1
|
Cheng Y, Zhai Y, Yuan Y, Wang Q, Li S, Sun H. The Contributions of Thrombospondin-1 to Epilepsy Formation. Neurosci Bull 2024; 40:658-672. [PMID: 38528256 PMCID: PMC11127911 DOI: 10.1007/s12264-024-01194-2] [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: 06/27/2023] [Accepted: 01/27/2024] [Indexed: 03/27/2024] Open
Abstract
Epilepsy is a neural network disorder caused by uncontrolled neuronal hyperexcitability induced by an imbalance between excitatory and inhibitory networks. Abnormal synaptogenesis plays a vital role in the formation of overexcited networks. Recent evidence has confirmed that thrombospondin-1 (TSP-1), mainly secreted by astrocytes, is a critical cytokine that regulates synaptogenesis during epileptogenesis. Furthermore, numerous studies have reported that TSP-1 is also involved in other processes, such as angiogenesis, neuroinflammation, and regulation of Ca2+ homeostasis, which are closely associated with the occurrence and development of epilepsy. In this review, we summarize the potential contributions of TSP-1 to epilepsy development.
Collapse
Affiliation(s)
- Yao Cheng
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Yujie Zhai
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Yi Yuan
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Qiaoyun Wang
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Shucui Li
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China.
| | - Hongliu Sun
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China.
| |
Collapse
|
2
|
Shan S, Liu F, Ford E, Caldwell RB, Narayanan SP, Somanath PR. Triciribine attenuates pathological neovascularization and vascular permeability in a mouse model of proliferative retinopathy. Biomed Pharmacother 2023; 162:114714. [PMID: 37080089 DOI: 10.1016/j.biopha.2023.114714] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/11/2023] [Accepted: 04/14/2023] [Indexed: 04/22/2023] Open
Abstract
Proliferative retinopathies are the leading cause of irreversible blindness in all ages, and there is a critical need to identify novel therapies. We investigated the impact of triciribine (TCBN), a tricyclic nucleoside analog and a weak Akt inhibitor, on retinal neurovascular injury, vascular permeability, and inflammation in oxygen-induced retinopathy (OIR). Post-natal day 7 (P7) mouse pups were subjected to OIR, and treated (i.p.) with TCBN or vehicle from P14-P16 and compared with age-matched, normoxic, vehicle or TCBN-treated controls. P17 retinas were processed for flat mounts, immunostaining, Western blotting, and qRT-PCR studies. Fluorescein angiography, electroretinography, and spectral domain optical coherence tomography were performed on days P21, P26, and P30, respectively. TCBN treatment significantly reduced pathological neovascularization, vaso-obliteration, and inflammation marked by reduced TNFα, IL6, MCP-1, Iba1, and F4/80 (macrophage/microglia markers) expression compared to the vehicle-treated OIR mouse retinas. Pathological expression of VEGF (vascular endothelial growth factor), and claudin-5 compromised the blood-retinal barrier integrity in the OIR retinas correlating with increased vascular permeability and neovascular tuft formation, which were blunted by TCBN treatment. Of note, there were no changes in the retinal architecture or retinal cell function in response to TCBN in the normoxia or OIR mice. We conclude that TCBN protects against pathological neovascularization, restores blood-retinal barrier homeostasis, and reduces retinal inflammation without adversely affecting the retinal structure and neuronal function in a mouse model of OIR. Our data suggest that TCBN may provide a novel therapeutic option for proliferative retinopathy.
Collapse
Affiliation(s)
- Shengshuai Shan
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, 30912, USA; Research Department, Charlie Norwood VA Medical Center, Augusta, GA, 30901, USA; Vascular Biology Center, Augusta University, Augusta, GA, 30912, USA; Culver Vision Discovery Institute, Augusta University, Augusta, GA, 30912, USA
| | - Fang Liu
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, 30912, USA; Research Department, Charlie Norwood VA Medical Center, Augusta, GA, 30901, USA; Vascular Biology Center, Augusta University, Augusta, GA, 30912, USA; Culver Vision Discovery Institute, Augusta University, Augusta, GA, 30912, USA
| | - Edith Ford
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, 30912, USA
| | - Ruth B Caldwell
- Vascular Biology Center, Augusta University, Augusta, GA, 30912, USA; Culver Vision Discovery Institute, Augusta University, Augusta, GA, 30912, USA
| | - S Priya Narayanan
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, 30912, USA; Research Department, Charlie Norwood VA Medical Center, Augusta, GA, 30901, USA; Vascular Biology Center, Augusta University, Augusta, GA, 30912, USA; Culver Vision Discovery Institute, Augusta University, Augusta, GA, 30912, USA.
