1
|
Vara-Messler M, Trevisi L, Zulato E, Ramaschi GE, Risé P, Pinna C, Indraccolo S, Sala A, Bolego C. Aspirin-triggered DHA metabolites inhibit angiogenesis. Front Pharmacol 2025; 16:1524980. [PMID: 40070577 PMCID: PMC11893558 DOI: 10.3389/fphar.2025.1524980] [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: 11/08/2024] [Accepted: 02/04/2025] [Indexed: 03/14/2025] Open
Abstract
Background and aim Blood vessels supply oxygen, nutrients and provide gateways for immune surveillance. Since this network nourishes all tissues, vessel abnormalities contribute to many diseases, such as cancer. One of the potential targets for Docosahexaenoic Acid (DHA) in cancer is suppressing angiogenesis, a process of new blood vessel formation within tumors. In addition, aspirin (ASA) has antineoplastic effects that may be mediated, at least in part, by metabolites derived from acetylated COX-2. We aimed at determining the effect of DHA as well as its metabolites in angiogenesis, using in vitro as well as in vivo models. Methods Endothelial cell (EC) proliferation, motility and capillary-like tube formation were determined by MTT, wound healing, Boyden and Matrigel assays, respectively. In vivo angiogenesis was measured by the Matrigel sponge model in mice. The biosynthesis of proresolving lipid mediators by ECs was determined by LC-MS-MS. Results and conclusion DHA, but not arachidonic acid (AA), at concentrations consistent with those reached in blood after fish oil supplementation, decreased EC migration in a time- and concentration-dependent manner. Pretreatment with ASA modulated cell migration already after 24 h, while both DHA and ASA decreased migration at longer incubation times without affecting viability. 17-hydroxy-DHA was detected upon incubation with DHA, and increased amounts were observed upon combined treatment with DHA and ASA, an increase that was associated to a synergic effect on EC migration. 17(R)-hydroxy-DHA (17R-HDHA), the metabolite resulting from acetylated COX-2 activity of DHA, reduced EC migration in a concentration-dependent manner. DHA in the presence of ASA, as well as 17R-HDHA, also reduced EC tube formation. These results were confirmed in vivo where both 17R-HDHA or its downstream metabolite 17RResolvinD1 were able to decrease microvessels density in a Matrigel sponge model. Overall, we demonstrated that DHA in the presence of ASA-dependent acetylation of COX-2 showed increased antiangiogenic effects, possibly resulting from its conversion to its hydroxylated derivatives.
Collapse
Affiliation(s)
- M. Vara-Messler
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UC Louvain), Brussels, Belgium
| | - L. Trevisi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - E. Zulato
- Basic and Translational Oncology Unit, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
| | - G. E. Ramaschi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - P. Risé
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - C. Pinna
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - S. Indraccolo
- Basic and Translational Oncology Unit, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - A. Sala
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - C. Bolego
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| |
Collapse
|
2
|
Yao J, Yao W, Zhu J, Liu Y, Liu J, Ji Y, Ni X, Mu W, Yan B. Targeting tRNA-Derived Non-Coding RNA Alleviates Diabetes-Induced Visual Impairment through Protecting Retinal Neurovascular Unit. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411042. [PMID: 39513253 PMCID: PMC11714213 DOI: 10.1002/advs.202411042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Indexed: 11/15/2024]
Abstract
Diabetes is a major risk factor for compromised visual health, leading to retinal vasculopathy and neuropathy, both of which are hallmarks of neurovascular unit dysfunction. Despite the critical impact of diabetic retinopathy, the precise mechanism underlying neurovascular coupling and effective strategies to suppress neurovascular dysfunction remain unclear. In this study, the up-regulation of a tRNA-derived stress-induced RNA, 5'tiRNA-His-GTG, in response to diabetic stress is revealed. 5'tiRNA-His-GTG directly regulates Müller glia action and indirectly alters endothelial angiogenic effects and retinal ganglion cell (RGC) survival in vitro. Downregulation of 5'tiRNA-His-GTG alleviates diabetes-induced retinal neurovascular dysfunction, characterized by reduced retinal vascular dysfunction, decreased retinal neurodegeneration, and improved visually-guided behaviors in vivo. Mechanistically, 5'tiRNA-His-GTG acts as a key regulator of retinal neurovascular dysfunction, primarily by modulating arachidonic acid (AA) metabolism via the CYPs pathway. The 5'tiRNA-His-GTG-CYP2E1-19(S)-HETE signaling axis is identified as a key driver of retinal neurovascular dysfunction. Thus, targeting 5'tiRNA-His-GTG presents a promising therapeutic strategy for treating vasculopathy and neuropathy associated with diabetes mellitus. Modulating this novel signaling pathway can open up new avenues for intervention in diabetic retinopathy and its related complications.
