1
|
Ma B, Gu C, Lu R, Lian P, Wang W, Huang Z, Su Z, Wang H. Inhibition of KPNA2 by ivermectin reduces E2F1 nuclear translocation to attenuate keratinocyte proliferation and ameliorate psoriasis-like lesions. Int Immunopharmacol 2024; 143:113360. [PMID: 39388894 DOI: 10.1016/j.intimp.2024.113360] [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: 08/28/2024] [Revised: 09/25/2024] [Accepted: 10/05/2024] [Indexed: 10/12/2024]
Abstract
Psoriasis is a chronic, immune-mediated skin disease with a significant global prevalence. Karyopherin subunit alpha 2 (KPNA2), a nuclear transport protein involved in cellular activities such as differentiation, proliferation, apoptosis, and immune response, has emerged as a potential biomarker in several diseases. Our study found that KPNA2 was significantly upregulated in psoriasis patients and in imiquimod (IMQ)-induced psoriasis mouse models by bioinformatics and molecular biotechnology. In vivo, treatment with ivermectin, a KPNA2 inhibitor, significantly improved psoriasis symptoms in mice as evidenced by reduced erythema, desquamation, and skin thickness. Histopathological staining revealed decreased expression of KPNA2, K17, and Ki67 in ivermectin-treated mice, suggesting reduced abnormal differentiation and proliferation of keratinocytes. Transcriptome data and immunoblotting analysis showed that KPNA2 inhibition reduced inflammation and keratinocyte proliferation and differentiation in IMQ-induced mice. In vitro, EdU (5-ethynyl-2'-deoxyuridine) and flow cytometry experiments demonstrated that the downregulation of KPNA2 expression in HaCaT cells was capable of inhibiting the EGF (Epidermal Growth Factor)-induced activation of AKT/STAT3 signaling and keratinocytes proliferation. In addition, nuclear-cytoplasmic protein separation and immunofluorescence localization experiments showed that KPNA2 inhibition affected the nuclear translocation of E2F transcription factor 1 (E2F1), a process critical for keratinocyte proliferation. This study elucidated the role of KPNA2 in the pathogenesis of psoriasis and highlighted its potential as a target for future psoriasis therapies. These findings provide new insights into targeted therapy for psoriasis and have significant implications for future clinical treatment.
Collapse
Affiliation(s)
- Bojie Ma
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, PR China
| | - Chaode Gu
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, PR China
| | - Renwei Lu
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, PR China
| | - Panpan Lian
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, PR China
| | - Wentong Wang
- Department of Dermatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Zhiqiang Huang
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, PR China.
| | - Zhonglan Su
- Department of Dermatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China.
| | - Hongwei Wang
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, PR China.
| |
Collapse
|
2
|
Wei J, Gao C, Lu C, Wang L, Dong D, Sun M. The E2F family: a ray of dawn in cardiomyopathy. Mol Cell Biochem 2024:10.1007/s11010-024-05063-4. [PMID: 38985251 DOI: 10.1007/s11010-024-05063-4] [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: 03/21/2024] [Accepted: 06/29/2024] [Indexed: 07/11/2024]
Abstract
Cardiomyopathies are a group of heterogeneous diseases, characterized by abnormal structure and function of the myocardium. For many years, it has been a hot topic because of its high morbidity and mortality as well as its complicated pathogenesis. The E2Fs, a group of transcription factors found extensively in eukaryotes, play a crucial role in governing cell proliferation, differentiation, and apoptosis, meanwhile their deregulated activity can also cause a variety of diseases. Based on accumulating evidence, E2Fs play important roles in cardiomyopathies. In this review, we describe the structural and functional characteristics of the E2F family and its role in cardiomyocyte processes, with a focus on how E2Fs are associated with the onset and development of cardiomyopathies. Moreover, we discuss the great potential of E2Fs as biomarkers and therapeutic targets, aiming to provide a reference for future research.
Collapse
Affiliation(s)
- Jinwen Wei
- College of Exercise and Health, Shenyang Sport University, No.36 Jinqiansong East Road, Shenyang, 110102, Liaoning, People's Republic of China
| | - Can Gao
- College of Exercise and Health, Shenyang Sport University, No.36 Jinqiansong East Road, Shenyang, 110102, Liaoning, People's Republic of China
| | - Changxu Lu
- College of Exercise and Health, Shenyang Sport University, No.36 Jinqiansong East Road, Shenyang, 110102, Liaoning, People's Republic of China
| | - Lijie Wang
- Department of Cardiology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110033, Liaoning, People's Republic of China
| | - Dan Dong
- College of Basic Medical Science, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, People's Republic of China
| | - Mingli Sun
- College of Exercise and Health, Shenyang Sport University, No.36 Jinqiansong East Road, Shenyang, 110102, Liaoning, People's Republic of China.
| |
Collapse
|
3
|
Premnath P, Lun T, Siddiqui H, Stahl AR, Ardebili AA, Olsen A, Krawetz R. Absence of E2f1 Negates Pro-osteogenic Impacts of p21 Absence. Calcif Tissue Int 2024; 114:625-637. [PMID: 38643416 DOI: 10.1007/s00223-024-01210-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 03/03/2024] [Indexed: 04/22/2024]
Abstract
Loss of p21 leads to increased bone formation post-injury; however, the mechanism(s) by which this occurs remains undetermined. E2f1 is downstream of p21 and as a transcription factor can act directly on gene expression; yet it is unknown if E2f1 plays a role in the osteogenic effects observed when p21 is differentially regulated. In this study we aimed to investigate the interplay between p21 and E2f1 and determine if the pro-regenerative osteogenic effects observed with the loss of p21 are E2f1 dependent. To accomplish this, we employed knockout p21 and E2f1 mice and additionally generated a p21/E2f1 double knockout. These mice underwent burr-hole injuries to their proximal tibiae and healing was assessed over 7 days via microCT imaging. We found that p21 and E2f1 play distinct roles in bone regeneration where the loss of p21 increased trabecular bone formation and loss of E2f1 increased cortical bone formation, yet loss of E2f1 led to poorer bone repair overall. Furthermore, when E2f1 was absent, either individually or simultaneously with p21, there was a dramatic decrease of the number of osteoblasts, osteoclasts, and chondrocytes at the site of injury compared to p21-/- and C57BL/6 mice. Together, these results suggest that E2f1 regulates the cell populations required for bone repair and has a distinct role in bone formation/repair compared to p21-/-E2f1-/-. These results highlight the possibility of cell cycle and/or p21/E2f1 being potential druggable targets that could be leveraged in clinical therapies to improve bone healing in pathologies such as osteoporosis.
Collapse
Affiliation(s)
- Priyatha Premnath
- Department of Biomedical Engineering, University of Wisconsin-Milwaukee, Milwaukee, WI, USA.
| | - Theodore Lun
- Cumming School of Medicine, McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Humza Siddiqui
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Alana Ruth Stahl
- Cumming School of Medicine, McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Aria Ahadzadeh Ardebili
- Cumming School of Medicine, McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Department of Biomedical Engineering, University of Calgary, Calgary, AB, Canada
| | - Alexandra Olsen
- Cumming School of Medicine, McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Department of Biomedical Engineering, University of Calgary, Calgary, AB, Canada
| | - Roman Krawetz
- Cumming School of Medicine, McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
4
|
Wang H, Dou L. Single-cell RNA sequencing reveals hub genes of myocardial infarction-associated endothelial cells. BMC Cardiovasc Disord 2024; 24:70. [PMID: 38267885 PMCID: PMC10809747 DOI: 10.1186/s12872-024-03727-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 01/14/2024] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND Myocardial infarction (MI) is a cardiovascular disease that seriously threatens human health. Dysangiogenesis of endothelial cells (ECs) primarily inhibits recovery from MI, but the specific mechanism remains to be further elucidated. METHODS In this study, the single-cell RNA-sequencing data from both MI and Sham mice were analyzed by the Seurat Package (3.2.2). The number of ECs in MI and Sham groups were compared by PCA and tSNE algorithm. FindMarkers function of Seurat was used to analyze the DEGs between the MI and Sham groups. Then, the ECs was further clustered into 8 sub-clusters for trajectory analysis. The BEAM was used to analyze the branch point 3 and cluster the results. In addition, the DEGs in the microarray data set of MI and Sham mice were cross-linked, and the cross-linked genes were used to construct PPI networks. The key genes with the highest degree were identified and analyzed for functional enrichment. Finally, this study cultured human umbilical vein endothelial cells (HUVECs), established hypoxia models, and interfered with hub gene expression in cells. The impact of hub genes on the migration and tube formation of hypoxic-induced HUVECs were verified by Wound healing assays and tubule formation experiments. RESULTS The number and proportion of ECs in the MI group were significantly lower than those in the Sham group. Meantime, 225 DEGs were found in ECs between the MI and Sham groups. Through trajectory analysis, EC4 was found to play an important role in MI. Then, by using BEAM to analyze the branch point 3, and clustering the results, a total of 495 genes were found to be highly expressed in cell Fate2 (mainly EC4). In addition, a total of 194 DEGs were identified in Micro array dataset containing both MI and Sham mice. The hub genes (Timp1 and Fn1) with the highest degree were identified. Inhibiting Timp1 and Fn1 expression promoted the migration and tube formation of HUVECs. CONCLUSIONS Our data highlighted the non-linear dynamics of ECs in MI, and provided a foothold for analyzing cardiac homeostasis and pro-angiogenesis in MI.