| | - Payaningal R Somanath
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, 30912, USA; Research Department, Charlie Norwood VA Medical Center, Augusta, GA, 30901, USA; Culver Vision Discovery Institute, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
3
|
Yu Y, Zheng C, Lu X, Deng C, Xu Q, Guo W, Wu Q, Wang Q, Liu C, Huang X, Song J. GB1a Ameliorates Ulcerative Colitis via Regulation of the NF-κB and Nrf2 Signaling Pathways in an Experimental Model. Front Med (Lausanne) 2021; 8:654867. [PMID: 34557497 PMCID: PMC8452853 DOI: 10.3389/fmed.2021.654867] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 08/09/2021] [Indexed: 11/13/2022] Open
Abstract
Ulcerative colitis (UC) is an inflammatory bowel disease. The intake of African Garcinia Kola nuts has been reported as a therapy for diarrhea and dysentery in the African population. However, the mechanism of action through which Garcinia Kola nuts act to ameliorates UC remains unknown. GB1a is the main active component of Garcinia Kola nuts. In this study, we explored the therapeutic effects and underlying mechanism of GB1a on dextran sodium sulfate (DSS)-induced UC. Human Colonic Epithelial Cells (HCoEpic) were challenged with TNF-α to test the effects of GB1a in protecting against oxidative stress and inflammation in vitro. Our data showed that GB1a significantly attenuated DSS-induced colonic inflammatory injury manifested as reversed loss of body weight and disease activity index (DAI) scores in UC mice. We also showed that GB1a improved the permeability of the intestinal epithelium by modulating the expression of tight junction proteins (ZO-1, Occludin). Mechanistically, GB1a may activate the Nrf2 antioxidant signaling pathway and suppress the nuclear translocation of NF-κB in reduced oxidative stress and expression of inflammatory genes induced by TNF-α in HCoEpic cells. Our study suggests that GB1a alleviates inflammation, oxidative stress and the permeability of the colonic epithelial mucosa in UC mice via the repression of NF-κB and activation of Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Yuanyuan Yu
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Congmin Zheng
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xu Lu
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Changsheng Deng
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,The First Affiliated Hospital and The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qin Xu
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,The First Affiliated Hospital and The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenfeng Guo
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,The First Affiliated Hospital and The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qingye Wu
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Wang
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Changhui Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinan Huang
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,The First Affiliated Hospital and The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianping Song
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,The First Affiliated Hospital and The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
4
|
Adil MS, Verma A, Rudraraju M, Narayanan SP, Somanath PR. Akt-independent effects of triciribine on ACE2 expression in human lung epithelial cells: Potential benefits in restricting SARS-CoV2 infection. J Cell Physiol 2021; 236:6597-6606. [PMID: 33624300 PMCID: PMC8014085 DOI: 10.1002/jcp.30343] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 02/05/2023]
Abstract
The severe acute respiratory syndrome coronavirus 2 that causes coronavirus disease 2019 (COVID-19) binds to the angiotensin-converting enzyme 2 (ACE2) to gain cellular entry. Akt inhibitor triciribine (TCBN) has demonstrated promising results in promoting recovery from advanced-stage acute lung injury in preclinical studies. In the current study, we tested the direct effect of TCBN on ACE2 expression in human bronchial (H441) and lung alveolar (A549) epithelial cells. Treatment with TCBN resulted in the downregulation of both messenger RNA and protein levels of ACE2 in A549 cells. Since HMGB1 plays a vital role in the inflammatory response in COVID-19, and because hyperglycemia has been linked to increased COVID-19 infections, we determined if HMGB1 and hyperglycemia have any effect on ACE2 expression in lung epithelial cells and whether TCBN has any effect on reversing HMGB1- and hyperglycemia-induced ACE2 expression. We observed increased ACE2 expression with both HMGB1 and hyperglycemia treatment in A549 as well as H441 cells, which were blunted by TCBN treatment. Our findings from this study, combined with our previous reports on the potential benefits of TCBN in the treatment of acute lung injury, generate reasonable optimism on the potential utility of TCBN in the therapeutic management of patients with COVID-19.
Collapse
Affiliation(s)
- Mir S. Adil
- Clinical and Experimental Therapeutics, Department of Clinical and Administrative Pharmacy, College of Pharmacy, and Charlie Norwood VA Medical CenterUniversity of GeorgiaAugustaGeorgiaUSA
| | - Arti Verma
- Clinical and Experimental Therapeutics, Department of Clinical and Administrative Pharmacy, College of Pharmacy, and Charlie Norwood VA Medical CenterUniversity of GeorgiaAugustaGeorgiaUSA
| | - Madhuri Rudraraju
- Clinical and Experimental Therapeutics, Department of Clinical and Administrative Pharmacy, College of Pharmacy, and Charlie Norwood VA Medical CenterUniversity of GeorgiaAugustaGeorgiaUSA
| | - S. Priya Narayanan
- Clinical and Experimental Therapeutics, Department of Clinical and Administrative Pharmacy, College of Pharmacy, and Charlie Norwood VA Medical CenterUniversity of GeorgiaAugustaGeorgiaUSA,Vision Discovery Institute, Medical College of GeorgiaAugusta UniversityAugustaGeorgiaUSA
| | - Payaningal R. Somanath
- Clinical and Experimental Therapeutics, Department of Clinical and Administrative Pharmacy, College of Pharmacy, and Charlie Norwood VA Medical CenterUniversity of GeorgiaAugustaGeorgiaUSA,Vascular Biology Center, Medical College of GeorgiaAugusta UniversityAugustaGeorgiaUSA,Department of MedicineAugusta UniversityAugustaGeorgiaUSA
| |
Collapse
|
5
|
Yamagata K. Prevention of Endothelial Dysfunction and Cardiovascular Disease by n-3 Fatty Acids-Inhibiting Action on Oxidative Stress and Inflammation. Curr Pharm Des 2021; 26:3652-3666. [PMID: 32242776 DOI: 10.2174/1381612826666200403121952] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/11/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND Prospective cohort studies and randomized controlled trials have shown the protective effect of n-3 fatty acids against cardiovascular disease (CVD). The effect of n-3 fatty acids on vascular endothelial cells indicates their possible role in CVD prevention. OBJECTIVE Here, we describe the effect of docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) on endothelial dysfunction-caused by inflammation and oxidative stress-and their role in the development of CVD. METHODS We reviewed epidemiological studies done on n-3 fatty acids in CVD. The effect of DHA and EPA on vascular endothelial cells was examined with regard to changes in various markers, such as arteriosclerosis, inflammation, and oxidative stress, using cell and animal models. RESULTS Epidemiological studies revealed that dietary intake of EPA and DHA was associated with a reduced risk of various CVDs. EPA and DHA inhibited various events involved in arteriosclerosis development by preventing oxidative stress and inflammation associated with endothelial cell damage. In particular, EPA and DHA prevented endothelial cell dysfunction mediated by inflammatory responses and oxidative stress induced by events related to CVD. DHA and EPA also increased eNOS activity and induced nitric oxide production. CONCLUSION The effects of DHA and EPA on vascular endothelial cell damage and dysfunction may involve the induction of nitric oxide, in addition to antioxidant and anti-inflammatory effects. n-3 fatty acids inhibit endothelial dysfunction and prevent arteriosclerosis. Therefore, the intake of n-3 fatty acids may prevent CVDs, like myocardial infarction and stroke.
