1
|
Huang K, Yang W, Shi M, Wang S, Li Y, Xu Z. The Role of TPM3 in Protecting Cardiomyocyte from Hypoxia-Induced Injury via Cytoskeleton Stabilization. Int J Mol Sci 2024; 25:6797. [PMID: 38928503 PMCID: PMC11203979 DOI: 10.3390/ijms25126797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/10/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Ischemic heart disease (IHD) remains a major global health concern, with ischemia-reperfusion injury exacerbating myocardial damage despite therapeutic interventions. In this study, we investigated the role of tropomyosin 3 (TPM3) in protecting cardiomyocytes against hypoxia-induced injury and oxidative stress. Using the AC16 and H9c2 cell lines, we established a chemical hypoxia model by treating cells with cobalt chloride (CoCl2) to simulate low-oxygen conditions. We found that CoCl2 treatment significantly upregulated the expression of hypoxia-inducible factor 1 alpha (HIF-1α) in cardiomyocytes, indicating the successful induction of hypoxia. Subsequent morphological and biochemical analyses revealed that hypoxia altered cardiomyocyte morphology disrupted the cytoskeleton, and caused cellular damage, accompanied by increased lactate dehydrogenase (LDH) release and malondialdehyde (MDA) levels, and decreased superoxide dismutase (SOD) activity, indicative of oxidative stress. Lentivirus-mediated TPM3 overexpression attenuated hypoxia-induced morphological changes, cellular damage, and oxidative stress imbalance, while TPM3 knockdown exacerbated these effects. Furthermore, treatment with the HDAC1 inhibitor MGCD0103 partially reversed the exacerbation of hypoxia-induced injury caused by TPM3 knockdown. Protein-protein interaction (PPI) network and functional enrichment analysis suggested that TPM3 may modulate cardiac muscle development, contraction, and adrenergic signaling pathways. In conclusion, our findings highlight the therapeutic potential of TPM3 modulation in mitigating hypoxia-associated cardiac injury, suggesting a promising avenue for the treatment of ischemic heart disease and other hypoxia-related cardiac pathologies.
Collapse
Affiliation(s)
- Ke Huang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730030, China;
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou 730030, China; (W.Y.); (M.S.); (S.W.)
| | - Weijia Yang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou 730030, China; (W.Y.); (M.S.); (S.W.)
| | - Mingxuan Shi
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou 730030, China; (W.Y.); (M.S.); (S.W.)
| | - Shiqi Wang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou 730030, China; (W.Y.); (M.S.); (S.W.)
| | - Yi Li
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou 730030, China; (W.Y.); (M.S.); (S.W.)
| | - Zhaoqing Xu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730030, China;
| |
Collapse
|
2
|
Surilige, Hu P, Bai T, Xiu Z, Hujiya, Li M, Zhang Q, Wan Q. Exploring the role of aquaporin proteins in the pre-protective action of Sanwei sandalwood decoction from adriamycin-induced chronic heart failure: A mechanistic study. Heliyon 2023; 9:e22718. [PMID: 38058452 PMCID: PMC10696208 DOI: 10.1016/j.heliyon.2023.e22718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 11/16/2023] [Accepted: 11/16/2023] [Indexed: 12/08/2023] Open
Abstract
This study employed network pharmacology, molecular docking technology, and modern pharmacological research methods to explore the pre-protective effect and underlying mechanism, Sanwei sandalwood decoction, against Adriamycin-induced Chronic Heart Failure, with a particular focus on the involvement of aquaporins. Additionally, the study highlighted aquaporins as a significant factor, affecting processes such as cell proliferation and response to reactive oxygen species. The results of in vivo experiments demonstrated that the administration of Sanwei sandalwood decoction in rats with chronic heart failure led to an enhancement in the ejection fraction and improved heart ejection function. Additionally, the decoction significantly reduced the serum levels of Creatine Kinase, Creatine Kinase-MB, and N-terminal pro-B-type natriuretic peptide. Furthermore, the relative expression of Aquarporin-1, 4, and 7mRNAs and proteins in the hearts of rats with chronic heart failure was down-regulated upon treatment. Overall, Sanwei sandalwood decoction can have an effective cardioprotective effect in preventing Adriamycin-induced Chronic Heart Failure in rats.
Collapse
Affiliation(s)
- Surilige
- Affiliated Hospital of Inner Mongolia University for Nationalities Inner Mongolia, China
| | - Pengfei Hu
- Affiliated Hospital of Inner Mongolia University for Nationalities Inner Mongolia, China
| | - Tingting Bai
- Key Laboratory of Mongolian Medicine Pharmacology for Cardio Inner Mongolia, China
| | - Zhi Xiu
- Clinical Medical College of Inner Mongolia University for Nationalities Inner Mongolia, China
| | - Hujiya
- Clinical Medical College of Inner Mongolia University for Nationalities Inner Mongolia, China
| | - Ming Li
- Clinical Medical College of Inner Mongolia University for Nationalities Inner Mongolia, China
| | - Qingshan Zhang
- Affiliated Hospital of Inner Mongolia University for Nationalities Inner Mongolia, China
| | - Quan Wan
- Affiliated Hospital of Inner Mongolia University for Nationalities Inner Mongolia, China
| |
Collapse
|
3
|
Eltobshy SAG, Messiha R, Metias E, Sarhan M, El-Gamal R, El-Shaieb A, Ghalwash M. Effect of SGLT2 Inhibitor on Cardiomyopathy in a Rat Model of T2DM: Possible involvement of Cardiac Aquaporins. Tissue Cell 2023; 85:102200. [PMID: 37660414 DOI: 10.1016/j.tice.2023.102200] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 08/17/2023] [Accepted: 08/19/2023] [Indexed: 09/05/2023]
Abstract
Diabetic cardiomyopathy (DCM) causes arrhythmia, heart failure, and sudden death. Empagliflozin, an SGLT-2 (Sodium glucose co-transporter) inhibitor, is an anti-diabetic medication that decreases blood glucose levels by stimulating urinary glucose excretion. Several aquaporins (AQPs) including AQP-1-3 and - 4 and their involvement in the pathogenesis in different cardiac diseases were detected. In the current study the effect of Empagliflozin on diabetic cardiomyopathy and the possible involvement of cardiac AQPs were investigated. METHODS 56 adult male Sprague-Dawley rats were divided into 4 groups: Control, DCM: type 2 diabetic rats, low EMPA+DCM received empagliflozin (10 mg/kg/day) and high EMPA+DCM received empagliflozin (30 mg/kg/day) for 6 weeks. RESULTS Administration of both EMPA doses, especially in high dose group, led to significant improvement in ECG parameters. Also, a significant improvement in biochemical and cardiac oxidative stress markers (significant decrease in serum CK-MB, and malondialdehyde while increasing catalase) with decreased fibrosis and edema in histopathological examination and a significant attenuation in apoptosis (caspase-3) and edema (AQP-1& -4). CONCLUSION Both doses of Empagliflozin have a cardioprotective effect and reduced myocardial tissue edema with high dose having a greater effect. This might be due to attenuation of oxidative stress, fibrosis and edema mediated through AQP-1, - 3& - 4 expression.
Collapse
Affiliation(s)
- Somaia A G Eltobshy
- Department of Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Refka Messiha
- Department of Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Emile Metias
- Department of Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Mohamed Sarhan
- Department of Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Randa El-Gamal
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt; Medical Experimental Research Center, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Ahmed El-Shaieb
- Department of Pathology, Faculty of Veterinary Medicine, Mansoura university, Mansoura 35516, Egypt
| | - Mohammad Ghalwash
- Department of Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
4
|
Lin H, Duan X, Li L, Ye J, Xiao H. Neuromyelitis Optica Spectrum Disorder with Sick Sinus Syndrome: Two Cases and a Literature Review. Healthcare (Basel) 2023; 11:2810. [PMID: 37957955 PMCID: PMC10649943 DOI: 10.3390/healthcare11212810] [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: 09/06/2023] [Revised: 10/17/2023] [Accepted: 10/20/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND AND OBJECTIVE Neuromyelitis optica spectrum disorder (NMOSD) is a rare immune-mediated demyelinating disease of the central nervous system (CNS). There is a lack of reports of sick sinus syndrome (SSS) associated with NMOSD; thus, we hereby report two cases of patients with NMOSD who developed SSS. CASES PRESENTATION The patients were both male and presented with area postrema syndrome. Brain MRI showed lesions in the dorsal part of their medulla oblongata. They were diagnosed with NMOSD when aquaporin-4 antibodies were found in their serum. Slow heart rates and several episodes of syncope were also observed in case 1 during hospitalization, while Holter monitoring showed sinus pauses (10-11 s) and SSS was diagnosed. A pacemaker was fitted. Case 2 had a respiratory arrest followed by a subsequent cardiac arrest. He was successfully resuscitated with epinephrine injection and cardiopulmonary resuscitation. Through immunotherapy, their neurological functions became stable and heart rate and blood pressure returned to the baseline. CONCLUSIONS Since sick sinus syndrome is a life-threatening complication, serious heart arrhythmias should be considered as a potential result of area postrema syndrome associated with NMOSD.
Collapse
Affiliation(s)
- Huiting Lin
- Department of Neurology, Neuromedicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518000, China; (H.L.); (X.D.); (L.L.); (J.Y.)
| | - Xinyi Duan
- Department of Neurology, Neuromedicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518000, China; (H.L.); (X.D.); (L.L.); (J.Y.)
- Shenzhen University Medical School, Shenzhen University, Shenzhen 518000, China
| | - Lina Li
- Department of Neurology, Neuromedicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518000, China; (H.L.); (X.D.); (L.L.); (J.Y.)
| | - Jinhao Ye
- Department of Neurology, Neuromedicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518000, China; (H.L.); (X.D.); (L.L.); (J.Y.)
| | - Haibing Xiao
- Department of Neurology, Neuromedicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518000, China; (H.L.); (X.D.); (L.L.); (J.Y.)
| |
Collapse
|
5
|
da Silva IV, Soveral G. Aquaporins in Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1398:289-302. [PMID: 36717502 DOI: 10.1007/978-981-19-7415-1_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Obesity is one of the most important metabolic disorders of this century and is associated with a cluster of the most dangerous cardiovascular disease risk factors, such as insulin resistance and diabetes, dyslipidemia, and hypertension, collectively named Metabolic Syndrome. The role of aquaporins (AQP) in glycerol metabolism facilitating glycerol release from the adipose tissue and distribution to various tissues and organs unveils these membrane channels as important players in lipid balance and energy homeostasis and points to their involvement in a variety of pathophysiological mechanisms including insulin resistance, obesity, and diabetes. This review summarizes the physiologic role of aquaglyceroporins in glycerol metabolism and lipid homeostasis, describing their specific tissue distribution, involvement in glycerol balance, and implication in obesity and fat-related metabolic complications. The development of specify pharmacologic modulators able to regulate aquaglyceroporins expression and function, in particular AQP7 in adipose tissue, might constitute a novel approach for controlling obesity and other metabolic disorders.
Collapse
Affiliation(s)
- Inês V da Silva
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
- Department Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Graça Soveral
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal.
- Department Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
6
|
Rojo‐García AV, Vanmunster M, Pacolet A, Suhr F. Physical inactivity by tail suspension alters markers of metabolism, structure, and autophagy of the mouse heart. Physiol Rep 2023; 11:e15574. [PMID: 36695670 PMCID: PMC9875748 DOI: 10.14814/phy2.15574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 12/28/2022] [Accepted: 12/29/2022] [Indexed: 06/17/2023] Open
Abstract
Sedentary behavior has become ingrained in our society and has been linked to cardiovascular diseases. Physical inactivity is the main characteristic of sedentary behavior. However, its impact on cardiovascular disease is not clear. Therefore, we investigated the effect of physical inactivity in an established mouse model on gene clusters associated with cardiac fibrosis, electrophysiology, cell regeneration, and tissue degradation/turnover. We investigated a sedentary group (CTR, n = 10) versus a tail suspension group (TS, n = 11) that caused hindlimb unloading and consequently physical inactivity. Through histological, protein content, and transcript analysis approaches, we found that cardiac fibrosis-related genes partly change, with significant TS-associated increases in Tgfb1, but without changes in Col1a1 and Fn1. These changes are not translated into fibrosis at tissue level. We further detected TS-mediated increases in protein degradation- (Trim63, p < 0.001; Fbxo32, p = 0.0947 as well as in biosynthesis-related [P70s6kb1, p < 0.01]). Corroborating these results, we found increased expression of autophagy markers such as Atg7 (p < 0.01) and ULK1 (p < 0.05). Two cardiomyocyte regeneration- and sarcomerogenesis-related genes, Yap (p = 0.0535) and Srf (p < 0.001), increased upon TS compared to CTR conditions. Finally, we found significant upregulation of Gja1 (p < 0.05) and a significant downregulation of Aqp1 (p < 0.05). Our data demonstrate that merely 2 weeks of reduced physical activity induce changes in genes associated with cardiac structure and electrophysiology. Hence, these data should find the basis for novel research directed to evaluate the interplay of cardiac functioning and physical inactivity.
