1
|
Zhang J, Yan H, Wang Y, Yue X, Wang M, Liu L, Qiao P, Zhu Y, Li Z. Emerging insights into pulmonary hypertension: the potential role of mitochondrial dysfunction and redox homeostasis. Mol Cell Biochem 2024:10.1007/s11010-024-05096-9. [PMID: 39254871 DOI: 10.1007/s11010-024-05096-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 08/14/2024] [Indexed: 09/11/2024]
Abstract
Pulmonary hypertension (PH) is heterogeneous diseases that can lead to death due to progressive right heart failure. Emerging evidence suggests that, in addition to its role in ATP production, changes in mitochondrial play a central role in their pathogenesis, regulating integrated metabolic and signal transduction pathways. This review focuses on the basic principles of mitochondrial redox status in pulmonary vascular and right ventricular disorders, a series of dysfunctional processes including mitochondrial quality control (mitochondrial biogenesis, mitophagy, mitochondrial dynamics, mitochondrial unfolded protein response) and mitochondrial redox homeostasis. In addition, we will summarize how mitochondrial renewal and dynamic changes provide innovative insights for studying and evaluating PH. This will provide us with a clearer understanding of the initial signal transmission of mitochondria in PH, which would further improve our understanding of the pathogenesis of PH.
Collapse
Affiliation(s)
- Junming Zhang
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Huimin Yan
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Yan Wang
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Xian Yue
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Meng Wang
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Limin Liu
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Pengfei Qiao
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Yixuan Zhu
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Zhichao Li
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China.
| |
Collapse
|
2
|
Zhang X, Yang Z, Su S, Nan X, Xie X, Li Z, Lu D. Kaempferol ameliorates pulmonary vascular remodeling in chronic hypoxia-induced pulmonary hypertension rats via regulating Akt-GSK3β-cyclin axis. Toxicol Appl Pharmacol 2023; 466:116478. [PMID: 36940862 DOI: 10.1016/j.taap.2023.116478] [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: 10/25/2022] [Revised: 03/02/2023] [Accepted: 03/16/2023] [Indexed: 03/23/2023]
Abstract
Excessive proliferation of pulmonary artery smooth muscle cells (PASMCs) is considered a major contributor to elevated pulmonary vascular resistance and a key mechanism of vascular remodeling in hypoxia-induced pulmonary hypertension (HPH). Kaempferol is a natural flavonoid compound and can be derived from numerous common medicinal herbs and vegetables, which exhibit antiproliferative and proapoptotic properties, however, the effects of kaempferol on vascular remodeling in HPH remain unexplored. In this study, SD rats were placed in a hypobaric hypoxia chamber for four weeks to establish a pulmonary hypertension model and given either kaempferol or sildenafil (an inhibitor of PDE-5) during days 1-28, after which the hemodynamic parameter and pulmonary vascular morphometry were assessed. Furthermore, primary rat PASMCs were exposed to hypoxic conditions to generate a cell proliferation model, then incubated with either kaempferol or LY294002 (an inhibitor of PI3K). Immunoblotting and real-time quantitative PCR assessed the protein and mRNA expression levels in HPH rat lungs and PASMCs. We found that kaempferol reduced pulmonary artery pressure and pulmonary vascular remodeling, and alleviated right ventricular hypertrophy in HPH rats. The mechanistic analysis demonstrated that kaempferol reduced the protein levels of phosphorylation of Akt and GSK3β, leading to decreased expression of pro-proliferation (CDK2, CDK4, Cyclin D1, and PCNA) and anti-apoptotic related proteins (Bcl-2) and increased expression of pro-apoptosis proteins (Bax and cleaved caspase 3). These results collectively demonstrate that kaempferol ameliorates HPH in rats by inhibiting PASMC proliferation and pro-apoptosis via modulation of the Akt/GSK3β/CyclinD axis.
Collapse
Affiliation(s)
- Xiaonan Zhang
- Research Center for High Altitude Medicine, Key Laboratory for High Altitude Medicine (Ministry of Education), Laboratory for High Altitude Medicine of Qinghai Province, Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining 810001, China; Qinghai Provincial People's Hospital, Xining 810007, China
| | - Zhanting Yang
- Research Center for High Altitude Medicine, Key Laboratory for High Altitude Medicine (Ministry of Education), Laboratory for High Altitude Medicine of Qinghai Province, Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining 810001, China
| | - Shanshan Su
- Xining Customs Technical Center, Key Laboratory of Food Safety Research in Qinghai Province, Qinghai, Xining 810003, China
| | - Xingmei Nan
- Research Center for High Altitude Medicine, Key Laboratory for High Altitude Medicine (Ministry of Education), Laboratory for High Altitude Medicine of Qinghai Province, Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining 810001, China
| | - Xin Xie
- School of Ecological and Environmental Engineering, Qinghai University, Xining 810016, China
| | - Zhanqiang Li
- Research Center for High Altitude Medicine, Key Laboratory for High Altitude Medicine (Ministry of Education), Laboratory for High Altitude Medicine of Qinghai Province, Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining 810001, China.
| | - Dianxiang Lu
- Research Center for High Altitude Medicine, Key Laboratory for High Altitude Medicine (Ministry of Education), Laboratory for High Altitude Medicine of Qinghai Province, Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining 810001, China; Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610086, China.
| |
Collapse
|
3
|
Proteasome Inhibitors Decrease the Viability of Pulmonary Arterial Smooth Muscle Cells by Restoring Mitofusin-2 Expression under Hypoxic Conditions. Biomedicines 2022; 10:biomedicines10040873. [PMID: 35453623 PMCID: PMC9030547 DOI: 10.3390/biomedicines10040873] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/01/2022] [Accepted: 04/07/2022] [Indexed: 02/04/2023] Open
Abstract
Pulmonary hypertension (PH) is a severe progressive disease, and the uncontrolled proliferation of pulmonary artery smooth muscle cells (PASMCs) is one of the main causes. Mitofusin-2 (MFN2) profoundly inhibits cell growth and proliferation in a variety of tumor cell lines and rat vascular smooth muscle cells. Down-regulation of MFN2 is known to contribute to PH. Proteasome inhibitors have been shown to inhibit the proliferation of PASMCs; however, there is no study on the regulation of proteasome inhibitors through MFN-2 in the proliferation of PASMCs, a main pathophysiology of PH. In this study, PASMCs were exposed to hypoxic conditions and the expression of MFN2 and cleaved-PARP1 were detected by Western blotting. The effects of hypoxia and proteasome inhibitors on the cell viability of PASMC cells were detected by CCK8 assay. The results indicated that hypoxia increases the viability and reduces the expression of MFN2 in a PASMCs model. MFN2 overexpression inhibits the hypoxia-induced proliferation of PASMCs. In addition, proteasome inhibitors, bortezomib and marizomib, restored the decreased expression of MFN2 under hypoxic conditions, inhibited hypoxia-induced proliferation and induced the expression of cleaved-PARP1. These results suggest that bortezomib and marizomib have the potential to improve the hypoxia-induced proliferation of PASMCs by restoring MFN2 expression.
Collapse
|
4
|
Shi HH, Zhang LY, Chen LP, Yang JY, Wang CC, Xue CH, Wang YM, Zhang TT. EPA-Enriched Phospholipids Alleviate Renal Interstitial Fibrosis in Spontaneously Hypertensive Rats by Regulating TGF-β Signaling Pathways. Mar Drugs 2022; 20:md20020152. [PMID: 35200681 PMCID: PMC8879699 DOI: 10.3390/md20020152] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/12/2022] [Accepted: 02/17/2022] [Indexed: 11/29/2022] Open
Abstract
Hypertensive nephropathy is a chronic kidney disease caused by hypertension. Eicosapentaenoic acid (EPA) has been reported to possess an antihypertensive effect, and our previous study suggested that EPA-enriched phospholipid (EPA-PL) had more significant bioactivities compared with traditional EPA. However, the effect of dietary EPA-PL on hypertensive nephropathy has not been studied. The current study was designed to examine the protection of EPA-PL against kidney damage in spontaneously hypertensive rats (SHRs). Treatment with EPA-PL for three weeks significantly reduced blood pressure through regulating the renin–angiotensin system in SHRs. Moreover, dietary EPA-PL distinctly alleviated kidney dysfunction in SHRs, evidenced by reduced plasma creatinine, blood urea nitrogen, and 24 h proteinuria. Histology results revealed that treatment of SHRs with EPA-PL alleviated renal injury and reduced tubulointerstitial fibrosis. Further mechanistic studies indicated that dietary EPA-PL remarkably inhibited the activation of TGF-β and Smad 3, elevated the phosphorylation level of PI3K/AKT, suppressed the activation of NF-κB, reduced the expression of pro-inflammatory cytokines, including IL-1β and IL-6, and repressed the oxidative stress and the mitochondria-mediated apoptotic signaling pathway in the kidney. These results indicate that EPA-PL has potential value in the prevention and alleviation of hypertensive nephropathy.
