1
|
Zhang Y, Youn JY, Huang K, Zhang Y, Cai H. Alleviation of accelerated diabetic atherogenesis in STZ-treated apoE/NOX1 DKO mice, apoE -/-/tg-EC-DHFR mice, and by folic acid. Redox Biol 2025; 82:103570. [PMID: 40184644 DOI: 10.1016/j.redox.2025.103570] [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: 02/19/2025] [Accepted: 02/24/2025] [Indexed: 04/07/2025] Open
Abstract
We and others have previously shown that uncoupling of endothelial nitric oxide synthase (eNOS) induces oxidative stress in diabetes, contributing to endothelial dysfunction. Activation of NADPH oxidase (NOX) isoform NOX1 by angiotensin II (Ang II) triggers eNOS uncoupling via deficiency in dihydrofolate reductase (DHFR) in streptozotocin (STZ)-induced type 1 diabetic mice. Presently, we investigated whether accelerated atherosclerosis is attenuated in apoE/NOX1 double knockout, and whether mice overexpressing DHFR in the endothelium (tg-EC-DHFR, generated in house) recouples eNOS to alleviate diabetic atherogenesis. At baseline, endothelial-specific DHFR overexpression recoupled eNOS and improved vasorelaxation in the aortas and mesenteric arteries of STZ-induced diabetic mice. Accelerated atherogenesis in STZ/high-fat diet (HFD) treated apoE-/- mice was markedly abrogated in tg-EC-DHFR background, establishing an important role of endothelial DHFR in maintaining vascular function and protecting from diabetic atherogenesis. Moreover, by crossing apoE-/- with NOX1 null mice (NOX1-/y), we found that NOX1 deletion markedly diminished atherosclerotic lesion formation in HFD + STZ-treated apoE-/-/NOX1-/y mice, indicating that NOX1 lies upstream of eNOS uncoupling in facilitating diabetic atherogenesis. Oral administration with folic acid (FA), shown to upregulate DHFR, robustly attenuated atherosclerotic lesion formation in HFD + STZ-treated apoE-/- mice similarly to NOX1 deletion. Taken together, our data for the first time demonstrate that endothelial DHFR plays an important role in the preservation of endothelial function and inhibition of atherosclerosis in diabetes, deficiency of which consequent to NOX1 activation mediates eNOS uncoupling driven lesion formation. Strategies targeting uncoupled eNOS prove to be robust treatment options for diabetic endothelial dysfunction and atherogenesis.
Collapse
Affiliation(s)
- Yixuan Zhang
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, California, 90095, USA
| | - Ji Youn Youn
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, California, 90095, USA
| | - Kai Huang
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, California, 90095, USA
| | - Yuhan Zhang
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, California, 90095, USA
| | - Hua Cai
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, California, 90095, USA.
| |
Collapse
|
2
|
Laumonnerie C, Shamambo M, Stabley DR, Lewis TL, Trivedi N, Howell D, Solecki DJ. Siah2 antagonism of Pard3/JamC modulates Ntn1-Dcc signaling to regulate cerebellar granule neuron germinal zone exit. Nat Commun 2025; 16:355. [PMID: 39774925 PMCID: PMC11706986 DOI: 10.1038/s41467-024-55400-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
Exiting a germinal zone (GZ) initiates a cascade of events that promote neuronal maturation and circuit assembly. Developing neurons and their progenitors must interpret various niche signals-such as morphogens, guidance molecules, extracellular matrix components, and adhesive cues-to navigate this region. How differentiating neurons in mouse brains integrate and adapt to multiple cell-extrinsic niche cues with their cell-intrinsic machinery in exiting a GZ is unknown. We establish cooperation between cell polarity-regulated adhesion and Netrin-1 signaling comprises a coincidence detection circuit repelling maturing neurons from their GZ. In this circuit, the Partitioning defective 3 (Pard3) polarity protein and Junctional adhesion molecule-C (JamC) adhesion molecule promote, while the Seven in absentia 2 (Siah2) ubiquitin ligase inhibits, Deleted in colorectal cancer (Dcc) receptor surface recruitment to gate differentiation linked repulsion to GZ Netrin-1. These results demonstrate cell polarity as a central integrator of adhesive- and guidance cues cooperating to spur GZ exit.
Collapse
Affiliation(s)
- Christophe Laumonnerie
- Neuronal Cell Biology Division, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38104, USA
| | - Maleelo Shamambo
- Neuronal Cell Biology Division, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38104, USA
| | - Daniel R Stabley
- Neuronal Cell Biology Division, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38104, USA
| | - Tommy L Lewis
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK, 73104, USA
| | - Niraj Trivedi
- Neuronal Cell Biology Division, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38104, USA
| | - Danielle Howell
- Neuronal Cell Biology Division, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38104, USA
| | - David J Solecki
- Neuronal Cell Biology Division, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38104, USA.
| |
Collapse
|
3
|
Laumonnerie C, Shamambo M, Stabley DR, Lewis TL, Trivedi N, Howell D, Solecki DJ. Antagonistic action of Siah2 and Pard3/JamC to promote germinal zone exit of differentiated cerebellar granule neurons by modulating Ntn1 signaling via Dcc. RESEARCH SQUARE 2024:rs.3.rs-1819367. [PMID: 39399669 PMCID: PMC11469462 DOI: 10.21203/rs.3.rs-1819367/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Exiting a germinal zone (GZ) initiates a cascade of events that promote neuronal maturation and circuit assembly. Developing neurons and their progenitors must interpret various niche signals-such as morphogens, guidance molecules, extracellular matrix components, and adhesive cues-to navigate this region. How differentiating neurons integrate and adapt to multiple cell-extrinsic niche cues with their cell-intrinsic machinery in exiting a GZ is unknown. We establish cooperation between cell polarity-regulated adhesion and Netrin-1 (Ntn-1) signaling comprises a coincidence detection circuit repelling maturing neurons from their GZ. In this circuit, the Partitioning defective 3 (Pard3) polarity protein and Junctional adhesion molecule-C (JamC) adhesion protein promote, while the Seven in absentia 2 (Siah2) ubiquitin ligase inhibits, Deleted in colorectal cancer (Dcc) receptor surface recruitment to gate differentiation linked repulsion to GZ Ntn-1. These results demonstrate cell polarity as a central integrator of adhesive- and guidance cues cooperating to spur GZ exit.
