1
|
Homilius C, Seefeldt JM, Hansen J, Nielsen R, de Paoli FV, Boedtkjer E. Lactate orchestrates metabolic hemodynamic adaptations through a unique combination of venocontraction, artery relaxation, and positive inotropy. Acta Physiol (Oxf) 2025; 241:e70037. [PMID: 40167405 PMCID: PMC11960580 DOI: 10.1111/apha.70037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/14/2025] [Accepted: 03/14/2025] [Indexed: 04/02/2025]
Abstract
AIM H+ facilitates metabolic blood flow regulation while negatively impacting cardiac contractility. Cardiovascular consequences of conjugate bases accumulating alongside H+ remain unclear. Here, we evaluate the cardiovascular effects of nine prominent carboxylates-particularly lactate, 3-hydroxybutyrate, and butyrate-linked to metabolic and microbial activity. METHODS Comparing the actions of pH-adjusted Na-carboxylates to equiosmolar NaCl, we study arteries and veins isolated from healthy rats and humans with ischaemic heart disease, isolated perfused rat hearts, and rat cardiovascular function in vivo. RESULTS The tested carboxylates generally relax arteries and veins. L-lactate relaxes human and rat arteries up to 70% (EC50 = 10.1 mM) and rat brachial and mesenteric veins up to 30% of pre-contractions, yet stands out by augmenting contractions of rat femoral, saphenous, and lateral marginal veins and human internal thoracic and great saphenous veins up to 50%. D-lactate shows only minor actions. In isolated perfused hearts, 10 mM L-lactate increases coronary flow (17.1 ± 7.7%) and left ventricular developed pressure (10.1 ± 3.0%) without affecting heart rate. L-lactate infusion in rats-reaching 3.7 ± 0.3 mM in the circulation-increases left ventricular end-diastolic volume (11.3 ± 2.8%), stroke volume (22.6 ± 3.0%), cardiac output (23.4 ± 3.5%), and ejection fraction (10.6 ± 2.0%), and lowers systemic vascular resistance (34.1 ± 3.7%) without influencing blood pressure or heart rate. The ketone body 3-hydroxybutyrate causes lactate accumulation and elevates left ventricular end-diastolic volume in vivo. CONCLUSION Carboxylate metabolites generally relax arteries and veins. L-lactate relaxes arteries, lowering systemic vascular resistance, causes preferential venocontraction with increased ventricular diastolic filling, and elevates cardiac contractility and cardiac output. We propose that L-lactate optimizes cardiovascular function during metabolic disturbances.
Collapse
Affiliation(s)
| | - Jacob M. Seefeldt
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of CardiologyAarhus University HospitalAarhusDenmark
| | - Jakob Hansen
- Department of Forensic MedicineAarhus UniversityAarhusDenmark
| | - Roni Nielsen
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of CardiologyAarhus University HospitalAarhusDenmark
| | - Frank V. de Paoli
- Department of BiomedicineAarhus UniversityAarhusDenmark
- Department of Cardiothoracic and Vascular SurgeryAarhus University HospitalAarhusDenmark
| | | |
Collapse
|
2
|
Ibañez AM, Godoy Coto J, Martínez VR, Del Milagro Yeves A, Dolcetti FJC, Cervellini S, Echavarría L, Velez-Rueda JO, Lofeudo JM, Portiansky EL, Bellini MJ, Aiello EA, Ennis IL, De Giusti VC. Cardioprotection and neurobehavioral impact of swimming training in ovariectomized rats. GeroScience 2025; 47:2317-2334. [PMID: 39527177 PMCID: PMC11979057 DOI: 10.1007/s11357-024-01422-7] [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: 01/30/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Cardiovascular (CV) disease is the major cause of mortality. Estrogens (E) exert multiple CV and neuroprotective effects. During menopause, CV and cognitive pathologies increase dramatically. At present, it is known that E exert many of their beneficial effects through the G protein-coupled estrogen receptor (GPER). Exercise reduces the risk of developing CV diseases. Sodium/proton exchanger (NHE-1) is overexpressed in ovariectomized (OVX) rats, probably due to the increase in reactive oxidative species (ROS). Insulin-like growth factor 1 (IGF-1), the main humoral mediator of exercise, inhibits the NHE-1. We aim to explore the subcellular mechanisms involved in the heart and brain impact of physiological exercise in OVX rats. We speculate that physical training, via IGF-1, prevents the increase in ROS, improving heart and brain physiological functions during menopause. Exercise diminished cardiac ROS production and increased catalase (CAT) activity in OVX rats. In concordance, IGF-1 treatment reduces brain ROS, surely contributing to the improvement in brain behavior. Moreover, the aerobic routine was able to prevent, and IGF-1 therapy to revert, NHE-1 hyperactivity in OVX rats. Finally, our results confirm the proposed signaling pathway as IGF-1/PI3K-AKT/NO. Surprisingly, GPER inhibitor (G36) was able to abolish the IGF-1 effect, suggesting that directly or indirectly GPER is part of the IGF-1 pathway. We propose that IGF-1 is the main responsible for the protective effect of aerobic training both in the heart and brain in OVX rats. Moreover, we showed that not only it is possible to prevent but also to revert the menopause-induced NHE-1 hyperactivity by exercise/IGF-1 cascade.
Collapse
Affiliation(s)
- Alejandro Martín Ibañez
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani" La Plata- Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, Calle 60 y 120, 1900, La Plata, Argentina
| | - Joshua Godoy Coto
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani" La Plata- Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, Calle 60 y 120, 1900, La Plata, Argentina
| | - Valeria Romina Martínez
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani" La Plata- Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, Calle 60 y 120, 1900, La Plata, Argentina
| | - Alejandra Del Milagro Yeves
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani" La Plata- Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, Calle 60 y 120, 1900, La Plata, Argentina
| | - Franco Juan Cruz Dolcetti
- Instituto de Investigaciones Bioquímicas de La Plata "Prof. Dr. Rodolfo R. Brenner"- Facultad de Ciencias Médicas, Universidad Nacionalde LaPlata-CONICET, La Plata, Argentina
| | - Sofía Cervellini
- Instituto de Investigaciones Bioquímicas de La Plata "Prof. Dr. Rodolfo R. Brenner"- Facultad de Ciencias Médicas, Universidad Nacionalde LaPlata-CONICET, La Plata, Argentina
| | - Lucía Echavarría
- Instituto de Investigaciones Bioquímicas de La Plata "Prof. Dr. Rodolfo R. Brenner"- Facultad de Ciencias Médicas, Universidad Nacionalde LaPlata-CONICET, La Plata, Argentina
| | - Jorge Omar Velez-Rueda
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani" La Plata- Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, Calle 60 y 120, 1900, La Plata, Argentina
| | - Juan Manuel Lofeudo
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani" La Plata- Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, Calle 60 y 120, 1900, La Plata, Argentina
| | - Enrique Leo Portiansky
- Cátedra de Patología General- Facultad de Ciencias Veterinarias, Universidad Nacional de La Plata- CONICET, La Plata, Argentina
| | - María José Bellini
- Instituto de Investigaciones Bioquímicas de La Plata "Prof. Dr. Rodolfo R. Brenner"- Facultad de Ciencias Médicas, Universidad Nacionalde LaPlata-CONICET, La Plata, Argentina
| | - Ernesto Alejandro Aiello
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani" La Plata- Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, Calle 60 y 120, 1900, La Plata, Argentina
| | - Irene Lucía Ennis
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani" La Plata- Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, Calle 60 y 120, 1900, La Plata, Argentina
| | - Verónica Celeste De Giusti
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani" La Plata- Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, Calle 60 y 120, 1900, La Plata, Argentina.