Collapse
Affiliation(s)
- Hao-Hao Shi
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China; (H.-H.S.); (L.-Y.Z.); (L.-P.C.); (J.-Y.Y.); (C.-C.W.); (C.-H.X.); (Y.-M.W.)
| | - Ling-Yu Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China; (H.-H.S.); (L.-Y.Z.); (L.-P.C.); (J.-Y.Y.); (C.-C.W.); (C.-H.X.); (Y.-M.W.)
- College of Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Li-Pin Chen
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China; (H.-H.S.); (L.-Y.Z.); (L.-P.C.); (J.-Y.Y.); (C.-C.W.); (C.-H.X.); (Y.-M.W.)
| | - Jin-Yue Yang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China; (H.-H.S.); (L.-Y.Z.); (L.-P.C.); (J.-Y.Y.); (C.-C.W.); (C.-H.X.); (Y.-M.W.)
| | - Cheng-Cheng Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China; (H.-H.S.); (L.-Y.Z.); (L.-P.C.); (J.-Y.Y.); (C.-C.W.); (C.-H.X.); (Y.-M.W.)
| | - Chang-Hu Xue
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China; (H.-H.S.); (L.-Y.Z.); (L.-P.C.); (J.-Y.Y.); (C.-C.W.); (C.-H.X.); (Y.-M.W.)
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, China
| | - Yu-Ming Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China; (H.-H.S.); (L.-Y.Z.); (L.-P.C.); (J.-Y.Y.); (C.-C.W.); (C.-H.X.); (Y.-M.W.)
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, China
| | - Tian-Tian Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China; (H.-H.S.); (L.-Y.Z.); (L.-P.C.); (J.-Y.Y.); (C.-C.W.); (C.-H.X.); (Y.-M.W.)
- Correspondence: ; Tel.: +86-0532-8203-2597; Fax: +86-0532-8203-2468
| |
Collapse
|
5
|
Ling M, Quan L, Lai X, Lang L, Li F, Yang X, Fu Y, Feng S, Yi X, Zhu C, Gao P, Zhu X, Wang L, Shu G, Jiang Q, Wang S. VEGFB Promotes Myoblasts Proliferation and Differentiation through VEGFR1-PI3K/Akt Signaling Pathway. Int J Mol Sci 2021; 22:13352. [PMID: 34948148 PMCID: PMC8707860 DOI: 10.3390/ijms222413352] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/05/2021] [Accepted: 12/09/2021] [Indexed: 01/06/2023] Open
Abstract
It has been demonstrated that vascular endothelial growth factor B (VEGFB) plays a vital role in regulating vascular biological function. However, the role of VEGFB in regulating skeletal muscle cell proliferation and differentiation remains unclear. Thus, this study aimed to investigate the effects of VEGFB on C2C12 myoblast proliferation and differentiation and to explore the underlying mechanism. For proliferation, VEGFB significantly promoted the proliferation of C2C12 myoblasts with the upregulating expression of cyclin D1 and PCNA. Meanwhile, VEGFB enhanced vascular endothelial growth factor receptor 1 (VEGFR1) expression and activated the PI3K/Akt signaling pathway in a VEGFR1-dependent manner. In addition, the knockdown of VEGFR1 and inhibition of PI3K/Akt totally abolished the promotion of C2C12 proliferation induced by VEGFB, suggesting that VEGFB promoted C2C12 myoblast proliferation through the VEGFR1-PI3K/Akt signaling pathway. Regarding differentiation, VEGFB significantly stimulated the differentiation of C2C12 myoblasts via VEGFR, with elevated expressions of MyoG and MyHC. Furthermore, the knockdown of VEGFR1 rather than NRP1 eliminated the VEGFB-stimulated C2C12 differentiation. Moreover, VEGFB activated the PI3K/Akt/mTOR signaling pathway in a VEGFR1-dependent manner. However, the inhibition of PI3K/Akt/mTOR blocked the promotion of C2C12 myoblasts differentiation induced by VEGFB, indicating the involvement of the PI3K/Akt pathway. To conclude, these findings showed that VEGFB promoted C2C12 myoblast proliferation and differentiation via the VEGFR1-PI3K/Akt signaling pathway, providing new insights into the regulation of skeletal muscle development.
Collapse
Affiliation(s)
- Mingfa Ling
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.L.); (L.Q.); (X.L.); (L.L.); (F.L.); (X.Y.); (Y.F.); (S.F.); (X.Y.); (C.Z.); (P.G.); (X.Z.); (L.W.); (G.S.); (Q.J.)
- National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Lulu Quan
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.L.); (L.Q.); (X.L.); (L.L.); (F.L.); (X.Y.); (Y.F.); (S.F.); (X.Y.); (C.Z.); (P.G.); (X.Z.); (L.W.); (G.S.); (Q.J.)
- National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Xumin Lai
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.L.); (L.Q.); (X.L.); (L.L.); (F.L.); (X.Y.); (Y.F.); (S.F.); (X.Y.); (C.Z.); (P.G.); (X.Z.); (L.W.); (G.S.); (Q.J.)
- National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Limin Lang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.L.); (L.Q.); (X.L.); (L.L.); (F.L.); (X.Y.); (Y.F.); (S.F.); (X.Y.); (C.Z.); (P.G.); (X.Z.); (L.W.); (G.S.); (Q.J.)
- National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Fan Li
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.L.); (L.Q.); (X.L.); (L.L.); (F.L.); (X.Y.); (Y.F.); (S.F.); (X.Y.); (C.Z.); (P.G.); (X.Z.); (L.W.); (G.S.); (Q.J.)
- National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Xiaohua Yang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.L.); (L.Q.); (X.L.); (L.L.); (F.L.); (X.Y.); (Y.F.); (S.F.); (X.Y.); (C.Z.); (P.G.); (X.Z.); (L.W.); (G.S.); (Q.J.)
- National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Yiming Fu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.L.); (L.Q.); (X.L.); (L.L.); (F.L.); (X.Y.); (Y.F.); (S.F.); (X.Y.); (C.Z.); (P.G.); (X.Z.); (L.W.); (G.S.); (Q.J.)
- National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Shengchun Feng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.L.); (L.Q.); (X.L.); (L.L.); (F.L.); (X.Y.); (Y.F.); (S.F.); (X.Y.); (C.Z.); (P.G.); (X.Z.); (L.W.); (G.S.); (Q.J.)
- National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Xin Yi
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.L.); (L.Q.); (X.L.); (L.L.); (F.L.); (X.Y.); (Y.F.); (S.F.); (X.Y.); (C.Z.); (P.G.); (X.Z.); (L.W.); (G.S.); (Q.J.)
- National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Canjun Zhu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.L.); (L.Q.); (X.L.); (L.L.); (F.L.); (X.Y.); (Y.F.); (S.F.); (X.Y.); (C.Z.); (P.G.); (X.Z.); (L.W.); (G.S.); (Q.J.)
- National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Ping Gao
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.L.); (L.Q.); (X.L.); (L.L.); (F.L.); (X.Y.); (Y.F.); (S.F.); (X.Y.); (C.Z.); (P.G.); (X.Z.); (L.W.); (G.S.); (Q.J.)
- National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Xiaotong Zhu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.L.); (L.Q.); (X.L.); (L.L.); (F.L.); (X.Y.); (Y.F.); (S.F.); (X.Y.); (C.Z.); (P.G.); (X.Z.); (L.W.); (G.S.); (Q.J.)
- National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Lina Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.L.); (L.Q.); (X.L.); (L.L.); (F.L.); (X.Y.); (Y.F.); (S.F.); (X.Y.); (C.Z.); (P.G.); (X.Z.); (L.W.); (G.S.); (Q.J.)
- National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Gang Shu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.L.); (L.Q.); (X.L.); (L.L.); (F.L.); (X.Y.); (Y.F.); (S.F.); (X.Y.); (C.Z.); (P.G.); (X.Z.); (L.W.); (G.S.); (Q.J.)
- National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Qingyan Jiang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.L.); (L.Q.); (X.L.); (L.L.); (F.L.); (X.Y.); (Y.F.); (S.F.); (X.Y.); (C.Z.); (P.G.); (X.Z.); (L.W.); (G.S.); (Q.J.)
