1
|
Kapteijn MY, Yanovska M, Laghmani EH, Postma RJ, van Duinen V, Ünlü B, Queiroz K, van Zonneveld AJ, Versteeg HH, Rondon AMR. Modeling cancer-associated hypercoagulability using glioblastoma spheroids in microfluidic chips. Res Pract Thromb Haemost 2024; 8:102475. [PMID: 39268353 PMCID: PMC11391032 DOI: 10.1016/j.rpth.2024.102475] [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: 02/13/2024] [Revised: 05/02/2024] [Accepted: 06/11/2024] [Indexed: 09/15/2024] Open
Abstract
Background Cancer increases the risk of venous thromboembolism, and glioblastoma is one of the cancer types with the highest risk of venous thromboembolism (10%-30%). Tumor-intrinsic features are believed to affect vascular permeability and hypercoagulability, but novel models are required to study the pathophysiological dynamics underlying cancer-associated thrombosis at the molecular level. Objectives We have developed a novel cancer-on-a-chip model to examine the effects of glioblastoma cells on the deregulation of blood coagulation. Methods This was accomplished by coculturing vessel-forming human umbilical vein endothelial cells with glioblastoma spheroids overexpressing tissue factor (TF), the initiator of coagulation (U251 lentivirus, LV-TF) or an LV-control (U251 LV-Ctrl) in an OrganoPlate Graft platform. Results Using a modified thrombin generation assay inside the cancer-on-a-chip, we found that U251 LV-Ctrl and U251 LV-TF spheroids promoted an increased procoagulant state in plasma, as was shown by a 3.1- and 7.0-fold increase in endogenous thrombin potential, respectively. Furthermore, the anticoagulant drug rivaroxaban and TF coagulation-blocking antibody 5G9 inhibited the activation of blood coagulation in U251 LV-TF spheroid-containing graft plates, as was shown by a reduced endogenous thrombin potential (4.0- and 4.4-fold, respectively). Conclusion With this study, we present a novel 3-dimensional cancer-on-a-chip model that has the potential to be used in the discovery of new anticoagulant drugs and identification of optimal anticoagulant strategies for glioblastoma and other cancer types.
Collapse
Affiliation(s)
- Maaike Y Kapteijn
- Division of Thrombosis and Hemostasis, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Monika Yanovska
- Division of Thrombosis and Hemostasis, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - El Houari Laghmani
- Division of Thrombosis and Hemostasis, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Rudmer J Postma
- Division of Nephrology, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Vincent van Duinen
- Division of Nephrology, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
- Mimetas BV, Oegstgeest, the Netherlands
| | - Betül Ünlü
- Division of Thrombosis and Hemostasis, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Anton Jan van Zonneveld
- Division of Nephrology, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Henri H Versteeg
- Division of Thrombosis and Hemostasis, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Araci M R Rondon
- Division of Thrombosis and Hemostasis, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
2
|
Hu F, Qu Z, Chen K, Zhang P, Wang B, Jiang R, Zuo Y, Xia P, Chen H. Lipoxin A4 Ameliorates Imiquimod-Induced Psoriasis-Like Dermatitis via Promoting the Regression of Inflammation. Clin Cosmet Investig Dermatol 2023; 16:2103-2111. [PMID: 37575152 PMCID: PMC10422962 DOI: 10.2147/ccid.s418467] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 08/03/2023] [Indexed: 08/15/2023]
Abstract
Introduction As a mediator of inflammation resolution, lipoxin A4 (LXA4) mainly plays an anti-inflammatory role and promotes inflammation resolution. LXA4 plays an inhibiting inflammatory role in a variety of diseases, tissues and cells, including keratinocytes. Psoriasis is a chronic inflammatory skin disease mediated by dysregulation of inflammation of immune cells and keratinocytes. However, the expression and role of LXA4 in psoriasis-like mouse models are still unclear. Methods Imiquimod (IMQ) topical treatment of dorsal skin induces psoriasis-like dermatitis in BALB/c mice, pretreated intraperitoneally with or without LXA4 prior to IMQ application. Severity of dorsal lesions is assessed by using a modified human scoring system and histopathology. The concentration of LXA4 and the expression of ALOX15 (a key gene in LXA4 metabolic synthesis) in lesional skins were detected by ELISA and Western blot. Quantitative PCR and ELISA were conducted to detect the mRNA and secretion levels of inflammatory cytokines. The proportion of IL-17A-producing γδT cells in skin and skin draining cervical lymph nodes and helper (Th) 17 cells in spleens was evaluated by flow cytometry. Western blotting was used to analyze the expressions of p-STAT3 and TRAF6. Results The concentration of LXA4 and the expression of ALOX15 were decreased in IMQ-induced lesional skin. LXA4 significantly relieved psoriasis-like lesions in IMQ-induced mouse models. Furthermore, LXA4 decreased IMQ-induced systemic inflammation, including reduced the proportion of IL-17A-producing gdT cells in skin and skin draining cervical lymph nodes and Th17 cells in spleens, the secretion and expression of CCL20, IL-17A, IL-1β, and TNF-α in skin and serum. LXA4 markedly inhibited IMQ-induced expression of TRAF6 and p-STAT3. Conclusion LXA4 significantly ameliorates IMQ-induced psoriasis-like inflammation, and LXA4 can be used as a target for psoriasis treatment.