Collapse
Affiliation(s)
- Christophe Laumonnerie
- Neuronal Cell Biology Division, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38104
| | - Maleelo Shamambo
- Neuronal Cell Biology Division, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38104
| | - Daniel R Stabley
- Neuronal Cell Biology Division, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38104
| | - Tommy L Lewis
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK 73104
| | - Niraj Trivedi
- Neuronal Cell Biology Division, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38104
| | - Danielle Howell
- Neuronal Cell Biology Division, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38104
| | - David J Solecki
- Neuronal Cell Biology Division, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38104
| |
Collapse
|
4
|
Murugesan P, Zhang Y, Huang Y, Chenggong Zong N, Youn JY, Chen W, Wang C, Loscalzo J, Cai H. Reversal of Pulmonary Hypertension in a Human-Like Model: Therapeutic Targeting of Endothelial DHFR. Circ Res 2024; 134:351-370. [PMID: 38299369 PMCID: PMC10880947 DOI: 10.1161/circresaha.123.323090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 01/06/2024] [Accepted: 01/15/2024] [Indexed: 02/02/2024]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a progressive disorder characterized by remodeling of the pulmonary vasculature and elevated mean pulmonary arterial pressure, resulting in right heart failure. METHODS Here, we show that direct targeting of the endothelium to uncouple eNOS (endothelial nitric oxide synthase) with DAHP (2,4-diamino 6-hydroxypyrimidine; an inhibitor of GTP cyclohydrolase 1, the rate-limiting synthetic enzyme for the critical eNOS cofactor tetrahydrobiopterin) induces human-like, time-dependent progression of PH phenotypes in mice. RESULTS Critical phenotypic features include progressive elevation in mean pulmonary arterial pressure, right ventricular systolic blood pressure, and right ventricle (RV)/left ventricle plus septum (LV+S) weight ratio; extensive vascular remodeling of pulmonary arterioles with increased medial thickness/perivascular collagen deposition and increased expression of PCNA (proliferative cell nuclear antigen) and alpha-actin; markedly increased total and mitochondrial superoxide production, substantially reduced tetrahydrobiopterin and nitric oxide bioavailabilities; and formation of an array of human-like vascular lesions. Intriguingly, novel in-house generated endothelial-specific dihydrofolate reductase (DHFR) transgenic mice (tg-EC-DHFR) were completely protected from the pathophysiological and molecular features of PH upon DAHP treatment or hypoxia exposure. Furthermore, DHFR overexpression with a pCMV-DHFR plasmid transfection in mice after initiation of DAHP treatment completely reversed PH phenotypes. DHFR knockout mice spontaneously developed PH at baseline and had no additional deterioration in response to hypoxia, indicating an intrinsic role of DHFR deficiency in causing PH. RNA-sequencing experiments indicated great similarity in gene regulation profiles between the DAHP model and human patients with PH. CONCLUSIONS Taken together, these results establish a novel human-like murine model of PH that has long been lacking in the field, which can be broadly used for future mechanistic and translational studies. These data also indicate that targeting endothelial DHFR deficiency represents a novel and robust therapeutic strategy for the treatment of PH.
Collapse
Affiliation(s)
- Priya Murugesan
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (P.M., Y.Z., Y.H., N.C.Z., J.Y.Y., H.C.)
| | - Yixuan Zhang
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (P.M., Y.Z., Y.H., N.C.Z., J.Y.Y., H.C.)
| | - Yuanli Huang
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (P.M., Y.Z., Y.H., N.C.Z., J.Y.Y., H.C.)
| | - Nobel Chenggong Zong
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (P.M., Y.Z., Y.H., N.C.Z., J.Y.Y., H.C.)
| | - Ji Youn Youn
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (P.M., Y.Z., Y.H., N.C.Z., J.Y.Y., H.C.)
| | - Wenhui Chen
- Peking Union Medical College and Chinese Academy of Medical Sciences, Department of Respiratory Medicine, China-Japan Friendship Hospital, Beijing (W.C., C.W.)
| | - Chen Wang
- Peking Union Medical College and Chinese Academy of Medical Sciences, Department of Respiratory Medicine, China-Japan Friendship Hospital, Beijing (W.C., C.W.)
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA (J.L.)
| | - Hua Cai
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (P.M., Y.Z., Y.H., N.C.Z., J.Y.Y., H.C.)
| |
Collapse
|
5
|
Wu J, Yan Y. SIAH1 Promotes the Pyroptosis of Cardiomyocytes in Diabetic Cardiomyopathy via Regulating IκB-α/NF-κВ Signaling. Crit Rev Eukaryot Gene Expr 2024; 34:45-57. [PMID: 38842203 DOI: 10.1615/critreveukaryotgeneexpr.2024052773] [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: 06/07/2024]
Abstract
Inflammation-mediated dysfunction of cardiomyocytes is the main cause of diabetic cardiomyopathy (DCM). The present study aimed to investigate the roles of siah E3 ubiquitin protein ligase 1 (SIAH1) in DCM. The online dataset GSE4172 was used to analyze the differentially expressed genes in myocardial inflammation of DCM patients. RT-qPCR was conducted to detect mRNA levels. Enzyme-Linked Immunosorbent Assay (ELISA) was performed to detect cytokine release. Western blot was used to detect protein expression. Lactate dehydrogenase (LDH) assay was used to determine cytotoxicity. In vitro ubiquitination assay was applied to determine the ubiquitination of nuclear factor kappa B inhibitor alpha (1κВ-α). Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay was used to detect the death of cardiomyocytes. Flow cytometry was applied for determining cardiomyocyte pyroptosis. The results showed that SIAH1 was overexpressed in human inflammatory cardiomyopathy. High expression of SIAH1 was associated with inflammatory response. SIAH1 was also overexpressed lipopolysaccharide (LPS)-induced inflammatory cardiomyopathy model in vitro. However, SIAH1 knockdown suppressed the inflammatory-related pyroptosis of cardiomyocytes. SIAH1 promoted the ubiquitination of 1κВ-α and activated nuclear factor kappa В (NF-κВ) signaling, which promoted the pyroptosis of cardiomyocytes. In conclusion, SIAH1 exacerbated the progression of human inflammatory cardiomyopathy via inducing the ubiquitination of 1κВ-α and activation of NF-κВ signaling. Therefore, SIAHI/IκB-α/NF-κB signaling may be a potential target for human inflammatory cardiomyopathy.
Collapse
Affiliation(s)
| | - Yaoming Yan
- Laboratory Department, Peking University Shenzhen Hospital, Shenzhen 518036, China
| |
Collapse
|
6
|
Heck-Swain KL, Li J, Ruan W, Yuan X, Wang Y, Koeppen M, Eltzschig HK. Myeloid hypoxia-inducible factor HIF1A provides cardio-protection during ischemia and reperfusion via induction of netrin-1. Front Cardiovasc Med 2022; 9:970415. [PMID: 36247475 PMCID: PMC9554136 DOI: 10.3389/fcvm.2022.970415] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/22/2022] [Indexed: 12/03/2022] Open
Abstract
The transcription factor hypoxia-inducible factor HIF1A induces cardioprotection from ischemia and reperfusion injury. Here, we investigate tissue-specific pathways that are critical for HIF1A-elicited tissue protection. Initial studies showed that mice with induced global Hif1a deletion (Hif1aloxP/loxP UbiquitinCre+) have exaggerated myocardial injury during in situ ischemia and reperfusion. Surprisingly, this phenotype was mirrored only in mice with myeloid-specific Hif1a deletion (Hif1a loxP/loxP LysM Cre+). In contrast, mice with myocardial specific (Hif1aloxP/loxP Myosin Cre+), or vascular Hif1a deletion (Hif1aloxP/loxP VEcadherin Cre+) experienced similar levels of injury as controls. Subsequent studies using adoptive transfer of Hif1a-deficient polymorphonuclear neutrophils (PMNs) prior to myocardial injury demonstrated increased reperfusion injury. On the contrary, the adoptive transfer of PMNs treated ex vivo with the hypoxia inducible factor (HIF) stabilizer dimethyloxalylglycine (DMOG) was associated with attenuated myocardial injury. Furthermore, DMOG-mediated cardioprotection was abolished in Hif1aloxP/loxP LysM Cre+ mice, but not in Hif2aloxP/loxP LysM Cre+ mice. Finally, studies of PMN-dependent HIF1A target genes implicated the neuronal guidance molecule netrin-1 in mediating the cardioprotective effects of myeloid HIF1A. Taken together, the present studies identified a functional role for myeloid-expressed HIF1A in providing cardioprotection during ischemia and reperfusion injury, which is mediated, at least in part, by the induction of the netrin-1 neuronal guidance molecule in neutrophils.