| |
Collapse
|
3
|
Chen Y, Liu P, Zhong Z, Zhang H, Sun A, Wang Y. STIM1 functions as a proton sensor to coordinate cytosolic pH with store-operated calcium entry. J Biol Chem 2024; 300:107924. [PMID: 39454952 PMCID: PMC11626807 DOI: 10.1016/j.jbc.2024.107924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/02/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
The meticulous regulation of intracellular pH (pHi) is crucial for maintaining cellular function and homeostasis, impacting physiological processes such as heart rhythm, cell migration, proliferation, and differentiation. Dysregulation of pHi is implicated in various pathologies such as arrhythmias, cancer, and neurodegenerative diseases. Here, we explore the role of STIM1, an ER calcium (Ca2+) sensor mediating Store Operated Ca2+ Entry (SOCE), in sensing pHi changes. Our study reveals that STIM1 functions as a sensor for pHi changes, independent of its Ca2+-binding state. Through comprehensive experimental approaches including confocal microscopy, FRET-based sensors, and mutagenesis, we demonstrate that changes in pHi induce conformational alterations in STIM1, thereby modifying its subcellular localization and activity. We identify two conserved histidines within STIM1 essential for sensing pHi shifts. Moreover, intracellular alkalization induced by agents such as Angiotensin II or NH4Cl enhances STIM1-mediated SOCE, promoting cardiac hypertrophy. These findings reveal a novel facet of STIM1 as a multi-modal stress sensor that coordinates cellular responses to both Ca2+ and pH fluctuations. This dual functionality underscores its potential as a therapeutic target for diseases associated with pH and Ca2+ dysregulation.
Collapse
Affiliation(s)
- Yilan Chen
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Panpan Liu
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Ziyi Zhong
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Hanhan Zhang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Aomin Sun
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China.
| | - Youjun Wang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China; Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China.
| |
Collapse
|
4
|
Yasutake R, Nagoshi T, Yoshii A, Takahashi H, Oi Y, Kimura H, Kashiwagi Y, Tanaka TD, Tanaka Y, Yoshimura M. Suppression of B-type natriuretic peptide gene expression in cardiomyocytes under anoxic conditions. Peptides 2024; 182:171316. [PMID: 39490746 DOI: 10.1016/j.peptides.2024.171316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/19/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
Several cell biology studies have focused on the effects of hypoxic environments on cardiomyocytes. However, the effect of anoxic conditions on cardiomyocytes remains largely unexplored. In the present study, we investigated the direct effects of anoxia on B-type natriuretic peptide (BNP) gene expression in cardiomyocytes. Neonatal rat cardiomyocytes (NRCMs) were exposed to anoxia using an airtight chamber saturated with 95 % N2/5 % CO2. BNP mRNA levels in NRCM were substantially reduced after more than 8 h of anoxia exposure, whereas after reoxygenation, BNP gene expression levels recovered in a time-dependent manner and significantly increased after 24 h of reoxygenation. BNP mRNA levels suppressed under anoxic conditions were significantly increased by aldosterone-induced activation of sodium-proton exchanger 1 (NHE1), which was canceled by an NHE1 inhibitor, suggesting that anoxia reduces BNP gene expression, at least in part, in an NHE1-dependent manner. In summary, we found that BNP gene expression in cardiomyocytes decreases under anoxic conditions, in contrast to previous research findings that BNP expression increases under hypoxic conditions. These findings reveal a new insight that, within a single heart tissue in various cardiovascular diseases, such as myocardial infarction, the biological responses of cardiomyocytes are fundamentally different in regions of anoxia and hypoxia.
Collapse
Affiliation(s)
- Rei Yasutake
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Tomohisa Nagoshi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan.
| | - Akira Yoshii
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Hirotake Takahashi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Yuhei Oi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Haruka Kimura
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Yusuke Kashiwagi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Toshikazu D Tanaka
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Yoshiro Tanaka
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Michihiro Yoshimura
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| |
Collapse
|
5
|
Ahmad M, Flerin M, Tay HM, Thompson AL, Duarte F, Langton MJ. Stimuli-responsive anion transport utilising caged hydrazone-based anionophores. NANOSCALE 2024; 16:21545-21553. [PMID: 39480659 DOI: 10.1039/d4nr03220a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2024]
Abstract
Ion transport across biological membranes, facilitated by naturally occurring ion channels and pumps, plays a crucial role in biological processes. Gating is an important aspect of these systems, whereby transport is regulated by a range of external stimuli such as light, ligands and membrane potential. While synthetic ion transport systems, especially those with gating mechanisms, are rare, they have garnered significant attention due to their potential applications in targeted therapeutics as anticancer agents or to treat channelopathies. In this work, we report stimuli-responsive anion transporters based on dynamic hydrogen bonding interactions of hydroxyl-functionalised hydrazone anionophores. Caging of the hydroxyl groups with moities that are responsive to light and H2S locks the hydrazone protons through intramolecular hydrogen bonding, rendering them unavailable for anion binding and transport. Upon decaging with light or H2S, the hydrogen bonding pattern is reversed, rendering the hydrazone protons available for anion binding, and leading to efficient switch-on of ion transport across the lipid bilayer membrane.
Collapse
Affiliation(s)
- Manzoor Ahmad
- Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, UK.
| | - Martin Flerin
- Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, UK.
| | - Hui Min Tay
- Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, UK.
| | - Amber L Thompson
- Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, UK.
| | - Fernanda Duarte
- Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, UK.
| | | |
Collapse
|
6
|
Zhang H, Zhao J, Yu J, Zhang X, Ran S, Wang S, Ye W, Luo Z, Li X, Hao Y, Zong J, Li R, Lai L, Zheng K, Huang P, Zhou C, Wu J, Li Y, Xia J. Lactate metabolism and lactylation in cardiovascular disease: novel mechanisms and therapeutic targets. Front Cardiovasc Med 2024; 11:1489438. [PMID: 39664763 PMCID: PMC11631895 DOI: 10.3389/fcvm.2024.1489438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 11/11/2024] [Indexed: 12/13/2024] Open
Abstract
Cardiovascular disease (CVD) is responsible for approximately 30% of annual global mortality rates, yet existing treatments for this condition are considered less than ideal. Despite being previously overlooked, lactate, a byproduct of glycolysis, is now acknowledged for its crucial role in the cellular functions of the cardiovascular system. Recent studies have shown that lactate influences the proliferation, differentiation, and activation of immune cells through its modulation of post-translational protein modifications, thereby affecting the development and prognosis of cardiovascular disease. Consequently, there has been a notable increase in interest towards drug targets targeting lactylation in immune cells, prompting further exploration. In light of the swift advancements in this domain, this review article is dedicated to examining lactylation in cardiovascular disease and potential drug targets for regulating lactylation, with the aim of enhancing comprehension of this intricate field.