Collapse
Affiliation(s)
| | - Mathias Vanmunster
- Department of Movement SciencesExercise Physiology Research Group, KU LeuvenLeuvenBelgium
| | - Alexander Pacolet
- Department of Movement SciencesExercise Physiology Research Group, KU LeuvenLeuvenBelgium
| | - Frank Suhr
- Department of Movement SciencesExercise Physiology Research Group, KU LeuvenLeuvenBelgium
| |
Collapse
|
7
|
Shangzu Z, Dingxiong X, ChengJun M, Yan C, Yangyang L, Zhiwei L, Ting Z, Zhiming M, Yiming Z, Liying Z, Yongqi L. Aquaporins: Important players in the cardiovascular pathophysiology. Pharmacol Res 2022; 183:106363. [PMID: 35905892 DOI: 10.1016/j.phrs.2022.106363] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 11/15/2022]
Abstract
Aquaporin is a membrane channel protein widely expressed in body tissues, which can control the input and output of water in cells. AQPs are differentially expressed in different cardiovascular tissues and participate in water transmembrane transport, cell migration, metabolism, inflammatory response, etc. The aberrant expression of AQPs highly correlates with the onset of ischemic heart disease, myocardial ischemia-reperfusion injury, heart failure, etc. Despite much attention to the regulatory role of AQPs in the cardiovascular system, the translation of AQPs into clinical application still faces many challenges, including clarification of the localization of AQPs in the cardiovascular system and mechanisms mediating cardiovascular pathophysiology, as well as the development of cardiovascular-specific AQPs modulators.Therefore, in this study, we comprehensively reviewed the critical roles of AQP family proteins in maintaining cardiovascular homeostasis and described the underlying mechanisms by which AQPs mediated the outcomes of cardiovascular diseases. Meanwhile, AQPs serve as important therapeutic targets, which provide a wide range of opportunities to investigate the mechanisms of cardiovascular diseases and the treatment of those diseases.
Collapse
Affiliation(s)
- Zhang Shangzu
- Gansu University of traditional Chinese Medicine, LanZhou, China
| | - Xie Dingxiong
- Gansu Institute of Cardiovascular Diseases, LanZhou,China
| | - Ma ChengJun
- Gansu University of traditional Chinese Medicine, LanZhou, China
| | - Chen Yan
- Gansu University of traditional Chinese Medicine, LanZhou, China
| | - Li Yangyang
- Gansu University of traditional Chinese Medicine, LanZhou, China
| | - Liu Zhiwei
- Gansu University of traditional Chinese Medicine, LanZhou, China
| | - Zhou Ting
- Gansu University of traditional Chinese Medicine, LanZhou, China
| | - Miao Zhiming
- Gansu University of traditional Chinese Medicine, LanZhou, China
| | - Zhang Yiming
- Gansu University of traditional Chinese Medicine, LanZhou, China
| | - Zhang Liying
- Gansu University of traditional Chinese Medicine, LanZhou, China; Gansu Institute of Cardiovascular Diseases, LanZhou,China.
| | - Liu Yongqi
- Gansu University of traditional Chinese Medicine, LanZhou, China; Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities Gansu University of Chinese Medicine, Lanzhou, China; Key Laboratory of Dunhuang Medicine and Transformation at Provincial and Ministerial Level, Lanzhou, China.
| |
Collapse
|
8
|
Segura-Anaya E, Martínez-Gómez A, Dent MA. Differences in the localization of AQP1 and expression patterns of AQP isoforms in rat and mouse sciatic nerve and changes in rat AQPs expression after nerve crush injury. IBRO Neurosci Rep 2022; 12:82-89. [PMID: 35036988 PMCID: PMC8749057 DOI: 10.1016/j.ibneur.2021.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/14/2021] [Accepted: 12/16/2021] [Indexed: 12/01/2022] Open
Abstract
In the peripheral nervous system aquaporins (AQPs) have been reported in both peripheral neurons and glial cells. Previously we described the precise localization of AQP1 in the rat sciatic nerve, which is present in both Remak and myelin Schwann cells, and is enriched in the Schmidt-Lanterman incisures. In this work, we found that AQP1 in mouse is only present in Remak cells, showing a different localization between these species. However, after nerve crush injury the level of AQP1 mRNA expression remains constant at all times studied in rat and mouse. We then performed RT-PCR of nine AQP (AQP1-9) isoforms from rat and mouse sciatic nerve, we found that in rat only five AQPs are present (AQP1, AQP4, AQP5, AQP7 and AQP9), whereas in mouse all AQPs except AQP8 are expressed. Then, we studied the expression by RT-PCR of AQPs in rat after nerve crush injury, showing that AQP1, AQP4 and AQP7 expression remain constant at all times studied, while AQP2, AQP5 and AQP9 are upregulated after injury. Therefore, these two closely related rodents show different AQP1 localization and have different AQPs expression patterns in the sciatic nerve, possibly due to a difference in the regulation of these AQPs. The expression of AQP1 in Remak cells supports the involvement of AQP1 in pain perception. Also, in rat the upregulation of AQP2, AQP5 and AQP7 after nerve injury suggests a possible role for these AQPs in promoting regeneration following injury.
Collapse
Affiliation(s)
- Edith Segura-Anaya
- Laboratorio de Neurociencias, Facultad de Medicina, Universidad Autónoma del Estado de México, Paseo Tollocan y Jesús Carranza, Toluca, Edo. de México CP 50180, México
| | - Alejandro Martínez-Gómez
- Laboratorio de Neurociencias, Facultad de Medicina, Universidad Autónoma del Estado de México, Paseo Tollocan y Jesús Carranza, Toluca, Edo. de México CP 50180, México
| | - Myrna A.R. Dent
- Laboratorio de Neurociencias, Facultad de Medicina, Universidad Autónoma del Estado de México, Paseo Tollocan y Jesús Carranza, Toluca, Edo. de México CP 50180, México
| |
Collapse
|
9
|
Wagner K, Unger L, Salman MM, Kitchen P, Bill RM, Yool AJ. Signaling Mechanisms and Pharmacological Modulators Governing Diverse Aquaporin Functions in Human Health and Disease. Int J Mol Sci 2022; 23:1388. [PMID: 35163313 PMCID: PMC8836214 DOI: 10.3390/ijms23031388] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 02/07/2023] Open
Abstract
The aquaporins (AQPs) are a family of small integral membrane proteins that facilitate the bidirectional transport of water across biological membranes in response to osmotic pressure gradients as well as enable the transmembrane diffusion of small neutral solutes (such as urea, glycerol, and hydrogen peroxide) and ions. AQPs are expressed throughout the human body. Here, we review their key roles in fluid homeostasis, glandular secretions, signal transduction and sensation, barrier function, immunity and inflammation, cell migration, and angiogenesis. Evidence from a wide variety of studies now supports a view of the functions of AQPs being much more complex than simply mediating the passive flow of water across biological membranes. The discovery and development of small-molecule AQP inhibitors for research use and therapeutic development will lead to new insights into the basic biology of and novel treatments for the wide range of AQP-associated disorders.
Collapse
Affiliation(s)
- Kim Wagner
- School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia;
| | - Lucas Unger
- College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK; (L.U.); (P.K.)
| | - Mootaz M. Salman
- Department of Physiology Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK;
- Oxford Parkinson’s Disease Centre, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Philip Kitchen
- College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK; (L.U.); (P.K.)
| | - Roslyn M. Bill
- College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK; (L.U.); (P.K.)
| | - Andrea J. Yool
- School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia;
| |
Collapse
|
10
|
A Cardioplegic Solution with an Understanding of a Cardiochannelopathy. Antioxidants (Basel) 2021; 10:antiox10121878. [PMID: 34942981 PMCID: PMC8698488 DOI: 10.3390/antiox10121878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/16/2021] [Accepted: 11/23/2021] [Indexed: 01/11/2023] Open
Abstract
Cardiac surgeries have been improved by accompanying developing cardioplegia solutions. However, the cardioplegia application presents an ongoing challenge with a view of a sufficiently restored cardiac function. In this review, we focus on the cardioplegia-induced mechanism and summarize the findings of studies undertaken to improve cardioprotective strategies. Currently, and somewhat surprisingly, relatively little is known about cardiac electrolyte regulation through channel physiology. We hope that an improved understanding of the electrolyte transport through ion channels/transporters and modulations of water channel aquaporins will provide an insight into cardiac channel physiology and a channel-based cardiac pathology of a cardiochannelopathy.
Collapse
|
11
|
Oztopuz O, Coskun O, Buyuk B. Alterations in aquaporin gene expression level on cyclophosphamide-induced cardiac injury and possible protective role of Ganoderma lucidum. Biologia (Bratisl) 2021. [DOI: 10.1007/s11756-021-00817-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
12
|
Montiel V, Bella R, Michel LYM, Esfahani H, De Mulder D, Robinson EL, Deglasse JP, Tiburcy M, Chow PH, Jonas JC, Gilon P, Steinhorn B, Michel T, Beauloye C, Bertrand L, Farah C, Dei Zotti F, Debaix H, Bouzin C, Brusa D, Horman S, Vanoverschelde JL, Bergmann O, Gilis D, Rooman M, Ghigo A, Geninatti-Crich S, Yool A, Zimmermann WH, Roderick HL, Devuyst O, Balligand JL. Inhibition of aquaporin-1 prevents myocardial remodeling by blocking the transmembrane transport of hydrogen peroxide. Sci Transl Med 2021; 12:12/564/eaay2176. [PMID: 33028705 DOI: 10.1126/scitranslmed.aay2176] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 12/24/2019] [Accepted: 08/31/2020] [Indexed: 12/31/2022]
Abstract
Pathological remodeling of the myocardium has long been known to involve oxidant signaling, but strategies using systemic antioxidants have generally failed to prevent it. We sought to identify key regulators of oxidant-mediated cardiac hypertrophy amenable to targeted pharmacological therapy. Specific isoforms of the aquaporin water channels have been implicated in oxidant sensing, but their role in heart muscle is unknown. RNA sequencing from human cardiac myocytes revealed that the archetypal AQP1 is a major isoform. AQP1 expression correlates with the severity of hypertrophic remodeling in patients with aortic stenosis. The AQP1 channel was detected at the plasma membrane of human and mouse cardiac myocytes from hypertrophic hearts, where it colocalized with NADPH oxidase-2 and caveolin-3. We show that hydrogen peroxide (H2O2), produced extracellularly, is necessary for the hypertrophic response of isolated cardiac myocytes and that AQP1 facilitates the transmembrane transport of H2O2 through its water pore, resulting in activation of oxidant-sensitive kinases in cardiac myocytes. Structural analysis of the amino acid residues lining the water pore of AQP1 supports its permeation by H2O2 Deletion of Aqp1 or selective blockade of the AQP1 intrasubunit pore inhibited H2O2 transport in mouse and human cells and rescued the myocyte hypertrophy in human induced pluripotent stem cell-derived engineered heart muscle. Treatment of mice with a clinically approved AQP1 inhibitor, Bacopaside, attenuated cardiac hypertrophy. We conclude that cardiac hypertrophy is mediated by the transmembrane transport of H2O2 by the water channel AQP1 and that inhibitors of AQP1 represent new possibilities for treating hypertrophic cardiomyopathies.