Collapse
Affiliation(s)
- Hao Wang
- Department of Cardiovascular Medicine, Zhejiang Greentown Cardiovascular Hospital, No.409 Gudun Road, Hangzhou, 310000, Zhejiang, China
| | - Liping Dou
- Department of Geriatrics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, No. 318 Chaowang Road, Hangzhou, 310005, Zhejiang, China.
| |
Collapse
|
5
|
Yu H, Wu Y, Zhang B, Xiong M, Yi Y, Zhang Q, Wu M. Exosomes Derived from E2F1 -/- Adipose-Derived Stem Cells Promote Skin Wound Healing via miR-130b-5p/TGFBR3 Axis. Int J Nanomedicine 2023; 18:6275-6292. [PMID: 37941530 PMCID: PMC10629453 DOI: 10.2147/ijn.s431725] [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: 07/21/2023] [Accepted: 10/21/2023] [Indexed: 11/10/2023] Open
Abstract
Background Skin wound is a widespread health problem and brings extraordinary burdens to patients. Exosomes derived from adipose-derived stem cells (ADSC-Exos) are considered promising strategies for repairing skin wounds. E2F1 is a member of the E2F family of transcription factors involved in cell growth and apoptosis. E2F1 deficiency in mice enhances wound healing by improving collagen deposition and angiogenesis. Additionally, E2F1 can regulate the transcription and paracrine activity of multiple miRNAs, which will inevitably reshape the paracrine expression profile of ADSC-Exos. This study aimed to investigate the impact of transcription factor E2F1 deficiency on the functions of ADSC-Exos in promoting wound healing. Methods First, we obtained ADSCs from subcutaneous adipose tissues of WT and E2F1-/- C57BL/6 mice and separated their exosomes, denoted as ADSCWT-Exos and ADSCE2F1-/--Exos. The wound healing effects of ADSCWT-Exos and ADSCE2F1-/--Exos in full-thickness skin wound models were investigated by wound images, H&E staining, and immunohistochemical staining. For the in vitro study, the abilities of ADSCWT-Exos and ADSCE2F1-/--Exos to promote cell activities, collagen formation, and angiogenesis were evaluated. The potential mechanism by which ADSCE2F1-/--Exos promote wound healing was determined by miRNA sequencing, ChIP‒qPCR, and dual-luciferase assays. Results ADSCE2F1-/--Exos accelerated wound healing by promoting collagen formation and angiogenesis. As a result, compared with the lower wound healing rate of 30.5% within 7 days in the control group and 42.3% in the ADSCWT-Exo group, ADSCE2F1-/--Exos significantly increased the wound healing rate to 72.5%. In vitro, ADSCE2F1-/--Exos activated the function of fibroblasts and vascular endothelial cells. The loss of E2F1 promoted miR-130b-5p expression in ADSCE2F1-/--Exos through transcriptional regulation. MiRNA high-throughput sequencing identified 12 differently expressed miRNAs between ADSCE2F1-/- and ADSCWT. ADSCE2F1-/--Exos enhanced fibroblast activities via the miR-130b-5p/TGFBR3 axis and TGF-β activation. Conclusion Our results indicated that ADSCE2F1-/--Exos effectively promoted wound healing by regulating the miR-130b-5p/TGFBR3 axis, thus providing a novel strategy of gene-engineered stem cell exosomes for accelerating wound healing.
Collapse
Affiliation(s)
- Honghao Yu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Yiping Wu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Boyu Zhang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Mingchen Xiong
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Yi Yi
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Qi Zhang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Min Wu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| |
Collapse
|
6
|
Cheng M, Chen C, Yu K, Lv X, Zeng Q, Dong N, Zhu F. Ablation of CXCR4 expression in cardiomyocytes exacerbates isoproterenol‑induced cell death and heart failure. Int J Mol Med 2022; 51:13. [PMID: 36579657 PMCID: PMC9869727 DOI: 10.3892/ijmm.2022.5216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 11/21/2022] [Indexed: 12/30/2022] Open
Abstract
CXCR4 is a seven‑transmembrane‑spanning Gi‑coupled receptor for the SDF‑1 chemokine and plays a critical role in cardiovascular development and post‑injury repair. However, the specific role of CXCR4 in cardiomyocytes is incompletely understood. It was hypothesized that CXCR4 activation in cardiomyocytes antagonizes β‑adrenoceptor/Gs signaling‑induced cardiac dysfunction. Cardiomyocyte‑specific CXCR4 knockout (CXCR4‑CMKO) mice were generated by crossing CXCR4fl/fl and MHC‑Cre+/‑ mice. Their cardiac structure and function in the basal state are equivalent to that of the control MHC‑Cre+/‑ littermates until at least 4 months old. However, following continuous subcutaneous administration of isoproterenol (Iso) via an osmotic mini‑pump, the ventricular myocardial contractility, dilation, cardiomyocyte apoptosis, and interstitial fibrosis are worse in CXCR4‑CMKO mice than in MHC‑Cre+/‑ littermates. In the cultured H9C2 cardiomyocytes, SDF‑1 treatment markedly attenuated Iso‑induced apoptosis and reduction in phospho‑Akt, and this protective effect was lost by knockdown of CXCR4 or by co‑treatment with Gi inhibitors. In conclusion, CXCR4 promotes cardiomyocyte survival and heart function during β‑adrenergic stress.
Collapse
Affiliation(s)
- Min Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China,Correspondence to: Dr Min Cheng or Dr Feng Zhu, Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1377 Jiefang Avenue, Wuhan, Hubei 430022, P.R. China, E-mail: , E-mail:
| | - Can Chen
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Kunwu Yu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Xiao Lv
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Qiutang Zeng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Nianguo Dong
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Feng Zhu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China,Correspondence to: Dr Min Cheng or Dr Feng Zhu, Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1377 Jiefang Avenue, Wuhan, Hubei 430022, P.R. China, E-mail: , E-mail:
| |
Collapse
|
7
|
Zhang L, Chen L, You X, Li M, Shi H, Sun W, Leng Y, Xue Y, Wang H. Naoxintong capsule limits myocardial infarct expansion by inhibiting platelet activation through the ERK5 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 98:153953. [PMID: 35092875 DOI: 10.1016/j.phymed.2022.153953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/08/2022] [Accepted: 01/18/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND In the clinic, Naoxintong capsule (NXT) has been applied in two level prevention of ischemic disease. However, its mechanism of action requires further study. PURPOSE This study investigated whether NXT could affect platelet function and activation under ischemic pathological conditions. MATERIALS AND METHODS Wistar rats were divided into six groups, sham, saline, NXT (250, 500, 1000 mg/kg), and aspirin group (10 mg/kg). For the pre-treatment assays, MI model was established after pre-administration of saline, NXT-L, NXT-M, NXT-H, and aspirin respectively for 14 days, and after surgery, there were no continuous treatments. For the post-treatment assay, rats were orally administered for 3 days after MI. FeCl3-induced thrombosis model was applied to determine the thrombus wet weight. Bleeding time was used to assess the ability of the platelets to develop a hemostatic plug. RESULTS NXT decreased infarct size, decreased LDH, CK, and CK-MB values, and improved cardiac function. NXT inhibited platelets activation through reducing CD62P-positive platelets and inhibited infarct expansion by decreasing the number of CD45-positive cells and the amount of MMP9 secreted into the heart tissue. Mechanistically, NXT inhibited platelets activation through decreasing ROS levels, decreasing ERK5 phosphorylation, and increasing RAC1 phosphorylation in MI rats. Pre-treatment with NXT decreased thrombus formation and had normal bleeding times. CONCLUSION NXT showed obviously preventive effects, which was associated with negative control of platelet activation. The above results provide a basis for clinically expanding application of NXT.