Collapse
Affiliation(s)
- Ka Lin Heck-Swain
- Department of Anesthesiology and Intensive Care Medicine, Tübingen University Hospital, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Jiwen Li
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
- Department of Cardiac Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wei Ruan
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
- Department of Anesthesiology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoyi Yuan
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Yanyu Wang
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Michael Koeppen
- Department of Anesthesiology and Intensive Care Medicine, Tübingen University Hospital, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Holger K. Eltzschig
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
7
|
Murugesan P, Zhang Y, Youn JY, Cai H. Novel and robust treatment of pulmonary hypertension with netrin-1 and netrin-1-derived small peptides. Redox Biol 2022; 55:102348. [PMID: 35830752 PMCID: PMC9287481 DOI: 10.1016/j.redox.2022.102348] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/11/2022] [Accepted: 05/18/2022] [Indexed: 11/07/2022] Open
Abstract
Limited medical therapies have been implemented for the treatment of the devastating cardiorespiratory disease of pulmonary hypertension (PH) while none of which is sufficiently effective to stop or regress development of PH. We have previously shown that netrin-1, an axon-guiding protein during development, protects against ischemia reperfusion injury induced myocardial infarction via modest and stable production of nitric oxide (NO) and attenuation of oxidative stress. Since NO deficiency and oxidative stress-mediated vascular remodeling play important roles in the pathogenesis of PH, our present study investigated therapeutic effects on PH of netrin-1 and its derived small peptides. Infused into mice for 3 weeks during exposure to hypoxia, netrin-1 and netrin-1 derived small peptides V1, V2 or V3 substantially alleviated pathophysiological and molecular features of PH, as indicated by abrogated increases in mean pulmonary artery pressure (mPAP) and right ventricular systolic pressure (RVSP), attenuated right ventricular hypertrophy, diminished vascular remodeling of medial thickening and upregulation in smooth muscle alpha-actin (SMA) and proliferative cell nuclear antigen (PCNA), and alleviated perivascular and peribronchial fibrosis reflected by collagen deposition. NO bioavailability was substantially improved by treatment with netrin-1 and netrin-1 derived small peptides, while hypoxia induced increases in total superoxide production and eNOS uncoupling activity were all attenuated. These dual mechanisms of increasing NO bioavailability and decreasing oxidative stress at the same time, underlie robust protective effects on PH of netrin-1 and its derived small peptides, which are different from existing medications that primarily target NO signaling alone. Our data for the first time demonstrate intriguing findings that netrin-1 and netrin-1 derived small peptides can be used as novel and robust therapeutics for the treatment of PH.
Collapse
Affiliation(s)
- Priya Murugesan
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, United States
| | - Yixuan Zhang
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, United States
| | - Ji Youn Youn
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, United States
| | - Hua Cai
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, United States.
| |
Collapse
|
8
|
Yuan X, Mills T, Doursout MF, Evans SE, Vidal Melo MF, Eltzschig HK. Alternative adenosine Receptor activation: The netrin-Adora2b link. Front Pharmacol 2022; 13:944994. [PMID: 35910389 PMCID: PMC9334855 DOI: 10.3389/fphar.2022.944994] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 06/28/2022] [Indexed: 11/25/2022] Open
Abstract
During hypoxia or inflammation, extracellular adenosine levels are elevated. Studies using pharmacologic approaches or genetic animal models pertinent to extracellular adenosine signaling implicate this pathway in attenuating hypoxia-associated inflammation. There are four distinct adenosine receptors. Of these, it is not surprising that the Adora2b adenosine receptor functions as an endogenous feedback loop to control hypoxia-associated inflammation. First, Adora2b activation requires higher adenosine concentrations compared to other adenosine receptors, similar to those achieved during hypoxic inflammation. Second, Adora2b is transcriptionally induced during hypoxia or inflammation by hypoxia-inducible transcription factor HIF1A. Studies seeking an alternative adenosine receptor activation mechanism have linked netrin-1 with Adora2b. Netrin-1 was originally discovered as a neuronal guidance molecule but also functions as an immune-modulatory signaling molecule. Similar to Adora2b, netrin-1 is induced by HIF1A, and has been shown to enhance Adora2b signaling. Studies of acute respiratory distress syndrome (ARDS), intestinal inflammation, myocardial or hepatic ischemia and reperfusion implicate the netrin-Adora2b link in tissue protection. In this review, we will discuss the potential molecular linkage between netrin-1 and Adora2b, and explore studies demonstrating interactions between netrin-1 and Adora2b in attenuating tissue inflammation.
Collapse
Affiliation(s)
- Xiaoyi Yuan
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Tingting Mills
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Marie-Francoise Doursout
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Scott E. Evans
- Department of Pulmonology, MD Anderson Cancer Center, Houston, TX, United States
| | | | - Holger K. Eltzschig
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
9
|
Li K, Li J, Ye M, Jin X. The role of Siah2 in tumorigenesis and cancer therapy. Gene 2022; 809:146028. [PMID: 34687788 DOI: 10.1016/j.gene.2021.146028] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/11/2021] [Accepted: 10/14/2021] [Indexed: 12/12/2022]
Abstract
Seven in absentia homolog 2 (Siah2), an RING E3 ubiquitin ligases, has been characterized to play the vital role in tumorigenesis and cancer progression. Numerous studies have determined that Siah2 promotes tumorigenesis in a variety of human malignancies such as prostate, lung, gastric, and liver cancers. However, several studies revealed that Siah2 exhibited tumor suppressor function by promoting the proteasome-mediated degradation of several oncoproteins, suggesting that Siah2 could exert its biological function according to different stages of tumor development. Moreover, Siah2 is subject to complex regulation, especially the phosphorylation of Siah2 by a variety of protein kinases to regulate its stability and activity. In this review, we describe the structure and regulation of Siah2 in human cancer. Moreover, we highlight the critical role of Siah2 in tumorigenesis. Furthermore, we note that the potential clinical applications of targeting Siah2 in cancer therapy.
