1
|
Ulaş N, Üstündağ H, Özkanlar S, Erbaş E, Kara A, Özkanlar Y. D-carvone attenuates LPS-induced acute lung injury via TLR4/NF-κB and Nrf2/HO-1 signaling pathways in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04024-y. [PMID: 40116872 DOI: 10.1007/s00210-025-04024-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 03/05/2025] [Indexed: 03/23/2025]
Abstract
Acute lung injury (ALI) is a severe respiratory disorder associated with high morbidity and mortality. Lipopolysaccharide (LPS) is widely used to induce ALI in animal models. D-carvone, a natural monoterpene, has been reported to possess anti-inflammatory and antioxidant properties. This study aimed to investigate the protective effects of D-carvone on LPS-induced ALI in rats. Thirty-six male rats were randomly divided into six groups (n = 6): control, D-carvone (10 mg/kg and 20 mg/kg p.o.), LPS (10 mg/kg E. coli lipopolysaccharide i.p.), and LPS + D-carvone (LPS with either 10 or 20 mg/kg D-carvone). D-carvone was administered orally once daily for 10 days. On day 10, sepsis was induced with LPS administration, and samples were collected after 6 h under deep anesthesia. LPS administration caused significant lung injury, as evidenced by increased histopathological scores, upregulation of pro-inflammatory markers (TLR4, IL-1β, TNF-α), and oxidative stress (increased MDA, decreased GSH and SOD). Treatment with D-carvone at both doses significantly attenuated these changes. D-carvone downregulated pro-inflammatory markers, upregulated anti-inflammatory (NRF2) and anti-apoptotic (Bcl-2) proteins, and reduced the levels of pro-inflammatory cytokines (IL-1β, TNF-α, IL-8) in lung tissues. In conclusion, D-carvone protects against LPS-induced ALI in rats, possibly through its anti-inflammatory and antioxidant properties. These findings suggest that D-carvone could be a potential therapeutic candidate for preventing and treating ALI.
Collapse
Affiliation(s)
- Nergis Ulaş
- Department of Internal Medicine, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
| | - Hilal Üstündağ
- Department of Physiology, Faculty of Medicine, Erzincan Binali Yıldırım University, Erzincan, Turkey.
| | - Seçkin Özkanlar
- Department of Biochemistry, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
| | - Elif Erbaş
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Adem Kara
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical University, Erzurum, Turkey
| | - Yunusemre Özkanlar
- Department of Internal Medicine, Faculty of Veterinary, Ondokuz Mayis University, Samsun, Turkey
| |
Collapse
|
2
|
Wang C, Yin L, Fu P, Lu G, Zhai X, Yang C. Anti-inflammatory effect of ApoE23 on Salmonella typhimurium-induced sepsis in mice. Open Med (Wars) 2023; 18:20230767. [PMID: 37533741 PMCID: PMC10390754 DOI: 10.1515/med-2023-0767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 07/06/2023] [Accepted: 07/06/2023] [Indexed: 08/04/2023] Open
Abstract
Two independent experiments were performed with three groups each (sepsis control, sepsis, and sepsis with apoE23 treatment) to investigate the anti-inflammatory effect of apolipoprotein 23 (apoE23) in a mouse model of sepsis induced by S. typhimurium. Survival rates; plasma level variations in tumor necrosis factor (TNF)-α, interleukin (IL)-6, and lipopolysaccharide (LPS); S. typhimurium colony-forming units in the spleen tissue; and mRNA and protein expression levels of low-density lipoprotein receptor (LDLR), LDLR-related protein (LRP), syndecan-1, and scavenger receptor B1 were evaluated in the livers of mice from the three groups. Results found that the survival rate of septic mice treated with apoE23 was 100% within 48 h, while it was only 40% in septic mice without apoE23 treatment (P < 0.001). The plasma LPS, TNF-α, and IL-6 levels and the S. typhimurium load in mice in the apoE23-treated group were significantly lower than those in septic mice (P < 0.05). Moreover, apoE23 restored the downregulated expression of LDLR and LRP in the liver tissue of septic mice. So apoE23 exhibits an anti-inflammatory effect in the mouse model of S. typhimurium-induced sepsis. Further studies are required to understand the mechanisms underlying the anti-inflammatory effects of apoE23.
Collapse
Affiliation(s)
- Chuanqing Wang
- Department of Nosocomial Infection Control and the Clinical Microbiology Laboratory, Children’s Hospital of Fudan University, Shanghai200032, China
- Department of Nosocomial Infection Control, Children’s Hospital of Fudan University, Shanghai200032, China
| | - Lijun Yin
- Department of Nosocomial Infection Control, Children’s Hospital of Fudan University, Shanghai200032, China
| | - Pan Fu
- Department of the Clinical Microbiology Laboratory, Children’s Hospital of Fudan University, Shanghai200032, China
| | - Guoping Lu
- Department of Pediatric Intensive Care Unit, Children’s Hospital of Fudan University, Shanghai200032, China
| | - Xiaowen Zhai
- Department of Hematology, Children’s Hospital of Fudan University, Shanghai, 399 Wanyuan Road, Shanghai200032, China
| | - Changsheng Yang
- The Institute of Cardiovascular Diseases of Shanghai, Key Laboratory of Viral Heart Diseases, Ministry of Health, Zhongshan Hospital of Fudan University, 180 Fenglin Road, Shanghai200032, China
| |
Collapse
|
3
|
Niu K, Qu S, Yang L, Zhang H, Yuan J, Fan H, Li X, Tao K. Protective effect of HDACIs in improves survival and organ injury after CLP-induced sepsis. Surg Open Sci 2023; 12:35-42. [PMID: 36936452 PMCID: PMC10015250 DOI: 10.1016/j.sopen.2023.03.003] [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: 02/17/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
Introduction The effects of isoform-specific histone deacetylase inhibitors (HDACIs) and the non-selective HDACI on sepsis have been profoundly reported. However, the best HDAC classes have not been fully evaluated. Therefore, this study aimed to determine which HDACIs are responsible for survival and beneficial for organ injury. Methods Experiment I, SD rats were subjected to cecal ligation and puncture and randomly assigned to the no treatment, dimethyl sulfoxide (DMSO) only, MS-275, LMK-235, tubastatinA (TubA), trichostatin-A (TSA), and sirtinol groups (n = 5). Survival was monitored for 48 h. Experiment II, the animals were monitored for 12 h, then, blood and tissues sample were collected. Interleukin (IL)-6, IL-1β, tumour necrosis factor (TNF)-α, alanine aminotransferase (ALT), aspartate aminotransferase (AST), creatine kinase (CK) and lactate dehydrogenase (LDH) expressions were evaluated using ELISA. Liver, heart and lung tissues were analysed via hematoxylin and eosin staining. Liver and heart tissue lysates were analysed for acetylated histones H3, H4, a-tubulin and nuclear factor kappa B (NF-κB), IL-6, IL-1β, and TNF-α using western blotting. Splenocytes were examined via flow cytometry to analyse the immune cell population. Results MS-275, TubA and TSA treatments significantly prolonged survival. MS-275, LMK-235, TubA and TSA significantly reduced the histopathological scores and AST, ALT, CK, LDH, IL-6, IL-1β and TNF-α levels, significantly increased acetylated of NF-κB and changed the immune cell proportion. Conclusion Our results indicated that HDACI classes I and IIb and non-selective HDACI can significantly prolong survival. Moreover, non-selective and isoform-specific class I and IIa/IIb HDACIs can attenuate inflammation and organ injury.
Collapse
|
4
|
Soni M, Handa M, Singh KK, Shukla R. Recent nanoengineered diagnostic and therapeutic advancements in management of Sepsis. J Control Release 2022; 352:931-945. [PMID: 36273527 PMCID: PMC9665001 DOI: 10.1016/j.jconrel.2022.10.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/15/2022] [Accepted: 10/17/2022] [Indexed: 11/17/2022]
Abstract
COVID-19 acquired symptoms have affected the worldwide population and increased the load of Intensive care unit (ICU) patient admissions. A large number of patients admitted to ICU end with a deadly fate of mortality. A high mortality rate of patients was reported with hospital-acquired septic shock that leads to multiple organ failures and ultimately ends with death. The patients who overcome this septic shock suffer from morbidity that also affects their caretakers. To overcome these situations, scientists are exploring progressive theragnostic techniques with advanced techniques based on biosensors, biomarkers, biozymes, vesicles, and others. These advanced techniques pave the novel way for early detection of sepsis-associated symptoms and timely treatment with appropriate antibiotics and immunomodulators and prevent the undue effect on other parts of the body. There are other techniques like externally modulated electric-based devices working on the principle of piezoelectric mechanism that not only sense the endotoxin levels but also target them with a loaded antibiotic to neutralize the onset of inflammatory response. Recently researchers have developed a lipopolysaccharide (LPS) neutralizing cartridge that not only senses the LPS but also appropriately neutralizes with dual mechanistic insights of antibiotic and anti-inflammatory effects. This review will highlight recent developments in the new nanotechnology-based approaches for the diagnosis and therapeutics of sepsis that is responsible for the high number of deaths of patients suffering from this critical disease.