Collapse
Affiliation(s)
- Lusha Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Lu Chen
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin, 301617, China; Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xingyu You
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin, 301617, China
| | - Mengyao Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Hong Shi
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin, 301617, China; Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Wei Sun
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yuze Leng
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yuejin Xue
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Hong Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin, 301617, China; Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, 301617, China; School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
8
|
The Role of the VEGF Family in Atherosclerosis Development and Its Potential as Treatment Targets. Int J Mol Sci 2022; 23:ijms23020931. [PMID: 35055117 PMCID: PMC8781560 DOI: 10.3390/ijms23020931] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/09/2022] [Accepted: 01/14/2022] [Indexed: 02/07/2023] Open
Abstract
The vascular endothelial growth factor (VEGF) family, the crucial regulator of angiogenesis, lymphangiogenesis, lipid metabolism and inflammation, is involved in the development of atherosclerosis and further CVDs (cardiovascular diseases). This review discusses the general regulation and functions of VEGFs, their role in lipid metabolism and atherosclerosis development and progression. These functions present the great potential of applying the VEGF family as a target in the treatment of atherosclerosis and related CVDs. In addition, we discuss several modern anti-atherosclerosis VEGFs-targeted experimental procedures, drugs and natural compounds, which could significantly improve the efficiency of atherosclerosis and related CVDs' treatment.
Collapse
|
9
|
Xiong M, Hu W, Tan Y, Yu H, Zhang Q, Zhao C, Yi Y, Wang Y, Wu Y, Wu M. Transcription Factor E2F1 Knockout Promotes Mice White Adipose Tissue Browning Through Autophagy Inhibition. Front Physiol 2021; 12:748040. [PMID: 34819874 PMCID: PMC8606532 DOI: 10.3389/fphys.2021.748040] [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: 08/10/2021] [Accepted: 10/20/2021] [Indexed: 12/03/2022] Open
Abstract
Obesity is associated with energy metabolic disturbance and is caused by long-term excessive energy storage in white adipose tissue (WAT). The WAT browning potentially reduces excessive energy accumulation, contributing an attractive target to combat obesity. As a pivotal regulator of cell growth, the transcription factor E2F1 activity dysregulation leads to metabolic complications. The regulatory effect and underlying mechanism of E2F1 knockout on WAT browning, have not been fully elucidated. To address this issue, in this study, the in vivo adipose morphology, mitochondria quantities, uncoupling protein 1 (UCP-1), autophagy-related genes in WAT of wild-type (WT) and E2F1–/– mice were detected. Furthermore, we evaluated the UCP-1, and autophagy-related gene expression in WT and E2F1–/– adipocyte in vitro. The results demonstrated that E2F1 knockout could increase mitochondria and UCP-1 expression in WAT through autophagy suppression in mice, thus promoting WAT browning. Besides, adipocytes lacking E2F1 showed upregulated UCP-1 and downregulated autophagy-related genes expression in vitro. These results verified that E2F1 knockout exerted effects on inducing mice WAT browning through autophagy inhibition in vivo and in vitro. These findings regarding the molecular mechanism of E2F1-modulated autophagy in controlling WAT plasticity, provide a novel insight into the functional network with the potential therapeutic application against obesity.
Collapse
Affiliation(s)
- Mingchen Xiong
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weijie Hu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yufang Tan
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Honghao Yu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Zhang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chongru Zhao
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Yi
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yichen Wang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiping Wu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Wu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
10
|
Zhou Y, Zhu X, Cui H, Shi J, Yuan G, Shi S, Hu Y. The Role of the VEGF Family in Coronary Heart Disease. Front Cardiovasc Med 2021; 8:738325. [PMID: 34504884 PMCID: PMC8421775 DOI: 10.3389/fcvm.2021.738325] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 07/27/2021] [Indexed: 01/04/2023] Open
Abstract
The vascular endothelial growth factor (VEGF) family, the regulator of blood and lymphatic vessels, is mostly investigated in the tumor and ophthalmic field. However, the functions it enjoys can also interfere with the development of atherosclerosis (AS) and further diseases like coronary heart disease (CHD). The source, regulating mechanisms including upregulation and downregulation, target cells/tissues, and known functions about VEGF-A, VEGF-B, VEGF-C, and VEGF-D are covered in the review. VEGF-A can regulate angiogenesis, vascular permeability, and inflammation by binding with VEGFR-1 and VEGFR-2. VEGF-B can regulate angiogenesis, redox, and apoptosis by binding with VEGFR-1. VEGF-C can regulate inflammation, lymphangiogenesis, angiogenesis, apoptosis, and fibrogenesis by binding with VEGFR-2 and VEGFR-3. VEGF-D can regulate lymphangiogenesis, angiogenesis, fibrogenesis, and apoptosis by binding with VEGFR-2 and VEGFR-3. These functions present great potential of applying the VEGF family for treating CHD. For instance, angiogenesis can compensate for hypoxia and ischemia by growing novel blood vessels. Lymphangiogenesis can degrade inflammation by providing exits for accumulated inflammatory cytokines. Anti-apoptosis can protect myocardium from impairment after myocardial infarction (MI). Fibrogenesis can promote myocardial fibrosis after MI to benefit cardiac recovery. In addition, all these factors have been confirmed to keep a link with lipid metabolism, the research about which is still in the early stage and exact mechanisms are relatively obscure. Because few reviews have been published about the summarized role of the VEGF family for treating CHD, the aim of this review article is to present an overview of the available evidence supporting it and give hints for further research.
Collapse
Affiliation(s)
- Yan Zhou
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China
| | - Xueping Zhu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hanming Cui
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jingjing Shi
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guozhen Yuan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shuai Shi
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuanhui Hu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
11
|
Yang L, Deng J, Ma W, Qiao A, Xu S, Yu Y, Boriboun C, Kang X, Han D, Ernst P, Zhou L, Shi J, Zhang E, Li TS, Qiu H, Nakagawa S, Blackshaw S, Zhang J, Qin G. Ablation of lncRNA Miat attenuates pathological hypertrophy and heart failure. Am J Cancer Res 2021; 11:7995-8007. [PMID: 34335976 PMCID: PMC8315059 DOI: 10.7150/thno.50990] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 04/29/2021] [Indexed: 12/11/2022] Open
Abstract
Rationale: The conserved long non-coding RNA (lncRNA) myocardial infarction associate transcript (Miat) was identified for its multiple single-nucleotide polymorphisms that are strongly associated with susceptibility to MI, but its role in cardiovascular biology remains elusive. Here we investigated whether Miat regulates cardiac response to pathological hypertrophic stimuli. Methods: Both an angiotensin II (Ang II) infusion model and a transverse aortic constriction (TAC) model were used in adult WT and Miat-null knockout (Miat-KO) mice to induce pathological cardiac hypertrophy. Heart structure and function were evaluated by echocardiography and histological assessments. Gene expression in the heart was evaluated by RNA sequencing (RNA-seq), quantitative real-time RT-PCR (qRT-PCR), and Western blotting. Primary WT and Miat-KO mouse cardiomyocytes were isolated and used in Ca2+ transient and contractility measurements. Results: Continuous Ang II infusion for 4 weeks induced concentric hypertrophy in WT mice, but to a lesser extent in Miat-KO mice. Surgical TAC for 6 weeks resulted in decreased systolic function and heart failure in WT mice but not in Miat-KO mice. In both models, Miat-KO mice displayed reduced heart-weight to tibia-length ratio, cardiomyocyte cross-sectional area, cardiomyocyte apoptosis, and cardiac interstitial fibrosis and a better-preserved capillary density, as compared to WT mice. In addition, Ang II treatment led to significantly reduced mRNA and protein expression of the Ca2+ cycling genes Sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2a (SERCA2a) and ryanodine receptor 2 (RyR2) and a dramatic increase in global RNA splicing events in the left ventricle (LV) of WT mice, and these changes were largely blunted in Miat-KO mice. Consistently, cardiomyocytes isolated from Miat-KO mice demonstrated more efficient Ca2+ cycling and greater contractility. Conclusions: Ablation of Miat attenuates pathological hypertrophy and heart failure, in part, by enhancing cardiomyocyte contractility.