Collapse
Affiliation(s)
- Virginie Montiel
- Institute of Experimental and Clinical Research (IREC), Pharmacology and Therapeutics (FATH), Cliniques Universitaires St Luc and Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Ramona Bella
- Institute of Experimental and Clinical Research (IREC), Pharmacology and Therapeutics (FATH), Cliniques Universitaires St Luc and Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Lauriane Y M Michel
- Institute of Experimental and Clinical Research (IREC), Pharmacology and Therapeutics (FATH), Cliniques Universitaires St Luc and Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Hrag Esfahani
- Institute of Experimental and Clinical Research (IREC), Pharmacology and Therapeutics (FATH), Cliniques Universitaires St Luc and Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Delphine De Mulder
- Institute of Experimental and Clinical Research (IREC), Pharmacology and Therapeutics (FATH), Cliniques Universitaires St Luc and Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Emma L Robinson
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KULeuven, 3000 Leuven, Belgium
| | - Jean-Philippe Deglasse
- Institute of Experimental and Clinical Research (IREC), Endocrinology, Diabetes and Nutrition (EDIN), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Malte Tiburcy
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, 37075 Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Pak Hin Chow
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia
| | - Jean-Christophe Jonas
- Institute of Experimental and Clinical Research (IREC), Endocrinology, Diabetes and Nutrition (EDIN), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Patrick Gilon
- Institute of Experimental and Clinical Research (IREC), Endocrinology, Diabetes and Nutrition (EDIN), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Benjamin Steinhorn
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 2115, USA
| | - Thomas Michel
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 2115, USA
| | - Christophe Beauloye
- Institute of Experimental and Clinical Research (IREC), Pole of Cardiovascular Research (CARD), Cliniques Universitaires St Luc and Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Luc Bertrand
- Institute of Experimental and Clinical Research (IREC), Pole of Cardiovascular Research (CARD), Cliniques Universitaires St Luc and Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Charlotte Farah
- Institute of Experimental and Clinical Research (IREC), Pharmacology and Therapeutics (FATH), Cliniques Universitaires St Luc and Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Flavia Dei Zotti
- Institute of Experimental and Clinical Research (IREC), Pharmacology and Therapeutics (FATH), Cliniques Universitaires St Luc and Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Huguette Debaix
- Institute of Experimental and Clinical Research (IREC), Nephrology (NEFR), Cliniques Universitaires St Luc and Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.,Institute of Physiology, University of Zürich, CH 8057 Zürich, Switzerland
| | - Caroline Bouzin
- 2IP-IREC Imaging Platform, Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Davide Brusa
- Flow Cytometry Platform, Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Sandrine Horman
- Institute of Experimental and Clinical Research (IREC), Pole of Cardiovascular Research (CARD), Cliniques Universitaires St Luc and Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Jean-Louis Vanoverschelde
- Institute of Experimental and Clinical Research (IREC), Pole of Cardiovascular Research (CARD), Cliniques Universitaires St Luc and Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Olaf Bergmann
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, 01062 Dresden, Germany.,Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Dimitri Gilis
- Computational Biology and Bioinformatics (3BIO-BioInfo), Université Libre de Bruxelles (ULB), 1000 Brussels, Belgium
| | - Marianne Rooman
- Computational Biology and Bioinformatics (3BIO-BioInfo), Université Libre de Bruxelles (ULB), 1000 Brussels, Belgium
| | - Alessandra Ghigo
- Molecular Biotechnology Center, Università di Torino, 10124 Torino, Italy
| | | | - Andrea Yool
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia
| | - Wolfram H Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, 37075 Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany.,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, 37075 Göttingen, Germany
| | - H Llewelyn Roderick
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KULeuven, 3000 Leuven, Belgium
| | - Olivier Devuyst
- Institute of Experimental and Clinical Research (IREC), Nephrology (NEFR), Cliniques Universitaires St Luc and Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.,Institute of Physiology, University of Zürich, CH 8057 Zürich, Switzerland
| | - Jean-Luc Balligand
- Institute of Experimental and Clinical Research (IREC), Pharmacology and Therapeutics (FATH), Cliniques Universitaires St Luc and Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.
| |
Collapse
|
13
|
Exploring the role of Aquaporins (AQPs) in LPS induced systemic inflammation and the ameliorative effect of Garcinia in male Wistar rat. Inflammopharmacology 2021; 29:801-823. [PMID: 34106384 DOI: 10.1007/s10787-021-00832-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 05/29/2021] [Indexed: 12/16/2022]
Abstract
The Aquaporins (AQPs) could prove to be striking targets of inflammation. The aim of this study was to study the involvement of AQPs and explore the anti-inflammatory activity of Garcinia extract in LPS induced acute systemic inflammation in Wistar rats. Adult male Wistar rats (n = 6) were pretreated with Garcinia orally twice for 7 days, followed by a single intraperitoneal dose (5.5 mg/kgbw) of LPS. Serum ALT, AST, ALP, Creatinine, Urea and BUN, nitric oxide, prostaglandin, cytokine and chemokine levels were measured. LC-MS analysis of Garcinia was performed to identify the phytoconstituents present. The iNOS and COX enzyme activity were determined in the target tissues. qPCR analysis of inos, cox-2 and aqps was performed. Relative protein expression of AQPs was studied by Western blot analysis. Molecular docking studies were performed to study the interaction of garcinol and hydroxycitric acid, the two important phytoconstituents of Garcinia with AQP. The qPCR analysis showed tissue-specific up-regulation of aqp1, aqp3, aqp4 and aqp8 in LPS induced rats. Garcinia extract treatment effectively lowered the mRNA expression of these AQPs. Garcinia extract significantly inhibited the LPS-induced NO, prostaglandin, cytokine and chemokine production in serum and also decreased tissue-specific transcript level of inos and cox-2, thus suggesting the anti-inflammatory role of Garcinia. Also, docking studies revealed interactions of garcinol and hydroxycitric acid with AQP1, 3, 4 and 8. Therefore, the present study suggests the possible involvement of AQP1, 3, 4 and 8 in inflammation and the efficacy of Garcinia extract as an anti-inflammatory agent. Therefore, AQPs can act as prognostic markers of inflammation and can be targeted with Garcinia extract.
Collapse
|
14
|
Azad AK, Raihan T, Ahmed J, Hakim A, Emon TH, Chowdhury PA. Human Aquaporins: Functional Diversity and Potential Roles in Infectious and Non-infectious Diseases. Front Genet 2021; 12:654865. [PMID: 33796134 PMCID: PMC8007926 DOI: 10.3389/fgene.2021.654865] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 02/22/2021] [Indexed: 12/11/2022] Open
Abstract
Aquaporins (AQPs) are integral membrane proteins and found in all living organisms from bacteria to human. AQPs mainly involved in the transmembrane diffusion of water as well as various small solutes in a bidirectional manner are widely distributed in various human tissues. Human contains 13 AQPs (AQP0-AQP12) which are divided into three sub-classes namely orthodox aquaporin (AQP0, 1, 2, 4, 5, 6, and 8), aquaglyceroporin (AQP3, 7, 9, and 10) and super or unorthodox aquaporin (AQP11 and 12) based on their pore selectivity. Human AQPs are functionally diverse, which are involved in wide variety of non-infectious diseases including cancer, renal dysfunction, neurological disorder, epilepsy, skin disease, metabolic syndrome, and even cardiac diseases. However, the association of AQPs with infectious diseases has not been fully evaluated. Several studies have unveiled that AQPs can be regulated by microbial and parasitic infections that suggest their involvement in microbial pathogenesis, inflammation-associated responses and AQP-mediated cell water homeostasis. This review mainly aims to shed light on the involvement of AQPs in infectious and non-infectious diseases and potential AQPs-target modulators. Furthermore, AQP structures, tissue-specific distributions and their physiological relevance, functional diversity and regulations have been discussed. Altogether, this review would be useful for further investigation of AQPs as a potential therapeutic target for treatment of infectious as well as non-infectious diseases.
Collapse
Affiliation(s)
- Abul Kalam Azad
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Topu Raihan
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Jahed Ahmed
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Al Hakim
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Tanvir Hossain Emon
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | | |
Collapse
|
15
|
Politi MT, Ochoa F, Netti V, Ferreyra R, Bortman G, Sanjuan N, Morales C, Piazza A, Capurro C. Changes in cardiac Aquaporin expression during aortic valve replacement surgery with cardiopulmonary bypass. Eur J Cardiothorac Surg 2020; 57:556-564. [PMID: 31535145 DOI: 10.1093/ejcts/ezz249] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 07/21/2019] [Accepted: 08/13/2019] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVES Cardiopulmonary bypass (CPB) use is an essential strategy for many cardiovascular surgeries. However, its use and duration have been associated with a higher rate of postoperative complications, such as low cardiac output syndrome due to myocardial oedema and dysfunction. Though Aquaporin water channels have been implicated in myocardial water balance, their specific role in this clinical scenario has not been established. METHODS In a consecutive study of 17 patients with severe aortic stenosis undergoing aortic valve replacement surgery, 2 myocardial biopsies of the left ventricle were taken: 1 before and 1 after CPB use. Sociodemographic, clinical and laboratory data were collected. Western blot and immunohistochemistry studies were performed. RESULTS After CPB use, there was a mean increase of ∼62% in Aquaporin 1 protein levels (P = 0.001) and a mean reduction of ∼38% in Aquaporin 4 protein levels (P = 0.030). In immunohistochemistry assays, Aquaporin 1 was found lining small blood vessels, while Aquaporin 4 formed a circular label in cardiomyocytes. There were no changes in the localization of either protein following CPB use. During the observed on-pump time interval, there was a 1.7%/min mean increase in Aquaporin 1 (P = 0.021) and a 2.5%/min mean decrease in Aquaporin 4 (P = 0.018). Myocardial interstitial oedema increased by 42% (95% confidence interval 31-54%) after CPB use. Patients who developed low cardiac output syndrome were in the upper half of the median percentage change of Aquaporin expression. CONCLUSION Time-dependent changes in cardiac Aquaporin expression may be associated with myocardial oedema and dysfunction related to CPB use.
Collapse
Affiliation(s)
- María Teresa Politi
- School of Medicine, University of Buenos Aires, National Scientific and Technical Research Council, Institute of Physiology and Biophysics "Bernardo Houssay" (IFIBIO-HOUSSAY), Buenos Aires, Argentina
| | - Federico Ochoa
- School of Medicine, University of Buenos Aires, National Scientific and Technical Research Council, Institute of Physiology and Biophysics "Bernardo Houssay" (IFIBIO-HOUSSAY), Buenos Aires, Argentina
| | - Vanina Netti
- School of Medicine, University of Buenos Aires, National Scientific and Technical Research Council, Institute of Physiology and Biophysics "Bernardo Houssay" (IFIBIO-HOUSSAY), Buenos Aires, Argentina
| | - Raúl Ferreyra
- Department of Cardiology, Sanatorio de la Trinidad-Mitre, Buenos Aires, Argentina
| | - Guillermo Bortman
- Department of Cardiology, Sanatorio de la Trinidad-Mitre, Buenos Aires, Argentina
| | - Norberto Sanjuan
- Laboratory of Experimental Pathology, Department of Microbiology (IMPaM-CONICET), School of Medicine, University of Buenos Aires, National Scientific and Technical Research Council, Buenos Aires, Argentina
| | - Celina Morales
- School of Medicine, Facultad de Medicina, University of Buenos Aires, Institute of Cardiovascular Pathophysiology (INFICA), Buenos Aires, Argentina
| | - Antonio Piazza
- Department of Cardiovascular Surgery, Sanatorio de la Trinidad-Mitre, Buenos Aires, Argentina
| | - Claudia Capurro
- School of Medicine, University of Buenos Aires, National Scientific and Technical Research Council, Institute of Physiology and Biophysics "Bernardo Houssay" (IFIBIO-HOUSSAY), Buenos Aires, Argentina
| |
Collapse
|
16
|
Fujii M, Ota K, Bessho R. Cardioprotective effect of hyperkalemic cardioplegia in an aquaporin 7-deficient murine heart. Gen Thorac Cardiovasc Surg 2019; 68:578-584. [PMID: 31707553 DOI: 10.1007/s11748-019-01243-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 10/25/2019] [Indexed: 11/30/2022]
Abstract
BACKGROUND Hyperkalemic cardioplegia using St. Thomas' Hospital solution No. 2 (STH2) is commonly used to protect the myocardium during surgery. Mice deficient in the myocyte channel aquaporin 7 (AQP7) show significantly reduced glycerol and ATP contents and develop obesity; however, the influence of AQP7 on cardioplegia effectiveness remains unclear. METHODS After determining the influence of ischemic duration on cardiac function, isolated hearts of male wild-type (WT) and AQP7-knockout (KO) mice (> 13 weeks old) were aerobically Langendorff-perfused with bicarbonate buffer, and randomly allocated to the control group (25 min of global ischemia) and STH2 group (5 min of STH2 infusion before 20 min of global ischemia, followed by 60 min of reperfusion). RESULTS Final recovery of left ventricular developed pressure (LVDP) of WT and AQP7-KO hearts in the control group was 24.5 ± 12.4% and 20.6 ± 8.4%, respectively, which were significantly lower than those of the STH2 group (96.4 ± 12.7% and 92.9 ± 27.6%). Troponin T levels of WT and AQP-KO hearts significantly decreased in the STH2 groups (142.9 ± 27.2 and 219.9 ± 197.3) compared to those of the control (1725.0 ± 768.6 and 1710 ± 819.9). CONCLUSIONS AQP7 was not involved in the protective efficacy of STH2 in this mouse model, suggesting its clinical utility even in complications of metabolic disease.