Collapse
Affiliation(s)
- Mukesh Soni
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, U.P., India
| | - Mayank Handa
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, U.P., India
| | - Kamalinder K. Singh
- School of Pharmacy and Biomedical Sciences, Faculty of Clinical and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK,Correspondence to: Prof. Kamalinder K. Singh, School of Pharmacy and Biomedical Sciences, Faculty of Clinical and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, United Kingdom
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, U.P., India,School of Pharmacy and Biomedical Sciences, Faculty of Clinical and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK,Correspondence to: Dr. Rahul Shukla (M. Pharm. PhD), National Institute of Pharmaceutical Education and Research (NIPER-Raebareli), Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow 226002, UP, India
| |
Collapse
|
5
|
Rosier F, Nuñez NF, Torres M, Loriod B, Rihet P, Pradel LC. Transcriptional Response in a Sepsis Mouse Model Reflects Transcriptional Response in Sepsis Patients. Int J Mol Sci 2022; 23:ijms23020821. [PMID: 35055007 PMCID: PMC8776114 DOI: 10.3390/ijms23020821] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/06/2022] [Accepted: 01/09/2022] [Indexed: 02/06/2023] Open
Abstract
Mortality due to sepsis remains unacceptably high, especially for septic shock patients. Murine models have been used to better understand pathophysiology mechanisms. However, the mouse model is still under debate. Herein we investigated the transcriptional response of mice injected with lipopolysaccharide (LPS) and compared it to either human cells stimulated in vitro with LPS or to the blood cells of septic patients. We identified a molecular signature composed of 2331 genes with an FDR median of 0%. This molecular signature is highly enriched in regulated genes in peritoneal macrophages stimulated with LPS. There is significant enrichment in several inflammatory signaling pathways, and in disease terms, such as pneumonia, sepsis, systemic inflammatory response syndrome, severe sepsis, an inflammatory disorder, immune suppression, and septic shock. A significant overlap between the genes upregulated in mouse and human cells stimulated with LPS has been demonstrated. Finally, genes upregulated in mouse cells stimulated with LPS are enriched in genes upregulated in human cells stimulated in vitro and in septic patients, who are at high risk of death. Our results support the hypothesis of common molecular and cellular mechanisms between mouse and human sepsis.
Collapse
Affiliation(s)
| | | | | | | | - Pascal Rihet
- Correspondence: (P.R.); (L.C.P.); Tel.: +33-491828723 (P.R.); +33-491828745 (L.C.P.)
| | - Lydie C. Pradel
- Correspondence: (P.R.); (L.C.P.); Tel.: +33-491828723 (P.R.); +33-491828745 (L.C.P.)
| |
Collapse
|
6
|
Kim E, Ku SK, Yang S, Lee BS, Kim GJ, Choi H, Bae JS. Collismycin C reduces HMGB1-mediated septic responses and improves survival rate in septic mice. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2021; 23:55-72. [PMID: 31888389 DOI: 10.1080/10286020.2019.1706497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 12/15/2019] [Indexed: 06/10/2023]
Abstract
We examined the effects of a 2,2'-bipyridine containing natural product, collismycin C on high mobility group box 1 (HMGB1, septic mediator)-mediated septic responses and survival rate in a mouse sepsis model. Collismycin C inhibited the HMGB1 release and downregulated HMGB1-mediated inflammatory responses in human endothelial cells. Collismycin C also inhibited HMGB1-induced hyperpermeability and leukocyte migration in mice. In addition, collismycin C treatment reduced CLP-induced HMGB1 release and sepsis-related mortality and pulmonary damage in vivo. Our results indicate that collismycin C is a potential therapeutic agent for the treatment of severe vascular inflammatory diseases by inhibiting HMGB1 signaling pathway.
Collapse
Affiliation(s)
- Eonmi Kim
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Sae-Kwang Ku
- Department of Histology and Anatomy, College of Korean Medicine, Daegu Haany University, Gyeongsan-si 38610, Republic of Korea
| | - Sumin Yang
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, BK21 plus KNU Multi-Omics based Creative Drug Research Team, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Bong-Seon Lee
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, BK21 plus KNU Multi-Omics based Creative Drug Research Team, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Geum Jin Kim
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Hyukjae Choi
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Jong-Sup Bae
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, BK21 plus KNU Multi-Omics based Creative Drug Research Team, Kyungpook National University, Daegu 41566, Republic of Korea
| |
Collapse
|
7
|
Manandhar S, Sinha P, Ejiwale G, Bhatia M. Hydrogen Sulfide and its Interaction with Other Players in Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1315:129-159. [PMID: 34302691 DOI: 10.1007/978-981-16-0991-6_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hydrogen sulfide (H2S) plays a vital role in human physiology and in the pathophysiology of several diseases. In addition, a substantial role of H2S in inflammation has emerged. This chapter will discuss the involvement of H2S in various inflammatory diseases. Furthermore, the contribution of reactive oxygen species (ROS), adhesion molecules, and leukocyte recruitment in H2S-mediated inflammation will be discussed. The interrelationship of H2S with other gasotransmitters in inflammation will also be examined. There is mixed literature on the contribution of H2S to inflammation due to studies reporting both pro- and anti-inflammatory actions. These apparent discrepancies in the literature could be resolved with further studies.
Collapse
Affiliation(s)
- Sumeet Manandhar
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Priyanka Sinha
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Grace Ejiwale
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Madhav Bhatia
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand.
| |
Collapse
|
8
|
Affiliation(s)
- Daniel Hollander
- Division of Digestive Diseases, Department of Medicine, UCLA School of Medicine, Los Angeles, CA 90024, USA
| | - Jonathan D. Kaunitz
- Division of Digestive Diseases, Department of Medicine, UCLA School of Medicine, Los Angeles, CA 90024, USA,Department of Surgery, UCLA School of Medicine, Los Angeles, CA 90024, USA,Gastroenterology Section, Medical Service, West Los Angeles VAMC, Los Angeles, CA 90073, USA
| |
Collapse
|
9
|
Jeong SY, Kim M, Park EK, Kim JS, Hahn D, Bae JS. Inhibitory Functions of Novel Compounds from Dioscorea batatas Decne Peel on HMGB1-mediated Septic Responses. BIOTECHNOL BIOPROC E 2020. [DOI: 10.1007/s12257-019-0382-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
10
|
Suppressive Effects of GSS on Lipopolysaccharide-Induced Endothelial Cell Injury and ALI via TNF- α and IL-6. Mediators Inflamm 2019; 2019:4251394. [PMID: 32082076 PMCID: PMC7012263 DOI: 10.1155/2019/4251394] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 09/01/2019] [Accepted: 10/23/2019] [Indexed: 12/21/2022] Open
Abstract
Background. Under septic conditions, LPS induced lung vascular endothelial cell (EC) injury, and the release of inflammatory mediator launches and aggravates acute lung injury (ALI). There are no effective therapeutic options for ALI. Genistein-3'-sodium sulfonate (GSS) is a derivative of native soy isoflavone, which exhibits neuroprotective effects via its antiapoptosis property. However, whether GSS protect against sepsis-induced EC injury and release of inflammatory mediators has not been determined. In this study, we found that GSS not only downregulated the levels of TNF-α and IL-6 in the lung and serum of mice in vivo but also inhibited the expression and secretion of TNF-α and IL-6 in ECs. Importantly, we also found that GSS blocked LPS-induced TNF-α and IL-6 expression in ECs via the Myd88/NF-κB signaling pathway. Taken together, our results demonstrated that GSS might be a promising candidate for sepsis-induced ALI via its regulating effects on inflammatory response in lung ECs.
Collapse
|
11
|
Fei QL, Zhang XY, Qi RJ, Huang YF, Han YX, Li XM, Cai RL, Gao Y, Qi Y. Canscora lucidissima, a Chinese folk medicine, exerts anti-inflammatory activities by inhibiting the phosphorylation of ERK1/2 in LPS-activated macrophages. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 19:371. [PMID: 31842849 PMCID: PMC6916437 DOI: 10.1186/s12906-019-2783-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 11/29/2019] [Indexed: 02/02/2023]
Abstract
Background Canscora lucidissima (Levl. & Vaniot) Hand.-Mazz. (C. lucidissima), mainly distributed in southern China, has been shown to be effective in the treatment of inflammatory diseases. However, the underlying mechanism of its anti-inflammatory effect is not fully understood. Methods In this study, we investigated the anti-inflammatory mechanism of ethanol extract of C. lucidissima (Cl-EE) in lipopolysaccharide (LPS)-induced inflammatory models. ELISA, real-time PCR, Western blot and luciferase reporter assay were used for the experiments in vitro, and ICR mouse endotoxemia model was used for in vivo test. Results Our data showed that Cl-EE reduced the production of NO by down-regulating the mRNA and protein expression of inducible nitric oxide synthase (iNOS) in LPS-activated RAW 264.7 cells. Meanwhile, it potently decreased other proinflammatory mediators, such as TNF-α, IL-6, MCP-1 and IL-1β at the transcriptional and translational levels. Further study indicated that Cl-EE did not affect NF-κB signaling pathway but significantly suppressed the phosphorylation of ERK1/2, rather than JNK or p38. In a LPS-induced endotoxemia mouse model, a single intraperitoneal injection of Cl-EE (75–300 mg/kg) could lower circulatory TNF-α, IL-6 and MCP-1 levels. Conclusions Collectively, our results indicated that Cl-EE suppressed the phosphorylation level of ERK1/2 thus reducing the transcription and translation of inflammatory genes, thereby exerted anti-inflammatory activity. This study reveals the anti-inflammatory mechanism of C. lucidissima and may provide an effective treatment option for a variety of inflammatory diseases.
Collapse
|
12
|
Tunctan B, Senol SP, Temiz-Resitoglu M, Guden DS, Sahan-Firat S, Falck JR, Malik KU. Eicosanoids derived from cytochrome P450 pathway of arachidonic acid and inflammatory shock. Prostaglandins Other Lipid Mediat 2019; 145:106377. [PMID: 31586592 DOI: 10.1016/j.prostaglandins.2019.106377] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 09/06/2019] [Accepted: 09/18/2019] [Indexed: 12/14/2022]
Abstract
Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection. Septic shock, the most common form of vasodilatory shock, is a subset of sepsis in which circulatory and cellular/metabolic abnormalities are severe enough to increase mortality. Inflammatory shock constitutes the hallmark of sepsis, but also a final common pathway of any form of severe long-term tissue hypoperfusion. The pathogenesis of inflammatory shock seems to be due to circulating substances released by pathogens (e.g., bacterial endotoxins) and host immuno-inflammatory responses (e.g., changes in the production of histamine, bradykinin, serotonin, nitric oxide [NO], reactive nitrogen and oxygen species, and arachidonic acid [AA]-derived eicosanoids mainly through NO synthase, cyclooxygenase, and cytochrome P450 [CYP] pathways, and proinflammatory cytokine formation). Therefore, refractory hypotension to vasoconstrictors with end-organ hypoperfusion is a life threatening feature of inflammatory shock. This review summarizes the current knowledge regarding the role of eicosanoids derived from CYP pathway of AA in animal models of inflammatory shock syndromes with an emphasis on septic shock in addition to potential therapeutic strategies targeting specific CYP isoforms responsible for proinflammatory/anti-inflammatory mediator production.