Collapse
|
12
|
Wu X, Reboll MR, Korf-Klingebiel M, Wollert KC. Angiogenesis after acute myocardial infarction. Cardiovasc Res 2020; 117:1257-1273. [PMID: 33063086 DOI: 10.1093/cvr/cvaa287] [Citation(s) in RCA: 163] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/09/2020] [Accepted: 09/30/2020] [Indexed: 12/16/2022] Open
Abstract
Acute myocardial infarction (MI) inflicts massive injury to the coronary microcirculation leading to vascular disintegration and capillary rarefication in the infarct region. Tissue repair after MI involves a robust angiogenic response that commences in the infarct border zone and extends into the necrotic infarct core. Technological advances in several areas have provided novel mechanistic understanding of postinfarction angiogenesis and how it may be targeted to improve heart function after MI. Cell lineage tracing studies indicate that new capillary structures arise by sprouting angiogenesis from pre-existing endothelial cells (ECs) in the infarct border zone with no meaningful contribution from non-EC sources. Single-cell RNA sequencing shows that ECs in infarcted hearts may be grouped into clusters with distinct gene expression signatures, likely reflecting functionally distinct cell populations. EC-specific multicolour lineage tracing reveals that EC subsets clonally expand after MI. Expanding EC clones may arise from tissue-resident ECs with stem cell characteristics that have been identified in multiple organs including the heart. Tissue repair after MI involves interactions among multiple cell types which occur, to a large extent, through secreted proteins and their cognate receptors. While we are only beginning to understand the full complexity of this intercellular communication, macrophage and fibroblast populations have emerged as major drivers of the angiogenic response after MI. Animal data support the view that the endogenous angiogenic response after MI can be boosted to reduce scarring and adverse left ventricular remodelling. The improved mechanistic understanding of infarct angiogenesis therefore creates multiple therapeutic opportunities. During preclinical development, all proangiogenic strategies should be tested in animal models that replicate both cardiovascular risk factor(s) and the pharmacotherapy typically prescribed to patients with acute MI. Considering that the majority of patients nowadays do well after MI, clinical translation will require careful selection of patients in need of proangiogenic therapies.
Collapse
Affiliation(s)
- Xuekun Wu
- Division of Molecular and Translational Cardiology, Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| | - Marc R Reboll
- Division of Molecular and Translational Cardiology, Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| | - Mortimer Korf-Klingebiel
- Division of Molecular and Translational Cardiology, Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| | - Kai C Wollert
- Division of Molecular and Translational Cardiology, Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| |
Collapse
|
13
|
Yang C, Wu Y, Yi Z, Zeng N, Ren Y, Xu Y, Zeng H, Deng P, Zhang Q, Wu M. Knockout of E2F1 Inhibits Adipose Stem Cell Proliferation and Differentiation in Fat Transplantation by Repressing Peroxisome Proliferator-Activated Receptor Gamma Expression. Transplant Proc 2020; 53:466-473. [PMID: 32482448 DOI: 10.1016/j.transproceed.2020.02.151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 02/07/2020] [Accepted: 02/15/2020] [Indexed: 10/24/2022]
Abstract
Adipose-derived stem cells (ADSCs) possess pluripotent differentiation potential and self-replication ability, which is highly significant in the field of tissue engineering. Cell-assisted lipotransfer (CAL) with ADSCs benefits fat survival. In this study, we focus on the effect of transcription factor E2F1 during CAL. The wild-type (WT) ADSCs were mixed with WT adipocytes, and the E2F1-/- ADSCs were mixed with E2F1-/- adipocytes. Then 2 cell mixtures were inoculated on the back 2 sides of E2F1-/- mice, respectively denoted as the WT group (WT ADSCs + WT adipose cells) and E2F1-/- group (E2F1-/- ADSCs + E2F1-/- adipose cells). At week 4, the fat graft was heavier in the WT group, with less necrotic area, more survival of mature adipocytes, and more proliferating ADSCs, compared with the E2F1-/- group. More capillaries were transformed from ADSCs in the WT group than in the E2F1-/- group. The in vitro protein levels of peroxisome proliferator-activated receptor gamma (PPAR-γ) were higher in WT ADSCs than in E2F1-/- ADSCs. Therefore, these findings suggest that knockout of E2F1 could affect ADSCs to inhibit the survival of fat grafts by downregulating PPAR-γ expression.
Collapse
Affiliation(s)
- Changchun Yang
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiping Wu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhen Yi
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ning Zeng
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuping Ren
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Xu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Zeng
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pei Deng
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Zhang
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Wu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
14
|
Xiao H, Wu YP, Yang CC, Yi Z, Zeng N, Xu Y, Zeng H, Deng P, Zhang Q, Wu M. Knockout of E2F1 enhances the polarization of M2 phenotype macrophages to accelerate the wound healing process. Kaohsiung J Med Sci 2020; 36:692-698. [PMID: 32349192 DOI: 10.1002/kjm2.12222] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/17/2020] [Accepted: 04/08/2020] [Indexed: 01/03/2023] Open
Abstract
Wound healing is a complex process, which is classically divided into inflammation, proliferation, and remodeling phases. Macrophages play a key role in wound healing, however, whether E2F1 mediates the M1/M2 polarization during the wound healing process is not known. Skin wounds were surgically induced in E2F1-/- mice and their WT littermates. At day 2 and day 7 post-surgery, the wounded skin tissues including 2 to 3 mm normal skin were obtained. The wounded skin tissues were used for the analyses of immunofluorescence staining (CD68, iNOS, CD206), western blotting (CD68, iNOS, CD206, PPAR-γ) and Co-immunoprecipitation (E2F1-PPAR-γ interactions). E2F1-/- mice exhibited faster wound healing process. At day 2, the M2 macrophages were remarkably increased in the E2F1-/- mice. Surprisingly, in the border zone of the wound, E2F1-/- mice had also more M2 macrophages and fewer M1 macrophages at day 7 post-surgery, suggesting a certain degree of polarization amongst the M1 and M2 phenotypes. Co-IP revealed that E2F1 indeed interacted with PPAR-γ, meanwhile western blotting and RT-PCR showed higher expression of PPAR-γ in the E2F1-/- mice as compared to that in the WT mice. Therefore, the findings suggest that wound healing process could be accelerated with enhanced M2 polarization through increased PPAR-γ expression in E2F1 knockout mice.
Collapse
Affiliation(s)
- Hui Xiao
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi-Ping Wu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chang-Chun Yang
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhen Yi
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ning Zeng
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Xu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Zeng
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pei Deng
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Zhang
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Wu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
15
|
Resveratrol Attenuates High Glucose-Induced Vascular Endothelial Cell Injury by Activating the E2F3 Pathway. BIOMED RESEARCH INTERNATIONAL 2020; 2020:6173618. [PMID: 32420356 PMCID: PMC7204347 DOI: 10.1155/2020/6173618] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 03/21/2020] [Accepted: 04/10/2020] [Indexed: 11/22/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is the most common metabolic disease. High glucose-induced macrovascular disease and microangiopathy are major complications of diabetes. E2F3, a member of the E2F transcription factor family, is closely related to cardiovascular diseases. Resveratrol, a nonflavonoid polyphenolic compound widely found in plants, has been shown to have cardiovascular protection. However, there are few studies on whether resveratrol can effectively treat diabetic angiopathy, and the specific mechanism involved needs further study. This study investigated whether E2F3 transcription factors are involved in the process of vascular endothelial injury induced by high glucose and observed its effects on the proliferation of vascular endothelial cells. Then, it analyzed whether resveratrol can inhibit high glucose-induced vascular endothelial cell injury by regulating the E2F3 pathway. We demonstrated that the expression level of the E2F3 transcription factor was significantly inhibited in high glucose state. Resveratrol inhibited high glucose-induced vascular endothelial cell injury by upregulating the E2F3 pathway. High glucose can induce vascular endothelial injury by inhibiting E2F3 gene expression, while resveratrol can inhibit high glucose-induced vascular endothelial injury by activating the E2F3 pathway.