Collapse
Affiliation(s)
- Feng Hu
- Department of Dermatology, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Province & Key Laboratory of Skin Infection and Immunity, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Zilu Qu
- Department of Dermatology, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Province & Key Laboratory of Skin Infection and Immunity, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Kai Chen
- Department of Dermatology, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Province & Key Laboratory of Skin Infection and Immunity, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Ping Zhang
- Department of Dermatology, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Bei Wang
- Department of Dermatology, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Ruili Jiang
- Department of Dermatology, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Yuyue Zuo
- Department of Dermatology, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Ping Xia
- Department of Dermatology, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Hongxiang Chen
- Department of Dermatology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, People’s Republic of China
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| |
Collapse
|
3
|
Neutrophils: As a Key Bridge between Inflammation and Thrombosis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:1151910. [PMID: 36408343 PMCID: PMC9668459 DOI: 10.1155/2022/1151910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 07/14/2022] [Accepted: 09/29/2022] [Indexed: 11/10/2022]
Abstract
Immunothrombosis is a mechanism of defense of the organism against pathogenic microorganisms that increases their recognition, limitation, and clearance and is part of the innate immune defense. Physiological immunothrombosis is beneficial to the body against the invasion of pathogenic microorganisms, but when immunothrombosis is out of control, it is easy to cause thrombotic diseases, thus, causing unpredictable consequences to the body. Neutrophils play a pivotal role in this process. Understanding the mechanism of neutrophils in immune thrombosis and out-of-control is particularly important for the treatment of related thrombotic diseases. In this review, we studied the role of neutrophils in immune thrombosis and each link out of control (including endothelial cell dysfunction; activation of platelets; activation of coagulation factor; inhibition of the anticoagulation system; and inhibition of the fibrinolysis system).
Collapse
|
4
|
Pro- and anti-inflammatory bioactive lipids imbalance contributes to the pathobiology of autoimmune diseases. Eur J Clin Nutr 2022:10.1038/s41430-022-01173-8. [PMID: 35701524 DOI: 10.1038/s41430-022-01173-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 04/22/2022] [Accepted: 05/26/2022] [Indexed: 12/27/2022]
Abstract
Autoimmune diseases are driven by TH17 cells that secrete pro-inflammatory cytokines, especially IL-17. Under normal physiological conditions, autoreactive T cells are suppressed by TGF-β and IL-10 secreted by microglia and dendritic cells. When this balance is upset due to injury, infection and other causes, leukocyte recruitment and macrophage activation occurs resulting in secretion of pro-inflammatory IL-6, TNF-α, IL-17 and PGE2, LTs (leukotrienes) accompanied by a deficiency of anti-inflammatory LXA4, resolvins, protecting, and maresins. PGE2 facilitates TH1 cell differentiation and promotes immune-mediated inflammation through TH17 expansion. There is evidence to suggest that autoimmune diseases can be suppressed by anti-inflammatory bioactive lipids LXA4, resolvins, protecting, and maresins. These results imply that systemic and/or local application of LXA4, resolvins, protecting, and maresins and administration of their precursors AA/EPA/DHA could form a potential therapeutic approach in the prevention and treatment of autoimmune diseases.
