1
|
Abdelaziz HM, Abdelmageed ME, Suddek GM. Molsidomine ameliorates DEXA-induced insulin resistance: Involvement of HMGB1/JAK1/STAT3 signaling pathway. Eur J Pharmacol 2025:177832. [PMID: 40490173 DOI: 10.1016/j.ejphar.2025.177832] [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: 09/18/2024] [Revised: 06/01/2025] [Accepted: 06/06/2025] [Indexed: 06/11/2025]
Abstract
Insulin resistance (IR) is a serious clinical syndrome that establishes the basis for illnesses like type 2 diabetes (T2D). In this study, the effectiveness of molsidomine (MOLS) which is a nitric oxide (NO) doner, on dexamethasone (DEXA)- induced IR in rats was examined. Male Wistar rats were managed with MOLS (5 and 10 mg/kg) orally once daily for 7 days before DEXA injection (1 mg/kg, intraperitoneally (i.p.)) and 7 days concurrent with DEXA injection. The findings showed that MOLS reduced low-density lipoprotein cholesterol (LDL-C), fasting serum glucose and insulin, homeostatic model assessment of insulin resistance (HOMA-IR), alanine transaminase (ALT), aspartate transaminase (AST), oral glucose tolerance test (OGTT), and triglycerides (TGs). These findings revealed that MOLS was successful in reducing DEXA-induced IR. Moreover, MOLS was associated with a large increase in reduced glutathione (GSH) and superoxide dismutase (SOD) activity as well as a significant decrease in the levels of malondialdehyde (MDA) in hepatic and aortic tissues. When compared to rats treated with DEXA, MOLS significantly decreased the levels of pro-inflammatory cytokine interlukin-6 (IL-6), high mobility group box 1 (HMGB1), phosphorylated Janus kinase1/phosphorylated signal transducer and activator of transcription 3 (p-JAK1/p-STAT3), and nuclear factor kappa-B-p65 subunit (NF-κB-p65) in hepatic tissues. Additionally, MOLS reduced inflammation and necrosis and increased B cell/lymphoma 2 (BCL-2) and lowered caspase-3 levels and attenuated liver histopathological changes. Moreover, aortic expression levels of NF-κB-p65 and IL-6 were reduced upon MOLS treatment. All these findings show that MOLS protects rats from DEXA-induced IR by inhibiting HMGB1/JAK1/STAT3 signaling, regulating oxidative stress and inflammatory pathways, and having an antioxidant and anti-inflammatory effect.
Collapse
Affiliation(s)
- Howida M Abdelaziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516 Mansoura, Egypt.
| | - Marwa E Abdelmageed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516 Mansoura, Egypt.
| | - Ghada M Suddek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516 Mansoura, Egypt.
| |
Collapse
|
2
|
Zhao JY, Zhou Y, Zhou CW, Zhan KB, Yang M, Wen M, Zhu LQ. Revisiting the critical roles of reactive microglia in traumatic brain injury. Int J Surg 2025; 111:3942-3978. [PMID: 40358653 PMCID: PMC12165506 DOI: 10.1097/js9.0000000000002420] [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: 11/06/2024] [Accepted: 04/11/2025] [Indexed: 05/15/2025]
Abstract
Traumatic brain injury (TBI) triggers a complex neuroinflammatory cascade, with microglia serving as key regulators of both pathological damage and tissue structural restoration. Despite extensive research, the precise temporal evolution of microglial activation and its implications for long-term neurological outcomes remain incompletely understood. Here, we provide a comprehensive review of the molecular and cellular mechanisms underlying microglial responses in TBI, highlighting their role in neuroinflammation, neurogenesis, and tissue remodeling. We systematically compare clinical and preclinical TBI classifications, lesion patterns, and animal modeling strategies, evaluating their translational relevance. Furthermore, we explore the limitations of the conventional M1/M2 dichotomy and emphasize recent insights from single-cell transcriptomic analyses that reveal distinct microglial subpopulations across different injury phases. Finally, we discuss current therapeutic strategies targeting microglial modulation and propose future directions for neuroimmune interventions in TBI. By integrating findings from experimental and clinical studies, this review aims to bridge mechanistic insights with therapeutic advancements, paving the way for precision-targeted neuroimmune therapies.
Collapse
Affiliation(s)
- Jing-Yu Zhao
- Department of Neurosurgery, Wuhan Hankou Hospital, Hankou Hospital Affiliated to Wuhan University of Science and Technology, Jiang`an District, Wuhan, People’s Republic of China
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yang Zhou
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Chao-Wen Zhou
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Ke-Bin Zhan
- Department of Neurology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
| | - Ming Yang
- Department of Neurosurgery, Wuhan Hankou Hospital, Hankou Hospital Affiliated to Wuhan University of Science and Technology, Jiang`an District, Wuhan, People’s Republic of China
| | - Ming Wen
- Department of Neurosurgery, Wuhan Hankou Hospital, Hankou Hospital Affiliated to Wuhan University of Science and Technology, Jiang`an District, Wuhan, People’s Republic of China
| | - Ling-Qiang Zhu
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
3
|
Higashi Y, Munesue S, Saeki M, Harashima A, Kimura K, Oshima Y, Takei R, Takada S, Nakanuma S, Makino I, Ohta T, Yagi S, Tajima H, Yamamoto Y. Platelet aggregation elicits FasL expression and hepatocyte apoptosis in sinusoidal obstruction syndrome. Sci Rep 2025; 15:18859. [PMID: 40442395 PMCID: PMC12122828 DOI: 10.1038/s41598-025-03839-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 05/22/2025] [Indexed: 06/02/2025] Open
Abstract
Sinusoidal obstruction syndrome (SOS) is a fatal liver condition resulting from sinusoidal endothelial cell injury and hepatocyte death, following liver or hematopoietic stem cell transplantation as well as chemotherapy. We showed evidence of platelet displacement and aggregation within the space of Disse in SOS. However, the relationship between platelets and hepatocyte death remains unclear. Using a mouse SOS model by intraperitoneal monocrotaline (270 mg/kg; a pyrrolizidine alkaloide plant toxin) administration, we observed positive stains for terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and cleaved caspase-3, which are markers for apoptosis, in the liver by immunohistochemistry. At 48 h of the SOS liver, aggregated platelets and hepatocytes around zone 3 were found to express Fas ligand (FasL) and Fas, respectively. Human peripheral blood platelets, when aggregated, could induce expression of FASL on themselves and then lead to apoptosis in co-cultured HepG2 cells. Treatment of recombinant soluble thrombomodulin (rTM), an anticoagulant and vascular endothelium-protective drug, prevented the hepatocyte death in the SOS mice. These findings suggest that the prevention of platelet aggregation is a potential therapeutic intervention against hepatocyte death and severe liver damage in SOS.
Collapse
Grants
- 20K07323 Grant-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology, Japan
- 20K09029 Grant-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology, Japan
- 21H02695 Grant-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology, Japan
Collapse
Affiliation(s)
- Yuri Higashi
- Department of Hepato-Biliary-Pancreatic Surgery and Transplantation/Pediatric Surgery, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, 920-8641, Japan
| | - Seiichi Munesue
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, 920-8640, Japan
| | - Masakazu Saeki
- Department of Hepato-Biliary-Pancreatic Surgery and Transplantation/Pediatric Surgery, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, 920-8641, Japan
| | - Ai Harashima
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, 920-8640, Japan
| | - Kumi Kimura
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, 920-8640, Japan
| | - Yu Oshima
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, 920-8640, Japan
| | - Ryohei Takei
- Department of Hepato-Biliary-Pancreatic Surgery and Transplantation/Pediatric Surgery, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, 920-8641, Japan
| | - Satoshi Takada
- Department of Hepato-Biliary-Pancreatic Surgery and Transplantation/Pediatric Surgery, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, 920-8641, Japan
| | - Shinichi Nakanuma
- Department of Hepato-Biliary-Pancreatic Surgery and Transplantation/Pediatric Surgery, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, 920-8641, Japan
| | - Isamu Makino
- Department of Hepato-Biliary-Pancreatic Surgery and Transplantation/Pediatric Surgery, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, 920-8641, Japan
| | - Tetsuo Ohta
- Department of Hepato-Biliary-Pancreatic Surgery and Transplantation/Pediatric Surgery, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, 920-8641, Japan
| | - Shintaro Yagi
- Department of Hepato-Biliary-Pancreatic Surgery and Transplantation/Pediatric Surgery, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, 920-8641, Japan
| | - Hidehiro Tajima
- Department of Gastroenterological Surgery, Dokkyo Medical University Saitama Medical Center, 2-1-50 Minami-Koshigaya, Koshigaya, 343-8555, Japan
| | - Yasuhiko Yamamoto
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, 920-8640, Japan.
| |
Collapse
|
4
|
Yan F, Qiao Y, Pan S, Kang A, Chen H, Bai Y. RIPK1: A Promising Target for Intervention Neuroinflammation. J Neuroimmune Pharmacol 2025; 20:59. [PMID: 40418439 DOI: 10.1007/s11481-025-10208-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/16/2025] [Indexed: 05/27/2025]
Abstract
Necroptosis is a novel mode of cell death that differs from traditional apoptosis, characterized by distinct molecular mechanisms and physiopathological features. Recent research has increasingly underscored the pivotal role of necroptosis in various neurological diseases, including stroke, Alzheimer's disease and multiple sclerosis. A defining hallmark of these conditions is neuroinflammation, a complex inflammatory response that critically influences neuronal survival. This review provides a comprehensive analysis of the mechanistic underpinnings of necroptosis and its intricate interplay with neuroinflammation, exploring the interrelationship between the two processes and their impact on neurological disorders. In addition, we discuss potential therapeutic strategies that target the intervention of necroptosis and neuroinflammation, offering novel avenues for intervention. By deepening our understanding of these interconnected processes, the development of more effective treatments approaches holds significant promise for improving patient outcomes in neurological disorders.
Collapse
Affiliation(s)
- Feixing Yan
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Yujun Qiao
- Department of Pharmacy, Lanzhou University Second Hospital, Lanzhou, 730000, China
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Shunli Pan
- Department of Pharmacy, Lanzhou University Second Hospital, Lanzhou, 730000, China
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Anjuan Kang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Haile Chen
- Department of Pharmacy, Lanzhou University Second Hospital, Lanzhou, 730000, China
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Yinliang Bai
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China.
- Department of Pharmacy, Lanzhou University Second Hospital, Lanzhou, 730000, China.
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730000, China.
| |
Collapse
|
5
|
Stojanović SD, Thum T, Bauersachs J. Anti-senescence therapies: a new concept to address cardiovascular disease. Cardiovasc Res 2025; 121:730-747. [PMID: 40036821 PMCID: PMC12101330 DOI: 10.1093/cvr/cvaf030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/16/2024] [Accepted: 01/22/2025] [Indexed: 03/06/2025] Open
Abstract
Accumulation of senescent cells is an increasingly recognized factor in the development and progression of cardiovascular (CV) disease (CVD). Senescent cells of different types display a pro-inflammatory and matrix remodelling molecular programme, known as the 'senescence-associated secretory phenotype' (SASP), which has roots in (epi)genetic changes. Multiple therapeutic options (senolytics, anti-SASP senomorphics, and epigenetic reprogramming) that delete or ameliorate cellular senescence have recently emerged. Some drugs routinely used in the clinics also have anti-senescence effects. However, multiple challenges hinder the application of novel anti-senescence therapeutics in the clinical setting. Understanding the biology of cellular senescence, advantages and pitfalls of anti-senescence treatments, and patients who can profit from these interventions is necessary to introduce this novel therapeutic modality into the clinics. We provide a guide through the molecular machinery of senescent cells, systematize anti-senescence treatments, and propose a pathway towards senescence-adapted clinical trial design to aid future efforts.
Collapse
Affiliation(s)
- Stevan D Stojanović
- Department of Cardiology and Angiology, Hannover Medical School, Carl Neuberg Str. 1, Hannover 30625, Germany
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl Neuberg Str. 1, Hannover 30625, Germany
- PRACTIS Clinician Scientist Program, Dean’s Office for Academic Career Development, Hannover Medical School, Carl Neuberg Str. 1, Hannover 30625, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl Neuberg Str. 1, Hannover 30625, Germany
- Center for Translational Regenerative Medicine, Hannover Medical School, Carl Neuberg Str. 1, Hannover 30625, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Carl Neuberg Str. 1, Hannover 30625, Germany
- Center for Translational Regenerative Medicine, Hannover Medical School, Carl Neuberg Str. 1, Hannover 30625, Germany
| |
Collapse
|
6
|
Esperante I, Banzan C, Munuera JZ, Lima A, Hunt H, De Kloet ER, Deniselle MCG, De Nicola AF, Meyer M. The Selective Glucocorticoid Receptor Modulator Cort125329 Decreases Neuroinflammation and Gliosis and Enhances Myelination in the Wobbler Model of Amyotrophic Lateral Sclerosis. Mol Neurobiol 2025:10.1007/s12035-025-05048-4. [PMID: 40381165 DOI: 10.1007/s12035-025-05048-4] [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: 11/15/2024] [Accepted: 05/06/2025] [Indexed: 05/19/2025]
Abstract
The Wobbler mouse is a genetic model of familial amyotrophic lateral sclerosis. Wobblers show spinal cord neurodegeneration associated with gliosis, neuroinflammation, and demyelination. Like human neurodegenerative diseases, Wobblers show high levels of corticosterone in the blood and the nervous system. The role of glucocorticoids in neuropathology is suggested by the observation that pathological signs attenuate with treatment with glucocorticoid receptor (GR) antagonists/modulators. In the present study, we demonstrated in 5-month-old clinically afflicted Wobbler mice that the selective GR modulator CORT125329 decreased motoneuron degeneration, astro- and microgliosis, and levels of pro-inflammatory factors (HMGB1, toll-like receptor 4, tumor necrosis factor α, and its receptor). In addition, CORT125329 increased the acetylcholine-producing enzyme choline acetyltransferase, the neurotrophin brain-derived neurotrophic factor, and their cellular colocalization. Furthermore, the increased oligodendrocyte number and a healthier myelin ultrastructure are consistent with the enhanced axonal myelination after CORT125329 treatment. Finally, the high expression of immunoreactive protein and mRNA levels of aquaporin4 in Wobblers was decreased by CORT125329 treatment, implying this water channel is a glucocorticoid target involved in neuropathology. The beneficial effects of CORT125329 correlated with enhanced motor behavioral performance and trophic changes of the forelimbs. In conclusion, our results support further preclinical and clinical studies on GR modulators in sporadic amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Iván Esperante
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, Buenos Aires, Argentina
| | - Carolina Banzan
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, Buenos Aires, Argentina
| | - Jimena Zahn Munuera
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, Buenos Aires, Argentina
| | - Analía Lima
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, Buenos Aires, Argentina
| | - Hazel Hunt
- CORCEPT Therapeutics, Menlo Park, CA, USA
| | - E Ronald De Kloet
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
| | - María Claudia González Deniselle
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, Buenos Aires, Argentina
- Department of Physiology, Faculty of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Alejandro F De Nicola
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, Buenos Aires, Argentina.
- Department of Human Biochemistry, Faculty of Medicine, University of Buenos Aires, Buenos Aires, Argentina.
- Instituto de Biologia y Medicina Experimental, 2490, 1428, Buenos Aires, Obligado, Argentina.
| | - María Meyer
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, Buenos Aires, Argentina.
| |
Collapse
|
7
|
Zhang Y, Liu Y, Wan H, Deng H, Xu P, Cao L, Yang X, Li H. Silencing HMGB1 secretion inhibited EV71-induced blood-brain barrier dysfunction and neural inflammation by depressing astrocyte activation via sHh signal blockage. Int J Biochem Cell Biol 2025; 185:106797. [PMID: 40349912 DOI: 10.1016/j.biocel.2025.106797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 05/06/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025]
Abstract
It is unclear whether high mobility group protein B1 (HMGB1) is associated with the malignant characterization of hand, foot, and mouth disease (HFMD), and whether it plays a key regulatory role in the process of enterovirus 71 (EV71)-induced brain damage. Firstly, we analyzed the correlation between clinical information and HMGB1 concentrations in patients with mild and severe HFMD. Immunofluorescence was used to determine the expression level of HMGB1 in astrocytes. The levels of cellular inflammatory factors (IL-1β, IL-4, IL-6, TNF-α and TGF-β1), chemokines (CCL2, CXCL10 and CXCL12) and adhesion factors (integrin β, P-gp, VCAM-1 and ICAM-1) were detected by ELISA kits. Western blot was used to measure the levels of blood-brain barrier (BBB) stability related factors (retinoic acid (RA), ANG1, ApoE and IGF-1) in astrocytes and BBB structure related proteins (occluding, claudin, PTCH-1 and ZO-1) in endothelial cells. Clinical studies found that the expression of HMGB1 was closely related to the HFMD severity. Knockdown of HMGB1 alleviated EV71-induced neuron damage and inhibited cellular inflammation and apoptosis. Importantly, silencing HMGB1 depressed excessive proliferation and the inflammation response of astrocytes caused by EV71 infection. Furthermore, knockdown of HMGB1 enhanced BBB stability by improving astrocyte adhesion and endothelial tight junctions. Mechanistically, HMGB1 regulated the stability of BBB by regulating sHh signaling and secretion in astrocytes. In conclusion, the level of HMGB1 is closely related to the clinical symptoms of patients with HFMD, and inhibiting the expression of HMGB1 promotes BBB stability by promoting sHh signaling in astrocytes.
