1
|
Sugihara R, Hashimura T, Sakata Y. Goreisan as a successful adjuvant therapy of heart failure with preserved ejection fraction and advanced chronic kidney disease: a case report. Oxf Med Case Reports 2023; 2023:omad116. [PMID: 37881268 PMCID: PMC10597620 DOI: 10.1093/omcr/omad116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/31/2023] [Accepted: 09/14/2023] [Indexed: 10/27/2023] Open
Abstract
Atrial functional mitral and tricuspid regurgitation due to atrial fibrillation (AF) are common causes of heart failure with preserved ejection fraction, but standard treatment with conventional diuretics can often lead to renal dysfunction. Kampo Goreisan, a traditional Eastern-Asian herbal medicine that regulates body water balance via the aquaporin-incorporated water reabsorption system can be used as an alternative therapy without causing renal burden. In this report, we describe a case of successful treatment with Goreisan of heart failure with preserved ejection fraction (HFpEF) due to atrial functional mitral and tricuspid regurgitation (AFMR/TR) receiving guideline-directed medical-therapy. Goreisan could afford amelioration of regurgitation and improvement bilateral systolic ventricular function without renal dysfunction. Thus, Goreisan may be a promising therapeutic option for patients refractory to conventional diuretics.
Collapse
Affiliation(s)
- Ryuta Sugihara
- Department of Cardiovascular Medicine, Kawachi General Hospital, 1-31 Yokomakura, Higashiosaka, Osaka 578-0954, Japan
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Tatsuro Hashimura
- Department of Cardiovascular Medicine, Kawachi General Hospital, 1-31 Yokomakura, Higashiosaka, Osaka 578-0954, Japan
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
2
|
Nicosia M, Lee J, Beavers A, Kish D, Farr GW, McGuirk PR, Pelletier MF, Lathia JD, Fairchild RL, Valujskikh A. Water channel aquaporin 4 is required for T cell receptor mediated lymphocyte activation. J Leukoc Biol 2023; 113:544-554. [PMID: 36805947 PMCID: PMC10848298 DOI: 10.1093/jleuko/qiad010] [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: 01/09/2022] [Revised: 12/16/2022] [Accepted: 01/16/2023] [Indexed: 02/04/2023] Open
Abstract
Aquaporins are a family of ubiquitously expressed transmembrane water channels implicated in a broad range of physiological functions. We have previously reported that aquaporin 4 (AQP4) is expressed on T cells and that treatment with a small molecule AQP4 inhibitor significantly delays T cell mediated heart allograft rejection. Using either genetic deletion or small molecule inhibitor, we show that AQP4 supports T cell receptor mediated activation of both mouse and human T cells. Intact AQP4 is required for optimal T cell receptor (TCR)-related signaling events, including nuclear translocation of transcription factors and phosphorylation of proximal TCR signaling molecules. AQP4 deficiency or inhibition impairs actin cytoskeleton rearrangements following TCR crosslinking, causing inferior TCR polarization and a loss of TCR signaling. Our findings reveal a novel function of AQP4 in T lymphocytes and identify AQP4 as a potential therapeutic target for preventing TCR-mediated T cell activation.
Collapse
Affiliation(s)
- Michael Nicosia
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Juyeun Lee
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Ashley Beavers
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Danielle Kish
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - George W. Farr
- Aeromics Inc., 470 James Street Suite 007, New Haven, CT 06513, United States
| | - Paul R. McGuirk
- Aeromics Inc., 470 James Street Suite 007, New Haven, CT 06513, United States
| | - Marc F. Pelletier
- Aeromics Inc., 470 James Street Suite 007, New Haven, CT 06513, United States
| | - Justin D. Lathia
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Robert L. Fairchild
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Anna Valujskikh
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| |
Collapse
|
3
|
Kozlakidis Z, Shi P, Abarbanel G, Klein C, Sfera A. Recent Developments in Protein Lactylation in PTSD and CVD: Novel Strategies and Targets. BIOTECH 2023; 12:38. [PMID: 37218755 PMCID: PMC10204439 DOI: 10.3390/biotech12020038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/27/2023] [Accepted: 05/01/2023] [Indexed: 05/24/2023] Open
Abstract
In 1938, Corneille Heymans received the Nobel Prize in physiology for discovering that oxygen sensing in the aortic arch and carotid sinus was mediated by the nervous system. The genetics of this process remained unclear until 1991 when Gregg Semenza while studying erythropoietin, came upon hypoxia-inducible factor 1, for which he obtained the Nobel Prize in 2019. The same year, Yingming Zhao found protein lactylation, a posttranslational modification that can alter the function of hypoxia-inducible factor 1, the master regulator of cellular senescence, a pathology implicated in both post-traumatic stress disorder (PTSD) and cardiovascular disease (CVD). The genetic correlation between PTSD and CVD has been demonstrated by many studies, of which the most recent one utilizes large-scale genetics to estimate the risk factors for these conditions. This study focuses on the role of hypertension and dysfunctional interleukin 7 in PTSD and CVD, the former caused by stress-induced sympathetic arousal and elevated angiotensin II, while the latter links stress to premature endothelial cell senescence and early vascular aging. This review summarizes the recent developments and highlights several novel PTSD and CVD pharmacological targets. They include lactylation of histone and non-histone proteins, along with the related biomolecular actors such as hypoxia-inducible factor 1α, erythropoietin, acid-sensing ion channels, basigin, and Interleukin 7, as well as strategies to delay premature cellular senescence by telomere lengthening and resetting the epigenetic clock.