Collapse
Affiliation(s)
- Han Zhang
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jiulu Zhao
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jizhang Yu
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xi Zhang
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuan Ran
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Song Wang
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Weicong Ye
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zilong Luo
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaohan Li
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yanglin Hao
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Junjie Zong
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ran Li
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Longyong Lai
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Kexiao Zheng
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Pinyan Huang
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Cheng Zhou
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jie Wu
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, ChineseAcademy of Medical Sciences, Wuhan, Hubei, China
| | - Yuan Li
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, ChineseAcademy of Medical Sciences, Wuhan, Hubei, China
| | - Jiahong Xia
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, ChineseAcademy of Medical Sciences, Wuhan, Hubei, China
| |
Collapse
|
7
|
Tan NK, Chan H, Lu Z, Zreiqat H, Lakhwani G, Lesani P, New EJ. Ultrasensitive Dual Fluorophore-Conjugated Carbon Dots for Intracellular pH Sensing in 3D Tumor Models. ACS APPLIED MATERIALS & INTERFACES 2024; 16:47303-47313. [PMID: 39215383 DOI: 10.1021/acsami.4c10836] [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: 09/04/2024]
Abstract
The dysregulation of pH has been linked to the onset of chronic conditions, such as cancer and neurological diseases. Consequently, the development of a highly sensitive tool for intracellular pH sensing is imperative to investigate the interplay between pH and the biochemical changes accompanying disease pathogenesis. Here, we present the development of a ratiometric fluorescent nanoprobe, NpRhoDot, designed for precisely measuring pH levels. We demonstrate its efficacy in sensitively reporting intracellular pH in monolayer A549 lung cancer cells, primary fibroblast cells, and 3D tumor spheroids derived from the DLD-1 colorectal adenocarcinoma cell line. NpRhoDot leverages a novel design, where stable carbon dots are functionalized with a pH-responsive ratiometric fluorescent probe comprising a naphthalimide-rhodamine moiety, NpRho1. This design confers NpRhoDot with the high pH sensitivity characteristics of organic fluorescent probes, along with excellent photostability up to 1 h and biocompatibility of carbon dots. Through one-photon and two-photon fluorescence microscopy, we validate the reliability of NpRhoDot for biosensing intracellular pH in monolayer and three-dimensional tumor models from pH 4 to 7.
Collapse
Affiliation(s)
- Nian Kee Tan
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | - Hazel Chan
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | - Zufu Lu
- School of Biomedical Engineering, The University of Sydney, Sydney, NSW 2006, Australia
| | - Hala Zreiqat
- School of Biomedical Engineering, The University of Sydney, Sydney, NSW 2006, Australia
- The University of Sydney Nano Institute, Sydney, NSW 2006, Australia
| | - Girish Lakhwani
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
- The University of Sydney Nano Institute, Sydney, NSW 2006, Australia
- Australian Research Council Centre of Excellence in Exciton Science, The University of Sydney, Sydney, NSW 2006, Australia
| | - Pooria Lesani
- School of Biomedical Engineering, The University of Sydney, Sydney, NSW 2006, Australia
- The University of Sydney Nano Institute, Sydney, NSW 2006, Australia
- School of Science, STEM College, RMIT University, Melbourne, VIC 3000, Australia
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States of America
| | - Elizabeth J New
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
- The University of Sydney Nano Institute, Sydney, NSW 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
8
|
Jian L, Zhang Q, Yao D, Wang Q, Chen M, Xia Y, Li S, Shen Y, Cao M, Qin A, Li L, Cao Y. The structural insight into the functional modulation of human anion exchanger 3. Nat Commun 2024; 15:6134. [PMID: 39033175 PMCID: PMC11271275 DOI: 10.1038/s41467-024-50572-x] [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: 02/20/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024] Open
Abstract
Anion exchanger 3 (AE3) is pivotal in regulating intracellular pH across excitable tissues, yet its structural intricacies and functional dynamics remain underexplored compared to other anion exchangers. This study unveils the structural insights into human AE3, including the cryo-electron microscopy structures for AE3 transmembrane domains (TMD) and a chimera combining AE3 N-terminal domain (NTD) with AE2 TMD (hAE3NTD2TMD). Our analyzes reveal a substrate binding site, an NTD-TMD interlock mechanism, and a preference for an outward-facing conformation. Unlike AE2, which has more robust acid-loading capabilities, AE3's structure, including a less stable inward-facing conformation due to missing key NTD-TMD interactions, contributes to its moderated pH-modulating activity and increased sensitivity to the inhibitor DIDS. These structural differences underline AE3's distinct functional roles in specific tissues and underscore the complex interplay between structural dynamics and functional specificity within the anion exchanger family, enhancing our understanding of the physiological and pathological roles of the anion exchanger family.
Collapse
Affiliation(s)
- Liyan Jian
- Department of Orthopaedics, Shanghai Key Laboratory of Orthopaedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Precision Medicine, the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 115 Jinzun Road, Shanghai, China
| | - Qing Zhang
- Institute of Precision Medicine, the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 115 Jinzun Road, Shanghai, China
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, USA
| | - Deqiang Yao
- Institute of Precision Medicine, the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 115 Jinzun Road, Shanghai, China
- Institute of Aging & Tissue Regeneration, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Wang
- Institute of Precision Medicine, the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 115 Jinzun Road, Shanghai, China
| | - Moxin Chen
- Department of Ophthalmology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Ying Xia
- Institute of Precision Medicine, the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 115 Jinzun Road, Shanghai, China
| | - Shaobai Li
- Institute of Precision Medicine, the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 115 Jinzun Road, Shanghai, China
| | - Yafeng Shen
- Institute of Precision Medicine, the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 115 Jinzun Road, Shanghai, China
| | - Mi Cao
- Institute of Precision Medicine, the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 115 Jinzun Road, Shanghai, China
| | - An Qin
- Department of Orthopaedics, Shanghai Key Laboratory of Orthopaedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Department of Orthopaedics, Shanghai Frontiers Science Center of Degeneration and Regeneration in Skeletal System, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Lin Li
- Department of Ophthalmology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| | - Yu Cao
- Department of Orthopaedics, Shanghai Key Laboratory of Orthopaedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Institute of Precision Medicine, the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 115 Jinzun Road, Shanghai, China.