Collapse
|
5
|
Gimenez BT, Cezarette GN, Bomfim ADS, Monteiro WM, Russo EMDS, Frantz FG, Sampaio SV, Sartim MA. Role of crotoxin in coagulation: novel insights into anticoagulant mechanisms and impairment of inflammation-induced coagulation. J Venom Anim Toxins Incl Trop Dis 2020; 26:e20200076. [PMID: 33293940 PMCID: PMC7702976 DOI: 10.1590/1678-9199-jvatitd-2020-0076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 11/04/2020] [Indexed: 12/24/2022] Open
Abstract
Background: Snake venom phospholipases A2 (svPLA2) are
biologically active toxins, capable of triggering and modulating a wide
range of biological functions. Among the svPLA2s, crotoxin (CTX)
has been in the spotlight of bioprospecting research due to its role in
modulating immune response and hemostasis. In the present study, novel
anticoagulant mechanisms of CTX, and the modulation of inflammation-induced
coagulation were investigated. Methods: CTX anticoagulant activity was evaluated using platelet poor plasma (PPP)
and whole blood (WB), and also using isolated coagulation factors and
complexes. The toxin modulation of procoagulant and pro-inflammatory effects
was evaluated using the expression of tissue factor (TF) and cytokines in
lipopolysaccharide (LPS)-treated peripheral blood mononuclear cells (PBMC)
and in WB. Results: The results showed that CTX impaired clot formation in both PPP and WB, and
was responsible for the inhibition of both intrinsic (TF/factor VIIa) and
extrinsic (factor IXa/factor VIIIa) tenase complexes, but not for factor Xa
and thrombin alone. In addition, the PLA2 mitigated the
prothrombinase complex by modulating the coagulation phospholipid role in
the complex. In regards to the inflammation-coagulation cross talk, the
toxin was capable of reducing the production of the pro-inflammatory
cytokines IL-1β, IL-6 and TNF-α, and was followed by decreased levels of TF
and procoagulant activity from LPS-treated PBMC either isolated or in
WB. Conclusion: The results obtained in the present study recognize the toxin as a novel
medicinal candidate to be applied in inflammatory diseases with coagulation
disorders.
Collapse
Affiliation(s)
- Bruna Terada Gimenez
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Gabriel Neves Cezarette
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Aline de Sousa Bomfim
- Center for Cell-Based Therapy and Regional Blood Center of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Wuelton Marcelo Monteiro
- Tropical Medicine Graduate Program, Amazonas State University, Manaus, AM, Brazil.,Carlos Borborema Clinical Research Institute, Doutor Heitor Vieira Dourado Tropical Medicine Foundation, Manaus, AM, Brazil
| | - Elisa Maria de Sousa Russo
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil.,Center for Cell-Based Therapy and Regional Blood Center of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Fabiani Gai Frantz
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Suely Vilela Sampaio
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Marco Aurelio Sartim
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil.,Basic and Applied Immunology Graduate Program, Institute of Biological Sciences, Federal University of Amazonas, Manaus, AM, Brazil
| |
Collapse
|
6
|
Jin J, Xie Y, Shi C, Ma J, Wang Y, Qiao L, Li K, Sun T. Lipoxin A4 Inhibits NLRP3 Inflammasome Activation in Rats With Non-compressive Disc Herniation Through the JNK1/Beclin-1/PI3KC3 Pathway. Front Neurosci 2020; 14:799. [PMID: 33071721 PMCID: PMC7539067 DOI: 10.3389/fnins.2020.00799] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 07/07/2020] [Indexed: 01/05/2023] Open
Abstract
Background Non-compressive disc herniation is induced by an inflammatory response from the nucleus pulposus tissue and nerve roots. Lipoxins (LXs) are important endogenous anti-inflammatory mediators in the body, helping to inhibit neutrophil recruitment and stimulate autophagy in monocytes and macrophages. Here, we investigated the molecular mechanisms underlying the effects of exogenous lipoxin administration on rats with non-compressive disc herniation. Method A non-compressive disc herniation model was established in rats. Fifty rats were randomly divided into: sham group, model group, PI3K inhibitor (LY294002) group, lipoxin A4 group (LXA4), and PI3K inhibitor and lipoxin A4 group (LY294002 + LXA4). Similar groupings were established for rat spinal neurons. Changes in the mechanical pain threshold and thermal pain threshold were monitored at different times. The expression of proinflammatory and anti-inflammatory mediators was assessed by ELISA, while immunohistochemistry was employed to measure the expression levels of NLRP3 and p-JNK1. The expression levels of autophagy-related proteins were measured by western blot. Results In vivo, the pain threshold was markedly decreased in the model group at each time point examined compared with that in sham group. LY294002 treatment further reduced the pain threshold. After LXA4 injection, the pain threshold was significantly increased, and the effect of LY294002 was significantly weakened (p < 0.05). The levels of proinflammatory cytokines were increased in rats with non-compressive disc herniation, and these levels were further increased by LY294002 treatment (p < 0.05). However, treatment with LXA4 significantly reduced the levels of these proinflammatory cytokines in the model group (p < 0.05). The opposite effect was observed for anti-inflammatory mediators. The expression of NLRP3 was largely increased in the model group compared with that in the sham group (p < 0.05). Treatment with LY294002 also increased the NLRP3 expression level, while the administration of LXA4 elicited the opposite effect. Furthermore, western blot analysis showed that the expression of autophagy-related proteins was greatly decreased in the model group, whereas it was significantly increased in the LXA4 group (p < 0.05). The in vitro results were consistent with the outcomes observed in vivo. Conclusions These data suggested that LXA4 inhibited NLRP3 activation in rats with non-compressive disc herniation by regulating the JNK1/beclin-1/PI3KC3 pathway.