Collapse
Affiliation(s)
- Bahar Tunctan
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey.
| | - Sefika Pinar Senol
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | | | - Demet Sinem Guden
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Seyhan Sahan-Firat
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - John R Falck
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kafait U Malik
- Department of Pharmacology, College of Medicine, University of Tennessee, Center for Health Sciences, Memphis, TN, USA
| |
Collapse
|
13
|
Chithra MA, Ijinu TP, Kharkwal H, Sharma RK, Pushpangadan P, George V. Phenolic rich Cocos nucifera inflorescence extract ameliorates inflammatory responses in LPS-stimulated RAW264.7 macrophages and toxin-induced murine models. Inflammopharmacology 2019; 28:1073-1089. [DOI: 10.1007/s10787-019-00620-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 07/13/2019] [Indexed: 10/26/2022]
|
14
|
Yang S, Lee BS, Lee C, Ku SK, Bae JS. Anti-Septic Effects of Cudratricusxanthone A Against HMGB1-Mediated Septic Responses. Nat Prod Commun 2019. [DOI: 10.1177/1934578x19856793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- Sumin Yang
- College of Pharmacy CMRI, Research Institute of Pharmaceutical Sciences, BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, Kyungpook National University, Daegu, Republic of Korea
| | - Bong-Seon Lee
- College of Pharmacy CMRI, Research Institute of Pharmaceutical Sciences, BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, Kyungpook National University, Daegu, Republic of Korea
| | - Changhun Lee
- College of Pharmacy CMRI, Research Institute of Pharmaceutical Sciences, BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, Kyungpook National University, Daegu, Republic of Korea
| | - Sae-Kwang Ku
- Department of Anatomy and Histology, College of Korean Medicine, Daegu Haany University, Gyeongsan, Republic of Korea
| | - Jong-Sup Bae
- College of Pharmacy CMRI, Research Institute of Pharmaceutical Sciences, BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
15
|
Yang S, Lee W, Lee BS, Lee C, Park EK, Ku SK, Bae JS. Aloin Reduces HMGB1-Mediated Septic Responses and Improves Survival in Septic Mice by Activation of the SIRT1 and PI3K/Nrf2/HO-1 Signaling Axis. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2019; 47:613-633. [PMID: 30966773 DOI: 10.1142/s0192415x19500320] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
High mobility group box 1 (HMGB1) is recognized as a late mediator of sepsis, and the inhibition of HMGB1 release and recovery of vascular barrier integrity have emerged as attractive therapeutic strategies for the management of sepsis. We tested the hypothesis that aloin induces sirtuin 1 (SIRT1) and heme oxygenase (HO)-1, which inhibit HMGB1 release in lipopolysaccharide (LPS)-stimulated cells, thereby inhibiting HMGB1-induced hyperpermeability and increasing the survival of septic mice. Aloin was administered after LPS or HMGB1 challenge, and the antiseptic activity of aloin was determined from measurements of permeability, activation of pro-inflammatory proteins and production of markers for tissue injury in HMGB1-activated human umbilical vein endothelial cells (HUVECs) and a cecal ligation and puncture (CLP)-induced sepsis mouse model. Aloin significantly reduced HMGB1 release in LPS-activated HUVECs via SIRT1-mediated HMGB1 deacetylation and the PI3K/Nrf2/heme oxygenase (HO)-1 signaling axis. Aloin also suppressed the production of tumor necrosis factor (TNF)- α and interleukin (IL)-6, as well as the activation of nuclear factor (NF)- κ B and extracellular signal-regulated kinase 1/2 (ERK 1/2) by HMGB1. Moreover, aloin restored HMGB1-mediated vascular disruption and inhibited vascular hyperpermeability in mice. In addition, treatment with aloin reduced the CLP-induced release of HMGB1, sepsis-related mortality and tissue injury in vivo. Our results suggest that aloin reduces HMGB1 release and sepsis-related mortality by activating SIRT1 and PI3K/Nrf2/HO-1 signals, indicating that aloin has potential for the treatment of sepsis.
Collapse
Affiliation(s)
- Sumin Yang
- * College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, BK21 Plus KNU Multi-Omics based Creative Drug Research Team, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Wonhwa Lee
- * College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, BK21 Plus KNU Multi-Omics based Creative Drug Research Team, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Bong-Seon Lee
- * College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, BK21 Plus KNU Multi-Omics based Creative Drug Research Team, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Changhun Lee
- * College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, BK21 Plus KNU Multi-Omics based Creative Drug Research Team, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Eui Kyun Park
- † Department of Pathology and Regenerative Medicine, School of Dentistry, Kyungpook National University, Daegu 41940, Republic of Korea
| | - Sae-Kwang Ku
- ‡ Department of Histology and Anatomy, College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Republic of Korea
| | - Jong-Sup Bae
- * College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, BK21 Plus KNU Multi-Omics based Creative Drug Research Team, Kyungpook National University, Daegu 41566, Republic of Korea
| |
Collapse
|
16
|
Lee BS, Lee C, Yang S, Park EK, Ku SK, Bae JS. Suppressive effects of pelargonidin on lipopolysaccharide-induced inflammatory responses. Chem Biol Interact 2019; 302:67-73. [DOI: 10.1016/j.cbi.2019.02.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 02/06/2019] [Accepted: 02/08/2019] [Indexed: 12/20/2022]
|
17
|
Lee W, Yuseok O, Yang S, Lee BS, Lee JH, Park EK, Baek MC, Song GY, Bae JS. JH-4 reduces HMGB1-mediated septic responses and improves survival rate in septic mice. J Cell Biochem 2018; 120:6277-6289. [PMID: 30378167 DOI: 10.1002/jcb.27914] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 09/27/2018] [Indexed: 12/12/2022]
Abstract
Inhibition of high mobility group box 1 (HMGB1) and restoration of endothelial integrity are emerging as attractive therapeutic strategies for the management of severe vascular inflammatory diseases. Recently, we found that JH-4, a synthesized decursin derivative, exhibited a strong anti-Hutchinson-Gilford progeria syndrome by efficiently blocking progerin-lamin A/C binding. In this study, we examined the effects of JH-4 on HMGB1-mediated septic responses and the survival rate in a mouse sepsis model. The anti-inflammatory activities of JH-4 were monitored based on its effects on lipopolysaccharide- or cecal ligation and puncture (CLP)-mediated release of HMGB1. The antiseptic activities of JH-4 were determined by measuring permeability, leukocyte adhesion, migration, and the activation of proinflammatory proteins in HMGB1-activated human umbilical vein endothelial cells and mice. JH-4 inhibited the release of HMGB1 and downregulated HMGB1-dependent inflammatory responses in human endothelial cells. JH-4 also inhibited HMGB1-mediated hyperpermeability and leukocyte migration in mice. In addition, treatment with JH-4 reduced CLP-induced release of HMGB1, sepsis-related mortality, and pulmonary injury in vivo. Our results indicate that JH-4 is a possible therapeutic agent to treat various severe vascular inflammatory diseases via the inhibition of the HMGB1 signaling pathway.
Collapse
Affiliation(s)
- Wonhwa Lee
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - O Yuseok
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Sumin Yang
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, BK21 Plus KNU Multi-Omics based Creative Drug Research Team, Kyungpook National University, Daegu, Republic of Korea
| | - Bong-Seon Lee
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, BK21 Plus KNU Multi-Omics based Creative Drug Research Team, Kyungpook National University, Daegu, Republic of Korea
| | | | - Eui Kyun Park
- Department of Pathology and Regenerative Medicine, School of Dentistry, Kyungpook National University, Daegu, Republic of Korea
| | - Moon-Chang Baek
- Department of Molecular Medicine, CMRI, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Gyu-Yong Song
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea.,AREZ Co. Ltd., Sejong, Republic of Korea
| | - Jong-Sup Bae
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, BK21 Plus KNU Multi-Omics based Creative Drug Research Team, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
18
|
Lee IC, Kim DY, Bae JS. Sulforaphane Reduces HMGB1-Mediated Septic Responses and Improves Survival Rate in Septic Mice. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2017; 45:1253-1271. [PMID: 28830206 DOI: 10.1142/s0192415x17500690] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Sulforaphane (SFN), a natural isothiocyanate present in cruciferous vegetables such as broccoli and cabbage, is effective in preventing carcinogenesis, diabetes, and inflammatory responses. Inhibition of high mobility group box 1 (HMGB1) and restoration of endothelial integrity is emerging as an attractive therapeutic strategy in the management of severe sepsis or septic shock. In this study, we examined the effects of SFN on HMGB1-mediated septic responses and survival rate in a mouse sepsis model. The anti-inflammatory activities of SFN were monitored based on its effects on lipopolysaccharide (LPS)- or cecal ligation and puncture (CLP)-mediated release of HMGB1. The antiseptic activities of SFN were determined by measuring permeability, leukocyte adhesion and migration, and the activation of pro-inflammatory proteins in HMGB1-activated human umbilical vein endothelial cells (HUVECs) and mice. SFN inhibited the release of HMGB1 and downregulated HMGB1-dependent inflammatory responses in human endothelial cells. SFN also inhibited HMGB1-mediated hyperpermeability and leukocyte migration in mice. In addition, treatment with SFN reduced CLP-induced release of HMGB1 and sepsis-related mortality and pulmonary injury in vivo. Our results indicate that SFN is a possible therapeutic agent that can be used to treat various severe vascular inflammatory diseases via the inhibition of the HMGB1 signaling pathway.