| |
Collapse
|
9
|
Weng H, Zou W, Tian F, Xie H, Liu A, Liu W, Liu Y, Zhou N, Cai X, Wu J, Zheng Y, Shu X. Inhalable cardiac targeting peptide modified nanomedicine prevents pressure overload heart failure in male mice. Nat Commun 2024; 15:6058. [PMID: 39025877 PMCID: PMC11258261 DOI: 10.1038/s41467-024-50312-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 07/08/2024] [Indexed: 07/20/2024] Open
Abstract
Heart failure causes considerable morbidity and mortality worldwide. Clinically applied drugs for the treatment of heart failure are still severely limited by poor delivery efficiency to the heart and off-target consumption. Inspired by the high heart delivery efficiency of inhaled drugs, we present an inhalable cardiac-targeting peptide (CTP)-modified calcium phosphate (CaP) nanoparticle for the delivery of TP-10, a selective inhibitor of PDE10A. The CTP modification significantly promotes cardiomyocyte and fibroblast targeting during the pathological state of heart failure in male mice. TP-10 is subsequently released from TP-10@CaP-CTP and effectively attenuates cardiac remodelling and improved cardiac function. In view of these results, a low dosage (2.5 mg/kg/2 days) of inhaled medication exerted good therapeutic effects without causing severe lung injury after long-term treatment. In addition, the mechanism underlying the amelioration of heart failure is investigated, and the results reveal that the therapeutic effects of this system on cardiomyocytes and cardiac fibroblasts are mainly mediated through the cAMP/AMPK and cGMP/PKG signalling pathways. By demonstrating the targeting capacity of CTP and verifying the biosafety of inhalable CaP nanoparticles in the lung, this work provides a perspective for exploring myocardium-targeted therapy and presents a promising clinical strategy for the long-term management of heart failure.
Collapse
Affiliation(s)
- Haobo Weng
- Department of Echocardiography, Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, Shanghai, PR China
- Shanghai Key Laboratory of Neuro-Ultrasound for Diagnosis and Treatment, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Weijuan Zou
- Shanghai Key Laboratory of Neuro-Ultrasound for Diagnosis and Treatment, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Fangyan Tian
- Department of Echocardiography, Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, Shanghai, PR China
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai, PR China
- Department of Ultrasound Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Huilin Xie
- Department of Echocardiography, Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, Shanghai, PR China
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Ao Liu
- Department of Echocardiography, Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, Shanghai, PR China
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Wen Liu
- Department of Echocardiography, Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Yu Liu
- Department of Echocardiography, Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Nianwei Zhou
- Department of Echocardiography, Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Xiaojun Cai
- Shanghai Key Laboratory of Neuro-Ultrasound for Diagnosis and Treatment, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Jianrong Wu
- Shanghai Key Laboratory of Neuro-Ultrasound for Diagnosis and Treatment, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| | - Yuanyi Zheng
- Shanghai Key Laboratory of Neuro-Ultrasound for Diagnosis and Treatment, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| | - Xianhong Shu
- Department of Echocardiography, Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, Shanghai, PR China.
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai, PR China.
- Department of Ultrasound in Medicine, Shanghai Xuhui District Central Hospital, Shanghai, PR China.
| |
Collapse
|
10
|
Ahmad M, Johnson TG, Flerin M, Duarte F, Langton MJ. Responsive Anionophores with AND Logic Multi-Stimuli Activation. Angew Chem Int Ed Engl 2024; 63:e202403314. [PMID: 38517056 DOI: 10.1002/anie.202403314] [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: 02/16/2024] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 03/23/2024]
Abstract
Artificial ion transport systems have emerged as an important class of compounds that promise applications in chemotherapeutics as anticancer agents or to treat channelopathies. Stimulus-responsive systems that offer spatiotemporally controlled activity for targeted applications remain rare. Here we utilize dynamic hydrogen bonding interactions of a 4,6-dihydroxy-isophthalamide core to generate a modular platform enabling access to stimuli-responsive ion transporters that can be activated in response to a wide variety of external stimuli, including light, redox, and enzymes, with excellent OFF-ON activation profiles. Alkylation of the two free hydroxyl groups with stimulus-responsive moieties locks the amide bonds through intramolecular hydrogen bonding and hence makes them unavailable for anion binding and transport. Triggering using a particular stimulus to cleave both cages reverses the hydrogen bonding arrangement, to generate a highly preorganized anion binding cavity for efficient transmembrane transport. Integration of two cages that are responsive to orthogonal stimuli enables multi-stimuli activation, where both stimuli are required to trigger transport in an AND logic process. Importantly, the strategy provides a facile method to post-functionalize the highly active transporter core with a variety of stimulus-responsive moieties for targeted activation with multiple triggers.
Collapse
Affiliation(s)
- Manzoor Ahmad
- Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK
| | - Toby G Johnson
- Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK
| | - Martin Flerin
- Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK
| | - Fernanda Duarte
- Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK
| | - Matthew J Langton
- Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK
| |
Collapse
|
11
|
Scranton K, John S, Angelini M, Steccanella F, Umar S, Zhang R, Goldhaber JI, Olcese R, Ottolia M. Cardiac function is regulated by the sodium-dependent inhibition of the sodium-calcium exchanger NCX1. Nat Commun 2024; 15:3831. [PMID: 38714663 PMCID: PMC11076594 DOI: 10.1038/s41467-024-47850-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 04/15/2024] [Indexed: 05/10/2024] Open
Abstract
The Na+-Ca2+ exchanger (NCX1) is the dominant Ca2+ extrusion mechanism in cardiac myocytes. NCX1 activity is inhibited by intracellular Na+ via a process known as Na+-dependent inactivation. A central question is whether this inactivation plays a physiological role in heart function. Using CRISPR/Cas9, we inserted the K229Q mutation in the gene (Slc8a1) encoding for NCX1. This mutation removes the Na+-dependent inactivation while preserving transport properties and other allosteric regulations. NCX1 mRNA levels, protein expression, and protein localization are unchanged in K229Q male mice. However, they exhibit reduced left ventricular ejection fraction and fractional shortening, while displaying a prolonged QT interval. K229Q ventricular myocytes show enhanced NCX1 activity, resulting in action potential prolongation, higher incidence of aberrant action potentials, a faster decline of Ca2+ transients, and depressed cell shortening. The results demonstrate that NCX1 Na+-dependent inactivation plays an essential role in heart function by affecting both cardiac excitability and contractility.