Collapse
Affiliation(s)
- Jin Yao
- The Affiliated Eye HospitalNanjing Medical UniversityNanjing210000China
| | - Wen Yao
- The Affiliated Eye HospitalNanjing Medical UniversityNanjing210000China
| | - Jun‐Ya Zhu
- The Affiliated Eye HospitalNanjing Medical UniversityNanjing210000China
- School of MedicineSoutheast UniversityNanjing210009China
| | - Yan Liu
- The Affiliated Eye HospitalNanjing Medical UniversityNanjing210000China
| | - Jin‐Hong Liu
- The Affiliated Eye HospitalNanjing Medical UniversityNanjing210000China
| | - Yu‐Ke Ji
- The Affiliated Eye HospitalNanjing Medical UniversityNanjing210000China
| | - Xi‐Shen Ni
- The Affiliated Eye HospitalNanjing Medical UniversityNanjing210000China
| | - Wan Mu
- Department of OphthalmologyShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080China
- Eye Institute and Department of OphthalmologyEye and ENT HospitalFudan UniversityShanghai200031China
| | - Biao Yan
- Department of OphthalmologyShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080China
| |
Collapse
|
3
|
Shi Y, Yang Y, Feng M, Wu H. CYP2C19 loss-of-function variants are independent risk factors for premature cerebral infarction: a hospital based retrospective study. BMC Cardiovasc Disord 2024; 24:602. [PMID: 39472784 PMCID: PMC11520391 DOI: 10.1186/s12872-024-04269-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 10/15/2024] [Indexed: 11/02/2024] Open
Abstract
OBJECTIVE Cytochrome P450 2C19 (CYP2C19) plays an vital role in the course of cardiovascular and cerebrovascular diseases by affecting lipid metabolism. Triglyceride-glucose (TyG) is a comprehensive index composed of triglyceride and blood glucose, has relationship with some diseases. There was no research report on the association CYP2C19 polymorphisms, TyG with premature cerebral infarction (CI) (onset ≤ 65 years old) susceptibility. METHODS This study retrospectively analyzed 1953 CI patients aged ≤ 65 years old from December 2018 to March 2024, and 1919 age-matched individuals with non-CI as controls. The relationship between CYP2C19 polymorphisms, TyG and premature CI risk were analyzed. RESULTS The proportion of hypertension, and diabetes mellitus in patients with premature CI was higher than those in controls. The serum total cholesterol (TC), triglycerides (TG), low-density lipoprotein-cholesterol (LDL-C), and TyG levels in patients with premature CI were significantly higher than those in controls (all p < 0.05). The patients had lower CYP2C19 *1 allele frequency (63.3% vs. 69.6%, p < 0.001) and higher CYP2C19 *2 allele frequency (31.3% vs. 25.4%, p < 0.001) than controls. Logistic regression analysis showed that smoking history (odds ratio (OR): 1.193, 95% confidence interval (CI): 1.002-1.422, p = 0.048), hypertension (OR: 3.371, 95% CI: 2.914-3.898, p < 0.001), diabetes mellitus (OR: 1.911, 95% CI: 1.632-2.237, p < 0.001), CYP2C19 intermediate metabolizer (IM) + poor metabolizer (PM) phenotypes (OR: 1.424, 95% CI: 1.243-1.631, p < 0.001), and dyslipidemia (OR: 1.294, 95% CI: 1.077-1.554, p = 0.006) were independent risk factors for premature CI. CONCLUSIONS History of smoking, hypertension, diabetes mellitus, dyslipidemia, and CYP2C19 IM + PM phenotypes were independently associated with premature CI susceptibility.