Collapse
Affiliation(s)
- Jin Jin
- Department of Pain Management, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Yonggang Xie
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Cunxian Shi
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Jiahai Ma
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Yihao Wang
- Department of Anesthesiology, Qingdao Municipal Hospital, Qingdao, China
| | - Leyan Qiao
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Kezhong Li
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Tao Sun
- Department of Pain Management, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
7
|
AL-Megrin WA, Soliman D, Kassab RB, Metwally DM, Ahmed E. Abdel Moneim, El-Khadragy MF. Coenzyme Q10 Activates the Antioxidant Machinery and Inhibits the Inflammatory and Apoptotic Cascades Against Lead Acetate-Induced Renal Injury in Rats. Front Physiol 2020; 11:64. [PMID: 32116774 PMCID: PMC7020615 DOI: 10.3389/fphys.2020.00064] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 01/21/2020] [Indexed: 12/29/2022] Open
Abstract
The kidney is among the metabolic organs most susceptible to injury, particularly following exposure to xenobiotics and heavy metals. We aimed to explore the potential protective impacts of coenzyme Q10 (CoQ10) on lead acetate (PbAc)-induced nephrotoxicity in rats. Four experimental groups (n = 7) were applied as follows: control group, CoQ10 alone (10 mg/kg), PbAc alone (20 mg/kg), and PbAc with CoQ10. Exposure to PbAc led to the accumulation of Pb in the kidney and increased urea and creatinine serum levels. The deposition of Pb coupled with the elevation of malondialdehyde and nitrate/nitrite levels along with the upregulation of inducible nitric oxide synthase. Additionally, upon PbAc poisoning, glutathione content and the antioxidant enzymes were depleted along with the downregulation of Nrf2 and HO-1 expression. Moreover, PbAc injection increased the protein and mRNA levels of pro-inflammatory cytokines namely, tumor necrosis factor-alpha and interleukin-1 beta, while decreased the levels of interleukin-10, an anti-inflammatory cytokine, in the kidney. Furthermore, exposure to PbAc correlated with increased levels of pro-apoptotic markers, Bax and caspase-3, and reduced levels of the anti-apoptotic marker Bcl-2. The administration of CoQ10 alleviated the molecular, biochemical and histological changes following PbAc intoxication. Thus, CoQ10 reduces the deleterious cellular side effects of PbAc exposure due to its antioxidant, anti-inflammatory and anti-apoptotic effects.
Collapse
Affiliation(s)
- Wafa A. AL-Megrin
- Department of Biology, Faculty of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Doaa Soliman
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt
| | - Rami B. Kassab
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt
| | - Dina M. Metwally
- Department of Parasitology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
- Department of Zoology, Faculty of Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed E. Abdel Moneim
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt
| | - Manal F. El-Khadragy
- Department of Biology, Faculty of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt
| |
Collapse
|
8
|
Fu T, Mohan M, Brennan EP, Woodman OL, Godson C, Kantharidis P, Ritchie RH, Qin CX. Therapeutic Potential of Lipoxin A 4 in Chronic Inflammation: Focus on Cardiometabolic Disease. ACS Pharmacol Transl Sci 2020; 3:43-55. [PMID: 32259087 DOI: 10.1021/acsptsci.9b00097] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Indexed: 02/07/2023]
Abstract
Several studies have shown that failure to resolve inflammation may contribute to the progression of many chronic inflammatory disorders. It has been suggested targeting the resolution of inflammation might be a novel therapeutic approach for chronic inflammatory diseases, including inflammatory bowel disease, diabetic complications, and cardiometabolic disease. Lipoxins [LXs] are a class of endogenously generated mediators that promote the resolution of inflammation. Biological actions of LXs include inhibition of neutrophil infiltration, promotion of macrophage polarization, increase of macrophage efferocytosis, and restoration of tissue homeostasis. Recently, several studies have demonstrated that LXs and synthetic analogues protect tissues from acute and chronic inflammation. The mechanism includes down-regulation of pro-inflammatory cytokines and chemokines (e.g., interleukin-1β and tumor necrosis factor-α), inhibition of the activation of the master pro-inflammatory pathway (e.g., nuclear factor κ-light-chain-enhancer of activated B cells pathway) and increased release of the pro-resolving cytokines (e.g., interleukin-10). Three generations of LXs analogues are well described in the literature, and more recently a fourth generation has been generated that appears to show enhanced potency. In this review, we will briefly discuss the potential therapeutic opportunity provided by lipoxin A4 as a novel approach to treat chronic inflammatory disorders, focusing on cardiometabolic disease and the current drug development in this area.