Collapse
Affiliation(s)
- Kazuo Yamagata
- College of Bioresource Science, Nihon University (UNBS), Kanagawa, Japan
| |
Collapse
|
6
|
Rudraraju M, Narayanan SP, Somanath PR. Regulation of blood-retinal barrier cell-junctions in diabetic retinopathy. Pharmacol Res 2020; 161:105115. [PMID: 32750417 PMCID: PMC7755666 DOI: 10.1016/j.phrs.2020.105115] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/22/2020] [Accepted: 07/27/2020] [Indexed: 12/15/2022]
Abstract
Loss of the blood-retinal barrier (BRB) integrity and subsequent damage to the neurovascular unit in the retina are the underlying reasons for diabetic retinopathy (DR). Damage to BRB eventually leads to severe visual impairment in the absence of prompt intervention. Diabetic macular edema and proliferative DR are the advanced stages of the disease where BRB integrity is altered. Primary mechanisms contributing to BRB dysfunction include loss of cell-cell barrier junctions, vascular endothelial growth factor, advanced glycation end products-induced damage, and oxidative stress. Although much is known about the involvement of adherens and tight-junction proteins in the regulation of vascular permeability in various diseases, there is a significant gap in our knowledge on the junctional proteins expressed in the BRB and how BRB function is modulated in the diabetic retina. In this review article, we present our current understanding of the molecular composition of BRB, the changes in the BRB junctional protein turnover in DR, and how BRB functional modulation affects vascular permeability and macular edema in the diabetic retina.
Collapse
Affiliation(s)
- Madhuri Rudraraju
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, United States; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA 30912, United States
| | - S Priya Narayanan
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, United States; Vascular Biology Center, Augusta University, Augusta, GA 30912, United States; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA 30912, United States
| | - Payaningal R Somanath
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, United States; Vascular Biology Center, Augusta University, Augusta, GA 30912, United States; Department of Medicine, Augusta University, Augusta, GA 30912, United States.
| |
Collapse
|
7
|
Sabbineni H, Verma A, Artham S, Anderson D, Amaka O, Liu F, Narayanan SP, Somanath PR. Pharmacological inhibition of β-catenin prevents EndMT in vitro and vascular remodeling in vivo resulting from endothelial Akt1 suppression. Biochem Pharmacol 2019; 164:205-215. [PMID: 30991049 DOI: 10.1016/j.bcp.2019.04.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 04/12/2019] [Indexed: 12/31/2022]
Abstract
Endothelial to mesenchymal transition (EndMT), where endothelial cells acquire mesenchymal characteristics has been implicated in several cardiopulmonary, vascular and fibrotic diseases. The most commonly studied molecular mechanisms involved in EndMT include TGFβ, Notch, interleukin, and interferon-γ signaling. As of today, the contributions of Akt1, an important mediator of TGFβ signaling and a key regulator of endothelial barrier function to EndMT remains unclear. By using the ShRNA based gene silencing approach and endothelial-specific inducible Akt1 knockdown (ECKOAkt1) mice, we studied the role of Akt1 in EndMT in vitro and pathological vascular remodeling in vivo. Stable, Akt1 silenced (ShAkt1) human microvascular endothelial cells (HMECs) indicated increased expression of mesenchymal markers such as N-cadherin and α-SMA, phosphorylation of Smad2/3, cellular stress via activation of p38 MAP Kinase and the loss of endothelial nitric oxide synthase (eNOS) accompanied by a change in the morphology of HMECs in vitro and co-localization of endothelial and mesenchymal markers promoting EndMT in vivo. EndMT as a result of Akt1 loss was associated with increased expression of TGFβ2, a potent inducer of EndMT and mesenchymal transcription factors Snail1, and FoxC2. We observed that hypoxia-induced lung vascular remodeling is exacerbated in ECKOAkt1 mice, which was reversed by pharmacological inhibition of β-catenin. Thus, we provide novel insights into the role of Akt1-mediated β-catenin signaling in EndMT and pathological vascular remodeling, and present β-catenin as a potential target for therapy for various cardiopulmonary diseases involving vascular remodeling.
