy JG. Assessment of the Monocyte Subpopulations and M1/M2 Macrophage Ratio in Concentrated Bone Marrow Aspirate. Cartilage 2024:19476035241304308. [PMID: 39651680 PMCID: PMC11626554 DOI: 10.1177/19476035241304308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/29/2024] [Accepted: 11/17/2024] [Indexed: 12/11/2024] Open
Abstract
OBJECTIVE The purpose of this study was to determine the M1/M2 macrophage ratio in concentrated bone marrow aspirate (cBMA) in patients undergoing surgical intervention augmented with cBMA for osteochondral lesions of the talus (OLTs). DESIGN Samples of peripheral blood (PB), bone marrow aspirate (BMA), and cBMA were collected during the procedure. The samples were analyzed by automated cell counting and multicolor fluorescence-activated cell sorting with specific antibodies recognizing monocytes (CD14+ CD16+) and the M1 (CD86+) and M2 (CD163+CD206+) populations within that monocyte population. Cytokine concentrations within the samples were evaluated with enzyme-linked immunosorbent assay (ELISA). The composition of cBMA was compared between 2 commercially available BMA concentration systems. RESULTS Thirty-eight patients with a mean age of 43.2 ± 10.1 years old undergoing a surgical procedure for the treatment of OLTs involving the use of cBMA were included. cBMA had a mean fold increase of 4.7 for all white blood cells, 6.1 for monocytes, 7.9 for lymphocytes, 2.4 for neutrophils, and 9.6 for platelets when compared to BMA. The mean M1/M2 ratio for PB, BMA, and cBMA was 15.2 ± 12.0, 20.8 ± 13.3, and 22.1 ± 16.0, respectively. There was a statistically significant higher concentration of interleukin-1 receptor antagonist (IL-1Ra) in the cBMA sample (8243.3 ± 14,837.4 pg/mL) compared to both BMA (3143.0 ± 2218.5 pg/mL) and PB (1847.5 ± 1520.4 pg/mL) samples. The IL-1Ra/IL-1β ratio for PB, BMA, and cBMA was 790.6 ± 581.9, 764.7 ± 675.2, and 235.7 ± 192.1, respectively. There was no difference in the cBMA M1/M2 ratio (19.0 ± 11.1 vs 24.0 ± 18.3) between the Magellan (Isto Biologics, Hopkinton, Massachusetts) and Angel systems (Arthrex Inc, Naples, Florida). CONCLUSION This prospective study found that the M1/M2 ratio in cBMA was 22.1 ± 16.0, with significant patient to patient variation observed. Overall, there was no statistically significant difference in the M1/M2 ratio across PB, BMA, and cBMA samples. This is the first study to characterize the macrophage subpopulation within cBMA, which may have significant clinical implications in future studies.
Collapse
Affiliation(s)
- James J. Butler
- Foot and Ankle Division, Department of Orthopaedic Surgery, NYU Langone Health, New York City, NY, USA
| | - John F. Dankert
- Foot and Ankle Division, Department of Orthopaedic Surgery, NYU Langone Health, New York City, NY, USA
| | - Laura E. Keller
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Mohammad T. Azam
- Foot and Ankle Division, Department of Orthopaedic Surgery, NYU Langone Health, New York City, NY, USA
| | - Jari Dahmen
- Department of Orthopaedic Surgery and Sports Medicine, Amsterdam Movement Sciences, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Gino M. M. J. Kerkhoffs
- Academic Center for Evidence-Based Sports Medicine, Amsterdam UMC, Amsterdam, The Netherlands
- Amsterdam Collaboration for Health and Safety in Sports, International Olympic Committee Research Center, Amsterdam UMC, Amsterdam, The Netherlands
| | - John G. Kennedy
- Foot and Ankle Division, Department of Orthopaedic Surgery, NYU Langone Health, New York City, NY, USA
| |
Collapse
|
23
|
Kindschuh WF, Austin GI, Meydan Y, Park H, Urban JA, Watters E, Pollak S, Saade GR, Chung J, Mercer BM, Grobman WA, Haas DM, Silver RM, Serrano M, Buck GA, McNeil R, Nandakumar R, Reddy U, Wapner RJ, Kav AB, Uhlemann AC, Korem T. Early prediction of preeclampsia using the first trimester vaginal microbiome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.01.626267. [PMID: 39677801 PMCID: PMC11642775 DOI: 10.1101/2024.12.01.626267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Preeclampsia is a severe obstetrical syndrome which contributes to 10-15% of all maternal deaths. Although the mechanisms underlying systemic damage in preeclampsia-such as impaired placentation, endothelial dysfunction, and immune dysregulation-are well studied, the initial triggers of the condition remain largely unknown. Furthermore, although the pathogenesis of preeclampsia begins early in pregnancy, there are no early diagnostics for this life-threatening syndrome, which is typically diagnosed much later, after systemic damage has already manifested. Here, we performed deep metagenomic sequencing and multiplex immunoassays of vaginal samples collected during the first trimester from 124 pregnant individuals, including 62 who developed preeclampsia with severe features. We identified multiple significant associations between vaginal immune factors, microbes, clinical factors, and the early pathogenesis of preeclampsia. These associations vary with BMI, and stratification revealed strong associations between preeclampsia and Bifidobacterium spp., Prevotella timonensis, and Sneathia vaginalis. Finally, we developed machine learning models that predict the development of preeclampsia using this first trimester data, collected ~5.7 months prior to clinical diagnosis, with an auROC of 0.78. We validated our models using data from an independent cohort (MOMS-PI), achieving an auROC of 0.80. Our findings highlight robust associations among the vaginal microbiome, local host immunity, and early pathogenic processes of preeclampsia, paving the way for early detection, prevention and intervention for this devastating condition.
Collapse
Affiliation(s)
- William F. Kindschuh
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - George I. Austin
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY, USA
| | - Yoli Meydan
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Heekuk Park
- Division of Infectious Diseases, Columbia University Irving Medical Center, New York, NY, USA
| | - Julia A. Urban
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Emily Watters
- Division of Infectious Diseases, Columbia University Irving Medical Center, New York, NY, USA
| | - Susan Pollak
- Biomarkers Core, Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York, NY, USA
| | - George R. Saade
- Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Judith Chung
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of California Irvine, CA, USA
| | - Brian M. Mercer
- Departments of Obstetrics and Gynecology, Case Western Reserve University, Cleveland, OH, USA
| | | | - David M. Haas
- Department of Obstetrics and Gynecology, Indiana University, Indianapolis, IN, USA
| | - Robert M. Silver
- Department of Obstetrics and Gynecology, University of Utah, Salt Lake City, UT, USA
| | - Myrna Serrano
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
- Center for Microbiome Engineering and Data Analysis, Virginia Commonwealth University, Richmond, VA, USA
| | - Gregory A. Buck
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
- Center for Microbiome Engineering and Data Analysis, Virginia Commonwealth University, Richmond, VA, USA
- Department of Computer Science, School of Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | | | - Renu Nandakumar
- Biomarkers Core, Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York, NY, USA
| | - Uma Reddy
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Ronald J. Wapner
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Aya Brown Kav
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Anne-Catrin Uhlemann
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Division of Infectious Diseases, Columbia University Irving Medical Center, New York, NY, USA
| | - Tal Korem
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
24
|
Wan Y, Jiang G, Shan H, Lin Y, Xia W, Yin F, Jiang C, Shi Z. F-Box and WD repeat domain containing 7 induces infectious osteomyelitis by regulating MYB stability and ubiquitination. Scand J Immunol 2024; 100:e13414. [PMID: 39487565 DOI: 10.1111/sji.13414] [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: 02/15/2024] [Revised: 08/29/2024] [Accepted: 10/13/2024] [Indexed: 11/04/2024]
Abstract
Osteomyelitis is a bone inflammation initiated by invading pathogens. Macrophages and inflammation play essential roles in osteomyelitis. F-Box and WD repeat domain containing 7 (Fbxw7) is a tumour suppressor and E3 ubiquitin ligase. In the present study, the potential roles of Fbxw7 in osteomyelitis were explored. The mRNA level of Fbxw7 was measured in bone marrow cells from patients with osteomyelitis and Staphylococcus aureus (S. aureus)-infected macrophages. The conditional knockout mice with Fbxw7 deficiency in myeloid cells were generated. The expression of interleukin (IL)-6, IL-23a and nitric oxide synthase 2 (Nos2) was measured in S. aureus-infected Fbxw7-deficient bone marrow-derived macrophages (BMDMs). The body weight loss, bacterial burden, bone loss and formation and serum level of IL-6, IL-23 and TNF-α were measured in S. aureus-infected Fbxw7 conditional KO mice. The interacting partners of Fbxw7 were predicted using STRING and the interaction were tested. Elevated expression of Fbxw7 was observed in bone marrow cells from patients with osteomyelitis and in S. aureus-infected macrophages. The expression of IL-6, IL-23a and Nos2 was remarkably suppressed in S. aureus-infected Fbxw7-deficient BMDMs. Fbxw7 conditional knockout mice had less body weight loss, higher bacterial burden, less bone loss and formation and decreased serum level of cytokines. Fbxw7 interacted with MYB. S. aureus-infected Fbxw7-deficient BMDMs had higher level of MYB and less ubiquitination of MYB. Fbxw7 promotes osteomyelitis symptoms by regulating ubiquitination and stability of MYB.