Collapse
Affiliation(s)
- Masahiro Fujii
- Department of Cardiovascular Surgery, Nippon Medical School Chiba Hokusoh Hospital, 1715 Kamagari, Inzai, Chiba, 270-1694, Japan.
| | - Keisuke Ota
- Department of Cardiovascular Surgery, Nippon Medical School Chiba Hokusoh Hospital, 1715 Kamagari, Inzai, Chiba, 270-1694, Japan
| | - Ryuzo Bessho
- Department of Cardiovascular Surgery, Nippon Medical School Chiba Hokusoh Hospital, 1715 Kamagari, Inzai, Chiba, 270-1694, Japan
| |
Collapse
|
17
|
Rivera MA, Fahey TD. Association Between aquaporin-1 and Endurance Performance: A Systematic Review. SPORTS MEDICINE-OPEN 2019; 5:40. [PMID: 31486928 PMCID: PMC6728102 DOI: 10.1186/s40798-019-0213-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 08/19/2019] [Indexed: 12/28/2022]
Abstract
Background There is abundant and mounting information related to the molecular and biological structure and function of the Aquaporin-1 (AQP1) gene and the AQP1-Aquaporin channel. Regulation of water flow across cell membranes is essential for supporting inter- and intracellular fluid balance, which is critical for health and exercise performance. The transmembrane water channel AQP1 is important for cardiorespiratory endurance (CE) because it influences fluid transfers in erythrocytes, endothelial, and pulmonary cells and is vital for transport of ammonium, bicarbonate, carbon dioxide, glycerol, nitric oxide, potassium ion, water, and trans-epithelial and renal water. Very recent publications suggest the association between a DNA sequence variant, rs1049305 (C > G), in the 3′-untranslated region of the AQP1 gene and CE performance. Other reports indicate further significant associations between AQP1 channel and CE phenotypes. The purposes of this systematic review were to examine the extent of the associations between the AQP1 rs1049305 genotype and CE exercise performance and body fluid loss in long-distance runners and AQP1 channel associations with other CE phenotypes. Methods Data sources: A comprehensive review was conducted using PubMed, EMBASE, CINAHL, and Cochrane electronic databases. The search ranged from January 1, 1988, to December 31, 2018. Studies reported in English, French, and Spanish were considered. Eligibility criteria: The criteria for inclusion in the review were (a) case-control study; (b) unequivocal definition of cases and controls; (c) CE was defined as performance in endurance events, laboratory tests, and/or maximal oxygen consumption; (d) exclusion criteria of known causes; (e) genotyping performed by PCR or sequencing; (f) genotype frequencies reported; and (g) no deviation of genotype frequencies from Hardy-Weinberg equilibrium in the control group. Study appraisal: The systematic review included studies examining the AQP1 gene and AQP1 channel structure and function, associations between the AQP1 gene sequence variant rs1049305 (C > G) and CE performance, body fluid loss in long-distance runners, and other studies reporting on the AQP1 gene and channel CE phenotype associations. Synthesis methods: For each selected study, the following data were extracted: authors, year of publication, sample size and number of cases and controls, CE definition, exclusion criteria, inclusion criteria for cases and controls, methods used for genotyping, genotype, allele frequencies and HWE for genotype frequencies in cases and control groups, and method of AQP1 gene and AQP1 channel analysis. Results The initial databases search found 172 pertinent studies. Of those, 46 studies were utilized in the final synthesis of the systematic review. The most relevant findings were (a) the identification of an independent replication of the association between AQP1 gene sequence variant rs1049305 (C > G) and CE performance; (b) the association of the rs1049305 C-allele with faster CE running performance; (c) in knockout model, using a linear regression analysis of distance run as a function of Aqp1 status (Aqp1-null vs. wild-type mice) and conditions of hypoxia (ambient [O2] = 16%), normoxia (21%), and hyperoxia (40%) indicated that the Aqp1 knockout ran less distance than the wild-type mice (p < 0.001); (d) in vitro, a reduced AQP1 expression was associated with the presence of the rs1049305 G-allele; (e) AQP1 null humans led normal lives and were entirely unaware of any physical limitations. However, they could not support fluid homeostasis when exposed to chronic fluid overload. The limited number of studies with “adequate sample sizes” in various racial and ethnic groups precluding to perform proper in-depth statistical analysis. Conclusions The AQP1 gene and AQP1 channel seems to support homeostatic mechanisms, yet to be totally understood, that are auxiliary in achieving an advantage during endurance exercise. AQP1 functions are vital during exercise and have a profound influence on endurance running performance. AQP1s are underappreciated structures that play vital roles in cellular homeostasis at rest and during CE endurance running exercise. The outcome of the present systematic review provide support to the statement of hypotheses and further research endeavors on the likely influence of AQP1 gene and AQP1 channel on CE performance. Registration: The protocol is not registered.
Collapse
Affiliation(s)
- Miguel A Rivera
- Department of Physical Medicine, Rehabilitation & Sports Medicine, School of Medicine, University of Puerto Rico, Main Building Office A204, San Juan, PR, 00936, USA.
| | - Thomas D Fahey
- Department of Kinesiology, California State University, Chico, 95929-0330, CA, USA
| |
Collapse
|
18
|
Verkerk AO, Lodder EM, Wilders R. Aquaporin Channels in the Heart-Physiology and Pathophysiology. Int J Mol Sci 2019; 20:ijms20082039. [PMID: 31027200 PMCID: PMC6514906 DOI: 10.3390/ijms20082039] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 04/19/2019] [Accepted: 04/23/2019] [Indexed: 12/11/2022] Open
Abstract
Mammalian aquaporins (AQPs) are transmembrane channels expressed in a large variety of cells and tissues throughout the body. They are known as water channels, but they also facilitate the transport of small solutes, gasses, and monovalent cations. To date, 13 different AQPs, encoded by the genes AQP0–AQP12, have been identified in mammals, which regulate various important biological functions in kidney, brain, lung, digestive system, eye, and skin. Consequently, dysfunction of AQPs is involved in a wide variety of disorders. AQPs are also present in the heart, even with a specific distribution pattern in cardiomyocytes, but whether their presence is essential for proper (electro)physiological cardiac function has not intensively been studied. This review summarizes recent findings and highlights the involvement of AQPs in normal and pathological cardiac function. We conclude that AQPs are at least implicated in proper cardiac water homeostasis and energy balance as well as heart failure and arsenic cardiotoxicity. However, this review also demonstrates that many effects of cardiac AQPs, especially on excitation-contraction coupling processes, are virtually unexplored.
Collapse
Affiliation(s)
- Arie O Verkerk
- Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
- Department of Experimental Cardiology, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Elisabeth M Lodder
- Department of Experimental Cardiology, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Ronald Wilders
- Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| |
Collapse
|
19
|
PEDF decreases cardiomyocyte edema during oxygen‑glucose deprivation and recovery via inhibiting lactate accumulation and expression of AQP1. Int J Mol Med 2019; 43:1979-1990. [PMID: 30864707 PMCID: PMC6445592 DOI: 10.3892/ijmm.2019.4132] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 02/28/2019] [Indexed: 01/13/2023] Open
Abstract
Myocardial edema is divided into cellular edema and interstitial edema; however, the dynamic change of cardiomyocyte edema has not been described in detail. Pigment epithelium-derived factor (PEDF) is known for its protective effects on ischemic cardiomyocytes; however, the association between PEDF and cardiomyocyte edema remains to be fully elucidated. In the present study, rat neonatal left ventricular cardiomyocytes were isolated and treated with oxygen-glucose deprivation (OGD) and recovery. During OGD and recovery, the cardiomyocytes exhibited significant edema following 30 min of OGD (OGD 30 min) and OGD 30 min with recovery for 6 h. PEDF significantly decreased the lactate content and extracellular acidification rate of the OGD-treated cardiomyocytes, thereby reducing cellular osmotic gradients and preventing the occurrence of cell edema. In addition, the glycolytic agonist, fructose-1, 6-diphosphate, eliminated the effect of PEDF on inhibiting edema in the OGD-treated cardiomyocytes. Furthermore, PEDF reduced the protein and mRNA expression of aquaporin 1 (AQP1), and thus downregulated cardiomyocyte edema during the OGD/recovery period. The addition of AQP1 agonist, arginine vasopressin, inhibited the inhibitory effect of PEDF on cardiomyocyte edema during OGD/recovery. In conclusion, the present study revealed a novel mechanism for the regulation of cardiomyocyte edema by PEDF involving lactate levels and the expression of AQP1 during OGD/recovery. The reduction of lactate content during OGD was associated with a decrease in the protein level of AQP1 during OGD/recovery; therefore, PEDF decreased cardiomyocyte edema and cellular apoptosis, prolonging the viability of the cells.
Collapse
|
20
|
Lee SY, Ha EJ, Cho HW, Kim HR, Lee D, Eom YB. Potential forensic application of receptor for advanced glycation end products (RAGE) and aquaporin 5 (AQP5) as novel biomarkers for diagnosis of drowning. J Forensic Leg Med 2019; 62:56-62. [PMID: 30677703 DOI: 10.1016/j.jflm.2019.01.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 10/15/2018] [Accepted: 01/11/2019] [Indexed: 02/08/2023]
Abstract
Drowning is the most common cause of unnatural death worldwide. There is no single biomarker to diagnose drowning, so the diagnosis of drowning is one of the most difficult tasks in forensic medicine. Especially, distinguishing a victim of drowning from a body disposed of in water following death remains a problem. The objective of this study was to identify specific biomarkers of drowning compared with other causes of death such as hypoxia and postmortem submersion. The present study investigated the intrapulmonary expression of receptor for advanced glycation end products (RAGE), aquaporin-5 (AQP5), surfactant protein-A (SP-A), interleukin 6 (IL-6) and interleukin 1β (IL-1β) as markers of drowning. In animal experiments, all rats (n = 45) were classified into four groups (drowning, postmortem-submersion, hypoxia and control group). The lungs of experimental animals were analyzed as mRNA expression, immunoblot expression and immunohistochemical staining. qRT-PCR demonstrated increased mRNA expression of RAGE and AQP5 in drowning group compared with that in control, hypoxia and postmortem-submersion group, but not other molecules. Western blotting also showed high expression of RAGE and AQP5 in drowning group, immunostaining of RAGE and AQP5 was highly detected in a linear pattern in type I alveolar epithelial cells, compared with control and postmortem-submersion group. These observations indicate a difference of expression in pulmonary molecular pathology compared with other causes, suggesting RAGE and AQP5 may be useful for differentiation between drowning and postmortem-submersion.
Collapse
Affiliation(s)
- So-Yeon Lee
- Department of Medical Science, College of Medical Sciences, Soonchunhyang University, Asan, Chungnam, 31538, Republic of Korea.
| | - Eun-Ju Ha
- Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, Asan, Chungnam, 31538, Republic of Korea.
| | - Hye-Won Cho
- Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, Asan, Chungnam, 31538, Republic of Korea.
| | - Hye-Rim Kim
- Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, Asan, Chungnam, 31538, Republic of Korea.
| | - Dongsup Lee
- Department of Clinical Laboratory Science, Hyejeon College, Hongseoung, Chungnam, 32244, Republic of Korea.
| | - Yong-Bin Eom
- Department of Medical Science, College of Medical Sciences, Soonchunhyang University, Asan, Chungnam, 31538, Republic of Korea; Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, Asan, Chungnam, 31538, Republic of Korea.
| |
Collapse
|
21
|
Molecular Basis of Aquaporin-7 Permeability Regulation by pH. Cells 2018; 7:cells7110207. [PMID: 30423801 PMCID: PMC6262577 DOI: 10.3390/cells7110207] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 11/05/2018] [Accepted: 11/07/2018] [Indexed: 01/09/2023] Open
Abstract
The aquaglyceroporin AQP7, a family member of aquaporin membrane channels, facilitates the permeation of water and glycerol through cell membranes and is crucial for body lipid and energy homeostasis. Regulation of glycerol permeability via AQP7 is considered a promising therapeutic strategy towards fat-related metabolic complications. Here, we used a yeast aqy-null strain for heterologous expression and functional analysis of human AQP7 and investigated its regulation by pH. Using a combination of in vitro and in silico approaches, we found that AQP7 changes from fully permeable to virtually closed at acidic pH, and that Tyr135 and His165 facing the extracellular environment are crucial residues for channel permeability. Moreover, instead of reducing the pore size, the protonation of key residues changes AQP7’s protein surface electrostatic charges, which, in turn, may decrease glycerol’s binding affinity to the pore, resulting in decreased permeability. In addition, since some pH-sensitive residues are located at the monomer-monomer interface, decreased permeability may result from cooperativity between AQP7’s monomers. Considering the importance of glycerol permeation via AQP7 in multiple pathophysiological conditions, this mechanism of hAQP7 pH-regulation may help the design of selective modulators targeting aquaglyceroporin-related disorders.