Collapse
Affiliation(s)
- Kyle Scranton
- Department of Anesthesiology & Perioperative Medicine, Division of Molecular Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Scott John
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Marina Angelini
- Department of Anesthesiology & Perioperative Medicine, Division of Molecular Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Federica Steccanella
- Department of Anesthesiology & Perioperative Medicine, Division of Molecular Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Soban Umar
- Department of Anesthesiology & Perioperative Medicine, Division of Molecular Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Rui Zhang
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Joshua I Goldhaber
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Riccardo Olcese
- Department of Anesthesiology & Perioperative Medicine, Division of Molecular Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Michela Ottolia
- Department of Anesthesiology & Perioperative Medicine, Division of Molecular Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
12
|
Sharma R, Sarkar S, Chattopadhayay S, Mondal J, Talukdar P. A Halogen-Bond-Driven Artificial Chloride-Selective Channel Constructed from 5-Iodoisophthalamide-based Molecules. Angew Chem Int Ed Engl 2024; 63:e202319919. [PMID: 38299773 DOI: 10.1002/anie.202319919] [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: 12/22/2023] [Revised: 01/25/2024] [Accepted: 02/01/2024] [Indexed: 02/02/2024]
Abstract
Despite considerable emphasis on advancing artificial ion channels, progress is constrained by the limited availability of small molecules with the necessary attributes of self-assembly and ion selectivity. In this study, a library of small molecules based on 5-haloisophthalamide and a non-halogenated isophthalamide were examined for their ion transport properties across the lipid bilayer membranes, and the finding demonstrates that the di-hexyl-substituted 5-iodoisophthalamide derivative exhibits the highest level of activity. Furthermore, it was established that the highest active compound facilitates the selective chloride transport that occurs via an antiport-mediated mechanism. The crystal structure of the compound unveils a distinctive self-assembly of molecules, forming a zig-zag channel pore that is well-suited for the permeation of anions. Planar bilayer conductance measurements proved the formation of chloride selective channels. A molecular dynamics simulation study, relying on the self-assembled component derived from the crystal structure, affirmed the paramount significance of intermolecular hydrogen bonding in the formation of supramolecular barrel-rosette structures that span the bilayer. Furthermore, it was demonstrated that the transport of chloride across the lipid bilayer membrane is facilitated by the synergistic effects of halogen bonding and hydrogen bonding within the channel.
Collapse
Affiliation(s)
- Rashmi Sharma
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Dr. Homi Bhabha Road, Pashan, Pune, 411008, Maharashtra, India
| | - Susmita Sarkar
- Center for Interdisciplinary Sciences, Tata Institute of Fundamental Research, Hyderabad, 500046 Telangana, India
| | - Sandip Chattopadhayay
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Dr. Homi Bhabha Road, Pashan, Pune, 411008, Maharashtra, India
| | - Jagannath Mondal
- Center for Interdisciplinary Sciences, Tata Institute of Fundamental Research, Hyderabad, 500046 Telangana, India
| | - Pinaki Talukdar
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Dr. Homi Bhabha Road, Pashan, Pune, 411008, Maharashtra, India
| |
Collapse
|
13
|
Das S, Kumar P. Exploring the carbonic anhydrase-mimetic [(PMDTA) 2ZnII2(OH -) 2] 2+ for nitric oxide monooxygenation. Dalton Trans 2024; 53:6173-6177. [PMID: 38501600 DOI: 10.1039/d4dt00407h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
In biology, nitrite (NO2-) serves as a storage pool of nitric oxide (NO); however, the formation of NO2- from NO is still under investigation. Here, we report the NO monooxygenation (NOM) reaction of a ZnII-hydroxide complex (1), producing a ZnII-nitrito complex {2, (ZnII-NO2-)} + H2.
Collapse
Affiliation(s)
- Sandip Das
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Tirupati 517507, India.
| | - Pankaj Kumar
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Tirupati 517507, India.
| |
Collapse
|
14
|
Tabata Fukushima C, Dancil IS, Clary H, Shah N, Nadtochiy SM, Brookes PS. Reactive oxygen species generation by reverse electron transfer at mitochondrial complex I under simulated early reperfusion conditions. Redox Biol 2024; 70:103047. [PMID: 38295577 PMCID: PMC10844975 DOI: 10.1016/j.redox.2024.103047] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 02/02/2024] Open
Abstract
Ischemic tissues accumulate succinate, which is rapidly oxidized upon reperfusion, driving a burst of mitochondrial reactive oxygen species (ROS) generation that triggers cell death. In isolated mitochondria with succinate as the sole metabolic substrate under non-phosphorylating conditions, 90 % of ROS generation is from reverse electron transfer (RET) at the Q site of respiratory complex I (Cx-I). Together, these observations suggest Cx-I RET is the source of pathologic ROS in reperfusion injury. However, numerous factors present in early reperfusion may impact Cx-I RET, including: (i) High [NADH]; (ii) High [lactate]; (iii) Mildly acidic pH; (iv) Defined ATP/ADP ratios; (v) Presence of the nucleosides adenosine and inosine; and (vi) Defined free [Ca2+]. Herein, experiments with mouse cardiac mitochondria revealed that under simulated early reperfusion conditions including these factors, total mitochondrial ROS generation was only 56 ± 17 % of that seen with succinate alone (mean ± 95 % confidence intervals). Of this ROS, only 52 ± 20 % was assignable to Cx-I RET. A further 14 ± 7 % could be assigned to complex III, with the remainder (34 ± 11 %) likely originating from other ROS sources upstream of the Cx-I Q site. Together, these data suggest the relative contribution of Cx-I RET ROS to reperfusion injury may be overestimated, and other ROS sources may contribute a significant fraction of ROS in early reperfusion.
Collapse
Affiliation(s)
- Caio Tabata Fukushima
- Departments of Anesthesiology, University of Rochester Medical Center, USA; Departments of Biochemistry, University of Rochester Medical Center, USA; Pharmacology and Physiology, University of Rochester Medical Center, USA
| | - Ian-Shika Dancil
- Departments of Anesthesiology, University of Rochester Medical Center, USA
| | - Hannah Clary
- Departments of Biochemistry, University of Rochester Medical Center, USA
| | - Nidhi Shah
- Pharmacology and Physiology, University of Rochester Medical Center, USA
| | - Sergiy M Nadtochiy
- Departments of Anesthesiology, University of Rochester Medical Center, USA
| | - Paul S Brookes
- Departments of Anesthesiology, University of Rochester Medical Center, USA; Pharmacology and Physiology, University of Rochester Medical Center, USA.