Collapse
Affiliation(s)
- Ting Fu
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia.,Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Muthukumar Mohan
- Department of Diabetes, Central Clinical School, Monash University, Clayton, Victoria 3800, Australia
| | - Eoin P Brennan
- UCD Diabetes Complications Research Centre, UCD Conway Institute, UCD School of Medicine, University College Dublin, Dublin, 4, Ireland
| | - Owen L Woodman
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia
| | - Catherine Godson
- UCD Diabetes Complications Research Centre, UCD Conway Institute, UCD School of Medicine, University College Dublin, Dublin, 4, Ireland
| | - Phillip Kantharidis
- Department of Diabetes, Central Clinical School, Monash University, Clayton, Victoria 3800, Australia
| | - Rebecca H Ritchie
- Department of Diabetes, Central Clinical School, Monash University, Clayton, Victoria 3800, Australia.,Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia.,Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria 3010, Australia.,Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Cheng Xue Qin
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia.,Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria 3010, Australia.,Department of Diabetes, Central Clinical School, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
9
|
Wang N, Zhang X, Ma Z, Niu J, Ma S, Wenjie W, Chen J. Combination of tanshinone IIA and astragaloside IV attenuate atherosclerotic plaque vulnerability in ApoE(-/-) mice by activating PI3K/AKT signaling and suppressing TRL4/NF-κB signaling. Biomed Pharmacother 2019; 123:109729. [PMID: 31887543 DOI: 10.1016/j.biopha.2019.109729] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 11/23/2019] [Accepted: 12/04/2019] [Indexed: 12/28/2022] Open
Abstract
Tanshinone IIA (TS IIA) and Astragaloside IV (AS IV) are natural herbal products which exert anti-inflammatory and anti-oxidant effects in order to eliminate unstable plaque in atherosclerosis. However, the combined effect of these two drugs on atherosclerotic plaque vulnerability and its molecular mechanism remains unclear. In the current study, we evaluate the effects of TS IIA and AS IV on atherosclerotic unstable plaque stability, and then further explore the mechanism of TS IIA and AS IV intervention on unstable plaque in vivo and in vitro. Histological characterization of atherosclerotic plaques was measured by Hematoxylin-Eosin (HE), Masson's Trichrome and Oil Red O staining. Cellular lipid droplet was measured by Oil Red O staining. The size of atherosclerotic lesion areas and content of lipids and collagen in the right common carotid arteries of apoE-/- mice were examined by Hematoxylin-Eosin (HE), Oil-red O, and Masson staining, respectively. The protein expression levels of interleukin (IL)-6, tumor necrosis factor (TNF)-α and C-reactive protein (CRP) in ApoE-/- mice and RAW264.7 cells were determined by enzyme-linked immunosorbent assay. The protein expression levels of matrix metalloproteinase-9 (MMP-9) and endothelial nitric oxide synthase (eNOS) in ApoE-/- mice and RAW264.7 cells were determined by western blotting. In addition, the PI3K/AKT and TRL4/NF-κB signaling were determined by western blotting. Our results revealed that the combination of TS IIA and AS IV significantly decreased lipid areas, increased collagen content and thickened fibrous cap in the right common carotid arteries compared with ApoE (-/-) mice model group. TS IIA and AS IV visibly reduced the cytoplasmic lipid droplet accumulation induced by oxLDL in RAW 264.7 macrophages. The ApoE-/- mice model group and oxLDL -stimulated RAW 264.7 macrophages treated with TS IIA and AS IV showed a downregulation in IL-6, MMP-9, TNF-α and CRP protein expression and upregulation in eNOS protein expression. Furthermore, TSIIA and AS IV may activate PI3K/AKT signaling and suppress TLR4/NF-κB signaling in vivo and in vitro. Additionally, blocking the PI3K/Akt signaling enhanced the translocation of NF-κB to the nucleus, TLR4, IL-6, MMP-9, TNF-α and CRP expression and inhibited eNOS expression in TS IIA and AS IV-treated RAW 264.7 macrophages. Therefore, the present study implicates that TS IIA and AS IV reinforces plaque stability via PI3K/AKT and TLR4/NF-κB signaling. TS IIA and AS IV administration may provide the basis for a potential therapeutic approach for the inhibition of vulnerable atherosclerotic plaques.