Collapse
Affiliation(s)
- Yufeng Zhang
- Department of Neonatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Department of Infectious Diseases, Xi'an Children's Hospital, Xi'an, Shaanxi 710003, China
| | - Yanfang Liu
- Department of Ophthalmology, Xi'an Children's Hospital, Xi'an, Shaanxi 710003, China
| | - Han Wan
- Department of General Surgery, Xi'an No.3 Hospital, Xi'an, Shaanxi 710021, China
| | - Huiling Deng
- Department of Infectious Diseases, Xi'an Children's Hospital, Xi'an, Shaanxi 710003, China
| | - Pengfei Xu
- Department of Infectious Diseases, Xi'an Children's Hospital, Xi'an, Shaanxi 710003, China
| | - Lu Cao
- Department of Infectious Diseases, Xi'an Children's Hospital, Xi'an, Shaanxi 710003, China
| | - Xiru Yang
- Department of Infectious Diseases, Xi'an Children's Hospital, Xi'an, Shaanxi 710003, China
| | - Hui Li
- Department of Neonatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Department of Neonatology, Xi'an Children's Hospital, Xi'an, Shaanxi 710003, China.
| |
Collapse
|
8
|
Zhang J, Mou D, Zhu L, Zhou J, Yu Q, Yang G, Luo C, Meng J, Mao K, Liu J, Yan B, Yang X. Curcumin Inhibits Lipopolysaccharide-Induced Inflammation Through the HMGB1/NF-κB Signaling Pathway to Promote the Osteogenic Differentiation of Bone Marrow Mesenchymal Stem Cells. Mol Biotechnol 2025:10.1007/s12033-025-01437-1. [PMID: 40310592 DOI: 10.1007/s12033-025-01437-1] [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: 02/22/2025] [Accepted: 03/20/2025] [Indexed: 05/02/2025]
Abstract
Curcumin has strong anti-inflammatory properties and promotes the osteogenic differentiation of human bone marrow mesenchymal stem cells (hBMSCs). The aim of this study was to explore the role and potential molecular mechanism of curcumin in ameliorating osteogenic differentiation disorders caused by inflammation. We used LPS to induce an inflammatory response in hBMSCs. The expression of related mRNAs and proteins was detected by RT‒qPCR, Western blotting, immunofluorescence, and ELISA. The osteogenic differentiation of hBMSCs was detected by alkaline phosphatase (ALP) staining and alizarin red S (ARS) staining. The results showed that after LPS treatment, the levels of the inflammatory cytokines TNF-α, IL-6 and IL-1β in hBMSCs increased, and the activity of ALP, the level of calcium salt deposition and the expression levels of the osteogenic proteins Runx2, COL1, OCN and OPN significantly decreased. The curcumin treatment alleviated this effect. These results indicated that curcumin improved the LPS-induced inflammation and osteogenic differentiation disorder in hBMSCs. Further studies revealed that the therapeutic effect of curcumin was caused by the inhibition of HMGB1 expression. From a mechanistic perspective, curcumin inhibits LPS-induced inflammation by inhibiting the expression of HMGB1, thereby inhibiting the NF-κB pathway and activating the NRF2 pathway, thereby improving the disordered osteogenic differentiation of hBMSCs. In conclusion, curcumin can reduce the LPS-induced inflammation of hBMSCs and ameliorate their osteogenic differentiation disorders.
Collapse
Affiliation(s)
- Jimei Zhang
- Gastroenterology Department, Chenggong Hospital, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, 650505, Yunnan, China
| | - Donggang Mou
- Pain Department, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, 650055, Yunnan, China
| | - Ling Zhu
- Orthopedics Department, Chenggong Hospital, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, 650505, Yunnan, China
| | - Jianping Zhou
- Orthopedics Department, Chenggong Hospital, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, 650505, Yunnan, China
| | - Qunying Yu
- Obstetrics Department, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650000, Yunnan, China
| | - Guangyuan Yang
- Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Chaoli Luo
- Operating Room, Yunnan Pain Disease Hospital, Kunming, 650000, Yunnan, China
| | - Jianguo Meng
- Orthopedics Department, Guang Nan Hospital of Traditional Chinese Medcine, Guangnan, 663300, Yunnan, China
| | - Kewang Mao
- Orthopedics Department, Hua Ning Country Hospital, Kunming, 652800, Yunnan, China
| | - Jing Liu
- Orthopedics Department, Chenggong Hospital, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, 650505, Yunnan, China
| | - Bo Yan
- Orthopedics Department, Kunming City Resort District DaYu Street Community Health Service Center, Kunming, 650500, Yunnan, China
| | - Xuming Yang
- Orthopedics Department, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, 650055, Yunnan, China.
| |
Collapse
|
9
|
Tsukiyama N, Tanaka Y, Yamane H, Tanimine N, Kuroda S, Tahara H, Ohira M, Ide K, Kobayashi T, Ohdan H. Impacts of high mobility group box protein 1 gene polymorphisms on morbidity and mortality after living donor liver transplantation. Transpl Immunol 2025; 90:102225. [PMID: 40157616 DOI: 10.1016/j.trim.2025.102225] [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: 11/18/2024] [Revised: 03/25/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
We investigated the effect of single-nucleotide polymorphisms (SNPs) in the high mobility group box 1 (HMGB1) gene on morbidity and mortality after liver transplantation (LT). Among 120 LT recipients and their living donors, the genotypes of HMGB1, and the SNPs rs2249825, rs1045411, rs1412125, and rs1360485 were determined. There were no significant associations between these four SNPs and the incidence of rejection or mortality. However, the incidence of early allograft dysfunction (EAD) (n = 43), which presents as functional insufficiency within 1 week of LT, was significantly higher in recipients with the GC + CC allele of rs2249825 (n = 17/34) than in those with the GG allele (n = 26/86) (p = 0.044). Although the impact of donor HMGB1 SNPs on the incidence of EAD was not statistically significant, recipients with the GC + CC allele of rs2249825 who received liver grafts from donors with the same genotype had the highest incidence of EAD (p = 0.052). In contrast, the donor TC + CC allele of rs1412125 was an independent risk factor for the development of sepsis (n = 33) in LT recipient (OR = 3.05, 95 % CI = 1.18-7.87, p = 0.021). Thus, the SNPs of the HMGB1 gene in either recipients or donors were not associated with mortality but influenced the incidence of EAD and sepsis, likely being a predictive biomarker for the risk of serious complications after LT.
Collapse
Affiliation(s)
- Naofumi Tsukiyama
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Yuka Tanaka
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan.
| | - Hiroaki Yamane
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Naoki Tanimine
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Shintaro Kuroda
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Hiroyuki Tahara
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Masahiro Ohira
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Kentaro Ide
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Tsuyoshi Kobayashi
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Hideki Ohdan
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan.
| |
Collapse
|
10
|
Wang YT, Moura AK, Zuo R, Roudbari K, Hu JZ, Khan SA, Wang Z, Shentu Y, Wang M, Li PL, Hao J, Zhang Y, Li X. Cardiovascular dysfunction and altered lysosomal signaling in a murine model of acid sphingomyelinase deficiency. J Mol Med (Berl) 2025; 103:599-617. [PMID: 40232391 DOI: 10.1007/s00109-025-02542-z] [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: 09/26/2024] [Revised: 03/18/2025] [Accepted: 04/02/2025] [Indexed: 04/16/2025]
Abstract
Niemann-Pick Disease (NPD) is a rare autosomal recessive lysosomal storage disorder (LSD) caused by the deficiency of acid sphingomyelinase (ASMD), which is encoded by the Smpd1 gene. ASMD impacts multiple organ systems in the body, including the cardiovascular system. This study is the first to characterize cardiac pathological changes in ASMD mice under baseline conditions, offering novel insights into the cardiac implications of NPD. Using histological analysis, biochemical assays, and echocardiography, we assessed cardiac pathological changes and function in Smpd1-/- mice compared to Smpd1+/+ littermate controls. Immunofluorescence and biochemical assays demonstrated that ASMD induced lysosomal dysfunction, as evidenced by the accumulation of lysosomal-associated membrane proteins, lysosomal protease, and autophagosomes in pericytes and cardiomyocytes. This lysosomal dysfunction was accompanied by pericytes and cardiomyocytes inflammation, characterized by increased expression of caspase1 and inflammatory cytokines, and infiltration of inflammatory cells in the cardiac tissues of Smpd1-/- mice. In addition, histological analysis revealed increased lipid deposition and cardiac steatosis, along with pericyte-to-myofibroblast transition (PMT) and interstitial fibrosis in Smpd1-/- mice. Moreover, echocardiography further demonstrated that Smpd1-/- mice developed coronary microvascular dysfunction (CMD), as evidenced by decreased coronary blood flow velocity and increased coronary arteriolar wall thickness. Additionally, these mice exhibited significant impairments in systolic and diastolic cardiac function, as shown by a reduced ejection fraction and prolonged left ventricular relaxation time constant (Tau value). These findings suggest that ASMD induces profound pathological changes and vascular dysfunction in the myocardium, potentially driven by mechanisms involving lysosomal dysfunction as well as both pericytes and cardiac inflammation. KEY MESSAGES: Lysosomal dysfunction in ASMD leads to impaired autophagic flux in cardiac pericytes ASMD causes cardiac inflammation with leukocyte and M2 macrophage infiltration Lipid buildup in the pericytes, fibroblasts and myocardium lead to cardiac steatosis Enhanced cardiac fibrosis in ASMD links to pericyte-to-myofibroblast transition ASMD results in coronary microvascular and diastolic and systolic cardiac dysfunction.
Collapse
Affiliation(s)
- Yun-Ting Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204 - 5056, USA
| | - Alexandra K Moura
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204 - 5056, USA
| | - Rui Zuo
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204 - 5056, USA
| | - Kiana Roudbari
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204 - 5056, USA
| | - Jenny Z Hu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204 - 5056, USA
| | - Saher A Khan
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204 - 5056, USA
| | - Zhengchao Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204 - 5056, USA
- Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou, China
| | - Yangping Shentu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204 - 5056, USA
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Mi Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204 - 5056, USA
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Jiukuan Hao
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204 - 5056, USA
| | - Yang Zhang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204 - 5056, USA.
| | - Xiang Li
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204 - 5056, USA.
| |
Collapse
|
11
|
Dong Y, Li Y, Tang W, Chen Q, Kong C. Increased Trophoblast Cell Ferroptosis via HMGB1/ACSL4 Pathway Is Associated with Spontaneous Abortion. Reprod Sci 2025; 32:1713-1722. [PMID: 39994144 PMCID: PMC12041038 DOI: 10.1007/s43032-025-01817-7] [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: 07/27/2024] [Accepted: 02/07/2025] [Indexed: 02/26/2025]
Abstract
INTRODUCTION Trophoblast cells undergo ferroptosis in pregnancy-related diseases. HMGB1 participates in pathological ferroptosis. However, whether lipopolysaccharide (LPS) -mediated HMGB1 expression induces the ferroptosis of trophoblast cells and further spontaneous abortion (SA) remains unknown. METHODS HMGB1 and ACSL4 expression were measured in villous tissues from 20 women with SA and 20 women with elective abortion. Human HTR-8/SVneo cells were treated with LPS to establish an in vitro abortion model. The hallmarks of ferroptosis including MDA, GSH, Fe2+ and ROS were detected using indicated assay kits. RESULTS The levels of HMGB1 and ACSL4 in villous tissues from SA women were significantly higher than those in the normal control group. HMGB1 interacts with and stabilizes ACSL4 to promote the ferroptosis of trophoblast cells. Conversely, HMGB1 and/or ACSL4 inhibition attenuated LPS-induced trophoblast cells ferroptosis. CONCLUSIONS An HMGB1/ACSL4 axis is engaged in LPS-induced ferroptosis of trophoblast cells, and may be targeted to design treatments preventing SA.
Collapse
Affiliation(s)
- Yishan Dong
- Department of Gynecology and Obstetrics, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China
- Department of Gynecology and Obstetrics, Changzhou Maternity and Child Health Care Hospital, Changzhou, China
| | - Yong Li
- Department of Gynecology and Obstetrics, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| | - Wenjie Tang
- Department of Gynecology and Obstetrics, Changzhou Maternity and Child Health Care Hospital, Changzhou, China
| | - Qin Chen
- Department of Gynecology and Obstetrics, Maoming Maternity and Child Health Care Hospital, Maoming, China
| | - Chengcai Kong
- Department of Gynecology and Obstetrics, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China.
- Department of Gynecology and Obstetrics, Changzhou Maternity and Child Health Care Hospital, Changzhou, China.
| |
Collapse
|
12
|
Mahi Imam Mollah M. Knockdown of Dorsal switch protein 1 Effect on Growth, Development, and survivability of Tenebrio molitor (Coleoptera: Tenebrionidae). J Invertebr Pathol 2025; 211:108338. [PMID: 40228737 DOI: 10.1016/j.jip.2025.108338] [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: 07/09/2024] [Revised: 04/09/2025] [Accepted: 04/11/2025] [Indexed: 04/16/2025]
Abstract
Dorsal switch protein 1 (DSP1) of insects and high mobility group box 1 (HMGB1) protein of vertebrates are homologous. Both HMGB1 and DSP1 act as damage-associated molecular pattern (DAMP) molecules. Previous studies reported that DSP1 plays a DAMP role in mealworm, Tenebrio molitor, by activating immune genes like antimicrobial peptides (AMPs), phospholipase A2 (PLA2), phenoloxidase (PO) and regulating nodulation. However, RNAi of Tm-DSP1 suppresses these genes and their immune responses. While the immune role of DSP1 is established, its function in growth, development and survivability of T. molitor larvae is unknown. This study was designed to assess the role of DSP1 in the above-mentioned aspects. Tm-DSP1 gene was expressed in all the developmental stages and tissues of T. molitor larvae. The highest expression was observed in L6 larval stage and fat body tissue. Tm-DSP1 detection by western blotting supports its expression in T. molitor samples. Following bacterial challenge, DSP1 expression was upregulated in immune-related tissues, including hemocyte, fat body, and midgut. Detection of increased DSP1 in bacteria-challenged plasma ensures the release of DSP1 in plasma from the cell nucleus of immune-challenged larvae. Additionally, RNAi knockdown of Tm-DSP1 led to reduced larval growth (body length) and development (body weight) of T. molitor larvae resulting in increased mortality of the larvae. These findings suggest that DSP1 regulates the growth, development, and survival of T. molitor. However, whether DSP1 directly plays a role in these physiological processes or whether it interrupts any other genes to achieve these effects is still unknown. Further study is required to clarify this issue.
Collapse
Affiliation(s)
- Md Mahi Imam Mollah
- Department of Entomology, Faculty of Agriculture, Patuakhali Science and Technology University, Dumki, Patuakhali 8602, Bangladesh; Department of Plant Medicals, College of Life Science, Andong National University, Andong 1275, Republic of Korea.
| |
Collapse
|
13
|
Liu J, Zhao T, Cui H, Tian Y, Miao X, Xing L, Wang X, Huang J, Liu Q, Zhang W, Shi K, Liu Y, Jia B, Kang L, Tian Y, Yuan W, He S, Feng X, Liu S. HMGB1 Encapsulated in Podocyte-Derived Exosomes Plays a Central Role in Glomerular Endothelial Cell Injury in Lupus Nephritis by Regulating TRIM27 Expression. J Transl Med 2025; 105:104096. [PMID: 39848602 DOI: 10.1016/j.labinv.2025.104096] [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: 05/30/2024] [Revised: 12/16/2024] [Accepted: 01/09/2025] [Indexed: 01/25/2025] Open
Abstract
Exosomes play a role in cell communication by transporting content between cells. Here, we tested whether renal podocyte-derived exosomes affect the injury of glomerular endothelial cells in lupus nephritis (LN). We found that exosomes containing high levels of high mobility group protein B1 (HMGB1) were released from podocytes in patients with LN, BALB/c mice injected with pristane (which induces lupus-like disease in mice), and cultured human renal glomerular endothelial cells (HRGECs) treated with LN plasma. In vitro, GW4869 (an inhibitor of exosome biogenesis/release) or exosome removal alleviated the injury of HRGECs induced by LN plasma. Additionally, leptomycin B or knockdown of HMGB1 in podocyte-derived exosomes reduced endothelial cell injury and the expression of tripartite motif-containing protein 27 (TRIM27). Knockdown or overexpression of TRIM27 attenuated or promoted the damage of HRGECs treated with LN plasma. In vivo, knockdown of HMGB1 in podocytes ameliorated the injury of glomerular endothelial cells in a mouse model of LN. Furthermore, the injection of podocyte-derived exosomes into mice caused glomerular endothelial cell dysfunction. In conclusion, our study revealed that podocyte-derived exosomes may mediate the injury of glomerular endothelial cells seen in LN.