Collapse
Affiliation(s)
- Yuliang Shi
- Department of Neurology, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China
| | - Yuxian Yang
- Department of Neurology, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China
| | - Miaoling Feng
- Department of Neurology, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China
| | - Heming Wu
- Department of Prenatal Diagnostic Center, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China.
| |
Collapse
|
4
|
Schaich KM. Epoxides: an underestimated lipid oxidation product. Free Radic Res 2024; 58:517-564. [PMID: 38124354 DOI: 10.1080/10715762.2023.2277142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 07/27/2023] [Accepted: 08/09/2023] [Indexed: 12/23/2023]
Abstract
Immense gains in understanding of mechanisms and effects of lipid oxidation have been achieved in the nearly 90 years over which lipid oxidation has been an active research focus. Even so, the substantial questions still being raised about lipid oxidation in this special issue show clearly that missing pieces remain and must be considered for full accounting of this important reaction in any system. In this context, epoxides are spotlighted as a critical overlooked product of lipid autoxidation - underestimated in analysis, underestimated in presence as a functionally active and competitive intermediate and product of lipid oxidation, and underestimated in potential contributions to impact of lipid oxidation on other molecules and cell functions. Logical reasons for ignoring or not finding epoxides are offered in historical development of lipid oxidation knowledge. Reactions generating lipid epoxides in autoxidation are reviewed, limitations in detecting and tracking epoxides are outlined to explain why epoxides may not be detected when they should be present, and justifications for increased research and analysis of epoxides are argued. The main goal is to provide a context for recognizing epoxides as critical products that must be accounted for in determining the state rather than extent of lipid oxidation and in tracking its consequences in oils, foods, personal care products, and tissues. A secondary goal is to stimulate new research using contemporary analyses to fill in the gaps of knowledge about epoxide formation, structure, and reactions in lipid autoxidation.
Collapse
Affiliation(s)
- Karen M Schaich
- Department of Food Science, Rutgers University, New Brunswick, NJ, USA
| |
Collapse
|
5
|
Penkov S, Fedorova M. Membrane Epilipidome-Lipid Modifications, Their Dynamics, and Functional Significance. Cold Spring Harb Perspect Biol 2024; 16:a041417. [PMID: 38253416 PMCID: PMC11216179 DOI: 10.1101/cshperspect.a041417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Lipids are characterized by extremely high structural diversity translated into a wide range of physicochemical properties. As such, lipids are vital for many different functions including organization of cellular and organelle membranes, control of cellular and organismal energy metabolism, as well as mediating multiple signaling pathways. To maintain the lipid chemical diversity and to achieve rapid lipid remodeling required for the responsiveness and adaptability of cellular membranes, living systems make use of a network of chemical modifications of already existing lipids that complement the rather slow biosynthetic pathways. Similarly to biopolymers, which can be modified epigenetically and posttranscriptionally (for nucleic acids) or posttranslationally (for proteins), lipids can also undergo chemical alterations through oxygenation, nitration, phosphorylation, glycosylation, etc. In this way, an expanded collective of modified lipids that we term the "epilipidome," provides the ultimate level of complexity to biological membranes and delivers a battery of active small-molecule compounds for numerous regulatory processes. As many lipid modifications are tightly controlled and often occur in response to extra- and intracellular stimuli at defined locations, the emergence of the epilipidome greatly contributes to the spatial and temporal compartmentalization of diverse cellular processes. Accordingly, epilipid modifications are observed in all living organisms and are among the most consistent prerequisites for complex life.