- National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Songbo Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.L.); (L.Q.); (X.L.); (L.L.); (F.L.); (X.Y.); (Y.F.); (S.F.); (X.Y.); (C.Z.); (P.G.); (X.Z.); (L.W.); (G.S.); (Q.J.)
- National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
6
|
Tettey A, Jiang Y, Li X, Li Y. Therapy for Pulmonary Arterial Hypertension: Glance on Nitric Oxide Pathway. Front Pharmacol 2021; 12:767002. [PMID: 34867394 PMCID: PMC8633825 DOI: 10.3389/fphar.2021.767002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/25/2021] [Indexed: 01/11/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe disease with a resultant increase of the mean pulmonary arterial pressure, right ventricular hypertrophy and eventual death. Research in recent years has produced various therapeutic options for its clinical management but the high mortality even under treatment remains a big challenge attributed to the complex pathophysiology. Studies from clinical and non-clinical experiments have revealed that the nitric oxide (NO) pathway is one of the key pathways underlying the pathophysiology of PAH. Many of the essential drugs used in the management of PAH act on this pathway highlighting its significant role in PAH. Meanwhile, several novel compounds targeting on NO pathway exhibits great potential to become future therapy medications. Furthermore, the NO pathway is found to interact with other crucial pathways. Understanding such interactions could be helpful in the discovery of new drug that provide better clinical outcomes.
Collapse
Affiliation(s)
- Abraham Tettey
- Department of Pharmacology, School of Pharmaceutical Science, Central South University, Changsha, China
| | - Yujie Jiang
- Department of Pharmacology, School of Pharmaceutical Science, Central South University, Changsha, China
| | - Xiaohui Li
- Department of Pharmacology, School of Pharmaceutical Science, Central South University, Changsha, China
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha, China
| | - Ying Li
- Department of Health Management, The Third Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha, China
| |
Collapse
|
7
|
Ahn SY, Song J, Kim YC, Kim MH, Hyun YM. Mitofusin-2 Promotes the Epithelial-Mesenchymal Transition-Induced Cervical Cancer Progression. Immune Netw 2021; 21:e30. [PMID: 34522443 PMCID: PMC8410987 DOI: 10.4110/in.2021.21.e30] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/14/2021] [Accepted: 07/17/2021] [Indexed: 12/18/2022] Open
Abstract
High expression of mitofusin-2 (MFN2), a mitochondrial fusion protein, has been frequently associated with poor prognosis of patients with cervical cancer. Here, we aimed to identify the function of MFN2 in cervical cancer to understand its influence on disease prognosis. To this end, from cervical adenocarcinoma, we performed an MTT assay and quantitative RT-PCR (qRT-PCR) analysis to assess the effects of MFN2 on the proliferation and of HeLa cells. Then, colony-formation ability and tumorigenesis were evaluated using a tumor xenograft mouse model. The migration ability related to MFN2 was also measured using a wound healing assay. Consequently, epithelial-mesenchymal transition (EMT) of MFN2-knockdowned HeLa cells originating from adenocarcinoma. markers related to MFN2 were assessed by qRT-PCR. Clinical data were analyzed using cBioPortal and The Cancer Genome Atlas. We found that MFN2 knockdown reduced the proliferation, colony formation ability, migration, and in vivo tumorigenesis of HeLa cells. Primarily, migration of MFN2-knockdowned HeLa cells decreased through the suppression of EMT. Thus, we concluded that MFN2 facilitates cancer progression and in vivo tumorigenesis in HeLa cells. These findings suggest that MFN2 could be a novel target to regulate the EMT program and tumorigenic potential in HeLa cells and might serve as a therapeutic target for cervical cancer. Taken together, this study is expected to contribute to the treatment of patients with cervical cancer.
Collapse
Affiliation(s)
- Sung Yong Ahn
- Department of Anatomy, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Jiwon Song
- Department of Anatomy, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea.,Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Yu Cheon Kim
- Department of Anatomy, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea.,Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Myoung Hee Kim
- Department of Anatomy, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea.,Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Young-Min Hyun
- Department of Anatomy, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea.,Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
8
|
Xu X, Su YL, Shi JY, Lu Q, Chen C. MicroRNA-17-5p Promotes Cardiac Hypertrophy by Targeting Mfn2 to Inhibit Autophagy. Cardiovasc Toxicol 2021; 21:759-771. [PMID: 34120306 DOI: 10.1007/s12012-021-09667-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 06/03/2021] [Indexed: 12/21/2022]
Abstract
Pathological cardiac hypertrophy is the leading cause of heart failure, and miRNAs have been recognized as key factors in cardiac hypertrophy. This study aimed to elucidate whether miR-17-5p affects cardiac hypertrophy by targeting the mitochondrial fusion protein mitofusin 2 (Mfn2)-mediated phosphatidylinositol-3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway and regulating autophagy. miR-17-5p expression was shown to be upregulated both in vivo and in vitro. In addition, a miR-17-5p inhibitor significantly reversed AngII-induced cell hypertrophy in neonatal rat left ventricle myocytes (NRVMs). In contrast to miR-17-5p expression, Mfn2 expression was inhibited in rat hearts at 4 weeks after transverse aortic constriction (TAC) and in an Ang II-induced cell hypertrophy model. We examined miR-17-5p targeting of Mfn2 by dual luciferase reporter and Western blot assays. In addition, we also verified the relationship between Mfn2 and the PI3K/AKT/mTOR pathway. Mfn2 overexpression attenuated miR-17-5p-induced cell hypertrophy, and in rat myocardial tissue, miR-17-5p induced autophagy inhibition. In summary, the results of the present study demonstrated that miR-17-5p inhibits Mfn2 expression, activates the PI3K/AKT/mTOR pathway and suppresses autophagy to promote cardiac hypertrophy.
Collapse
Affiliation(s)
- Xuan Xu
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Yi-Ling Su
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Jia-Yu Shi
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Qi Lu
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
| | - Chu Chen
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
9
|
Meng L, Yuan W, Chi H, Han R, Zhang Y, Pan X, Meng J, Liu Y, Song J, Zhong J, Liu X. Genetic deletion of CMG2 exacerbates systemic-to-pulmonary shunt-induced pulmonary arterial hypertension. FASEB J 2021; 35:e21421. [PMID: 33749907 DOI: 10.1096/fj.202000299r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 12/30/2020] [Accepted: 01/21/2021] [Indexed: 11/11/2022]
Abstract
Pulmonary arterial hypertension (PAH) secondary to congenital heart disease (CHD-PAH) with systemic-to-pulmonary shunt (SPS) is characterized by proliferative vascular remodeling. Capillary morphogenesis gene-2 (CMG2) plays a key role in cell proliferation and apoptosis. This study aimed to determine the role of CMG2 in the pathogenesis of SPS-induced PAH. CMG2 levels were significantly downregulated in pulmonary arterioles from patients with Eisenmenger syndrome and rats with SPS-induced PAH. CMG2 was highly expressed in several cells including human pulmonary arterial smooth muscle cells (HPASMCs). CMG2-/- rats exhibited more severe PAH and pulmonary vascular remodeling than wild-type rats when exposed to SPS for 8 weeks. Overexpression of CMG2 significantly inhibited proliferation and promoted apoptosis of HPASMCs, while knockdown of CMG2 promoted cell proliferation and inhibited cell apoptosis. Next-generation sequencing and subsequent validation results suggested that PI3K-AKT was the most prominent signaling pathway regulated by differentially expressed genes (DEGs) in CMG2-/- rat lungs. Our work identified a novel role for CMG2 in SPS-induced PAH based on the findings that CMG2 deficiency exacerbates SPS-induced vascular remodeling in the development of PAH, indicating that CMG2 might act as a potential target for the treatment of CHD-PAH.