Collapse
Affiliation(s)
- Jinxi Liu
- Department of Pathology, Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China; Key Laboratory of Kidney Diseases of Hebei Province, Hebei Medical University, Shijiazhuang, China
| | - Tongyu Zhao
- Department of Pathology, Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China; Key Laboratory of Kidney Diseases of Hebei Province, Hebei Medical University, Shijiazhuang, China; Department of Pathology, the First Hospital of Hebei Medical University; Shijiazhuang, China
| | - Huixin Cui
- Department of Pathology, Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China; Key Laboratory of Kidney Diseases of Hebei Province, Hebei Medical University, Shijiazhuang, China
| | - Yuexin Tian
- Department of Pathology, Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China; Key Laboratory of Kidney Diseases of Hebei Province, Hebei Medical University, Shijiazhuang, China
| | - Xinyan Miao
- Department of Pathology, Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China; Key Laboratory of Kidney Diseases of Hebei Province, Hebei Medical University, Shijiazhuang, China
| | - Lingling Xing
- Department of Nephrology, the Second Affiliated Hospital of Hebei Medical University; Shijiazhuang, China
| | - Xiaorong Wang
- Department of Nephrology, the Second Affiliated Hospital of Hebei Medical University; Shijiazhuang, China
| | - Jie Huang
- Department of Pathology, Shijiazhuang Obstetrics and Gynecology Hospital, China
| | - Qingjuan Liu
- Department of Pathology, Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China; Key Laboratory of Kidney Diseases of Hebei Province, Hebei Medical University, Shijiazhuang, China
| | - Wei Zhang
- Department of Pathology, Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China; Key Laboratory of Kidney Diseases of Hebei Province, Hebei Medical University, Shijiazhuang, China
| | - Ke Shi
- Department of Pathology, Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China; Key Laboratory of Kidney Diseases of Hebei Province, Hebei Medical University, Shijiazhuang, China; Department of Oncology, the Fourth Hospital of Hebei Medical University; Shijiazhuang, China
| | - Yunhe Liu
- Department of Pathology, Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China; Key Laboratory of Kidney Diseases of Hebei Province, Hebei Medical University, Shijiazhuang, China
| | - Baiyun Jia
- Department of Pathology, Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China; Key Laboratory of Kidney Diseases of Hebei Province, Hebei Medical University, Shijiazhuang, China
| | - Lihua Kang
- Department of Pathology, Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China; Key Laboratory of Kidney Diseases of Hebei Province, Hebei Medical University, Shijiazhuang, China
| | - Yu Tian
- Department of Pathology, Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China; Department of Rheumatology, the Second Affiliated Hospital of Hebei Medical University; Shijiazhuang, China
| | - Weicheng Yuan
- Clinical Medicine, Hebei Medical University; Shijiazhuang, China
| | - Shiwei He
- Clinical Medicine, Hebei Medical University; Shijiazhuang, China
| | - Xiaojuan Feng
- Department of Pathology, Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China; Key Laboratory of Kidney Diseases of Hebei Province, Hebei Medical University, Shijiazhuang, China.
| | - Shuxia Liu
- Department of Pathology, Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China; Key Laboratory of Kidney Diseases of Hebei Province, Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
14
|
Oğlak SC, Aşır F, Yılmaz EZ, Bolluk G, Korak T, Ağaçayak E. The Immunohistochemical and Bioinformatics Analysis of the Placental Expressions of Vascular Cell Adhesion Protein 1 (VCAM-1) and High Mobility Group Box 1 (HMGB1) Proteins in Gestational Diabetic Mothers. Z Geburtshilfe Neonatol 2025; 229:90-98. [PMID: 39532125 DOI: 10.1055/a-2451-2223] [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: 11/16/2024]
Abstract
OBJECTIVE We aimed to examine both the expression levels of high mobility group box 1 (HMGB1) and vascular cell adhesion molecule-1 (VCAM-1) proteins in the placentas of pregnant women with gestational diabetes mellitus (GDM) and control groups by immunohistochemical (IHC) method. MATERIAL AND METHODS An experimental case-control study was conducted, including 40 pregnant women complicated with GDM and 40 healthy pregnant women. Placental tissues obtained following cesarean delivery were subjected to routine tissue monitoring. The placental sections were stained with VCAM-1 and HMGB1 immunostains and subjected to IHC examination under a light microscope. H-score (HS) was used to evaluate the results of IHC staining by semi-quantitative analysis. Pathway analysis in Cytoscape software identified GDM-associated proteins within HMGB1 and VCAM-1 interaction networks, followed by GO analysis to explore associated biological processes. RESULTS Placental HGMB1 expression was significantly increased in the GDM group compared to the control group (p<0.001). However, placental VCAM-1 expression was found to be statistically similar in GDM and control groups (p=0.584). The shared 19 proteins were identified between HMGB1 and GDM, and 13 between VCAM-1 and GDM, with notable GO biological process terms such as immune system activation for HMGB1 and interleukin-6 regulation for VCAM-1 associated with GDM. CONCLUSION We consider that GDM-related inflammation and oxidative stress may contribute to tissue damage and inflammation by increasing placental HMGB1 expression. The blockade of HMGB1 and its receptors might represent a promising therapeutic approach to control inflammation in GDM. Understanding the distinct roles of HMGB1 and VCAM-1 may provide valuable insights for the development of targeted therapies aimed at mitigating the inflammatory processes associated with GDM and improving maternal and fetal outcomes.
Collapse
Affiliation(s)
- Süleyman Cemil Oğlak
- Obstetrics and Gynecology, Diyarbakir Gazi Yasargil Training and Research Hospital, Diyarbakır, Turkey
| | - Fırat Aşır
- Histology and Embryology, Dicle University, Diyarbakir, Turkey
| | | | - Gökhan Bolluk
- Perinatology, TC Sağlık Bakanlığ Başakşehir Çam ve Sakura Şehir Hastanesi, Basaksehir, Turkey
| | - Tuğcan Korak
- Medical Biology, Kocaeli University, Kocaeli, Turkey
| | - Elif Ağaçayak
- Obstetrics and Gynecology, Dicle University, Diyarbakir, Turkey
| |
Collapse
|
15
|
Wang YT, Moura AK, Zuo R, Roudbari K, Hu JZ, Khan SA, Wang Z, Shentu Y, Wang M, Li PL, Hao J, Zhang Y, Li X. Cardiovascular Dysfunction and Altered Lysosomal Signaling in a Murine Model of Acid Sphingomyelinase Deficiency. RESEARCH SQUARE 2025:rs.3.rs-5154105. [PMID: 40166006 PMCID: PMC11957194 DOI: 10.21203/rs.3.rs-5154105/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Niemann-Pick Disease (NPD) is a rare autosomal recessive lysosomal storage disorder (LSD) caused by the deficiency of acid sphingomyelinase (ASMD), which is encoded by the Smpd1 gene. ASMD impacts multiple organ systems in the body, including the cardiovascular system. This study is the first to characterize cardiac pathological changes in ASMD mice under baseline conditions, offering novel insights into the cardiac implications of NPD. Using histological analysis, biochemical assays, and echocardiography, we assessed cardiac pathological changes and function in Smpd1 -/- mice compared to Smpd1 +/+ littermate controls. Immunofluorescence and biochemical assays demonstrated that ASMD induced lysosomal dysfunction, as evidenced by the accumulation of lysosomal-associated membrane proteins, lysosomal protease, and autophagosomes in pericytes and cardiomyocytes. This lysosomal dysfunction was accompanied by pericytes and cardiomyocytes inflammation, characterized by increased expression of caspase1 and inflammatory cytokines, and infiltration of inflammatory cells in the cardiac tissues of Smpd1 -/- mice. In addition, histological analysis revealed increased lipid deposition and cardiac steatosis, along with pericyte-to-myofibroblast transition (PMT) and interstitial fibrosis in Smpd1 -/- mice. Moreover, echocardiography further demonstrated that Smpd1 -/- mice developed coronary microvascular dysfunction (CMD), as evidenced by decreased coronary blood flow velocity and increased coronary arteriolar wall thickness. Additionally, these mice exhibited significant impairments in systolic and diastolic cardiac function, as shown by a reduced ejection fraction and prolonged left ventricular relaxation time constant (Tau value). These findings suggest that ASMD induces profound pathological changes and vascular dysfunction in the myocardium, potentially driven by mechanisms involving lysosomal dysfunction as well as both pericytes and cardiac inflammation.
Collapse
|
16
|
Miki K, Fujieda H, Ueno Y, Arakane T, Fujii Y. Investigation of Inflammatory Reduction During Extracorporeal Membrane Oxygenation Using a Novel Cytokine Adsorption Column: A Rat Model Study. J Clin Med 2025; 14:1686. [PMID: 40095706 PMCID: PMC11900994 DOI: 10.3390/jcm14051686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/23/2025] [Accepted: 02/27/2025] [Indexed: 03/19/2025] Open
Abstract
Background: Cardiopulmonary bypass (CPB) and extracorporeal membrane oxygenation (ECMO) are widely used. Previous methods to reduce inflammation have shown inconsistent results. We developed a cytokine adsorption column using polymethyl methacrylate (PMMA) and investigated its anti-inflammatory effects during ECMO. Materials and Methods: Male Sprague-Dawley rats were divided into three groups (seven rats in each group): SHAM, ECMO, and ECMO with PMMA (PMMA group). Experiments comprised 180 min of cannulation only in the SHAM group and 60 min of ECMO followed by 120 min of observation in the ECMO and PMMA groups. PMMA adsorption was conducted from 30 min after ECMO initiation to completion in the PMMA group. Blood parameters and cytokines were measured during experiments. Lung tissues were collected after the experiment for evaluation of tissue edema. Results: The PMMA group showed significantly lower levels of tumor necrosis factor alpha (TNF-α) and interleukin(IL)-6 compared to the ECMO group at 120 min after completing ECMO. However, there were no significant differences in IL-10 levels between the ECMO group and the PMMA group at the same time points. Lung edema incidence was significantly lower in the PMMA group. Conclusions: The PMMA column effectively suppressed systemic inflammatory reactions during ECMO.
Collapse
Affiliation(s)
- Kota Miki
- Medical Engineering Center, Shimane University Hospital, Enya-cho 89-1, Izumo-shi 693-8501, Shimane, Japan
- Graduate School, Niigata University of Health and Welfare, Shimamicho1398, Kitaku 950-3198, Niigata, Japan;
| | - Hiroaki Fujieda
- Toray Industries, Inc., Nihonbashi 2-1-1, Muromachi, Chuo-ku, Tokyo 103-8666, Japan; (H.F.); (Y.U.); (T.A.)
| | - Yoshiyuki Ueno
- Toray Industries, Inc., Nihonbashi 2-1-1, Muromachi, Chuo-ku, Tokyo 103-8666, Japan; (H.F.); (Y.U.); (T.A.)
| | - Toru Arakane
- Toray Industries, Inc., Nihonbashi 2-1-1, Muromachi, Chuo-ku, Tokyo 103-8666, Japan; (H.F.); (Y.U.); (T.A.)
| | - Yutaka Fujii
- Graduate School, Niigata University of Health and Welfare, Shimamicho1398, Kitaku 950-3198, Niigata, Japan;
- Department of Clinical Engineering and Medical Technology, Niigata University of Health and Welfare, Shimamicho1398, Kitaku 950-3198, Niigata, Japan
| |
Collapse
|
17
|
Miserocchi G. Physiopathology of High-Altitude Pulmonary Edema. High Alt Med Biol 2025; 26:1-12. [PMID: 39331568 DOI: 10.1089/ham.2024.0037] [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: 09/29/2024] Open
Abstract
Miserocchi, Giuseppe. Physiopathology of high-altitude pulmonary edema. High Alt Med Biol. 26:1-12, 2025.-The air-blood barrier is well designed to accomplish the matching of gas diffusion with blood flow. This function is achieved by maintaining its thickness at ∼0.5 µm, a feature implying to keep extravascular lung water to the minimum. Exposure to hypobaric hypoxia, especially when associated with exercise, is a condition potentially leading to the development of the so-called high-altitude pulmonary edema (HAPE). This article presents a view of the physiopathology of HAPE by merging available data in humans exposed to high altitude with data from animal experimental approaches. A model is also presented to characterize HAPE nonsusceptible versus susceptible individuals based on the efficiency of alveolar-capillary oxygen uptake and estimated morphology of the air-blood barrier.
Collapse
Affiliation(s)
- Giuseppe Miserocchi
- Department of Medicine and Surgery, School of Medicine, University of Milano Bicocca, Monza, Italy
| |
Collapse
|
18
|
Lu S, Li M, Cheng Z, Liang Y, Huang J, Huang J, Wang K, Yao D, Chen E, Wang P, Li Y, Huang L. HMGB1-mediated macrophage regulation of NF-κB activation and MMP3 upregulation in nucleus pulposus cells: A critical mechanism in the vicious cycle of intervertebral disc degeneration. Cell Signal 2025; 127:111628. [PMID: 39880103 DOI: 10.1016/j.cellsig.2025.111628] [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: 11/28/2024] [Revised: 01/09/2025] [Accepted: 01/24/2025] [Indexed: 01/31/2025]
Abstract
Intervertebral disc degeneration (IVDD) is a leading cause of low back pain, primarily driven by inflammatory processes within the disc, particularly involving the infiltration and activity of macrophages. High Mobility Group Box 1 (HMGB1) has been identified as a crucial mediator in this inflammatory cascade, yet its precise role in macrophage-induced disc degeneration remains unclear. In this study, we employed a combination of in vivo and in vitro models, including genetically engineered mice with macrophage-specific overexpression of HMGB1, a rat model of IVDD, and cultured macrophages and nucleus pulposus cells (NPCs), to elucidate the role of HMGB1 in IVDD. Our findings reveal that HMGB1 overexpression in macrophages significantly accelerates IVDD progression by enhancing NF-κB activation and upregulating MMP3 expression in NPCs. Furthermore, the administration of glycyrrhizin (GL), an HMGB1 inhibitor, effectively mitigated these effects, delaying IVDD progression. This study not only uncovers the critical mechanisms by which HMGB1 regulates the interactions between macrophages and NPCs in the inflammatory microenvironment but also provides a theoretical framework for targeting HMGB1 as a potential therapeutic strategy for IVDD. Thus, our findings suggest a promising novel approach for the treatment of this condition.
Collapse
Affiliation(s)
- Shixin Lu
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou 510120, PR China; Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, 3025 Shennan Middle Road, Shenzhen 518033, PR China
| | - Ming Li
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou 510120, PR China
| | - Ziying Cheng
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, No.555 Youyi East Road, Xi'an 710000, Shanxi, PR China
| | - Yuwei Liang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou 510120, PR China
| | - Junshen Huang
- Department of Spine Surgery, People's Hospital of Longhua, 38 Jinglong Jianshe Road, Longhua District, Shenzhen 518000, PR China
| | - Jiajun Huang
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, 3025 Shennan Middle Road, Shenzhen 518033, PR China
| | - Kun Wang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou 510120, PR China
| | - Dengbo Yao
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, No.37 Guoxue Lane, Wuhou District, Chengdu 610000, Sichuan, PR China
| | - Enming Chen
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou 510120, PR China
| | - Peng Wang
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, 3025 Shennan Middle Road, Shenzhen 518033, PR China
| | - Yuxi Li
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou 510120, PR China.
| | - Lin Huang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou 510120, PR China.
| |
Collapse
|
19
|
Borges Paes Lemes J, Panichkina A, Franco Malange K, Morado-Urbina CE, Dochnal SA, Jadhav S, Dolmat M, Pagliusi M, Navia-Pealez JM, Corr M, Miller YI, Yaksh TL. Chronic Pain Induced by Social Defeat Stress in Juvenile Mice Depends on TLR4. Cells 2025; 14:350. [PMID: 40072079 PMCID: PMC11898947 DOI: 10.3390/cells14050350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 02/07/2025] [Accepted: 02/18/2025] [Indexed: 03/15/2025] Open
Abstract
A significant portion of adolescents suffer from mental illnesses and persistent pain due to repeated stress. The components of the nervous system that link stress and pain in early life remain unclear. Prior studies in adult mice implicated the innate immune system, specifically Toll-like receptors (TLRs), as critical for inducing long-term anxiety and pain-like behaviors in social defeat stress (SDS) models. In this work, we investigated the pain and anxiety behavioral phenotypes of wild-type and TLR4-deficient juvenile mice subjected to repeated SDS and evaluated the engagement of TLR4 by measuring dimerization in the spinal cord, dorsal root ganglia, and prefrontal cortex. Male juvenile (4-week-old) mice (C57BL/6J or Tlr4-/-) underwent six social defeat sessions with adult aggressor (CD1) mice. In WT mice, SDS promotes chronic mechanical allodynia and thermal hyperalgesia assessed via von Frey testing and the Hargreaves test, respectively. In parallel, the stressed WT mice exhibited transient anxiety-like behavior and long-lasting locomotor activity reduction in the open-field test. Tlr4-/--stressed animals were resistant to the induction of pain-like behavior but had a remnant of anxious behavior, spending less time in the center of the arena. In WT SDS, there were concordant robust increases in TLR4 dimerization in dorsal root ganglia macrophages and spinal cord microglia, indicating TLR4 activation. These results suggest that the chronic pain phenotype and locomotor impairment induced by SDS in juvenile mice depends on TLR4 engagement evidenced by dimerization in immune cells of the dorsal root ganglia and spinal cord.