Collapse
|
22
|
Song D, Liu X, Diao Y, Sun Y, Gao G, Zhang T, Chen K, Pei L. Hydrogen‑rich solution against myocardial injury and aquaporin expression via the PI3K/Akt signaling pathway during cardiopulmonary bypass in rats. Mol Med Rep 2018; 18:1925-1938. [PMID: 29956781 PMCID: PMC6072160 DOI: 10.3892/mmr.2018.9198] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 01/05/2018] [Indexed: 12/19/2022] Open
Abstract
Myocardial ischemia, hypoxia and reperfusion injury are induced by aortic occlusion, cardiac arrest and resuscitation during cardiopulmonary bypass (CPB), which can severely affect cardiac function. The aim of the present study was to investigate the effects of hydrogen-rich solution (HRS) and aquaporin (AQP) on cardiopulmonary bypass (CPB)-induced myocardial injury, and determine the mechanism of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway. Sprague Dawley rats were divided into a sham operation group, a CPB surgery group and a HRS group. A CPB model was established, and the hemodynamic parameters were determined at the termination of CPB. The myocardial tissues were observed by hematoxylin and eosin, and Masson staining. The levels of myocardial injury markers [adult cardiac troponin I (cTnI), lactate dehydrogenase (LDH), creatine kinase MB (CK-MB) and brain natriuretic peptide (BNP)], inflammatory factors [interleukin (IL)-1β, IL-6 and tumor necrosis factor-α (TNF-α)] and oxidative stress indicators [superoxide dismutase (SOD), malondialdehyde (MDA) and myeloperoxidase (MPO)] were determined by ELISA. Furthermore, H9C2 cells were treated with HRS following hypoxia/reoxygenation. Cell viability and cell apoptosis were investigated. The expression of apoptosis regulator Bcl-2 (Bcl-2), apoptosis regulator Bax (Bax), caspase 3, AQP-1, AQP-4, phosphorylated (p)-Akt, heme oxygenase 1 (HO-1) and nuclear factor erythroid 2-related factor 2 (Nrf2) were investigated using western blotting and quantitative-polymerase chain reaction of tissues and cells. Following CPB, myocardial cell arrangement was disordered, myocardial injury markers (cTnI, LDH, CK-MB and BNP), inflammatory cytokines (IL-1β, IL-6 and TNF-α) and MDA levels were significantly increased compared with the sham group; whereas the SOD levels were significantly downregulated following CPB compared with the sham group. HRS attenuated myocardial injury, reduced the expression levels of cTnI, LDH, CK-MB, BNP, IL-1β, IL-6, TNF-α, MDA and MPO, and increased SOD release. Levels of Bcl-2, AQP-1, AQP-4, p-Akt, HO-1 and Nrf2 were significantly increased following HRS; whereas Bax and caspase-3 expression levels were significantly reduced following CPB. HRS treatment significantly increased the viability of myocardial cells, reduced the rate of myocardial cell apoptosis and the release of MDA and LDH compared with the CPB group. A PI3K inhibitor (LY294002) was revealed to reverse the protective effect of HRS treatment. HRS was demonstrated to attenuate CPB-induced myocardial injury, suppress AQP-1 and AQP-4 expression following CPB treatment and protect myocardial cells via the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Dandan Song
- Department of Anesthesiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110016, P.R. China
| | - Xuelei Liu
- Department of Clinical Laboratory, General Hospital of Shenyang Military Area Command, Shenyang, Liaoning 110016, P.R. China
| | - Yugang Diao
- Department of Anesthesiology, General Hospital of Shenyang Military Area Command, Shenyang, Liaoning 110016, P.R. China
| | - Yingjie Sun
- Department of Anesthesiology, General Hospital of Shenyang Military Area Command, Shenyang, Liaoning 110016, P.R. China
| | - Guangjie Gao
- Department of Anesthesiology, The 463rd Hospital of People's Liberation Army China, Shenyang, Liaoning 110012, P.R. China
| | - Tiezheng Zhang
- Department of Anesthesiology, General Hospital of Shenyang Military Area Command, Shenyang, Liaoning 110016, P.R. China
| | - Keyan Chen
- Department of Laboratory Animal Science, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Ling Pei
- Department of Anesthesiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110016, P.R. China
| |
Collapse
|
23
|
Edhager AV, Povlsen JA, Løfgren B, Bøtker HE, Palmfeldt J. Proteomics of the Rat Myocardium during Development of Type 2 Diabetes Mellitus Reveals Progressive Alterations in Major Metabolic Pathways. J Proteome Res 2018; 17:2521-2532. [PMID: 29847139 DOI: 10.1021/acs.jproteome.8b00276] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Congestive heart failure and poor clinical outcome after myocardial infarction are known complications in patients with type-2 diabetes mellitus (T2DM). Protein alterations may be involved in the mechanisms underlying these disarrays in the diabetic heart. Here we map proteins involved in intracellular metabolic pathways in the Zucker diabetic fatty rat heart as T2DM develops using MS based proteomics. The prediabetic state only induced minor pathway changes, whereas onset and late T2DM caused pronounced perturbations. Two actin-associated proteins, ARPC2 and TPM3, were up-regulated at the prediabetic state indicating increased actin dynamics. All differentially regulated proteins involved in fatty acid metabolism, both peroxisomal and mitochondrial, were up-regulated at late T2DM, whereas enzymes of branched chain amino acid degradation were all down-regulated. At both onset and late T2DM, two members of the serine protease inhibitor superfamily, SERPINA3K and SERPINA3L, were down-regulated. Furthermore, we found alterations in proteins involved in clearance of advanced glycation end-products and lipotoxicity, DCXR and CBR1, at both onset and late T2DM. These proteins deserve elucidation with regard to their role in T2DM pathogenesis and their respective role in the deterioration of the diabetic heart. Data are available via ProteomeXchange with identifiers PXD009538, PXD009554, and PXD009555.
Collapse
Affiliation(s)
- Anders Valdemar Edhager
- Research Unit for Molecular Medicine, Department of Clinical Medicine , Aarhus University and Aarhus University Hospital , 8200 , Aarhus N , Denmark
| | | | - Bo Løfgren
- Department of Cardiology , Aarhus University Hospital , 8200 , Aarhus N , Denmark.,Institute for Experimental Clinical Research , Aarhus University , 8000 , Aarhus C , Denmark
| | - Hans Erik Bøtker
- Department of Cardiology , Aarhus University Hospital , 8200 , Aarhus N , Denmark
| | - Johan Palmfeldt
- Research Unit for Molecular Medicine, Department of Clinical Medicine , Aarhus University and Aarhus University Hospital , 8200 , Aarhus N , Denmark
| |
Collapse
|
24
|
da Silva IV, Rodrigues JS, Rebelo I, Miranda JPG, Soveral G. Revisiting the metabolic syndrome: the emerging role of aquaglyceroporins. Cell Mol Life Sci 2018; 75:1973-1988. [PMID: 29464285 PMCID: PMC11105723 DOI: 10.1007/s00018-018-2781-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 02/05/2018] [Accepted: 02/15/2018] [Indexed: 02/07/2023]
Abstract
The metabolic syndrome (MetS) includes a group of medical conditions such as insulin resistance (IR), dyslipidemia and hypertension, all associated with an increased risk for cardiovascular disease. Increased visceral and ectopic fat deposition are also key features in the development of IR and MetS, with pathophysiological sequels on adipose tissue, liver and muscle. The recent recognition of aquaporins (AQPs) involvement in adipose tissue homeostasis has opened new perspectives for research in this field. The members of the aquaglyceroporin subfamily are specific glycerol channels implicated in energy metabolism by facilitating glycerol outflow from adipose tissue and its systemic distribution and uptake by liver and muscle, unveiling these membrane channels as key players in lipid balance and energy homeostasis. Being involved in a variety of pathophysiological mechanisms including IR and obesity, AQPs are considered promising drug targets that may prompt novel therapeutic approaches for metabolic disorders such as MetS. This review addresses the interplay between adipose tissue, liver and muscle, which is the basis of the metabolic syndrome, and highlights the involvement of aquaglyceroporins in obesity and related pathologies and how their regulation in different organs contributes to the features of the metabolic syndrome.
Collapse
Affiliation(s)
- Inês Vieira da Silva
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, 1649-003, Lisbon, Portugal
- Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, 1649-003, Lisbon, Portugal
| | - Joana S Rodrigues
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, 1649-003, Lisbon, Portugal
- Department of Toxicological and Bromatological Sciences, Faculty of Pharmacy, Universidade de Lisboa, 1649-003, Lisbon, Portugal
| | - Irene Rebelo
- UCIBIO, REQUIMTE, Department of Biological Sciences, Faculty of Pharmacy, Universidade do Porto, Porto, Portugal
| | - Joana P G Miranda
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, 1649-003, Lisbon, Portugal
- Department of Toxicological and Bromatological Sciences, Faculty of Pharmacy, Universidade de Lisboa, 1649-003, Lisbon, Portugal
| | - Graça Soveral
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, 1649-003, Lisbon, Portugal.
- Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, 1649-003, Lisbon, Portugal.
| |
Collapse
|
25
|
Iena FM, Lebeck J. Implications of Aquaglyceroporin 7 in Energy Metabolism. Int J Mol Sci 2018; 19:ijms19010154. [PMID: 29300344 PMCID: PMC5796103 DOI: 10.3390/ijms19010154] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 12/29/2017] [Accepted: 12/31/2017] [Indexed: 12/14/2022] Open
Abstract
The aquaglyceroporin AQP7 is a pore-forming transmembrane protein that facilitates the transport of glycerol across cell membranes. Glycerol is utilized both in carbohydrate and lipid metabolism. It is primarily stored in white adipose tissue as part of the triglyceride molecules. During states with increased lipolysis, such as fasting and diabetes, glycerol is released from adipose tissue and metabolized in other tissues. AQP7 is expressed in adipose tissue where it facilitates the efflux of glycerol, and AQP7 deficiency has been linked to increased glycerol kinase activity and triglyceride accumulation in adipose tissue, leading to obesity and secondary development of insulin resistance. However, AQP7 is also expressed in a wide range of other tissues, including kidney, muscle, pancreatic β-cells and liver, where AQP7 also holds the potential to influence whole body energy metabolism. The aim of the review is to summarize the current knowledge on AQP7 in adipose tissue, as well as AQP7 expressed in other tissues where AQP7 might play a significant role in modulating whole body energy metabolism.
Collapse
Affiliation(s)
- Francesco Maria Iena
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Allé 3, 8000 Aarhus, Denmark.
| | - Janne Lebeck
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Allé 3, 8000 Aarhus, Denmark.
| |
Collapse
|
26
|
Arias-Hidalgo M, Al-Samir S, Weber N, Geers-Knörr C, Gros G, Endeward V. CO 2 permeability and carbonic anhydrase activity of rat cardiomyocytes. Acta Physiol (Oxf) 2017; 221:115-128. [PMID: 28429509 DOI: 10.1111/apha.12887] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 04/11/2017] [Accepted: 04/14/2017] [Indexed: 11/30/2022]
Abstract
AIM To determine the CO2 permeability (PCO2 ) of plasma membranes of cardiomyocytes. These cells were chosen because heart possesses the highest rate of O2 consumption/CO2 production in the body. METHODS Cardiomyocytes were isolated from rat hearts using the Langendorff technique. Cardiomyocyte suspensions exhibited a vitality of 2-14% and were studied by the previously described mass spectrometric 18 O-exchange technique deriving PCO2 . We showed by mass spectrometry and by carbonic anhydrase (CA) staining that non-vital cardiomyocytes are free of CA and thus do not contribute to the mass spectrometric signal, which is determined exclusively by the fully functional vital cardiomyocytes. RESULTS Lysed cardiomyocytes yielded an intracellular CA activity for vital cells of 5070; that is, the rate of CO2 hydration inside the cell is accelerated 5071-fold. Using this number, analyses of the mass spectrometric recordings from cardiomyocyte suspensions yield a PCO2 of 0.10 cm s-1 (SD ± 0.06, n = 15) at 37 °C. CONCLUSION In comparison with the PCO2 of other cells, this value is quite high and about identical to that of the human red cell membrane. As no major protein CO2 channels such as aquaporins 1 and 4 are present in rat cardiac sarcolemma, the high PCO2 of this membrane is likely due to its low cholesterol content of about 0.2 (mol cholesterol)·(mol total membrane lipids)-1 . Previous work predicted a PCO2 of ≥0.1 cm s-1 from this level of cholesterol. We conclude that the low cholesterol establishes a PCO2 high enough to render the membrane resistance to CO2 diffusion almost negligible, even under conditions of maximal O2 consumption of the heart.