Collapse
Affiliation(s)
- Harika Sabbineni
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, United States
| | - Arti Verma
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, United States
| | - Sandeep Artham
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, United States
| | - Daniel Anderson
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, United States
| | - Oge Amaka
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, United States
| | - Fang Liu
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, United States
| | - Subhadra P Narayanan
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, United States
| | - Payaningal R Somanath
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, United States; Department of Medicine, Vascular Biology Center and Cancer Center, Augusta University, Augusta, GA 30912, United States.
| |
Collapse
|
8
|
Artham S, Gao F, Verma A, Alwhaibi A, Sabbineni H, Hafez S, Ergul A, Somanath PR. Endothelial stromelysin1 regulation by the forkhead box-O transcription factors is crucial in the exudative phase of acute lung injury. Pharmacol Res 2019; 141:249-263. [PMID: 30611853 DOI: 10.1016/j.phrs.2019.01.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 12/11/2018] [Accepted: 01/02/2019] [Indexed: 12/18/2022]
Abstract
Enhanced vascular permeability is associated with inflammation and edema in alveoli during the exudative phase of acute respiratory distress syndrome (ARDS). Mechanisms leading to the endothelial contribution on the early exudative stage of ARDS are not precise. We hypothesized that modulation of endothelial stromelysin1 expression and activity by Akt1-forkhead box-O transcription factors 1/3a (FoxO1/3a) pathway could play a significant role in regulating pulmonary edema during the initial stages of acute lung injury (ALI). We utilized lipopolysaccharide (LPS)-induced mouse ALI model in vivo and endothelial barrier resistance measurements in vitro to determine the specific role of the endothelial Akt1-FoxO1/3a-stromelysin1 pathway in ALI. LPS treatment of human pulmonary endothelial cells resulted in increased stromelysin1 and reduced tight junction claudin5 involving FoxO1/3a, associated with decreased trans-endothelial barrier resistance as determined by electric cell-substrate impedance sensing technology. In vivo, LPS-induced lung edema was significantly higher in endothelial Akt1 knockdown (EC-Akt1-/-) compared to wild-type mice, which was reversed upon treatment with FoxO inhibitor (AS1842856), stromelysin1 inhibitor (UK356618) or with shRNA-mediated FoxO1/3a depletion in the mouse lungs. Overall, our study provides the hope that targeting FoxO and styromelysin1 could be beneficial in the treatment of ALI.
Collapse
Affiliation(s)
- Sandeep Artham
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, United States
| | - Fei Gao
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, United States; Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Arti Verma
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, United States
| | - Abdulrahman Alwhaibi
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, United States
| | - Harika Sabbineni
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, United States
| | - Sherif Hafez
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, United States; Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, Miami, FL 33169, United States
| | - Adviye Ergul
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, United States; Department of Physiology, Augusta University, Augusta, GA 30912, United States
| | - Payaningal R Somanath
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, United States; Department of Medicine, Vascular Biology Center and Cancer Center, Augusta University, Augusta, GA 30912, United States.
| |
Collapse
|
9
|
Giacomelli C, Natali L, Nisi M, De Leo M, Daniele S, Costa B, Graziani F, Gabriele M, Braca A, Trincavelli ML, Martini C. Negative effects of a high tumour necrosis factor-α concentration on human gingival mesenchymal stem cell trophism: the use of natural compounds as modulatory agents. Stem Cell Res Ther 2018; 9:135. [PMID: 29751776 PMCID: PMC5948671 DOI: 10.1186/s13287-018-0880-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 04/08/2018] [Accepted: 04/19/2018] [Indexed: 12/31/2022] Open
Abstract
Background Adult mesenchymal stem cells (MSCs) play a crucial role in the maintenance of tissue homeostasis and in regenerative processes. Among the different MSC types, the gingiva-derived mesenchymal stem cells (GMSCs) have arisen as a promising tool to promote the repair of damaged tissues secreting trophic mediators that affect different types of cells involved in regenerative processes. Tumour necrosis factor (TNF)-α is one of the key mediators of inflammation that could affect tissue regenerative processes and modify the MSC properties in in-vitro applications. To date, no data have been reported on the effects of TNF-α on GMSC trophic activities and how its modulation with anti-inflammatory agents from natural sources could modulate the GMSC properties. Methods GMSCs were isolated and characterized from healthy subjects. The effects of TNF-α were evaluated on GMSCs and on the well-being of endothelial cells. The secretion of cytokines was measured and related to the modification of GMSC-endothelial cell communication using a conditioned-medium method. The ability to modify the inflammatory response was evaluated in the presence of Ribes nigrum bud extract (RBE). Results TNF-α differently affected GMSC proliferation and the expression of inflammatory-related proteins (interleukin (IL)-6, IL-10, transforming growth factor (TGF)-β, and cyclooxygenase (COX)-2) dependent on its concentration. A high TNF-α concentration decreased the GMSC viability and impaired the positive cross-talk between GMSCs and endothelial cells, probably by enhancing the amount of pro-inflammatory cytokines in the GMSC secretome. RBE restored the beneficial effects of GMSCs on endothelial viability and motility under inflammatory conditions. Conclusions A high TNF-α concentration decreased the well-being of GMSCs, modifying their trophic activities and decreasing endothelial cell healing. These data highlight the importance of controlling TNF-α concentrations to maintain the trophic activity of GMSCs. Furthermore, the use of natural anti-inflammatory agents restored the regenerative properties of GMSCs on endothelial cells, opening the way to the use and development of natural extracts in wound healing, periodontal regeneration, and tissue-engineering applications that use MSCs. Electronic supplementary material The online version of this article (10.1186/s13287-018-0880-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chiara Giacomelli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Letizia Natali
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Marco Nisi
- Department of Surgical, Medical, Molecular and Critical Area Pathology, University of Pisa, Via Savi 10, 56126, Pisa, Italy
| | - Marinella De Leo
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy.,Centro Interdipartimentale di Ricerca "Nutraceutica e Alimentazione per la Salute", University of Pisa, Via del Borghetto 80, 56124, Pisa, Italy
| | - Simona Daniele
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Barbara Costa
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Filippo Graziani
- Department of Surgical, Medical, Molecular and Critical Area Pathology, University of Pisa, Via Savi 10, 56126, Pisa, Italy.,Centro Interdipartimentale di Ricerca "Nutraceutica e Alimentazione per la Salute", University of Pisa, Via del Borghetto 80, 56124, Pisa, Italy
| | - Mario Gabriele
- Department of Surgical, Medical, Molecular and Critical Area Pathology, University of Pisa, Via Savi 10, 56126, Pisa, Italy
| | - Alessandra Braca
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy.,Centro Interdipartimentale di Ricerca "Nutraceutica e Alimentazione per la Salute", University of Pisa, Via del Borghetto 80, 56124, Pisa, Italy
| | - M Letizia Trincavelli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy. .,Centro Interdipartimentale di Ricerca "Nutraceutica e Alimentazione per la Salute", University of Pisa, Via del Borghetto 80, 56124, Pisa, Italy.