Collapse
Affiliation(s)
- Zisis Kozlakidis
- International Agency for Research on Cancer, World Health Organization (IARC/WHO), 69372 Lyon, France
| | - Patricia Shi
- Department of Psychiatry, Loma Linda University, Loma Linda, CA 92350, USA
| | - Ganna Abarbanel
- Patton State Hospital, University of California, Riverside, CA 92521, USA
| | | | - Adonis Sfera
- Patton State Hospital, University of California, Riverside, CA 92521, USA
- Department of Psychiatry, University of California, Riverside, CA 92521, USA
| |
Collapse
|
4
|
Iglesias M, Brennan DC, Larsen CP, Raimondi G. Targeting inflammation and immune activation to improve CTLA4-Ig-based modulation of transplant rejection. Front Immunol 2022; 13:926648. [PMID: 36119093 PMCID: PMC9478663 DOI: 10.3389/fimmu.2022.926648] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022] Open
Abstract
For the last few decades, Calcineurin inhibitors (CNI)-based therapy has been the pillar of immunosuppression for prevention of organ transplant rejection. However, despite exerting effective control of acute rejection in the first year post-transplant, prolonged CNI use is associated with significant side effects and is not well suited for long term allograft survival. The implementation of Costimulation Blockade (CoB) therapies, based on the interruption of T cell costimulatory signals as strategy to control allo-responses, has proven potential for better management of transplant recipients compared to CNI-based therapies. The use of the biologic cytotoxic T-lymphocyte associated protein 4 (CTLA4)-Ig is the most successful approach to date in this arena. Following evaluation of the BENEFIT trials, Belatacept, a high-affinity version of CTLA4-Ig, has been FDA approved for use in kidney transplant recipients. Despite its benefits, the use of CTLA4-Ig as a monotherapy has proved to be insufficient to induce long-term allograft acceptance in several settings. Multiple studies have demonstrated that events that induce an acute inflammatory response with the consequent release of proinflammatory cytokines, and an abundance of allograft-reactive memory cells in the recipient, can prevent the induction of or break established immunomodulation induced with CoB regimens. This review highlights advances in our understanding of the factors and mechanisms that limit CoB regimens efficacy. We also discuss recent successes in experimentally designing complementary therapies that favor CTLA4-Ig effect, affording a better control of transplant rejection and supporting their clinical applicability.
Collapse
Affiliation(s)
- Marcos Iglesias
- Vascularized and Composite Allotransplantation (VCA) Laboratory, Department of Plastic and Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- *Correspondence: Giorgio Raimondi, ; Marcos Iglesias,
| | - Daniel C. Brennan
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Christian P. Larsen
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States
| | - Giorgio Raimondi
- Vascularized and Composite Allotransplantation (VCA) Laboratory, Department of Plastic and Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- *Correspondence: Giorgio Raimondi, ; Marcos Iglesias,
| |
Collapse
|
5
|
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
|
6
|
The role of AQP3 and AQP4 channels in cisplatin-induced cardiovascular edema and the protective effect of melatonin. Mol Biol Rep 2021; 48:7457-7465. [PMID: 34657253 DOI: 10.1007/s11033-021-06763-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 09/15/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND The present study evaluates the development of edema, the change in the AQP3, AQP4, p53 and Bax gene expressions, and the protective effects of melatonin in rat hearts administered with cisplatin. METHODS AND RESULTS A total of 28 Wistar albino rats were divided into four groups. The vehicle was administered intraperitoneally (i.p.) to the rats in the control group. The melatonin group (Mel) received melatonin at a dose of 10 mg/kg for 13 days. The cisplatin group (Cis) received cisplatin on days 1, 5, 9 and 13 at a dose of 4 mg/kg. The rats in the cisplatin + melatonin (Cis+Mel) group underwent the procedures both in the Mel and Cis groups. Blood and left ventricular samples were taken and analyzed on day 14 of the study. AQP3, p53 and Bax gene expressions were found to be significantly increased following cisplatin administration compared to the control, while melatonin administration significantly decreased the expression of these genes (p < 0.05). Melatonin administration also significantly decreased the level of AQP4 gene expression compared to the cis. On histological examination, congestion, hemorrhage, extracellular and intracellular edema, and degenerative changes were significantly more common in the Cis than in the control. Melatonin administration significantly decreased intracellular edema (p = 0.010) and degenerative changes (p = 0.010), and the improvement in extracellular edema was close to statistical significance (p = 0.051) in melatonin. CONCLUSIONS These results indicate that melatonin had an ameliorative effect on myocardial edema and AQP channels, and that it may be used as a protective molecule against myocardial edema secondary to cisplatin administration.