Collapse
Affiliation(s)
- M. Arias-Hidalgo
- Molekular- und Zellphysiologie and AG Vegetative Physiologie; Medizinische Hochschule Hannover; Hannover Germany
| | - S. Al-Samir
- Molekular- und Zellphysiologie and AG Vegetative Physiologie; Medizinische Hochschule Hannover; Hannover Germany
| | - N. Weber
- Molekular- und Zellphysiologie and AG Vegetative Physiologie; Medizinische Hochschule Hannover; Hannover Germany
| | - C. Geers-Knörr
- Molekular- und Zellphysiologie and AG Vegetative Physiologie; Medizinische Hochschule Hannover; Hannover Germany
| | - G. Gros
- Molekular- und Zellphysiologie and AG Vegetative Physiologie; Medizinische Hochschule Hannover; Hannover Germany
| | - V. Endeward
- Molekular- und Zellphysiologie and AG Vegetative Physiologie; Medizinische Hochschule Hannover; Hannover Germany
| |
Collapse
|
27
|
Sutka M, Amodeo G, Ozu M. Plant and animal aquaporins crosstalk: what can be revealed from distinct perspectives. Biophys Rev 2017; 9:545-562. [PMID: 28871493 DOI: 10.1007/s12551-017-0313-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 08/02/2017] [Indexed: 01/03/2023] Open
Abstract
Aquaporins (AQPs) can be revisited from a distinct and complementary perspective: the outcome from analyzing them from both plant and animal studies. (1) The approach in the study. Diversity found in both kingdoms contrasts with the limited number of crystal structures determined within each group. While the structure of almost half of mammal AQPs was resolved, only a few were resolved in plants. Strikingly, the animal structures resolved are mainly derived from the AQP2-lineage, due to their important roles in water homeostasis regulation in humans. The difference could be attributed to the approach: relevance in animal research is emphasized on pathology and in consequence drug screening that can lead to potential inhibitors, enhancers and/or regulators. By contrast, studies on plants have been mainly focused on the physiological role that AQPs play in growth, development and stress tolerance. (2) The transport capacity. Besides the well-described AQPs with high water transport capacity, large amount of evidence confirms that certain plant AQPs can carry a large list of small solutes. So far, animal AQP list is more restricted. In both kingdoms, there is a great amount of evidence on gas transport, although there is still an unsolved controversy around gas translocation as well as the role of the central pore of the tetramer. (3) More roles than expected. We found it remarkable that the view of AQPs as specific channels has evolved first toward simple transporters to molecules that can experience conformational changes triggered by biochemical and/or mechanical signals, turning them also into signaling components and/or behave as osmosensor molecules.
Collapse
Affiliation(s)
- Moira Sutka
- Departamento de Biodiversidad y Biología Experimental, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires e Instituto de Biodiversidad y Biología Experimental, Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Gabriela Amodeo
- Departamento de Biodiversidad y Biología Experimental, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires e Instituto de Biodiversidad y Biología Experimental, Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina.
| | - Marcelo Ozu
- Departamento de Biodiversidad y Biología Experimental, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires e Instituto de Biodiversidad y Biología Experimental, Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina.
| |
Collapse
|
28
|
Abstract
Obesity is one of the most important metabolic disorders of this century and is associated with a cluster of the most dangerous cardiovascular disease risk factors, such as insulin resistance and diabetes , dyslipidemia and hypertension , collectively named Metabolic Syndrome. The role of aquaporins in glycerol metabolism facilitating glycerol release from the adipose tissue and distribution to various tissues and organs, unveils these membrane channels as important players in lipid balance and energy homeostasis and points to their involvement in a variety of pathophysiological mechanisms including insulin resistance, obesity and diabetes.This review summarizes the physiologic role of aquaglyceroporins in glycerol metabolism and lipid homeostasis, describing their specific tissue distribution, their involvement in glycerol balance and their implication in obesity and fat-related metabolic complications. The development of specify pharmacologic modulators able to regulate aquaglyceroporins expression and function , in particular AQP7 in adipose tissue, might constitute a novel approach for controlling obesity and other metabolic disorders.
Collapse
Affiliation(s)
- Inês Vieira da Silva
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, 1649-003, Portugal
- Department of Bioquimica e Biologia Humana, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Graça Soveral
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, 1649-003, Portugal.
- Department of Bioquimica e Biologia Humana, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
29
|
Netti VA, Iovane AN, Vatrella MC, Zotta E, Fellet AL, Balaszczuk AM. Effects of nitric oxide system and osmotic stress on Aquaporin-1 in the postnatal heart. Biomed Pharmacother 2016; 81:225-234. [DOI: 10.1016/j.biopha.2016.03.050] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 03/31/2016] [Accepted: 03/31/2016] [Indexed: 11/25/2022] Open
|
30
|
Investigation of the effects of aging on the expression of aquaporin 1 and aquaporin 4 protein in heart tissue. Anatol J Cardiol 2016; 17:18-23. [PMID: 27443479 PMCID: PMC5324856 DOI: 10.14744/anatoljcardiol.2016.7033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Objective: Aquaporin (AQP) 1 and AQP 4 are expressed in human heart and several studies have been focused on these two aquaporins. For this purpose, the present study is aimed to research the effects of aging on AQP 1 and AQP 4 in heart tissue. Methods: In this study, 14 Balb/C type white mice were used. Animals were divided into two equal groups. Group I consisted of 2-month-old young animals (n=7), and group II consisted of 18-month-old animals (n=7). To determine the AQP1 and AQP4 expression in the myocardium, the heart tissue was removed to perform western blotting and immunohistochemical and histopathological evaluations. Results: Muscle fibers of the heart in aged animals were more irregular and loosely organized in hematoxylin–eosin (H&E) stained sections. H-score analysis revealed that AQP1 and AQP4 immunoreactivity significantly increased in heart tissues of old mice compared with those of young mice (p<0.001). In addition, AQP1 and AQP4 protein expressions in the tissues of old animals were increased significantly according to western blot analysis (p=0.018 and p<0.001 for AQP1 and AQP4, respectively). Conclusion: Increased AQP1 and AQP4 levels in the heart tissue may be correlated with the maintenance of water and electrolytes balance, which decreases with aging. In this context, it might be the result of a compensatory response to decreased AQP4 functions. In addition, this increase with aging as demonstrated in our study might be one of the factors that increases the tendency of ischemia in elder people.
Collapse
|
31
|
Palabiyik O, Karaca A, Taştekin E, Yamasan BE, Tokuç B, Sipahi T, Vardar SA. The Effect of a High-Protein Diet and Exercise on Cardiac AQP7 and GLUT4 Gene Expression. Biochem Genet 2016; 54:731-45. [PMID: 27294385 DOI: 10.1007/s10528-016-9753-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 06/08/2016] [Indexed: 12/18/2022]
Abstract
High-protein (HP) diets are commonly consumed by athletes despite their potential health hazard, which is postulated to enforce a negative effect on bone and renal health. However, its effects on heart have not been known yet. Aquaporin-7 (AQP7) is an aquaglyceroporin that facilitates glycerol and water transport. Glycerol is an important cardiac energy production substrate, especially during exercise, in conjunction with fatty acids and glucose. Glucose transporter 4 (GLUT4) is an insulin-sensitive glucose transporter in heart. We aimed to investigate the effect of HPD on AQP7 and GLUT4 levels in the rat heart subjected to exercise. Male Sprague-Dawley rats were divided into control (n = 12), exercise (E) training (n = 10), HPD (n = 12), and HPD-E training (n = 9) groups. The HPD groups were fed a 45 % protein-containing diet 5 weeks. The HPD-E and E groups were performed the treadmill exercise during the 5-week study period. Real-time polymerase chain reaction and immunohistochemistry techniques were used to determine the gene expression and localization of AQP7 and GLUT4 in heart tissue. Results of relative gene expression were calculated by the 'Pfaffl' mathematical method using the REST program. Differences in AQP7 and GLUT4 gene expression were expressed as fold change compared to the control group. Heart weight/tibia ratio and ventricular wall thickness were evaluated as markers of cardiac hypertrophy. Further, serum glucose, glycerol, and insulin levels were also measured. AQP7 gene expression was found to be increased in the E (3.47-fold, p < 0.001), HPD (5.59-fold, p < 0.001), and HPD-E (3.87-fold, p < 0.001) groups compared to the control group. AQP7 protein expression was also increased in the HPD and HPD-E groups (p < 0.001). Additionally, cardiac mRNA expression levels of GLUT4 showed a significant increase in the E (2.16-fold, p < 0.003), HPD (7.14-fold, p < 0.001), and HPD-E (3.43-fold, p < 0.001) groups compared to the control group. GLUT4 protein expression was significantly increased in the E, HPD, and HPD-E groups compared to the control group (p = 0.024, p < 0.001, and p < 0.001, respectively). Furthermore, Serum glucose levels were significantly different between groups (p < 0.005). This difference was observed between the HPD groups and normal-protein diet groups (C and E). Serum insulin levels were higher for HPD groups compared with the normal-protein diet groups (p < 0.001), whereas no differences were observed between the exercise and sedentary groups (p = 0.111). Serum glycerol levels were significantly increased in the HPD groups compared with control and E groups (p < 0.05 and p < 0.05, respectively). Consumption of HPD supplementation caused the increased effects on AQP7 and GLUT4 expression in rat heart.
Collapse
Affiliation(s)
- Orkide Palabiyik
- Department of Biophysics, Faculty of Medicine, Trakya University, Edirne, Turkey.
| | - Aziz Karaca
- Department of Physiology, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Ebru Taştekin
- Department of Pathology, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Bilge Eren Yamasan
- Department of Biophysics, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Burcu Tokuç
- Department of Public Health, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Tammam Sipahi
- Department of Biophysics, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Selma Arzu Vardar
- Department of Physiology, Faculty of Medicine, Trakya University, Edirne, Turkey
| |
Collapse
|
32
|
Laforenza U, Bottino C, Gastaldi G. Mammalian aquaglyceroporin function in metabolism. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1858:1-11. [PMID: 26456554 DOI: 10.1016/j.bbamem.2015.10.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 10/05/2015] [Accepted: 10/07/2015] [Indexed: 11/26/2022]
Abstract
Aquaglyceroporins are integral membrane proteins that are permeable to glycerol as well as water. The movement of glycerol from a tissue/organ to the plasma and vice versa requires the presence of different aquaglyceroporins that can regulate the entrance or the exit of glycerol across the plasma membrane. Actually, different aquaglyceroporins have been discovered in the adipose tissue, small intestine, liver, kidney, heart, skeletal muscle, endocrine pancreas and capillary endothelium, and their differential expression could be related to obesity and the type 2 diabetes. Here we describe the expression and function of different aquaglyceroporins in physiological condition and in obesity and type 2 diabetes, suggesting they are potential therapeutic targets for metabolic disorders.
Collapse
Affiliation(s)
| | - Cinzia Bottino
- Department of Molecular Medicine, University of Pavia, Italy
| | - Giulia Gastaldi
- Department of Molecular Medicine, University of Pavia, Italy
| |
Collapse
|
33
|
Aquaporin-1 Deficiency Protects Against Myocardial Infarction by Reducing Both Edema and Apoptosis in Mice. Sci Rep 2015; 5:13807. [PMID: 26348407 PMCID: PMC4562302 DOI: 10.1038/srep13807] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 08/06/2015] [Indexed: 01/26/2023] Open
Abstract
Many studies have determined that AQP1 plays an important role in edema formation and resolution in various tissues via water transport across the cell membrane. The aim of this research was to determine both if and how AQP1 is associated with cardiac ischemic injury, particularly the development of edema following myocardial infarction (MI). AQP1+/+ and AQP1−/− mice were used to create the MI model. Under physiological conditions, AQP1−/− mice develop normally; however, in the setting of MI, they exhibit cardioprotective properties, as shown by reduced cardiac infarct size determined via NBT staining, improved cardiac function determined via left ventricular catheter measurements, decreased AQP1-dependent myocardial edema determined via water content assays, and decreased apoptosis determined via TUNEL analysis. Cardiac ischemia caused by hypoxia secondary to AQP1 deficiency stabilized the expression of HIF-1α in endothelial cells and subsequently decreased microvascular permeability, resulting in the development of edema. The AQP1-dependent myocardial edema and apoptosis contributed to the development of MI. AQP1 deficiency protected cardiac function from ischemic injury following MI. Furthermore, AQP1 deficiency reduced microvascular permeability via the stabilization of HIF-1α levels in endothelial cells and decreased cellular apoptosis following MI.