| | - Claudia Martini
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy.,Centro Interdipartimentale di Ricerca "Nutraceutica e Alimentazione per la Salute", University of Pisa, Via del Borghetto 80, 56124, Pisa, Italy
| |
Collapse
|
10
|
DeVallance E, Branyan KW, Lemaster K, Olfert IM, Smith DM, Pistilli EE, Frisbee JC, Chantler PD. Aortic dysfunction in metabolic syndrome mediated by perivascular adipose tissue TNFα- and NOX2-dependent pathway. Exp Physiol 2018; 103:590-603. [PMID: 29349831 DOI: 10.1113/ep086818] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 01/15/2018] [Indexed: 12/17/2022]
Abstract
NEW FINDINGS What is the central question of this study? Tumour necrosis factor-α (TNFα) has been shown to impair vascular function, but the impact of thoracic aorta perivascular adipose tissue (tPVAT)-derived TNFα on tPVAT and aortic function in metabolic syndrome is unknown. What is the main finding and its importance? Release of TNFα by tPVAT causes production of reactive oxygen species in tPVAT through activation of an NADPH-oxidase 2 (NOX2)-dependent pathway, activates production of aortic reactive oxygen species and mediates aortic stiffness, potentially through matrix metalloproteinase 9 activity. Neutralization of TNFα and/or inhibition of NOX2 blocks the tPVAT-induced impairment of aortic function. These data partly implicate tPVAT NOX2 and TNFα in mediating the vascular pathology of metabolic syndrome. ABSTRACT Perivascular adipose tissue (PVAT) is recognized for its vasoactive effects, but it is unclear how metabolic syndrome impacts thoracic aorta (t)PVAT and the subsequent effect on functional and structural aortic stiffness. Thoracic aorta and tPVAT were removed from 16- to 17-week-old lean (LZR, n = 16) and obese Zucker rats (OZR, n = 16). The OZR presented with aortic endothelial dysfunction, assessed by wire myography, and increased aortic stiffness, assessed by elastic modulus. The OZR tPVAT exudate further exacerbated the endothelial dysfunction, reducing nitric oxide and endothelium-dependent relaxation (P < 0.05). Additionally, OZR tPVAT exudate had increased MMP9 activity (P < 0.05) and further increased the elastic modulus of the aorta after 72 h of co-culture (P < 0.05). We found that the observed aortic dysfunction caused by OZR tPVAT was mediated through increased production and release of tumour necrosis factor-α (TNFα; P < 0.01), which was dependent on tPVAT NADPH-oxidase 2 (NOX2) activity. The OZR tPVAT release of reactive oxygen species and subsequent aortic dysfunction were inhibited by TNFα neutralization and/or inhibition of NOX2. Additionally, we found that OZR tPVAT had reduced activity of the active sites of the 20S proteasome (P < 0.05) and reduced superoxide dismutase activity (P < 0.01). In conclusion, metabolic syndrome causes tPVAT dysfunction through an interplay between TNFα and NOX2 that leads to tPVAT-mediated aortic stiffness by activation of aortic reactive oxygen species and increased MMP9 activity.