Collapse
|
16
|
SLMP53-1 Inhibits Tumor Cell Growth through Regulation of Glucose Metabolism and Angiogenesis in a P53-Dependent Manner. Int J Mol Sci 2020; 21:ijms21020596. [PMID: 31963392 PMCID: PMC7013701 DOI: 10.3390/ijms21020596] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/13/2020] [Accepted: 01/15/2020] [Indexed: 01/10/2023] Open
Abstract
The Warburg effect is an emerging hallmark of cancer, which has the tumor suppressor p53 as its major regulator. Herein, we unveiled that p53 activation by (S)-tryptophanol-derived oxazoloisoindolinone (SLMP53-1) mediated the reprograming of glucose metabolism in cancer cells and xenograft human tumor tissue, interfering with angiogenesis and migration. Particularly, we showed that SLMP53-1 regulated glycolysis by downregulating glucose transporter 1 (GLUT1), hexokinase-2 (HK2), and phosphofructokinase-2 isoform 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase-3 (PFKFB3) (key glycolytic enzymes), while upregulating the mitochondrial markers synthesis of cytochrome c oxidase 2 (SCO2), cytochrome c oxidase subunit 4 (COX4), and OXPHOS mitochondrial complexes. SLMP53-1 also downregulated the monocarboxylate transporter 4 (MCT4), causing the subsequent reduction of lactate export by cancer cells. Besides the acidification of the extracellular environment, SLMP53-1 further increased E-cadherin and reduced metalloproteinase-9 (MMP-9) expression levels in both cancer cells and xenograft human tumor tissue, which suggested the interference of SLMP53-1 in extracellular matrix remodeling and epithelial-to-mesenchymal transition. Consistently, SLMP53-1 depleted angiogenesis, decreasing endothelial cell tube formation and vascular endothelial growth factor (VEGF) expression levels. SLMP53-1 also exhibited synergistic growth inhibitory activity in combination with the metabolic modulator dichloroacetic acid. These data reinforce the promising application of the p53-activating agent SLMP53-1 in cancer therapy, by targeting p53-mediated pathways of growth and dissemination.
Collapse
|
17
|
Lam B, Roudier E. Considering the Role of Murine Double Minute 2 in the Cardiovascular System? Front Cell Dev Biol 2020; 7:320. [PMID: 31921839 PMCID: PMC6916148 DOI: 10.3389/fcell.2019.00320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 11/21/2019] [Indexed: 01/26/2023] Open
Abstract
The E3 ubiquitin ligase Murine double minute 2 (MDM2) is the main negative regulator of the tumor protein p53 (TP53). Extensive studies over more than two decades have confirmed MDM2 oncogenic role through mechanisms both TP53-dependent and TP53-independent oncogenic function. These studies have contributed to designate MDM2 as a therapeutic target of choice for cancer treatment and the number of patents for MDM2 antagonists has increased immensely over the last years. However, the question of the physiological functions of MDM2 has not been fully resolved yet, particularly when expressed and regulated physiologically in healthy tissue. Cardiovascular complications are almost an inescapable side-effect of anti-cancer therapies. While several MDM2 antagonists are entering phase I, II and even III of clinical trials, this review proposes to bring awareness on the physiological role of MDM2 in the cardiovascular system.
Collapse
Affiliation(s)
- Brian Lam
- Angiogenesis Research Group, School of Kinesiology and Health Sciences, Muscle Health Research Center, Faculty of Health, York University, Toronto, ON, Canada
| | - Emilie Roudier
- Angiogenesis Research Group, School of Kinesiology and Health Sciences, Muscle Health Research Center, Faculty of Health, York University, Toronto, ON, Canada
| |
Collapse
|
18
|
Morrell BC, Zhang L, Schütz LF, Perego MC, Maylem ERS, Spicer LJ. Regulation of the transcription factor E2F8 gene expression in bovine ovarian cells. Mol Cell Endocrinol 2019; 498:110572. [PMID: 31493442 DOI: 10.1016/j.mce.2019.110572] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 09/01/2019] [Accepted: 09/03/2019] [Indexed: 12/31/2022]
Abstract
Overexpression of the transcription factor, E2F8, has been associated with ovarian cancer. Objectives of this study were to determine: 1) if E2F8 gene expression in granulosa cells (GC) and theca cells (TC) change with follicular development, and 2) if E2F8 mRNA abundance in TC and GC is hormonally regulated. Using real-time PCR, E2F8 mRNA abundance in GC and TC was greater (P < 0.05) in small than large follicles. FGF9 induced an increase (P < 0.05) in E2F8 mRNA abundance by 1.6- to 7-fold in large-follicle (8-20 mm) TC and GC as well as in small-follicle (1-5 mm) GC. Abundance of E2F8 mRNA in TC was increased (P < 0.05) with FGF2, FGF9 or VEGFA treatments alone in vitro, and concomitant treatment of VEGFA with FGF9 increased (P < 0.05) abundance of E2F8 mRNA above any of the singular treatments; BMP4, WNT3A and LH were without effect. IGF1 amplified the stimulatory effect of FGF9 on E2F8 mRNA abundance by 2.7-fold. Collectively, our studies show for the first time that follicular E2F8 is developmentally and hormonally regulated indicating that E2F8 may be involved in follicular development.
Collapse
Affiliation(s)
- Breanne C Morrell
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK, 74078, USA
| | - Lingna Zhang
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK, 74078, USA
| | - Luis F Schütz
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK, 74078, USA
| | - M Chiara Perego
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK, 74078, USA
| | - Excel Rio S Maylem
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK, 74078, USA
| | - Leon J Spicer
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK, 74078, USA.