| |
Collapse
|
15
|
Holmberg SR, Sakamoto Y, Kato A, Romero MF. The role of Na +-coupled bicarbonate transporters (NCBT) in health and disease. Pflugers Arch 2024; 476:479-503. [PMID: 38536494 PMCID: PMC11338471 DOI: 10.1007/s00424-024-02937-w] [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: 12/15/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 04/11/2024]
Abstract
Cellular and organism survival depends upon the regulation of pH, which is regulated by highly specialized cell membrane transporters, the solute carriers (SLC) (For a comprehensive list of the solute carrier family members, see: https://www.bioparadigms.org/slc/ ). The SLC4 family of bicarbonate (HCO3-) transporters consists of ten members, sorted by their coupling to either sodium (NBCe1, NBCe2, NBCn1, NBCn2, NDCBE), chloride (AE1, AE2, AE3), or borate (BTR1). The ionic coupling of SLC4A9 (AE4) remains controversial. These SLC4 bicarbonate transporters may be controlled by cellular ionic gradients, cellular membrane voltage, and signaling molecules to maintain critical cellular and systemic pH (acid-base) balance. There are profound consequences when blood pH deviates even a small amount outside the normal range (7.35-7.45). Chiefly, Na+-coupled bicarbonate transporters (NCBT) control intracellular pH in nearly every living cell, maintaining the biological pH required for life. Additionally, NCBTs have important roles to regulate cell volume and maintain salt balance as well as absorption and secretion of acid-base equivalents. Due to their varied tissue expression, NCBTs have roles in pathophysiology, which become apparent in physiologic responses when their expression is reduced or genetically deleted. Variations in physiological pH are seen in a wide variety of conditions, from canonically acid-base related conditions to pathologies not necessarily associated with acid-base dysfunction such as cancer, glaucoma, or various neurological diseases. The membranous location of the SLC4 transporters as well as recent advances in discovering their structural biology makes them accessible and attractive as a druggable target in a disease context. The role of sodium-coupled bicarbonate transporters in such a large array of conditions illustrates the potential of treating a wide range of disease states by modifying function of these transporters, whether that be through inhibition or enhancement.
Collapse
Affiliation(s)
- Shannon R Holmberg
- Physiology & Biomedical Engineering, Mayo Clinic College of Medicine & Science, 200 1st Street SW, Rochester, MN 55905, USA
- Biochemistry & Molecular Biology, Mayo Clinic College of Medicine & Science, 200 1st Street SW, Rochester, MN, USA
| | - Yohei Sakamoto
- School of Life Science and Technology, Tokyo Institute of Technology, Midori-Ku, Yokohama, 226-8501, Japan
| | - Akira Kato
- School of Life Science and Technology, Tokyo Institute of Technology, Midori-Ku, Yokohama, 226-8501, Japan
| | - Michael F Romero
- Physiology & Biomedical Engineering, Mayo Clinic College of Medicine & Science, 200 1st Street SW, Rochester, MN 55905, USA.
- Nephrology & Hypertension, Mayo Clinic College of Medicine & Science, 200 1st Street SW, Rochester, MN, USA.
| |
Collapse
|
16
|
Park KC, Crump NT, Louwman N, Krywawych S, Cheong YJ, Vendrell I, Gill EK, Gunadasa-Rohling M, Ford KL, Hauton D, Fournier M, Pires E, Watson L, Roseman G, Holder J, Koschinski A, Carnicer R, Curtis MK, Zaccolo M, Hulikova A, Fischer R, Kramer HB, McCullagh JSO, Trefely S, Milne TA, Swietach P. Disrupted propionate metabolism evokes transcriptional changes in the heart by increasing histone acetylation and propionylation. NATURE CARDIOVASCULAR RESEARCH 2023; 2:1221-1245. [PMID: 38500966 PMCID: PMC7615744 DOI: 10.1038/s44161-023-00365-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 10/15/2023] [Indexed: 03/20/2024]
Abstract
Propiogenic substrates and gut bacteria produce propionate, a post-translational protein modifier. In this study, we used a mouse model of propionic acidaemia (PA) to study how disturbances to propionate metabolism result in histone modifications and changes to gene expression that affect cardiac function. Plasma propionate surrogates were raised in PA mice, but female hearts manifested more profound changes in acyl-CoAs, histone propionylation and acetylation, and transcription. These resulted in moderate diastolic dysfunction with raised diastolic Ca2+, expanded end-systolic ventricular volume and reduced stroke volume. Propionate was traced to histone H3 propionylation and caused increased acetylation genome-wide, including at promoters of Pde9a and Mme, genes related to contractile dysfunction through downscaled cGMP signaling. The less severe phenotype in male hearts correlated with β-alanine buildup. Raising β-alanine in cultured myocytes treated with propionate reduced propionyl-CoA levels, indicating a mechanistic relationship. Thus, we linked perturbed propionate metabolism to epigenetic changes that impact cardiac function.
Collapse
Affiliation(s)
- Kyung Chan Park
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK
| | - Nicholas T. Crump
- MRC Molecular Haematology Unit, Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Present Address: Hugh and Josseline Langmuir Centre for Myeloma Research, Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Niamh Louwman
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK
| | - Steve Krywawych
- Department of Chemical Pathology, Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Yuen Jian Cheong
- Epigenetics & Signalling Programmes, Babraham Institute, Cambridge, UK
| | - Iolanda Vendrell
- Nuffield Department of Medicine, Target Discovery Institute, Oxford, UK
- Nuffield Department of Medicine, Chinese Academy for Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
| | - Eleanor K. Gill
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK
| | | | - Kerrie L. Ford
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK
| | - David Hauton
- Department of Chemistry, University of Oxford, Oxford, UK
| | | | | | - Lydia Watson
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK
| | - Gerald Roseman
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK
| | - James Holder
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Andreas Koschinski
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK
| | - Ricardo Carnicer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - M. Kate Curtis
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK
| | - Manuela Zaccolo
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK
| | - Alzbeta Hulikova
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK
| | - Roman Fischer
- Nuffield Department of Medicine, Target Discovery Institute, Oxford, UK
- Nuffield Department of Medicine, Chinese Academy for Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
| | - Holger B. Kramer
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | | | - Sophie Trefely
- Epigenetics & Signalling Programmes, Babraham Institute, Cambridge, UK
| | - Thomas A. Milne
- MRC Molecular Haematology Unit, Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Pawel Swietach
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
17
|
Costa ADS, Ghouri I, Johnston A, McGlynn K, McNair A, Bowman P, Malik N, Hurren J, Bingelis T, Dunne M, Smith GL, Kemi OJ. Electrically stimulated in vitro heart cell mimic of acute exercise reveals novel immediate cellular responses to exercise: Reduced contractility and metabolism, but maintained calcium cycling and increased myofilament calcium sensitivity. Cell Biochem Funct 2023; 41:1147-1161. [PMID: 37665041 PMCID: PMC10947300 DOI: 10.1002/cbf.3847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 08/14/2023] [Accepted: 08/21/2023] [Indexed: 09/05/2023]
Abstract
Cardiac cellular responses to acute exercise remain undescribed. We present a model for mimicking acute aerobic endurance exercise to freshly isolated cardiomyocytes by evoking exercise-like contractions over prolonged periods of time with trains of electrical twitch stimulations. We then investigated immediate contractile, Ca2+ , and metabolic responses to acute exercise in perfused freshly isolated left ventricular rat cardiomyocytes, after a matrix-design optimized protocol and induced a mimic for acute aerobic endurance exercise by trains of prolonged field twitch stimulations. Acute exercise decreased cardiomyocyte fractional shortening 50%-80% (p < .01). This was not explained by changes to intracellular Ca2+ handling (p > .05); rather, we observed a weak insignificant Ca2+ transient increase (p = .11), while myofilament Ca2+ sensitivity increased 20%-70% (p < .05). Acidic pH 6.8 decreased fractional shortening 20%-70% (p < .05) because of 20%-30% decreased Ca2+ transients (p < .05), but no difference occurred between control and acute exercise (p > .05). Addition of 1 or 10 mM La- increased fractional shortening in control (1 mM La- : no difference, p > .05; 10 mM La- : 20%-30%, p < .05) and acute exercise (1 mM La- : 40%-90%, p < .01; 10 mM La- : 50%-100%, p < .01) and rendered acute exercise indifferent from control (p > .05). Intrinsic autofluorescence showed a resting NADstate of 0.59 ± 0.04 and FADstate of 0.17 ± 0.03, while acute exercise decreased NADH/FAD ratio 8% (p < .01), indicating intracellular oxidation. In conclusion, we show a novel approach for studying immediate acute cardiomyocyte responses to aerobic endurance exercise. We find that acute exercise in cardiomyocytes decreases contraction, but Ca2+ handling and myofilament Ca2+ sensitivity compensate for this, while acidosis and reduced energy substrate and mitochondrial ATP generation explain this.