Collapse
Affiliation(s)
- Evan DeVallance
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Kayla W Branyan
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Kent Lemaster
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - I Mark Olfert
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - David M Smith
- Department of Biochemistry, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Emidio E Pistilli
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Jefferson C Frisbee
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.,Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Paul D Chantler
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA
| |
Collapse
|
11
|
A function-blocking CD47 antibody modulates extracellular vesicle-mediated intercellular signaling between breast carcinoma cells and endothelial cells. J Cell Commun Signal 2017; 12:157-170. [PMID: 29188480 DOI: 10.1007/s12079-017-0428-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 10/25/2017] [Indexed: 12/25/2022] Open
Abstract
Tumor cells release extracellular vesicles (EVs) into the tumor microenvironment that may facilitate malignant progression and metastasis. Breast carcinoma EVs express high levels of the thrombospondin-1 and signal regulatory protein-α receptor CD47, which is the target of several experimental therapeutics currently in clinical trials. We analyzed changes in gene expression and function in human umbilical vein endothelial cells (HUVEC) induced by treatment with EVs derived from breast carcinoma cells and the effects of the function-blocking CD47 antibody B6H12 on the resulting intercellular communication. CD47+ EVs exhibited greater uptake by HUVEC compared to CD47- EVs, but the CD47 antibody did not inhibit their uptake. Global and targeted analyses of transcripts demonstrated that treatment of HUVEC with EVs derived from MDA-MB-231 breast carcinomas cells altered pathways associated with tumor necrosis factor-α signaling, angiogenesis, lymphangiogenesis, endothelial-mesenchymal transition, and extracellular matrix. EVs from triple-negative MDA-MB-231 cells were more active than EVs from less metastatic breast carcinoma cell lines. Treatment with MDA-MB-231 EVs down-regulated VEGFR2 mRNA expression and tyrosine phosphorylation while enhancing phosphorylation of the tyrosine phosphatase SHP2. VEGFR2 expression and phosphorylation in HUVEC was further inhibited by the CD47 antibody. Consistent with the observed changes in endothelial-mesenchymal transition genes and SHP2, treatment with MDA-MB-231-derived EVs decreased Zeb1 protein levels in HUVEC, whereas the CD47 antibody increased Zeb1 levels. The induction of E-selectin and other known targets of tumor necrosis factor-α signaling by EVs was also enhanced by the CD47 antibody, and E-selectin was the most up-regulated transcript following CD47 antibody treatment alone. These studies reveal several mechanisms by which therapeutics targeting CD47 could modulate tumor growth by altering the cross talk between cancer-derived EVs and nonmalignant cells in the tumor stroma.
Collapse
|
12
|
Chang PY, Zhang BY, Cui S, Qu C, Shao LH, Xu TK, Qu YQ, Dong LH, Wang J. MSC-derived cytokines repair radiation-induced intra-villi microvascular injury. Oncotarget 2017; 8:87821-87836. [PMID: 29152123 PMCID: PMC5675675 DOI: 10.18632/oncotarget.21236] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 08/26/2017] [Indexed: 02/06/2023] Open
Abstract
Microvascular injury initiates the pathogenesis of radiation enteropathy. As previously demonstrated, the secretome from mesenchymal stem cells contains various angiogenic cytokines that exhibited therapeutic potential for ischemic lesions. As such, the present study aimed to investigate whether cytokines derived from mesenchymal stem cells can repair endothelial injuries from irradiated intestine. Here, serum-free medium was conditioned by human adipose-derived mesenchymal stem cells, and we found that there were several angiogenic cytokines in the medium, including IL-8, angiogenin, HGF and VEGF. This medium promoted the formation of tubules between human umbilical cord vein endothelial cells and protected these cells against radiation-induced apoptosis in vitro. Likewise, our in vivo results revealed that repeated injections of mesenchymal stem cell-conditioned medium could accelerate the recovery of irradiated mice by reducing the serum levels of pro-inflammatory cytokines, including IL-1α, IL-6 and TNF-α, and promoting intra-villi angiogenesis. Herein, intervention by conditioned medium could increase the number of circulating endothelial progenitors, whereas neutralizing SDF-1α and/or inhibiting PI3K would hamper the recruitment of endothelial progenitors to the injured sites. Such results suggested that SDF-1α and PI3K-mediated phosphorylation were required for intra-villi angiogenesis. To illustrate this, we found that conditioned medium enabled endothelial cells to increase intracellular levels of phosphorylated Akt Ser473, both under irradiated and steady state conditions, and to up-regulate the expression of the CXCR4 and CXCR7 genes. Collectively, the present results revealed the therapeutic effects of mesenchymal stem cell-derived cytokines on microvascular injury of irradiated intestine.