Collapse
Affiliation(s)
- Sider Penkov
- Lipid Metabolism: Analysis and Integration, Center of Membrane Biochemistry and Lipid Research, Faculty of Medicine Carl Gustav Carus of TU Dresden, Dresden 01307, Germany
| | - Maria Fedorova
- Lipid Metabolism: Analysis and Integration, Center of Membrane Biochemistry and Lipid Research, Faculty of Medicine Carl Gustav Carus of TU Dresden, Dresden 01307, Germany
| |
Collapse
|
6
|
Zhou Z, Li Y, Wu S, Liu T, Jiang J. Host-microbiota interactions in collagen-induced arthritis rats treated with human umbilical cord mesenchymal stem cell exosome and ginsenoside Rh2. Biomed Pharmacother 2024; 174:116515. [PMID: 38569276 DOI: 10.1016/j.biopha.2024.116515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/19/2024] [Accepted: 03/28/2024] [Indexed: 04/05/2024] Open
Abstract
Mesenchymal stem cell exosome (MSCs-exo) is a class of products secreted by mesenchymal stem cells (MSCs) that contain various biologically active substances. MSCs-exo is a promising alternative to MSCs due to their lower immunogenicity and lack of ethical constraints. Ginsenoside Rh2 (Rh2) is a hydrolyzed component of the primary active substance of ginsenosides. Rh2 has a variety of pharmacological functions, including anti-inflammatory, anti-tumor, and antioxidant. Studies have demonstrated that gut microbiota and metabolites are critical in developing rheumatoid arthritis (RA). In this study, we constructed a collagen-induced arthritis (CIA) model in rats. We used MSCs-exo combined with Rh2 to treat CIA rats. To observe the effect of MSCs-exo combined with Rh2 on joint inflammation, rat feces were collected for 16 rRNA amplicon sequencing and untargeted metabolomics analysis. The results showed that the arthritis index score and joint swelling of CIA rats treated with MSCs-exo in combination with Rh2 were significantly lower than those of the model and MSCs-exo alone groups. MSCs-exo and Rh2 significantly ameliorated the disturbed gut microbiota in CIA rats. The regulation of Candidatus_Saccharibacteria and Clostridium_XlVb regulation may be the most critical. Rh2 enhanced the therapeutic effect of MSCs-exo compared with the MSCs-exo -alone group. Furthermore, significant changes in gut metabolites were observed in the CIA rat group, and these differentially altered metabolites may act as messengers for host-microbiota interactions. These differential metabolites were enriched into relevant critical metabolic pathways, revealing possible pathways for host-microbiota interactions.
Collapse
Affiliation(s)
- Zhongsheng Zhou
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yang Li
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Shuhui Wu
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Te Liu
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China; Yibin Jilin University Research Institute, Jilin University, Yibin, Sichuan, China.
| | - Jinlan Jiang
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China.
| |
Collapse
|
7
|
Niknafs S, Meijer MMY, Khaskheli AA, Roura E. In ovo delivery of oregano essential oil activated xenobiotic detoxification and lipid metabolism at hatch in broiler chickens. Poult Sci 2024; 103:103321. [PMID: 38100943 PMCID: PMC10762474 DOI: 10.1016/j.psj.2023.103321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/20/2023] [Accepted: 11/22/2023] [Indexed: 12/17/2023] Open
Abstract
In ovo interventions are used to improve embryonic development and robustness of chicks. The objective of this study was to identify the optimal dose for in ovo delivery of oregano essential oil (OEO), and to investigate metabolic impacts. Broiler chickens Ross 308 fertile eggs were injected with 7 levels of OEO (0, 5, 10, 20, 30, 40, and 50 µL) into the amniotic fluid at embryonic d 17.5 (E17.5) (n = 48). Chick quality was measured by navel score (P < 0.05) and/or hatchability rates (P < 0.01) were significantly decreased at doses at or above 10 or 20 µL/egg, respectively, indicating potential toxicity. However, no effects were observed at the 5 µL/egg, suggesting that compensatory mechanisms were effective to maintain homeostasis in the developing embryo. To pursue a better understanding of these mechanisms, transcriptomic analyses of the jejunum were performed comparing the control injected with saline and the group injected with 5 µL of OEO. The transcriptomic analyses identified that 167 genes were upregulated and 90 were downregulated in the 5 µL OEO compared to the control group injected with saline (P < 0.01). Functional analyses of the differentially expressed genes (DEG) showed that metabolic pathways related to the epoxygenase cytochrome P450 pathway associated with xenobiotic catabolic processes were significantly upregulated (P < 0.05). In addition, long-chain fatty acid metabolism associated with ATP binding transporters was also upregulated in the OEO treated group (P < 0.05). The results indicated that low doses of OEO in ovo have the potential to increase lipid metabolism in late stages (E17.5) of embryonic development. In conclusion, in ovo delivery of 5 µL OEO did not show any negative impact on hatchability and chick quality. OEO elevated expression of key enzymes and receptors involved in detoxification pathways and lipid metabolism in the jejunum of hatchling broiler chicks.