Collapse
|
7
|
Cibelli A, Veronica Lopez-Quintero S, Mccutcheon S, Scemes E, Spray DC, Stout RF, Suadicani SO, Thi MM, Urban-Maldonado M. Generation and Characterization of Immortalized Mouse Cortical Astrocytes From Wildtype and Connexin43 Knockout Mice. Front Cell Neurosci 2021; 15:647109. [PMID: 33790744 PMCID: PMC8005635 DOI: 10.3389/fncel.2021.647109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 02/18/2021] [Indexed: 11/17/2022] Open
Abstract
We transduced mouse cortical astrocytes cultured from four litters of embryonic wildtype (WT) and connexin43 (Cx43) null mouse pups with lentiviral vector encoding hTERT and measured expression of astrocyte-specific markers up to passage 10 (p10). The immortalized cell lines thus generated (designated IWCA and IKOCA, respectively) expressed biomarkers consistent with those of neonatal astrocytes, including Cx43 from wildtype but not from Cx43-null mice, lack of Cx30, and presence of Cx26. AQP4, the water channel that is found in high abundance in astrocyte end-feet, was expressed at moderately high levels in early passages, and its mRNA and protein declined to low but still detectable levels by p10. The mRNA levels of the astrocyte biomarkers aldehyde dehydrogenase 1L1 (ALDH1L1), glutamine synthetase (GS) and glial fibrillary acidic protein (GFAP) remained relatively constant during successive passages. GS protein expression was maintained while GFAP declined with cell passaging but was still detectable at p10. Both mRNA and protein levels of glutamate transporter 1 (GLT-1) declined with passage number. Immunostaining at corresponding times was consistent with the data from Western blots and provided evidence that these proteins were expressed at appropriate intracellular locations. Consistent with our goal of generating immortalized cell lines in which Cx43 was either functionally expressed or absent, IWCA cells were found to be well coupled with respect to intercellular dye transfer and similar to primary astrocyte cultures in terms of time course of junction formation, electrical coupling strength and voltage sensitivity. Moreover, barrier function was enhanced in co-culture of the IWCA cell line with bEnd.3 microvascular endothelial cells. In addition, immunostaining revealed oblate endogenous Cx43 gap junction plaques in IWCA that were similar in appearance to those plaques obtained following transfection of IKOCA cells with fluorescent protein tagged Cx43. Re-expression of Cx43 in IKOCA cells allows experimental manipulation of connexins and live imaging of interactions between connexins and other proteins. We conclude that properties of these cell lines resemble those of primary cultured astrocytes, and they may provide useful tools in functional studies by facilitating genetic and pharmacological manipulations in the context of an astrocyte-appropriate cellular environment.
Collapse
Affiliation(s)
- Antonio Cibelli
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States
| | | | - Sean Mccutcheon
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States
| | - Eliana Scemes
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States
| | - David C. Spray
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States,Department of Medicine (Cardiology), Albert Einstein College of Medicine, New York, NY, United States,*Correspondence: David C. Spray,
| | - Randy F. Stout
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States,Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, New York, NY, United States,Randy J. Stout Jr.,
| | - Sylvia O. Suadicani
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States,Department of Urology, Albert Einstein College of Medicine, New York, NY, United States,Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY, United States
| | - Mia M. Thi
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States,Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY, United States,Department of Orthopaedic Surgery, Albert Einstein College of Medicine, New York, NY, United States
| | - Marcia Urban-Maldonado
- Department of Urology, Albert Einstein College of Medicine, New York, NY, United States,Department of Orthopaedic Surgery, Albert Einstein College of Medicine, New York, NY, United States
| |
Collapse
|
8
|
Construction and analysis for differentially expressed long non-coding RNAs and mRNAs in acute myocardial infarction. Sci Rep 2020; 10:6989. [PMID: 32332808 PMCID: PMC7181872 DOI: 10.1038/s41598-020-63840-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 04/06/2020] [Indexed: 12/18/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) are transcripts longer than 200 nucleotides. Some lncRNAs are related to acute myocardial infarction (AMI) and can serve as blood-based biomarkers for AMI detection. To identify whether new lncRNAs participate in AMI, the expression of lncRNAs and mRNAs was analysed by microarray analysis (Agilent human array) with the limma package in R in two series: five paired peripheral blood mononuclear cell (PBMC) samples and four paired plasma samples from different AMI patients. In PBMCs, a total of 2677 upregulated and 458 downregulated lncRNAs were significantly differentially expressed; additionally, 1168 mRNAs were upregulated and 1334 mRNAs were downregulated between the AMI patients and controls. In plasma, we found 41 upregulated and 51 downregulated lncRNAs that were differentially expressed, as well as 9 mRNAs that were upregulated and 9 mRNAs that were downregulated among the two groups. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed using the clusterProfiler package in R, and differentially expressed mRNAs were functionally annotated. The top differentially expressed mRNAs were associated with circadian rhythm, the NF-kB pathway, the p53 pathway and the metabolism pathway. We further performed target gene prediction and coexpression analysis and revealed the interrelationships among the significantly differentially expressed lncRNAs and mRNAs. The expression of four lncRNAs (uc002ddj.1, NR_047662, ENST00000581794.1 and ENST00000509938.1) was validated in the newly diagnosed AMI and control groups by quantitative real-time PCR (qRT-PCR). Our study demonstrated that the clustered expression of lncRNAs between PBMCs and plasma showed tremendous differences. The newly screened lncRNAs may play indispensable roles in the development of AMI, although their biological functions need to be further validated.
Collapse
|
9
|
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
|
10
|
Nicosia M, Miyairi S, Beavers A, Farr GW, McGuirk PR, Pelletier MF, Valujskikh A. Aquaporin 4 inhibition alters chemokine receptor expression and T cell trafficking. Sci Rep 2019; 9:7417. [PMID: 31092872 PMCID: PMC6520372 DOI: 10.1038/s41598-019-43884-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 04/26/2019] [Indexed: 11/16/2022] Open
Abstract
Aquaporins (AQPs) are water channels that mediate a variety of biological processes. However, their role in the immune system is poorly understood. We recently reported that AQP4 is expressed by naïve and memory T cells and that AQP4 blockade with a small molecule inhibitor prolongs murine heart allograft survival at least partially through diminishing T cell activation, proliferation and trafficking. The goal of this study was to determine how AQP4 function impacts T cells in the absence of antigen stimulation. AQP4 inhibition transiently reduced the number of circulating CD4+ and CD8+ T cells in naïve non-transplanted mice in the absence of systemic T cell depletion. Adoptive transfer studies demonstrated T cell intrinsic effect of AQP4 inhibition. AQP4 blockade altered T cell gene and protein expression of chemokine receptors S1PR1 and CCR7, and their master regulator KLF-2, and reduced chemotaxis toward S1P and CCL21. Consistent with the in vitro data, in vivo AQP4 inhibition reduced T lymphocyte numbers in the lymph nodes with simultaneous accumulation in the liver. Our findings indicate that blocking AQP4 reversibly alters T lymphocyte trafficking pattern. This information can be explored for the treatment of undesirable immune responses in transplant recipients or in patients with autoimmune diseases.