Collapse
Affiliation(s)
- Liukun Meng
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Wen Yuan
- Medical Research Center & Beijing Key Laboratory of Hypertension Research, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Hongjie Chi
- Heart Center & Beijing Key Laboratory of Hypertension Research, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Ruijuan Han
- Department of Cardiology, Baotou Central hospital, Inner Mongolia, China
| | - Yeping Zhang
- Heart Center & Beijing Key Laboratory of Hypertension Research, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xiangbin Pan
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jian Meng
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Ying Liu
- Heart Center & Beijing Key Laboratory of Hypertension Research, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Jiawei Song
- Heart Center & Beijing Key Laboratory of Hypertension Research, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Jiuchang Zhong
- Heart Center & Beijing Key Laboratory of Hypertension Research, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xiaoyan Liu
- Medical Research Center & Beijing Key Laboratory of Hypertension Research, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.,Heart Center & Beijing Key Laboratory of Hypertension Research, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
10
|
Li D, Yang S, Xing Y, Pan L, Zhao R, Zhao Y, Liu L, Wu M. Novel Insights and Current Evidence for Mechanisms of Atherosclerosis: Mitochondrial Dynamics as a Potential Therapeutic Target. Front Cell Dev Biol 2021; 9:673839. [PMID: 34307357 PMCID: PMC8293691 DOI: 10.3389/fcell.2021.673839] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 06/16/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease (CVD) is the main cause of death worldwide. Atherosclerosis is the underlying pathological basis of CVD. Mitochondrial homeostasis is maintained through the dynamic processes of fusion and fission. Mitochondria are involved in many cellular processes, such as steroid biosynthesis, calcium homeostasis, immune cell activation, redox signaling, apoptosis, and inflammation, among others. Under stress conditions, mitochondrial dynamics, mitochondrial cristae remodeling, and mitochondrial ROS (mitoROS) production increase, mitochondrial membrane potential (MMP) decreases, calcium homeostasis is imbalanced, and mitochondrial permeability transition pore open (mPTP) and release of mitochondrial DNA (mtDNA) are activated. mtDNA recognized by TLR9 can lead to NF-κB pathway activation and pro-inflammatory factor expression. At the same time, TLR9 can also activate NLRP3 inflammasomes and release interleukin, an event that eventually leads to tissue damage and inflammatory responses. In addition, mitochondrial dysfunction may amplify the activation of NLRP3 through the production of mitochondrial ROS, which together aggravate accumulating mitochondrial damage. In addition, mtDNA defects or gene mutation can lead to mitochondrial oxidative stress. Finally, obesity, diabetes, hypertension and aging are risk factors for the progression of CVD, which are closely related to mitochondrial dynamics. Mitochondrial dynamics may represent a new target in the treatment of atherosclerosis. Antioxidants, mitochondrial inhibitors, and various new therapies to correct mitochondrial dysfunction represent a few directions for future research on therapeutic intervention and amelioration of atherosclerosis.
Collapse
Affiliation(s)
- Dan Li
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shengjie Yang
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanwei Xing
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Limin Pan
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ran Zhao
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yixi Zhao
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Longtao Liu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Min Wu
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
11
|
Lopez-Crisosto C, Arias-Carrasco R, Sepulveda P, Garrido-Olivares L, Maracaja-Coutinho V, Verdejo HE, Castro PF, Lavandero S. Novel molecular insights and public omics data in pulmonary hypertension. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166200. [PMID: 34144090 DOI: 10.1016/j.bbadis.2021.166200] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 06/02/2021] [Accepted: 06/07/2021] [Indexed: 12/21/2022]
Abstract
Pulmonary hypertension is a rare disease with high morbidity and mortality which mainly affects women of reproductive age. Despite recent advances in understanding the pathogenesis of pulmonary hypertension, the high heterogeneity in the presentation of the disease among different patients makes it difficult to make an accurate diagnosis and to apply this knowledge to effective treatments. Therefore, new studies are required to focus on translational and personalized medicine to overcome the lack of specificity and efficacy of current management. Here, we review the majority of public databases storing 'omics' data of pulmonary hypertension studies, from animal models to human patients. Moreover, we review some of the new molecular mechanisms involved in the pathogenesis of pulmonary hypertension, including non-coding RNAs and the application of 'omics' data to understand this pathology, hoping that these new approaches will provide insights to guide the way to personalized diagnosis and treatment.
Collapse
Affiliation(s)
- Camila Lopez-Crisosto
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago 8380492, Chile; Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8380492, Chile
| | - Raul Arias-Carrasco
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago 8380492, Chile
| | - Pablo Sepulveda
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8380492, Chile; Division of Cardiovascular Diseases, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Luis Garrido-Olivares
- Cardiovascular Surgery, Division of Surgery, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Vinicius Maracaja-Coutinho
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago 8380492, Chile
| | - Hugo E Verdejo
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8380492, Chile; Division of Cardiovascular Diseases, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo F Castro
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8380492, Chile; Division of Cardiovascular Diseases, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago 8380492, Chile; Department of Internal Medicine, Cardiology Division, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA.
| |
Collapse
|
12
|
Wu P, Xie X, Chen M, Sun J, Cai L, Wei J, Yang L, Huang X, Wang L. Elucidation of the Mechanisms and Molecular Targets of Qishen Yiqi Formula for the Treatment of Pulmonary Arterial Hypertension using a Bioinformatics/Network Topology-based Strategy. Comb Chem High Throughput Screen 2021; 24:701-715. [PMID: 33076804 DOI: 10.2174/1386207323666201019145354] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/06/2020] [Accepted: 09/12/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND AND OBJECTIVE Qishen Yiqi formula (QSYQ) is used to treat cardiovascular disease in the clinical practice of traditional Chinese medicine. However, few studies have explored whether QSYQ affects pulmonary arterial hypertension (PAH), and the mechanisms of action and molecular targets of QSYQ for the treatment of PAH are unclear. A bioinformatics/network topology-based strategy was used to identify the bioactive ingredients, putative targets, and molecular mechanisms of QSYQ in PAH. METHODS A network pharmacology-based strategy was employed by integrating active component gathering, target prediction, PAH gene collection, network topology, and gene enrichment analysis to systematically explore the multicomponent synergistic mechanisms. RESULTS In total, 107 bioactive ingredients of QSYQ and 228 ingredient targets were identified. Moreover, 234 PAH-related differentially expressed genes with a |fold change| >2 and an adjusted P value < 0.005 were identified between the PAH patient and control groups, and 266 therapeutic targets were identified. The pathway enrichment analysis indicated that 85 pathways, including the PI3K-Akt, MAPK, and HIF-1 signaling pathways, were significantly enriched. TP53 was the core target gene, and 7 other top genes (MAPK1, RELA, NFKB1, CDKN1A, AKT1, MYC, and MDM2) were the key genes in the gene-pathway network based on the effects of QSYQ on PAH. CONCLUSION An integrative investigation based on network pharmacology may elucidate the multicomponent synergistic mechanisms of QSYQ in PAH and lay a foundation for further animal experiments, human clinical trials and rational clinical applications of QSYQ.
Collapse
Affiliation(s)
- Peiliang Wu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Xiaona Xie
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Mayun Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Junwei Sun
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Luqiong Cai
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Jinqiu Wei
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Lin Yang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Xiaoying Huang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Liangxing Wang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| |
Collapse
|
13
|
Xin Y, Li J, Wu W, Liu X. Mitofusin-2: A New Mediator of Pathological Cell Proliferation. Front Cell Dev Biol 2021; 9:647631. [PMID: 33869201 PMCID: PMC8049505 DOI: 10.3389/fcell.2021.647631] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/02/2021] [Indexed: 02/05/2023] Open
Abstract
Cell proliferation is an important cellular process for physiological tissue homeostasis and remodeling. The mechanisms of cell proliferation in response to pathological stresses are not fully understood. Mitochondria are highly dynamic organelles whose shape, number, and biological functions are modulated by mitochondrial dynamics, including fusion and fission. Mitofusin-2 (Mfn-2) is an essential GTPase-related mitochondrial dynamics protein for maintaining mitochondrial network and bioenergetics. A growing body of evidence indicates that Mfn-2 has a potential role in regulating cell proliferation in various cell types. Here we review these new functions of Mfn-2, highlighting its crucial role in several signaling pathways during the process of pathological cell proliferation. We conclude that Mfn-2 could be a new mediator of pathological cell proliferation and a potential therapeutic target.