Collapse
Affiliation(s)
- Shahram Niknafs
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, Brisbane, Qld 4072, Australia
| | - Mila M Y Meijer
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, Brisbane, Qld 4072, Australia
| | - Asad A Khaskheli
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, Brisbane, Qld 4072, Australia
| | - Eugeni Roura
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, Brisbane, Qld 4072, Australia.
| |
Collapse
|
8
|
Luo Z, Yao J, Wang Z, Xu J. Mitochondria in endothelial cells angiogenesis and function: current understanding and future perspectives. J Transl Med 2023; 21:441. [PMID: 37407961 DOI: 10.1186/s12967-023-04286-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 06/19/2023] [Indexed: 07/07/2023] Open
Abstract
Endothelial cells (ECs) angiogenesis is the process of sprouting new vessels from the existing ones, playing critical roles in physiological and pathological processes such as wound healing, placentation, ischemia/reperfusion, cardiovascular diseases and cancer metastasis. Although mitochondria are not the major sites of energy source in ECs, they function as important biosynthetic and signaling hubs to regulate ECs metabolism and adaptations to local environment, thus affecting ECs migration, proliferation and angiogenic process. The understanding of the importance and potential mechanisms of mitochondria in regulating ECs metabolism, function and the process of angiogenesis has developed in the past decades. Thus, in this review, we discuss the current understanding of mitochondrial proteins and signaling molecules in ECs metabolism, function and angiogeneic signaling, to provide new and therapeutic targets for treatment of diverse cardiovascular and angiogenesis-dependent diseases.
Collapse
Affiliation(s)
- Zhen Luo
- Shanghai Key Laboratory of Veterinary Biotechnology/Shanghai Collaborative Innovation Center of Agri-Seeds, School of Agriculture and Biology, Shanghai Jiao Tong University, Dongchuan Road 800, Minhang District, Shanghai, China
| | - Jianbo Yao
- Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, West Virginia, USA
| | - Zhe Wang
- Shanghai Key Laboratory of Veterinary Biotechnology/Shanghai Collaborative Innovation Center of Agri-Seeds, School of Agriculture and Biology, Shanghai Jiao Tong University, Dongchuan Road 800, Minhang District, Shanghai, China
| | - Jianxiong Xu
- Shanghai Key Laboratory of Veterinary Biotechnology/Shanghai Collaborative Innovation Center of Agri-Seeds, School of Agriculture and Biology, Shanghai Jiao Tong University, Dongchuan Road 800, Minhang District, Shanghai, China.
| |
Collapse
|
9
|
Ortiz-Placín C, Castillejo-Rufo A, Estarás M, González A. Membrane Lipid Derivatives: Roles of Arachidonic Acid and Its Metabolites in Pancreatic Physiology and Pathophysiology. Molecules 2023; 28:4316. [PMID: 37298790 PMCID: PMC10254454 DOI: 10.3390/molecules28114316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/17/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023] Open
Abstract
One of the most important constituents of the cell membrane is arachidonic acid. Lipids forming part of the cellular membrane can be metabolized in a variety of cellular types of the body by a family of enzymes termed phospholipases: phospholipase A2, phospholipase C and phospholipase D. Phospholipase A2 is considered the most important enzyme type for the release of arachidonic acid. The latter is subsequently subjected to metabolization via different enzymes. Three enzymatic pathways, involving the enzymes cyclooxygenase, lipoxygenase and cytochrome P450, transform the lipid derivative into several bioactive compounds. Arachidonic acid itself plays a role as an intracellular signaling molecule. Additionally, its derivatives play critical roles in cell physiology and, moreover, are involved in the development of disease. Its metabolites comprise, predominantly, prostaglandins, thromboxanes, leukotrienes and hydroxyeicosatetraenoic acids. Their involvement in cellular responses leading to inflammation and/or cancer development is subject to intense study. This manuscript reviews the findings on the involvement of the membrane lipid derivative arachidonic acid and its metabolites in the development of pancreatitis, diabetes and/or pancreatic cancer.