Collapse
Affiliation(s)
- Yongbo Wan
- Department of Orthopaedic Surgery, Haikou Orthopedic and Diabetes Hospital of Shanghai Sixth People's Hospital, Haikou, Hainan, China
| | - Gehan Jiang
- Department of Orthopaedic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Haojie Shan
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiwei Lin
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenyang Xia
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fuli Yin
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chaolai Jiang
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhongmin Shi
- Department of Orthopaedic Surgery, Haikou Orthopedic and Diabetes Hospital of Shanghai Sixth People's Hospital, Haikou, Hainan, China
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
25
|
Gabitto MI, Travaglini KJ, Rachleff VM, Kaplan ES, Long B, Ariza J, Ding Y, Mahoney JT, Dee N, Goldy J, Melief EJ, Agrawal A, Kana O, Zhen X, Barlow ST, Brouner K, Campos J, Campos J, Carr AJ, Casper T, Chakrabarty R, Clark M, Cool J, Dalley R, Darvas M, Ding SL, Dolbeare T, Egdorf T, Esposito L, Ferrer R, Fleckenstein LE, Gala R, Gary A, Gelfand E, Gloe J, Guilford N, Guzman J, Hirschstein D, Ho W, Hupp M, Jarsky T, Johansen N, Kalmbach BE, Keene LM, Khawand S, Kilgore MD, Kirkland A, Kunst M, Lee BR, Leytze M, Mac Donald CL, Malone J, Maltzer Z, Martin N, McCue R, McMillen D, Mena G, Meyerdierks E, Meyers KP, Mollenkopf T, Montine M, Nolan AL, Nyhus JK, Olsen PA, Pacleb M, Pagan CM, Peña N, Pham T, Pom CA, Postupna N, Rimorin C, Ruiz A, Saldi GA, Schantz AM, Shapovalova NV, Sorensen SA, Staats B, Sullivan M, Sunkin SM, Thompson C, Tieu M, Ting JT, Torkelson A, Tran T, Valera Cuevas NJ, Walling-Bell S, Wang MQ, Waters J, Wilson AM, Xiao M, Haynor D, Gatto NM, Jayadev S, Mufti S, Ng L, Mukherjee S, Crane PK, Latimer CS, Levi BP, Smith KA, et alGabitto MI, Travaglini KJ, Rachleff VM, Kaplan ES, Long B, Ariza J, Ding Y, Mahoney JT, Dee N, Goldy J, Melief EJ, Agrawal A, Kana O, Zhen X, Barlow ST, Brouner K, Campos J, Campos J, Carr AJ, Casper T, Chakrabarty R, Clark M, Cool J, Dalley R, Darvas M, Ding SL, Dolbeare T, Egdorf T, Esposito L, Ferrer R, Fleckenstein LE, Gala R, Gary A, Gelfand E, Gloe J, Guilford N, Guzman J, Hirschstein D, Ho W, Hupp M, Jarsky T, Johansen N, Kalmbach BE, Keene LM, Khawand S, Kilgore MD, Kirkland A, Kunst M, Lee BR, Leytze M, Mac Donald CL, Malone J, Maltzer Z, Martin N, McCue R, McMillen D, Mena G, Meyerdierks E, Meyers KP, Mollenkopf T, Montine M, Nolan AL, Nyhus JK, Olsen PA, Pacleb M, Pagan CM, Peña N, Pham T, Pom CA, Postupna N, Rimorin C, Ruiz A, Saldi GA, Schantz AM, Shapovalova NV, Sorensen SA, Staats B, Sullivan M, Sunkin SM, Thompson C, Tieu M, Ting JT, Torkelson A, Tran T, Valera Cuevas NJ, Walling-Bell S, Wang MQ, Waters J, Wilson AM, Xiao M, Haynor D, Gatto NM, Jayadev S, Mufti S, Ng L, Mukherjee S, Crane PK, Latimer CS, Levi BP, Smith KA, Close JL, Miller JA, Hodge RD, Larson EB, Grabowski TJ, Hawrylycz M, Keene CD, Lein ES. Integrated multimodal cell atlas of Alzheimer's disease. Nat Neurosci 2024; 27:2366-2383. [PMID: 39402379 PMCID: PMC11614693 DOI: 10.1038/s41593-024-01774-5] [Show More Authors] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 08/28/2024] [Indexed: 10/19/2024]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in older adults. Although AD progression is characterized by stereotyped accumulation of proteinopathies, the affected cellular populations remain understudied. Here we use multiomics, spatial genomics and reference atlases from the BRAIN Initiative to study middle temporal gyrus cell types in 84 donors with varying AD pathologies. This cohort includes 33 male donors and 51 female donors, with an average age at time of death of 88 years. We used quantitative neuropathology to place donors along a disease pseudoprogression score. Pseudoprogression analysis revealed two disease phases: an early phase with a slow increase in pathology, presence of inflammatory microglia, reactive astrocytes, loss of somatostatin+ inhibitory neurons, and a remyelination response by oligodendrocyte precursor cells; and a later phase with exponential increase in pathology, loss of excitatory neurons and Pvalb+ and Vip+ inhibitory neuron subtypes. These findings were replicated in other major AD studies.
Collapse
Affiliation(s)
- Mariano I Gabitto
- Allen Institute for Brain Science, Seattle, WA, USA
- Department of Statistics, University of Washington, Seattle, WA, USA
| | | | - Victoria M Rachleff
- Allen Institute for Brain Science, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | | | - Brian Long
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Jeanelle Ariza
- Allen Institute for Brain Science, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Yi Ding
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Nick Dee
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Erica J Melief
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Anamika Agrawal
- Center for Data-Driven Discovery for Biology, Allen Institute, Seattle, WA, USA
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Omar Kana
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | - John Campos
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | | | | | | | | | - Jonah Cool
- Chan Zuckerberg Initiative, Redwood City, CA, USA
| | | | - Martin Darvas
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | | | - Tim Dolbeare
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Tom Egdorf
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - Rohan Gala
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Amanda Gary
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Jessica Gloe
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - Windy Ho
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Madison Hupp
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Tim Jarsky
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Brian E Kalmbach
- Allen Institute for Brain Science, Seattle, WA, USA
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Lisa M Keene
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Sarah Khawand
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Mitchell D Kilgore
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Amanda Kirkland
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | | | - Brian R Lee
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - Zoe Maltzer
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Naomi Martin
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Rachel McCue
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Gonzalo Mena
- Department of Statistics and Data Science, Carnegie Mellon University, Pittsburgh, PA, USA
| | | | - Kelly P Meyers
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | | | - Mark Montine
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Amber L Nolan
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | | | - Paul A Olsen
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Maiya Pacleb
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | | | | | | | | | - Nadia Postupna
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | | | | | | | - Aimee M Schantz
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | | | | | - Brian Staats
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - Michael Tieu
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Tracy Tran
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - Jack Waters
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Angela M Wilson
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Ming Xiao
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - David Haynor
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Nicole M Gatto
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Suman Jayadev
- Department of Neurology, University of Washington, Seattle, WA, USA
| | - Shoaib Mufti
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Lydia Ng
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Paul K Crane
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Caitlin S Latimer
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Boaz P Levi
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | - Eric B Larson
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Thomas J Grabowski
- Department of Radiology, University of Washington, Seattle, WA, USA
- Department of Neurology, University of Washington, Seattle, WA, USA
| | | | - C Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.
| | - Ed S Lein
- Allen Institute for Brain Science, Seattle, WA, USA.
| |
Collapse
|
26
|
Wang P, Okada-Rising S, Scultetus AH, Bailey ZS. The Relevance and Implications of Monoclonal Antibody Therapies on Traumatic Brain Injury Pathologies. Biomedicines 2024; 12:2698. [PMID: 39767605 PMCID: PMC11672875 DOI: 10.3390/biomedicines12122698] [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: 09/25/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
Traumatic brain injury (TBI) is a global public health concern. It remains one of the leading causes of morbidity and mortality. TBI pathology involves complex secondary injury cascades that are associated with cellular and molecular dysfunction, including oxidative stress, coagulopathy, neuroinflammation, neurodegeneration, neurotoxicity, and blood-brain barrier (BBB) dysfunction, among others. These pathological processes manifest as a diverse array of clinical impairments. They serve as targets for potential therapeutic intervention not only in TBI but also in other diseases. Monoclonal antibodies (mAbs) have been used as key therapeutic agents targeting these mechanisms for the treatment of diverse diseases, including neurological diseases such as Alzheimer's disease (AD). MAb therapies provide a tool to block disease pathways with target specificity that may be capable of mitigating the secondary injury cascades following TBI. This article reviews the pathophysiology of TBI and the molecular mechanisms of action of mAbs that target these shared pathological pathways in a wide range of diseases. Publicly available databases for various applications of mAb therapy were searched and further classified to assess relevance to TBI pathology and evaluate current stages of development. The authors intend for this review to highlight the potential impact of current mAb technology within pathological TBI processes.
Collapse
Affiliation(s)
- Ping Wang
- Brain Trauma Neuroprotection, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (S.O.-R.); (A.H.S.); (Z.S.B.)
| | | | | | | |
Collapse
|
27
|
Kang B, Song B, Shin H, Lee IS. Downregulation of nuclear receptor-binding SET domain protein 1 induces proinflammatory cytokine expression via mitogen-activated protein kinase pathways in U87MG cells. Biochem Biophys Res Commun 2024; 734:150638. [PMID: 39236589 DOI: 10.1016/j.bbrc.2024.150638] [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: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 09/07/2024]
Abstract
Haploinsufficiency of the nuclear receptor binding SET domain-containing protein 1 gene (NSD1) leads to a neurodevelopmental disorder known as Sotos syndrome (SOTOS). This study investigated the effects of NSD1 knockdown in glial cells. U87MG glioma cells were transfected with siRNA targeting NSD1, which resulted in morphological changes characteristic of activated astrocytes. These activated phenotypes were accompanied by specific activation of mitogen-activated protein kinase (MAPK) signaling pathways, particularly those mediated by p38 MAPK and c-Jun N-terminal kinase (JNK). Transcriptome analysis showed increased expression of proinflammatory cytokine genes, particularly interleukin (IL)-1α, IL-1β, and IL-6, following NSD1 knockdown. Treatment with MAPK inhibitors significantly reduced the cytokine induction caused by NSD1 knockdown, with the p38 MAPK inhibitor being more effective than the JNK inhibitor. These findings provide new insights into the role of NSD1 loss in neurological dysfunctions associated with SOTOS.