Collapse
Affiliation(s)
- Kailang Li
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo 315020, China; Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathphysiology, Medical School of Ningbo University, Ningbo 315211, China
| | - Jinyun Li
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo 315020, China; Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathphysiology, Medical School of Ningbo University, Ningbo 315211, China
| | - Meng Ye
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo 315020, China; Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathphysiology, Medical School of Ningbo University, Ningbo 315211, China.
| | - Xiaofeng Jin
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo 315020, China; Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathphysiology, Medical School of Ningbo University, Ningbo 315211, China.
| |
Collapse
|
10
|
Netrin-1: An Emerging Player in Inflammatory Diseases. Cytokine Growth Factor Rev 2022; 64:46-56. [DOI: 10.1016/j.cytogfr.2022.01.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 01/12/2022] [Indexed: 12/14/2022]
|
11
|
Therapeutic application of estrogen for COVID-19: Attenuation of SARS-CoV-2 spike protein and IL-6 stimulated, ACE2-dependent NOX2 activation, ROS production and MCP-1 upregulation in endothelial cells. Redox Biol 2021; 46:102099. [PMID: 34509916 PMCID: PMC8372492 DOI: 10.1016/j.redox.2021.102099] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 01/08/2023] Open
Abstract
The outbreak of COVID-19 has remained uncontained with urgent need for robust therapeutics. We have previously reported sex difference of COVID-19 for the first time indicating male predisposition. Males are more susceptible than females, and more often to develop into severe cases with higher mortality. This predisposition is potentially linked to higher prevalence of cigarette smoking. Nonetheless, we found for the first time that cigarette smoking extract (CSE) had no effect on angiotensin converting enzyme 2 (ACE2) and transmembrane protease serine 2 (TMPRSS2) expression in endothelial cells. The otherwise observed worse outcomes in smokers is likely linked to baseline respiratory diseases associated with chronic smoking. Instead, we hypothesized that estrogen mediated protection might underlie lower morbidity, severity and mortality of COVID-19 in females. Of note, endothelial inflammation and barrier dysfunction are major mediators of disease progression, and development of acute respiratory distress syndrome (ARDS) and multi-organ failure in patients with COVID-19. Therefore, we investigated potential protective effects of estrogen on endothelial cells against oxidative stress induced by interleukin-6 (IL-6) and SARS-CoV-2 spike protein (S protein). Indeed, 17β-estradiol completely reversed S protein-induced selective activation of NADPH oxidase isoform 2 (NOX2) and reactive oxygen species (ROS) production that are ACE2-dependent, as well as ACE2 upregulation and induction of pro-inflammatory gene monocyte chemoattractant protein-1 (MCP-1) in endothelial cells to effectively attenuate endothelial dysfunction. Effects of IL-6 on activating NOX2-dependent ROS production and upregulation of MCP-1 were also completely attenuated by 17β-estradiol. Of note, co-treatment with CSE had no additional effects on S protein stimulated endothelial oxidative stress, confirming that current smoking status is likely unrelated to more severe disease in chronic smokers. These data indicate that estrogen can serve as a novel therapy for patients with COVID-19 via inhibition of initial viral responses and attenuation of cytokine storm induced endothelial dysfunction, to substantially alleviate morbidity, severity and mortality of the disease, especially in men and post-menopause women. Short-term administration of estrogen can therefore be readily applied to the clinical management of COVID-19 as a robust therapeutic option.
Collapse
|
12
|
Li J, Conrad C, Mills TW, Berg NK, Kim B, Ruan W, Lee JW, Zhang X, Yuan X, Eltzschig HK. PMN-derived netrin-1 attenuates cardiac ischemia-reperfusion injury via myeloid ADORA2B signaling. J Exp Med 2021; 218:212023. [PMID: 33891683 PMCID: PMC8077173 DOI: 10.1084/jem.20210008] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 02/26/2021] [Accepted: 03/19/2021] [Indexed: 01/03/2023] Open
Abstract
Previous studies implicated the neuronal guidance molecule netrin-1 in attenuating myocardial ischemia-reperfusion injury. However, the tissue-specific sources and receptor signaling events remain elusive. Neutrophils are among the first cells responding to an ischemic insult and can be associated with tissue injury or rescue. We found netrin-1 levels were elevated in the blood of patients with myocardial infarction, as well as in mice exposed to myocardial ischemia-reperfusion. Selectively increased infarct sizes and troponin levels were found in Ntn1loxP/loxP Lyz2 Cre+ mice, but not in mice with conditional netrin-1 deletion in other tissue compartments. In vivo studies using neutrophil depletion identified neutrophils as the main source for elevated blood netrin-1 during myocardial injury. Finally, pharmacologic studies using treatment with recombinant netrin-1 revealed a functional role for purinergic signaling events through the myeloid adenosine A2b receptor in mediating netrin-1-elicited cardioprotection. These findings suggest an autocrine signaling loop with a functional role for neutrophil-derived netrin-1 in attenuating myocardial ischemia-reperfusion injury through myeloid adenosine A2b signaling.
Collapse
Affiliation(s)
- Jiwen Li
- Department of Anesthesiology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX.,Department of Cardiac Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Catharina Conrad
- Department of Anesthesiology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX.,Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Tingting W Mills
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX
| | - Nathaniel K Berg
- Department of Anesthesiology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX
| | - Boyun Kim
- Department of Anesthesiology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX
| | - Wei Ruan
- Department of Anesthesiology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX.,Department of Anesthesiology, Second Xiangya Hospital, Central South University, Hunan, China
| | - Jae W Lee
- Department of Anesthesiology, Yale University School of Medicine, New Haven, CT
| | - Xu Zhang
- Center for Clinical and Translational Sciences, The University of Texas Health Science Center at Houston, Houston, TX
| | - Xiaoyi Yuan
- Department of Anesthesiology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX
| | - Holger K Eltzschig
- Department of Anesthesiology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX
| |
Collapse
|
13
|
Abstract
Cardiovascular pathologies are often induced by inflammation. The associated changes in the inflammatory response influence vascular endothelial biology; they complicate the extent of ischaemia and reperfusion injury, direct the migration of immune competent cells and activate platelets. The initiation and progression of inflammation is regulated by the classical paradigm through the system of cytokines and chemokines. Therapeutic approaches have previously used this knowledge to control the extent of cardiovascular changes with varying degrees of success. Neuronal guidance proteins (NGPs) have emerged in recent years and have been shown to be significantly involved in the control of tissue inflammation and the mechanisms of immune cell activation. Therefore, proteins of this class might be used in the future as targets to control the extent of inflammation in the cardiovascular system. In this review, we describe the role of NGPs during cardiovascular inflammation and highlight potential therapeutic options that could be explored in the future.
Collapse
|
14
|
Targeting feed-forward signaling of TGFβ/NOX4/DHFR/eNOS uncoupling/TGFβ axis with anti-TGFβ and folic acid attenuates formation of aortic aneurysms: Novel mechanisms and therapeutics. Redox Biol 2020; 38:101757. [PMID: 33126053 PMCID: PMC7585948 DOI: 10.1016/j.redox.2020.101757] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/29/2020] [Accepted: 10/08/2020] [Indexed: 01/09/2023] Open
Abstract
In the present study we aimed to identify novel mechanisms and therapeutics for thoracic aortic aneurysm (TAA) in Fbn1C1039G/+ Marfan Syndrome (MFS) mice. The expression of mature/active TGFβ and its downstream effector NOX4 were upregulated while tetrahydrobiopterin (H4B) salvage enzyme dihydrofolate reductase (DHFR) was downregulated in Fbn1C1039G/+ mice. In vivo treatment with anti-TGFβ completely attenuated NOX4 expression, restored DHFR protein abundance, reduced ROS production, recoupled eNOS and attenuated aneurysm formation. Intriguingly, oral administration with folic acid (FA) to recouple eNOS markedly alleviated expansion of aortic roots and abdominal aortas in Fbn1C1039G/+ mice, which was attributed to substantially upregulated DHFR expression and activity in the endothelium to restore tissue and circulating levels of H4B. Notably, circulating H4B levels were accurately predictive of tissue H4B bioavailability, and negatively associated with expansion of aortic roots, indicating a novel biomarker role of circulating H4B for TAA. Furthermore, FA diet abrogated TGFβ and NOX4 expression, disrupting the feed-forward loop to inactivate TGFβ/NOX4/DHFR/eNOS uncoupling axis in vivo and in vitro, while PTIO, a NO scavenger, reversed this effect in cultured human aortic endothelial cells (HAECs). Besides, expression of the rate limiting H4B synthetic enzyme GTP cyclohydrolase 1 (GTPCHI), was downregulated in Fbn1C1039G/+ mice at baseline. In cultured HAECs, RNAi inhibition of fibrillin resulted in reduced GTPCHI expression, while this response was abrogated by anti-TGFβ, indicating TGFβ-dependent downregulation of GTPCHI in response to fibrillin deficiency. Taken together, our data for the first time reveal that uncoupled eNOS plays a central role in TAA formation, while anti-TGFβ and FA diet robustly abolish aneurysm formation via inactivation of a novel TGFβ/NOX4/DHFR/eNOS uncoupling/TGFβ feed-forward pathway. Correction of fibrillin deficiency is additionally beneficial via preservation of GTPCHI function.