| |
Collapse
|
19
|
Liu E, Kaidonis G, McComish BJ, Gillies MC, Abhary S, Essex RW, Chang JH, Pal B, Daniell M, Lake S, Petrovsky N, Hewitt AW, Jenkins A, Lamoureux EL, Gleadle JM, Craig JE, Burdon KP. MicroRNA-Related Genetic Variants Are Associated With Diabetic Retinopathy in Type 1 Diabetes Mellitus. ACTA ACUST UNITED AC 2019; 60:3937-3942. [DOI: 10.1167/iovs.18-25570] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Ebony Liu
- Department of Ophthalmology, Flinders University, College of Medicine and Public Health, Flinders Medical Centre, Adelaide, South Australia, Australia
| | - Georgia Kaidonis
- Department of Ophthalmology, Flinders University, College of Medicine and Public Health, Flinders Medical Centre, Adelaide, South Australia, Australia
| | - Bennet J. McComish
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Mark C. Gillies
- Save Sight Institute, Clinical Ophthalmology and Eye Health, the University of Sydney, Sydney, New South Wales, Australia
| | - Sotoodeh Abhary
- Department of Ophthalmology, Flinders University, College of Medicine and Public Health, Flinders Medical Centre, Adelaide, South Australia, Australia
| | - Rohan W. Essex
- Academic Unit of Ophthalmology, Australian National University, Canberra, Australia
| | - John H. Chang
- School of Medical Sciences, University of NSW, Sydney, New South Wales, Australia
- Medical Retina Service, Moorfields Eye Hospital, London, United Kingdom
| | - Bishwanath Pal
- Medical Retina Service, Moorfields Eye Hospital, London, United Kingdom
| | - Mark Daniell
- Department of Ophthalmology, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Stewart Lake
- Department of Ophthalmology, Flinders University, College of Medicine and Public Health, Flinders Medical Centre, Adelaide, South Australia, Australia
| | - Nikolai Petrovsky
- Department of Endocrinology, Flinders University, Flinders Medical Centre, Adelaide, South Australia, Australia
| | - Alex W. Hewitt
- Centre for Eye Research Australia, University of Melbourne, Melbourne, Victoria, Australia
| | - Alicia Jenkins
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, New South Wales, Australia
- St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Ecosse L. Lamoureux
- Centre for Eye Research Australia, University of Melbourne, Melbourne, Victoria, Australia
- Singapore Eye Research Institute, Singapore
- Duke-NUS Medical School, Singapore
| | - Jonathan M. Gleadle
- Renal Medicine, College of Medicine and Public Health, Flinders University, Flinders Medical Centre, Adelaide, South Australia
| | - Jamie E. Craig
- Department of Ophthalmology, Flinders University, College of Medicine and Public Health, Flinders Medical Centre, Adelaide, South Australia, Australia
| | - Kathryn P. Burdon
- Department of Ophthalmology, Flinders University, College of Medicine and Public Health, Flinders Medical Centre, Adelaide, South Australia, Australia
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
20
|
Nichols JA, Perego MC, Schütz LF, Hemple AM, Spicer LJ. Hormonal regulation of vascular endothelial growth factor A (VEGFA) gene expression in granulosa and theca cells of cattle1. J Anim Sci 2019; 97:3034-3045. [PMID: 31077271 DOI: 10.1093/jas/skz164] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 05/10/2019] [Indexed: 12/13/2022] Open
Abstract
Vascular endothelial growth factor A (VEGFA) stimulates angiogenesis and is associated with increased vascularity in ovarian follicles of cattle. The objectives of this study were to investigate the developmental and hormonal regulation of VEGFA expression in ovarian granulosa and theca cells (TC) of cattle. Bovine ovaries were collected from a local slaughterhouse and granulosa cells (GC) and TC were collected from small (SM; 1 to 5 mm) and large (LG; 8 to 20 mm) follicles. Cells were collected fresh or cultured in serum-free medium and treated with various factors that regulate angiogenesis and follicular development. RNA was collected for analysis of VEGFA mRNA abundance via quantitative PCR. In SM-follicle GC (SMGC), prostaglandin E2 (PGE2) and FSH decreased (P < 0.05) VEGFA mRNA abundance by 30 to 46%, whereas in LG-follicle GC (LGGC), PGE2 and FSH were without effect (P > 0.10). In SMGC, dihydrotestosterone (DHT), sonic hedgehog (SHH), and growth differentiation factor-9 (GDF9) decreased (P < 0.05) VEGFA expression by 30 to 40%. Fibroblast growth factor-9 (FGF9) and estradiol (E2) were without effect (P > 0.10) on VEGFA mRNA in both SMGC and LGGC, whereas progesterone increased (P < 0.05) VEGFA mRNA in LGGC but had no effect in LGTC. Bone morphogenetic protein-4 (BMP4), LH, and FGF9 increased (P < 0.05) abundance of VEGFA mRNA by 1.5- to 1.9-fold in LGTC. Insulin-like growth factor-1 (IGF1) was without effect (P > 0.10) on VEGFA mRNA in both TC and GC. An E2F transcription factor inhibitor, HLM0064741 (E2Fi), dramatically (i.e., 8- to 13-fold) stimulated (P < 0.01) the expression of VEGFA mRNA expression in both SMGC and LGTC. Abundance of VEGFA mRNA was greater (P < 0.05) in LGGC and SMGC than in LGTC. Also, SMTC had greater (P < 0.05) abundance of VEGFA mRNA than LGTC. In conclusion, VEGFA mRNA abundance was greater in GC than TC, and VEGFA expression decreased in TC during follicle development. Some treatments either suppressed, stimulated, or had no effect on VEGFA expression depending on the cell type. The inhibition of E2F transcription factors had the greatest stimulatory effect of all treatments evaluated, and thus, E2Fs may play an important role in regulating angiogenesis during follicle growth in cattle.
Collapse
Affiliation(s)
- Jacqueline A Nichols
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK, 74078
| | - Maria Chiara Perego
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK, 74078
| | - Luis F Schütz
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK, 74078
| | - Amber M Hemple
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK, 74078
| | - Leon J Spicer
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK, 74078
| |
Collapse
|
21
|
Dassanayaka S, Brittian KR, Jurkovic A, Higgins LA, Audam TN, Long BW, Harrison LT, Militello G, Riggs DW, Chitre MG, Uchida S, Muthusamy S, Gumpert AM, Jones SP. E2f1 deletion attenuates infarct-induced ventricular remodeling without affecting O-GlcNAcylation. Basic Res Cardiol 2019; 114:28. [PMID: 31152247 DOI: 10.1007/s00395-019-0737-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/20/2019] [Indexed: 01/05/2023]
Abstract
Several post-translational modifications figure prominently in ventricular remodeling. The beta-O-linkage of N-acetylglucosamine (O-GlcNAc) to proteins has emerged as an important signal in the cardiovascular system. Although there are limited insights about the regulation of the biosynthetic pathway that gives rise to the O-GlcNAc post-translational modification, much remains to be elucidated regarding the enzymes, such as O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA), which regulate the presence/absence of O-GlcNAcylation. Recently, we showed that the transcription factor, E2F1, could negatively regulate OGT and OGA expression in vitro. The present study sought to determine whether E2f1 deletion would improve post-infarct ventricular function by de-repressing expression of OGT and OGA. Male and female mice were subjected to non-reperfused myocardial infarction (MI) and followed for 1 or 4 week. MI significantly increased E2F1 expression. Deletion of E2f1 alone was not sufficient to alter OGT or OGA expression in a naïve setting. Cardiac dysfunction was significantly attenuated at 1-week post-MI in E2f1-ablated mice. During chronic heart failure, E2f1 deletion also attenuated cardiac dysfunction. Despite the improvement in function, OGT and OGA expression was not normalized and protein O-GlcNAcyltion was not changed at 1-week post-MI. OGA expression was significantly upregulated at 4-week post-MI but overall protein O-GlcNAcylation was not changed. As an alternative explanation, we also performed guided transcriptional profiling of predicted targets of E2F1, which indicated potential differences in cardiac metabolism, angiogenesis, and apoptosis. E2f1 ablation increased heart size and preserved remote zone capillary density at 1-week post-MI. During chronic heart failure, cardiomyocytes in the remote zone of E2f1-deleted hearts were larger than wildtype. These data indicate that, overall, E2f1 exerts a deleterious effect on ventricular remodeling. Thus, E2f1 deletion improves ventricular remodeling with limited impact on enzymes regulating O-GlcNAcylation.
Collapse
Affiliation(s)
- Sujith Dassanayaka
- Division of Cardiovascular Medicine, Department of Medicine, Diabetes and Obesity Center, Institute of Molecular Cardiology, University of Louisville, 580 South Preston Street-321F, Delia Baxter Building-321F, Louisville, KY, 40202, USA
| | - Kenneth R Brittian
- Division of Cardiovascular Medicine, Department of Medicine, Diabetes and Obesity Center, Institute of Molecular Cardiology, University of Louisville, 580 South Preston Street-321F, Delia Baxter Building-321F, Louisville, KY, 40202, USA
| | - Andrea Jurkovic
- Division of Cardiovascular Medicine, Department of Medicine, Diabetes and Obesity Center, Institute of Molecular Cardiology, University of Louisville, 580 South Preston Street-321F, Delia Baxter Building-321F, Louisville, KY, 40202, USA
| | - Lauren A Higgins
- Division of Cardiovascular Medicine, Department of Medicine, Diabetes and Obesity Center, Institute of Molecular Cardiology, University of Louisville, 580 South Preston Street-321F, Delia Baxter Building-321F, Louisville, KY, 40202, USA
| | - Timothy N Audam
- Division of Cardiovascular Medicine, Department of Medicine, Diabetes and Obesity Center, Institute of Molecular Cardiology, University of Louisville, 580 South Preston Street-321F, Delia Baxter Building-321F, Louisville, KY, 40202, USA
| | - Bethany W Long
- Division of Cardiovascular Medicine, Department of Medicine, Diabetes and Obesity Center, Institute of Molecular Cardiology, University of Louisville, 580 South Preston Street-321F, Delia Baxter Building-321F, Louisville, KY, 40202, USA
| | - Linda T Harrison
- Division of Cardiovascular Medicine, Department of Medicine, Diabetes and Obesity Center, Institute of Molecular Cardiology, University of Louisville, 580 South Preston Street-321F, Delia Baxter Building-321F, Louisville, KY, 40202, USA
| | - Giuseppe Militello
- Division of Cardiovascular Medicine, Department of Medicine, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA
| | - Daniel W Riggs
- Division of Cardiovascular Medicine, Department of Medicine, Diabetes and Obesity Center, Institute of Molecular Cardiology, University of Louisville, 580 South Preston Street-321F, Delia Baxter Building-321F, Louisville, KY, 40202, USA
| | - Mitali G Chitre
- Division of Cardiovascular Medicine, Department of Medicine, Diabetes and Obesity Center, Institute of Molecular Cardiology, University of Louisville, 580 South Preston Street-321F, Delia Baxter Building-321F, Louisville, KY, 40202, USA
| | - Shizuka Uchida
- Division of Cardiovascular Medicine, Department of Medicine, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA
| | - Senthilkumar Muthusamy
- Division of Cardiovascular Medicine, Department of Medicine, Diabetes and Obesity Center, Institute of Molecular Cardiology, University of Louisville, 580 South Preston Street-321F, Delia Baxter Building-321F, Louisville, KY, 40202, USA
| | - Anna M Gumpert
- Division of Cardiovascular Medicine, Department of Medicine, Diabetes and Obesity Center, Institute of Molecular Cardiology, University of Louisville, 580 South Preston Street-321F, Delia Baxter Building-321F, Louisville, KY, 40202, USA
| | - Steven P Jones
- Division of Cardiovascular Medicine, Department of Medicine, Diabetes and Obesity Center, Institute of Molecular Cardiology, University of Louisville, 580 South Preston Street-321F, Delia Baxter Building-321F, Louisville, KY, 40202, USA.