Collapse
Affiliation(s)
- In-Chul Lee
- * Department of Cosmetic Science and Technology, Seowon University, Cheongju 28674, Republic of Korea
| | - Dae Yong Kim
- † Department of Biology Education, Seowon University, Cheongju 28674, Republic of Korea
| | - Jong-Sup Bae
- ‡ College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, Kyungpook National University, Daegu 41566, Republic of Korea
| |
Collapse
|
19
|
Zhang J, Liu H, Li S, Wu J, Sun J. SERCA1 attenuates diaphragm relaxation and uptake rate of SERCA in rats with acute sepsis. Mol Med Rep 2017; 16:5015-5022. [PMID: 28765908 DOI: 10.3892/mmr.2017.7134] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 06/15/2017] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to investigate the effects of acute sepsis on diaphragm contractility and relaxation, via examining the Ca2+‑uptake function of sarco/endoplasmic reticulum Ca2+ adenosine triphosphatase (SERCA), and the protein levels of SERCA1, SERCA2 and the ryanodine receptor (RyR) of the sarcoplasmic reticulum (SR). A sepsis rat model was established through cecal ligation and puncture (CLP). A total of 6 and 12 h following CLP, the isometric contractile and relaxation parameters of the diaphragm were measured. In addition, Ca2+ uptake and release from the SR, and the protein expression levels of SERCA1, SERCA2 and RyR in diaphragm muscle tissue were investigated. At 6 and 12 h post‑CLP, the diaphragm half‑relaxation time was prolonged and the maximum rate of tension decline was decreased and the Ca2+‑uptake function of SERCA was markedly reduced. The maximum rate of twitch force development, the maximal twitch and tetanic tension, and the release function of SR were decreased at 12 h post‑CLP. A total of 12 h following CLP, the protein expression levels of SERCA1 were significantly downregulated, and its activity was significantly reduced; conversely, the protein levels of SERCA2 remained unaltered. The present findings indicated that at the acute stage of sepsis induced by CLP the contractile and relaxation functions of the diaphragm were significantly compromised. The impairments in relaxation may be a result of the impaired uptake function of the SR and the downregulation in SERCA1 protein expression. Conversely, the compromised contractility may be a result of the impaired release function of the SR and the downregulation in RyR protein levels. This could provide some new insights into the treatment of sepsis. In acute stages of sepsis, the improvement of SERCA function could reduce the disequilibrium of calcium homeostasis to improve the critical illness myopathy and respiratory failure.
Collapse
Affiliation(s)
- Jianyou Zhang
- Department of Anesthesiology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu 225000, P.R. China
| | - Hui Liu
- Department of Endocrinology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu 225000, P.R. China
| | - Shitong Li
- Department of Anesthesiology, Shanghai General Hospital of Nanjing Medical University, Shanghai 200080, P.R. China
| | - Jin Wu
- Department of Anesthesiology, Shanghai General Hospital of Nanjing Medical University, Shanghai 200080, P.R. China
| | - Jianhong Sun
- Department of Anesthesiology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu 225000, P.R. China
| |
Collapse
|
20
|
Lee IC, Kim J, Bae JS. Anti-inflammatory effects of dabrafenib in vitro and in vivo. Can J Physiol Pharmacol 2017; 95:697-707. [DOI: 10.1139/cjpp-2016-0519] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The screening of bioactive compound libraries can be an effective approach for repositioning FDA-approved drugs or discovering new treatments for human diseases (drug repositioning). Drug repositioning refers to the development of existing drugs for new indications. Dabrafenib (DAB) is a B-Raf inhibitor and initially used for the treatment of metastatic melanoma therapy. Here, we tested the possible use of DAB in the treatment of lipopolysaccharide (LPS)-mediated vascular inflammatory responses. The anti-inflammatory activities of DAB were determined by measuring permeability, neutrophils adhesion and migration, and activation of pro-inflammatory proteins in LPS-activated human umbilical vein endothelial cells (HUVECs) and mice. We found that DAB inhibited LPS-induced barrier disruption, expression of cell adhesion molecules (CAMs), and adhesion and transendothelial migration of neutrophils to human endothelial cells. DAB also suppressed LPS-induced hyperpermeability and leukocytes migration in vivo. Furthermore, DAB suppressed the production of tumor necrosis factor-α (TNF-α) or interleukin (IL)-6 and the activation of nuclear factor-κB (NF-κB) or extracellular regulated kinases (ERK) 1/2 by LPS. Moreover, treatment with DAB resulted in reduced LPS-induced lethal endotoxemia. These results suggest that DAB possesses anti-inflammatory functions by inhibiting hyperpermeability, expression of CAMs, and adhesion and migration of leukocytes, thereby endorsing its usefulness as a therapy for vascular inflammatory diseases.
Collapse
Affiliation(s)
- In-Chul Lee
- Department of Cosmetic Science and Technology, Seowon University, Cheongju 28674, Republic of Korea
| | - Jongdoo Kim
- Cancer Control Team, Gachon University Gil Medical Center, Incheon 21565, Republic of Korea
| | - Jong-Sup Bae
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, BK21 Plus KNU Multi-Omics based Creative Drug Research Team, Kyungpook National University, Daegu 41566, Republic of Korea
| |
Collapse
|
21
|
Jung B, Kang H, Lee W, Noh HJ, Kim YS, Han MS, Baek MC, Kim J, Bae JS. Anti-septic effects of dabrafenib on HMGB1-mediated inflammatory responses. BMB Rep 2017; 49:214-9. [PMID: 26592934 PMCID: PMC4915240 DOI: 10.5483/bmbrep.2016.49.4.220] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Indexed: 12/26/2022] Open
Abstract
A nucleosomal protein, high mobility group box 1 (HMGB1) is known to be a late mediator of sepsis. Dabrafenib is a B-Raf inhibitor and initially used for the treatment of metastatic melanoma therapy. Inhibition of HMGB1 and renewal of vascular integrity is appearing as an engaging therapeutic strategy in the administration of severe sepsis or septic shock. Here, we examined the effects of dabrafenib (DAB) on the modulation of HMGB1-mediated septic responses. DAB inhibited the release of HMGB1 and downregulated HMGB1-dependent inflammatory responses by enhancing the expressions of cell adhesion molecules (CAMs) in human endothelial cells. In addition, treatment with DAB inhibited the HMGB1 secretion by CLP and sepsis-related mortality and pulmonary injury. This study demonstrated that DAB could be alternative therapeutic options for sepsis or septic shock via the inhibition of the HMGB1 signaling pathway. [BMB Reports 2016; 49(4): 214-219].
Collapse
Affiliation(s)
- Byeongjin Jung
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea
| | - Hyejin Kang
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea
| | - Wonhwa Lee
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566; Department of Biochemistry and Cell Biology, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Hyun Jin Noh
- Department of Biochemistry and Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 16499, Korea
| | - You-Sun Kim
- Department of Biochemistry and Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 16499, Korea
| | - Min-Su Han
- Laboratory for Arthritis and Bone Biology, Fatima Research Institute, Fatima Hospital, Daegu 41199, Korea
| | - Moon-Chang Baek
- Department of Molecular Medicine, CMRI, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Jaehong Kim
- Department of Biochemistry, School of Medicine, Gachon University, Incheon 21999, Korea
| | - Jong-Sup Bae
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea
| |
Collapse
|
22
|
Anti-septic effects of pelargonidin on HMGB1-induced responses in vitro and in vivo. Arch Pharm Res 2016; 39:1726-1738. [DOI: 10.1007/s12272-016-0834-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 09/07/2016] [Indexed: 10/20/2022]
|
23
|
Yang C, Liu H, Li X, Sui S, Liu Y. Protective effect of recombinant Trichinella 53-kDa protein in sepsis and the effect on macrophages. Exp Ther Med 2016; 12:965-968. [PMID: 27446304 PMCID: PMC4950598 DOI: 10.3892/etm.2016.3354] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 05/17/2016] [Indexed: 11/07/2022] Open
Abstract
The survival rate of the recombinant Trichinella 53-kDa protein infected by the cecal ligation and puncture (CLP) model of sepsis in rats and the expression difference of macrophages were analyzed. Eighteen clean SD rats were selected for the present study. The rats were divided into the sham operation (n=5), control (n=5) and experimental (n=8) groups. The rats in the sham operation group underwent cecum division and suture with routine therapy for cure. The rats in the control and experimental groups were placed in the CLP model of sepsis in rats. The experimental group was administered recombinant Trichinella 53-kDa protein in advance, and the control group was administered the same dose of placebo. The survival rate of the rats within 6 and 12 h, the macrophage expression ratio, and the differences of the expression levels Th1-type cytokines IFN-γ and tumor necrosis factor (TNF)-α, and the Th2 type cytokines interleukin-4 (IL-4) and IL-13 were analyzed. The survival rate of rats in the experimental group was higher than that of the control group with a statistically significant difference (P<0.05). The expression ratio of the macrophages received from the different handling methods of the rats in the experimental group was higher than that of the control group. The difference was statistically significant (P<0.05). The expression levels of the Th1-type cytokines IFN-γ and TNF-α of the rats in the experimental group was higher than that of the control group, while the expression level of the Th2-type cytokines IL-4 and IL-13 was higher than that of the control group. The difference was statistically significant (P<0.05). In conclusion, recombinant Trichinella 53-kDa protein can increase the survival rate following infection with CLP sepsis, which may be associated with the improvement of the macrophages and the adjustment of the expression of Th2 cytokines.