Collapse
Affiliation(s)
- Julia Borges Paes Lemes
- Department of Anesthesiology, University of California, San Diego, CA 92093, USA; (A.P.); (K.F.M.); (C.E.M.-U.); (S.A.D.); (S.J.); (T.L.Y.)
| | - Alisa Panichkina
- Department of Anesthesiology, University of California, San Diego, CA 92093, USA; (A.P.); (K.F.M.); (C.E.M.-U.); (S.A.D.); (S.J.); (T.L.Y.)
| | - Kaue Franco Malange
- Department of Anesthesiology, University of California, San Diego, CA 92093, USA; (A.P.); (K.F.M.); (C.E.M.-U.); (S.A.D.); (S.J.); (T.L.Y.)
| | - Carlos E. Morado-Urbina
- Department of Anesthesiology, University of California, San Diego, CA 92093, USA; (A.P.); (K.F.M.); (C.E.M.-U.); (S.A.D.); (S.J.); (T.L.Y.)
| | - Sara Anna Dochnal
- Department of Anesthesiology, University of California, San Diego, CA 92093, USA; (A.P.); (K.F.M.); (C.E.M.-U.); (S.A.D.); (S.J.); (T.L.Y.)
| | - Saee Jadhav
- Department of Anesthesiology, University of California, San Diego, CA 92093, USA; (A.P.); (K.F.M.); (C.E.M.-U.); (S.A.D.); (S.J.); (T.L.Y.)
| | - Maksim Dolmat
- Department of Chemical and Nano Engineering, University of California, San Diego, CA 92093, USA;
| | - Marco Pagliusi
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Sao Paulo 14049-900, Brazil;
| | - Juliana M. Navia-Pealez
- Department of Pharmacology and Physiology, Saint Louis University, Saint Louis, MO 63104, USA;
| | - Maripat Corr
- Department of Medicine, University of California, San Diego, CA 92093, USA; (M.C.); (Y.I.M.)
| | - Yury I. Miller
- Department of Medicine, University of California, San Diego, CA 92093, USA; (M.C.); (Y.I.M.)
| | - Tony L. Yaksh
- Department of Anesthesiology, University of California, San Diego, CA 92093, USA; (A.P.); (K.F.M.); (C.E.M.-U.); (S.A.D.); (S.J.); (T.L.Y.)
| |
Collapse
|
20
|
Irzan FI, Retinasamy T, Wen WR, Sheng ETM, Shaikh MF, Arulsamy A. The Role of HMGB1 in Infection-Related Cognitive Deficits. FRONT BIOSCI-LANDMRK 2025; 30:25544. [PMID: 40018921 DOI: 10.31083/fbl25544] [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: 07/04/2024] [Revised: 09/04/2024] [Accepted: 09/12/2024] [Indexed: 03/01/2025]
Abstract
Infectious diseases caused by fungi, viruses, or bacteria can have a profound impact on human cognition. This can be due to either direct spread to the central nervous system (CNS) or indirect neuroinflammation. Ultimately causing neuronal damage and even neurodegeneration. Deteriorations in cognition, such as poor encoding and attention deficits, have been reported secondary to infectious diseases. Preclinical studies have identified the underlying mechanisms of these infection-related cognitive effects, such as through blood-brain barrier (BBB) disruption and M1 microglial polarization. These mechanisms are spearheaded by inflammatory markers that are released/initiated by the pathogens over the course of the infection. Among them, the high mobility group box 1 (HMGB1) protein is a common biomarker implicated across several infection-related cognitive deficits. Understanding these effects and mechanisms is crucial for the development of strategies to prevent and treat infection-related cognitive impairment. This review will thus consolidate and elucidate the current knowledge on the potential role of HMGB1 as a therapeutic target for infection-related cognitive impairments. This review will not only advance scientific understanding but also have significant clinical and public health implications, especially considering recent global health challenges. Based on the selected articles, extracellular HMGB1, as opposed to intracellular HMGB1, acts as damage-associated molecular patterns (DAMPs) or alarmins when released in the peripheries secondary to inflammasome activation. Due to their low molecular weight, they then enter the CNS through routes such as retrograde transport along the afferent nerves, or simple diffusion across the impaired BBB. This results in further disruption of the brain microenvironment due to the dysregulation of other regulatory pathways. The outcome is structural neuronal changes and cognitive impairment. Given its key role in neuroinflammation, HMGB1 holds promise as both a biomarker for diagnostic detection and a potential therapeutic target candidate for preventing infection-related cognitive impairment.
Collapse
Affiliation(s)
- Fathima Ijaza Irzan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 47500 Petaling Jaya, Selangor, Malaysia
| | - Thaarvena Retinasamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 47500 Petaling Jaya, Selangor, Malaysia
| | - Wong Ruo Wen
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 47500 Petaling Jaya, Selangor, Malaysia
| | - Edward Ting Ming Sheng
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 47500 Petaling Jaya, Selangor, Malaysia
| | - Mohd Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 47500 Petaling Jaya, Selangor, Malaysia
- School of Dentistry and Medical Sciences, Charles Sturt University, Orange, NSW 2800, Australia
| | - Alina Arulsamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 47500 Petaling Jaya, Selangor, Malaysia
| |
Collapse
|
21
|
Alekseeva LA, Sen’kova AV, Sounbuli K, Savin IA, Zenkova MA, Mironova NL. Pulmozyme Ameliorates LPS-Induced Lung Fibrosis but Provokes Residual Inflammation by Modulating Cell-Free DNA Composition and Controlling Neutrophil Phenotype. Biomolecules 2025; 15:298. [PMID: 40001601 PMCID: PMC11853346 DOI: 10.3390/biom15020298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/28/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Pulmonary fibrosis, a chronic progressive lung disorder, can be the result of previous acute inflammation-associated lung injury and involves a wide variety of inflammatory cells, causing the deposition of extracellular matrix (ECM) components in the lungs. Such lung injury is often associated with excessive neutrophil function and the formation of DNA networks in the lungs, which are also some of the most important factors for fibrosis development. Acute lung injury with subsequent fibrosis was initiated in C57Bl/6 mice by a single intranasal (i.n.) administration of LPS. Starting from day 14, human recombinant DNase I in the form of Pulmozyme for topical administration was instilled i.n. twice a week at a dose of 50 U/mouse. Cell-free DNA (cfDNA), DNase activity, and cell content were analyzed in blood serum and bronchoalveolar lavage fluid (BALF). Inflammatory and fibrotic changes in lung tissue were evaluated by histological analysis. The gene expression profile in spleen-derived neutrophils was analyzed by RT-qPCR. We demonstrated that Pulmozyme significantly reduced connective tissue expansion in the lungs. However, despite the reliable antifibrotic effect, complete resolution of inflammation in the respiratory system of mice treated with Pulmozyme was not achieved, possibly due to enhanced granulocyte recruitment and changes in the nuclear/mitochondrial cfDNA balance in the BALF. Moreover, Pulmozyme introduction caused the enrichment of the spleen-derived neutrophil population by those with an unusual phenotype, combining pro-inflammatory and anti-inflammatory features, which can also maintain lung inflammation. Pulmozyme can be considered a promising drug for lung fibrosis management; however, the therapy may be accompanied by residual inflammation.
Collapse
Affiliation(s)
- Ludmila A. Alekseeva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences (SB RAS), Lavrentiev Ave., 8, Novosibirsk 630090, Russia; (L.A.A.); (A.V.S.); (K.S.); (I.A.S.); (M.A.Z.)
| | - Aleksandra V. Sen’kova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences (SB RAS), Lavrentiev Ave., 8, Novosibirsk 630090, Russia; (L.A.A.); (A.V.S.); (K.S.); (I.A.S.); (M.A.Z.)
| | - Khetam Sounbuli
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences (SB RAS), Lavrentiev Ave., 8, Novosibirsk 630090, Russia; (L.A.A.); (A.V.S.); (K.S.); (I.A.S.); (M.A.Z.)
- Faculty of Natural Sciences, Novosibirsk State University, Pirogova St., 1, Novosibirsk 630090, Russia
| | - Innokenty A. Savin
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences (SB RAS), Lavrentiev Ave., 8, Novosibirsk 630090, Russia; (L.A.A.); (A.V.S.); (K.S.); (I.A.S.); (M.A.Z.)
| | - Marina A. Zenkova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences (SB RAS), Lavrentiev Ave., 8, Novosibirsk 630090, Russia; (L.A.A.); (A.V.S.); (K.S.); (I.A.S.); (M.A.Z.)
| | - Nadezhda L. Mironova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences (SB RAS), Lavrentiev Ave., 8, Novosibirsk 630090, Russia; (L.A.A.); (A.V.S.); (K.S.); (I.A.S.); (M.A.Z.)
| |
Collapse
|
22
|
Rykova EY, Klimontov VV, Shmakova E, Korbut AI, Merkulova TI, Kzhyshkowska J. Anti-Inflammatory Effects of SGLT2 Inhibitors: Focus on Macrophages. Int J Mol Sci 2025; 26:1670. [PMID: 40004134 PMCID: PMC11854991 DOI: 10.3390/ijms26041670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/04/2025] [Accepted: 02/06/2025] [Indexed: 02/21/2025] Open
Abstract
A growing body of evidence indicates that nonglycemic effects of sodium-glucose cotransporter 2 (SGLT2) inhibitors play an important role in the protective effects of these drugs in diabetes, chronic kidney disease, and heart failure. In recent years, the anti-inflammatory potential of SGLT2 inhibitors has been actively studied. This review summarizes results of clinical and experimental studies on the anti-inflammatory activity of SGLT2 inhibitors, with a special focus on their effects on macrophages, key drivers of metabolic inflammation. In patients with type 2 diabetes, therapy with SGLT2 inhibitors reduces levels of inflammatory mediators. In diabetic and non-diabetic animal models, SGLT2 inhibitors control low-grade inflammation by suppressing inflammatory activation of tissue macrophages, recruitment of monocytes from the bloodstream, and macrophage polarization towards the M1 phenotype. The molecular mechanisms of the effects of SGLT2 inhibitors on macrophages include an attenuation of inflammasome activity and inhibition of the TLR4/NF-κB pathway, as well as modulation of other signaling pathways (AMPK, PI3K/Akt, ERK 1/2-MAPK, and JAKs/STAT). The review discusses the state-of-the-art concepts and prospects of further investigations that are needed to obtain a deeper insight into the mechanisms underlying the effects of SGLT2 inhibitors on the molecular, cellular, and physiological levels.
Collapse
Affiliation(s)
- Elena Y. Rykova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (IC&G SB RAS), Lavrentjev Prospect 10, 630090 Novosibirsk, Russia; (E.Y.R.); (V.V.K.); (E.S.); (A.I.K.); (T.I.M.)
| | - Vadim V. Klimontov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (IC&G SB RAS), Lavrentjev Prospect 10, 630090 Novosibirsk, Russia; (E.Y.R.); (V.V.K.); (E.S.); (A.I.K.); (T.I.M.)
- Research Institute of Clinical and Experimental Lymphology, Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (RICEL—Branch of IC&G SB RAS), Timakov Str. 2, 630060 Novosibirsk, Russia
| | - Elena Shmakova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (IC&G SB RAS), Lavrentjev Prospect 10, 630090 Novosibirsk, Russia; (E.Y.R.); (V.V.K.); (E.S.); (A.I.K.); (T.I.M.)
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 634050 Tomsk, Russia
| | - Anton I. Korbut
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (IC&G SB RAS), Lavrentjev Prospect 10, 630090 Novosibirsk, Russia; (E.Y.R.); (V.V.K.); (E.S.); (A.I.K.); (T.I.M.)
- Research Institute of Clinical and Experimental Lymphology, Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (RICEL—Branch of IC&G SB RAS), Timakov Str. 2, 630060 Novosibirsk, Russia
| | - Tatyana I. Merkulova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (IC&G SB RAS), Lavrentjev Prospect 10, 630090 Novosibirsk, Russia; (E.Y.R.); (V.V.K.); (E.S.); (A.I.K.); (T.I.M.)
| | - Julia Kzhyshkowska
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (IC&G SB RAS), Lavrentjev Prospect 10, 630090 Novosibirsk, Russia; (E.Y.R.); (V.V.K.); (E.S.); (A.I.K.); (T.I.M.)
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 634050 Tomsk, Russia
- Institute of Transfusion Medicine and Immunology, Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| |
Collapse
|
23
|
Hur CJE, Steinberg BE. Targeting NINJ1-mediated cell rupture to treat inflammatory diseases. Mol Med 2025; 31:60. [PMID: 39953391 PMCID: PMC11829424 DOI: 10.1186/s10020-025-01113-9] [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: 10/14/2024] [Accepted: 02/03/2025] [Indexed: 02/17/2025] Open
Abstract
Cell death can terminate in plasma membrane rupture to release potent pro-inflammatory intracellular contents thereby contributing to inflammatory diseases. Cell rupture is an active process, mediated by the membrane protein ninjurin-1 (NINJ1) in pyroptosis, post-apoptosis lysis, ferroptosis, and forms of necrosis. Once activated, NINJ1 clusters into large oligomers within the membrane to initiate cellular lysis. Recent preclinical studies have demonstrated that inhibiting NINJ1 is a new strategy for treating immune-mediated diseases. Indeed, both small molecule inhibitors and neutralizing antibodies can target NINJ1 clustering to preserve plasma membrane integrity and mitigate disease pathogenesis. In this Perspective, we provide a summary of the current state of knowledge and recent developments in targeting cellular integrity during cell death through NINJ1 inhibition to treat inflammatory disease, with a focus on liver injury. As these NINJ1-mediated cell death pathways are pivotal in maintaining health and contribute to disease pathogenesis when dysregulated, the studies discussed within have broad implications across the immunologic basis of molecular medicine.
Collapse
Affiliation(s)
- Claire Ju-Eun Hur
- Program in Neuroscience and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Benjamin Ethan Steinberg
- Program in Neuroscience and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada.
- Department of Physiology, University of Toronto, Toronto, ON, Canada.
- Department of Anesthesia and Pain Medicine, The Hospital for Sick Children, Toronto, ON, Canada.
| |
Collapse
|
24
|
Su E, Song X, Wei L, Xue J, Cheng X, Xie S, Jiang H, Liu M. Endothelial GSDMD underlies LPS-induced systemic vascular injury and lethality. JCI Insight 2025; 10:e182398. [PMID: 39927458 PMCID: PMC11948583 DOI: 10.1172/jci.insight.182398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 12/17/2024] [Indexed: 02/11/2025] Open
Abstract
Endothelial injury destroys endothelial barrier integrity, triggering organ dysfunction and ultimately resulting in sepsis-related death. Considerable attention has been focused on identifying effective targets for inhibiting damage to endothelial cells to treat endotoxemia-induced septic shock. Global gasdermin D (Gsdmd) deletion reportedly prevents death caused by endotoxemia. However, the role of endothelial GSDMD in endothelial injury and lethality in lipopolysaccharide-induced (LPS-induced) endotoxemia and the underlying regulatory mechanisms are unknown. Here, we show that LPS increases endothelial GSDMD level in aortas and lung microvessels. We demonstrated that endothelial Gsdmd deficiency, but not myeloid cell Gsdmd deletion, protects against endothelial injury and death in mice with endotoxemia or sepsis. In vivo experiments suggested that hepatocyte GSDMD mediated the release of high-mobility group box 1, which subsequently binds to the receptor for advanced glycation end products in endothelial cells to cause systemic vascular injury, ultimately resulting in acute lung injury and lethality in shock driven by endotoxemia or sepsis. Additionally, inhibiting endothelial GSDMD activation via a polypeptide inhibitor alleviated endothelial damage and improved survival in a mouse model of endotoxemia or sepsis. These data suggest that endothelial GSDMD is a viable pharmaceutical target for treating endotoxemia and endotoxemia-induced sepsis.
Collapse
Affiliation(s)
- Enyong Su
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, NHC Key Laboratory of Ischemic Heart Diseases, Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, National Clinical Research Center for Interventional Medicine, Shanghai, China
- Shanghai Engineering Research Center of AI Technology for Cardiopulmonary Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoyue Song
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, NHC Key Laboratory of Ischemic Heart Diseases, Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Lili Wei
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, NHC Key Laboratory of Ischemic Heart Diseases, Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, National Clinical Research Center for Interventional Medicine, Shanghai, China
- Shanghai Engineering Research Center of AI Technology for Cardiopulmonary Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Cardiology, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai, China
| | - Junqiang Xue
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, NHC Key Laboratory of Ischemic Heart Diseases, Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Xuelin Cheng
- Department of Health Management Center, Zhongshan Hospital, and
- Department of General Practice, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shiyao Xie
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hong Jiang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, NHC Key Laboratory of Ischemic Heart Diseases, Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, National Clinical Research Center for Interventional Medicine, Shanghai, China
- Shanghai Engineering Research Center of AI Technology for Cardiopulmonary Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
- Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Ming Liu
- Shanghai Engineering Research Center of AI Technology for Cardiopulmonary Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Health Management Center, Zhongshan Hospital, and
- Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| |
Collapse
|
25
|
Shirey KA, Joseph J, Coughlan L, Nijhuis H, Varley AW, Blanco JCG, Vogel SN. An adenoviral vector encoding an inflammation-inducible antagonist, HMGB1 Box A, as a novel therapeutic approach to inflammatory diseases. mBio 2025; 16:e0338724. [PMID: 39699172 PMCID: PMC11796352 DOI: 10.1128/mbio.03387-24] [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: 10/30/2024] [Accepted: 11/26/2024] [Indexed: 12/20/2024] Open
Abstract
Influenza, as well as other respiratory viruses, can trigger local and systemic inflammation resulting in an overall "cytokine storm" that produces serious outcomes such as acute lung injury (ALI) or acute respiratory distress syndrome (ARDS). We hypothesized that gene therapy platforms could be useful in these cases if the production of an anti-inflammatory protein reflects the intensity and duration of the inflammatory condition. The recombinant protein would be produced and released only in the presence of the inciting stimulus, avoiding immunosuppression or other unwanted side effects that may occur when treating infectious diseases with anti-inflammatory drugs. To test this hypothesis, we developed AdV.C3-Tat/HIV-Box A, an inflammation-inducible cassette that remains innocuous in the absence of inflammation but releases HMGB1 Box A, an antagonist of high mobility group box 1 (HMGB1), in response to inflammatory stimuli such as lipopolysaccharide (LPS) or influenza virus infection. We report here that this novel inflammation-inducible HMGB1 Box A construct in a non-replicative adenovirus (AdV) vector mitigates lung and systemic inflammation therapeutically in response to influenza infection. We anticipate that this strategy will apply to the treatment of multiple diseases in which HMGB1-mediated signaling is a central driver of inflammation.IMPORTANCEMany inflammatory diseases are mediated by the action of a host-derived protein, HMGB1, on Toll-like receptor 4 (TLR4) to elicit an inflammatory response. We have engineered a non-replicative AdV vector that produces HMGB1 Box A, an antagonist of HMGB1-induced inflammation, under the control of an endogenous complement component C3 (C3) promoter sequence, that is inducible by LPS and influenza in vitro and ex vivo in macrophages (Mϕ) and protects mice and cotton rats therapeutically against infection with mouse-adapted and human non-adapted influenza strains, respectively, in vivo. We anticipate that this novel strategy will apply to the treatment of multiple infectious and non-infectious diseases in which HMGB1-mediated TLR4 signaling is a central driver of inflammation.