Collapse
Affiliation(s)
- Yanguo Xin
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| | - Junli Li
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Wenchao Wu
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaojing Liu
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China.,Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
14
|
Hu Z, Song Q, Ma H, Guo Y, Zhang T, Xie H, Luo X. TRIM32 inhibits the proliferation and migration of pulmonary artery smooth muscle cells through the inactivation of PI3K/Akt pathway in pulmonary arterial hypertension. J Bioenerg Biomembr 2021; 53:309-320. [PMID: 33694017 DOI: 10.1007/s10863-021-09880-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 02/07/2021] [Indexed: 01/27/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive and fetal cardiovascular disease. Tripartite motif 32 (TRIM32) is a member of TRIM family that has been found to be involved in cardiovascular disease. However, the role of TRIM32 in PAH remains unclear. Here we investigated the effects of TRIM32 on hypoxia-induced pulmonary artery smooth muscle cells (PASMCs) in vitro. Our results showed that TRIM32 protein level in the plasma samples from PAH patients was decreased as compared with healthy volunteers. Exposure to hypoxia condition caused a significant decrease in TRIM32 expression in PASMCs. Overexpression of TRIM32 inhibited hypoxia-induced proliferation and migration of PASMCs. TRIM32 overexpression elevated the increased apoptotic rate and caspase-3 activity in hypoxia-induced PASMCs. Moreover, overexpression of TRIM32 reversed hypoxia-induced down-regulation of myocardin, SM 22 and calponin, as well as up-regulation of osteopontin (OPN). Whereas, TRIM32 knockdown shwed the opposite effect. Furthermore, overexpression of TRIM32 inhibited hypoxia-induced activation of PI3K/Akt with decreased phosphorylated level of PI3K and Akt. Additionally, activation of PI3K/Akt by IGF-1 treatment reversed the effects of TRIM32 on hypoxia-induced PASMCs. In conclusion, these findings indicated that TRIM32 was involved in the development of PAH through regulating the proliferation, migration, apoptosis and dedifferentiation of PASMCs, which might be mediated by the PI3K/Akt signaling pathway. Thus, TRIM32 might be a potential target for PAH treatment.
Collapse
Affiliation(s)
- Zhi Hu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 West Yanta Road, Xi'an, Shaanxi, 710061, China.
| | - Qiang Song
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Hui Ma
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Yaozhang Guo
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Tingting Zhang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Hang Xie
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Xiaohui Luo
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 West Yanta Road, Xi'an, Shaanxi, 710061, China
| |
Collapse
|
15
|
Zhao H, Lin J, Sieck G, Haddad GG. Neuroprotective Role of Akt in Hypoxia Adaptation in Andeans. Front Neurosci 2021; 14:607711. [PMID: 33519361 PMCID: PMC7843528 DOI: 10.3389/fnins.2020.607711] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/11/2020] [Indexed: 11/13/2022] Open
Abstract
Chronic mountain sickness (CMS) is a disease that potentially threatens a large segment of high-altitude populations during extended living at altitudes above 2,500 m. Patients with CMS suffer from severe hypoxemia, excessive erythrocytosis and neurologic deficits. The cellular mechanisms underlying CMS neuropathology remain unknown. We previously showed that iPSC-derived CMS neurons have altered mitochondrial dynamics and increased susceptibility to hypoxia-induced cell death. Genome analysis from the same population identified many ER stress-related genes that play an important role in hypoxia adaptation or lack thereof. In the current study, we showed that iPSC-derived CMS neurons have increased expression of ER stress markers Grp78 and XBP1s under normoxia and hyperphosphorylation of PERK under hypoxia, alleviating ER stress does not rescue the hypoxia-induced CMS neuronal cell death. Akt is a cytosolic regulator of ER stress with PERK as a direct target of Akt. CMS neurons exhibited lack of Akt activation and lack of increased Parkin expression as compared to non-CMS neurons under hypoxia. By enhancing Akt activation and Parkin overexpression, hypoxia-induced CMS neuronal cell death was reduced. Taken together, we propose that increased Akt activation protects non-CMS from hypoxia-induced cell death. In contrast, impaired adaptive mechanisms including failure to activate Akt and increase Parkin expression render CMS neurons more susceptible to hypoxia-induced cell death.
Collapse
Affiliation(s)
- Helen Zhao
- Department of Pediatrics (Respiratory Medicine), University of California, San Diego, La Jolla, CA, United States
| | - Jonathan Lin
- Department of Pathology, University of California, San Diego, La Jolla, CA, United States
- Department of Pathology, Stanford University, Stanford, CA, United States
- VA Palo Alto Healthcare System, Palo Alto, CA, United States
| | - Gary Sieck
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| | - Gabriel G. Haddad
- Department of Pediatrics (Respiratory Medicine), University of California, San Diego, La Jolla, CA, United States
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States
- The Rady Children’s Hospital, San Diego, CA, United States
| |
Collapse
|
16
|
Zhang S, Wang J, Qi X, Tao X, Xie W, Wan J, Shen YH, Zhai Z. Plasminogen activator Inhibitor-2 inhibits pulmonary arterial smooth muscle cell proliferation in pulmonary arterial hypertension via PI3K/Akt and ERK signaling. Exp Cell Res 2020; 398:112392. [PMID: 33227315 DOI: 10.1016/j.yexcr.2020.112392] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 11/09/2020] [Accepted: 11/19/2020] [Indexed: 12/25/2022]
Abstract
BACKGROUND The proliferation of pulmonary arterial smooth muscle cells (PASMCs) and subsequent pulmonary vascular remodeling leads to pulmonary arterial hypertension (PAH). Understanding the underlying mechanisms and identifying molecules that can suppress PASMCs proliferation is critical for developing effective pharmacological treatment. We previously showed that plasminogen activator inhibitor-2 (PAI-2) inhibited human PASMC (hPASMCs) proliferation in vitro. However, its inhibitory effect on PAH remains to be determined, and the mechanism remains to be illustrated. METHODS We compared serum PAI-2 levels between PAH patients and healthy controls, and examined the correlation between PAI-2 level and disease severity. In monocrotaline-induced PAH rats, we examined the effects of exogenous PAI-2 administration on pulmonary vascular remodeling and PAH development. The effect of PAI-2 and potential mechanisms was further examined in cultured hPASMCs. RESULTS The serum PAI-2 was decreased in PAH patients compared with controls. PAI-2 level was negatively correlated with mean pulmonary arterial pressure and estimated systolic pulmonary arterial pressure in ultrasonic cardiogram, while positively correlated with 6-min walking distance. In rats, administration of exogenous PAI-2 significantly reversed monocrotaline-induced PAH, as indicated by the decrease in right ventricle systolic pressure, right ventricular hypertrophy index and percent media thickness of pulmonary arterioles. Further mechanistic investigation in hPASMCs showed that PAI-2 inhibited cell proliferation by preventing the activation of PI3K/Akt and ERK pathways. CONCLUSION PAI-2 is downregulated in PAH patients. PAI-2 attenuates PAH development by suppressing hPASMCs proliferation via the inhibition of PI3K/Akt and ERK pathways. PAI-2 may serve as a potential biomarker and therapeutic target for PAH.
Collapse
Affiliation(s)
- Shuai Zhang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China; National Clinical Research Center for Respiratory Diseases, Beijing, China
| | - Jing Wang
- Department of Pulmonary and Critical Care Medicine, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Xianmei Qi
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China; Department of Respiratory Medicine, Capital Medical University, Beijing, China
| | - Xincao Tao
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China; National Clinical Research Center for Respiratory Diseases, Beijing, China
| | - Wanmu Xie
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China; National Clinical Research Center for Respiratory Diseases, Beijing, China
| | - Jun Wan
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China; National Clinical Research Center for Respiratory Diseases, Beijing, China
| | - Ying H Shen
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA; Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA; Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX, USA
| | - Zhenguo Zhai
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China; National Clinical Research Center for Respiratory Diseases, Beijing, China.
| |
Collapse
|
17
|
Huang S, Yue Y, Feng K, Huang X, Li H, Hou J, Yang S, Huang S, Liang M, Chen G, Wu Z. Conditioned medium from M2b macrophages modulates the proliferation, migration, and apoptosis of pulmonary artery smooth muscle cells by deregulating the PI3K/Akt/FoxO3a pathway. PeerJ 2020; 8:e9110. [PMID: 32411539 PMCID: PMC7207208 DOI: 10.7717/peerj.9110] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 04/10/2020] [Indexed: 01/20/2023] Open
Abstract
Background Immunity and inflammation are considered to be central features of pulmonary artery hypertension (PAH), in which macrophages are one of the main components of inflammatory cell infiltration around the pulmonary artery. M2b macrophages, which are different from M1 and M2 macrophages, are believed to have immunomodulatory activities and produce little fibrosis. The purpose of this study was to explore the effect of M2b macrophages on pulmonary artery smooth muscle cells (PASMCs) derived from monocrotaline-induced PAH rats. Methods PASMCs were cultured in serum-free medium, the supernatant of M0 macrophages, or the supernatant of M2b macrophages for 24 hours. Then cell proliferation was assessed by cell counting kit-8 and cell migration ability was detected by wound healing and transwell assays. The apoptosis rate of cells was determined by TUNEL staining and annexin V-PE/7-ADD staining. Western blot was used to detect the expression of Bcl-2 family proteins, cleaved caspase-9 and PI3K/Akt/FoxO3a pathway. LY294002 (a specific inhibitor of PI3K) was used to investigate its effect on PASMCs and its relationship with M2b macrophages. Results Conditioned medium from M2b macrophages significantly inhibited the proliferation and migration of PASMCs compared with the control group and M0 macrophage group. Furthermore, conditioned medium from M2b macrophages promote PASMC apoptosis and increased the expression of pro-apoptotic proteins Bax and cleaved caspase-9, inhibited the expression of anti-apoptotic proteins Bcl-2 and Bcl-xl. Finally, conditioned medium from M2b macrophages inhibited the PI3K/Akt/FoxO3a pathway. Inhibition of PI3K/Akt/FoxO3a pathway also significantly inhibit the proliferation, migration, and apoptosis resistance of PASMCs. Conclusion Conditioned medium from M2b macrophages can inhibit the proliferation, migration, and apoptosis resistance of PASMCs, which may be at least partially by deregulating the PI3K/Akt/FoxO3a pathway.