Collapse
Affiliation(s)
- Byungjun Kang
- Department of Biological Sciences, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Bokyeong Song
- Department of Biological Sciences, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Hyewon Shin
- Department of Biological Sciences, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Im-Soon Lee
- Department of Biological Sciences, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea.
| |
Collapse
|
28
|
Wang H, Song X, Wang Y, Yang T, Liu W, Mou Y, Ren C, Song X. Interleukin 1β Mediates the Pathogenesis of Nasal Mucosal Epithelial Barrier Dysfunction in Allergic Rhinitis. J Inflamm Res 2024; 17:9071-9085. [PMID: 39588138 PMCID: PMC11586497 DOI: 10.2147/jir.s488340] [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: 07/23/2024] [Accepted: 11/14/2024] [Indexed: 11/27/2024] Open
Abstract
Background The nasal mucosal epithelial barrier is the primary site of allergic rhinitis (AR). Interleukin-1β (IL-1β), as a crucial factor in immune inflammation, not only plays a crucial role in hypersensitivity reactions but also affects the digestive mucosa and skin epithelial barrier. However, the role of IL-1β in the nasal mucosal epithelial barrier in AR has not been reported, and this study aimed to investigate the effect and possible mechanisms involved. Methods Dermatophagoides pteronyssinus 1 was used as an allergen to construct an AR mouse model and stimulate human nasal mucosal epithelial cells (HNEpCs) and observe the expression changes of IL-1β and epithelial barrier indicators CLDN1 and OCLN in mouse nasal mucosa and HNEpCs. Then, the possible mechanisms of action were explored via exogenous IL-1β stimulation and pharmacological inhibition of IL-1β or its receptor interleukin-1 receptor type 1 (IL-1R1). Results The results showed that Dermatophagoides pteronyssinus 1-primed mouse nasal mucosa or human HENpCs had increased expression of IL-1β and decreased CLDN1 and OCLN, and IL-1β could directly lead to reduced expression of epithelial barrier indexes in HNEpCs. In addition, inhibition of IL-1β or IL-1R1 can effectively alleviate the damage to the epithelial barrier. Conclusion IL-1β has a destructive effect on the nasal mucosal epithelial barrier in AR, and inhibition of IL-1β or its receptor IL-1R1 can effectively protect the nasal mucosal barrier. IL-1β is a potential target for the treatment of AR.
Collapse
Affiliation(s)
- Hanrui Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, People’s Republic of China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, People’s Republic of China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, People’s Republic of China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, People’s Republic of China
| | - Xiaoyu Song
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, People’s Republic of China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, People’s Republic of China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, People’s Republic of China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, People’s Republic of China
| | - Yao Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, People’s Republic of China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, People’s Republic of China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, People’s Republic of China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, People’s Republic of China
| | - Ting Yang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, People’s Republic of China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, People’s Republic of China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, People’s Republic of China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, People’s Republic of China
| | - Wanchen Liu
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, People’s Republic of China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, People’s Republic of China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, People’s Republic of China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, People’s Republic of China
| | - Yakui Mou
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, People’s Republic of China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, People’s Republic of China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, People’s Republic of China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, People’s Republic of China
| | - Chao Ren
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, People’s Republic of China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, People’s Republic of China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, People’s Republic of China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, People’s Republic of China
- Department of Neurology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, People’s Republic of China
| | - Xicheng Song
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, People’s Republic of China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, People’s Republic of China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, People’s Republic of China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, People’s Republic of China
| |
Collapse
|
29
|
Pei J, Wang Y, Shi S, Chen L, Yin J, Nan F. Synthesis and Biological Activity of Tetracyclic Dispiro Core Derivatives of Natural Products Dispirocochlearoids A-C. Org Lett 2024; 26:9654-9658. [PMID: 39485012 DOI: 10.1021/acs.orglett.4c03358] [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: 11/03/2024]
Abstract
Natural products dispirocochlearoids A-C, which are meroterpenoids derived from Ganoderma fungi, feature a 6/6/5/6/6/6 ring system and exhibit selective COX-2 inhibitory activity. Herein, the concise total synthesis of the tetracyclic core structure of dispirocochlearoids A-C was achieved through an aldol reaction/cyclization/deprotection/cyclization cascade sequence. A series of simplified tetracyclic analogues was successfully constructed and their anti-inflammatory activity was further explored, with several tetracyclic analogues (such as compound 8ab) exhibiting strong inhibitory activity against IL-1β expression in lipopolysaccharide-stimulated bone marrow-derived macrophage cells (IC50 = 2.8 μM).
Collapse
Affiliation(s)
- Junping Pei
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Ying Wang
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
| | - Shengjie Shi
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
| | - Linhai Chen
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jianpeng Yin
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
| | - Fajun Nan
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
30
|
Lian J, Tang X, Gui Y, Lu S, Song Y, Deng Y. Impact of formulation parameters and circulation time on PEGylated liposomal doxorubicin related hand-foot syndrome. Int J Pharm 2024; 665:124659. [PMID: 39260752 DOI: 10.1016/j.ijpharm.2024.124659] [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: 05/23/2024] [Revised: 08/24/2024] [Accepted: 09/01/2024] [Indexed: 09/13/2024]
Abstract
PEGylated liposomal doxorubicin (PLD) has effectively reduced the cardiac toxicity of free doxorubicin (DOX) due to its unique nanoscale properties. However, an unexpected accumulation of PLD in the skin has led to hand-foot syndrome (HFS), negatively impacting quality of life and psychological well-being. In this study, self-limiting HFS rat models were created to mimic human symptoms through varying dosing schedules and intensities of PLD. The effects of PLD formulation parameters on HFS were also investigated. The results demonstrated that replacing ammonium sulfate with citric buffer, increasing liposome size, or reducing DSPE-mPEG2000 modification density alleviated HFS. Additionally, liposomes without DSPE-mPEG2000 modification completely avoided HFS, suggesting that PEGylated phospholipid was the key formulation parameter contributing to PLD-induced HFS. Furthermore, the correlation between liposome pharmacokinetics and HFS indicated that PEGylation, rather than the extended circulation time of liposomes, may mediated PLD-related HFS. Better understanding of the formulation parameters that trigger HFS can guide reformulation strategies to mitigate or prevent this syndrome.
Collapse
Affiliation(s)
- Jiawei Lian
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, PR China
| | - Xueying Tang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, PR China
| | - Yangxu Gui
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, PR China
| | - Shuang Lu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, PR China
| | - Yanzhi Song
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, PR China.
| | - Yihui Deng
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, PR China.
| |
Collapse
|
31
|
Wu Y, Sun X, Kang K, Yang Y, Li H, Zhao A, Niu T. Hemophagocytic lymphohistiocytosis: current treatment advances, emerging targeted therapy and underlying mechanisms. J Hematol Oncol 2024; 17:106. [PMID: 39511607 PMCID: PMC11542428 DOI: 10.1186/s13045-024-01621-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 10/14/2024] [Indexed: 11/15/2024] Open
Abstract
Hemophagocytic lymphohistiocytosis (HLH) is a rapidly progressing, life-threatening syndrome characterized by excessive immune activation, often presenting as a complex cytokine storm. This hyperactive immune response can lead to multi-organ failure and systemic damage, resulting in an extremely short survival period if left untreated. Over the past decades, although HLH has garnered increasing attention from researchers, there have been few advancements in its treatment. The cytokine storm plays a crucial role in the treatment of HLH. Investigating the detailed mechanisms behind cytokine storms offers insights into targeted therapeutic approaches, potentially aiding in early intervention and improving the clinical outcome of HLH patients. To date, there is only one targeted therapy, emapalumab targeting interferon-γ, that has gained approval for primary HLH. This review aims to summarize the current treatment advances, emerging targeted therapeutics and underlying mechanisms of HLH, highlighting its newly discovered targets potentially involved in cytokine storms, which are expected to drive the development of novel treatments and offer fresh perspectives for future studies. Besides, multi-targeted combination therapy may be essential for disease control, but further trials are required to determine the optimal treatment mode for HLH.
Collapse
Affiliation(s)
- Yijun Wu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- National Facility for Translational Medicine (Sichuan), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xu Sun
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- National Facility for Translational Medicine (Sichuan), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kai Kang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- National Facility for Translational Medicine (Sichuan), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuqi Yang
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - He Li
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- National Facility for Translational Medicine (Sichuan), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ailin Zhao
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- National Facility for Translational Medicine (Sichuan), West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Ting Niu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- National Facility for Translational Medicine (Sichuan), West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
32
|
Pirzada RH, Yasmeen F, Haseeb M, Javaid N, Kim E, Choi S. Small molecule inhibitors of IL-1R1/IL-1β interaction identified via transfer machine learning QSAR modelling. Int J Biol Macromol 2024; 282:137295. [PMID: 39515709 DOI: 10.1016/j.ijbiomac.2024.137295] [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: 09/22/2024] [Revised: 11/03/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
The human interleukin-1 receptor I (IL-1R1) is a cytokine receptor recognized by interleukin 1β (IL-1β), among other cytokines. Over activation of IL-1R1 has been implicated in various inflammatory conditions. This research aims to identify small-molecule inhibitors targeting the hIL1R1/IL1β interaction, employing a multi-task transfer learning approach for quantitative structure-activity relationship (QSAR) modelling. A comprehensive bioactivity dataset from functionally related proteins was utilised to build a robust ensemble machine learning model for predicting IC50 values against the target protein. Despite the availability of antibody-based therapies, the absence of orally available small-molecule inhibitors necessitates their development. By combining model predictions with docking and simulation approaches, the interleukin-1 receptor inhibitor (IRI-1) emerged as a lead compound. It potently inhibited human IL1-R1 with micromolar activity in THP-1 and Saos-2 cells and demonstrated good biocompatibility. Western blot analysis revealed that IRI-1 inhibits IL-1β-mediated phosphorylation of IL1-R1, JNK, IRAK-4, and ERK in THP-1 cells. Furthermore, molecular dynamics simulations confirmed the structural stability of the protein-ligand complexes. This study highlights the effectiveness of multi-task transfer learning approaches for building robust QSAR models against novel proteins or those with limited bioactivity data, such as hIL-1β/IL-1R1 protein.