Collapse
|
34
|
Méndez-Giménez L, Rodríguez A, Balaguer I, Frühbeck G. Role of aquaglyceroporins and caveolins in energy and metabolic homeostasis. Mol Cell Endocrinol 2014; 397:78-92. [PMID: 25008241 DOI: 10.1016/j.mce.2014.06.017] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 06/25/2014] [Accepted: 06/26/2014] [Indexed: 12/23/2022]
Abstract
Aquaglyceroporins and caveolins are submicroscopic integral membrane proteins that are particularly abundant in many mammalian cells. Aquaglyceroporins (AQP3, AQP7, AQP9 and AQP10) encompass a subfamily of aquaporins that allow the movement of water, but also of small solutes, such as glycerol, across cell membranes. Glycerol constitutes an important metabolite as a substrate for de novo synthesis of triacylglycerols and glucose as well as an energy substrate to produce ATP via the mitochondrial oxidative phosphorylation. In this sense, the control of glycerol influx/efflux in metabolic organs by aquaglyceroporins plays a crucial role with the dysregulation of these glycerol channels being associated with metabolic diseases, such as obesity, insulin resistance, non-alcoholic fatty liver disease and cardiac hypertrophy. On the other hand, caveolae have emerged as relevant plasma membrane sensors implicated in a wide range of cellular functions, including endocytosis, apoptosis, cholesterol homeostasis, proliferation and signal transduction. Caveolae-coating proteins, namely caveolins and cavins, can act as scaffolding proteins within caveolae by concentrating signaling molecules involved in free fatty acid and cholesterol uptake, proliferation, insulin signaling or vasorelaxation, among others. The importance of caveolae in whole-body homeostasis is highlighted by the link between homozygous mutations in genes encoding caveolins and cavins with metabolic diseases, such as lipodystrophy, dyslipidemia, muscular dystrophy and insulin resistance in rodents and humans. The present review focuses on the role of aquaglyceroporins and caveolins on lipid and glucose metabolism, insulin secretion and signaling, energy production and cardiovascular homeostasis, outlining their potential relevance in the development and treatment of metabolic diseases.
Collapse
Affiliation(s)
- Leire Méndez-Giménez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Pamplona, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Pamplona, Spain.
| | - Inmaculada Balaguer
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Pamplona, Spain; Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain.
| |
Collapse
|
35
|
Rutkovskiy A, Mariero LH, Vaage J. Deletion of the aquaporin-4 gene alters expression and phosphorylation of protective kinases in the mouse heart. Scandinavian Journal of Clinical and Laboratory Investigation 2014; 74:500-5. [PMID: 24792367 DOI: 10.3109/00365513.2014.905698] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
AIM Aquaporins are channel-forming proteins highly permeable to water and some small molecular solutes. We have previously shown that aquaporin-4 knockout mice have increased tolerance to ischemia. However, the mechanism of cardioprotection was unclear. The aim of the current study was to investigate the effects of aquaporin-4 deletion on baseline expression and phosphorylation of some cardioprotective protein kinases. METHODS Proteins were extracted from hearts of aquaporin-4 knockout mice and littermate wild-type controls and analyzed with Western blot. Samples were taken from young (≤ 6 months of age), and old (≥ 1 year) mice. RESULTS Western blots showed three different isoforms of aquaporin-4 in wild types, likely representing M1, M23, and Mz. Total AMP-dependent kinase expression was decreased in aquaporin-4 knockout hearts by 18 ± 13% (p = 0.02), while the expression of Akt kinase, extracellular signal regulated kinase 1/2, protein kinase C-epsilon, mitogen-associated kinase P38 and C-Jun N-terminal kinase was unchanged. The phosphorylation of Akt kinase was reduced in hearts from knockout mice by 41 ± 16% (p = 0.01). No other alterations in phosphorylation were found. These effects were only detected in young mice. CONCLUSION Deletion of the aquaporin-4 gene decreased AMP-dependent kinase expression and Akt kinase phosphorylation in the heart. These changes may influence energy metabolism and endogenous cardioprotection.
Collapse
Affiliation(s)
- Arkady Rutkovskiy
- Department of Emergency and Intensive Care at the Institute of Clinical Medicine , Norway
| | | | | |
Collapse
|
36
|
De Mello WC. Angiotensin (1–7) re-establishes heart cell communication previously impaired by cell swelling: Implications for myocardial ischemia. Exp Cell Res 2014; 323:359-65. [DOI: 10.1016/j.yexcr.2014.03.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 03/06/2014] [Accepted: 03/09/2014] [Indexed: 11/28/2022]
|
37
|
Transplantation of Mesenchymal Cells Improves Peripheral Limb Ischemia in Diabetic Rats. Mol Biotechnol 2014; 56:438-48. [DOI: 10.1007/s12033-014-9735-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
38
|
Abstract
Aquaporins are a group of proteins with high-selective permeability for water. A subgroup called aquaglyceroporins is also permeable to glycerol, urea and a few other solutes. Aquaporin function has mainly been studied in the brain, kidney, glands and skeletal muscle, while the information about aquaporins in the heart is still scarce. The current review explores the recent advances in this field, bringing aquaporins into focus in the context of myocardial ischemia, reperfusion, and blood osmolarity disturbances. Since the amount of data on aquaporins in the heart is still limited, examples and comparisons from better-studied areas of aquaporin biology have been used. The human heart expresses aquaporin-1, -3, -4 and -7 at the protein level. The potential roles of aquaporins in the heart are discussed, and some general phenomena that the myocardial aquaporins share with aquaporins in other organs are elaborated. Cardiac aquaporin-1 is mostly distributed in the microvasculature. Its main role is transcellular water flux across the endothelial membranes. Aquaporin-4 is expressed in myocytes, both in cardiac and in skeletal muscle. In addition to water flux, its function is connected to the calcium signaling machinery. It may play a role in ischemia-reperfusion injury. Aquaglyceroporins, especially aquaporin-7, may serve as a novel pathway for nutrient delivery into the heart. They also mediate toxicity of various poisons. Aquaporins cannot influence permeability by gating, therefore, their function is regulated by changes of expression-on the levels of transcription, translation (by microRNAs), post-translational modification, membrane trafficking, ubiquitination and subsequent degradation. Studies using mice genetically deficient for aquaporins have shown rather modest changes in the heart. However, they might still prove to be attractive targets for therapy directed to reduce myocardial edema and injury caused by ischemia and reperfusion.
Collapse
|
39
|
Day RE, Kitchen P, Owen DS, Bland C, Marshall L, Conner AC, Bill RM, Conner MT. Human aquaporins: regulators of transcellular water flow. Biochim Biophys Acta Gen Subj 2013; 1840:1492-506. [PMID: 24090884 DOI: 10.1016/j.bbagen.2013.09.033] [Citation(s) in RCA: 196] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 09/19/2013] [Accepted: 09/23/2013] [Indexed: 02/06/2023]
Abstract
BACKGROUND Emerging evidence supports the view that (AQP) aquaporin water channels are regulators of transcellular water flow. Consistent with their expression in most tissues, AQPs are associated with diverse physiological and pathophysiological processes. SCOPE OF REVIEW AQP knockout studies suggest that the regulatory role of AQPs, rather than their action as passive channels, is their critical function. Transport through all AQPs occurs by a common passive mechanism, but their regulation and cellular distribution varies significantly depending on cell and tissue type; the role of AQPs in cell volume regulation (CVR) is particularly notable. This review examines the regulatory role of AQPs in transcellular water flow, especially in CVR. We focus on key systems of the human body, encompassing processes as diverse as urine concentration in the kidney to clearance of brain oedema. MAJOR CONCLUSIONS AQPs are crucial for the regulation of water homeostasis, providing selective pores for the rapid movement of water across diverse cell membranes and playing regulatory roles in CVR. Gating mechanisms have been proposed for human AQPs, but have only been reported for plant and microbial AQPs. Consequently, it is likely that the distribution and abundance of AQPs in a particular membrane is the determinant of membrane water permeability and a regulator of transcellular water flow. GENERAL SIGNIFICANCE Elucidating the mechanisms that regulate transcellular water flow will improve our understanding of the human body in health and disease. The central role of specific AQPs in regulating water homeostasis will provide routes to a range of novel therapies. This article is part of a Special Issue entitled Aquaporins.
Collapse
Affiliation(s)
- Rebecca E Day
- Biomedical Research Centre, Sheffield Hallam University, Howard Street, Sheffield S1 1WB, UK
| | - Philip Kitchen
- Molecular Organisation and Assembly in Cells Doctoral Training Centre, University of Warwick, Coventry CV4 7AL, UK
| | - David S Owen
- Biomedical Research Centre, Sheffield Hallam University, Howard Street, Sheffield S1 1WB, UK
| | - Charlotte Bland
- School of Life and Health Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Lindsay Marshall
- School of Life and Health Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Alex C Conner
- School of Clinical and Experimental Medicine, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| | - Roslyn M Bill
- School of Life and Health Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK.
| | - Matthew T Conner
- Biomedical Research Centre, Sheffield Hallam University, Howard Street, Sheffield S1 1WB, UK.
| |
Collapse
|
40
|
Chai RC, Jiang JH, Wong AYK, Jiang F, Gao K, Vatcher G, Hoi Yu AC. AQP5 is differentially regulated in astrocytes during metabolic and traumatic injuries. Glia 2013; 61:1748-65. [PMID: 23922257 DOI: 10.1002/glia.22555] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 05/30/2013] [Accepted: 06/17/2013] [Indexed: 01/14/2023]
Abstract
Water movement plays vital roles in both physiological and pathological conditions in the brain. Astrocytes are responsible for regulating this water movement and are the major contributors to brain edema in pathological conditions. Aquaporins (AQPs) in astrocytes play critical roles in the regulation of water movement in the brain. AQP1, 3, 4, 5, 8, and 9 have been reported in the brain. Compared with AQP1, 4, and 9, AQP3, 5, and 8 are less studied. Among the lesser known AQPs, AQP5, which has multiple functions identified outside the central nervous system, is also indicated to be involved in hypoxia injury in astrocytes. In our study, AQP5 expression could be detected both in primary cultures of astrocytes and neurons, and AQP5 expression in astrocytes was confirmed in 1- to 4-week old primary cultures of astrocytes. AQP5 was localized on the cytoplasmic membrane and in the cytoplasm of astrocytes. AQP5 expression was downregulated during ischemia treatment and upregulated after scratch-wound injury, which was also confirmed in a middle cerebral artery occlusion model and a stab-wound injury model in vivo. The AQP5 increased after scratch injury was polarized to the migrating processes and cytoplasmic membrane of astrocytes in the leading edge of the scratch-wound, and AQP5 over-expression facilitated astrocyte process elongation after scratch injury. Taken together, these results indicate that AQP5 might be an important water channel in astrocytes that is differentially expressed during various brain injuries.