Collapse
|
24
|
Lee W, Ku SK, Bae JS. Anti-inflammatory effects of Baicalin, Baicalein, and Wogonin in vitro and in vivo. Inflammation 2015; 38:110-25. [PMID: 25249339 DOI: 10.1007/s10753-014-0013-0] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Here, three structurally related polyphenols found in the Chinese herb Huang Qui, namely baicalin, baicalein, and wogonin, were examined for its effects on inflammatory responses by monitoring the effects of baicalin, baicalein, and wogonin on lipopolysaccharide (LPS)-mediated vascular inflammatory responses. We found that each compound inhibited LPS-induced barrier disruption, expression of cell adhesion molecules (CAMs), and adhesion/transendothelial migration of monocytes to human endothelial cells. Each compound induced potent inhibition of phorbol-12-myristate 13-acetate and LPS-induced endothelial cell protein C receptor shedding. It also suppressed LPS-induced hyperpermeability and leukocytes migration in vivo. Furthermore, each compound suppressed the production of tumor necrosis factor-α or interleukin-6 and the activation of nuclear factor-κB or extracellular regulated kinases 1/2 by LPS. Moreover, treatment with each compound resulted in reduced LPS-induced lethal endotoxemia. These results suggest that baicalin, baicalein, and wogonin posses anti-inflammatory functions by inhibiting hyperpermeability, expression of CAMs, and adhesion and migration of leukocytes, thereby endorsing its usefulness as a therapy for vascular inflammatory diseases.
Collapse
Affiliation(s)
- Wonhwa Lee
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, 80 Dahak-ro, Buk-gu, Daegu, 702-701, Republic of Korea
| | | | | |
Collapse
|
25
|
Abstract
Inflammation is a response to traumatic, infectious, post-ischemic, toxic, or autoimmune injury. However, uncontrolled inflammation can lead to disease, and inflammation is now believed to be responsible for several disease conditions. Research in our laboratory has shown that hydrogen sulfide (H2S) acts as a novel mediator of inflammation. At present, work in several research groups worldwide is focused on determining the role of H2S in inflammation. H2S has been implicated in different inflammatory conditions. Most of this research involved working with animal models of disease and in vitro systems. Recent research, however, points to a role of H2S in clinical inflammatory disease as well. This chapter describes our current understanding of the role of H2S in inflammation.
Collapse
Affiliation(s)
- Madhav Bhatia
- Department of Pathology, University of Otago, Christchurch, 2 Riccarton Avenue, 4345, Christchurch, 8140, New Zealand,
| |
Collapse
|
26
|
Anti-inflammatory effects of methylthiouracil in vitro and in vivo. Toxicol Appl Pharmacol 2015; 288:374-86. [DOI: 10.1016/j.taap.2015.08.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 07/25/2015] [Accepted: 08/14/2015] [Indexed: 01/11/2023]
|
27
|
Anti-inflammatory effects of vicenin-2 and scolymoside in vitro and in vivo. Inflamm Res 2015; 64:1005-21. [DOI: 10.1007/s00011-015-0886-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 09/28/2015] [Accepted: 10/06/2015] [Indexed: 12/21/2022] Open
|
28
|
Micro-concentration Lipopolysaccharide as a Novel Stimulator of Megakaryocytopoiesis that Synergizes with IL-6 for Platelet Production. Sci Rep 2015; 5:13748. [PMID: 26330186 PMCID: PMC4557119 DOI: 10.1038/srep13748] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 08/04/2015] [Indexed: 01/07/2023] Open
Abstract
Lipopolysaccharide (LPS) induces platelet activation and enhances platelet sensitivity to aggregation, which might alter platelet counts. We found that serial doses of micro-concentration LPS significantly increased the platelet count in mice treated with kanamycin, along with increased expression of IL-6 compared with IL-3 and TPO in megakaryocytes obtained from the mouse bone morrow following LPS administration. Furthermore, LPS at lower levels ranging plus IL-6 effectively stimulated CFU-MK formation and increased CD41 expression and megakaryocyte polyploidization. Meanwhile, there was a sustained rise in the percentage of reticulated platelets in the whole blood in response to low-dosage LPS combined with IL-6. In vivo experiments also demonstrated that the administration of LPS combined with IL-6 substantially enhanced the number of circulating platelets in normal and thrombocytopenic mice. Notably, the optimal LPS concentration in combination with IL-6 might be a novel stimulator of TLR4 and IL-6R expression in Dami cell lines, which initially occurs through TLR4-IL-6R crosstalk and then involves the activation of NF-κB and phosphorylation of p38 MAPK. These data suggest a new paradigm for the regulation of megakaryocytopoiesis and platelet production via a synergistic effect of LPS and IL-6, which has the potential to be used for the design of new therapies.
Collapse
|
29
|
Kwak S, Ku SK, Kang H, Baek MC, Bae JS. Methylthiouracil, a new treatment option for sepsis. Vascul Pharmacol 2015; 88:1-10. [PMID: 26239884 DOI: 10.1016/j.vph.2015.07.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 06/17/2015] [Accepted: 07/07/2015] [Indexed: 11/28/2022]
Abstract
The screening of bioactive compound libraries can be an effective approach for repositioning FDA-approved drugs or discovering new treatments for human diseases. Inhibition of high mobility group box 1 (HMGB1) and restoration of endothelial integrity are emerging as an attractive therapeutic strategies in the management of severe sepsis or septic shock. Here, we examined the effects of methylthiouracil (MTU), used as antithyroid drug, by monitoring the effects on lipopolysaccharide (LPS)- or cecal ligation and puncture (CLP)-mediated release of HMGB1, and on the modulation of HMGB1-mediated inflammatory responses. The anti-inflammatory activities of MTU were determined by measuring permeability, leukocyte adhesion and migration, and the activation of pro-inflammatory proteins in HMGB1-activated HUVECs and mice. MTU inhibited the release of HMGB1 and downregulated HMGB1-dependent inflammatory responses in human endothelial cells. MTU also inhibited HMGB1-mediated hyperpermeability and leukocyte migration in mice. In addition, treatment with MTU reduced CLP-induced release of HMGB1 and sepsis-related mortality and pulmonary injury. Our results indicate that MTUs could be candidate therapeutic agents for various severe vascular inflammatory diseases via the inhibition of the HMGB1 signaling pathway.
Collapse
Affiliation(s)
- Soyoung Kwak
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 702-701, Republic of Korea
| | - Sae-Kwang Ku
- Department of Anatomy and Histology, College of Korean Medicine, Daegu Haany University, Gyeongsan 712-715, Republic of Korea
| | - Hyejin Kang
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 702-701, Republic of Korea
| | - Moon-Chang Baek
- Department of Molecular Medicine, CMRI, School of Medicine, Kyungpook National University, Daegu 700-422, Republic of Korea.
| | - Jong-Sup Bae
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 702-701, Republic of Korea.
| |
Collapse
|
30
|
Abstract
Sepsis is a state of disrupted inflammatory homeostasis that is initiated by infection. High mobility group box 1 (HMGB1) protein acting as a late mediator of severe vascular inflammatory conditions, such as sepsis and endothelial cell protein C receptor (EPCR), is involved in vascular inflammation. Fisetin, an active compound from the family Fabaceae, was reported to have antiviral, neuroprotective, and anti-inflammatory activities. Here, we determined the anti-septic effects of fisetin on HMGB1-mediated inflammatory responses and on the shedding of EPCR in vitro and in vivo, for the first time. First, we monitored the effects of post-treatment fisetin on lipopolysaccharide (LPS) and cecal ligation and puncture (CLP)-mediated release of HMGB1 and HMGB1-mediated regulation of pro-inflammatory responses in human umbilical vein endothelial cells (HUVECs) and septic mice. Post-treatment fisetin was found to suppress LPS-mediated release of HMGB1 and HMGB1-mediated cytoskeletal rearrangements. Fisetin also inhibited HMGB1-mediated hyperpermeability and leukocyte migration in septic mice. Fisetin induced potent inhibition of phorbol-12-myristate 13-acetate (PMA) and CLP-induced EPCR. Fisetin also inhibited the expression and activity of tumor necrosis factor-α converting enzyme, induced by PMA in endothelial cells. In addition, fisetin inhibited the production of tumor necrosis factor-α and the activation of AKT, nuclear factor-κB, and extracellular regulated kinases 1/2 by HMGB1 in HUVECs. Fisetin also down-regulated CLP-induced release of HMGB1, production of interleukin 1β, and reduced septic mortality. Collectively, these results suggest that fisetin may be a candidate therapeutic agent for the treatment of vascular inflammatory diseases via inhibition of the HMGB1 signaling pathway.
Collapse
|
31
|
Lee W, Ku SK, Park EJ, Na DH, Kim KM, Bae JS. Exendin-4 inhibits HMGB1-induced inflammatory responses in HUVECs and in murine polymicrobial sepsis. Inflammation 2015; 37:1876-88. [PMID: 24826914 DOI: 10.1007/s10753-014-9919-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Exendin-4 (EX4) has been reported to attenuate myocardial ischemia and reperfusion (I/R) injury and inflammatory and oxidative responses. Nuclear DNA-binding protein high-mobility group box 1 (HMGB1) protein acts as a late mediator of severe vascular inflammatory conditions. However, the effect of EX4 on HMGB1-induced inflammatory response has not been studied. First, we accessed this question by monitoring the effects of posttreatment EX4 on lipopolysaccharide (LPS) and cecal ligation and puncture (CLP)-mediated release of HMGB1 and HMGB1-mediated regulation of proinflammatory responses in human umbilical vein endothelial cells (HUVECs) and septic mice. Posttreatment EX4 was found to suppress LPS-mediated release of HMGB1 and inhibited HMGB1-mediated hyperpermeability and leukocyte migration in septic mice. EX4 also induced downregulation of CLP-induced release of HMGB1, production of IL-6, and mortality. Collectively, these results suggest that EX4 may be regarded as a candidate therapeutic agent for treatment of vascular inflammatory diseases via inhibition of the HMGB1 signaling pathway.