Collapse
Affiliation(s)
- Michael Nicosia
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, 44195, USA
| | - Satoshi Miyairi
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, 44195, USA
| | - Ashley Beavers
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, 44195, USA
| | | | | | | | - Anna Valujskikh
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, 44195, USA.
| |
Collapse
|
11
|
Farr GW, Hall CH, Farr SM, Wade R, Detzel JM, Adams AG, Buch JM, Beahm DL, Flask CA, Xu K, LaManna JC, McGuirk PR, Boron WF, Pelletier MF. Functionalized Phenylbenzamides Inhibit Aquaporin-4 Reducing Cerebral Edema and Improving Outcome in Two Models of CNS Injury. Neuroscience 2019; 404:484-498. [PMID: 30738082 DOI: 10.1016/j.neuroscience.2019.01.034] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/18/2019] [Accepted: 01/21/2019] [Indexed: 01/23/2023]
Abstract
Cerebral edema in ischemic stroke can lead to increased intracranial pressure, reduced cerebral blood flow and neuronal death. Unfortunately, current therapies for cerebral edema are either ineffective or highly invasive. During the development of cytotoxic and subsequent ionic cerebral edema water enters the brain by moving across an intact blood brain barrier and through aquaporin-4 (AQP4) at astrocyte endfeet. Using AQP4-expressing cells, we screened small molecule libraries for inhibitors that reduce AQP4-mediated water permeability. Additional functional assays were used to validate AQP4 inhibition and identified a promising structural series for medicinal chemistry. These efforts improved potency and revealed a compound we designated AER-270, N-[3,5-bis (trifluoromethyl)phenyl]-5-chloro-2-hydroxybenzamide. AER-270 and a prodrug with enhanced solubility, AER-271 2-{[3,5-Bis(trifluoromethyl) phenyl]carbamoyl}-4-chlorophenyl dihydrogen phosphate, improved neurological outcome and reduced swelling in two models of CNS injury complicated by cerebral edema: water intoxication and ischemic stroke modeled by middle cerebral artery occlusion.
Collapse
Affiliation(s)
- George W Farr
- Aeromics, Inc., Cleveland, OH 44106, USA; Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| | | | | | - Ramon Wade
- Aeromics, Inc., Cleveland, OH 44106, USA
| | | | | | | | - Derek L Beahm
- Department of Biology, Buffalo State College, Buffalo, NY 14222, USA
| | - Christopher A Flask
- Departments of Radiology, Biomedical Engineering and Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Kui Xu
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Joseph C LaManna
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | | | - Walter F Boron
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | | |
Collapse
|
12
|
Association of Genetic Variation at AQP4 Locus with Vascular Depression. Biomolecules 2018; 8:biom8040164. [PMID: 30563176 PMCID: PMC6316852 DOI: 10.3390/biom8040164] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 11/27/2018] [Accepted: 11/27/2018] [Indexed: 02/07/2023] Open
Abstract
Despite its substantial clinical importance, specific genetic variants associated with depression have not yet been identified. We sought to identify genetic variants associated with depression by (a) focusing on a more homogenous subsample (vascular depression) and (b) applying a three-stage approach. First, we contacted 730 participants with a confirmed atherosclerotic disease (coronary artery disease) from a population-based study population (German Myocardial Infarction Family Study IV) for psychiatric assessment with the Mini International Neuropsychiatric Interview. Second, we genotyped these patients using genome-wide single nucleotide polymorphism (SNP) arrays. Third, we characterized the SNP via in-silico analysis. The final sample consisted of 342 patients (78.3% male, age = 63.2 ± 9.9 years), 22.8% with a severe depressive disorder. Variant rs528732638 on chromosome 18q11.2 was a genome-wide significant variant and was associated with 3.6-fold increase in the odds of lifetime depression. The locus belongs to a linkage disequilibrium block showing expression quantitative trait loci effects on three putative cis-regulated genes, including the aquaporin 4 (AQP4) locus. AQP4 is already known to mediate the formation of ischemic edema in the brain and heart, increasing the size and extent of resulting lesions. Our findings indicate that AQP4 may also play a role in the etiopathology of vascular depression.