Collapse
Affiliation(s)
- Rameez Hassan Pirzada
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea.
| | - Farzana Yasmeen
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Muhammad Haseeb
- S&K Therapeutics, Ajou University Campus Plaza 418, 199 Worldcup-ro, Yeongtong-gu, Suwon 16502, Republic of Korea
| | - Nasir Javaid
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Eunha Kim
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea; S&K Therapeutics, Ajou University Campus Plaza 418, 199 Worldcup-ro, Yeongtong-gu, Suwon 16502, Republic of Korea.
| |
Collapse
|
33
|
Taru V, Szabo G, Mehal W, Reiberger T. Inflammasomes in chronic liver disease: Hepatic injury, fibrosis progression and systemic inflammation. J Hepatol 2024; 81:895-910. [PMID: 38908436 PMCID: PMC11881887 DOI: 10.1016/j.jhep.2024.06.016] [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: 02/18/2024] [Revised: 05/23/2024] [Accepted: 06/17/2024] [Indexed: 06/24/2024]
Abstract
Chronic liver disease leads to hepatocellular injury that triggers a pro-inflammatory state in several parenchymal and non-parenchymal hepatic cell types, ultimately resulting in liver fibrosis, cirrhosis, portal hypertension and liver failure. Thus, an improved understanding of inflammasomes - as key molecular drivers of liver injury - may result in the development of novel diagnostic or prognostic biomarkers and effective therapeutics. In liver disease, innate immune cells respond to hepatic insults by activating cell-intrinsic inflammasomes via toll-like receptors and NF-κB, and by releasing pro-inflammatory cytokines (such as IL-1β, IL-18, TNF-α and IL-6). Subsequently, cells of the adaptive immune system are recruited to fuel hepatic inflammation and hepatic parenchymal cells may undergo gasdermin D-mediated programmed cell death, termed pyroptosis. With liver disease progression, there is a shift towards a type 2 inflammatory response, which promotes tissue repair but also fibrogenesis. Inflammasome activation may also occur at extrahepatic sites, such as the white adipose tissue in MASH (metabolic dysfunction-associated steatohepatitis). In end-stage liver disease, flares of inflammation (e.g., in severe alcohol-related hepatitis) that spark on a dysfunctional immune system, contribute to inflammasome-mediated liver injury and potentially result in organ dysfunction/failure, as seen in ACLF (acute-on-chronic liver failure). This review provides an overview of current concepts regarding inflammasome activation in liver disease progression, with a focus on related biomarkers and therapeutic approaches that are being developed for patients with liver disease.
Collapse
Affiliation(s)
- Vlad Taru
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria; Christian-Doppler Laboratory for Portal Hypertension and Liver Fibrosis, Medical University of Vienna, Vienna, Austria; Iuliu Hatieganu University of Medicine and Pharmacy, 4(th) Dept. of Internal Medicine, Cluj-Napoca, Romania
| | - Gyongyi Szabo
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Wajahat Mehal
- Section of Digestive Diseases, Yale School of Medicine, New Haven, CT, USA; West Haven Veterans Medical Center, West Haven, CT, USA.
| | - Thomas Reiberger
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria; Christian-Doppler Laboratory for Portal Hypertension and Liver Fibrosis, Medical University of Vienna, Vienna, Austria; Center for Molecular Medicine (CeMM) of the Austrian Academy of Science, Vienna, Austria
| |
Collapse
|
34
|
Edin C, Ekstedt M, Karlsson M, Wegmann B, Warntjes M, Swahn E, Östgren CJ, Ebbers T, Lundberg P, Carlhäll CJ. Liver fibrosis is associated with left ventricular remodeling: insight into the liver-heart axis. Eur Radiol 2024; 34:7492-7502. [PMID: 38795131 PMCID: PMC11519090 DOI: 10.1007/s00330-024-10798-1] [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: 11/01/2023] [Revised: 03/21/2024] [Accepted: 04/04/2024] [Indexed: 05/27/2024]
Abstract
OBJECTIVE In nonalcoholic fatty liver disease (NAFLD), liver fibrosis is the strongest predictor of adverse outcomes. We sought to investigate the relationship between liver fibrosis and cardiac remodeling in participants from the general population using magnetic resonance imaging (MRI), as well as explore potential mechanistic pathways by analyzing circulating cardiovascular biomarkers. METHODS In this cross-sectional study, we prospectively included participants with type 2 diabetes and individually matched controls from the SCAPIS (Swedish CArdioPulmonary bioImage Study) cohort in Linköping, Sweden. Between November 2017 and July 2018, participants underwent MRI at 1.5 Tesla for quantification of liver proton density fat fraction (spectroscopy), liver fibrosis (stiffness from elastography), left ventricular (LV) structure and function, as well as myocardial native T1 mapping. We analyzed 278 circulating cardiovascular biomarkers using a Bayesian statistical approach. RESULTS In total, 92 participants were enrolled (mean age 59.5 ± 4.6 years, 32 women). The mean liver stiffness was 2.1 ± 0.4 kPa. 53 participants displayed hepatic steatosis. LV concentricity increased across quartiles of liver stiffness. Neither liver fat nor liver stiffness displayed any relationships to myocardial tissue characteristics (native T1). In a regression analysis, liver stiffness was related to increased LV concentricity. This association was independent of diabetes and liver fat (Beta = 0.26, p = 0.0053), but was attenuated (Beta = 0.17, p = 0.077) when also adjusting for circulating levels of interleukin-1 receptor type 2. CONCLUSION MRI reveals that liver fibrosis is associated to structural LV remodeling, in terms of increased concentricity, in participants from the general population. This relationship could involve the interleukin-1 signaling. CLINICAL RELEVANCE STATEMENT Liver fibrosis may be considered a cardiovascular risk factor in patients without cirrhosis. Further research on the mechanisms that link liver fibrosis to left ventricular concentricity may reveal potential therapeutic targets in patients with non-alcoholic fatty liver disease (NAFLD). KEY POINTS Previously, studies on liver fibrosis and cardiac remodeling have focused on advanced stages of liver fibrosis. Liver fibrosis is associated with left ventricular (LV) concentricity and may relate to interleukin-1 receptor type 2. Interleukin-1 signaling is a potential mechanistic interlink between early liver fibrosis and LV remodeling.
Collapse
Affiliation(s)
- Carl Edin
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
- Department of Clinical Physiology in Linköping, and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Mattias Ekstedt
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Markus Karlsson
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
- Department of Radiation Physics, and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Bertil Wegmann
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
- Department of Computer and Information Science, Linköping University, Linköping, Sweden
| | - Marcel Warntjes
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Eva Swahn
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
- Department of Cardiology in Linköping, and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Carl Johan Östgren
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
- Division of Prevention, Rehabilitation and Community Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Tino Ebbers
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Peter Lundberg
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
- Department of Radiation Physics, and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Carl-Johan Carlhäll
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden.
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden.
- Department of Clinical Physiology in Linköping, and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden.
| |
Collapse
|
35
|
González-Cubero E, González-Fernández ML, Esteban-Blanco M, Pérez-Castrillo S, Pérez-Fernández E, Navasa N, Aransay AM, Anguita J, Villar-Suárez V. The Therapeutic Potential of Adipose-Derived Mesenchymal Stem Cell Secretome in Osteoarthritis: A Comprehensive Study. Int J Mol Sci 2024; 25:11287. [PMID: 39457070 PMCID: PMC11508730 DOI: 10.3390/ijms252011287] [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: 09/17/2024] [Revised: 10/06/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease characterized by cartilage degradation and inflammation. This study investigates the therapeutic potential of secretome derived from adipose tissue mesenchymal stem cells (ASCs) in mitigating inflammation and promoting cartilage repair in an in vitro model of OA. Our in vitro model comprised chondrocytes inflamed with TNF. To assess the therapeutic potential of secretome, inflamed chondrocytes were treated with it and concentrations of pro-inflammatory cytokines, metalloproteinases (MMPs) and extracellular matrix markers were measured. In addition, secretome-treated chondrocytes were subject to a microarray analysis to determine which genes were upregulated and which were downregulated. Treating TNF-inflamed chondrocytes with secretome in vitro inhibits the NF-κB pathway, thereby mediating anti-inflammatory and anti-catabolic effects. Additional protective effects of secretome on cartilage are revealed in the inhibition of hypertrophy markers such as RUNX2 and COL10A1, increased production of COL2A1 and ACAN and upregulation of SOX9. These findings suggest that ASC-derived secretome can effectively reduce inflammation, promote cartilage repair, and maintain chondrocyte phenotype. This study highlights the potential of ASC-derived secretome as a novel, non-cell-based therapeutic approach for OA, offering a promising alternative to current treatments by targeting inflammation and cartilage repair mechanisms.