Collapse
Affiliation(s)
- Nanding Wang
- Xi'an Hospital of Traditional Chinese Medicine, Xi'an, 710021, Shaanxi Province, China
| | - Xiaofeng Zhang
- Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi'an, 710021, Shaanxi Province, China
| | - Zhen Ma
- Xi'an Hospital of Traditional Chinese Medicine, Xi'an, 710021, Shaanxi Province, China
| | - Jinghu Niu
- Xi'an Hospital of Traditional Chinese Medicine, Xi'an, 710021, Shaanxi Province, China
| | - Shihang Ma
- Xi'an Hospital of Traditional Chinese Medicine, Xi'an, 710021, Shaanxi Province, China
| | - Wang Wenjie
- Shaanxi university of Chinese medicine, Xi'an, 712046, Shaanxi Province, China
| | - Jun Chen
- Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi'an, 710021, Shaanxi Province, China.
| |
Collapse
|
10
|
Ma N, Wei W, Fan X, Ci X. Farrerol Attenuates Cisplatin-Induced Nephrotoxicity by Inhibiting the Reactive Oxygen Species-Mediated Oxidation, Inflammation, and Apoptotic Signaling Pathways. Front Physiol 2019; 10:1419. [PMID: 31849693 PMCID: PMC6901966 DOI: 10.3389/fphys.2019.01419] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 11/04/2019] [Indexed: 12/20/2022] Open
Abstract
Cisplatin is a chemotherapy drug that is often used in clinical practice, but its frequent use often leads to nephrotoxicity. Therefore, we urgently need a drug that reduces the nephrotoxicity induced by cisplatin. Farrerol reportedly has antioxidant potential, but its renal protective effects and potential mechanisms remain unclear. In this study, we used both cell and mouse models to determine the mechanism of farrerol in cisplatin-induced nephrotoxicity. The in vitro experiments revealed that farrerol improved cisplatin-induced nephrotoxicity and reactive oxygen species (ROS) production via nuclear factor erythrocyte 2-related factor 2 (Nrf2) activation. Moreover, farrerol effectively activated Nrf2 and subsequently increased the expression of Nrf2-targeted antioxidant enzymes, including heme oxygenase-1 (HO-1) and NAD(P)H quinone oxidoreductase-1 (NQO1), but inhibited Kelch-like ECH-associated protein 1 (Keap1) and NADPH oxidase type 4 (NOX4). Furthermore, farrerol attenuated the phosphorylation of C-Jun N-terminal kinase (JNK), extracellular signal-regulated kinase (ERK), and p38 mitogen-activated protein kinase (p38); the activation of phosphorylated nuclear factor-κB (p-NF-κB) and nucleotide-binding domain (NOD)-like receptor protein 3 (NLRP3); and the expression of phosphorylated p53 (p-p53), Bax, and cleaved caspase-3. In vivo, farrerol significantly improved cisplatin-induced renal damage, as demonstrated by the recovery of blood urea nitrogen (BUN), serum creatinine (SCr), kidney injury molecule-1 (KIM-1), neutrophil gelatinase-associated lipocalin (NGAL), and pathological damage. Moreover, farrerol inhibited inflammatory and apoptotic protein expression in vivo. Notably, farrerol exerted slight protection in Nrf2-knockout mice compared with wild-type mice. These findings indicate that farrerol can effectively activate Nrf2 and can serve as a therapeutic target in the treatment of acute kidney injury (AKI).
Collapse
Affiliation(s)
- Ning Ma
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China.,Department of Urology, The First Hospital, Jilin University, Changchun, China
| | - Wei Wei
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
| | - Xiaoye Fan
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
| | - Xinxin Ci
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
| |
Collapse
|