Collapse
|
13
|
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
|
14
|
Cheng Y, Chao J, Dai D, Dai Y, Zhu D, Liu B. AQP4-knockout aggravation of isoprenaline-induced myocardial injury is mediated by p66Shc and endoplasmic reticulum stress. Clin Exp Pharmacol Physiol 2018; 44:1106-1115. [PMID: 28688153 DOI: 10.1111/1440-1681.12812] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 06/13/2017] [Accepted: 06/22/2017] [Indexed: 12/14/2022]
Abstract
Aquaporin 4 (AQP4) is a type of water channel protein that maintains the water balance of cardiomyocytes. However, the physiological role of AQP4 in cardiovascular disease is poorly understood. We wanted to explore whether p66Shc and endoplasmic reticulum stress participates in AQP4 knockout (KO)-mediated cardiac injury. There were two types of mice: AQP4 knockout and wild-type mice. Each type was randomly divided into three groups: Control group, isoprenaline stimulation group (ISO, 1 mg/kg, s.c., 5 days), and apocynin treatment group (APO, 100 mg/kg, p.o., 3 days). H9c2 rat cardiomyocytes were cultured for RNA interference of AQP4. Results showed increased left ventricular weight index and more severe myocardial inflammation were induced in AQP4 knockout mice relative to wild-type mice, accompanied by significantly increased levels of the oxidative stress biomarkers MDA and NOX4. In addition, the expressions of p66Shc, ER stress markers PERK, GRP78 and CHOP and proinflammatory factors such as ETA , IL6 and TNFα were upregulated in the myocardium of AQP4 knockout mice or AQP4 siRNA treated cardiomyocytes, whereas CASQ2 was downregulated. ISO stimulation aggravated these abnormalities, which were significantly attenuated by apocynin. This study showed that AQP4 knockout mice were susceptible to cardiac injury induced by ISO. The mechanism was closely connected with p66Shc and proinflammatory factors. Endoplasmic reticulum stress was also involved in the pathological process.
Collapse
Affiliation(s)
- Yusi Cheng
- Institute of Nephrology, Medical School of Southeast University, Nanjing, China.,Department of Physiology, Medical School of Southeast University, Nanjing, China
| | - Jie Chao
- Department of Physiology, Medical School of Southeast University, Nanjing, China
| | - Dezai Dai
- Research Division of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Yin Dai
- Research Division of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Dongdong Zhu
- Institute of Nephrology, Medical School of Southeast University, Nanjing, China
| | - Bicheng Liu
- Institute of Nephrology, Medical School of Southeast University, Nanjing, China
| |
Collapse
|
15
|
Ayasoufi K, Kohei N, Nicosia M, Fan R, Farr GW, McGuirk PR, Pelletier MF, Fairchild RL, Valujskikh A. Aquaporin 4 blockade improves survival of murine heart allografts subjected to prolonged cold ischemia. Am J Transplant 2018; 18:1238-1246. [PMID: 29243390 PMCID: PMC5910181 DOI: 10.1111/ajt.14624] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 11/30/2017] [Accepted: 12/03/2017] [Indexed: 01/25/2023]
Abstract
Prolonged cold ischemia storage (CIS) is a leading risk factor for poor transplant outcome. Existing strategies strive to minimize ischemia-reperfusion injury in transplanted organs, yet there is a need for novel approaches to improve outcomes of marginal allografts and expand the pool of donor organs suitable for transplantation. Aquaporins (AQPs) are a family of water channels that facilitate homeostasis, tissue injury, and inflammation. We tested whether inhibition of AQP4 improves the survival of fully MHC-mismatched murine cardiac allografts subjected to 8 hours of CIS. Administration of a small molecule AQP4 inhibitor during donor heart collection and storage and for a short-time posttransplantation improves the viability of donor graft cells, diminishes donor-reactive T cell responses, and extends allograft survival in the absence of other immunosuppression. Furthermore, AQP4 inhibition is synergistic with cytotoxic T lymphocyte-associated antigen 4-Ig in prolonging survival of 8-hour CIS heart allografts. AQP4 blockade markedly reduced T cell proliferation and cytokine production in vitro, suggesting that the improved graft survival is at least in part mediated through direct effects on donor-reactive T cells. These results identify AQPs as a promising target for diminishing donor-specific alloreactivity and improving the survival of high-risk organ transplants.
Collapse
Affiliation(s)
- Katayoun Ayasoufi
- Glickman Urological Institute and Department of Immunology, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Naoki Kohei
- Glickman Urological Institute and Department of Immunology, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Michael Nicosia
- Glickman Urological Institute and Department of Immunology, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Ran Fan
- Glickman Urological Institute and Department of Immunology, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | | | | | | | - Robert L. Fairchild
- Glickman Urological Institute and Department of Immunology, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Anna Valujskikh
- Glickman Urological Institute and Department of Immunology, Cleveland Clinic, Cleveland, Ohio 44195, USA
| |
Collapse
|
16
|
Rutkovskiy A, Sagave J, Czibik G, Baysa A, Zihlavnikova Enayati K, Hillestad V, Dahl CP, Fiane A, Gullestad L, Gravning J, Ahmed S, Attramadal H, Valen G, Vaage J. Connective tissue growth factor and bone morphogenetic protein 2 are induced following myocardial ischemia in mice and humans. Scandinavian Journal of Clinical and Laboratory Investigation 2017; 77:321-331. [PMID: 28460577 DOI: 10.1080/00365513.2017.1318447] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
We aimed to study the cardiac expression of bone morphogenetic protein 2, its receptor 1 b, and connective tissue growth factor, factors implicated in cardiac embryogenesis, following ischemia/hypoxia, heart failure, and in remodeling hearts from humans and mice. Biopsies from the left ventricle of patients with end-stage heart failure due to dilated cardiomyopathy or coronary artery disease were compared with donor hearts and biopsies from patients with normal heart function undergoing coronary artery bypass grafting. Mouse model of post-infarction remodeling was made by permanent ligation of the left coronary artery. Hearts were analyzed by real-time polymerase chain reaction and Western blotting after 24 hours and after 2 and 4 weeks. Patients with dilated cardiomyopathy and mice post-infarction had increased cardiac expression of connective tissue growth factor. Bone morphogenetic protein 2 was increased in human hearts failing due to coronary artery disease and in mice post-infarction. Gene expression of bone morphogenetic protein receptor 1 beta was reduced in hearts of patients with failure, but increased two weeks following permanent ligation of the left coronary artery in mice. In conclusion, connective tissue growth factor is upregulated in hearts of humans with dilated cardiomyopathy, bone morphogenetic protein 2 is upregulated in remodeling due to myocardial infarction while its receptor 1 b in human failing hearts is downregulated. A potential explanation might be an attempt to engage regenerative processes, which should be addressed by further, mechanistic studies.