Collapse
Affiliation(s)
| | | | | | - Antonio González
- Instituto de Biomarcadores de Patologías Moleculares, Departamento de Fisiología, Universidad de Extremadura, 10003 Cáceres, Spain; (C.O.-P.); (A.C.-R.); (M.E.)
| |
Collapse
|
10
|
Gao L, Chen W, Li L, Li J, Kongling W, Zhang Y, Yang X, Zhao Y, Bai J, Wang F. Targeting soluble epoxide hydrolase promotes osteogenic-angiogenic coupling via activating SLIT3/HIF-1α signalling pathway. Cell Prolif 2023:e13403. [PMID: 36636821 DOI: 10.1111/cpr.13403] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 01/03/2023] [Accepted: 01/05/2023] [Indexed: 01/14/2023] Open
Abstract
Type H vessels have recently been identified to modulate osteogenesis. Epoxyeicostrioleic acids (EETs) have an essential contribution to vascular homeostasis. However, whether increased EETs with soluble epoxide hydrolase (sEH) inhibitor TPPU enhance the coupling of angiogenesis and osteogenesis remains largely unknown. The effects of TPPU on cross-talk between co-cultured human umbilical vein endothelial cells (HUVECs) and human dental pulp stem cells (hDPSCs), and on long bone growth and calvarial defect repair in mice were investigated in vitro and in vivo. TPPU enhanced osteogenic differentiation of co-cultured HUVECs and hDPSCs in vitro and increased type H vessels, and long bone growth and bone repair of calvarial defect. Mechanistically, TPPU promoted cell proliferation and angiogenesis, reclined cell apoptosis, and significantly increased CD31hi EMCNhi endothelial cells (ECs) and SLIT3 and HIF-1α expression levels in co-cultured HUVECs and hDPSCs. Knockdown of Slit3 in hDPSCs or Hif-1α in HUVECs impaired the formation of CD31hi EMCNhi ECs and reversed TPPU-induced osteogenesis. We defined a previously unidentified effect of TPPU coupling angiogenesis and osteogenesis. TPPU induced type H vessels by upregulating the expression of hDPSCs-derived SLIT3, which resulted in the activation of ROBO1/YAP1/HIF-1α signalling pathway in ECs. Targeting metabolic pathways of EETs represents a new strategy to couple osteogenesis and angiogenesis, sEH is a promising therapeutic target for bone regeneration and repair.
Collapse
Affiliation(s)
- Lu Gao
- School of Stomatology, Dalian Medical University, Dalian, China.,Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Dalian, China.,The Affiliated Stomatological Hospital of Dalian Medical University School of Stomatology, Dalian, China
| | - Weixian Chen
- School of Stomatology, Dalian Medical University, Dalian, China.,Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Dalian, China
| | - Lijun Li
- School of Stomatology, Dalian Medical University, Dalian, China.,Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Dalian, China
| | - Juanjuan Li
- School of Stomatology, Dalian Medical University, Dalian, China.,Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Dalian, China
| | - Wenyao Kongling
- School of Stomatology, Dalian Medical University, Dalian, China.,Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Dalian, China
| | - Yaoyang Zhang
- School of Stomatology, Dalian Medical University, Dalian, China.,The Affiliated Stomatological Hospital of Dalian Medical University School of Stomatology, Dalian, China
| | - Xueping Yang
- School of Stomatology, Dalian Medical University, Dalian, China.,Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Dalian, China
| | - Yanrong Zhao
- School of Stomatology, Dalian Medical University, Dalian, China.,Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Dalian, China
| | - Jie Bai
- School of Stomatology, Dalian Medical University, Dalian, China.,Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Dalian, China
| | - Fu Wang
- School of Stomatology, Dalian Medical University, Dalian, China.,Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Dalian, China.,The Affiliated Stomatological Hospital of Dalian Medical University School of Stomatology, Dalian, China
| |
Collapse
|
11
|
Lipid mediators generated by the cytochrome P450—Epoxide hydrolase pathway. ADVANCES IN PHARMACOLOGY 2023; 97:327-373. [DOI: 10.1016/bs.apha.2022.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
12
|
Lopez M, Malacarne PF, Ramanujam DP, Warwick T, Müller N, Hu J, Dewenter M, Weigert A, Günther S, Gilsbach R, Engelhardt S, Brandes RP, Rezende F. Endothelial deletion of the cytochrome P450 reductase leads to cardiac remodelling. Front Physiol 2022; 13:1056369. [DOI: 10.3389/fphys.2022.1056369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/08/2022] [Indexed: 12/05/2022] Open
Abstract