Collapse
Affiliation(s)
- Peng-Yu Chang
- State Key Laboratory of Electroanalytical Chemistry, Chinese Academy of Sciences, Changchun Jilin 130022, P.R. China.,Department of Radiation Oncology, First Bethune Hospital of Jilin University, Changchun 130021, P.R. China
| | - Bo-Yin Zhang
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, P.R. China
| | - Shuang Cui
- Department of Radiation Oncology, First Bethune Hospital of Jilin University, Changchun 130021, P.R. China
| | - Chao Qu
- Department of Radiation Oncology, First Bethune Hospital of Jilin University, Changchun 130021, P.R. China
| | - Li-Hong Shao
- Department of Radiation Oncology, First Bethune Hospital of Jilin University, Changchun 130021, P.R. China
| | - Tian-Kai Xu
- Department of Radiation Oncology, First Bethune Hospital of Jilin University, Changchun 130021, P.R. China
| | - Ya-Qin Qu
- Department of Radiation Oncology, First Bethune Hospital of Jilin University, Changchun 130021, P.R. China
| | - Li-Hua Dong
- Department of Radiation Oncology, First Bethune Hospital of Jilin University, Changchun 130021, P.R. China
| | - Jin Wang
- State Key Laboratory of Electroanalytical Chemistry, Chinese Academy of Sciences, Changchun Jilin 130022, P.R. China.,Department of Chemistry and Physics, State University of New York at Stony Brook, Stony Brook, NY 11794-3400, USA
| |
Collapse
|
13
|
Gao F, Sabbineni H, Artham S, Somanath PR. Modulation of long-term endothelial-barrier integrity is conditional to the cross-talk between Akt and Src signaling. J Cell Physiol 2017; 232:2599-2609. [PMID: 28075016 DOI: 10.1002/jcp.25791] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 01/06/2017] [Accepted: 01/10/2017] [Indexed: 12/30/2022]
Abstract
Although numerous studies have implicated Akt and Src kinases in vascular endothelial growth factor (VEGF) and Angiopoietin-1 (Ang-1)-induced endothelial-barrier regulation, a link between these two pathways has never been demonstrated. We determined the long-term effects of Akt inhibition on Src activity and vice versa, and in turn, on the human microvascular endothelial cell (HMEC) barrier integrity at the basal level, and in response to growth factors. Our data showed that Akt1 gene knockdown increases gap formation in HMEC monolayer at the basal level. Pharmacological inhibition of Akt, but not Src resulted in exacerbated VEGF-induced vascular leakage and impaired Ang-1-induced HMEC-barrier protection in vitro at 24 hr. Whereas inhibition of Akt had no effect on VEGF-induced HMEC gap formation in the short term, inhibition of Src blunted this process. In contrast, inhibition of Akt disrupted the VEGF and Ang-1 stabilized barrier integrity in the long-term while inhibition of Src did not. Interestingly, both long-term Akt inhibition and Akt1 gene knockdown in HMECs resulted in increased Tyr416 phosphorylation of Src. Treatment of HMECs with transforming growth factor-β1 (TGFβ1) that inhibited Akt Ser473 phosphorylation in the long-term, activated Src through increased Tyr416 phosphorylation and decreased HMEC-barrier resistance. The effect of TGFβ1 on endothelial-barrier breakdown was blunted in Akt1 deficient HMEC monolayers, where endothelial-barrier resistance was already impaired compared to the control. To our knowledge, this is the first report demonstrating a direct cross-talk between Akt and Src in endothelial-barrier regulation.
Collapse
Affiliation(s)
- Fei Gao
- Clinical and Experimental Therapeutics, University of Georgia and Charlie Norwood VA Medical Center, Augusta, Georgia.,Department of Urology, The First Affiliated Hospital of Chongqing University, Chongqing, China
| | - Harika Sabbineni
- Clinical and Experimental Therapeutics, University of Georgia and Charlie Norwood VA Medical Center, Augusta, Georgia
| | - Sandeep Artham
- Clinical and Experimental Therapeutics, University of Georgia and Charlie Norwood VA Medical Center, Augusta, Georgia
| | - Payaningal R Somanath
- Clinical and Experimental Therapeutics, University of Georgia and Charlie Norwood VA Medical Center, Augusta, Georgia.,Department of Medicine and Vascular Biology Center, Augusta University, Augusta, Georgia
| |
Collapse
|
14
|
Li Y, Turpin CP, Wang S. Role of thrombospondin 1 in liver diseases. Hepatol Res 2017; 47:186-193. [PMID: 27492250 PMCID: PMC5292098 DOI: 10.1111/hepr.12787] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 07/13/2016] [Accepted: 08/01/2016] [Indexed: 02/06/2023]
Abstract
Thrombospondin 1 (TSP1) is a matricellular glycoprotein that can be secreted by many cell types. Through binding to extracellular proteins and/or cell surface receptors, TSP1 modulates a variety of cellular functions. Since its discovery in 1971, TSP1 has been found to play important roles in multiple biological processes including angiogenesis, apoptosis, latent transforming growth factor-β activation, and immune regulation. Thrombospondin 1 is also involved in regulating many organ functions. However, the role of TSP1 in liver diseases has not been extensively addressed. In this review, we summarize the findings about the possible role that TSP1 plays in chronic liver diseases focusing on non-alcoholic fatty liver diseases, liver fibrosis, and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Yanzhang Li
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
- Medical College of Henan University, Kaifeng, Henan 475004, China
| | - Courtney P Turpin
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Shuxia Wang
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
15
|
Yang LT, Li XX, Qiu SQ, Zeng L, Li LJ, Feng BS, Zheng PY, Liu ZG, Yang PC. Micro RNA-19a suppresses thrombospondin-1 in CD35 + B cells in the intestine of mice with food allergy. Am J Transl Res 2016; 8:5503-5511. [PMID: 28078021 PMCID: PMC5209501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 11/29/2016] [Indexed: 06/06/2023]
Abstract
Disruption of immune tolerance is associated in the pathogenesis of allergy. Thrombospondin-1 (TSP1) plays a role in the maintenance of immune tolerance, which is compromised in allergic disorders. Micro RNA (miR) is involved in the regulation of immune responses. This study tests a hypothesis that miR-17-92 cluster is involved in the regulation of TSP1 in the intestinal CD35+ B cells. In this study, a food allergy mouse model was developed. The intestinal B cells were isolated to be analyzed for the expression of a miR-17-92 cluster and TSP1. The role of miR-19a in the suppression of TSP1 in B cells was tested in a cell culture model. We observed that the levels of TSP1 were significantly decreased; the levels of miR-19a were significantly increased in intestinal CD35+ B cells of mice sensitized to ovalbumin (OVA) as compared with naïve controls. Exposure to interleukin (IL)-4 suppressed the expression of TSP1 in B cells, which was abolished by inhibition of miR-19a. miR-19a mediated the effects of IL-4 on repressing TSP1 expression in B cells. We conclude that IL-4 suppresses the expression of TSP1 in the intestinal CD35+ B cells via up regulating miR-19a. The miR-19a may be a target to regulate the immune tolerant status in the body.