Collapse
Affiliation(s)
- Kari Ann Shirey
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland, USA
| | - John Joseph
- Sigmovir Biosystems Inc., Rockville, Maryland, USA
| | - Lynda Coughlan
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland, USA
- Center for Vaccine Development and Global Health (CVD), University of Maryland, School of Medicine, Baltimore, Maryland, USA
| | - Haye Nijhuis
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland, USA
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| | | | | | - Stefanie N. Vogel
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
26
|
Jiang R, Yang X. Prognostic value of serum high-mobility group box 1 in neonates with neonatal encephalopathy. Pediatr Res 2025; 97:1079-1084. [PMID: 39009766 DOI: 10.1038/s41390-024-03408-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/21/2024] [Accepted: 06/29/2024] [Indexed: 07/17/2024]
Abstract
BACKGROUND This study aimed to investigate the diagnostic potential of serum high-mobility group box 1 (HMGB1) in neonatal encephalopathy (NE). METHODS A retrospective study was conducted, analyzing 216 neonates diagnosed with NE. The neonates were divided into two groups based on their outcomes at 28 days. Serum HMGB1 levels were compared between the two groups. ROC analysis was used to determine the predictive value of HMGB1. RESULTS At 28 days, 174 infants had a good prognosis, while 42 had a poor prognosis. Infants with a poor prognosis had higher serum HMGB1 concentrations within 24 h of birth. Multifactorial analysis revealed that extremely preterm birth, extremely low birth weight, an Apgar score of 0-3 at 5 min, premature rupture of membranes by the mother, moderate to severe NE, and serum HMGB1 > 6.14 ng/mL are independent risk factors for poor prognosis. HMGB1 has predictive value for short-term prognosis with an area under the curve of 0.79. Elevated HMGB1 levels in the acute phase of NE are associated with poor short-term neonatal outcomes. The decrease in HMGB1 concentrations over time correlates with a good prognosis; whereas an increase suggests a poor prognosis. CONCLUSION Early measurement of serum HMGB1 could aid in the prognostic assessment of neonates with NE. IMPACT STATEMENT Although serum HMGB1 has emerged as a potential predictor of neonatal outcomes in neonatal encephalopathy, the relationship of HMGB1 levels to neonatal encephalopathy severity remains unclear. The current results demonstrate that infants with a poor prognosis had higher serum HMGB1 concentrations within 24 h of birth. Importantly, elevated serum HMGB1 levels in the acute phase of neonatal encephalopathy are associated with poor short-term neonatal outcomes. Our findings reveal the clinical values of HMGB1 in the prediction of neonatal outcomes in NE patients.
Collapse
Affiliation(s)
- Rui Jiang
- Department of Neonatology, Cangzhou Central Hospital, No.16 Xinhua West Road, Cangzhou, 061000, Hebei, China.
| | - Xinxin Yang
- Department of Endocrinology, Cangzhou Central Hospital, No.16 Xinhua West Road, Cangzhou, 061000, Hebei, China
| |
Collapse
|
27
|
Krishna AA, Abhirami BL, Kumaran A. Pain in rheumatoid arthritis: Emerging role of high mobility group box 1 protein-HMGB1. Life Sci 2025; 362:123361. [PMID: 39761742 DOI: 10.1016/j.lfs.2024.123361] [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: 10/14/2024] [Revised: 12/20/2024] [Accepted: 12/31/2024] [Indexed: 01/11/2025]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease where pain, driven by both inflammatory and non-inflammatory processes, is a major concern for patients. This pain can persist even after joint inflammation subsides. High mobility group box-1 (HMGB1) is a non-histone-DNA binding protein located in the nucleus that plays a key role in processes such as DNA transcription, recombination, and replication. HMGB1 can be released into the extracellular space through both passive and active mechanisms. Extracellular HMGB1 contributes to synovial inflammation, bone degradation, and the production of cytokines in RA by binding to toll-like receptors (TLRs) and receptors for advanced glycation end products (RAGE). It also forms complexes with molecules like lipopolysaccharide (LPS) and IL-1β, amplifying inflammatory responses. Due to its central role in these processes, HMGB1 is considered a promising therapeutic target in RA. It also acts as a nociceptive molecule in mediating pain in diseases such as diabetes and bone cancer. In this review, we explore how HMGB1 contributes to chronic pain in RA, supported by both in vitro and in vivo models. We begin by providing an overview of the mechanisms of pain in RA, the structure of HMGB1, its release mechanisms, and the therapeutic potential of targeting HMGB1 in RA. Following this, we highlight its role in peripheral and central pain sensitization through direct activation of the TLR4/MAPK/NF-κB pathway, as well as indirectly through downstream mediators, underscoring its potential as a target for managing RA pain.
Collapse
Affiliation(s)
- Anithakumari Aswathy Krishna
- Agroprocessing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram 695019, Kerala, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Beena Levakumar Abhirami
- Agroprocessing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram 695019, Kerala, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Alaganandam Kumaran
- Agroprocessing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram 695019, Kerala, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
28
|
Herrera TES, Tello IPS, Mustafa MA, Jamil NY, Alaraj M, Atiyah Altameem KK, Alasheqi MQ, Hamoody AHM, Alkhafaji AT, Shakir MN, Alshahrani MY, Alawadi A. Kaempferol: Unveiling its anti-inflammatory properties for therapeutic innovation. Cytokine 2025; 186:156846. [PMID: 39754793 DOI: 10.1016/j.cyto.2024.156846] [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: 09/15/2024] [Revised: 11/24/2024] [Accepted: 12/23/2024] [Indexed: 01/06/2025]
Abstract
Inflammation, driven by various stimuli such as pathogens, cellular damage, or vascular injury, plays a central role in numerous acute and chronic conditions. Current treatments are being re-evaluated, prompting interest in naturally occurring compounds like kaempferol, a flavonoid prevalent in fruits and vegetables, for their anti-inflammatory properties. This study explores the therapeutic potential of kaempferol, focusing on its ability to modulate pro-inflammatory cytokines and its broader effects on inflammatory signaling pathways. Comprehensive reviews of in vitro and in vivo studies were conducted to elucidate the mechanisms underlying its anti-inflammatory and antioxidant actions. Kaempferol effectively inhibits the production of key inflammatory mediators, including cytokines and enzymes such as COX-2 and iNOS, while also targeting oxidative stress pathways like Nrf2 activation. The compound demonstrated protective effects in various inflammatory conditions, including sepsis, neurodegenerative disorders, cardiovascular diseases, and autoimmune conditions, by modulating pathways such as NF-κB, MAPK, and STAT. Despite its promise, kaempferol's clinical application faces challenges related to its bioavailability and stability, underscoring the need for advanced formulation strategies. These findings position kaempferol as a promising candidate for anti-inflammatory therapy, with the potential to improve patient outcomes across a wide range of inflammatory diseases. Further clinical studies are required to validate its efficacy, optimize dosage, and address pharmacokinetic limitations.
Collapse
Affiliation(s)
| | - Iván Patricio Salgado Tello
- Facultad de Ciencias Pecuarias, Escuela Superior Politécnica de Chimborazo (ESPOCH), Riobamba 060106, Ecuador
| | | | - Nawfal Yousif Jamil
- Department of Radiology & Sonar Techniques, Al-Noor University College, Nineveh, Iraq
| | - Mohd Alaraj
- School of Pharmacy-Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University, Gangoh, Uttar Pradesh-247341, India; Department of Pharmacy, Arka Jain University, Jamshedpur, Jharkhand- 831001, India
| | | | | | | | | | - Maha Noori Shakir
- Department of Medical Laboratories Technology, Al-Nisour University College/ Baghdad/, Iraq
| | - Mohammad Y Alshahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia.
| | - Ahmed Alawadi
- College of technical engineering, the Islamic University, Najaf, Iraq; College of technical engineering, The Islamic University of Al Diwaniyah, Diwaniya, Iraq; College of technical engineering, The Islamic University of Babylon, Hillah, Iraq
| |
Collapse
|
29
|
Qi S, Wu Q, Xiang P, Hou C, Kang Z, Chen M, Yi C, Bai X, Li T, Li Z, Xie W. HMGB1 in Septic Muscle Atrophy: Roles and Therapeutic Potential for Muscle Atrophy and Regeneration. J Cachexia Sarcopenia Muscle 2025; 16:e13711. [PMID: 39963819 PMCID: PMC11833301 DOI: 10.1002/jcsm.13711] [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: 08/01/2024] [Revised: 11/14/2024] [Accepted: 01/02/2025] [Indexed: 02/20/2025] Open
Abstract
Currently, the treatment of septic myopathy presents significant challenges with implications for increased mortality rates and prolonged hospitalizations. Effective therapeutic strategies for septic myopathy remain elusive, highlighting an urgent need for novel therapeutic approaches. High-mobility group box 1 (HMGB1) is a conserved nonhistone nuclear protein that is released passively from deceased cells or actively secreted by activated immune cells, influencing both infectious and noninfectious inflammatory responses. Studies have indicated that HMGB1 likely plays a pivotal role in the pathogenesis of septic myopathy by crucial pathways associated with muscle atrophy and contributing to muscle regeneration under certain conditions. This review aims to summarize the possible mechanisms of HMGB1 in muscle atrophy and its potential in muscle regeneration, providing a theoretical basis for HMGB1 treatment of septic myopathy. Research shows that the dual role of HMGB1 is related to its specific forms, which are influenced to varying degrees by environmental factors. HMGB1 is a key participant in septic muscle atrophy, whereas HMGB1 shows therapeutic potential in muscle regeneration. One key mechanism by which HMGB1 contributes to septic muscle atrophy is through the exacerbation of inflammation. HMGB1 can amplify the inflammatory response by promoting the release of pro-inflammatory cytokines, which further damages muscle tissue. HMGB1 is also involved in promoting cell death in sepsis, which contributes to muscle degradation. Another important mechanism is the regulation of protein degradation systems. HMGB1 can activate the ubiquitin-proteasome system and autophagy-lysosome pathway, both of which are crucial for the breakdown of muscle proteins during atrophy. Conversely, targeting HMGB1 has shown the potential to ameliorate muscle atrophy in various diseases. For instance, HMGB1 has been shown to promote muscle vascular regeneration, modify stem cell status and enhance stem cell migration and differentiation, all of which are beneficial for muscle repair and recovery. Pharmacological inhibition of HMGB1 has been explored, with several drugs demonstrating efficacy in reducing inflammation and muscle degradation in sepsis models. These findings suggest that HMGB1 inhibition could be a viable therapeutic approach for septic myopathy. However, the function of promoting muscle regeneration in septic myopathy needs further research. HMGB1 emerges as a promising therapeutic target for the treatment of muscle atrophy in sepsis. This review focuses on identifying the correlation between HMGB1 and septic myopathy, analysing the possible role of HMGB1 in disease development and examining the feasibility of HMGB1 as a therapeutic target.
Collapse
Affiliation(s)
- Si‐Yuan Qi
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Qiqi Wu
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Peng‐Hui Xiang
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Chao‐Yao Hou
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Zhaofeng Kang
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Meng‐Qi Chen
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Chengla Yi
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiangjun Bai
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Tianyu Li
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Zhanfei Li
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Wei‐Ming Xie
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Trauma CenterPeking University People's HospitalBeijingChina
- Key Laboratory of Trauma Treatment and Neural Regeneration (Peking University)Ministry of EducationBeijingChina
- National Center for Trauma Medicine of ChinaBeijingChina
| |
Collapse
|
30
|
Date S, Bhatt LK. Targeting high-mobility-group-box-1-mediated inflammation: a promising therapeutic approach for myocardial infarction. Inflammopharmacology 2025; 33:767-784. [PMID: 39487941 DOI: 10.1007/s10787-024-01586-w] [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: 08/30/2024] [Accepted: 10/11/2024] [Indexed: 11/04/2024]
Abstract
Myocardial ischemia, resulting from coronary artery blockage, precipitates cardiac arrhythmias, myocardial structural changes, and heart failure. The pathophysiology of MI is mainly based on inflammation and cell death, which are essential in aggravating myocardial ischemia and reperfusion injury. Emerging research highlights the functionality of high mobility group box-1, a non-histone nucleoprotein functioning as a chromosomal stabilizer and inflammatory mediator. HMGB1's release into the extracellular compartment during ischemia acts as damage-associated molecular pattern, triggering immune reaction by pattern recognition receptors and exacerbating tissue inflammation. Its involvement in signaling pathways like PI3K/Akt, TLR4/NF-κB, and RAGE/HMGB1 underscores its significance in promoting angiogenesis, apoptosis, and reducing inflammation, which is crucial for MI treatment strategies. This review highlights the complex function of HMGB1 in the pathogenesis of myocardial infarction by summarizing novel findings on the protein in ischemic situations. Understanding the mechanisms underlying HMGB1 could widen the way to specific treatments that minimize the severity of MI and enhance patient outcomes.
Collapse
Affiliation(s)
- Shrutika Date
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India.
| |
Collapse
|
31
|
Sasaki R, Luo Y, Kishi S, Ogata R, Nishiguchi Y, Sasaki T, Ohmori H, Fujiwara-Tani R, Kuniyasu H. Oxidative High Mobility Group Box-1 Accelerates Mitochondrial Transfer from Mesenchymal Stem Cells to Colorectal Cancer Cells Providing Cancer Cell Stemness. Int J Mol Sci 2025; 26:1192. [PMID: 39940960 PMCID: PMC11818411 DOI: 10.3390/ijms26031192] [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: 12/25/2024] [Revised: 01/25/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025] Open
Abstract
Mitochondria are important organelles for cell metabolism and tissue survival. Their cell-to-cell transfer is important for the fate of recipient cells. Recently, bone marrow mesenchymal stem cells (BM-MSCs) have been reported to provide mitochondria to cancer cells and rescue mitochondrial dysfunction in cancer cells. However, the details of the mechanism have not yet been fully elucidated. In this study, we investigated the humoral factors inducing mitochondrial transfer (MT) and the mechanisms. BM-MSCs produced MT in colorectal cancer (CRC) cells damaged by 5-fluorouracil (5-FU), but were suppressed by the anti-high mobility group box-1 (HMGB1) antibody. BM-MSCs treated with oxidized HMGB1 had increased expression of MT-associated genes, whereas reduced HMGB1 did not. Inhibition of nuclear factor-κB, a downstream factor of HMGB1 signaling, significantly decreased MT-associated gene expression. CRC cells showed increased stemness and decreased 5-FU sensitivity in correlation with MT levels. In a mouse subcutaneous tumor model of CRC, 5-FU sensitivity decreased and stemness increased by the MT from host mouse BM-MSCs. These results suggest that oxidized HMGB1 induces MTs from MSCs to CRC cells and promotes cancer cell stemness. Targeting of oxidized HMGB1 may attenuate stemness of CRCs.
Collapse
Grants
- 19K16564 Ministry of Education, Culture, Sports, Science and Technology
- 23K10481 Ministry of Education, Culture, Sports, Science and Technology
- 22K11396 Ministry of Education, Culture, Sports, Science and Technology
- 21K06926 Ministry of Education, Culture, Sports, Science and Technology
- 23K19900 Ministry of Education, Culture, Sports, Science and Technology
- 20K21659 Ministry of Education, Culture, Sports, Science and Technology
Collapse
Affiliation(s)
- Rika Sasaki
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan; (R.S.); (Y.L.); (S.K.); (R.O.); (Y.N.); (T.S.); (H.O.); (R.F.-T.)
| | - Yi Luo
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan; (R.S.); (Y.L.); (S.K.); (R.O.); (Y.N.); (T.S.); (H.O.); (R.F.-T.)
| | - Shingo Kishi
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan; (R.S.); (Y.L.); (S.K.); (R.O.); (Y.N.); (T.S.); (H.O.); (R.F.-T.)
- Pathology Laboratory, Research Institute, Tokushukai Nozaki Hospital, 2-10-50 Tanigawa, Daito 574-0074, Osaka, Japan
| | - Ruiko Ogata
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan; (R.S.); (Y.L.); (S.K.); (R.O.); (Y.N.); (T.S.); (H.O.); (R.F.-T.)
| | - Yukiko Nishiguchi
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan; (R.S.); (Y.L.); (S.K.); (R.O.); (Y.N.); (T.S.); (H.O.); (R.F.-T.)
| | - Takamitsu Sasaki
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan; (R.S.); (Y.L.); (S.K.); (R.O.); (Y.N.); (T.S.); (H.O.); (R.F.-T.)
| | - Hitoshi Ohmori
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan; (R.S.); (Y.L.); (S.K.); (R.O.); (Y.N.); (T.S.); (H.O.); (R.F.-T.)
| | - Rina Fujiwara-Tani
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan; (R.S.); (Y.L.); (S.K.); (R.O.); (Y.N.); (T.S.); (H.O.); (R.F.-T.)
| | - Hiroki Kuniyasu
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan; (R.S.); (Y.L.); (S.K.); (R.O.); (Y.N.); (T.S.); (H.O.); (R.F.-T.)
| |
Collapse
|
32
|
Yang Y, Hu Y, Yang Y, Liu Q, Zheng P, Yang Z, Duan B, He J, Li W, Li D, Zheng X, Wang M, Fu Y, Long Q, Ma Y. Tumor Vaccine Exploiting Membranes with Influenza Virus-Induced Immunogenic Cell Death to Decorate Polylactic Coglycolic Acid Nanoparticles. ACS NANO 2025; 19:3115-3134. [PMID: 39806805 DOI: 10.1021/acsnano.4c00654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Immunogenic cell death (ICD) of tumor cells, which is characterized by releasing immunostimulatory "find me" and "eat me" signals, expressing proinflammatory cytokines and providing personalized and broad-spectrum tumor antigens draws increasing attention in developing a tumor vaccine. In this study, we aimed to investigate whether the influenza virus (IAV) is efficient enough to induce ICD in tumor cells and an extra modification of IAV components such as hemeagglutinin (HA) will be helpful for the ICD-induced cells to elicit robust antitumor effects; in addition, to evaluate whether the membrane-engineering polylactic coglycolic acid nanoparticles (PLGA NPs) simulating ICD immune stimulation mechanisms hold the potential to be a promising vaccine candidate, a mouse melanoma cell line (B16-F10 cell) was infected with IAV rescued by the reverse genetic system, and the prepared cells and membrane-modified PLGA NPs were used separately to immunize the melanoma-bearing mice. IAV-infected tumor cells exhibit dying status, releasing high mobility group box-1 (HMGB1) and adenosine triphosphate (ATP), and exposing calreticulin (CRT), IAV hemeagglutinin (HA), and tumor antigens like tyrosinase-related protein 2 (TRP2). IAV-induced ICD cells enhance biomass-derived carbon (BMDCs) migration, antigen uptake, cross-presentation, and maturation in vitro. Furthermore, immunization with IAV-induced ICD cells effectively suppressed tumor growth in melanoma-bearing mice. The isolated cell membrane inherited the immunological characteristics from the ICD cells and elicited robust antitumor immune responses through decorating PLGA NPs loading with a tumor-specific helper T-cell peptide and supplemented with ATP in a hydrogel system. This study indicated a promising strategy for developing cell-based and personalized tumor vaccines through fully taking advantage of the immune stimulation mechanisms of ICD occurrence in tumor cells, IAV modification, and nanoscale delivery.