Collapse
|
15
|
Zhou T, Chen Y, Zhao B, Hu S, Li J, Liu M, Liang S, Bao Z, Wu X. Characterization and functional analysis of SIAH1 during skin and hair follicle development in the angora rabbit (Oryctolagus cuniculus). Hereditas 2020; 157:10. [PMID: 32252830 PMCID: PMC7137485 DOI: 10.1186/s41065-020-00126-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 03/24/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Seven in absentia homolog 1 (SIAH1) is an E3 ubiquitin ligase containing a RING-finger domain and a key regulator of normal development. Skin and hair follicle development is a complex and special process of morphogenesis involving multiple signaling pathways. SIAH1 is enriched in the Wnt signaling pathway and potentially related to hair follicle cycle and skin development. This study aims to provide evidence for the role of SIAH1 in skin and hair development. RESULTS Full-length cloning and analysis of SIAH1 was conducted to better understand its function. Phylogenetically, the sequence of SIAH1 in the rabbit shares the greatest homology with Home sapiens, Pongo abelii and Mus mulatta. Based on the rabbit hair follicle synchronization model, we found that the expression level of SIAH1 in the regressive period of the rabbit hair cycle is significantly lower than in the active growth and rest periods. In addition, the mRNA expression levels of skin and hair follicle development-related genes changed significantly when SIAH1 was overexpressed and silenced. After SIAH1 overexpression, the expression levels of WNT2, LEF1 and FGF2 decreased, and those of SFRP2 and DKK1 increased (P < 0.05). After interference of SIAH1, the expression levels of WNT2, LEF1 and FGF2 increased (P < 0.05), and SFRP2 and DKK1 decreased. CONCLUSIONS SIAH1 can affect skin and hair follicle development and exert an inhibitory effect. These results could provide foundamental insights into the role of SIAH1 as a target gene in rabbit skin and hair follicle development.
Collapse
Affiliation(s)
- Tong Zhou
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Yang Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, China
| | - Bohao Zhao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Shuaishuai Hu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Jiali Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Ming Liu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Shuang Liang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Zhiyuan Bao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Xinsheng Wu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China. .,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, China.
| |
Collapse
|
16
|
Wang Q, Shi W, Zhang Q, Feng W, Wang J, Zhai C, Yan X, Li M. Inhibition of Siah2 ubiquitin ligase ameliorates monocrotaline-induced pulmonary arterial remodeling through inactivation of YAP. Life Sci 2019; 242:117159. [PMID: 31837334 DOI: 10.1016/j.lfs.2019.117159] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 11/23/2019] [Accepted: 12/09/2019] [Indexed: 12/11/2022]
Abstract
AIMS It has been shown that up-regulation of E3 ubiquitin ligase seven-in-absentia-homolog 2 (Siah2) and activation of Hippo signaling pathway effector yes-associated protein (YAP) are involved in the development of pulmonary arterial hypertension (PAH). However, it is still unclear whether Siah2 activates YAP in monocrotaline (MCT)-induced PAH rat models. MAIN METHODS Intraperitoneal injection of MCT was used to induce PAH rat models. The right ventricular systolic pressure (RVSP), right ventricle hypertrophy index (RVHI), percentage of medial wall thickness (%MT), α-SMA, Ki-67 and TUNEL staining were performed to evaluate the development of PAH. Protein levels of Siah2, Lats1/2, YAP phosphorylation and total YAP, and the subcellular localization of YAP were examined using immunoblotting. Proteasome activity was measured by an assay kit. KEY FINDINGS The protein level of Siah2 was significantly increased in MCT-induced PAH rats, this was accompanied with the proteasome-dependent degradation of Lats1/2 and subsequent up-regulation and dephosphorylation of YAP and its nuclear localization. Administration of PAH rats with Siah2 inhibitor Vitamin K3 or proteasome inhibitor MG-132 dramatically suppressed MCT-induced down-regulation of Lats1/2 and activation of YAP, finally reduced RVSP, RVHI, %MT, pulmonary arterial muscularization, pulmonary arterial smooth muscle cells (PASMCs) proliferation and enhanced PASMCs apoptosis in PAH rats. SIGNIFICANCE Siah2 contributes to the development of MCT-induced PAH by destabilizing Lats1/2 and subsequently stimulating YAP activation. Inhibition of Siah2 or proteasome alleviates pulmonary arterial remodeling through inactivation of YAP, indicating Siah2 ubiquitin ligase as a novel target might have potential value in the management of PAH.
Collapse
Affiliation(s)
- Qingting Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Wenhua Shi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Qianqian Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Wei Feng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jian Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Cui Zhai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xin Yan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
17
|
NADPH oxidases and oxidase crosstalk in cardiovascular diseases: novel therapeutic targets. Nat Rev Cardiol 2019; 17:170-194. [PMID: 31591535 DOI: 10.1038/s41569-019-0260-8] [Citation(s) in RCA: 350] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/19/2019] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS)-dependent production of ROS underlies sustained oxidative stress, which has been implicated in the pathogenesis of cardiovascular diseases such as hypertension, aortic aneurysm, hypercholesterolaemia, atherosclerosis, diabetic vascular complications, cardiac ischaemia-reperfusion injury, myocardial infarction, heart failure and cardiac arrhythmias. Interactions between different oxidases or oxidase systems have been intensively investigated for their roles in inducing sustained oxidative stress. In this Review, we discuss the latest data on the pathobiology of each oxidase component, the complex crosstalk between different oxidase components and the consequences of this crosstalk in mediating cardiovascular disease processes, focusing on the central role of particular NADPH oxidase (NOX) isoforms that are activated in specific cardiovascular diseases. An improved understanding of these mechanisms might facilitate the development of novel therapeutic agents targeting these oxidase systems and their interactions, which could be effective in the prevention and treatment of cardiovascular disorders.