Collapse
Affiliation(s)
- Wonhwa Lee
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 702-701, Republic of Korea
| | | | | | | | | | | |
Collapse
|
32
|
Lee W, Yoon EK, Kim KM, Park DH, Bae JS. Antiseptic effect of vicenin-2 and scolymoside from Cyclopia subternata (honeybush) in response to HMGB1 as a late sepsis mediator in vitro and in vivo. Can J Physiol Pharmacol 2015; 93:709-20. [PMID: 26243020 DOI: 10.1139/cjpp-2015-0021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cyclopia subternata is a medicinal plant commonly used in traditional medicine to relieve pain. In this study, we investigated the antiseptic effects and underlying mechanisms of vicenin-2 and scolymoside, which are 2 active compounds from C. subternata that act against high mobility group box 1 (HMGB1)-mediated septic responses in human umbilical vein endothelial cells (HUVECs) and mice. The antiseptic activities of vicenin-2 and scolymoside were determined by measuring permeability, neutrophil adhesion and migration, and activation of proinflammatory proteins in HMGB1-activated HUVECs and mice. According to the results, vicenin-2 and scolymoside effectively inhibited lipopolysaccharide-induced release of HMGB1, and suppressed HMGB1-mediated septic responses such as hyperpermeability, the adhesion and migration of leukocytes, and the expression of cell adhesion molecules. In addition, vicenin-2 and scolymoside suppressed the production of tumor necrosis factor-α and interleukin 6, and activation of nuclear factor-κB and extracellular regulated kinases 1/2 by HMGB1. Collectively, these results indicate that vicenin-2 and scolymoside could be a potential therapeutic agents for the treatment of various severe vascular inflammatory diseases via inhibition of the HMGB1 signaling pathway.
Collapse
Affiliation(s)
- Wonhwa Lee
- a College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, 80 Dahak-ro, Buk-gu, Daegu 702-701, Republic of Korea.,b Department of Biochemistry and Cell Biology, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu 702-701, Republic of Korea
| | - Eun-Kyung Yoon
- a College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, 80 Dahak-ro, Buk-gu, Daegu 702-701, Republic of Korea
| | - Kyung-Min Kim
- c Division of Plant Biosciences, School of Applied BioSciences, College of Agriculture and Life Science, Kyungpook National University, Daegu 702-701, Republic of Korea
| | - Dong Ho Park
- d Department of Ophthalmology, School of Medicine, Kyungpook National University, 50 Samduk-dong-2-ga, Jung-gu, Daegu 700-721, Republic of Korea
| | - Jong-Sup Bae
- a College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, 80 Dahak-ro, Buk-gu, Daegu 702-701, Republic of Korea
| |
Collapse
|
33
|
Lee W, Ku SK, Na DH, Bae JS. Anti-Inflammatory Effects of Lysozyme Against HMGB1 in Human Endothelial Cells and in Mice. Inflammation 2015; 38:1911-24. [DOI: 10.1007/s10753-015-0171-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
34
|
Cordioli RL, Cordioli E, Negrini R, Silva E. Sepsis and pregnancy: do we know how to treat this situation? Rev Bras Ter Intensiva 2015; 25:334-44. [PMID: 24553516 PMCID: PMC4031877 DOI: 10.5935/0103-507x.20130056] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 11/21/2013] [Indexed: 12/18/2022] Open
Abstract
Sepsis is defined as an acute inflammatory response syndrome secondary to an
infectious focus. It has a high incidence, morbidity and mortality, causing
substantial financial costs, especially due to complications such as septic shock and
multiple organ dysfunction. The pathogen toxins associated with individual
susceptibility culminate with cytokine release, which promotes a systemic
inflammatory response that can progress to multiple organ dysfunction and eventual
patient death. Specifically, sepsis incidence, morbidity and mortality are lower in pregnant women,
as this group is typically younger with fewer comorbidities having a polymicrobial
etiology resulting in sepsis. Pregnant women exhibit physiological characteristics that may confer specific
clinical presentation and laboratory patterns during the sepsis course. Thus, a
better understanding of these changes is critical for better identification and
management of these patients. The presence of a fetus also requires unique approaches
in a pregnant woman with sepsis. Sepsis treatment is based on certain guidelines that were established after major
clinical trials, which, unfortunately, all classified pregnancy as a exclusion
criteria. Thus, the treatment of sepsis in the general population has been extrapolated to the
pregnant population, with the following main goals: maintenance of tissue perfusion
with fluid replacement and vasoactive drugs (initial resuscitation), adequate
oxygenation, control of the infection source and an early start of antibiotic
therapy, corticosteroid infusion and blood transfusion when properly indicated,
prophylaxis, and specifically monitoring and maintenance of fetal heath.
Collapse
Affiliation(s)
- Ricardo Luiz Cordioli
- Corresponding author: Ricardo Luiz Cordioli, Unidade de Terapia
Intensiva do Hospital Israelita Albert Einstein, Avenida Albert Einstein, 627/701,
5º Andar - Morumbi, Zip code: 05652-900 - São Paulo (SP), Brazil.
E-mail:
| | | | | | | |
Collapse
|
35
|
Lee W, Park EJ, Kwak S, Kim Y, Na DH, Bae JS. PEGylated lysozymes with anti-septic effects in human endothelial cells and in mice. Biochem Biophys Res Commun 2015; 459:662-7. [PMID: 25769950 DOI: 10.1016/j.bbrc.2015.02.167] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 02/28/2015] [Indexed: 11/19/2022]
Abstract
High mobility group box 1 (HMGB1) was recently shown to be an important extracellular mediator of severe vascular inflammatory disease, sepsis. Lysozyme (LYZ) has been shown to bind to bacterial lipopolysaccharide (LPS) and have a potential for playing a role in the therapy of inflammatory diseases. However, the effect of LYZ on HMGB1-induced septic response has not been investigated. Moreover, PEGylation effects on the antiseptic activity of LYZ are not known. Here, we show, for the first time, the anti-septic effects of PEGylated LYZ (PEG-LYZ) in HMGB1-mediated inflammatory responses in vitro and in vivo. Among four mono-PEGylated LYZs with different PEGylation sites (N-terminus, Lys(13), Lys(33), and Lys(97)), N-terminally PEGylated LYZ showed the highest activity. Subsequently, among three N-terminally PEGylated LYZs prepared with aldehyde-activated PEGs of 5, 10, and 20 kDa, 5 kDa-PEG-conjugated LYZ (P5-K(1)-LYZ) showed the highest antiseptic activity. The data showed that P5-K(1)-LYZ post-treatment effectively suppressed LPS-mediated release of HMGB1. P5-K(1)-LYZ also inhibited HMGB1-mediated hyperpermeability in human endothelial cells. Furthermore, P5-K(1)-LYZ reduced the cecal ligation and puncture (CLP)-induced release of HMGB1 and septic mortality. Collectively, these results suggest P5-K(1)-LYZ as a candidate therapeutic agent for the treatment of vascular inflammatory diseases via inhibition of the HMGB1 signaling pathway.
Collapse
Affiliation(s)
- Wonhwa Lee
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 702-701, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu 702-701, Republic of Korea
| | - Eun Ji Park
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 702-701, Republic of Korea
| | - Soyoung Kwak
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 702-701, Republic of Korea
| | - Yejin Kim
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 702-701, Republic of Korea
| | - Dong Hee Na
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 702-701, Republic of Korea.
| | - Jong-Sup Bae
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 702-701, Republic of Korea.
| |
Collapse
|
36
|
Lee W, Bae JS. Anti-inflammatory Effects of Aspalathin and Nothofagin from Rooibos (Aspalathus linearis) In Vitro and In Vivo. Inflammation 2015; 38:1502-16. [DOI: 10.1007/s10753-015-0125-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
37
|
Zhu L, Meng Q, Liang S, Ma Y, Li R, Li G, Zeng H. The transcription factor GFI1 negatively regulates NLRP3 inflammasome activation in macrophages. FEBS Lett 2015; 588:4513-9. [PMID: 25447538 DOI: 10.1016/j.febslet.2014.10.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 10/13/2014] [Accepted: 10/22/2014] [Indexed: 12/13/2022]
Abstract
Interleukin-1β (IL-1β) secretion downstream of Toll-like receptor (TLR) activation is tightly controlled at the transcriptional and post-translational levels. NLRP3 inflammasome is involved in the maturation of pro-IL-1β, with NLRP3 expression identified as the limiting factor for inflammasome activation. Previously, we had demonstrated that the zinc-finger protein GFI1 inhibits pro-IL-1β transcription. Here, we show that GFI1 inhibits NLRP3 inflammasome activation and IL-1β secretion in macrophages. GFI1 suppressed Nlrp3 transcription via two mechanisms: (1) by binding to the Gli-responsive element 1 (GRE1) in the Nlrp3 promoter; and (2) by antagonizing the nuclear factor-κB (NF-κB) transcriptional activity. Thus, GFI1 negatively regulates TLR-mediated IL-1β production at both transcriptional and post-translational levels.
Collapse
Affiliation(s)
- Liuluan Zhu
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
38
|
Filgueiras LR, Brandt SL, Wang S, Wang Z, Morris DL, Evans-Molina C, Mirmira RG, Jancar S, Serezani CH. Leukotriene B4-mediated sterile inflammation promotes susceptibility to sepsis in a mouse model of type 1 diabetes. Sci Signal 2015; 8:ra10. [PMID: 25628460 DOI: 10.1126/scisignal.2005568] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Type 1 diabetes mellitus (T1DM) is associated with chronic systemic inflammation and enhanced susceptibility to systemic bacterial infection (sepsis). We hypothesized that low insulin concentrations in T1DM trigger the enzyme 5-lipoxygenase (5-LO) to produce the lipid mediator leukotriene B4 (LTB4), which triggers systemic inflammation that may increase susceptibility to polymicrobial sepsis. Consistent with chronic inflammation, peritoneal macrophages from two mouse models of T1DM had greater abundance of the adaptor MyD88 (myeloid differentiation factor 88) and its direct transcriptional effector STAT-1 (signal transducer and activator of transcription 1) than macrophages from nondiabetic mice. Expression of Alox5, which encodes 5-LO, and the concentration of the proinflammatory cytokine interleukin-1β (IL-1β) were also increased in peritoneal macrophages and serum from T1DM mice. Insulin treatment reduced LTB4 concentrations in the circulation and Myd88 and Stat1 expression in the macrophages from T1DM mice. T1DM mice treated with a 5-LO inhibitor had reduced Myd88 mRNA in macrophages and increased abundance of IL-1 receptor antagonist and reduced production of IL-β in the circulation. T1DM mice lacking 5-LO or the receptor for LTB4 also produced less proinflammatory cytokines. Compared to wild-type or untreated diabetic mice, T1DM mice lacking the receptor for LTB4 or treated with a 5-LO inhibitor survived polymicrobial sepsis, had reduced production of proinflammatory cytokines, and had decreased bacterial counts. These results uncover a role for LTB4 in promoting sterile inflammation in diabetes and the enhanced susceptibility to sepsis in T1DM.