Collapse
Affiliation(s)
- Li-Tao Yang
- The Research Center of Allergy & Immunology, Shenzhen University School of MedicineShenzhen 518060, China
- Shenzhen ENT Institute, Longgang ENT HospitalShenzhen 518116, China
- Brain Body Institute, McMaster UniversityHamilton, ON L8N 4A6, Canada
| | - Xiao-Xi Li
- The Research Center of Allergy & Immunology, Shenzhen University School of MedicineShenzhen 518060, China
| | - Shu-Qi Qiu
- The Research Center of Allergy & Immunology, Shenzhen University School of MedicineShenzhen 518060, China
- Shenzhen ENT Institute, Longgang ENT HospitalShenzhen 518116, China
| | - Lu Zeng
- The Research Center of Allergy & Immunology, Shenzhen University School of MedicineShenzhen 518060, China
| | - Lin-Jing Li
- Brain Body Institute, McMaster UniversityHamilton, ON L8N 4A6, Canada
- Department of Gastroenterology, The Second Hospital, Zhengzhou UniversityZhengzhou, China
| | - Bai-Sui Feng
- Department of Gastroenterology, The Second Hospital, Zhengzhou UniversityZhengzhou, China
| | - Peng-Yuan Zheng
- Department of Gastroenterology, The Fifth Hospital, Zhengzhou UniversityZhengzhou, China
| | - Zhi-Gang Liu
- The Research Center of Allergy & Immunology, Shenzhen University School of MedicineShenzhen 518060, China
| | - Ping-Chang Yang
- The Research Center of Allergy & Immunology, Shenzhen University School of MedicineShenzhen 518060, China
| |
Collapse
|
16
|
Risher ML, Sexton HG, Risher WC, Wilson WA, Fleming RL, Madison RD, Moore SD, Eroglu C, Swartzwelder HS. Adolescent Intermittent Alcohol Exposure: Dysregulation of Thrombospondins and Synapse Formation are Associated with Decreased Neuronal Density in the Adult Hippocampus. Alcohol Clin Exp Res 2015; 39:2403-13. [PMID: 26537975 PMCID: PMC4712076 DOI: 10.1111/acer.12913] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 09/22/2015] [Indexed: 01/18/2023]
Abstract
BACKGROUND Adolescent intermittent alcohol exposure (AIE) has profound effects on neuronal function. We have previously shown that AIE causes aberrant hippocampal structure and function that persists into adulthood. However, the possible contributions of astrocytes and their signaling factors remain largely unexplored. We investigated the acute and enduring effects of AIE on astrocytic reactivity and signaling on synaptic expression in the hippocampus, including the impact of the thrombospondin (TSP) family of astrocyte-secreted synaptogenic factors and their neuronal receptor, alpha2delta-1 (α2δ-1). Our hypothesis is that some of the influences of AIE on neuronal function may be secondary to direct effects on astrocytes. METHODS We conducted Western blot analysis on TSPs 1 to 4 and α2δ-1 from whole hippocampal lysates 24 hours after the 4th and 10th doses of AIE, then 24 days after the last dose (in adulthood). We used immunohistochemistry to assess astrocyte reactivity (i.e., morphology) and synaptogenesis (i.e., colocalization of pre- and postsynaptic puncta). RESULTS Adolescent AIE reduced α2δ-1 expression, and colocalized pre- and postsynaptic puncta after the fourth ethanol (EtOH) dose. By the 10th dose, increased TSP2 levels were accompanied by an increase in colocalized pre- and postsynaptic puncta, while α2δ-1 returned to control levels. Twenty-four days after the last EtOH dose (i.e., adulthood), TSP2, TSP4, and α2δ-1 expression were all elevated. Astrocyte reactivity, indicated by increased astrocytic volume and area, was also observed at that time. CONCLUSIONS Repeated EtOH exposure during adolescence results in long-term changes in specific astrocyte signaling proteins and their neuronal synaptogenic receptor. Continued signaling by these traditionally developmental factors in adulthood may represent a compensatory mechanism whereby astrocytes reopen the synaptogenic window and repair lost connectivity, and consequently contribute to the enduring maladaptive structural and functional abnormalities previously observed in the hippocampus after AIE.
Collapse
Affiliation(s)
- Mary-Louise Risher
- Durham VA Medical Center, Duke University Medical Center, Durham, North Carolina
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina
| | - Hannah G Sexton
- Durham VA Medical Center, Duke University Medical Center, Durham, North Carolina
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina
| | - W Christopher Risher
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina
| | - Wilkie A Wilson
- Social Sciences Research Institute, Duke University Medical Center, Durham, North Carolina
| | - Rebekah L Fleming
- Durham VA Medical Center, Duke University Medical Center, Durham, North Carolina
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina
| | - Roger D Madison
- Durham VA Medical Center, Duke University Medical Center, Durham, North Carolina
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Scott D Moore
- Durham VA Medical Center, Duke University Medical Center, Durham, North Carolina
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina
| | - Cagla Eroglu
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina
| | - H Scott Swartzwelder
- Durham VA Medical Center, Duke University Medical Center, Durham, North Carolina
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina
- Department of Psychology and Neuroscience, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|