Collapse
Affiliation(s)
- Suiqing Huang
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Yuan Yue
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Kangni Feng
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaolin Huang
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Huayang Li
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Jian Hou
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Song Yang
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China.,Department of Cardiosurgery Intensive Care Unit, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shaojie Huang
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Mengya Liang
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Guangxian Chen
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhongkai Wu
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
18
|
Effects of Dietary Supplementation of Lauric Acid on Lactation Function, Mammary Gland Development, and Serum Lipid Metabolites in Lactating Mice. Animals (Basel) 2020; 10:ani10030529. [PMID: 32235692 PMCID: PMC7143820 DOI: 10.3390/ani10030529] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/17/2020] [Accepted: 03/19/2020] [Indexed: 12/17/2022] Open
Abstract
Simple Summary Milk secreted from mammary glands is an important nutrition source for offspring after parturition. Mammary gland development and lactation ability have important effects on the growth and health of the offspring. Many studies have demonstrated that external factors, including the environment and nutrition influence the development of mammary glands. Lauric acid is a fatty acid that has many nutritional and physiological properties. In this study, we investigated the effects of dietary supplementation of lauric acid on lactation function and mammary gland development in lactating mice. We found that dietary supplementation of lauric acid during lactation might enhance the mammary development to promote the lactation function of mice. Through the study of mice, we hoped that the results could be applied to animal feed development and animal breeding production. Abstract Our previous studies demonstrated that lauric acid (LA) stimulated mammary gland development during puberty. However, the roles of LA on lactation in mice remain indeterminate. Thus, the aim of this study was to investigate the effects of dietary LA supplementation on lactation functioning and to study the potential mechanisms during lactation. in vivo, there was no effect of 1% LA dietary supplementation during lactation on the feed intake or body weight of breast-feeding mice. However, maternal LA supplementation significantly expanded the number of mammary gland alveoli of mice during lactation and the average body weight of the offspring, suggesting that LA supplementation enhanced the development and lactation function of the mammary glands. in vitro, 100 μM of LA significantly increased the content of triglycerides (TG) in the cell supernatant of induced HC11 cells, however, with no effect on the expression of the genes associated with fatty acid synthesis. LA also activated the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway. LA dietary supplementation significantly expanded the serum levels of lipid metabolites, including sphingomyelin and other metabolites with the sn-2 position of C12 and sn-1 position of C18 in the TG of the lactating mice. Taken together, dietary supplementation of LA during lactation could promote the lactation function of mice, which might be related to increasing the development of the mammary glands and alternation of serum lipid metabolites. These findings provided more theoretical and experimental basis for the application of lauric acid in the development of mammary glands and lactation function of lactating animals.
Collapse
|
19
|
Dasgupta A, Wu D, Tian L, Xiong PY, Dunham-Snary KJ, Chen KH, Alizadeh E, Motamed M, Potus F, Hindmarch CCT, Archer SL. Mitochondria in the Pulmonary Vasculature in Health and Disease: Oxygen-Sensing, Metabolism, and Dynamics. Compr Physiol 2020; 10:713-765. [PMID: 32163206 DOI: 10.1002/cphy.c190027] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In lung vascular cells, mitochondria serve a canonical metabolic role, governing energy homeostasis. In addition, mitochondria exist in dynamic networks, which serve noncanonical functions, including regulation of redox signaling, cell cycle, apoptosis, and mitochondrial quality control. Mitochondria in pulmonary artery smooth muscle cells (PASMC) are oxygen sensors and initiate hypoxic pulmonary vasoconstriction. Acquired dysfunction of mitochondrial metabolism and dynamics contribute to a cancer-like phenotype in pulmonary arterial hypertension (PAH). Acquired mitochondrial abnormalities, such as increased pyruvate dehydrogenase kinase (PDK) and pyruvate kinase muscle isoform 2 (PKM2) expression, which increase uncoupled glycolysis (the Warburg phenomenon), are implicated in PAH. Warburg metabolism sustains energy homeostasis by the inhibition of oxidative metabolism that reduces mitochondrial apoptosis, allowing unchecked cell accumulation. Warburg metabolism is initiated by the induction of a pseudohypoxic state, in which DNA methyltransferase (DNMT)-mediated changes in redox signaling cause normoxic activation of HIF-1α and increase PDK expression. Furthermore, mitochondrial division is coordinated with nuclear division through a process called mitotic fission. Increased mitotic fission in PAH, driven by increased fission and reduced fusion favors rapid cell cycle progression and apoptosis resistance. Downregulation of the mitochondrial calcium uniporter complex (MCUC) occurs in PAH and is one potential unifying mechanism linking Warburg metabolism and mitochondrial fission. Mitochondrial metabolic and dynamic disorders combine to promote the hyperproliferative, apoptosis-resistant, phenotype in PAH PASMC, endothelial cells, and fibroblasts. Understanding the molecular mechanism regulating mitochondrial metabolism and dynamics has permitted identification of new biomarkers, nuclear and CT imaging modalities, and new therapeutic targets for PAH. © 2020 American Physiological Society. Compr Physiol 10:713-765, 2020.
Collapse
Affiliation(s)
- Asish Dasgupta
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Danchen Wu
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Lian Tian
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Ping Yu Xiong
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | | | - Kuang-Hueih Chen
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Elahe Alizadeh
- Department of Medicine, Queen's Cardiopulmonary Unit (QCPU), Translational Institute of Medicine (TIME), Queen's University, Kingston, Ontario, Canada
| | - Mehras Motamed
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - François Potus
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Charles C T Hindmarch
- Department of Medicine, Queen's Cardiopulmonary Unit (QCPU), Translational Institute of Medicine (TIME), Queen's University, Kingston, Ontario, Canada
| | - Stephen L Archer
- Department of Medicine, Queen's University, Kingston, Ontario, Canada.,Kingston Health Sciences Centre, Kingston, Ontario, Canada.,Providence Care Hospital, Kingston, Ontario, Canada
| |
Collapse
|
20
|
Kim S, Song J, Ernst P, Latimer MN, Ha CM, Goh KY, Ma W, Rajasekaran NS, Zhang J, Liu X, Prabhu SD, Qin G, Wende AR, Young ME, Zhou L. MitoQ regulates redox-related noncoding RNAs to preserve mitochondrial network integrity in pressure-overload heart failure. Am J Physiol Heart Circ Physiol 2020; 318:H682-H695. [PMID: 32004065 PMCID: PMC7099446 DOI: 10.1152/ajpheart.00617.2019] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 01/29/2020] [Accepted: 01/29/2020] [Indexed: 01/04/2023]
Abstract
Evidence suggests that mitochondrial network integrity is impaired in cardiomyocytes from failing hearts. While oxidative stress has been implicated in heart failure (HF)-associated mitochondrial remodeling, the effect of mitochondrial-targeted antioxidants, such as mitoquinone (MitoQ), on the mitochondrial network in a model of HF (e.g., pressure overload) has not been demonstrated. Furthermore, the mechanism of this regulation is not completely understood with an emerging role for posttranscriptional regulation via long noncoding RNAs (lncRNAs). We hypothesized that MitoQ preserves mitochondrial fusion proteins (i.e., mitofusin), likely through redox-sensitive lncRNAs, leading to improved mitochondrial network integrity in failing hearts. To test this hypothesis, 8-wk-old C57BL/6J mice were subjected to ascending aortic constriction (AAC), which caused substantial left ventricular (LV) chamber remodeling and remarkable contractile dysfunction in 1 wk. Transmission electron microscopy and immunostaining revealed defective intermitochondrial and mitochondrial-sarcoplasmic reticulum ultrastructure in AAC mice compared with sham-operated animals, which was accompanied by elevated oxidative stress and suppressed mitofusin (i.e., Mfn1 and Mfn2) expression. MitoQ (1.36 mg·day-1·mouse-1, 7 consecutive days) significantly ameliorated LV dysfunction, attenuated Mfn2 downregulation, improved interorganellar contact, and increased metabolism-related gene expression. Moreover, our data revealed that MitoQ alleviated the dysregulation of an Mfn2-associated lncRNA (i.e., Plscr4). In summary, the present study supports a unique mechanism by which MitoQ improves myocardial intermitochondrial and mitochondrial-sarcoplasmic reticulum (SR) ultrastructural remodeling in HF by maintaining Mfn2 expression via regulation by an lncRNA. These findings underscore the important role of lncRNAs in the pathogenesis of HF and the potential of targeting them for effective HF treatment.NEW & NOTEWORTHY We have shown that MitoQ improves cardiac mitochondrial network integrity and mitochondrial-SR alignment in a pressure-overload mouse heart-failure model. This may be occurring partly through preventing the dysregulation of a redox-sensitive lncRNA-microRNA pair (i.e., Plscr4-miR-214) that results in an increase in mitofusin-2 expression.