Collapse
|
18
|
Deng Q, Hou J, Feng L, Lv A, Ke X, Liang H, Wang F, Zhang K, Chen K, Cui H. PHF19 promotes the proliferation, migration, and chemosensitivity of glioblastoma to doxorubicin through modulation of the SIAH1/β-catenin axis. Cell Death Dis 2018; 9:1049. [PMID: 30323224 PMCID: PMC6189144 DOI: 10.1038/s41419-018-1082-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 06/14/2018] [Accepted: 06/21/2018] [Indexed: 12/29/2022]
Abstract
PHD finger protein 19 (PHF19), a critical component of the polycomb repressive complex 2 (PRC2), is crucial for maintaining the repressive transcriptional activity of several developmental regulatory genes and plays essential roles in various biological processes. Abnormal expression of PHF19 causes dysplasia or serious diseases, including chronic myeloid disorders and tumors. However, the biological functions and molecular mechanisms of PHF19 in glioblastoma (GBM) remain unclear. Here, we demonstrated that PHF19 expression was positively associated with GBM progression, including cell proliferation, migration, invasion, chemosensitivity, and tumorigenesis. Using XAV-939, a Wnt/β-catenin inhibitor, we found that the effects of PHF19 on GBM cells were β-catenin-dependent. We also demonstrated that PHF19 expression was positively correlated with cytoplasmic β-catenin expression. PHF19 stabilized β-catenin by inhibiting the transcription of seven in absentia homolog 1 (SIAH1), an E3 ubiquitin ligase of β-catenin, through direct binding to the SIAH1 promoter region. Taken together, our results revealed the novel PHF19-SIAH1–β-catenin axis as a potential and promising therapeutic target.
Collapse
Affiliation(s)
- Qing Deng
- State Key Laboratory of Silkworm Genome Biology, The Institute of Sericulture and Systems Biology, Southwest University, 400716, Chongqing, People's Republic of China
| | - Jianbing Hou
- State Key Laboratory of Silkworm Genome Biology, The Institute of Sericulture and Systems Biology, Southwest University, 400716, Chongqing, People's Republic of China
| | - Liying Feng
- State Key Laboratory of Silkworm Genome Biology, The Institute of Sericulture and Systems Biology, Southwest University, 400716, Chongqing, People's Republic of China
| | - Ailing Lv
- State Key Laboratory of Silkworm Genome Biology, The Institute of Sericulture and Systems Biology, Southwest University, 400716, Chongqing, People's Republic of China
| | - Xiaoxue Ke
- State Key Laboratory of Silkworm Genome Biology, The Institute of Sericulture and Systems Biology, Southwest University, 400716, Chongqing, People's Republic of China
| | - Hanghua Liang
- State Key Laboratory of Silkworm Genome Biology, The Institute of Sericulture and Systems Biology, Southwest University, 400716, Chongqing, People's Republic of China
| | - Feng Wang
- State Key Laboratory of Silkworm Genome Biology, The Institute of Sericulture and Systems Biology, Southwest University, 400716, Chongqing, People's Republic of China
| | - Kui Zhang
- State Key Laboratory of Silkworm Genome Biology, The Institute of Sericulture and Systems Biology, Southwest University, 400716, Chongqing, People's Republic of China
| | - Kuijun Chen
- Department 6 of the Research Institute of Surgery, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, 400042, Chongqing, People's Republic of China
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, The Institute of Sericulture and Systems Biology, Southwest University, 400716, Chongqing, People's Republic of China.
| |
Collapse
|
19
|
Zhang H, Vreeken D, Bruikman CS, van Zonneveld AJ, van Gils JM. Understanding netrins and semaphorins in mature endothelial cell biology. Pharmacol Res 2018; 137:1-10. [PMID: 30240825 DOI: 10.1016/j.phrs.2018.09.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 09/15/2018] [Accepted: 09/15/2018] [Indexed: 02/07/2023]
Abstract
Netrins and semaphorins are known as neuronal guidance molecules that are important to the facilitate patterning of the nervous system in embryonic development. In recent years, their function has been broadened to guide development in other systems, including the vascular system, where netrins and semaphorins critically contribute to the development of the vascular system. Evidence is accumulating that these guidance cues are also of critical importance in the biology of the mature endothelium by regulating the maintenance of endothelial quiescence. Here we review our current insights into the roles of netrins and semaphorins in endothelial cell survival, self-renewing, barrier function, response to wall shear stress, and control of the vascular tone. We also provide suggestions for future research into the functions of netrins and semaphorins in mature endothelial cell biology.
Collapse
Affiliation(s)
- Huayu Zhang
- Department of Internal Medicine, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Dianne Vreeken
- Department of Internal Medicine, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Caroline S Bruikman
- Amsterdam UMC, University of Amsterdam, Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam, the Netherlands
| | - Anton Jan van Zonneveld
- Department of Internal Medicine, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Janine M van Gils
- Department of Internal Medicine, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands.
| |
Collapse
|
20
|
Toque HA, Fernandez-Flores A, Mohamed R, Caldwell RB, Ramesh G, Caldwell RW. Netrin-1 is a novel regulator of vascular endothelial function in diabetes. PLoS One 2017; 12:e0186734. [PMID: 29059224 PMCID: PMC5653335 DOI: 10.1371/journal.pone.0186734] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 10/08/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Netrin-1, a secreted laminin-like protein identified as an axon guidance molecule, has been shown to be of critical importance in the cardiovascular system. Recent studies have revealed pro-angiogenic, anti-apoptotic and anti-inflammatory properties of netrin-1 as well as cardioprotective actions against myocardial injury in diabetic mice. AIM To examine the role of netrin-1 in diabetes-and high glucose (HG)-induced vascular endothelial dysfunction (VED) using netrin-1 transgenic mice (Tg3) and cultured bovine aortic endothelial cells (BAEC). MAIN OUTCOME Overexpression of netrin-1 prevented diabetes-induced VED in aorta from diabetic mice and netrin-1 treatment attenuated HG-induced impairment of nitric oxide synthase (NOS) function in BAECs. METHODS AND RESULTS Experiments were performed in Tg3 and littermate control (WT) mice rendered diabetic with streptozotocin (STZ) and in BAECs treated with HG (25 mmol/L). Levels of netrin-1 and its receptor DCC, markers of inflammation and apoptosis and vascular function were assessed in aortas from diabetic and non-diabetic Tg3 and WT mice. Vascular netrin-1 in WT mice was reduced under diabetic conditions. Aortas from non-diabetic Tg3 and WT mice showed similar maximum endothelium-dependent relaxation (MEDR) (83% and 87%, respectively). MEDR was markedly impaired in aorta from diabetic WT mice (51%). This effect was significantly blunted in Tg3 diabetic aortas (70%). Improved vascular relaxation in Tg3 diabetic mice was associated with increased levels of phospho-ERK1/2 and reduced levels of oxidant stress, NFκB, COX-2, p16INK4A, cleaved caspase-3 and p16 and p53 mRNA. Netrin-1 treatment prevented the HG-induced decrease in NO production and elevation of oxidative stress and apoptosis in BAECs. CONCLUSIONS Diabetes decreases aortic levels of netrin-1. However, overexpression of netrin-1 attenuates diabetes-induced VED and limits the reduction of NO levels, while increasing expression of p-ERK1/2, and suppressing oxidative stress and inflammatory and apoptotic processes. Enhancement of netrin-1 function may be a useful therapeutic means for preventing vascular dysfunction in diabetes.