Collapse
Affiliation(s)
- Rui Chao Chai
- Neuroscience Research Institute & Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | | | | | | | | | | | | |
Collapse
|
41
|
Genetic deletion of aquaporin-1 results in microcardia and low blood pressure in mouse with intact nitric oxide-dependent relaxation, but enhanced prostanoids-dependent relaxation. Pflugers Arch 2013; 466:237-51. [PMID: 23873354 DOI: 10.1007/s00424-013-1325-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Revised: 06/27/2013] [Accepted: 06/28/2013] [Indexed: 12/21/2022]
Abstract
The water channels, aquaporins (AQPs) are key mediators of transcellular fluid transport. However, their expression and role in cardiac tissue is poorly characterized. Particularly, AQP1 was suggested to transport other molecules (nitric oxide (NO), hydrogen peroxide (H2O2)) with potential major bearing on cardiovascular physiology. We therefore examined the expression of all AQPs and the phenotype of AQP1 knockout mice (vs. wild-type littermates) under implanted telemetry in vivo, as well as endothelium-dependent relaxation in isolated aortas and resistance vessels ex vivo. Four aquaporins were expressed in wild-type heart tissue (AQP1, AQP7, AQP4, AQP8) and two aquaporins in aortic and mesenteric vessels (AQP1-AQP7). AQP1 was expressed in endothelial as well as cardiac and vascular muscle cells and co-segregated with caveolin-1. AQP1 knockout (KO) mice exhibited a prominent microcardia and decreased myocyte transverse dimensions despite no change in capillary density. Both male and female AQP1 KO mice had lower mean BP, which was not attributable to altered water balance or autonomic dysfunction (from baroreflex and frequency analysis of BP and HR variability). NO-dependent BP variability was unperturbed. Accordingly, endothelium-derived hyperpolarizing factor (EDH(F)) or NO-dependent relaxation were unchanged in aorta or resistance vessels ex vivo. However, AQP1 KO mesenteric vessels exhibited an increase in endothelial prostanoids-dependent relaxation, together with increased expression of COX-2. This enhanced relaxation was abrogated by COX inhibition. We conclude that AQP1 does not regulate the endothelial EDH or NO-dependent relaxation ex vivo or in vivo, but its deletion decreases baseline BP together with increased prostanoids-dependent relaxation in resistance vessels. Strikingly, this was associated with microcardia, unrelated to perturbed angiogenesis. This may raise interest for new inhibitors of AQP1 and their use to treat hypertrophic cardiac remodeling.
Collapse
|
42
|
Nakayama T, Oishi K. Influence of coffee (Coffea arabica) and galacto-oligosaccharide consumption on intestinal microbiota and the host responses. FEMS Microbiol Lett 2013; 343:161-8. [PMID: 23551139 DOI: 10.1111/1574-6968.12142] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 03/24/2013] [Accepted: 03/25/2013] [Indexed: 12/14/2022] Open
Abstract
Although studies have reported numerous effects of coffee on human health, few studies have examined its specific effects on gut microbiota. This study aimed to clarify the influence of coffee and galacto-oligosaccharide (GOS) consumption on gut microbiota and host responses. After mice consumed coffee and GOS, their intestines were sampled, and the bacterial counts were measured with quantitative RT-PCR. Results showed that GOS consumption significantly increased total bacteria counts in the proximal colon. Although Escherichia coli and Clostridium spp. counts significantly decreased in the proximal colon, Bifidobacterium spp. counts increased remarkably in the same area. A bacterial growth inhibition assay was also conducted, and the results showed that E. coli growth was inhibited only by a coffee agar. Host responses were also investigated, revealing that coffee and GOS consumption remarkably increased aquaporin8 expression in the proximal colon. In conclusion, coffee has antibiotic effects, and GOS significantly decreased E. coli and Clostridium spp. counts, but increased Bifidobacterium spp. counts remarkably. Aquaporin8 expression was also increased with a mixture of coffee and GOS consumption. This is the first study to demonstrate that coffee consumption can regulate gut microbiota and increase aquaporin8, both of which are necessary for maintaining intestinal balance.
Collapse
Affiliation(s)
- Tatsuya Nakayama
- International Research Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.
| | | |
Collapse
|
43
|
Netti VA, Vatrella MC, Chamorro MF, Rosón MI, Zotta E, Fellet AL, Balaszczuk AM. Comparison of cardiovascular aquaporin-1 changes during water restriction between 25- and 50-day-old rats. Eur J Nutr 2013; 53:287-95. [DOI: 10.1007/s00394-013-0527-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 04/17/2013] [Indexed: 11/24/2022]
|
44
|
Zhang HZ, Kim MH, Lim JH, Bae HR. Time-dependent expression patterns of cardiac aquaporins following myocardial infarction. J Korean Med Sci 2013; 28:402-8. [PMID: 23487012 PMCID: PMC3594604 DOI: 10.3346/jkms.2013.28.3.402] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 01/04/2013] [Indexed: 01/06/2023] Open
Abstract
Aquaporins (AQPs) are expressed in myocardium and the implication of AQPs in myocardial water balance has been suggested. We investigated the expression patterns of AQP subtypes in normal myocardium and their changes in the process of edema formation and cardiac dysfunction following myocardial infarction (MI). Immunostaining demonstrated abundant expression of AQP1, AQP4, and AQP6 in normal mouse heart; AQP1 in blood vessels and cardiac myocytes, AQP4 exclusively on the intercalated discs between cardiac myocytes and AQP6 inside the myocytes. However, neither AQP7 nor AQP9 proteins were expressed in CD1 mouse myocardium. Echocardiography revealed that cardiac function was reduced at 1 week and recovered at 4 weeks after MI, whereas myocardial water content determined by wet-to-dry weight ratio increased at 1 week and rather reduced below the normal at 4 weeks. The expression of cardiac AQPs was up-regulated in MI-induced groups compared with sham-operated control group, but their time-dependent patterns were different. The time course of AQP4 expression coincided with that of myocardial edema and cardiac dysfunction following MI. However, expression of both AQP1 and AQP6 increased persistently up to 4 weeks. Our findings suggest a different role for cardiac AQPs in the formation and reabsorption of myocardial edema after MI.
Collapse
Affiliation(s)
- Hong Zhe Zhang
- Department of Cardiology, Dong-A University College of Medicine, Busan, Korea
| | - Moo Hyun Kim
- Department of Cardiology, Dong-A University College of Medicine, Busan, Korea
| | - Ju Hyun Lim
- Department of Physiology, Dong-A University College of Medicine, Busan, Korea
| | - Hae-Rahn Bae
- Department of Physiology, Dong-A University College of Medicine, Busan, Korea
| |
Collapse
|
45
|
Yan Y, Huang J, Ding F, Mei J, Zhu J, Liu H, Sun K. Aquaporin 1 plays an important role in myocardial edema caused by cardiopulmonary bypass surgery in goat. Int J Mol Med 2013; 31:637-43. [PMID: 23292298 DOI: 10.3892/ijmm.2013.1228] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 12/03/2012] [Indexed: 11/06/2022] Open
Abstract
Myocardial stunning, which is closely related to myocardial edema, is a severe complication that may occur following cardiac surgery. In this study, we examined the expression of aquaporin 1 (AQP1) and Connexin 43 (Cx43) following cardiopulmonary bypass (CPB) surgery in goats. We assessed myocardial muscle tissue water content according to changes in dry-wet weight. Our results showed that AQP1 expression and myocardial muscle tissue water content increased significantly 6 h after CPB surgery, reaching peak levels 48 h after surgery; additionally, the protein expression of Cx43 was inversely correlated with AQP1 expression. Overexpression of AQP1 during CPB surgery enhanced the degree of myocardial edema, whereas the addition of water channel protein inhibitor Hg2+ in cold crystalloid cardioplegia and knockdown of AQP1 during surgery weakened the degree of myocardial edema. These findings revealed that the severity of myocardial edema after CPB surgery is correlated with AQP1 protein expression levels, suggesting the important role played by AQP1 protein in the regulation of Cx43 in the pathological progression of myocardial edema.
Collapse
Affiliation(s)
- Yumei Yan
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200092, PR China
| | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Visceral fat accumulation is located upstream of metabolic syndrome. Recent progress in adipocyte biology has clarified the molecular mechanism for pathophysiology of metabolic syndrome and its related disorders. In this review we summarize adiponectin and aquaporin 7 (AQP7) in the role of metabolic syndrome and cardiovascular diseases.
Collapse
Affiliation(s)
- Norikazu Maeda
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan.
| | | | | |
Collapse
|
47
|
Rutkovskiy A, Bliksøen M, Hillestad V, Amin M, Czibik G, Valen G, Vaage J, Amiry-Moghaddam M, Stensløkken KO. Aquaporin-1 in cardiac endothelial cells is downregulated in ischemia, hypoxia and cardioplegia. J Mol Cell Cardiol 2012; 56:22-33. [PMID: 23238222 DOI: 10.1016/j.yjmcc.2012.12.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Revised: 11/08/2012] [Accepted: 12/03/2012] [Indexed: 10/27/2022]
Abstract
Aquaporin-1 (AQP1) is expressed in human and mouse hearts, but little is known about its cellular and subcellular localization and regulation. The aim of this study was to investigate the localization of AQP1 in the mouse heart and to determine the effects of ischemia and hypoxia on its expression. Mouse myocardial cells were freshly isolated and split into cardiomyocyte and non-cardiomyocyte fractions. Isolated, Langendorff-perfused C57Bl6 mouse hearts (n=46) were harvested with no intervention, subjected to 35min of ischemia or ischemia followed by 60min of reperfusion. Eleven mouse hearts were perfusion-fixed for electron microscopy. Forty C57Bl6 mice were exposed to normobaric hypoxia for one or two weeks (n=12). Needle biopsies of human left ventricular myocardium were sampled (n=30) during coronary artery bypass surgery before cardioplegia and after 30min of reperfusion. Human umbilical vein endothelial cells (HUVECs) were subjected to 4h of hypoxia with reoxygenation for either 4 or 24h. AQP1 expression was studied by electron microscopy with immunogold labeling, Western blot, and qPCR. Expression of miR-214 and miR-320 in HUVECs with hypoxia was studied with qPCR. HUVECs were then transfected with precursors and inhibitors of miR-214. AQP1 expression was confined to cardiac endothelial cells, with no signal in cardiomyocytes or cardiac fibroblasts. Immunogold electron microscopy showed AQP1 expression in endothelial caveolae with equal distribution along the basal and apical membranes. Ischemia and reperfusion tended to decrease AQP1 mRNA expression in mouse hearts by 37±9% (p=0.06), while glycosylated AQP1 protein was reduced by 16±9% (p=0.03). No difference in expression was found between ischemia alone and ischemia-reperfusion. In human left ventricles AQP1 mRNA expression was reduced following cardioplegia and reperfusion (p=0.008). Hypoxia in mice reduced AQP1 mRNA expression by 20±7% (p<0.0001), as well as both glycosylated (-47±10%, p=0.03) and glycan-free protein (-34±16%, p=0.05). Hypoxia and reoxygenation in HUVECs downregulated glycan-free AQP1 protein (-34±24%, p=0.04) and upregulated miR-214 (+287±52%, p<0.05). HUVECs transfected with anti-miR-214 had increased glycosylated (1.5 fold) and glycan-free (2 fold) AQP1. AQP1 in mouse hearts is localized to endothelial cell membranes and caveolae. Cardioplegia, ischemia and hypoxia decrease AQP1 mRNA as well as total protein expression and glycosylation, possibly regulated by miR-214.
Collapse
Affiliation(s)
- Arkady Rutkovskiy
- Department of Emergency and Intensive Care at the Institute of Clinical Medicine, University of Oslo, Postbox 1171 Blindern, 0318 Oslo, Norway.
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Maeda N. Implications of aquaglyceroporins 7 and 9 in glycerol metabolism and metabolic syndrome. Mol Aspects Med 2012; 33:665-75. [DOI: 10.1016/j.mam.2012.02.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Revised: 02/28/2012] [Accepted: 02/28/2012] [Indexed: 10/28/2022]
|
49
|
Ding FB, Yan YM, Huang JB, Mei J, Zhu JQ, Liu H. The involvement of AQP1 in heart oedema induced by global myocardial ischemia. Cell Biochem Funct 2012; 31:60-4. [PMID: 22865611 DOI: 10.1002/cbf.2860] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Revised: 05/28/2012] [Accepted: 07/05/2012] [Indexed: 11/06/2022]
Affiliation(s)
- Fang-Bao Ding
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine; Shanghai Jiao Tong University; Shanghai; China
| | - Yu-Mei Yan
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine; Shanghai Jiao Tong University; Shanghai; China
| | - Jian-Bin Huang
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine; Shanghai Jiao Tong University; Shanghai; China
| | - Ju Mei
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine; Shanghai Jiao Tong University; Shanghai; China
| | - Jia-Quan Zhu
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine; Shanghai Jiao Tong University; Shanghai; China
| | - Hao Liu
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine; Shanghai Jiao Tong University; Shanghai; China
| |
Collapse
|
50
|
Rutkovskiy A, Mariero LH, Nygård S, Stensløkken KO, Valen G, Vaage J. Transient hyperosmolality modulates expression of cardiac aquaporins. Biochem Biophys Res Commun 2012; 425:70-5. [DOI: 10.1016/j.bbrc.2012.07.052] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 07/12/2012] [Indexed: 10/28/2022]
|