Collapse
Affiliation(s)
- Ying Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
| | - Yongmao Hu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
- Yunnan University, Kunming 650091, China
| | - Ying Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
- Kunming Medical University, Kunming 650500, China
| | - Qingwen Liu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
- Kunming Medical University, Kunming 650500, China
| | - Peng Zheng
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
| | - Zhongqian Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
| | - Biao Duan
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
- Kunming Medical University, Kunming 650500, China
| | - Jinrong He
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
| | - Weiran Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
| | - Duo Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
- Department of Acute Infectious Diseases Control and Prevention, Yunnan Provincial Center for Disease Control and Prevention, Kunming 650000, China
| | - Xiao Zheng
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
| | - Mengzhen Wang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
| | - Yuting Fu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
| | - Qiong Long
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
| | - Yanbing Ma
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
- State Key Laboratory of Respiratory Health and Multimorbidity, Kunming 650031, China
| |
Collapse
|
33
|
Matsushige T, Inoue H, Hoshide M, Kohno F, Kobayashi H, Ichihara K, Ichiyama T, Hasegawa S. Serial cerebrospinal fluid concentrations of high mobility group box 1 in bacterial meningitis: a retrospective cohort study. BMC Infect Dis 2025; 25:107. [PMID: 39849347 PMCID: PMC11756128 DOI: 10.1186/s12879-025-10476-7] [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: 09/16/2024] [Accepted: 01/09/2025] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND Bacterial meningitis (BM) is a life-threatening central nervous system infection with potential for severe neurological sequelae. High mobility group box 1 (HMGB1) is known as a late inflammatory mediator associated with lethal pathology. This study aims to investigate the serial cerebrospinal fluid (CSF) concentrations of HMGB1 in children with BM and its relationship to neurological prognosis. METHODS This retrospective cohort study included children with BM, aseptic meningitis (AM), and controls. CSF samples were collected serially from patients with BM and once from those with AM and controls. HMGB1 and interleukin-6 (IL-6) concentrations were measured using ELISA and bead-based multiplex assays, respectively. Statistical analyses included Mann-Whitney U tests, Kruskal-Wallis tests, and three-way ANOVA to evaluate differences among groups and over time. RESULTS HMGB1 levels in the CSF of children with BM were significantly higher than in those with AM and controls (p < 0.001). Inflammatory cytokine IL-6 levels decreased after treatment; however, HMGB1 levels remained elevated in half of the BM patients. Notably, a patient with neurological sequelae exhibited a delayed elevation of HMGB1 until the latest time points. Three-way ANOVA revealed significant differences in the time course of IL-6 and HMGB1 among individuals (p = 0.018). CONCLUSIONS Elevated CSF HMGB1 levels persist in some children with BM even after treatment, particularly in those with poor neurological outcomes. These findings suggest that delayed elevation of HMGB1 may contribute to severe inflammation and poor prognosis in BM. Further research into HMGB1 as a potential therapeutic target in BM is warranted.
Collapse
Affiliation(s)
- Takeshi Matsushige
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi, 755-8505, Japan.
| | - Hirofumi Inoue
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Madoka Hoshide
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Fumitaka Kohno
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Hikaru Kobayashi
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Kiyoshi Ichihara
- Department of Clinical Laboratory Sciences, Faculty of Health Sciences, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Takashi Ichiyama
- Division of Pediatrics, Tsudumigaura Medical Center for Children With Disabilities, 752-4 Kume, Shunan, Yamaguchi, 745-0801, Japan
| | - Shunji Hasegawa
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi, 755-8505, Japan
| |
Collapse
|
34
|
Li W, Chen Q, Peng C, Yang D, Liu S, Lv Y, Jiang L, Xu S, Huang L. Roles of the Receptor for Advanced Glycation End Products and Its Ligands in the Pathogenesis of Alzheimer's Disease. Int J Mol Sci 2025; 26:403. [PMID: 39796257 PMCID: PMC11721675 DOI: 10.3390/ijms26010403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/02/2025] [Accepted: 01/04/2025] [Indexed: 01/13/2025] Open
Abstract
The Receptor for Advanced Glycation End Products (RAGE), part of the immunoglobulin superfamily, plays a significant role in various essential functions under both normal and pathological conditions, especially in the progression of Alzheimer's disease (AD). RAGE engages with several damage-associated molecular patterns (DAMPs), including advanced glycation end products (AGEs), beta-amyloid peptide (Aβ), high mobility group box 1 (HMGB1), and S100 calcium-binding proteins. This interaction impairs the brain's ability to clear Aβ, resulting in increased Aβ accumulation, neuronal injury, and mitochondrial dysfunction. This further promotes inflammatory responses and oxidative stress, ultimately leading to a range of age-related diseases. Given RAGE's significant role in AD, inhibitors that target RAGE and its ligands hold promise as new strategies for treating AD, offering new possibilities for alleviating and treating this serious neurodegenerative disease. This article reviews the various pathogenic mechanisms of AD and summarizes the literature on the interaction between RAGE and its ligands in various AD-related pathological processes, with a particular focus on the evidence and mechanisms by which RAGE interactions with AGEs, HMGB1, Aβ, and S100 proteins induce cognitive impairment in AD. Furthermore, the article discusses the principles of action of RAGE inhibitors and inhibitors targeting RAGE-ligand interactions, along with relevant clinical trials.
Collapse
Affiliation(s)
- Wen Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (W.L.); (Q.C.); (C.P.); (D.Y.); (S.L.); (Y.L.); (L.J.)
| | - Qiuping Chen
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (W.L.); (Q.C.); (C.P.); (D.Y.); (S.L.); (Y.L.); (L.J.)
| | - Chengjie Peng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (W.L.); (Q.C.); (C.P.); (D.Y.); (S.L.); (Y.L.); (L.J.)
| | - Dan Yang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (W.L.); (Q.C.); (C.P.); (D.Y.); (S.L.); (Y.L.); (L.J.)
| | - Si Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (W.L.); (Q.C.); (C.P.); (D.Y.); (S.L.); (Y.L.); (L.J.)
| | - Yanwen Lv
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (W.L.); (Q.C.); (C.P.); (D.Y.); (S.L.); (Y.L.); (L.J.)
| | - Langqi Jiang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (W.L.); (Q.C.); (C.P.); (D.Y.); (S.L.); (Y.L.); (L.J.)
| | - Shijun Xu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (W.L.); (Q.C.); (C.P.); (D.Y.); (S.L.); (Y.L.); (L.J.)
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, China
- Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Lihua Huang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (W.L.); (Q.C.); (C.P.); (D.Y.); (S.L.); (Y.L.); (L.J.)
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, China
| |
Collapse
|
35
|
Sun K, Lin W, Hong Q, Chen S, Li J, Qiu S. Matrine: A Promising Treatment for Ulcerative Colitis by Targeting the HMGB1/NLRP3/Caspase-1 Pathway. Comb Chem High Throughput Screen 2025; 28:654-663. [PMID: 38347801 DOI: 10.2174/0113862073292384240209095838] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/23/2024] [Accepted: 01/26/2024] [Indexed: 04/12/2025]
Abstract
BACKGROUND Previous studies have found that matrine (MAT) effectively treated Ulcerative Colitis (UC). The purpose of this study is to explore its mechanism based on the HMGB1/NLRP3/Caspase-1 signaling pathway. METHODS MAT was administered intragastrically to DSS-induced UC mice for 14 days. The Disease Activity Index (DAI) and histological staining were measured to detect histopathological changes in colon. The levels of IL-1β, IL-6, and TNF-α in serum were measured by ELISA. The protein and mRNA expression of HMGB1/NLRP3/Caspase-1 in the colon were detected by immunohistochemistry, western Blotting or qRT-PCR. RESULTS MAT improved the histological pathological changes of UC mice, as assessed by DAI, colonic length, and colonic mucosal injury. MAT also reduced colonic inflammatory damage by reducing the serum IL-1β, IL-6, and TNF-α content and decreasing the expression of HMGB1, NLRP3, Caspase-1, and IL-1β and proteins and mRNA in the colon. CONCLUSION MAT could significantly alleviate DSS-induced UC symptoms by reducing the expressions of pro-inflammatory cytokines, such as IL-1β, TNF-α, and IL-6, the mechanism of which is related to the inhibition of HMGB1/NLRP3/Caspase-1 signaling pathway.
Collapse
Affiliation(s)
- Kexin Sun
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, 548 Binwen Rd, Hangzhou, 310053, P.R. China
| | - Weiye Lin
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, 548 Binwen Rd, Hangzhou, 310053, P.R. China
| | - Qianran Hong
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, 548 Binwen Rd, Hangzhou, 310053, P.R. China
| | - Shuangyu Chen
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, 548 Binwen Rd, Hangzhou, 310053, P.R. China
| | - Jiayang Li
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, 548 Binwen Rd, Hangzhou, 310053, P.R. China
| | - Shengliang Qiu
- Zhejiang Provincial Hospital of Chinese Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, 54 Youdian Rd, Hangzhou, 310006, P.R. China
| |
Collapse
|
36
|
Sandys O, Stokkers PCF, Te Velde AA. DAMP-ing IBD: Extinguish the Fire and Prevent Smoldering. Dig Dis Sci 2025; 70:49-73. [PMID: 38963463 PMCID: PMC11761125 DOI: 10.1007/s10620-024-08523-5] [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: 02/21/2024] [Accepted: 06/04/2024] [Indexed: 07/05/2024]
Abstract
In inflammatory bowel diseases (IBD), the most promising therapies targeting cytokines or immune cell trafficking demonstrate around 40% efficacy. As IBD is a multifactorial inflammation of the intestinal tract, a single-target approach is unlikely to solve this problem, necessitating an alternative strategy that addresses its variability. One approach often overlooked by the pharmaceutically driven therapeutic options is to address the impact of environmental factors. This is somewhat surprising considering that IBD is increasingly viewed as a condition heavily influenced by such factors, including diet, stress, and environmental pollution-often referred to as the "Western lifestyle". In IBD, intestinal responses result from a complex interplay among the genetic background of the patient, molecules, cells, and the local inflammatory microenvironment where danger- and microbe-associated molecular patterns (D/MAMPs) provide an adjuvant-rich environment. Through activating DAMP receptors, this array of pro-inflammatory factors can stimulate, for example, the NLRP3 inflammasome-a major amplifier of the inflammatory response in IBD, and various immune cells via non-specific bystander activation of myeloid cells (e.g., macrophages) and lymphocytes (e.g., tissue-resident memory T cells). Current single-target biological treatment approaches can dampen the immune response, but without reducing exposure to environmental factors of IBD, e.g., by changing diet (reducing ultra-processed foods), the adjuvant-rich landscape is never resolved and continues to drive intestinal mucosal dysregulation. Thus, such treatment approaches are not enough to put out the inflammatory fire. The resultant smoldering, low-grade inflammation diminishes physiological resilience of the intestinal (micro)environment, perpetuating the state of chronic disease. Therefore, our hypothesis posits that successful interventions for IBD must address the complexity of the disease by simultaneously targeting all modifiable aspects: innate immunity cytokines and microbiota, adaptive immunity cells and cytokines, and factors that relate to the (micro)environment. Thus the disease can be comprehensively treated across the nano-, meso-, and microscales, rather than with a focus on single targets. A broader perspective on IBD treatment that also includes options to adapt the DAMPing (micro)environment is warranted.
Collapse
Affiliation(s)
- Oliver Sandys
- Tytgat Institute for Liver and Intestinal Research, AmsterdamUMC, AGEM, University of Amsterdam, Amsterdam, The Netherlands
| | - Pieter C F Stokkers
- Department of Gastroenterology and Hepatology, OLVG West, Amsterdam, The Netherlands
| | - Anje A Te Velde
- Tytgat Institute for Liver and Intestinal Research, AmsterdamUMC, AGEM, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
37
|
Shen P, Zhang L, Jiang X, Yu B, Zhang J. Targeting HMGB1 and Its Interaction with Receptors: Challenges and Future Directions. J Med Chem 2024; 67:21671-21694. [PMID: 39648929 DOI: 10.1021/acs.jmedchem.4c01912] [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: 12/10/2024]
Abstract
High mobility group box 1 (HMGB1) is a nonhistone chromatin protein predominantly located in the nucleus. However, under pathological conditions, HMGB1 can translocate from the nucleus to the cytoplasm and subsequently be released into the extracellular space through both active secretion and passive release mechanisms. The distinct cellular locations of HMGB1 facilitate its interaction with various endogenous and exogenous factors, allowing it to perform diverse functions across a range of diseases. This Perspective provides a comprehensive overview of the structure, release mechanisms, and multifaceted roles of HMGB1 in disease contexts. Furthermore, it introduces the development of both small molecule and macromolecule inhibitors targeting HMGB1 and its interaction with receptors. A detailed analysis of the predicted pockets is also presented, aiming to establish a foundation for the future design and development of HMGB1 inhibitors.
Collapse
Affiliation(s)
- Pingping Shen
- Department of Resources Science of Traditional Chinese Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Libang Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Xuewa Jiang
- Department of Resources Science of Traditional Chinese Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Boyang Yu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Jian Zhang
- Department of Resources Science of Traditional Chinese Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 211198, P. R. China
| |
Collapse
|
38
|
Ge S, Wu S, Yin Q, Tan M, Wang S, Yang Y, Chen Z, Xu L, Zhang H, Meng C, Xia Y, Asakawa N, Wei W, Gong K, Pan X. Ecliptasaponin A protects heart against acute ischemia-induced myocardial injury by inhibition of the HMGB1/TLR4/NF-κB pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 335:118612. [PMID: 39047883 DOI: 10.1016/j.jep.2024.118612] [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: 03/20/2024] [Revised: 07/11/2024] [Accepted: 07/21/2024] [Indexed: 07/27/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Eclipta prostrata (Linn.) is a traditional medicinal Chinese herb that displays multiple biological activities, such as encompassing immunomodulatory, anti-inflammatory, anti-tumor, liver-protective, antioxidant, and lipid-lowering effects. Ecliptasaponin A (ESA), a pentacyclic triterpenoid saponin isolated from Eclipta prostrata (Linn.), has been demonstrated to exert superior anti-inflammatory activity against many inflammatory disorders. AIM OF THE STUDY Inflammation plays a critical role in acute myocardial infarction (AMI). This study aims to explore the treatment effects of ESA in AMI, as well as the underlying mechanism. METHODS An AMI mouse model was established in mice via left anterior descending coronary artery (LAD) ligation. After surgery, ESA was injected at doses of 0.5, 1.25, and 2.5 mg/kg, respectively. Myocardial infarction size, cardiomyocyte apoptosis and cardiac echocardiography were studied. The potential mechanism of action of ESA was investigated by RNA-seq, Western blot, surface plasmon resonance (SPR), molecular docking, and immunofluorescence staining. RESULTS ESA treatment not only significantly reduced myocardial infarct size, decreased myocardial cell apoptosis, and inhibited inflammatory cell infiltration, but also facilitated to improve cardiac function. RNA-seq and Western blot analysis proved that ESA treatment-induced differential expression genes mainly enriched in HMGB1/TLR4/NF-κB pathway. Consistently, ESA treatment resulted into the down-regulation of IL-1β, IL-6, and TNF-α levels after AMI. Furthermore, SPR and molecular docking results showed that ESA could bind directly to HMGB1, thereby impeding the activation of the downstream TLR4/NF-κB pathway. The immunofluorescence staining and Western blot results at the cellular level also demonstrated that ESA inhibited the activation of the HMGB1/TLR4/NF-κB pathway in H9C2 cells. CONCLUSION Our study was the first to demonstrate a cardiac protective role of ESA in AMI. Mechanism study indicated that the treatment effects of ESA are mainly attributed to its anti-inflammatory activity that was mediated by the HMGB1/TLR4/NF-κB pathway.