The cytochrome P450 reductase (POR) transfers electrons to all microsomal cytochrome P450 enzymes (CYP450) thereby driving their activity. In the vascular system, the POR/CYP450 system has been linked to the production of epoxyeicosatrienoic acids (EETs) but also to the generation of reactive oxygen species. In cardiac myocytes (CMs), EETs have been shown to modulate the cardiac function and have cardioprotective effects. The functional importance of the endothelial POR/CYP450 system in the heart is unclear and was studied here using endothelial cell-specific, inducible knockout mice of POR (ecPOR−/−). RNA sequencing of murine cardiac cells revealed a cell type-specific expression of different CYP450 homologues. Cardiac endothelial cells mainly expressed members of the CYP2 family which produces EETs, and of the CYP4 family that generates omega fatty acids. Tamoxifen-induced endothelial deletion of POR in mice led to cardiac remodelling under basal conditions, as shown by an increase in heart weight to body weight ratio and an increased CM area as compared to control animals. Endothelial deletion of POR was associated with a significant increase in endothelial genes linked to protein synthesis with no changes in genes of the oxidative stress response. CM of ecPOR−/− mice exhibited attenuated expression of genes linked to mitochondrial function and an increase in genes related to cardiac myocyte contractility. In a model of pressure overload (transverse aortic constriction, TAC with O-rings), ecPOR−/− mice exhibited an accelerated reduction in cardiac output (CO) and stroke volume (SV) as compared to control mice. These results suggest that loss of endothelial POR along with a reduction in EETs leads to an increase in vascular stiffness and loss in cardioprotection, resulting in cardiac remodelling.
Collapse
|
13
|
Kytikova OY, Denisenko YK, Novgorodtseva TP, Bocharova NV, Kovalenko IS. [Fatty acid epoxides in the regulation of the inflammation]. BIOMEDITSINSKAIA KHIMIIA 2022; 68:177-189. [PMID: 35717582 DOI: 10.18097/pbmc20226803177] [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] [Indexed: 06/15/2023]
Abstract
Cyclooxygenase and lipoxygenase derived lipid metabolites of polyunsaturated fatty acids (PUFAs), as well as their role in the inflammation, have been studied quite thoroughly. However, cytochrome P450 derived lipid mediators, as well as their participation in the regulation of the inflammation, need deeper understanding. In recent years, it has become known that PUFAs are oxidized by cytochrome P450 epoxygenases to epoxy fatty acids, which act as the extremely powerful lipid mediators involved in resolving inflammation. Recent studies have shown that the anti-inflammatory mechanisms of ω-3 PUFAs are also mediated by their conversion to the endocannabinoid epoxides. Thus, it is clear that a number of therapeutically relevant functions of PUFAs are due to their conversion to PUFA epoxides. However, with the participation of cytochrome P450 epoxygenases, not only PUFA epoxides, but also other metabolites are formed. They are further are converted by epoxide hydrolases into pro-inflammatory dihydroxy fatty acids and anti-inflammatory dihydroxyeicosatrienoic acids. The study of the role of PUFA epoxides in the regulation of the inflammation and pharmacological modeling of the activity of epoxide hydrolases are the promising strategies for the treatment of the inflammatory diseases. This review systematizes the current literature data of the fatty acid epoxides, in particular, the endocannabinoid epoxides. Their role in the regulation of inflammation is discussed.
Collapse
Affiliation(s)
- O Y Kytikova
- Vladivostok Branch of Far Eastern Scientific Center of Physiology and Pathology of Respiration - Institute of Medical Climatology and Rehabilitative Treatment, Vladivostok, Russia
| | - Y K Denisenko
- Vladivostok Branch of Far Eastern Scientific Center of Physiology and Pathology of Respiration - Institute of Medical Climatology and Rehabilitative Treatment, Vladivostok, Russia
| | - T P Novgorodtseva
- Vladivostok Branch of Far Eastern Scientific Center of Physiology and Pathology of Respiration - Institute of Medical Climatology and Rehabilitative Treatment, Vladivostok, Russia
| | - N V Bocharova
- Vladivostok Branch of Far Eastern Scientific Center of Physiology and Pathology of Respiration - Institute of Medical Climatology and Rehabilitative Treatment, Vladivostok, Russia
| | - I S Kovalenko
- Vladivostok Branch of Far Eastern Scientific Center of Physiology and Pathology of Respiration - Institute of Medical Climatology and Rehabilitative Treatment, Vladivostok, Russia
| |
Collapse
|