Collapse
Affiliation(s)
- Elsa González-Cubero
- Department of Neurosurgery, Stanford School of Medicine, Stanford University, Palo Alto, CA 94304, USA;
- Department of Anatomy, Faculty of Veterinary Sciences, Campus de Vegazana, University of Léon-Universidad de León, 24071 León, Spain; (M.L.G.-F.); (M.E.-B.); (S.P.-C.); (E.P.-F.)
| | - Maria Luisa González-Fernández
- Department of Anatomy, Faculty of Veterinary Sciences, Campus de Vegazana, University of Léon-Universidad de León, 24071 León, Spain; (M.L.G.-F.); (M.E.-B.); (S.P.-C.); (E.P.-F.)
| | - Marta Esteban-Blanco
- Department of Anatomy, Faculty of Veterinary Sciences, Campus de Vegazana, University of Léon-Universidad de León, 24071 León, Spain; (M.L.G.-F.); (M.E.-B.); (S.P.-C.); (E.P.-F.)
| | - Saúl Pérez-Castrillo
- Department of Anatomy, Faculty of Veterinary Sciences, Campus de Vegazana, University of Léon-Universidad de León, 24071 León, Spain; (M.L.G.-F.); (M.E.-B.); (S.P.-C.); (E.P.-F.)
| | - Esther Pérez-Fernández
- Department of Anatomy, Faculty of Veterinary Sciences, Campus de Vegazana, University of Léon-Universidad de León, 24071 León, Spain; (M.L.G.-F.); (M.E.-B.); (S.P.-C.); (E.P.-F.)
| | - Nicolás Navasa
- Department of Molecular Biology, Faculty of Veterinary Sciences, Campus de Vegazana, University of Léon-Universidad de León, 24071 León, Spain;
- Center for Cooperative Research in Biosciences (CIC bioGUNE)-Basque Research and Technology Alliance (BRTA), Parque Tecnológico de Bizkaia, Building 801-A, 48160 Derio, Spain; (A.M.A.); (J.A.)
| | - Ana M. Aransay
- Center for Cooperative Research in Biosciences (CIC bioGUNE)-Basque Research and Technology Alliance (BRTA), Parque Tecnológico de Bizkaia, Building 801-A, 48160 Derio, Spain; (A.M.A.); (J.A.)
- CIBERehd, ISCIII, 28029 Madrid, Spain
| | - Juan Anguita
- Center for Cooperative Research in Biosciences (CIC bioGUNE)-Basque Research and Technology Alliance (BRTA), Parque Tecnológico de Bizkaia, Building 801-A, 48160 Derio, Spain; (A.M.A.); (J.A.)
- IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain
| | - Vega Villar-Suárez
- Department of Anatomy, Faculty of Veterinary Sciences, Campus de Vegazana, University of Léon-Universidad de León, 24071 León, Spain; (M.L.G.-F.); (M.E.-B.); (S.P.-C.); (E.P.-F.)
- Institute of Biomedicine (IBIOMED), Faculty of Veterinary Sciences, Campus de Vegazana, University of León-Universidad de León, 24071 León, Spain
| |
Collapse
|
36
|
Fu W, Wang T, Lu Y, Shi T, Yang Q. The role of lactylation in plasma cells and its impact on rheumatoid arthritis pathogenesis: insights from single-cell RNA sequencing and machine learning. Front Immunol 2024; 15:1453587. [PMID: 39421742 PMCID: PMC11484267 DOI: 10.3389/fimmu.2024.1453587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024] Open
Abstract
Introduction Rheumatoid arthritis (RA) is a chronic autoimmune disorder characterized by persistent synovitis, systemic inflammation, and autoantibody production. This study aims to explore the role of lactylation in plasma cells and its impact on RA pathogenesis. Methods We utilized single-cell RNA sequencing (scRNA-seq) data and applied bioinformatics and machine learning techniques. A total of 10,163 cells were retained for analysis after quality control. Clustering analysis identified 13 cell clusters, with plasma cells displaying the highest lactylation scores. We performed pathway enrichment analysis to examine metabolic activity, such as oxidative phosphorylation and glycolysis, in highly lactylated plasma cells. Additionally, we employed 134 machine learning algorithms to identify seven core lactylation-promoting genes and constructed a diagnostic model with an average AUC of 0.918. Results The RA lactylation score (RAlac_score) was significantly elevated in RA patients and positively correlated with immune cell infiltration and immune checkpoint molecule expression. Differential expression analysis between two plasma cell clusters revealed distinct metabolic and immunological profiles, with cluster 2 demonstrating increased immune activity and extracellular matrix interactions. qRT-PCR validation confirmed that NDUFB3, NGLY1, and SLC25A4 are highly expressed in RA. Conclusion This study highlights the critical role of lactylation in plasma cells for RA pathogenesis and identifies potential biomarkers and therapeutic targets, which may offer insights for future therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | - Tiejun Shi
- Department of Orthopedics, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua Municipal Central Hospital, Jinhua, Zhejiang, China
| | - Qining Yang
- Department of Orthopedics, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua Municipal Central Hospital, Jinhua, Zhejiang, China
| |
Collapse
|
37
|
Sun J, Feng S, Ding T, Wang T, Du L, Kang W, Ge S. Fusobacterium nucleatum dysregulates inflammatory cytokines and NLRP3 inflammasomes in oral cells. Oral Dis 2024; 30:4767-4781. [PMID: 38409736 DOI: 10.1111/odi.14899] [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: 10/17/2023] [Revised: 01/14/2024] [Accepted: 02/06/2024] [Indexed: 02/28/2024]
Abstract
OBJECTIVE This study aimed to clarify the difference in Fusobacterium nucleatum (F. nucleatum) induced inflammatory cytokines and nod-like receptor protein 3 (NLRP3) inflammasomes dysregulation among three periodontal cells. METHODS Oral epithelial cells (HIOECs), THP-1 macrophages, and human gingival fibroblasts (HGFs) were exposed to F. nucleatum with/without adenosine triphosphate (ATP) and nigericin (Nig). Cell morphology was assessed by scanning electron microscopy. qRT-PCR, protein microarrays, and bioinformatic methods were used to evaluate the cytokines and their complex interplay. NLRP3 inflammasomes activation was detected by western blotting and ELISA. RESULTS F. nucleatum adhered to and invaded cells. In HIOECs, F. nucleatum enhanced interleukin (IL)-1α/1β/6/10/13, TNF-α, and interferon (IFN)-γ expression. In THP-1 macrophages, F. nucleatum up-regulated IL-1α/1β/6/10 and TNF-α levels. In HGFs, F. nucleatum increased IL-6 levels. F. nucleatum and ATP synergistically boosted IFN-γ level in THP-1 macrophages and IL-13 level in HGFs. IL-1α/1β/6, and TNF-α served as epicenters of the inflammatory response. Additionally, F. nucleatum activated NLRP3 inflammasomes in HIOECs, and ATP/Nig boosted the activation. F. nucleatum also triggered NLRP3 inflammasomes in THP-1 macrophages, but in HGFs, only NLRP3 and caspase-1 levels were elevated. CONCLUSION F. nucleatum infiltrated periodontal supporting cells and dysregulated inflammatory cytokines and NLRP3 inflammasomes.
Collapse
Affiliation(s)
- Jingzhuo Sun
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Susu Feng
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Tian Ding
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Ting Wang
- Department of General Dentistry, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Lingqian Du
- Department of Stomatology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Wenyan Kang
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Shaohua Ge
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| |
Collapse
|
38
|
Maskey AR, Kopulos D, Kwan M, Reyes N, Figueroa C, Mo X, Yang N, Tiwari R, Geliebter J, Li XM. Berberine Inhibits the Inflammatory Response Induced by Staphylococcus aureus Isolated from Atopic Eczema Patients via the TNF-α/Inflammation/RAGE Pathways. Cells 2024; 13:1639. [PMID: 39404402 PMCID: PMC11475634 DOI: 10.3390/cells13191639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/19/2024] Open
Abstract
Atopic eczema patients exhibit high levels of Staphylococcus aureus (S. aureus) skin colonization. S. aureus can stimulate macrophages and the expression of proinflammatory cytokines. Berberine (BBR), an alkaloid, attenuates S. aureus toxin production. This study investigated if BBR suppressed bacterial growth and inflammatory response induced by eczema-patient-derived S. aureus using murine macrophage (RAW 264.7) and human monocyte cell lines (U937). RAW 264.7 and U937 were treated with BBR at different concentrations and stimulated with heat-killed S. aureus (ATCC #33591) or S. aureus derived from severe eczema patients (EC01-EC10), who were undergoing topical steroid withdrawal, for 24 h. TNF-α protein levels were determined by ELISA, gene expression by qRT-PCR, cell cytotoxicity by trypan blue excursion, and reactive oxygen species (ROS) levels by fluorometric assay. BBR showed a bacteriostatic effect in S. aureus (ATCC strain #33591 and clinical isolates (EC01-EC10) and suppressed TNF-α production in RAW 264.7 and U937 cells exposed to heat-killed S. aureus (ATCC and clinical isolates) dose-dependently without any cell cytotoxicity. BBR (20 µg/mL) suppressed >90% of TNF-α production (p < 0.001), downregulated genes involved in inflammatory pathways, and inhibited S. aureus ROS production in U937 and RAW 264.7 cells (p < 0.01). BBR suppresses S. aureus-induced inflammation via inhibition of TNF-α release, ROS production, and expression of key genes involved in the inflammatory pathway.
Collapse
Affiliation(s)
- Anish R. Maskey
- Department of Pathology, Microbiology & Immunology, New York Medical College, Valhalla, NY 10595, USA; (A.R.M.); (D.K.); (M.K.); (N.R.); (C.F.); (X.M.); (R.T.)
| | - Daniel Kopulos
- Department of Pathology, Microbiology & Immunology, New York Medical College, Valhalla, NY 10595, USA; (A.R.M.); (D.K.); (M.K.); (N.R.); (C.F.); (X.M.); (R.T.)
| | - Matthew Kwan
- Department of Pathology, Microbiology & Immunology, New York Medical College, Valhalla, NY 10595, USA; (A.R.M.); (D.K.); (M.K.); (N.R.); (C.F.); (X.M.); (R.T.)
| | - Niradiz Reyes
- Department of Pathology, Microbiology & Immunology, New York Medical College, Valhalla, NY 10595, USA; (A.R.M.); (D.K.); (M.K.); (N.R.); (C.F.); (X.M.); (R.T.)
- Genetics and Molecular Biology Research Group, School of Medicine, University of Cartagena, Cartagena 130001, Colombia
| | - Christian Figueroa
- Department of Pathology, Microbiology & Immunology, New York Medical College, Valhalla, NY 10595, USA; (A.R.M.); (D.K.); (M.K.); (N.R.); (C.F.); (X.M.); (R.T.)