Collapse
Affiliation(s)
- Seulhee Kim
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jiajia Song
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Patrick Ernst
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| | - Mary N Latimer
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Chae-Myeong Ha
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Kah Yong Goh
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Wenxia Ma
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| | | | - Jianhua Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Xiaoguang Liu
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| | - Sumanth D Prabhu
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Gangjian Qin
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| | - Adam R Wende
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Martin E Young
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Lufang Zhou
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
21
|
Durgin BG, Straub AC. Redox control of vascular smooth muscle cell function and plasticity. J Transl Med 2018; 98:1254-1262. [PMID: 29463879 PMCID: PMC6102093 DOI: 10.1038/s41374-018-0032-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/15/2017] [Accepted: 12/18/2017] [Indexed: 02/07/2023] Open
Abstract
Vascular smooth muscle cells (SMC) play a major role in vascular diseases, such as atherosclerosis and hypertension. It has long been established in vitro that contractile SMC can phenotypically switch to function as proliferative and/or migratory cells in response to stimulation by oxidative stress, growth factors, and inflammatory cytokines. Reactive oxygen species (ROS) are oxidative stressors implicated in driving vascular diseases, shifting cell bioenergetics, and increasing SMC proliferation, migration, and apoptosis. In this review, we summarize our current knowledge of how disruptions to redox balance can functionally change SMC and how this may influence vascular disease pathogenesis. Specifically, we focus on our current understanding of the role of vascular nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOX) 1, 4, and 5 in SMC function. We also review the evidence implicating mitochondrial fission in SMC phenotypic transitions and mitochondrial fusion in maintenance of SMC homeostasis. Finally, we discuss the importance of the redox regulation of the soluble guanylate cyclase (sGC)-cyclic guanosine monophosphate (cGMP)-protein kinase G (PKG) pathway as a potential oxidative and therapeutic target for regulating SMC function.
Collapse
Affiliation(s)
- Brittany G Durgin
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Adam C Straub
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
22
|
Burgos-Ramos E, Canelles S, Rodríguez A, Frago LM, Gómez-Ambrosi J, Chowen JA, Frühbeck G, Argente J, Barrios V. The increase in fiber size in male rat gastrocnemius after chronic central leptin infusion is related to activation of insulin signaling. Mol Cell Endocrinol 2018; 470:48-59. [PMID: 28962893 DOI: 10.1016/j.mce.2017.09.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 09/18/2017] [Accepted: 09/25/2017] [Indexed: 01/20/2023]
Abstract
Insulin potentiates leptin effects on muscle accrual and glucose homeostasis. However, the relationship between leptin's central effects on peripheral insulin sensitivity and the associated structural changes remain unclear. We hypothesized that central leptin infusion modifies muscle size through activation of insulin signaling. Muscle insulin signaling, enzymes of fatty acid metabolism, mitochondrial respiratory chain complexes, proliferating cell nuclear antigen (PCNA) and fiber area were analyzed in the gastrocnemius of chronic central infused (L), pair-fed (PF) and control rats. PCNA-positive nuclei, fiber area, GLUT4 and glycogen levels and activation of Akt and mechanistic target of rapamycin were increased in L, with no changes in PF. Acetyl-CoA carboxylase-β mRNA levels and non-esterified fatty acid and triglyceride content were reduced and carnitine palmitoyltransferase-1b expression and mitochondrial complexes augmented in L. These results suggest that leptin promotes an increase in muscle size associated with improved insulin signaling favored by lipid profile.
Collapse
Affiliation(s)
- Emma Burgos-Ramos
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, E-28009, Madrid, Spain; Área de Bioquímica, Facultad de Ciencias Ambientales y Bioquímica, Universidad Castilla-La Mancha, E-45071, Toledo, Spain
| | - Sandra Canelles
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, E-28009, Madrid, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28009, Madrid, Spain
| | - Amaia Rodríguez
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28009, Madrid, Spain; Metabolic Research Laboratory, Clínica Universidad de Navarra, E-31008, Pamplona, Spain
| | - Laura M Frago
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, E-28009, Madrid, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28009, Madrid, Spain; Department of Pediatrics, Universidad Autónoma de Madrid, E-28009, Madrid, Spain
| | - Javier Gómez-Ambrosi
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28009, Madrid, Spain; Metabolic Research Laboratory, Clínica Universidad de Navarra, E-31008, Pamplona, Spain
| | - Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, E-28009, Madrid, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28009, Madrid, Spain
| | - Gema Frühbeck
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28009, Madrid, Spain; Metabolic Research Laboratory, Clínica Universidad de Navarra, E-31008, Pamplona, Spain
| | - Jesús Argente
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, E-28009, Madrid, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28009, Madrid, Spain; Department of Pediatrics, Universidad Autónoma de Madrid, E-28009, Madrid, Spain; IMDEA Food Institute, CEI UAM + CSIC, E-28049, Madrid, Spain
| | - Vicente Barrios
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, E-28009, Madrid, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28009, Madrid, Spain.