Collapse
Affiliation(s)
- Luciano Ribeiro Filgueiras
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Immunology Department, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508, Brazil
| | - Stephanie L Brandt
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Soujuan Wang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Zhuo Wang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - David L Morris
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Carmella Evans-Molina
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Raghavendra G Mirmira
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sonia Jancar
- Immunology Department, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508, Brazil
| | - C Henrique Serezani
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
39
|
Bae JS, Lee W, Son HN, Lee YM, Kim IS. Anti-transforming growth factor β-induced protein antibody ameliorates vascular barrier dysfunction and improves survival in sepsis. Acta Physiol (Oxf) 2014; 212:306-15. [PMID: 25219400 DOI: 10.1111/apha.12398] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 07/11/2014] [Accepted: 09/09/2014] [Indexed: 11/30/2022]
Abstract
AIM Sepsis is a systemic inflammatory response syndrome resulting from a microbial infection. Transforming growth factor β-induced protein (TGFBIp) is an extracellular matrix protein expressed by human endothelial cells and platelets that induces sepsis through interaction with integrin αvβ5. The aim of this study was to investigate the role of TGFBIp in vascular permeability and the underlying mechanisms using TGFBIp-neutralizing antibody. METHODS Mice were subjected to caecal ligation and puncture (CLP) with or without neutralizing anti-TGFBIp antibody (300 μg kg(-1), intravenously). Wild-type or integrin β5-null mice received TGFBIp (0.1 mg kg(-1), intravenously) or were subjected to CLP. Human umbilical vein endothelial cells were exposed to lipopolysaccharide (100 ng mL(-1)) with or without neutralizing anti-TGFBIp antibody (50 μg mL(-1)). RESULTS Administration of neutralizing anti-TGFBIp antibody in mice attenuated CLP-induced secretion of TGFBIp, leucocyte migration and vascular permeability and reduced septic mortality. Injected TGFBIp did not enhance vascular barrier permeability or leucocyte migration in β5-null mice. Finally, neutralizing anti-TGFBIp antibody inhibited the specific interactions between TGFBIp and its receptor, integrin αvβ5. CONCLUSION Our findings demonstrate that treatment with a TGFBIp-neutralizing antibody can ameliorate the deleterious effects of sepsis.
Collapse
Affiliation(s)
- J.-S. Bae
- College of Pharmacy; Research Institute of Pharmaceutical Sciences; Kyungpook National University; Daegu Korea
- BK21 Plus KNU Biomedical Convergence Program; Department of Biochemistry and Cell Biology; Cell and Matrix Research Institute; School of Medicine; Kyungpook National University; Daegu Korea
| | - W. Lee
- College of Pharmacy; Research Institute of Pharmaceutical Sciences; Kyungpook National University; Daegu Korea
- BK21 Plus KNU Biomedical Convergence Program; Department of Biochemistry and Cell Biology; Cell and Matrix Research Institute; School of Medicine; Kyungpook National University; Daegu Korea
| | - H.-N. Son
- BK21 Plus KNU Biomedical Convergence Program; Department of Biochemistry and Cell Biology; Cell and Matrix Research Institute; School of Medicine; Kyungpook National University; Daegu Korea
| | - Y.-M. Lee
- College of Pharmacy; Research Institute of Pharmaceutical Sciences; Kyungpook National University; Daegu Korea
| | - I.-S. Kim
- BK21 Plus KNU Biomedical Convergence Program; Department of Biochemistry and Cell Biology; Cell and Matrix Research Institute; School of Medicine; Kyungpook National University; Daegu Korea
- Biomedical Research Institute; Korea Institute of Science and Technology; Seoul Korea
| |
Collapse
|
40
|
Premkumar R, Phillips ARJ, Petrov MS, Windsor JA. The clinical relevance of obesity in acute pancreatitis: targeted systematic reviews. Pancreatology 2014; 15:25-33. [PMID: 25464938 DOI: 10.1016/j.pan.2014.10.007] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 09/26/2014] [Accepted: 10/15/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND With the rising prevalence of obesity, its impact on the severity and outcome of acute pancreatitis remains an important consideration when managing obese patients with acute pancreatitis. OBJECTIVE To determine the clinical relevance of obesity in acute pancreatitis. METHODS A series of clinically relevant questions were framed which formed the basis of our literature search using PubMed and EMBASE databases. These related to acute pancreatitis severity, systemic inflammatory response, mortality, local and systemic complications. The search was restricted to human studies. Studies were classified according to the Oxford Centre for Evidence Based Medicine levels of evidence 1 for prognostic studies. Obesity was defined according to the guidelines of the World Health Organization. In keeping with studies included the binary classification (mild and severe) of acute pancreatitis was used. RESULTS Obesity is associated with an amplified systemic inflammatory response in acute pancreatitis and is a prognostic factor for mortality, local, systemic complications and severity in acute pancreatitis. Obesity was not found to be an independent prognostic factor for mortality and organ failure in patients with acute pancreatitis. It was evident that further studies are required to determine whether incorporating obesity into existing scoring systems improves severity prediction. Emerging evidence suggests that an obesity paradox is present in patients with acute pancreatitis. CONCLUSION This review demonstrates that obesity has a clinically relevant impact on the course and outcome of acute pancreatitis.
Collapse
Affiliation(s)
- Rakesh Premkumar
- Surgical Center for Outcomes Research and Evaluation, Department of Surgery & Applied Surgery and Metabolism Laboratory, School of Biological Sciences, University of Auckland, Auckland, New Zealand.
| | - Anthony R J Phillips
- Surgical Center for Outcomes Research and Evaluation, Department of Surgery & Applied Surgery and Metabolism Laboratory, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Maxim S Petrov
- Surgical Center for Outcomes Research and Evaluation, Department of Surgery & Applied Surgery and Metabolism Laboratory, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - John A Windsor
- Surgical Center for Outcomes Research and Evaluation, Department of Surgery & Applied Surgery and Metabolism Laboratory, School of Biological Sciences, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
41
|
|
42
|
Lee W, Ku SK, Bae JS. Factor Xa inhibits HMGB1-induced septic responses in human umbilical vein endothelial cells and in mice. Thromb Haemost 2014; 112:757-69. [PMID: 25007770 DOI: 10.1160/th14-03-0233] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 05/12/2014] [Indexed: 12/24/2022]
Abstract
Nuclear DNA-binding protein high mobility group box 1 (HMGB1) acts as a late mediator of severe vascular inflammatory conditions, such as sepsis. Activated factor X (FXa) is an important player in the coagulation cascade responsible for thrombin generation, and it influences cell signalling in various cell types by activating protease-activated receptors (PARs). However, the effect of FXa on HMGB1-induced inflammatory response has not been studied. First, we addressed this issue by monitoring the effects of post-treatment with FXa on lipopolysaccharide (LPS)- and cecal ligation and puncture (CLP)-mediated release of HMGB1 and HMGB1-mediated regulation of pro-inflammatory responses in human umbilical vein endothelial cells (HUVECs) and septic mice. Post-treatment with FXa was found to suppress LPS-mediated release of HMGB1 and HMGB1-mediated cytoskeletal rearrangements. FXa also inhibited HMGB1-mediated hyperpermeability and leukocyte migration in septic mice. In addition, FXa inhibited the production of tumour necrosis factor-α and interleukin (IL)-1β. FXa also facilitated the downregulation of CLP-induced release of HMGB1, production of IL-6, and mortality. Collectively, these results suggest that FXa may be regarded as a candidate therapeutic agent for treating vascular inflammatory diseases by inhibiting the HMGB1 signalling pathway.
Collapse
Affiliation(s)
| | | | - Jong-Sup Bae
- Jong-Sup Bae, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 702-701, Republic of Korea, Tel.: +82 53 950 8570, Fax: +82 53 950 8557, E-mail:
| |
Collapse
|
43
|
Lee W, Ku SK, Bae JS. Vascular barrier protective effects of orientin and isoorientin in LPS-induced inflammation in vitro and in vivo. Vascul Pharmacol 2014; 62:3-14. [DOI: 10.1016/j.vph.2014.04.006] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 04/12/2014] [Accepted: 04/18/2014] [Indexed: 01/11/2023]
|
44
|
Lee W, Ku S, Yoo H, Song K, Bae J. Andrographolide inhibits HMGB1-induced inflammatory responses in human umbilical vein endothelial cells and in murine polymicrobial sepsis. Acta Physiol (Oxf) 2014; 211:176-87. [PMID: 24581270 DOI: 10.1111/apha.12264] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 12/06/2013] [Accepted: 02/24/2014] [Indexed: 01/18/2023]
Abstract
AIM Nuclear DNA-binding protein high-mobility group box 1 (HMGB1) protein acts as a late mediator of severe vascular inflammatory conditions, such as septic shock, upregulating pro-inflammatory cytokines. Andrographolide (AG) is isolated from the plant of Andrographis paniculata and used as a folk medicine for treatment of viral infection, diarrhoea, dysentery and fever. However, the effect of AG on HMGB1-induced inflammatory response has not been studied. METHODS Firstly, we accessed this question by monitoring the effects of post-treatment AG on lipopolysaccharide (LPS) and caecal ligation and puncture (CLP)-mediated release of HMGB1 and HMGB1-mediated regulation of pro-inflammatory responses in human umbilical vein endothelial cells (HUVECs) and septic mice. RESULTS Post-treatment AG was found to suppress LPS-mediated release of HMGB1 and HMGB1-mediated cytoskeletal rearrangements. AG also inhibited HMGB1-mediated hyperpermeability and leucocyte migration in septic mice. In addition, AG inhibited production of tumour necrosis factor-α (TNF-α) and activation of AKT, nuclear factor-κB (NF-κB) and extracellular-regulated kinases (ERK) 1/2 by HMGB1 in HUVECs. AG also induced downregulation of CLP-induced release of HMGB1, production of interleukin (IL) 1β/6/8 and mortality. CONCLUSION Collectively, these results suggest that AG may be regarded as a candidate therapeutic agent for the treatment of vascular inflammatory diseases via inhibition of the HMGB1 signalling pathway.