Collapse
Affiliation(s)
- Haroldo A. Toque
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
- * E-mail:
| | - Aracely Fernandez-Flores
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - Riyaz Mohamed
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - Ruth B. Caldwell
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States of America
| | - Ganesan Ramesh
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - R. William Caldwell
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| |
Collapse
|
21
|
Dissecting the Role of the Extracellular Matrix in Heart Disease: Lessons from the Drosophila Genetic Model. Vet Sci 2017; 4:vetsci4020024. [PMID: 29056683 PMCID: PMC5606597 DOI: 10.3390/vetsci4020024] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 02/15/2017] [Accepted: 04/20/2017] [Indexed: 12/16/2022] Open
Abstract
The extracellular matrix (ECM) is a dynamic scaffold within organs and tissues that enables cell morphogenesis and provides structural support. Changes in the composition and organisation of the cardiac ECM are required for normal development. Congenital and age-related cardiac diseases can arise from mis-regulation of structural ECM proteins (Collagen, Laminin) or their receptors (Integrin). Key regulators of ECM turnover include matrix metalloproteinases (MMPs) and their inhibitors, tissue inhibitors of matrix metalloproteinases (TIMPs). MMP expression is increased in mice, pigs, and dogs with cardiomyopathy. The complexity and longevity of vertebrate animals makes a short-lived, genetically tractable model organism, such as Drosophila melanogaster, an attractive candidate for study. We survey ECM macromolecules and their role in heart development and growth, which are conserved between Drosophila and vertebrates, with focus upon the consequences of altered expression or distribution. The Drosophila heart resembles that of vertebrates during early development, and is amenable to in vivo analysis. Experimental manipulation of gene function in a tissue- or temporally-regulated manner can reveal the function of adhesion or ECM genes in the heart. Perturbation of the function of ECM proteins, or of the MMPs that facilitate ECM remodelling, induces cardiomyopathies in Drosophila, including cardiodilation, arrhythmia, and cardia bifida, that provide mechanistic insight into cardiac disease in mammals.
Collapse
|
22
|
|
23
|
Attenuation of neointimal formation with netrin-1 and netrin-1 preconditioned endothelial progenitor cells. J Mol Med (Berl) 2016; 95:335-348. [PMID: 28004124 DOI: 10.1007/s00109-016-1490-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 10/10/2016] [Accepted: 11/15/2016] [Indexed: 10/20/2022]
Abstract
Restenosis after angioplasty is a serious clinical problem that can result in re-occlusion of the coronary artery. Although current drug-eluting stents have proved to be more effective in reducing restenosis, they have drawbacks of inhibiting reendothelialization to promote thrombosis. New treatment options are in urgent need. We have shown that netrin-1, an axon-guiding protein, promotes angiogenesis and cardioprotection via production of nitric oxide (NO). The present study examined whether and how netrin-1 attenuates neointimal formation in a femoral wire injury model. Infusion of netrin-1 into C57BL/6 mice markedly attenuated neointimal formation following wire injury of femoral arteries, measured by intimal to media ratio (from 1.94 ± 0.55 to 0.45 ± 0.86 at 4 weeks). Proliferation of VSMC in situ was largely reduced. This protective effect was absent in DCC+/- animals. NO production was increased by netrin-1 in both intact and injured femoral arteries, indicating netrin-1 stimulation of endogenous NO production from intact endothelium and remaining endothelial cells post-injury. VSMC migration was abrogated by netrin-1 via a NO/cGMP/p38 MAPK pathway, while timely EPC homing was induced. Injection of netrin-1 preconditioned wild-type EPCs, but not EPCs of DCC+/- animals, substantially attenuated neointimal formation. EPC proliferation, NO production, and resistance to oxidative stress induced apoptosis were augmented by netrin-1 treatment. In conclusion, our data for the first time demonstrate that netrin-1 is highly effective in reducing neointimal formation following vascular endothelial injury, which is dependent on DCC, and attributed to inhibition of VSMC proliferation and migration, as well as improved EPC function. These data may support usage of netrin-1 and netrin-1 preconditioned EPCs as novel therapies for post angioplasty restenosis. KEY MESSAGE Netrin-1 attenuates neointimal formation following post endothelial injury via DCC and NO. Netrin-1 inhibits VSMC proliferation in situ following endothelial injury. Netrin-1 inhibits VSMC migration via a NO/cGMP/p38 MAPK pathway. Netrin-1 augments proliferation of endothelial progenitor cells (EPCs) and EPC eNOS/NO activation. Netrin-1 enhances resistance of EPCs to oxidative stress, improving re-endothelialization following injury.
Collapse
|
24
|
Wang Z, Zhang XJ, Ji YX, Zhang P, Deng KQ, Gong J, Ren S, Wang X, Chen I, Wang H, Gao C, Yokota T, Ang YS, Li S, Cass A, Vondriska TM, Li G, Deb A, Srivastava D, Yang HT, Xiao X, Li H, Wang Y. The long noncoding RNA Chaer defines an epigenetic checkpoint in cardiac hypertrophy. Nat Med 2016; 22:1131-1139. [PMID: 27618650 PMCID: PMC5053883 DOI: 10.1038/nm.4179] [Citation(s) in RCA: 309] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 08/05/2016] [Indexed: 12/13/2022]
Abstract
Epigenetic reprogramming is a critical process of pathological gene induction during cardiac hypertrophy and remodeling, but the underlying regulatory mechanisms remain to be elucidated. Here we identified a heart-enriched long noncoding (lnc)RNA, named cardiac-hypertrophy-associated epigenetic regulator (Chaer), which is necessary for the development of cardiac hypertrophy. Mechanistically, Chaer directly interacts with the catalytic subunit of polycomb repressor complex 2 (PRC2). This interaction, which is mediated by a 66-mer motif in Chaer, interferes with PRC2 targeting to genomic loci, thereby inhibiting histone H3 lysine 27 methylation at the promoter regions of genes involved in cardiac hypertrophy. The interaction between Chaer and PRC2 is transiently induced after hormone or stress stimulation in a process involving mammalian target of rapamycin complex 1, and this interaction is a prerequisite for epigenetic reprogramming and induction of genes involved in hypertrophy. Inhibition of Chaer expression in the heart before, but not after, the onset of pressure overload substantially attenuates cardiac hypertrophy and dysfunction. Our study reveals that stress-induced pathological gene activation in the heart requires a previously uncharacterized lncRNA-dependent epigenetic checkpoint.