| |
Collapse
|
23
|
Zhang Q, Cao Y, Luo Q, Wang P, Shi P, Song C, E M, Ren J, Fu B, Sun H. The transient receptor potential vanilloid-3 regulates hypoxia-mediated pulmonary artery smooth muscle cells proliferation via PI3K/AKT signaling pathway. Cell Prolif 2018; 51:e12436. [PMID: 29359496 DOI: 10.1111/cpr.12436] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 12/01/2017] [Indexed: 01/01/2023] Open
Abstract
OBJECTVES Transient receptor potential vanilloid 3 (TRPV3) is a member of the TRP channels family of Ca2+ -permeant cation channels. In this study, we aim to investigate the role of TRPV3 in pulmonary vascular remodeling and PASMCs proliferation under hypoxia. MATERIALS AND METHODS The expression of TRPV3 was evaluated in patients with pulmonary arterial hypertension (PAH) and hypoxic rats, using hematoxylin and eosin (H&E) and immunohistochemistry. In vitro, MTT assay, flow cytometry, Western blotting and immunofluorescence were performed to investigate the effects of TRPV3 on proliferation of PASMCs. RESULTS We found that, in vivo, the expression of TRPV3 was increased in patients with PAH and hypoxic rats. Right ventricular hypertrophy measurements and pulmonary pathomorphology data show that the ratio of the heart weight/tibia length (HW/TL), the right ventricle/left ventricle plus septum (RV/LV+S) and the medial width of the pulmonary artery were increased in chronic hypoxic rats. Moreover, the expression of proliferating cell nuclear antigen (PCNA), Cyclin D, Cyclin E and Cyclin A, phospho-CaMKII (p-CaMKII) were induced by hypoxia. In vitro, we revealed that hypoxia promoted PASMCs viability, increased the expression of PCNA, Cyclin D, Cyclin E, Cyclin A p-CaMKII, made more cells from G0 /G1 phase to G2 /M + S phase, enhanced the microtubule formation, and increased [Ca2+ ]i , which could be suppressed by Ruthenium Red, an inhibitor of TRPV3, and TRPV3 silencing has similar effects. Furthermore, the up-regulated expression of PCNA, Cyclin D, Cyclin E and Cyclin A, the increased number of cells in G2 /M and S phase, and the enhanced activation and expression of PI3K and AKT proteins induced by hypoxia and in presence of carvacrol (an agonist of TRPV3), was significantly attenuated by incubation of LY 294002, a specific inhibitor for PI3K/AKT. CONCLUSIONS These findings suggest that TRPV3 is involved in hypoxia-induced pulmonary vascular remodeling and promotes proliferation of PASMCs and the effect is, at least in part, mediated via the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Qianlong Zhang
- Department of Physiology, Harbin Medical University-Daqing, Daqing, China
| | - Yonggang Cao
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, China
| | - Qian Luo
- Department of Physiology, Harbin Medical University-Daqing, Daqing, China
| | - Peng Wang
- Department of Physiology, Harbin Medical University-Daqing, Daqing, China
| | - Pilong Shi
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, China
| | - Chao Song
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, China
| | - Mingyao E
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, China
| | - Jing Ren
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, China
| | - Bowen Fu
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, China
| | - Hongli Sun
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, China
| |
Collapse
|
24
|
Epigallocatechin-3-gallate ameliorates hypoxia-induced pulmonary vascular remodeling by promoting mitofusin-2-mediated mitochondrial fusion. Eur J Pharmacol 2017; 809:42-51. [DOI: 10.1016/j.ejphar.2017.05.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 05/01/2017] [Accepted: 05/02/2017] [Indexed: 01/21/2023]
|
25
|
Jie Z, Baoqin W, Changan L, Xiangli T, Zegeng L. Qibai Pingfei capsule medicated serum inhibits the proliferation of hypoxia-induced pulmonary arterial smooth muscle cells via the Ca 2+ /calcineurin/nuclear factor of activated T-cells 3 pathway. J TRADIT CHIN MED 2017. [DOI: 10.1016/s0254-6272(17)30153-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
26
|
MiR-125a regulates mitochondrial homeostasis through targeting mitofusin 1 to control hypoxic pulmonary vascular remodeling. J Mol Med (Berl) 2017; 95:977-993. [PMID: 28593577 DOI: 10.1007/s00109-017-1541-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 03/14/2017] [Accepted: 04/28/2017] [Indexed: 10/19/2022]
Abstract
Abnormal pulmonary arterial smooth muscle cells (PASMCs) proliferation is an important pathological process in hypoxic pulmonary arterial hypertension. Mitochondrial dynamics and quality control have a central role in the maintenance of the cell proliferation-apoptosis balance. However, the molecular mechanism is still unknown. We used hypoxic animal models, cell biology, and molecular biology to determine the effect of mitofusin 1 (Mfn1) on hypoxia-mediated PASMCs mitochondrial homeostasis. We found that Mfn1 expression was increased in hypoxia, which was crucial for hypoxia-induced mitochondrial dysfunction and smooth muscle cell proliferation as well as hypoxia-stimulated cell-cycle transition from the G0/G1 phase to S phase. Subsequently, we studied the role of microRNAs in mitochondrial function associated with PASMC proliferation under hypoxic conditions. The promotive effect of Mfn1 on pulmonary vascular remodeling was alleviated in the presence of miR-125a agomir, and miR-125a antagomir mimicked the hypoxic damage effects to mitochondrial homeostasis. Moreover, in vivo and in vitro treatment with miR-125a agomir protected the pulmonary vessels from mitochondrial dysfunction and abnormal remodeling. In the present study, we determined that mitochondrial homeostasis, particularly Mfn1, played an important role in PASMCs proliferation. MiR-125a, an important underlying factor, which inhibited Mfn1 expression and decreased PASMCs disordered growth during hypoxia. These results provide a theoretical basis for the prevention and treatment of pulmonary vascular remodeling. KEY MESSAGES Hypoxia leads to upregulation of mitofusin 1 (Mfn1) both in vivo and in vitro. Mfn1 is involved in hypoxia-induced PASMCs proliferation. Mfn1-mediated mitochondrial homeostasis is regulated by miR-125a. MiR-125a plays a role in PASMCs oxidative phosphorylation and glycolysis.
Collapse
|
27
|
Ong SB, Kalkhoran SB, Hernández-Reséndiz S, Samangouei P, Ong SG, Hausenloy DJ. Mitochondrial-Shaping Proteins in Cardiac Health and Disease - the Long and the Short of It! Cardiovasc Drugs Ther 2017; 31:87-107. [PMID: 28190190 PMCID: PMC5346600 DOI: 10.1007/s10557-016-6710-1] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mitochondrial health is critically dependent on the ability of mitochondria to undergo changes in mitochondrial morphology, a process which is regulated by mitochondrial shaping proteins. Mitochondria undergo fission to generate fragmented discrete organelles, a process which is mediated by the mitochondrial fission proteins (Drp1, hFIS1, Mff and MiD49/51), and is required for cell division, and to remove damaged mitochondria by mitophagy. Mitochondria undergo fusion to form elongated interconnected networks, a process which is orchestrated by the mitochondrial fusion proteins (Mfn1, Mfn2 and OPA1), and which enables the replenishment of damaged mitochondrial DNA. In the adult heart, mitochondria are relatively static, are constrained in their movement, and are characteristically arranged into 3 distinct subpopulations based on their locality and function (subsarcolemmal, myofibrillar, and perinuclear). Although the mitochondria are arranged differently, emerging data supports a role for the mitochondrial shaping proteins in cardiac health and disease. Interestingly, in the adult heart, it appears that the pleiotropic effects of the mitochondrial fusion proteins, Mfn2 (endoplasmic reticulum-tethering, mitophagy) and OPA1 (cristae remodeling, regulation of apoptosis, and energy production) may play more important roles than their pro-fusion effects. In this review article, we provide an overview of the mitochondrial fusion and fission proteins in the adult heart, and highlight their roles as novel therapeutic targets for treating cardiac disease.
Collapse
Affiliation(s)
- Sang-Bing Ong
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore, 169857, Singapore.,National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
| | - Siavash Beikoghli Kalkhoran
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, London, UK
| | - Sauri Hernández-Reséndiz
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore, 169857, Singapore.,National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
| | - Parisa Samangouei
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, London, UK
| | - Sang-Ging Ong
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Derek John Hausenloy
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore, 169857, Singapore. .,National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore. .,The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, London, UK. .,The National Institute of Health Research, University College London Hospitals Biomedical Research Centre, London, UK.
| |
Collapse
|
28
|
Meng Y, Zhang J, Zhang F, Ai W, Zhu X, Shu G, Wang L, Gao P, Xi Q, Zhang Y, Liang X, Jiang Q, Wang S. Lauric Acid Stimulates Mammary Gland Development of Pubertal Mice through Activation of GPR84 and PI3K/Akt Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:95-103. [PMID: 27978622 DOI: 10.1021/acs.jafc.6b04878] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
It has been demonstrated that dietary fat affects pubertal mammary gland development. However, the role of lauric acid (LA) in this process remains unclear. Thus, this study aimed to investigate the effects of LA on mammary gland development in pubertal mice and to explore the underlying mechanism. In vitro, 100 μM LA significantly promoted proliferation of mouse mammary epithelial cell line HC11 by regulating expression of proliferative markers (cyclin D1/3, p21, PCNA). Meanwhile, LA activated the G protein-coupled receptor 84 (GPR84) and PI3K/Akt signaling pathway. In agreement, dietary 1% LA enhanced mammary duct development, increased the expression of GPR84 and cyclin D1, and activated PI3K/Akt in mammary gland of pubertal mice. Furthermore, knockdown of GPR84 or inhibition of PI3K/Akt totally abolished the promotion of HC11 proliferation induced by LA. These results showed that LA stimulated mammary gland development of pubertal mice through activation of GPR84 and PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Yingying Meng
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510642, P. R. China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, P. R. China
| | - Jing Zhang
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510642, P. R. China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, P. R. China
| | - Fenglin Zhang
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510642, P. R. China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, P. R. China
| | - Wei Ai
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510642, P. R. China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, P. R. China
| | - Xiaotong Zhu
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510642, P. R. China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, P. R. China
| | - Gang Shu
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510642, P. R. China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, P. R. China
| | - Lina Wang
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510642, P. R. China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, P. R. China
| | - Ping Gao
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510642, P. R. China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, P. R. China
| | - Qianyun Xi
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510642, P. R. China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, P. R. China
| | - Yongliang Zhang
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510642, P. R. China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, P. R. China
| | - Xingwei Liang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi High Education Laboratory for Animal Reproduction and Biotechnology, Guangxi University , Nanning 530004, P. R. China
| | - Qingyan Jiang
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510642, P. R. China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, P. R. China
| | - Songbo Wang
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510642, P. R. China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, P. R. China
| |
Collapse
|