Collapse
Affiliation(s)
- Sumin Ge
- Department of Cardiology, Department of Pediatrics, Central Laboratory, Cutting-edge Innovation Key Lab of Major CVD in Yangzhou, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, Jiangsu, China
| | - Sihua Wu
- Division of Molecular Science, Graduate School of Science and Technology, Gunma University, Kiryu, Gunma, Japan; State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Qin Yin
- Department of Cardiology, Department of Pediatrics, Central Laboratory, Cutting-edge Innovation Key Lab of Major CVD in Yangzhou, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, Jiangsu, China
| | - Meng Tan
- Department of Cardiology, Department of Pediatrics, Central Laboratory, Cutting-edge Innovation Key Lab of Major CVD in Yangzhou, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, Jiangsu, China
| | - Sichuan Wang
- Department of Cardiology, Department of Pediatrics, Central Laboratory, Cutting-edge Innovation Key Lab of Major CVD in Yangzhou, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, Jiangsu, China
| | - Yonghao Yang
- Department of Cardiology, Department of Pediatrics, Central Laboratory, Cutting-edge Innovation Key Lab of Major CVD in Yangzhou, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, Jiangsu, China
| | - Zixuan Chen
- Department of Cardiology, Department of Pediatrics, Central Laboratory, Cutting-edge Innovation Key Lab of Major CVD in Yangzhou, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, Jiangsu, China
| | - Lei Xu
- Department of Cardiology, Department of Pediatrics, Central Laboratory, Cutting-edge Innovation Key Lab of Major CVD in Yangzhou, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, Jiangsu, China
| | - Hui Zhang
- School of Medicine, Yangzhou University, Yangzhou 225000, Jiangsu, China
| | - Chuang Meng
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, 225000, Jiangsu, China
| | - Yufei Xia
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Naoki Asakawa
- Division of Molecular Science, Graduate School of Science and Technology, Gunma University, Kiryu, Gunma, Japan
| | - Wenping Wei
- Department of Cardiology, Department of Pediatrics, Central Laboratory, Cutting-edge Innovation Key Lab of Major CVD in Yangzhou, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, Jiangsu, China.
| | - Kaizheng Gong
- Department of Cardiology, Department of Pediatrics, Central Laboratory, Cutting-edge Innovation Key Lab of Major CVD in Yangzhou, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, Jiangsu, China.
| | - Xin Pan
- Department of Cardiology, Department of Pediatrics, Central Laboratory, Cutting-edge Innovation Key Lab of Major CVD in Yangzhou, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, Jiangsu, China.
| |
Collapse
|
39
|
Wang Y, Moura AK, Zuo R, Zhou W, Wang Z, Roudbari K, Hu JZ, Li P, Zhang Y, Li X. Coronary Microvascular Dysfunction Is Associated With Augmented Lysosomal Signaling in Hypercholesterolemic Mice. J Am Heart Assoc 2024; 13:e037460. [PMID: 39604023 PMCID: PMC11681558 DOI: 10.1161/jaha.124.037460] [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: 07/02/2024] [Accepted: 10/08/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Accumulating evidence indicates that coronary microvascular dysfunction (CMD) caused by hypercholesterolemia can lead to myocardial ischemia, with or without obstructive atherosclerotic coronary artery disease. However, the molecular pathways associated with compromised coronary microvascular function before the development of myocardial ischemic injury remain poorly defined. In this study, we investigated the effects of hypercholesterolemia on the function and integrity of the coronary microcirculation in mice and the underlying mechanisms. METHODS AND RESULTS Mice were fed a hypercholesterolemic Paigen's diet for 8 weeks. Echocardiography data showed that Paigen's diet caused CMD, characterized by significant reductions in coronary blood flow and coronary flow reserve, but did not affect cardiac remodeling or dysfunction. Immunofluorescence studies revealed that Paigen's diet-induced CMD was associated with activation of coronary arterioles inflammation and increased myocardial inflammatory cell infiltration. These pathological changes occurred in parallel with the upregulation of lysosomal signaling pathways in endothelial cells (ECs). Treating hypercholesterolemic mice with the cholesterol-lowering drug ezetimibe significantly ameliorated Paigen's diet-induced adverse effects, including hypercholesterolemia, steatohepatitis, reduced coronary flow reserve, coronary endothelial cell inflammation, and myocardial inflammatory cell infiltration. In cultured mouse cardiac ECs, 7-ketocholesterol increased mitochondrial reactive oxygen species and inflammatory responses. Meanwhile, 7-ketocholesterol induced the activation of transcriptional factor EB and lysosomal signaling in mouse cardiac ECs, whereas the lysosome inhibitor bafilomycin A1 blocked 7-ketocholesterol-induced transcriptional factor EB activation and exacerbated 7-ketocholesterol-induced inflammation and cell death. Interestingly, ezetimibe synergistically enhanced 7-ketocholesterol-induced transcriptional factor EB activation and attenuated 7-ketocholesterol-induced mitochondrial reactive oxygen species and inflammatory responses in mouse cardiac ECs. CONCLUSIONS These results suggest that CMD can develop and precede detectable cardiac functional or structural changes in the setting of hypercholesterolemia and that upregulation of transcriptional factor EB-mediated lysosomal signaling in endothelial cells plays a protective role against CMD.
Collapse
Affiliation(s)
- Yun‐Ting Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of PharmacyUniversity of HoustonTX
| | - Alexandra K. Moura
- Department of Pharmacological and Pharmaceutical Sciences, College of PharmacyUniversity of HoustonTX
| | - Rui Zuo
- Department of Pharmacological and Pharmaceutical Sciences, College of PharmacyUniversity of HoustonTX
| | - Wei Zhou
- Department of Pharmacological and Pharmaceutical Sciences, College of PharmacyUniversity of HoustonTX
- Department of Medical UltrasoundTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Zhengchao Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of PharmacyUniversity of HoustonTX
- Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life SciencesFujian Normal UniversityFuzhouChina
| | - Kiana Roudbari
- Department of Pharmacological and Pharmaceutical Sciences, College of PharmacyUniversity of HoustonTX
| | - Jenny Z. Hu
- Department of Pharmacological and Pharmaceutical Sciences, College of PharmacyUniversity of HoustonTX
| | - Pin‐Lan Li
- Department of Pharmacology and ToxicologyVirginia Commonwealth University, School of MedicineRichmondVA
| | - Yang Zhang
- Department of Pharmacological and Pharmaceutical Sciences, College of PharmacyUniversity of HoustonTX
| | - Xiang Li
- Department of Pharmacological and Pharmaceutical Sciences, College of PharmacyUniversity of HoustonTX
| |
Collapse
|
40
|
Carloni S, Nasoni MG, Casabianca A, Orlandi C, Capobianco L, Iaconisi GN, Cerioni L, Burattini S, Benedetti S, Reiter RJ, Balduini W, Luchetti F. Melatonin Reduces Mito-Inflammation in Ischaemic Hippocampal HT22 Cells and Modulates the cGAS-STING Cytosolic DNA Sensing Pathway and FGF21 Release. J Cell Mol Med 2024; 28:e70285. [PMID: 39707673 DOI: 10.1111/jcmm.70285] [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: 07/22/2024] [Revised: 11/20/2024] [Accepted: 11/27/2024] [Indexed: 12/23/2024] Open
Abstract
Mitochondrial dysfunction is a key event in many pathological conditions, including neurodegenerative processes. When mitochondria are damaged, they release damage-associated molecular patterns (DAMPs) that activate mito-inflammation. The present study assessed mito-inflammation after in vitro oxygen-glucose deprivation as a representation of ischaemia, followed by reoxygenation (OGD/R) of HT22 cells and modulation of the inflammatory response by melatonin. We observed that melatonin prevented mitochondrial structural damage and dysfunction caused by OGD/R. Melatonin reduced oxidative damage and preserved the enzymatic activity for complexes I, III and IV, encoded by mitochondrial DNA, which were reduced by OGD/R. No effect was observed on complex II activity encoded by nuclear DNA. The release of mtDNA into the cytosol was also prevented with a consequent reduction of the cGAS-STING pathway and IFNβ and IL-6 production. Interestingly, melatonin also increased the early release of the fibroblast growth factor-21 (FGF-21), a mitokine secreted in response to mitochondrial stress. These data indicate that melatonin reduces mito-inflammation and modulates FGF-21 release, further highlighting the key role of this molecule in preserving mitochondrial integrity in OGD/R deprivation-type ischaemic brain injury.
Collapse
Affiliation(s)
- Silvia Carloni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Maria Gemma Nasoni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Anna Casabianca
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
- Laboratorio Covid, University of Urbino Carlo Bo, Fano, Italy
| | - Chiara Orlandi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
- Laboratorio Covid, University of Urbino Carlo Bo, Fano, Italy
| | - Loredana Capobianco
- Department of Biological Science and Technology, University of Salento, Lecce, Italy
| | | | - Liana Cerioni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Sabrina Burattini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Serena Benedetti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, Long School of Medicine, UT Health, San Antonio, Texas, USA
| | - Walter Balduini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Francesca Luchetti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| |
Collapse
|
41
|
Ruggieri E, Di Domenico E, Locatelli AG, Isopo F, Damanti S, De Lorenzo R, Milan E, Musco G, Rovere-Querini P, Cenci S, Vénéreau E. HMGB1, an evolving pleiotropic protein critical for cellular and tissue homeostasis: Role in aging and age-related diseases. Ageing Res Rev 2024; 102:102550. [PMID: 39427887 DOI: 10.1016/j.arr.2024.102550] [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: 08/06/2024] [Revised: 10/05/2024] [Accepted: 10/13/2024] [Indexed: 10/22/2024]
Abstract
Aging is a universal biological process characterized by a progressive, cumulative decline in homeostatic capabilities and physiological functions, which inevitably increases vulnerability to diseases. A number of molecular pathomechanisms and hallmarks of aging have been recognized, yet we miss a thorough understanding of their complex interconnectedness. This review explores the molecular and cellular mechanisms underlying human aging, with a focus on the multiple roles of high mobility group Box 1 protein (HMGB1), the archetypal damage-associated molecular pattern (DAMP) molecule. In the nucleus, this non-histone chromatin-associated protein functions as a DNA chaperone and regulator of gene transcription, influencing DNA structure and gene expression. Moreover, this versatile protein can translocate to the cytoplasm to orchestrate other processes, such as autophagy, or be unconventionally secreted into the extracellular environment, where it acts as a DAMP, combining inflammatory and regenerative properties. Notably, lower expression of HMGB1 within the cell and its heightened extracellular release have been associated with diverse age-associated traits, making it a suitable candidate as a universal biomarker of aging. In this review, we outline the evidence implicating HMGB1 in aging, also in light of an evolutionary perspective on its functional pleiotropy, and propose critical issues that need to be addressed to gauge the value of HMGB1 as a potential biomarker across age-related diseases and therapeutic target to promote healthy longevity.
Collapse
Affiliation(s)
- Elena Ruggieri
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| | - Erika Di Domenico
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| | | | - Flavio Isopo
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| | - Sarah Damanti
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| | - Rebecca De Lorenzo
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| | - Enrico Milan
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| | | | - Patrizia Rovere-Querini
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| | - Simone Cenci
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy.
| | - Emilie Vénéreau
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy.
| |
Collapse
|
42
|
Hu P, Li H, Ji Z, Jing W, Li Z, Yu S, Shan X, Cui Y, Wang B, Dong H, Zhou Y, Wang Z, Xiong H, Zhang X, Li HC, Wang J, Tang J, Wang T, Xie K, Liu Y, Zhu H, Yu Q. Fructose-1,6-diphosphate inhibits viral replication by promoting the lysosomal degradation of HMGB1 and blocking the binding of HMGB1 to the viral genome. PLoS Pathog 2024; 20:e1012782. [PMID: 39693295 DOI: 10.1371/journal.ppat.1012782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 11/26/2024] [Indexed: 12/20/2024] Open
Abstract
Fructose-1,6-diphosphate (FBP), a key glycolytic metabolite, is recognized for its cytoprotective effects during stress. However, the role of FBP in viral infections is unknown. Here, we demonstrate that virus-infected cells exhibit elevated FBP levels. Exogenous FBP inhibits both RNA and DNA virus infections in vitro and in vivo. Modulating intracellular FBP levels by regulating the expression of the metabolic enzymes FBP1 and PFK1 significantly impacts viral infections. Mechanistically, the inhibitory effects of FBP are not a result of altered viral adhesion or entry and are largely independent of type I interferon-mediated immune responses; rather, they occur through modulation of HMGB1. During viral infections, FBP predominantly reduces the protein levels of HMGB1 by facilitating its lysosomal degradation. Furthermore, FBP interacts with HMGB1 and disrupts the binding of HMGB1 to viral genomes, thereby further inhibiting viral replication. Our findings underscore the potential of FBP as a therapeutic target for controlling viral infections.
Collapse
Affiliation(s)
- Penghui Hu
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Huiyi Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Department of Pathogen Biology, School of Basic Medicine and Life Science, Department of Clinical Laboratory of the Second Affiliated Hospital of Hainan Medical University, The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Hainan Medical University, Hainan, China
| | - Zemin Ji
- Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Weijia Jing
- Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Zihan Li
- Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Sujun Yu
- Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xiao Shan
- Department of Health Management Centre & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yan Cui
- Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Baochen Wang
- Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Hongyuan Dong
- Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yanzhao Zhou
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
- University of Electronic Science and Technology of China, Chengdu, China
| | - Zhe Wang
- Department of Health Management Centre & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Hui Xiong
- Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xiaomei Zhang
- Department of Health Management Centre & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Hui-Chieh Li
- Department of Health Management Centre & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jinrong Wang
- Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Jiuzhou Tang
- Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Ting Wang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, Tianjin Medical University, Tianjin, China
| | - Keliang Xie
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuping Liu
- Department of Health Management Centre & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Haizhen Zhu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Department of Pathogen Biology, School of Basic Medicine and Life Science, Department of Clinical Laboratory of the Second Affiliated Hospital of Hainan Medical University, The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Hainan Medical University, Hainan, China
| | - Qiujing Yu
- Department of Health Management Centre & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
43
|
Yang HM, Zhao XN, Li XL, Wang X, Pu Y, Wei DK, Li Z. A pan-cancer analysis of the oncogenic function of HMGB1 in human tumors. Biochem Biophys Rep 2024; 40:101851. [PMID: 39582753 PMCID: PMC11584604 DOI: 10.1016/j.bbrep.2024.101851] [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: 05/09/2024] [Revised: 09/30/2024] [Accepted: 10/16/2024] [Indexed: 11/26/2024] Open
Abstract
Background Although high mobility group box protein 1 (HMGB1) has been researched in relation to cancer in many investigations, a thorough investigation of its role in pan-cancer has yet to be conducted. With the objective of bridging this gap, we delved into the functions of HMGB1 in various tumors. Methods This investigation employed The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases to examine HMGB1 gene expression differences and correlation with survival across various human tumors. Then, genetic alterations of HMGB1 were analyzed by tool cBioPortal, and immune cell infiltration was assessed. Finally, we gathered clinial samples from 95 patients with various types of solid tumor and performed somatic mutation analysis using panel sequencing. This further highlighted the role of HMGB1 in different solid tumors. Results There was a notable elevation of HMGB1 gene expression in tumor tissues as opposed to non-cancerous tissues across the bulk of tumor types. Elevated HMGB1 gene expression had a connection with shorter overall survival, progression-free survival, and disease-free survival in specific tumor types. Genetic alterations of HMGB1 suggested that the amplifications and mutations of HMGB1 may impact the prognosis of breast cancer (BRCA) and liver hepatocellular carcinoma (LIHC). Both BRCA and mesothelioma (MESO) displayed a connection between the infiltration of cancer-associated fibroblasts (CAFs) and HMGB1 gene expression. Moreover, HMGB1 co-expression analysis revealed its association with genes involved in RNA splicing, mRNA processing, and modulation of mRNA metabolic processes. Additionally, a pathway analysis by use of the Kyoto Encyclopedia of Genes and Genomes (KEGG) unveiled that HMGB1 was implicated in the pathogenic mechanisms of "Hepatitis B," "Viral Carcinogenesis," and "Hepatocellular Carcinoma." Based on somatic mutation analysis of 95 patients with different solid tumors, we found that the frequency of HMGB1 mutations was higher in Liver cancer patients compared to other solid tumors. This finding is consistent with our in-silico study results. Additionally, we discovered that the frequency of HMGB1 mutations ranked among the top 20 mutated genes in the 95 patients' data, indicating that HMGB1 plays an important role in the development and prognosis of various solid tumors. Conclusion This pan-cancer study of HMGB1 underscores its potential as a signature marker and target for the management of various tumor types.
Collapse
Affiliation(s)
- Hui-min Yang
- Shanghai Singlera Medical Laboratory, 1rst Floor, No. 20 Building, 500 Furonghua Road, Pudong New District, Shanghai, China
| | - Xiang-ning Zhao
- Department of Surgical Oncology, Shanghai Mengchao Cancer Hospital, 118 Qianyang Road, Jiading District, Shanghai, China
| | | | - Xi Wang
- Suzhou Func Biotech Inc, Suzhou, Jiangsu, China
| | - Yu Pu
- Suzhou Func Biotech Inc, Suzhou, Jiangsu, China
| | | | - Zhe Li
- Department of Breast Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Pudong New District, Shanghai, China
| |
Collapse
|
44
|
Ali M, Kumar KG, Singh K, Rabyang S, Thinlas T, Mishra A. Evaluation of the cell death markers for aberrated cell free DNA release in high altitude pulmonary edema. Clin Sci (Lond) 2024; 138:1467-1480. [PMID: 39509268 DOI: 10.1042/cs20242052] [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: 09/10/2024] [Revised: 11/04/2024] [Accepted: 11/07/2024] [Indexed: 11/15/2024]
Abstract
The effect of high altitude (HA, altitude >2500 m) can trigger a maladaptive response in unacclimatized individuals, leading to various HA illnesses such as high altitude pulmonary edema (HAPE). The present study investigates circulating cell free (cf) DNA, a minimally invasive biomarker that can elicit a pro-inflammatory response. Our earlier study observed altered cfDNA fragment patterns in HAPE patients and the significant correlation of these patterns with peripheral oxygen saturation levels. However, the unclear release mechanisms of cfDNA in circulation limit its characterization and clinical utility. The present study not only observed a significant increase in cfDNA levels in HAPE patients (27.03 ± 1.37 ng/ml; n = 145) compared to healthy HA sojourners (controls, 14.57 ± 0.74 ng/ml; n = 65) and highlanders (HLs, 15.50 ± 0.8 ng/ml; n = 34) but also assayed the known cell death markers involved in cfDNA release at HA. The study found significantly elevated levels of the apoptotic marker, annexin A5, and secondary necrosis or late apoptotic marker, high mobility group box 1, in HAPE patients. In addition, we observed a higher oxidative DNA damage marker, 8-hydroxy-2'-deoxyguanosine, in HAPE compared with controls, suggestive of the role of oxidative DNA status in promoting the inflammatory potential of cfDNA fragments and their plausible role in manifesting HAPE pathophysiology. Extensive in vitro future assays can confirm the immunogenic role of cfDNA fragments that may act as a danger-associated molecular pattern and associate with markers of cellular stresses in HAPE.