| |
Collapse
|
22
|
McGrory CL, Ryan KM, Kolshus E, McLoughlin DM. Peripheral blood E2F1 mRNA in depression and following electroconvulsive therapy. Prog Neuropsychopharmacol Biol Psychiatry 2019; 89:380-385. [PMID: 30365982 DOI: 10.1016/j.pnpbp.2018.10.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/19/2018] [Accepted: 10/21/2018] [Indexed: 12/28/2022]
Abstract
The E2F transcription factors are a group of proteins that bind to the promotor region of the adenovirus E2 gene. E2F1, the first family member to be cloned, is linked to functions including cell proliferation and apoptosis, DNA repair, cell senescence and metabolism. We recently performed a deep sequencing study of micro-RNA changes in whole blood following ECT. Two micro-RNAs (miR-126-3p and miR-106a-5p) were identified and gene targeting analysis identified E2F1 as a shared target of these miRNAs. To our knowledge, no studies have examined E2F1 mRNA levels in patients with depression. Peripheral blood E2F1 mRNA levels were therefore examined in patients with depression, compared to healthy controls, and the effects of a course of ECT on peripheral blood E2F1 mRNA was investigated. Depressed patient and healthy control groups were balanced on the basis of age and sex. E2F1 mRNA levels were significantly lower in depressed patients in comparison to controls (p = .009) but did not change with ECT. There was no relationship between baseline E2F1 levels and depression severity, response to treatment, presence of psychosis or polarity of depression. There were no significant correlations between E2F1 levels and mood scores based on the HAM-D24. These results indicate that reduced peripheral blood E2F1 mRNA could be a trait feature of depression.
Collapse
Affiliation(s)
- Claire L McGrory
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland; Department of Psychiatry, Trinity College Dublin, St Patrick's University Hospital, Dublin 8, Ireland
| | - Karen M Ryan
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland; Department of Psychiatry, Trinity College Dublin, St Patrick's University Hospital, Dublin 8, Ireland
| | - Erik Kolshus
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland; Department of Psychiatry, Trinity College Dublin, St Patrick's University Hospital, Dublin 8, Ireland
| | - Declan M McLoughlin
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland; Department of Psychiatry, Trinity College Dublin, St Patrick's University Hospital, Dublin 8, Ireland.
| |
Collapse
|
23
|
Xu S, Tao J, Yang L, Zhang E, Boriboun C, Zhou J, Sun T, Cheng M, Huang K, Shi J, Dong N, Liu Q, Zhao TC, Qiu H, Harris RA, Chandel NS, Losordo DW, Qin G. E2F1 Suppresses Oxidative Metabolism and Endothelial Differentiation of Bone Marrow Progenitor Cells. Circ Res 2018; 122:701-711. [PMID: 29358228 DOI: 10.1161/circresaha.117.311814] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 12/18/2017] [Accepted: 01/19/2018] [Indexed: 12/13/2022]
Abstract
RATIONALE The majority of current cardiovascular cell therapy trials use bone marrow progenitor cells (BM PCs) and achieve only modest efficacy; the limited potential of these cells to differentiate into endothelial-lineage cells is one of the major barriers to the success of this promising therapy. We have previously reported that the E2F transcription factor 1 (E2F1) is a repressor of revascularization after ischemic injury. OBJECTIVE We sought to define the role of E2F1 in the regulation of BM PC function. METHODS AND RESULTS Ablation of E2F1 (E2F1 deficient) in mouse BM PCs increases oxidative metabolism and reduces lactate production, resulting in enhanced endothelial differentiation. The metabolic switch in E2F1-deficient BM PCs is mediated by a reduction in the expression of pyruvate dehydrogenase kinase 4 and pyruvate dehydrogenase kinase 2; overexpression of pyruvate dehydrogenase kinase 4 reverses the enhancement of oxidative metabolism and endothelial differentiation. Deletion of E2F1 in the BM increases the amount of PC-derived endothelial cells in the ischemic myocardium, enhances vascular growth, reduces infarct size, and improves cardiac function after myocardial infarction. CONCLUSION Our results suggest a novel mechanism by which E2F1 mediates the metabolic control of BM PC differentiation, and strategies that inhibit E2F1 or enhance oxidative metabolism in BM PCs may improve the effectiveness of cell therapy.
Collapse
Affiliation(s)
- Shiyue Xu
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Jun Tao
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Liu Yang
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Eric Zhang
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Chan Boriboun
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Junlan Zhou
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Tianjiao Sun
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Min Cheng
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Kai Huang
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Jiawei Shi
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Nianguo Dong
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Qinghua Liu
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Ting C Zhao
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Hongyu Qiu
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Robert A Harris
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Navdeep S Chandel
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Douglas W Losordo
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Gangjian Qin
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.).
| |
Collapse
|
24
|
Wang H, Qiu L, Ma Y, Zhang L, Chen L, Li C, Geng X, You X, Gao X. Naoxintong inhibits myocardial infarction injury by VEGF/eNOS signaling-mediated neovascularization. JOURNAL OF ETHNOPHARMACOLOGY 2017; 209:13-23. [PMID: 28669772 DOI: 10.1016/j.jep.2017.06.040] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 06/14/2017] [Accepted: 06/20/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Naoxintong capsules (NXT), a traditional Chinese Medical preparation, are widely used for treatment of cardiovascular diseases, while the mechanism is still unclear. MATERIALS AND METHODS Myocardial infarction (MI) was induced by ligation of the left coronary artery in mice. Echocardiographic measurements were performed to do physiological assessments of left ventricle (LV) function. Histological and immunohistochemical staining was used to determine infarct size, capillary density, tissue endothelial nitric oxide synthase (eNOS) expression. Bone Marrow Transplantation (BMT) model and flow cytometric (FCM) analyses were applied to assay endothelial progenitor cells (EPCs) mobilization. Quantitative Real-Time Reverse Transcription Polymerase Chain Reaction (qRT-PCR), Western blotting and enzyme-linked immunosorbent assay (ELISA) were performed to detect the expressions of vascular endothelial growth factor (VEGF), kinase domain region (KDR), phosphorylated-Akt (p-Akt), phosphorylated-eNOS (p-eNOS). RESULTS NXT administration reduced myocardium fibrosis and increased myocardium capillary density in response to MI. NXT increased circulating Sca1+/ Fetal liver kinase 1 (Flk1)+ mononuclear cells (MNCs) and soluble Kit ligand (sKitL) of bone marrow (BM) in response to MI. In mice transplanted with green fluorescent protein (GFP) BM cells, NXT increased the numbers of GFP-positive cells at the border zone of the ischemic region in MI-induced mice. NXT increased the numbers of eNOS-expressing BM-derived cells in tissues, which was involved in increased the expressions of VEGF, KDR, p-eNOS, p-Akt in the myocardium. CONCLUSION NXT-mediated recovery in MI-induced mice was involved in mobilization and incorporation of bone marrow-derived EPCs/circulating angiogenic cells (CACs) leading to enhancement of neovascularization via VEGF/eNOS signaling.