Collapse
Affiliation(s)
- Zhihua Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Animal Experiment Center–Animal Biosafety Level 3 Laboratory, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine, University of California at Los Angeles (UCLA), Los Angeles, California, USA
| | - Xiao-Jing Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Animal Experiment Center–Animal Biosafety Level 3 Laboratory, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Yan-Xiao Ji
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Animal Experiment Center–Animal Biosafety Level 3 Laboratory, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Peng Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Animal Experiment Center–Animal Biosafety Level 3 Laboratory, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Ke-Qiong Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Animal Experiment Center–Animal Biosafety Level 3 Laboratory, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Jun Gong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Animal Experiment Center–Animal Biosafety Level 3 Laboratory, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Shuxun Ren
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine, University of California at Los Angeles (UCLA), Los Angeles, California, USA
| | - Xinghua Wang
- Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Iris Chen
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine, University of California at Los Angeles (UCLA), Los Angeles, California, USA
| | - He Wang
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine, University of California at Los Angeles (UCLA), Los Angeles, California, USA
| | - Chen Gao
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine, University of California at Los Angeles (UCLA), Los Angeles, California, USA
| | - Tomohiro Yokota
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine, University of California at Los Angeles (UCLA), Los Angeles, California, USA
| | - Yen Sin Ang
- Gladstone Institute of Cardiovascular Diseases, San Francisco, California, USA
- University of California San Francisco, School of Medicine, San Francisco, California, USA
| | - Shen Li
- Department of Medicine, Cardiology Division, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, USA
- Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, USA
| | - Ashley Cass
- Department of Integrative Biology and Physiology, College of Life Sciences, Molecular Biology Institute, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, USA
- Bioinformatics Interdepartmental Program, University of California at Los Angeles, Los Angeles, California, USA
| | - Thomas M. Vondriska
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine, University of California at Los Angeles (UCLA), Los Angeles, California, USA
| | - Guangping Li
- Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Arjun Deb
- Department of Medicine, Cardiology Division, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, USA
- Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, USA
| | - Deepak Srivastava
- Gladstone Institute of Cardiovascular Diseases, San Francisco, California, USA
- University of California San Francisco, School of Medicine, San Francisco, California, USA
| | - Huang-Tian Yang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinshu Xiao
- Department of Integrative Biology and Physiology, College of Life Sciences, Molecular Biology Institute, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, USA
- Bioinformatics Interdepartmental Program, University of California at Los Angeles, Los Angeles, California, USA
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Animal Experiment Center–Animal Biosafety Level 3 Laboratory, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Yibin Wang
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine, University of California at Los Angeles (UCLA), Los Angeles, California, USA
- Department of Medicine, Cardiology Division, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, USA
- Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, USA
| |
Collapse
|
25
|
Teoh JP, Park KM, Broskova Z, Jimenez FR, Bayoumi AS, Archer K, Su H, Johnson J, Weintraub NL, Tang Y, Kim IM. Identification of gene signatures regulated by carvedilol in mouse heart. Physiol Genomics 2015; 47:376-85. [PMID: 26152686 DOI: 10.1152/physiolgenomics.00028.2015] [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] [Received: 03/30/2015] [Accepted: 07/06/2015] [Indexed: 01/14/2023] Open
Abstract
Chronic treatment with the β-blocker carvedilol has been shown to reduce established maladaptive left ventricle (LV) hypertrophy and to improve LV function in experimental heart failure. However, the detailed mechanisms by which carvedilol improves LV failure are incompletely understood. We previously showed that carvedilol is a β-arrestin-biased β1-adrenergic receptor ligand, which activates cellular pathways in the heart independent of G protein-mediated second messenger signaling. More recently, we have demonstrated by microRNA (miR) microarray analysis that carvedilol upregulates a subset of mature and pre-mature miRs, but not their primary miR transcripts in mouse hearts. Here, we next sought to identify the effects of carvedilol on LV gene expression on a genome-wide basis. Adult mice were treated with carvedilol or vehicle for 1 wk. RNA was isolated from LV tissue and hybridized for microarray analysis. Gene expression profiling analysis revealed a small group of genes differentially expressed after carvedilol treatment. Further analysis categorized these genes into pathways involved in tight junction, malaria, viral myocarditis, glycosaminoglycan biosynthesis, and arrhythmogenic right ventricular cardiomyopathy. Genes encoding proteins in the tight junction, malaria, and viral myocarditis pathways were upregulated in the LV by carvedilol, while genes encoding proteins in the glycosaminoglycan biosynthesis and arrhythmogenic right ventricular cardiomyopathy pathways were downregulated by carvedilol. These gene expression changes may reflect the molecular mechanisms that underlie the functional benefits of carvedilol therapy.
Collapse
Affiliation(s)
- Jian-Peng Teoh
- Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, Georgia
| | - Kyoung-Mi Park
- Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, Georgia
| | - Zuzana Broskova
- Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, Georgia
| | - Felix R Jimenez
- Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, Georgia
| | - Ahmed S Bayoumi
- Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, Georgia
| | - Krystal Archer
- Department of Medicine, Medical College of Georgia, Georgia Regents University, Augusta, Georgia; and
| | - Huabo Su
- Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, Georgia; Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia
| | - John Johnson
- Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia
| | - Neal L Weintraub
- Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, Georgia; Department of Medicine, Medical College of Georgia, Georgia Regents University, Augusta, Georgia; and
| | - Yaoliang Tang
- Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, Georgia; Department of Medicine, Medical College of Georgia, Georgia Regents University, Augusta, Georgia; and
| | - Il-Man Kim
- Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, Georgia; Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia
| |
Collapse
|
26
|
Cui MZ. Potential therapeutics for myocardial ischemia-reperfusion injury. Focus on "Induction of cardioprotection by small netrin-1-derived peptides". Am J Physiol Cell Physiol 2015; 309:C97-9. [PMID: 26040896 DOI: 10.1152/ajpcell.00150.2015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Mei-Zhen Cui
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee
| |
Collapse
|
27
|
Li Q, Cai H. Induction of cardioprotection by small netrin-1-derived peptides. Am J Physiol Cell Physiol 2015; 309:C100-6. [PMID: 25924621 DOI: 10.1152/ajpcell.00332.2014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 04/22/2015] [Indexed: 11/22/2022]
Abstract
We have shown that netrin-1 induces potent cardioprotection via extracellular signal-regulated kinases 1 and 2 (ERK1/2)-dependent endothelial nitric oxide synthase (eNOS)/NO activation. The present study investigated cardioprotective effects of small netrin-1-derived peptides. We synthesized three laminin (Lam) V peptides and found those time dependently increased phosphorylation of ERK1/2 and eNOSs1179 in endothelial cells at the same molar concentration used for netrin-1. Preperfusion with Lam V peptides induced a substantial reduction in infarct size (control 39.3 ± 0.2% vs. 14.6 ± 2.3%, 23.0 ± 2.8%, and 18.8 ± 0.8% for V1, V2, and V3, respectively). Furthermore, reperfusion with all three also induced potent cardioprotection (control 37.6 ± 1.3% vs. 17.6 ± 3.2%, 20.6 ± 1.7%, and 15.8 ± 2.0% for V1, V2, and V3, respectively), implicating that these peptides are consistently beneficial whenever they are delivered to the heart. Based on the sequence alignment, we found a region of high sequence homology and synthesized smaller peptides [V1-9 amino acid (aa), V2-10aa, and V3-11aa]. These smaller peptides markedly reduced infarct size during reperfusion (V1-9aa 16.8 ± 2.2%, V2-10aa 18.6 ± 1.7%, and V3-11aa 16.7 ± 3.0% vs. control 37.6 ± 1.3%). A negative control V3-16aa with no sequence homology failed to protect the heart. Of note, the core area has the characteristic sequence of: Cx(1-2)Cx(3-4)Tx(0-1)G. Furthermore, all three smaller peptides induced NO production in endothelial cells that could in turn diffuse to cardiomyocytes to promote survival. Combined applications of V1-9aa and V2-10aa synergistically induced more NO production. Taken together, these data strongly suggest that small netrin-1-derived peptides are highly effective in protecting the heart against myocardial ischemia-reperfusion injury and has the potential to be developed into peptide drugs directly applicable to the treatment of myocardial infarction.
Collapse
Affiliation(s)
- Qiang Li
- Divisions of Molecular Medicine and Cardiology, Departments of Anesthesiology and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California
| | - Hua Cai
- Divisions of Molecular Medicine and Cardiology, Departments of Anesthesiology and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California
| |
Collapse
|