Collapse
Affiliation(s)
- Ana Da Silva Costa
- School of Cardiovascular and Metabolic Health, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
- Graduate School, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Iffath Ghouri
- School of Cardiovascular and Metabolic Health, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Alexander Johnston
- School of Cardiovascular and Metabolic Health, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Karen McGlynn
- School of Cardiovascular and Metabolic Health, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Andrew McNair
- School of Cardiovascular and Metabolic Health, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Peter Bowman
- School of Cardiovascular and Metabolic Health, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Natasha Malik
- School of Cardiovascular and Metabolic Health, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Johanne Hurren
- School of Cardiovascular and Metabolic Health, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Tomas Bingelis
- School of Cardiovascular and Metabolic Health, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Michael Dunne
- School of Cardiovascular and Metabolic Health, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Godfrey L. Smith
- School of Cardiovascular and Metabolic Health, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Ole J. Kemi
- School of Cardiovascular and Metabolic Health, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| |
Collapse
|
18
|
Vetter VC, Bouten CVC, van der Pol A. Hydrogels for Cardiac Restorative Support: Relevance of Gelation Mechanisms for Prospective Clinical Use. Curr Heart Fail Rep 2023; 20:519-529. [PMID: 37812347 PMCID: PMC10746579 DOI: 10.1007/s11897-023-00630-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/20/2023] [Indexed: 10/10/2023]
Abstract
PURPOSE OF REVIEW Cardiac tissue regenerative strategies have gained much traction over the years, in particular those utilizing hydrogels. With our review, and with special focus on supporting post-myocardial infarcted tissue, we aim to provide insights in determining crucial design considerations of a hydrogel and the implications these could have for future clinical use. RECENT FINDINGS To date, two hydrogel delivery strategies are being explored, cardiac injection or patch, to treat myocardial infarction. Recent advances have demonstrated that the mechanism by which a hydrogel is gelated (i.e., physically or chemically cross-linked) not only impacts the biocompatibility, mechanical properties, and chemical structure, but also the route of delivery of the hydrogel and thus its effect on cardiac repair. With regard to cardiac regeneration, various hydrogels have been developed with the ability to function as a delivery system for therapeutic strategies (e.g., drug and stem cells treatments), as well as a scaffold to guide cardiac tissue regeneration following myocardial infarction. However, these developments remain within the experimental and pre-clinical realm and have yet to transition towards the clinical setting.
Collapse
Affiliation(s)
- Valentine C Vetter
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Carlijn V C Bouten
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Atze van der Pol
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands.
| |
Collapse
|
19
|
Fukushima CT, Dancil IS, Clary H, Shah N, Nadtochiy SM, Brookes PS. Reactive Oxygen Species Generation by Reverse Electron Transfer at Mitochondrial Complex I Under Simulated Early Reperfusion Conditions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.21.568136. [PMID: 38045326 PMCID: PMC10690194 DOI: 10.1101/2023.11.21.568136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Ischemic tissues accumulate succinate, which is rapidly oxidized upon reperfusion, driving a burst of mitochondrial reactive oxygen species (ROS) generation that triggers cell death. In isolated mitochondria with succinate as the sole metabolic substrate under non-phosphorylating conditions, 90% of ROS generation is from reverse electron transfer (RET) at the Q site of respiratory complex I (Cx-I). Together, these observations suggest Cx-I RET is the source of pathologic ROS in reperfusion injury. However, numerous factors present in early reperfusion may impact Cx-I RET, including: (i) High [NADH]; (ii) High [lactate]; (iii) Mildly acidic pH; (iv) Defined ATP/ADP ratios; (v) Presence of the nucleosides adenosine and inosine; and (vi) Defined free [Ca2+]. Herein, experiments with mouse cardiac mitochondria revealed that under simulated early reperfusion conditions including these factors, overall mitochondrial ROS generation was only 56% of that seen with succinate alone, and only 52% of this ROS was assignable to Cx-I RET. The residual non-RET ROS could be partially assigned to complex III (Cx-III) with the remainder likely originating from other ROS sources upstream of the Cx-I Q site. Together, these data suggest the relative contribution of Cx-I RET ROS to reperfusion injury may be overestimated, and other ROS sources may contribute a significant fraction of ROS in early reperfusion.
Collapse
Affiliation(s)
- Caio Tabata Fukushima
- Department of Anesthesiology, University of Rochester Medical Center
- Department of Biochemistry, University of Rochester Medical Center
- Department of Pharmacology and Physiology, University of Rochester Medical Center
| | - Ian-Shika Dancil
- Department of Anesthesiology, University of Rochester Medical Center
| | - Hannah Clary
- Department of Biochemistry, University of Rochester Medical Center
| | - Nidhi Shah
- Department of Pharmacology and Physiology, University of Rochester Medical Center
| | | | - Paul S. Brookes
- Department of Anesthesiology, University of Rochester Medical Center
- Department of Pharmacology and Physiology, University of Rochester Medical Center
| |
Collapse
|
20
|
Kim J, Shanmugasundaram A, Kim DS, Jeong YJ, Kanade PP, Kim ES, Lee BK, Lee DW. Quantitative assessment of cardiomyocyte mechanobiology through high-throughput cantilever-based functional well plate systems. Analyst 2023; 148:5133-5143. [PMID: 37695027 DOI: 10.1039/d3an01286g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Proper regulation of the in vitro cell culture environment is essential for disease modelling and drug toxicity screening. The main limitation of well plates used for cell culture is that they cannot accurately maintain energy sources and compounds needed during cell growth. Herein, to understand the importance of perfusion in cardiomyocyte culture, changes in contractile force and heart rate during cardiomyocyte growth are systematically investigated, and the results are compared with those of a perfusion-free system. The proposed perfusion system consists of a Peltier refrigerator, a peristaltic pump, and a functional well plate. A functional well plate with 12 wells is made through injection moulding, with two tubes integrated in the cover for each well to continuously circulate the culture medium. The contractile force of cardiomyocytes growing on the cantilever surface is analysed through changes in cantilever displacement. The maturation of cardiomyocytes is evaluated through fluorescence staining and western blot; cardiomyocytes cultured in the perfusion system show greater maturity than those cultured in a manually replaced culture medium. The pH of the culture medium manually replaced at intervals of 3 days decreases to 6.8, resulting in an abnormal heartbeat, while cardiomyocytes cultured in the perfusion system maintained at pH 7.4 show improved contractility and a uniform heart rate. Two well-known ion channel blockers, verapamil and quinidine, are used to measure changes in the contractile force of cardiomyocytes from the two systems. Cardiomyocytes in the perfusion system show greater stability during drug toxicity screening, proving that the perfusion system provides a better environment for cell growth.