Collapse
Affiliation(s)
- Hong Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Lizhen Qiu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yake Ma
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lusha Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lu Chen
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Chunxiao Li
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiao Geng
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xingyu You
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiumei Gao
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| |
Collapse
|
25
|
Cai H, Ma Y, Jiang L, Mu Z, Jiang Z, Chen X, Wang Y, Yang GY, Zhang Z. Hypoxia Response Element-Regulated MMP-9 Promotes Neurological Recovery via Glial Scar Degradation and Angiogenesis in Delayed Stroke. Mol Ther 2017; 25:1448-1459. [PMID: 28396199 DOI: 10.1016/j.ymthe.2017.03.020] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 03/10/2017] [Accepted: 03/10/2017] [Indexed: 12/14/2022] Open
Abstract
Matrix metalloproteinase 9 (MMP-9) plays a beneficial role in the delayed phase of middle cerebral artery occlusion (MCAO). However, the mechanism is obscure. Here, we constructed hypoxia response element (HRE)-regulated MMP-9 to explore its effect on glial scars and neurogenesis in delayed ischemic stroke. Adult male Institute of Cancer Research (ICR) mice underwent MCAO and received a stereotactic injection of lentivirus carrying HRE-MMP-9 or normal saline (NS)/lentivirus-GFP 7 days after ischemia. We found that HRE-MMP-9 improved neurological outcomes, reduced ischemia-induced brain atrophy, and degraded glial scars (p < 0.05). Furthermore, HRE-MMP-9 increased the number of microvessels in the peri-infarct area (p < 0.001), which may have been due to the accumulation of endogenous endothelial progenitor cells (EPCs) in the peri-infarct area after glial scar degradation. Finally, HRE-MMP-9 increased the number of bromodeoxyuridine-positive (BrdU+)/NeuN+ cells and the expression of PSD-95 in the peri-infarct area (p < 0.01). These changes could be blocked by vascular endothelial growth factor receptor 2 (VEGFR2) inhibitor SU5416 and MMP-9 inhibitor 2-[[(4-phenoxyphenyl)sulfonyl]methyl]-thiirane (SB-3CT). Our results provided a novel mechanism by which glial scar degradation and vascular endothelial growth factor (VEGF)/VEGFR2-dependent angiogenesis may be key procedures for neurological recovery in delayed ischemic stroke after HRE-MMP-9 treatment. Therefore, HRE-MMP-9 overexpression in the delayed ischemic brain is a promising approach for neurological recovery.
Collapse
Affiliation(s)
- Hongxia Cai
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Neurology, Yangzhou University Affiliated Hospital, Yangzhou No. 1 People's Hospital, Jiangsu Province, 225000, China
| | - Yuanyuan Ma
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Neuroscience and Neuroengineering Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Lu Jiang
- Neuroscience and Neuroengineering Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Zhihao Mu
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Neuroscience and Neuroengineering Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Zhen Jiang
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Neuroscience and Neuroengineering Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Xiaoyan Chen
- Neuroscience and Neuroengineering Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yongting Wang
- Neuroscience and Neuroengineering Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Guo-Yuan Yang
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Neuroscience and Neuroengineering Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Zhijun Zhang
- Neuroscience and Neuroengineering Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China.
| |
Collapse
|
26
|
Denechaud PD, Fajas L, Giralt A. E2F1, a Novel Regulator of Metabolism. Front Endocrinol (Lausanne) 2017; 8:311. [PMID: 29176962 PMCID: PMC5686046 DOI: 10.3389/fendo.2017.00311] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 10/26/2017] [Indexed: 01/09/2023] Open
Abstract
In the past years, several lines of evidence have shown that cell cycle regulatory proteins also can modulate metabolic processes. The transcription factor E2F1 is a central player involved in cell cycle progression, DNA-damage response, and apoptosis. Its crucial role in the control of cell fate has been extensively studied and reviewed before; however, here, we focus on the participation of E2F1 in the regulation of metabolism. We summarize recent findings about the cell cycle-independent roles of E2F1 in various tissues that contribute to global metabolic homeostasis and highlight that E2F1 activity is increased during obesity. Finally, coming back to the pivotal role of E2F1 in cancer development, we discuss how E2F1 links cell cycle progression with different metabolic adaptations required for cell growth and survival.
Collapse
Affiliation(s)
| | - Lluis Fajas
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Albert Giralt
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
- *Correspondence: Albert Giralt,
| |
Collapse
|
27
|
E2F1 Hinders Skin Wound Healing by Repressing Vascular Endothelial Growth Factor (VEGF) Expression, Neovascularization, and Macrophage Recruitment. PLoS One 2016; 11:e0160411. [PMID: 27490344 PMCID: PMC4973919 DOI: 10.1371/journal.pone.0160411] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 07/19/2016] [Indexed: 12/21/2022] Open
Abstract
Background Refractory surface of wound and dermal chronic ulcer are largely attributed to poor neovascularization. We have previously shown that E2F1 suppresses VEGF expression in the ischemic heart, and that genetic deletion of E2F1 leads to better cardiac recovery. However, whether E2F1 has a role in dermal wound healing is currently not known. Methods and Results Skin wounds were surgically induced in E2F1-null (E2F1–/–) mice and WT littermates. E2F1–/– displayed an accelerated wound healing including wound closure, dermal thickening and collagen deposition, which was associated with an increased endothelial cell proliferation and greater vessel density in the border zone of the wound. Furthermore, more macrophages were recruited to the skin lesions and the level of VEGF expression was markedly higher in E2F1–/– than in WT mice. Conclusions E2F1 hinders skin wound healing by suppressing VEGF expression, neovascularization, and macrophage recruitment. Strategies that target E2F1 may enhance wound healing.
Collapse
|
28
|
Ertosun MG, Hapil FZ, Osman Nidai O. E2F1 transcription factor and its impact on growth factor and cytokine signaling. Cytokine Growth Factor Rev 2016; 31:17-25. [PMID: 26947516 DOI: 10.1016/j.cytogfr.2016.02.001] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 02/27/2016] [Indexed: 12/13/2022]
Abstract
E2F1 is a transcription factor involved in cell cycle regulation and apoptosis. The transactivation capacity of E2F1 is regulated by pRb. In its hypophosphorylated form, pRb binds and inactivates DNA binding and transactivating functions of E2F1. The growth factor stimulation of cells leads to activation of CDKs (cyclin dependent kinases), which in turn phosphorylate Rb and hyperphosphorylated Rb is released from E2F1 or E2F1/DP complex, and free E2F1 can induce transcription of several genes involved in cell cycle entry, induction or inhibition of apoptosis. Thus, growth factors and cytokines generally utilize E2F1 to direct cells to either fate. Furthermore, E2F1 regulates expressions of various cytokines and growth factor receptors, establishing positive or negative feedback mechanisms. This review focuses on the relationship between E2F1 transcription factor and cytokines (IL-1, IL-2, IL-3, IL-6, TGF-beta, G-CSF, LIF), growth factors (EGF, KGF, VEGF, IGF, FGF, PDGF, HGF, NGF), and interferons (IFN-α, IFN-β and IFN-γ).
Collapse
Affiliation(s)
- Mustafa Gokhan Ertosun
- Akdeniz University, Faculty of Medicine, Department of Medical Biology and Genetic, Kampus, Antalya 07070, Turkey
| | - Fatma Zehra Hapil
- Akdeniz University, Faculty of Medicine, Department of Medical Biology and Genetic, Kampus, Antalya 07070, Turkey
| | - Ozes Osman Nidai
- Akdeniz University, Faculty of Medicine, Department of Medical Biology and Genetic, Kampus, Antalya 07070, Turkey.
| |
Collapse
|