Collapse
Affiliation(s)
- Arkady Rutkovskiy
- a Division of Physiology, Department of Molecular Medicine , Institute of Basic Medical Sciences, University of Oslo , Oslo , Norway.,b International Laboratory of Bioinformatics and Genomics, ITMO University , Saint-Petersburg , Russia
| | - Julia Sagave
- a Division of Physiology, Department of Molecular Medicine , Institute of Basic Medical Sciences, University of Oslo , Oslo , Norway
| | - Gabor Czibik
- a Division of Physiology, Department of Molecular Medicine , Institute of Basic Medical Sciences, University of Oslo , Oslo , Norway
| | - Anton Baysa
- a Division of Physiology, Department of Molecular Medicine , Institute of Basic Medical Sciences, University of Oslo , Oslo , Norway
| | - Katarina Zihlavnikova Enayati
- a Division of Physiology, Department of Molecular Medicine , Institute of Basic Medical Sciences, University of Oslo , Oslo , Norway
| | - Vigdis Hillestad
- c Institute for Experimental Medical Research, Oslo University Hospital, Ullevål , Oslo , Norway
| | | | - Arnt Fiane
- e Department of Cardiothoracic Surgery , Oslo University Hospital , Oslo , Norway
| | - Lars Gullestad
- d Department of Cardiology , Oslo University Hospital , Oslo , Norway
| | - Jørgen Gravning
- f Division of Medicine , Akershus University Hospital , Lørenskog , Norway
| | - Shakil Ahmed
- c Institute for Experimental Medical Research, Oslo University Hospital, Ullevål , Oslo , Norway
| | - Håvard Attramadal
- c Institute for Experimental Medical Research, Oslo University Hospital, Ullevål , Oslo , Norway
| | - Guro Valen
- a Division of Physiology, Department of Molecular Medicine , Institute of Basic Medical Sciences, University of Oslo , Oslo , Norway
| | - Jarle Vaage
- g Department of Emergency Medicine and Intensive Care , Oslo University Hospital , Oslo , Norway
| |
Collapse
|
17
|
Vilahur G, Gutiérrez M, Casani L, Varela L, Capdevila A, Pons-Lladó G, Carreras F, Carlsson L, Hidalgo A, Badimon L. Protective Effects of Ticagrelor on Myocardial Injury After Infarction. Circulation 2016; 134:1708-1719. [PMID: 27789556 DOI: 10.1161/circulationaha.116.024014] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 10/08/2016] [Indexed: 11/16/2022]
Abstract
BACKGROUND The P2Y12 receptor antagonist ticagrelor has been shown to be clinically superior to clopidogrel. Although the underlying mechanisms remain elusive, ticagrelor may exert off-target effects through adenosine-related mechanisms. We aimed to investigate whether ticagrelor reduces myocardial injury to a greater extent than clopidogrel after myocardial infarction (MI) at a similar level of platelet inhibition and to determine the underlying mechanisms. METHODS Pigs received the following before MI induction: (1) placebo-control; (2) a loading dose of clopidogrel (600 mg); (3) a loading dose of ticagrelor (180 mg); or (4) a loading dose of ticagrelor followed by an adenosine A1/A2-receptor antagonist [8-(p-sulfophenyl)theophylline, 4 mg/kg intravenous] to determine the potential contribution of adenosine in ticagrelor-related cardioprotection. Animals received the corresponding maintenance doses of the antiplatelet agents during the following 24 hours and underwent 3T-cardiac MRI analysis. Platelet inhibition was monitored by ADP-induced platelet aggregation. In the myocardium, we assessed the expression and activation of proteins known to modulate edema formation, including aquaporin-4 and AMP-activated protein kinase and its downstream effectors CD36 and endothelial nitric oxide synthase and cyclooxygenase-2 activity. RESULTS Clopidogrel and ticagrelor exerted a high and consistent antiplatelet effect (68.2% and 62.2% of platelet inhibition, respectively, on challenge with 20 μmol/L ADP) that persisted up to 24 hours post-MI (P<0.05). All groups showed comparable myocardial area-at-risk and cardiac worsening after MI induction. 3T-Cardiac MRI analysis revealed that clopidogrel- and ticagrelor-treated animals had a significantly smaller extent of MI than placebo-control animals (15.7 g left ventricle and 12.0 g left ventricle versus 22.8 g left ventricle, respectively). Yet, ticagrelor reduced infarct size to a significantly greater extent than clopidogrel (further 23.5% reduction; P=0.0026), an effect supported by troponin-I assessment and histopathologic analysis (P=0.0021). Furthermore, in comparison with clopidogrel, ticagrelor significantly diminished myocardial edema by 24.5% (P=0.004), which correlated with infarct mass (r=0.73; P<0.001). 8-(p-Sulfophenyl)theophylline administration abolished the cardioprotective effects of ticagrelor over clopidogrel. At a molecular level, aquaporin-4 expression decreased and the expression and activation of AMP-activated protein kinase signaling and cyclooxygenase-2 increased in the ischemic myocardium of ticagrelor- versus clopidogrel-treated animals (P<0.05). These protein changes were not observed in those animals administered the adenosine receptor blocker 8-(p-sulfophenyl)theophylline. CONCLUSIONS Ticagrelor, beyond its antiplatelet efficacy, exerts cardioprotective effects by reducing necrotic injury and edema formation via adenosine-dependent mechanisms.