- Department of Biology, University of Richmond, Richmond, VA 23173, USA
| | - Xian Mo
- Department of Pathology, Microbiology & Immunology, New York Medical College, Valhalla, NY 10595, USA; (A.R.M.); (D.K.); (M.K.); (N.R.); (C.F.); (X.M.); (R.T.)
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, Guangzhou 510120, China
| | - Nang Yang
- General Nutraceutical Technology, Elmsford, NY 10523, USA;
| | - Raj Tiwari
- Department of Pathology, Microbiology & Immunology, New York Medical College, Valhalla, NY 10595, USA; (A.R.M.); (D.K.); (M.K.); (N.R.); (C.F.); (X.M.); (R.T.)
- Department of Otolaryngology, New York Medical College, Valhalla, NY 10595, USA
| | - Jan Geliebter
- Department of Pathology, Microbiology & Immunology, New York Medical College, Valhalla, NY 10595, USA; (A.R.M.); (D.K.); (M.K.); (N.R.); (C.F.); (X.M.); (R.T.)
- Department of Otolaryngology, New York Medical College, Valhalla, NY 10595, USA
| | - Xiu-Min Li
- Department of Pathology, Microbiology & Immunology, New York Medical College, Valhalla, NY 10595, USA; (A.R.M.); (D.K.); (M.K.); (N.R.); (C.F.); (X.M.); (R.T.)
- Department of Otolaryngology, New York Medical College, Valhalla, NY 10595, USA
- Department of Dermatology, New York Medical College, Valhalla, NY 10595, USA
| |
Collapse
|
39
|
Abdalhadi HM, Chatham WW, Alduraibi FK. CAR-T-Cell Therapy for Systemic Lupus Erythematosus: A Comprehensive Overview. Int J Mol Sci 2024; 25:10511. [PMID: 39408836 PMCID: PMC11476835 DOI: 10.3390/ijms251910511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disorder characterized by the production of autoreactive B and T cells and cytokines, leading to chronic inflammation affecting multiple organs. SLE is associated with significant complications that substantially increase morbidity and mortality. Given its complex pathogenesis, conventional treatments for SLE often have significant side effects and limited efficacy, necessitating the exploration of novel therapeutic strategies. One promising approach is the use of chimeric antigen receptor (CAR)-T-cell therapy, which has shown remarkable success in treating refractory hematological malignancies. This review provides a comprehensive analysis of the current use of CAR-T-cell therapy in SLE.
Collapse
Affiliation(s)
- Haneen M. Abdalhadi
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Walter W. Chatham
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Nevada, Las Vegas, NV 89102, USA;
| | - Fatima K. Alduraibi
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
- Department of Medicine, Division of Clinical Immunology and Rheumatology, Harvard Teaching Hospital, Boston, MA 02215, USA
- Department of Medicine, Division of Clinical Immunology and Rheumatology, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| |
Collapse
|
40
|
Chen Z, Sun J, Shi T, Song C, Wu C, Wu Z, Lin J. Causal roles of circulating cytokines in sarcopenia-related traits: a Mendelian randomization study. Front Endocrinol (Lausanne) 2024; 15:1370985. [PMID: 39345889 PMCID: PMC11427268 DOI: 10.3389/fendo.2024.1370985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 08/28/2024] [Indexed: 10/01/2024] Open
Abstract
Background Epidemiological and experimental evidence suggests that chronic inflammation plays an important role in the onset and progression of sarcopenia. However, there is inconsistent data on the inflammatory cytokines involved in the pathogenesis of sarcopenia. Therefore, we performed a two-sample Mendelian randomization (MR) analysis to explore the causal relationship between circulating cytokines and sarcopenia-related traits. Methods The MR analysis utilized genetic data from genome-wide association study that included genetic variations in 41 circulating cytokines and genetic variant data for appendicular lean mass (ALM), hand grip strength, and usual walking pace. Causal associations were primarily explored using the inverse variance-weighted (IVW) method, supplemented by MR-Egger, simple mode, weighted median, and weighted mode analyses. Additionally, sensitivity analyses were also performed to ensure the reliability and stability of the results. Results Three cytokines [hepatocyte growth factor (HGF), interferon gamma-induced protein 10 (IP-10), and macrophage colony-stimulating factor (M-CSF)] were positively associated with ALM (β: 0.0221, 95% confidence interval (CI): 0.0071, 0.0372, P= 0.0039 for HGF; β: 0.0096, 95%CI: 4e-04, 0.0189, P= 0.0419 for IP-10; and β: 0.0100, 95%CI: 0.0035, 0.0165, P= 0.0025 for M-CSF). Conversely, higher levels of interleukin-7 (IL-7), monocyte chemotactic protein 3 (MCP-3), and regulated on activation, normal T cell expressed and secreted (RANTES) were associated with decreased hand grip strength (β: -0.0071, 95%CI: -0.0127, -0.0014, P= 0.0140 for IL-7; β: -0.0064, 95%CI: -0.0123, -6e-04, P= 0.0313 for MCP-3; and β: -0.0082, 95%CI: -0.0164, -1e-04, P= 0.0480 for RANTES). Similarly, interleukin 1 receptor antagonist (IL-1RA) was negatively correlated with usual walking pace (β: -0.0104, 95%CI: -0.0195, -0.0013, P= 0.0254). Sensitivity analysis confirmed the robustness of these findings. Conclusions Our study provides additional insights into the pivotal role of specific inflammatory cytokines in the pathogenesis of sarcopenia. Further research is required to determine whether these cytokines can be used as targets for the prevention and treatment of sarcopenia.
Collapse
Affiliation(s)
- Zhi Chen
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Jun Sun
- Department of Emergency, Zhaotong Traditional Chinese Medicine Hospital, Zhaotong, Yunnan, China
| | - Tengbin Shi
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Chenyang Song
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Chengjian Wu
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Zhengru Wu
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Jiajun Lin
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| |
Collapse
|
41
|
Zhu W, Guo S, Sun J, Zhao Y, Liu C. Lactate and lactylation in cardiovascular diseases: current progress and future perspectives. Metabolism 2024; 158:155957. [PMID: 38908508 DOI: 10.1016/j.metabol.2024.155957] [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: 12/06/2023] [Revised: 06/10/2024] [Accepted: 06/17/2024] [Indexed: 06/24/2024]
Abstract
Cardiovascular diseases (CVDs) are often linked to structural and functional impairments, such as heart defects and circulatory dysfunction, leading to compromised peripheral perfusion and heightened morbidity risks. Metabolic remodeling, particularly in the context of cardiac fibrosis and inflammation, is increasingly recognized as a pivotal factor in the pathogenesis of CVDs. Metabolic syndromes further predispose individuals to these conditions, underscoring the need to elucidate the metabolic underpinnings of CVDs. Lactate, a byproduct of glycolysis, is now recognized as a key molecule that connects cellular metabolism with the regulation of cellular activity. The transport of lactate between different cells is essential for metabolic homeostasis and signal transduction. Disruptions to lactate dynamics are implicated in various CVDs. Furthermore, lactylation, a novel post-translational modification, has been identified in cardiac cells, where it influences protein function and gene expression, thereby playing a significant role in CVD pathogenesis. In this review, we summarized recent advancements in understanding the role of lactate and lactylation in CVDs, offering fresh insights that could guide future research directions and therapeutic interventions. The potential of lactate metabolism and lactylation as innovative therapeutic targets for CVD is a promising avenue for exploration.
Collapse
Affiliation(s)
- Wengen Zhu
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China; Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou 510080, PR China.
| | - Siyu Guo
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China; Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou 510080, PR China
| | - Junyi Sun
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China
| | - Yudan Zhao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430023, PR China.
| | - Chen Liu
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China; Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou 510080, PR China.
| |
Collapse
|
42
|
Shen X, Zhang X, Li K, Huang G, Li X, Hou Y, Ge X. Combined bacterial translocation and cholestasis aggravates liver injury by activation pyroptosis in obstructive jaundice. Heliyon 2024; 10:e35793. [PMID: 39220957 PMCID: PMC11363856 DOI: 10.1016/j.heliyon.2024.e35793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 08/02/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024] Open
Abstract
This study explores the mechanism by which obstructive jaundice (OJ) induces liver damage through pyroptosis. We induced OJ in rats via bile duct ligation and assessed liver damage using serum biochemical markers and histological analysis of liver tissue. Pyroptosis was investigated through immunofluorescence, ELISA, Western blot, and quantitative RT-PCR techniques. Additionally, we examined intestinal function and fecal microbiota alterations in the rats using 16S rDNA sequencing. In vitro experiments involved co-culturing Kupffer cells and hepatocytes, which were then exposed to bile and lipopolysaccharide (LPS). Our findings indicated that OJ modified the gut microbiota, increasing LPS levels, which, in conjunction with bile, initiated a cycle of inflammation, fibrosis, and cell death in the liver. Mechanistically, OJ elevated necrotic markers such as ATP, which in turn activated pyroptotic pathways. Increased levels of pyroptosis-related molecules, including NLRP3, caspase-1, gasdermin D, and IL-18, were confirmed. In our co-cultured cell model, bile exposure resulted in cell death and ATP release, leading to the activation of the NLRP3 inflammasome and its downstream effectors, caspase-1 and IL-18. The combination of bile and LPS significantly intensified pyroptotic responses. This study is the first to demonstrate that LPS and bile synergistically exacerbate liver injury by promoting necrosis and pyroptosis, unveiling a novel mechanism of OJ-associated hepatic damage and suggesting avenues for potential preventive or therapeutic interventions.