Collapse
Affiliation(s)
- W. Lee
- College of Pharmacy; CMRI; Research Institute of Pharmaceutical Sciences; Kyungpook National University; Daegu Korea
- Department of Biochemistry and Cell Biology; School of Medicine; Kyungpook National University; Daegu Korea
| | - S. Ku
- Department of Anatomy and Histology; College of Korean Medicine; Daegu Haany University; Gyeongsan Korea
| | - H. Yoo
- College of Pharmacy; CMRI; Research Institute of Pharmaceutical Sciences; Kyungpook National University; Daegu Korea
| | - K. Song
- College of Pharmacy; CMRI; Research Institute of Pharmaceutical Sciences; Kyungpook National University; Daegu Korea
| | - J. Bae
- College of Pharmacy; CMRI; Research Institute of Pharmaceutical Sciences; Kyungpook National University; Daegu Korea
| |
Collapse
|
45
|
Yoo H, Ku SK, Lee T, Bae JS. Orientin Inhibits HMGB1-Induced Inflammatory Responses in HUVECs and in Murine Polymicrobial Sepsis. Inflammation 2014; 37:1705-17. [DOI: 10.1007/s10753-014-9899-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
46
|
Deutschman CS. Therapy for sepsis: Einstein once said... Br J Anaesth 2013; 111:693-5. [PMID: 24108726 DOI: 10.1093/bja/aet157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- C S Deutschman
- Perelman School of Medicine at the University of Pennsylvania - Anesthesiology and Critical Care, Dulles 781A/HUP, 3400 Spruce Street, Philadelphia, PA 19104-4283, USA
| |
Collapse
|
47
|
Lee W, Ku SK, Min BW, Lee S, Jee JG, Kim JA, Bae JS. Vascular barrier protective effects of pellitorine in LPS-induced inflammation in vitro and in vivo. Fitoterapia 2013; 92:177-87. [PMID: 24262867 DOI: 10.1016/j.fitote.2013.11.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 11/06/2013] [Accepted: 11/09/2013] [Indexed: 12/22/2022]
Abstract
Pellitorine (PT), an active amide compound, is well known to possess insecticidal, antibacterial and anticancer properties. In this study, we first investigated the possible barrier protective effects of pellitorine against pro-inflammatory responses induced by lipopolysaccharide (LPS) and the associated signaling pathways in vitro and in vivo. The barrier protective activities of PT were determined by measuring permeability, monocyte adhesion and migration, and activation of pro-inflammatory proteins in LPS-activated human umbilical vein endothelial cells (HUVECs) and in mice. We found that PT inhibited LPS-induced barrier disruption, expression of cell adhesion molecules (CAMs) and adhesion/transendothelial migration of monocytes to human endothelial cells. PT also suppressed LPS-induced hyperpermeability and leukocyte migration in vivo. Further studies revealed that PT suppressed the production of tumor necrosis factor-α (TNF-α) or Interleukin (IL)-6 and activation of nuclear factor-κB (NF-κB) or extracellular regulated kinases (ERK) 1/2 by LPS. Moreover, treatment with PT resulted in reduced LPS-induced lethal endotoxemia. These results suggest that PT protects vascular barrier integrity by inhibiting hyperpermeability, expression of CAMs, and adhesion and migration of leukocytes, thereby endorsing its usefulness as a therapy for vascular inflammatory diseases.
Collapse
Affiliation(s)
- Wonhwa Lee
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 702-701, Republic of Korea; Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu 702-701, Republic of Korea
| | - Sae-Kwang Ku
- Department of Anatomy and Histology, College of Korean Medicine, Daegu Haany University, Gyeongsan 712-715, Republic of Korea
| | - Byung-Woon Min
- Department of BioMedical Clinical Pathology, Hanlyo University, Gwangyang 545-704, Republic of Korea
| | - Sangkyu Lee
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 702-701, Republic of Korea
| | - Jun-Goo Jee
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 702-701, Republic of Korea
| | - Jeong Ah Kim
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 702-701, Republic of Korea
| | - Jong-Sup Bae
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 702-701, Republic of Korea.
| |
Collapse
|
48
|
Abstract
The receptor-interacting protein kinase 3 (RIP3/RIPK3) has emerged as a critical regulator of programmed necrosis/necroptosis, an inflammatory form of cell death with important functions in pathogen-induced and sterile inflammation. RIP3 activation is tightly regulated by phosphorylation, ubiquitination, and caspase-mediated cleavage. These post-translational modifications coordinately regulate the assembly of a macromolecular signaling complex termed the necrosome. Recently, several reports indicate that RIP3 can promote inflammation independent of its pronecrotic activity. Here, we review our current understanding of the mechanisms that drive RIP3-dependent necrosis and its role in different inflammatory diseases.
Collapse
Affiliation(s)
- Kenta Moriwaki
- Department of Pathology, Immunology and Microbiology Program, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | | |
Collapse
|
49
|
Lee W, Yoo H, Kim JA, Lee S, Jee JG, Lee MY, Lee YM, Bae JS. Barrier protective effects of piperlonguminine in LPS-induced inflammation in vitro and in vivo. Food Chem Toxicol 2013; 58:149-57. [PMID: 23619565 DOI: 10.1016/j.fct.2013.04.027] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Revised: 03/27/2013] [Accepted: 04/12/2013] [Indexed: 01/04/2023]
Abstract
Piperlonguminine (PL), an important component of Piper longum fruits, is well known to possess potent anti-hyperlipidemic, anti-platelet and anti-melanogenesis activities. In this study, we first investigated the possible barrier protective effects of piperlonguminine against proinflammatory responses induced by lipopolysaccharide (LPS) and the associated signaling pathways in vitro and in vivo. The barrier protective activities of PL were determined by measuring permeability, monocytes adhesion and migration, and activation of proinflammatory proteins in LPS-activated human umbilical vein endothelial cells (HUVECs) and in mice. We found that PL inhibited LPS-induced barrier disruption, expression of cell adhesion molecules (CAMs) and adhesion/transendothelial migration of monocytes to human endothelial cells. PL also suppressed LPS-induced hyperpermeability and leukocytes migration in vivo. Further studies revealed that PL suppressed the production of tumor necrosis factor-α (TNF-α) or Interleukin (IL)-6 and activation of nuclear factor-κB (NF-κB) or extracellular regulated kinases (ERK) 1/2 by LPS. Moreover, treatment with PL resulted in reduced LPS-induced septic mortality. Collectively, these results suggest that PL protects vascular barrier integrity by inhibiting hyperpermeability, expression of CAMs, adhesion and migration of leukocytes, thereby endorsing its usefulness as a therapy for vascular inflammatory diseases.
Collapse
Affiliation(s)
- Wonhwa Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 702-701, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Stampalija T, Romero R, Korzeniewski SJ, Chaemsaithong P, Miranda J, Yeo L, Dong Z, Hassan SS, Chaiworapongsa T. Soluble ST2 in the fetal inflammatory response syndrome: in vivo evidence of activation of the anti-inflammatory limb of the immune response. J Matern Fetal Neonatal Med 2013; 26:1384-93. [PMID: 23488731 DOI: 10.3109/14767058.2013.784258] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Inflammation is a mechanism of host response to infection, which can be harmful when inappropriately modulated. Soluble ST2 (sST2) is a decoy receptor of interleukin (IL)-33, and this complex modulates the balance in the Th1/Th2 immune response. Moreover, sST2 inhibits the production of pro-inflammatory cytokines in cooperation with an anti-inflammatory cytokine, IL-10. The objectives of this study were to: (1) determine whether umbilical cord plasma sST2 concentration differs between preterm neonates with and without funisitis and between those with and without the fetal inflammatory response syndrome (FIRS); and (2) evaluate the relationship between sST2 and IL-10 among neonates with funisitis and/or FIRS. METHODS Umbilical cord plasma was collected from neonates delivered prematurely due to preterm labor or preterm prelabor rupture of membranes with (n = 36), and without funisitis (n = 30). FIRS (umbilical cord IL-6 concentration ≥ 17.5 pg/mL) was identified in 29 neonates. Plasma sST2 and IL-10 concentrations were determined by enzyme linked immune sorbent assay. RESULTS The median umbilical cord plasma sST2 concentration was 6.7-fold higher in neonates with FIRS than in those without FIRS (median 44.6 ng/mL, interquartile range (IQR) 13.8-80.3 ng/mL versus median 6.7 ng/mL, IQR 5.6-20.1 ng/mL; p < 0.0001). Similarly, the median umbilical cord plasma sST2 concentration was 2.7-fold higher in neonates with funisitis than in those without funisitis (median 19.1 ng/mL; IQR 7.1-75.0 ng/mL versus median 7.2 ng/mL; IQR 5.9-23.1 ng/mL; p = 0.008). There was a strong positive correlation between sST2 and IL-10 in neonates with funisitis and/or FIRS (Spearman's Rho = 0.7, p < 0.0001). CONCLUSION FIRS and funisitis are associated with an elevation of umbilical cord plasma concentrations of soluble ST2. This protein represents an important mediator of the immune response in neonates diagnosed with FIRS by promoting an anti-inflammatory effect in association with IL-10.
Collapse
Affiliation(s)
- Tamara Stampalija
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD and Detroit, MI 48201, USA
| | | | | | | | | | | | | | | | | |
Collapse
|