Collapse
Affiliation(s)
- Gemma Vilahur
- From Cardiovascular Research Center (CSIC-ICCC), IIB-HSCSP, Barcelona, Spain (G.V., L.C., L.V., L.B.); Radiology Unit, Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (M.G., A.C., A.H.); Cardiology Unit. Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (G.P.-L., F.C.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden (L.C.); and Cardiovascular Research Chair UAB (Autonomous University of Barcelona), Spain (L.B.)
| | - Manuel Gutiérrez
- From Cardiovascular Research Center (CSIC-ICCC), IIB-HSCSP, Barcelona, Spain (G.V., L.C., L.V., L.B.); Radiology Unit, Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (M.G., A.C., A.H.); Cardiology Unit. Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (G.P.-L., F.C.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden (L.C.); and Cardiovascular Research Chair UAB (Autonomous University of Barcelona), Spain (L.B.)
| | - Laura Casani
- From Cardiovascular Research Center (CSIC-ICCC), IIB-HSCSP, Barcelona, Spain (G.V., L.C., L.V., L.B.); Radiology Unit, Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (M.G., A.C., A.H.); Cardiology Unit. Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (G.P.-L., F.C.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden (L.C.); and Cardiovascular Research Chair UAB (Autonomous University of Barcelona), Spain (L.B.)
| | - Lourdes Varela
- From Cardiovascular Research Center (CSIC-ICCC), IIB-HSCSP, Barcelona, Spain (G.V., L.C., L.V., L.B.); Radiology Unit, Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (M.G., A.C., A.H.); Cardiology Unit. Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (G.P.-L., F.C.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden (L.C.); and Cardiovascular Research Chair UAB (Autonomous University of Barcelona), Spain (L.B.)
| | - Antoni Capdevila
- From Cardiovascular Research Center (CSIC-ICCC), IIB-HSCSP, Barcelona, Spain (G.V., L.C., L.V., L.B.); Radiology Unit, Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (M.G., A.C., A.H.); Cardiology Unit. Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (G.P.-L., F.C.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden (L.C.); and Cardiovascular Research Chair UAB (Autonomous University of Barcelona), Spain (L.B.)
| | - Guillem Pons-Lladó
- From Cardiovascular Research Center (CSIC-ICCC), IIB-HSCSP, Barcelona, Spain (G.V., L.C., L.V., L.B.); Radiology Unit, Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (M.G., A.C., A.H.); Cardiology Unit. Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (G.P.-L., F.C.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden (L.C.); and Cardiovascular Research Chair UAB (Autonomous University of Barcelona), Spain (L.B.)
| | - Francesc Carreras
- From Cardiovascular Research Center (CSIC-ICCC), IIB-HSCSP, Barcelona, Spain (G.V., L.C., L.V., L.B.); Radiology Unit, Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (M.G., A.C., A.H.); Cardiology Unit. Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (G.P.-L., F.C.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden (L.C.); and Cardiovascular Research Chair UAB (Autonomous University of Barcelona), Spain (L.B.)
| | - Leif Carlsson
- From Cardiovascular Research Center (CSIC-ICCC), IIB-HSCSP, Barcelona, Spain (G.V., L.C., L.V., L.B.); Radiology Unit, Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (M.G., A.C., A.H.); Cardiology Unit. Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (G.P.-L., F.C.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden (L.C.); and Cardiovascular Research Chair UAB (Autonomous University of Barcelona), Spain (L.B.)
| | - Alberto Hidalgo
- From Cardiovascular Research Center (CSIC-ICCC), IIB-HSCSP, Barcelona, Spain (G.V., L.C., L.V., L.B.); Radiology Unit, Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (M.G., A.C., A.H.); Cardiology Unit. Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (G.P.-L., F.C.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden (L.C.); and Cardiovascular Research Chair UAB (Autonomous University of Barcelona), Spain (L.B.)
| | - Lina Badimon
- From Cardiovascular Research Center (CSIC-ICCC), IIB-HSCSP, Barcelona, Spain (G.V., L.C., L.V., L.B.); Radiology Unit, Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (M.G., A.C., A.H.); Cardiology Unit. Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (G.P.-L., F.C.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden (L.C.); and Cardiovascular Research Chair UAB (Autonomous University of Barcelona), Spain (L.B.).
| |
Collapse
|
18
|
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
|
19
|
Johnson ZI, Gogate SS, Day R, Binch A, Markova DZ, Chiverton N, Cole A, Conner M, Shapiro IM, Le Maitre CL, Risbud MV. Aquaporin 1 and 5 expression decreases during human intervertebral disc degeneration: Novel HIF-1-mediated regulation of aquaporins in NP cells. Oncotarget 2016; 6:11945-58. [PMID: 25844601 PMCID: PMC4494915 DOI: 10.18632/oncotarget.3631] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 03/05/2015] [Indexed: 01/07/2023] Open
Abstract
Objectives of this study were to investigate whether AQP1 and AQP5 expression is altered during intervertebral disc degeneration and if hypoxia and HIF-1 regulate their expression in NP cells. AQP expression was measured in human tissues from different degenerative grades; regulation by hypoxia and HIF-1 was studied using promoter analysis and gain- and loss-of-function experiments. We show that both AQPs are expressed in the disc and that mRNA and protein levels decline with human disease severity. Bioinformatic analyses of AQP promoters showed multiple evolutionarily conserved HREs. Surprisingly, hypoxia failed to induce promoter activity or expression of either AQP. While genomic chromatin immunoprecipitation showed limited binding of HIF-1α to conserved HREs, their mutation did not suppress promoter activities. Stable HIF-1α suppression significantly decreased mRNA and protein levels of both AQPs, but HIF-1α failed to induce AQP levels following accumulation. Together, our results demonstrate that AQP1 and AQP5 expression is sensitive to human disc degeneration and that HIF-1α uniquely maintains basal expression of both AQPs in NP cells, independent of oxemic tension and HIF-1 binding to promoter HREs. Diminished HIF-1 activity during degeneration may suppress AQP levels in NP cells, compromising their ability to respond to extracellular osmolarity changes.