Collapse
Affiliation(s)
- Xin Shen
- Department of Pharmacy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xin Zhang
- Luoyang Orthopedic-Traumatological Hospital of Henan Province (Henan Provincial Orthopedic Hospital), Luoyang, 471002, Henan, China
| | - Kaiyu Li
- Department of General Surgery, Heilongjiang Provincial Hospital, 82 Zhongshan Road, Harbin, 150036, Heilongjiang, China
| | - Guangming Huang
- Department of General Surgery, Heilongjiang Provincial Hospital, 82 Zhongshan Road, Harbin, 150036, Heilongjiang, China
| | - Xinyu Li
- Department of General Surgery, Heilongjiang Provincial Hospital, 82 Zhongshan Road, Harbin, 150036, Heilongjiang, China
| | - Yunlong Hou
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine, Shijiazhuang, 050000, Hebei, China
| | - Xin Ge
- Department of General Surgery, Heilongjiang Provincial Hospital, 82 Zhongshan Road, Harbin, 150036, Heilongjiang, China
| |
Collapse
|
43
|
Petruccioli E, Sbarra S, Vita S, Salmi A, Cuzzi G, De Marco P, Matusali G, Navarra A, Pierelli L, Grifoni A, Sette A, Maggi F, Nicastri E, Goletti D. Characterization of the Monkeypox Virus [MPX]-Specific Immune Response in MPX-Cured Individuals Using Whole Blood to Monitor Memory Response. Vaccines (Basel) 2024; 12:964. [PMID: 39339995 PMCID: PMC11436000 DOI: 10.3390/vaccines12090964] [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: 07/17/2024] [Revised: 08/13/2024] [Accepted: 08/23/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Monkeypox (Mpox) is a zoonotic disease caused by monkeypox virus (MPXV), an Orthopoxvirus (OPXV). Since we are observing the first MPXV outbreak outside the African continent, the general population probably does not have a pre-existing memory response for MPXV but may have immunity against the previous smallpox vaccine based on a live replicating Vaccinia strain (VACV). Using a whole blood platform, we aim to study the MPXV- T-cell-specific response in Mpox-cured subjects. METHODS We enrolled 16 subjects diagnosed with Mpox in the previous 3-7 months and 15 healthy donors (HD) with no recent vaccination history. Whole blood was stimulated overnight with MPXV and VACV peptides to elicit CD4 and CD8 T-cell-specific responses, which were evaluated by ELISA and multiplex assay. RESULTS Mpox-cured subjects showed a significant IFN-γ T-cell response to MPXV and VACV. Besides IFN-γ, IL-6, IP-10, IL-8, IL-2, G-CSF, MCP-1, MIP1-α, MIP-1β, IL-1Rα, and IL-5 were significantly induced after specific stimulation compared to the unstimulated control. The specific response was mainly induced by the CD4 peptides MPX-CD4-E and VACV-CD4. CONCLUSIONS We showed that MPXV-specific responses have a mixed Th1- and Th2-response in a whole blood platform assay, which may be useful for monitoring the specific immunity induced by vaccination or infection.
Collapse
Affiliation(s)
- Elisa Petruccioli
- Translational Research Unit, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy; (E.P.); (S.S.)
| | - Settimia Sbarra
- Translational Research Unit, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy; (E.P.); (S.S.)
| | - Serena Vita
- Highly Infectious Diseases Isolation Unit, Clinical Department, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| | - Andrea Salmi
- Translational Research Unit, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy; (E.P.); (S.S.)
| | - Gilda Cuzzi
- Translational Research Unit, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy; (E.P.); (S.S.)
| | - Patrizia De Marco
- Highly Infectious Diseases Isolation Unit, Clinical Department, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| | - Giulia Matusali
- Laboratory of Virology and Biosafety Laboratories, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| | - Assunta Navarra
- Clinical Epidemiology Unit, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| | - Luca Pierelli
- Unità Operativa Complessa (UOC) Transfusion Medicine and Stem Cell, San Camillo Forlanini Hospital, 00149 Rome, Italy
| | - Alba Grifoni
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Alessandro Sette
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Fabrizio Maggi
- Laboratory of Virology and Biosafety Laboratories, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| | - Emanuele Nicastri
- Highly Infectious Diseases Isolation Unit, Clinical Department, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy; (E.P.); (S.S.)
| |
Collapse
|
44
|
Li Z, Zeng M, Wu T, Wang Z, Sun Y, Zhang Z, Wu F, Chen Z, Fu M, Meng F. Causal Effects of COVID-19 on the Risk of Thrombosis: A Two-Sample Mendel Randomization Study. Thromb Haemost 2024; 124:709-720. [PMID: 38325400 PMCID: PMC11259497 DOI: 10.1055/a-2263-8514] [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: 07/19/2023] [Accepted: 11/17/2023] [Indexed: 02/09/2024]
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) and thrombosis are linked, but the biomolecular mechanism is unclear. We aimed to investigate the causal relationship between COVID-19 and thrombotic biomarkers. METHODS We used two-sample Mendelian randomization (MR) to assess the effect of COVID-19 on 20 thrombotic biomarkers. We estimated causality using inverse variance weighting with multiplicative random effect, and performed sensitivity analysis using weighted median, MR-Egger regression and MR Pleiotropy Residual Sum and Outlier (MR-PRESSO) methods. All the results were examined by false discovery rate (FDR) with the Benjamin and Hochberg method for this correction to minimize false positives. We used R language for the analysis. RESULTS All COVID-19 classes showed lower levels of tissue factor pathway inhibitor (TFPI) and interleukin-1 receptor type 1 (IL-1R1). COVID-19 significantly reduced TFPI (odds ratio [OR] = 0.639, 95% confidence interval [CI]: 0.435-0.938) and IL-1R1 (OR = 0.603, 95% CI = 0.417-0.872), nearly doubling the odds. We also found that COVID-19 lowered multiple coagulation factor deficiency protein 2 and increased C-C motif chemokine 3. Hospitalized COVID-19 cases had less plasminogen activator, tissue type (tPA) and P-selectin glycoprotein ligand 1 (PSGL-1), while severe cases had higher mean platelet volume (MPV) and lower platelet count. These changes in TFPI, tPA, IL-1R1, MPV, and platelet count suggested a higher risk of thrombosis. Decreased PSGL-1 indicated a lower risk of thrombosis. CONCLUSION TFPI, IL-1R, and seven other indicators provide causal clues of the pathogenesis of COVID-19 and thrombosis. This study demonstrated that COVID-19 causally influences thrombosis at the biomolecular level.
Collapse
Affiliation(s)
- Zhengran Li
- The Second Clinical Medicine School, Southern Medical University, Guangzhou, Guangdong, China
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Minghui Zeng
- Institute of Scientific Research, Southern Medical University, Guangzhou, China
| | - Tong Wu
- The First Clinical Medicine School, Southern Medical University, Guangzhou, Guangdong, China
| | - Zijin Wang
- The Second Clinical Medicine School, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuxin Sun
- The Second Clinical Medicine School, Southern Medical University, Guangzhou, Guangdong, China
| | - Ziran Zhang
- The Second Clinical Medicine School, Southern Medical University, Guangzhou, Guangdong, China
| | - Fanye Wu
- The Second Clinical Medicine School, Southern Medical University, Guangzhou, Guangdong, China
| | - Zejun Chen
- The Second Clinical Medicine School, Southern Medical University, Guangzhou, Guangdong, China
| | - Min Fu
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Fanke Meng
- Emergency Department, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
45
|
Zhang X, Liu T, Ran C, Wang W, Piao F, Yang J, Tian S, Li L, Zhao D. Immunoregulatory paracrine effect of mesenchymal stem cells and mechanism in the treatment of osteoarthritis. Front Cell Dev Biol 2024; 12:1411507. [PMID: 39129785 PMCID: PMC11310049 DOI: 10.3389/fcell.2024.1411507] [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: 04/03/2024] [Accepted: 07/08/2024] [Indexed: 08/13/2024] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease caused by chronic inflammation that damages articular cartilage. At present, the treatment of OA includes drug therapy to relieve symptoms and joint replacement therapy for advanced OA. However, these palliatives cannot truly block the progression of the disease from the immunological pathogenesis of OA. In recent years, bone marrow mesenchymal stem cell (BMSC) transplantation has shown great potential in tissue engineering repair. In addition, many studies have shown that BMSC paracrine signals play an important role in the treatment of OA through immune regulation and suppressing inflammation. At present, the mechanism of inflammation-induced OA and the use of BMSC transplantation in joint repair have been reviewed, but the mechanism and significance of BMSC paracrine signals in the treatment of OA have not been fully reviewed. Therefore, this article focused on the latest research progress on the paracrine effects of BMSCs in the treatment of OA and the related mechanisms by which BMSCs secrete cytokines to inhibit the inflammatory response, regulate immune balance, and promote cell proliferation and differentiation. In addition, the application potential of BMSC-Exos as a new type of cell-free therapy for OA is described. This review aimed to provide systematic theoretical support for the clinical application of BMSC transplantation in the treatment of OA.
Collapse
Affiliation(s)
- Xiuzhi Zhang
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Tianhao Liu
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Chunxiao Ran
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Weidan Wang
- Orthopaedic Medical Research Center, Dalian University, Dalian, Liaoning, China
| | - Fengyuan Piao
- Orthopaedic Medical Research Center, Dalian University, Dalian, Liaoning, China
| | - Jiahui Yang
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Simiao Tian
- Orthopaedic Medical Research Center, Dalian University, Dalian, Liaoning, China
| | - Lu Li
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Dewei Zhao
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| |
Collapse
|
46
|
Zhou CJ, Guo Y. Mini review on collagens in normal skin and pathological scars: current understanding and future perspective. Front Med (Lausanne) 2024; 11:1449597. [PMID: 39091289 PMCID: PMC11291465 DOI: 10.3389/fmed.2024.1449597] [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: 06/15/2024] [Accepted: 07/05/2024] [Indexed: 08/04/2024] Open
Abstract
Pathological scar tissues are characterized by the presence of overabundant collagens whose structure and organization are also different from those in unwounded skin. This causes scar tissues to lose some functions performed by normal skin, and currently, there are no effective measures to prevent scar formation. Inflammation has been shown to modulate fibroblast proliferation, differentiation, and function, hence collagen production and organization. In this minireview, we provide an overview of the current understanding of collagen, specifically collagen type I and III which are main collagens in skin, structure and fibre formation and highlight their differences between normal skin and pathological scars. We discuss the role that cytokines play in modulating fibroblast function. We also identify some potential research directions which could help to further our understanding of the complex and dynamic wound healing and scar formation process.