Collapse
Affiliation(s)
- Manzoor Ali
- Genomics and Genome Biology Unit, CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Krishna G Kumar
- Genomics and Genome Biology Unit, CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India
| | - Kanika Singh
- Genomics and Genome Biology Unit, CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Stanzen Rabyang
- Department of Medicine, Sonam Norboo Memorial Hospital, Leh 194101, India
| | - Tashi Thinlas
- Department of Medicine, Sonam Norboo Memorial Hospital, Leh 194101, India
| | - Aastha Mishra
- Genomics and Genome Biology Unit, CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
45
|
Abdelrahman RS, Abdelmageed ME. Hepatoprotective effects of the xanthine oxidase inhibitor Febuxostat against thioacetamide-induced liver injury in rats: The role of the Nrf2/ HO-1 and TLR4/ NF-κB pathways. Food Chem Toxicol 2024; 194:115087. [PMID: 39489394 DOI: 10.1016/j.fct.2024.115087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/27/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
Experimental models of liver injury have been established utilizing thioacetamide (TAA), a classic liver toxic chemical that causes organ damage via oxidative stress and inflammatory induction. This study examined the impact of Febuxostat (a xanthine oxidase inhibitor; Febu, 10-15 mg/kg, orally) against TAA (500 mg/kg, i.p.) -induced liver injury in rats. Febu significantly attenuated TAA-induced alterations in liver function parameters, in addition to promoting hepatic antioxidant effects through a significant elevation of Heme-oxygenase-1(HO-1), nuclear factor erythroid 2-related factor2 (Nrf2), reduced glutathione (GSH) and superoxide dismutase (SOD) levels and reduction in hepatic malondialdehyde (MDA) content. Moreover, Febu improved the hepatic anti-inflammatory status by increasing the anti-inflammatory cytokine Interleukin (IL-10) level and reducing the levels of the pro-inflammatory cytokines (Nuclear factor kappa B (NF-κB), IL-1β, high-mobility group box1 (HMGB1), receptor for advanced glycation end products (RAGE), and toll-like receptor4 (TLR4) levels, in addition to suppressing the increased protein and mRNA expression levels of tumor necrosis factor alpha (TNF-α) and IL-6, hepatic expression of TNF-α and activated mitogen-activated protein kinases (p-JNK/p-p38 MAPK). Histopathologically, Febu markedly normalized TAA-induced alteration in liver sections. In conclusion, Febu, in a dose-dependent fashion, refines TAA-induced hepatotoxicity by enhancing antioxidant capabilities and decreasing inflammatory signals.
Collapse
Affiliation(s)
- Rehab S Abdelrahman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516, Mansoura, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Taibah University, Al-Madina Al-Munawwarah, 30001, Saudi Arabia
| | - Marwa E Abdelmageed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516, Mansoura, Egypt.
| |
Collapse
|
46
|
Choi A, Woo JS, Park YS, Kim JH, Chung YE, Lee S, Beom JH, You JS. TARGETED TEMPERATURE MANAGEMENT AT 36°C IMPROVES SURVIVAL AND PROTECTS TISSUES BY MITIGATING THE DELETERIOUS INFLAMMATORY RESPONSE FOLLOWING HEMORRHAGIC SHOCK. Shock 2024; 62:716-727. [PMID: 39186053 DOI: 10.1097/shk.0000000000002453] [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: 08/27/2024]
Abstract
ABSTRACT Hemorrhagic shock (HS) is a life-threatening condition with high mortality rates despite current treatments. This study investigated whether targeted temperature management (TTM) could improve outcomes by modulating inflammation and protecting organs following HS. Using a rat model of HS, TTM was applied at 33°C and 36°C after fluid resuscitation. Surprisingly, TTM at 33°C increased mortality, while TTM at 36°C significantly improved survival rates. It also reduced histological damage in lung and kidney tissues, lowered serum lactate levels, and protected against apoptosis and excessive reactive oxygen species production. TTM at 36°C inhibited the release of high mobility group box 1 protein (HMGB1), a key mediator of inflammation, and decreased proinflammatory cytokine levels in the kidneys and lungs. Moreover, it influenced macrophage behavior, suppressing the harmful M1 phenotype while promoting the beneficial M2 polarization. Cytokine array analysis confirmed reduced levels of proinflammatory cytokines with TTM at 36°C. These results collectively highlight the potential of TTM at 36°C as a therapeutic approach to improve outcomes in HS. By addressing multiple aspects of injury and inflammation, including modulation of macrophage responses and cytokine profiles, TTM at 36°C offers promising implications for critical care management after HS, potentially reducing mortality and improving patient recovery.
Collapse
Affiliation(s)
- Arom Choi
- Department of Emergency Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ji Sun Woo
- Department of Emergency Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yoo Seok Park
- Department of Emergency Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ju Hee Kim
- Department of Emergency Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong Eun Chung
- Department of Radiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sojung Lee
- Class of 2025, Biology B.S., Emory University, Atlanta, Georgia
| | - Jin Ho Beom
- Department of Emergency Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Je Sung You
- Department of Emergency Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
47
|
Ge X, Subramaniyam N, Song Z, Desert R, Han H, Das S, Komakula SSB, Wang C, Lantvit D, Ge Z, Hoshida Y, Nieto N. Post-translational modifications drive the effects of HMGB1 in alcohol-associated liver disease. Hepatol Commun 2024; 8:e0549. [PMID: 39760999 PMCID: PMC11495752 DOI: 10.1097/hc9.0000000000000549] [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: 07/12/2024] [Accepted: 08/26/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND We previously identified that high-mobility group box-1 (HMGB1) is increased and undergoes post-translational modifications (PTMs) in response to alcohol consumption. Here, we hypothesized that specific PTMs, occurring mostly in hepatocytes and myeloid cells, could contribute to the pathogenesis of alcohol-associated liver disease (AALD). METHODS We used the Lieber-DeCarli (LD) model of early alcohol-induced liver injury, combined with engineered viral vectors and genetic approaches to regulate the expression of HMGB1, its PTMs (reduced [H], oxidized [O], acetylated [Ac], both [O + Ac]), and its receptors (RAGE, TLR4) in a cell-specific manner (hepatocytes and/or myeloid cells). RESULTS Hmgb1 ablation in hepatocytes or myeloid cells partially protected, while ablation in both prevented steatosis, inflammation, IL1B production, and alcohol-induced liver injury. Hepatocytes were a major source of [H], [O], and [Ac] HMGB1, whereas myeloid cells produced only [H] and [Ac] HMGB1. Neutralization of HMGB1 prevented, whereas injection of [H] HMGB1 increased AALD, which was worsened by injection of [O] HMGB1. While [O] HMGB1 induced liver injury, [Ac] HMGB1 protected and counteracted the effects of [O] HMGB1 in AALD. [O] HMGB1 stimulated macrophage (MF) migration, activation, IL1B production, and secretion. Ethanol-fed RageΔMye but not Tlr4ΔMye, RageΔHep, or Tlr4ΔHep mice were protected from AALD, indicating a crucial role of RAGE in myeloid cells for AALD. [O] HMGB1 recruited and activated myeloid cells through RAGE and contributed to steatosis, inflammation, and IL1B production in AALD. CONCLUSIONS These results provide evidence for targeting [O] HMGB1 of hepatocyte origin as a ligand for RAGE signaling in myeloid cells and a driver of steatosis, inflammatory cell infiltration, and IL1B production in AALD. Importantly, we reveal that [Ac] HMGB1 offsets the noxious effects of [O] HMGB1 in AALD.
Collapse
Affiliation(s)
- Xiaodong Ge
- Department of Pathology, University of Illinois Chicago, Chicago, Illinois, USA
| | | | - Zhuolun Song
- Department of Pathology, University of Illinois Chicago, Chicago, Illinois, USA
| | - Romain Desert
- Department of Pathology, University of Illinois Chicago, Chicago, Illinois, USA
| | - Hui Han
- Department of Pathology, University of Illinois Chicago, Chicago, Illinois, USA
| | - Sukanta Das
- Department of Pathology, University of Illinois Chicago, Chicago, Illinois, USA
| | | | - Chao Wang
- Department of Pathology, University of Illinois Chicago, Chicago, Illinois, USA
| | - Daniel Lantvit
- Department of Pathology, University of Illinois Chicago, Chicago, Illinois, USA
| | - Zhiyan Ge
- Department of Pathology, University of Illinois Chicago, Chicago, Illinois, USA
| | - Yujin Hoshida
- Department of Internal Medicine, Division of Digestive and Liver Diseases, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Natalia Nieto
- Department of Pathology, University of Illinois Chicago, Chicago, Illinois, USA
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Illinois Chicago, Chicago, Illinois, USA
- Research & Development Service, Jesse Brown Veterans Affairs Medical Center, Chicago, lllinois, USA
| |
Collapse
|
48
|
Castellanos-Molina A, Bretheau F, Boisvert A, Bélanger D, Lacroix S. Constitutive DAMPs in CNS injury: From preclinical insights to clinical perspectives. Brain Behav Immun 2024; 122:583-595. [PMID: 39222725 DOI: 10.1016/j.bbi.2024.07.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/28/2024] [Accepted: 07/04/2024] [Indexed: 09/04/2024] Open
Abstract
Damage-associated molecular patterns (DAMPs) are endogenous molecules released in tissues upon cellular damage and necrosis, acting to initiate sterile inflammation. Constitutive DAMPs (cDAMPs) have the particularity to be present within the intracellular compartments of healthy cells, where they exert diverse functions such as regulation of gene expression and cellular homeostasis. However, after injury to the central nervous system (CNS), cDAMPs are rapidly released by stressed, damaged or dying neuronal, glial and endothelial cells, and can trigger inflammation without undergoing structural modifications. Several cDAMPs have been described in the injured CNS, such as interleukin (IL)-1α, IL-33, nucleotides (e.g. ATP), and high-mobility group box protein 1. Once in the extracellular milieu, these molecules are recognized by the remaining surviving cells through specific DAMP-sensing receptors, thereby inducing a cascade of molecular events leading to the production and release of proinflammatory cytokines and chemokines, as well as cell adhesion molecules. The ensuing immune response is necessary to eliminate cellular debris caused by the injury, allowing for damage containment. However, seeing as some molecules associated with the inflammatory response are toxic to surviving resident CNS cells, secondary damage occurs, aggravating injury and exacerbating neurological and behavioral deficits. Thus, a better understanding of these cDAMPs, as well as their receptors and downstream signaling pathways, could lead to identification of novel therapeutic targets for treating CNS injuries such as SCI, TBI, and stroke. In this review, we summarize the recent literature on cDAMPs, their specific functions, and the therapeutic potential of interfering with cDAMPs or their signaling pathways.
Collapse
Affiliation(s)
- Adrian Castellanos-Molina
- Axe Neurosciences du Centre de recherche du Centre hospitalier universitaire (CHU) de Québec-Université Laval et Département de médecine moléculaire de l'Université Laval, Québec, QC G1V 4G2, Canada
| | - Floriane Bretheau
- Axe Neurosciences du Centre de recherche du Centre hospitalier universitaire (CHU) de Québec-Université Laval et Département de médecine moléculaire de l'Université Laval, Québec, QC G1V 4G2, Canada
| | - Ana Boisvert
- Axe Neurosciences du Centre de recherche du Centre hospitalier universitaire (CHU) de Québec-Université Laval et Département de médecine moléculaire de l'Université Laval, Québec, QC G1V 4G2, Canada
| | - Dominic Bélanger
- Axe Neurosciences du Centre de recherche du Centre hospitalier universitaire (CHU) de Québec-Université Laval et Département de médecine moléculaire de l'Université Laval, Québec, QC G1V 4G2, Canada
| | - Steve Lacroix
- Axe Neurosciences du Centre de recherche du Centre hospitalier universitaire (CHU) de Québec-Université Laval et Département de médecine moléculaire de l'Université Laval, Québec, QC G1V 4G2, Canada.
| |
Collapse
|
49
|
Wang B, Feng S, Jiang Y, Tang Y, Man Y, Wei N, Xiang L. Early anti-inflammatory polarization of macrophages ameliorates post-surgical inflammation and osseointegration around titanium implants in mice. Mol Immunol 2024; 175:155-163. [PMID: 39437620 DOI: 10.1016/j.molimm.2024.10.001] [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: 05/26/2024] [Revised: 09/26/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024]
Abstract
Dental implants are considered a superior option for the replacement of missing teeth. However, the invasive nature of the surgical procedure often results in significant postoperative inflammation, and the prolonged healing period of 3-6 months presents a notable disadvantage. High mobility group box 1 (HMGB1) is a critical mediator of acute inflammation following surgical injury, which can hinder the onset of osseointegration. This study aims to examine whether the inhibition of HMGB1 can mitigate acute inflammation and subsequently enhance osseointegration. The findings indicate that HMGB1 inhibition markedly reduces inflammation and promotes bone repair in murine models. Further in vitro investigations into the regulatory mechanisms of HMGB1 in macrophages reveal its role in increasing Yes-associated protein (YAP) activity, which contributes to pro-inflammatory polarization. Additionally, conditioned media derived from macrophages influenced by HMGB1 significantly impair the migratory and osteogenic capabilities of bone marrow-derived mesenchymal stem cells, which are essential for bone regeneration. In vivo experiments further validate that the administration of exogenous HMGB1 exacerbates postoperative acute inflammation and obstructs osseointegration. The study concludes that inhibiting HMGB1 fosters an anti-inflammatory polarization of macrophages, leading to diminished postoperative acute inflammation and expedited osseointegration around dental implants in mice.
Collapse
Affiliation(s)
- Bin Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China; Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.
| | - Shuqi Feng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| | - Yixuan Jiang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China; Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.
| | - Yufei Tang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| | - Yi Man
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| | - Na Wei
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| | - Lin Xiang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
50
|
Kusumo MHB, Prayitno A, Soetrisno, Laqif A. Synergistic therapeutic approach for hemorrhoids: integrating mesenchymal stem cells with diosmin-hesperidin to target tissue edema and inflammation. Arch Med Sci 2024; 20:1556-1566. [PMID: 39649264 PMCID: PMC11623161 DOI: 10.5114/aoms/183465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 01/30/2024] [Indexed: 12/10/2024] Open
Abstract
Introduction Mesenchymal stem cells (MSCs) have promising regenerative properties in tissue repair and anti-inflammatory responses. This study aimed to investigate the effects of MSCs and their combination with micronized purified flavonoid fraction (MPFF) in a croton oil-induced hemorrhoids model on tissue edema, inflammation, and underlying molecular mechanisms. Material and methods MSCs were isolated and characterized for their adherence, differentiation capacity, and immunophenotyping. Croton oil-induced hemorrhoid mouse models were established to assess tissue edema, inflammation, tumor necrosis factor (TNF-α) expression, transforming growth factor-β (TGF-β) expression, collagen ratio, and MMP-9 activity. The effects of MSCs and their combination with MPFF (diosmin-hesperidin) were evaluated through histological examinations, western blot analysis, and gelatin zymography. Results Characterization confirmed the MSCs' plastic adherence, osteogenic differentiation potential, and immunophenotype (positive for CD90 and CD29, negative for CD45 and CD31). Treatment with MSCs alone or in combination with MPFF significantly reduced tissue edema, inflammation, TNF-α expression, and MMP-9 activity. Additionally, MSCs increased TGF-β expression, and collagen type I/III ratio, and accelerated wound healing by resolving inflammation. Conclusions These findings suggest that MSCs play a crucial role in modulating TNF-α, TGF-β, collagen remodeling, and MMP-9 activity, highlighting their promising role in hemorrhoid treatment and wound healing processes. Further research is warranted to fully elucidate the intricate mechanisms and optimize MSC-based therapies for clinical applications in hemorrhoidal disease management.
Collapse
Affiliation(s)
- M. Hidayat Budi Kusumo
- Doctoral program of Medical Sciences, Faculty of Medicine, Universitas Sebelas Maret, Surakarta, Indonesia
- Department of Surgery, Faculty of Medicine, Universitas Muhammadiyah Purwokerto, Purwokerto, Indonesia
| | - Adi Prayitno
- Doctoral program of Medical Sciences, Faculty of Medicine, Universitas Sebelas Maret, Surakarta, Indonesia
- Department of Oral and Maxillofacial Pathology, Faculty of Medicine, Universitas Sebelas Maret, Surakarta, Indonesia
| | - Soetrisno
- Doctoral program of Medical Sciences, Faculty of Medicine, Universitas Sebelas Maret, Surakarta, Indonesia
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universitas Sebelas Maret, Surakarta, Indonesia
| | - Abdurahman Laqif
- Doctoral program of Medical Sciences, Faculty of Medicine, Universitas Sebelas Maret, Surakarta, Indonesia
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universitas Sebelas Maret, Surakarta, Indonesia
- Department of Obstetrics and Gynaecology, Moewardi General Hospital, Surakarta, Indonesia
| |
Collapse
|