Collapse
Affiliation(s)
- Zariel I Johnson
- Department of Orthopaedic Surgery and Graduate Program in Cell and Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Shilpa S Gogate
- Department of Orthopaedic Surgery and Graduate Program in Cell and Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Rebecca Day
- Biomedical Research Centre, Sheffield Hallam University, Sheffield, UK
| | - Abbie Binch
- Biomedical Research Centre, Sheffield Hallam University, Sheffield, UK
| | - Dessislava Z Markova
- Department of Orthopaedic Surgery and Graduate Program in Cell and Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Neil Chiverton
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Ashley Cole
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Matt Conner
- Biomedical Research Centre, Sheffield Hallam University, Sheffield, UK
| | - Irving M Shapiro
- Department of Orthopaedic Surgery and Graduate Program in Cell and Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Makarand V Risbud
- Department of Orthopaedic Surgery and Graduate Program in Cell and Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
20
|
LaMacchia JC, Roth MB. Aquaporins-2 and -4 regulate glycogen metabolism and survival during hyposmotic-anoxic stress in Caenorhabditis elegans. Am J Physiol Cell Physiol 2015; 309:C92-6. [PMID: 26017147 DOI: 10.1152/ajpcell.00131.2015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 05/20/2015] [Indexed: 12/22/2022]
Abstract
Periods of oxygen deprivation can lead to ion and water imbalances in affected tissues that manifest as swelling (edema). Although oxygen deprivation-induced edema is a major contributor to injury in clinical ischemic diseases such as heart attack and stroke, the pathophysiology of this process is incompletely understood. In the present study we investigate the impact of aquaporin-mediated water transport on survival in a Caenorhabditis elegans model of edema formation during complete oxygen deprivation (anoxia). We find that nematodes lacking aquaporin water channels in tissues that interface with the surrounding environment display decreased edema formation and improved survival rates in anoxia. We also find that these animals have significantly reduced demand for glycogen as an energetic substrate during anoxia. Together, our data suggest that reductions in membrane water permeability may be sufficient to induce a hypometabolic state during oxygen deprivation that reduces injury and extends survival limits.
Collapse
Affiliation(s)
- John C LaMacchia
- Molecular and Cellular Biology Program, University of Washington, Seattle, Washington; Medical Scientist Training Program, University of Washington, Seattle, Washington; and
| | - Mark B Roth
- Basic Science Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| |
Collapse
|
21
|
Xie AX, Petravicz J, McCarthy KD. Molecular approaches for manipulating astrocytic signaling in vivo. Front Cell Neurosci 2015; 9:144. [PMID: 25941472 PMCID: PMC4403552 DOI: 10.3389/fncel.2015.00144] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 03/27/2015] [Indexed: 12/26/2022] Open
Abstract
Astrocytes are the predominant glial type in the central nervous system and play important roles in assisting neuronal function and network activity. Astrocytes exhibit complex signaling systems that are essential for their normal function and the homeostasis of the neural network. Altered signaling in astrocytes is closely associated with neurological and psychiatric diseases, suggesting tremendous therapeutic potential of these cells. To further understand astrocyte function in health and disease, it is important to study astrocytic signaling in vivo. In this review, we discuss molecular tools that enable the selective manipulation of astrocytic signaling, including the tools to selectively activate and inactivate astrocyte signaling in vivo. Lastly, we highlight a few tools in development that present strong potential for advancing our understanding of the role of astrocytes in physiology, behavior, and pathology.
Collapse
Affiliation(s)
- Alison X Xie
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill Chapel Hill, NC, USA
| | - Jeremy Petravicz
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology Cambridge, MA, USA
| | - Ken D McCarthy
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill Chapel Hill, NC, USA
| |
Collapse
|
22
|
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
|
23
|
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
|
24
|
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
|
25
|
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
|
26
|
Cosgrove J, Alli S, Ramadan H, Ford HL. Myocarditis and diffuse skeletal muscle oedema: new features of neuromyelitis optica spectrum disorder? A case report. Mult Scler 2013; 20:120-2. [PMID: 23846353 DOI: 10.1177/1352458513495939] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
We present a case report of newly diagnosed neuromyelitis optica spectrum disorder (NMOSD) with associated myocarditis and diffuse oedema of the pelvic and anterior compartment thigh muscles on magnetic resonance imaging. Aquaporin 4 antibodies are expressed in skeletal myofibres but involvement of skeletal muscle is rarely reported in NMOSD and myocarditis has not previously been described in this context. This case highlights the need for further research into the involvement of cardiac and skeletal muscle in NMOSD.
Collapse
Affiliation(s)
- Jeremy Cosgrove
- Department of Neurology, Leeds General Infirmary, Leeds Teaching Hospitals NHS Trust, UK
| | | | | | | |
Collapse
|