Collapse
Affiliation(s)
| | - Yuan Guo
- School of Food Science and Nutrition, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
47
|
Schmauch E, Severin Y, Xing X, Mangold A, Conrad C, Johansen P, Kahlenberg JM, Mellett M, Navarini A, Nobbe S, Sarkar MK, Satyam A, Tsoi LC, French LE, Nilsson J, Linna-Kuosmanen S, Kaikkonen MU, Snijder B, Kellis M, Gudjonsson JE, Tsokos GC, Contassot E, Kolios AGA. Targeting IL-1 controls refractory pityriasis rubra pilaris. SCIENCE ADVANCES 2024; 10:eado2365. [PMID: 38959302 PMCID: PMC11221491 DOI: 10.1126/sciadv.ado2365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/29/2024] [Indexed: 07/05/2024]
Abstract
Pityriasis rubra pilaris (PRP) is a rare inflammatory skin disease with a poorly understood pathogenesis. Through a molecularly driven precision medicine approach and an extensive mechanistic pathway analysis in PRP skin samples, compared to psoriasis, atopic dermatitis, healed PRP, and healthy controls, we identified IL-1β as a key mediator, orchestrating an NF-κB-mediated IL-1β-CCL20 axis, including activation of CARD14 and NOD2. Treatment of three patients with the IL-1 antagonists anakinra and canakinumab resulted in rapid clinical improvement and reversal of the PRP-associated molecular signature with a 50% improvement in skin lesions after 2 to 3 weeks. This transcriptional signature was consistent with in vitro stimulation of keratinocytes with IL-1β. With the central role of IL-1β underscoring its potential as a therapeutic target, our findings propose a redefinition of PRP as an autoinflammatory keratinization disorder. Further clinical trials are needed to validate the efficacy of IL-1β antagonists in PRP.
Collapse
Affiliation(s)
- Eloi Schmauch
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Yannik Severin
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, 8049 Zurich, Switzerland
| | - Xianying Xing
- Departments of Internal Medicine and Dermatology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Aaron Mangold
- Department of Dermatology, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
| | - Curdin Conrad
- Department of Dermatology, CHUV University Hospital and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Pål Johansen
- Department of Dermatology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - J. Michelle Kahlenberg
- Departments of Internal Medicine and Dermatology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mark Mellett
- Department of Dermatology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Alexander Navarini
- Department of Biomedicine and Dermatology Department, University Hospital of Basel, Basel, Switzerland
| | - Stefan Nobbe
- Department of Dermatology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
- Department of Dermatology, Cantonal Hospital Frauenfeld, Frauenfeld, Switzerland
| | - Mrinal K. Sarkar
- Departments of Internal Medicine and Dermatology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Abhigyan Satyam
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Lam C. Tsoi
- Departments of Internal Medicine and Dermatology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lars E. French
- Department of Dermatology and Allergology, Ludwig Maximilian University of Munich, Munich, Germany
- Dr. Philip Frost, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL 33125, USA
| | - Jakob Nilsson
- Department of Immunology, University Hospital Zurich, Zurich, Switzerland
| | - Suvi Linna-Kuosmanen
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Minna U. Kaikkonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Berend Snijder
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, 8049 Zurich, Switzerland
| | - Manolis Kellis
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Johann E. Gudjonsson
- Departments of Internal Medicine and Dermatology, University of Michigan, Ann Arbor, MI 48109, USA
- Taubman Medical Research Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - George C. Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Emmanuel Contassot
- Department of Biomedicine and Dermatology Department, University Hospital of Basel, Basel, Switzerland
| | - Antonios G. A. Kolios
- Department of Dermatology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- University of Zurich, Zurich, Switzerland
| |
Collapse
|
48
|
Dal-Fabbro R, Yu M, Mei L, Sasaki H, Schwendeman A, Bottino MC. Synthetic high-density lipoprotein (sHDL): a bioinspired nanotherapeutics for managing periapical bone inflammation. Int J Oral Sci 2024; 16:50. [PMID: 38956025 PMCID: PMC11219839 DOI: 10.1038/s41368-024-00316-w] [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: 11/27/2023] [Revised: 06/06/2024] [Accepted: 06/11/2024] [Indexed: 07/04/2024] Open
Abstract
Apical periodontitis (AP) is a dental-driven condition caused by pathogens and their toxins infecting the inner portion of the tooth (i.e., dental pulp tissue), resulting in inflammation and apical bone resorption affecting 50% of the worldwide population, with more than 15 million root canals performed annually in the United States. Current treatment involves cleaning and decontaminating the infected tissue with chemo-mechanical approaches and materials introduced years ago, such as calcium hydroxide, zinc oxide-eugenol, or even formalin products. Here, we present, for the first time, a nanotherapeutics based on using synthetic high-density lipoprotein (sHDL) as an innovative and safe strategy to manage dental bone inflammation. sHDL application in concentrations ranging from 25 µg to 100 µg/mL decreases nuclear factor Kappa B (NF-κB) activation promoted by an inflammatory stimulus (lipopolysaccharide, LPS). Moreover, sHDL at 500 µg/mL concentration markedly decreases in vitro osteoclastogenesis (P < 0.001), and inhibits IL-1α (P = 0.027), TNF-α (P = 0.004), and IL-6 (P < 0.001) production in an inflammatory state. Notably, sHDL strongly dampens the Toll-Like Receptor signaling pathway facing LPS stimulation, mainly by downregulating at least 3-fold the pro-inflammatory genes, such as Il1b, Il1a, Il6, Ptgs2, and Tnf. In vivo, the lipoprotein nanoparticle applied after NaOCl reduced bone resorption volume to (1.3 ± 0.05) mm3 and attenuated the inflammatory reaction after treatment to (1 090 ± 184) cells compared to non-treated animals that had (2.9 ± 0.6) mm3 (P = 0.012 3) and (2 443 ± 931) cells (P = 0.004), thus highlighting its promising clinical potential as an alternative therapeutic for managing dental bone inflammation.
Collapse
Affiliation(s)
- Renan Dal-Fabbro
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Minzhi Yu
- Department of Pharmaceutical Sciences, College of Pharmacy and Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Ling Mei
- Department of Pharmaceutical Sciences, College of Pharmacy and Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Hajime Sasaki
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, College of Pharmacy and Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Marco C Bottino
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
49
|
Hu B, Korsos V, Palomba ML. Chimeric antigen receptor T-cell therapy for aggressive B-cell lymphomas. Front Oncol 2024; 14:1394057. [PMID: 39011476 PMCID: PMC11246842 DOI: 10.3389/fonc.2024.1394057] [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: 02/29/2024] [Accepted: 05/21/2024] [Indexed: 07/17/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is a revolutionary approach in the treatment of lymphoma. This review article provides an overview of the four FDA-approved CAR T-cell products for aggressive B-cell lymphoma, including diffuse large B-cell lymphoma and mantle cell lymphoma, highlighting their efficacy and toxicity as well as discussing future directions.
Collapse
Affiliation(s)
- Bei Hu
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute/Wake Forest School of Medicine, Charlotte, NC, United States
| | - Victoria Korsos
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - M. Lia Palomba
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
50
|
Tan X, Pan J, Cai J, Jiang S, Shu F, Xu M, Peng H, Tang J, Zhang H. Relevant Research of Inflammatory Cytokines Spectrum in Peripheral Blood of Sudden Hearing Loss. Laryngoscope 2024; 134:3293-3301. [PMID: 38193513 DOI: 10.1002/lary.31276] [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: 09/19/2023] [Revised: 12/24/2023] [Accepted: 12/27/2023] [Indexed: 01/10/2024]
Abstract
OBJECTIVE To investigate whether there is a correlation between the inflammatory state and the pathogenesis and clinical features of sudden hearing loss (SHL) by studying the expression of inflammation-related cytokines in the peripheral blood of patients with SHL. METHODS In this work, we analyzed the cytokine profiles of 48 analytes in 38 patients with SHL compared to 38 healthy donors using a multiplex immunoassay. This study used appropriate statistical methods to screen for inflammatory cytokines associated with the pathogenesis of SHL, to analyze their network correlation, and to analyze the relationship between clinical features of SHL and inflammatory cytokines. RESULTS Several cytokines, including CTACK, Eotaxin, HGF, INF-α2, IFN-β, IL-1β, IL-1ra, IL-2Rα, IL-4, IL-7, IL-8, IL-9, IL-10, IL-12(p40), IL-13, MIG, β-NGF, SCF, and TNF-α, exhibited significantly higher levels in the peripheral blood of the SHL group compared to the control group. An inflammatory network composed of multiple cytokines, including IL-1β, is a risk factor for the development of SHL. CONCLUSION This study identified several inflammatory cytokines with elevated expression, which may be linked with the onset of SHL. The results of this study also provide a basis for the theoretical hypothesis of inflammation in SHL. LEVEL OF EVIDENCE 3 Laryngoscope, 134:3293-3301, 2024.
Collapse
Affiliation(s)
- Xinyuan Tan
- Department of Otolaryngology Head and Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Ear Research Institute, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jing Pan
- Department of Otolaryngology Head and Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Ear Research Institute, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jieqing Cai
- Department of Otolaryngology Head and Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Ear Research Institute, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shanshan Jiang
- Department of Otolaryngology Head and Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Ear Research Institute, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Fan Shu
- Department of Otolaryngology Head and Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Ear Research Institute, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Muqing Xu
- Department of Otolaryngology Head and Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Ear Research Institute, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Hua Peng
- Department of Otolaryngology Head and Neck Surgery, General Hospital of Southern Theatre Command of PLA, Guangzhou, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jie Tang
- Ear Research Institute, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hongzheng Zhang
- Department of Otolaryngology Head and Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Ear Research Institute, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|