Collapse
Affiliation(s)
- Jongyun Kim
- MEMS and Nanotechnology Laboratory, School of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea.
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Arunkumar Shanmugasundaram
- MEMS and Nanotechnology Laboratory, School of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea.
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Dong-Su Kim
- MEMS and Nanotechnology Laboratory, School of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea.
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
- Green Energy & Nano Technology R&D Group, Korea Institute of Industrial Technology (KITECH), Gwangju, 61012, Republic of Korea
| | - Yun-Jin Jeong
- MEMS and Nanotechnology Laboratory, School of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea.
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Pooja P Kanade
- MEMS and Nanotechnology Laboratory, School of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea.
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Eung-Sam Kim
- Department of Biological Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea
- Center for Next-Generation Sensor Research and Development, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Bong-Kee Lee
- MEMS and Nanotechnology Laboratory, School of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea.
- Center for Next-Generation Sensor Research and Development, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Dong-Weon Lee
- MEMS and Nanotechnology Laboratory, School of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea.
- Center for Next-Generation Sensor Research and Development, Chonnam National University, Gwangju 61186, Republic of Korea
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
| |
Collapse
|
21
|
Shaul D, Lev-Cohain N, Sapir G, Sosna J, Gomori JM, Joskowicz L, Katz-Brull R. Real-time influence of intracellular acidification and Na + /H + exchanger inhibition on in-cell pyruvate metabolism in the perfused mouse heart: A 31 P-NMR and hyperpolarized 13 C-NMR study. NMR IN BIOMEDICINE 2023; 36:e4993. [PMID: 37424280 DOI: 10.1002/nbm.4993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/31/2023] [Accepted: 06/01/2023] [Indexed: 07/11/2023]
Abstract
Disruption of acid-base balance is linked to various diseases and conditions. In the heart, intracellular acidification is associated with heart failure, maladaptive cardiac hypertrophy, and myocardial ischemia. Previously, we have reported that the ratio of the in-cell lactate dehydrogenase (LDH) to pyruvate dehydrogenase (PDH) activities is correlated with cardiac pH. To further characterize the basis for this correlation, these in-cell activities were investigated under induced intracellular acidification without and with Na+ /H+ exchanger (NHE1) inhibition by zoniporide. Male mouse hearts (n = 30) were isolated and perfused retrogradely. Intracellular acidification was performed in two ways: (1) with the NH4 Cl prepulse methodology; and (2) by combining the NH4 Cl prepulse with zoniporide. 31 P NMR spectroscopy was used to determine the intracellular cardiac pH and to quantify the adenosine triphosphate and phosphocreatine content. Hyperpolarized [1-13 C]pyruvate was obtained using dissolution dynamic nuclear polarization. 13 C NMR spectroscopy was used to monitor hyperpolarized [1-13 C]pyruvate metabolism and determine enzyme activities in real time at a temporal resolution of a few seconds using the product-selective saturating excitation approach. The intracellular acidification induced by the NH4 Cl prepulse led to reduced LDH and PDH activities (-16% and -39%, respectively). This finding is in line with previous evidence of reduced myocardial contraction and therefore reduced metabolic activity upon intracellular acidification. Concomitantly, the LDH/PDH activity ratio increased with the reduction in pH, as previously reported. Combining the NH4 Cl prepulse with zoniporide led to a greater reduction in LDH activity (-29%) and to increased PDH activity (+40%). These changes resulted in a surprising decrease in the LDH/PDH ratio, as opposed to previous predictions. Zoniporide alone (without intracellular acidification) did not change these enzyme activities. A possible explanation for the enzymatic changes observed during the combination of the NH4 Cl prepulse and NHE1 inhibition may be related to mitochondrial NHE1 inhibition, which likely negates the mitochondrial matrix acidification. This effect, combined with the increased acidity in the cytosol, would result in an enhanced H+ gradient across the mitochondrial membrane and a temporarily higher pyruvate transport into the mitochondria, thereby increasing the PDH activity at the expense of the cytosolic LDH activity. These findings demonstrate the complexity of in-cell cardiac metabolism and its dependence on intracellular acidification. This study demonstrates the capabilities and limitations of hyperpolarized [1-13 C]pyruvate in the characterization of intracellular acidification as regards cardiac pathologies.
Collapse
Affiliation(s)
- David Shaul
- Department of Radiology, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Wohl Institute for Translational Medicine, Hadassah Medical Organization, Jerusalem, Israel
| | - Naama Lev-Cohain
- Department of Radiology, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Gal Sapir
- Department of Radiology, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Wohl Institute for Translational Medicine, Hadassah Medical Organization, Jerusalem, Israel
| | - Jacob Sosna
- Department of Radiology, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - J Moshe Gomori
- Department of Radiology, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Leo Joskowicz
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Rachel Katz-Brull
- Department of Radiology, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Wohl Institute for Translational Medicine, Hadassah Medical Organization, Jerusalem, Israel
| |
Collapse
|
22
|
Bustin A, Witschey WRT, van Heeswijk RB, Cochet H, Stuber M. Magnetic resonance myocardial T1ρ mapping : Technical overview, challenges, emerging developments, and clinical applications. J Cardiovasc Magn Reson 2023; 25:34. [PMID: 37331930 DOI: 10.1186/s12968-023-00940-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 05/15/2023] [Indexed: 06/20/2023] Open
Abstract
The potential of cardiac magnetic resonance to improve cardiovascular care and patient management is considerable. Myocardial T1-rho (T1ρ) mapping, in particular, has emerged as a promising biomarker for quantifying myocardial injuries without exogenous contrast agents. Its potential as a contrast-agent-free ("needle-free") and cost-effective diagnostic marker promises high impact both in terms of clinical outcomes and patient comfort. However, myocardial T1ρ mapping is still at a nascent stage of development and the evidence supporting its diagnostic performance and clinical effectiveness is scant, though likely to change with technological improvements. The present review aims at providing a primer on the essentials of myocardial T1ρ mapping, and to describe the current range of clinical applications of the technique to detect and quantify myocardial injuries. We also delineate the important limitations and challenges for clinical deployment, including the urgent need for standardization, the evaluation of bias, and the critical importance of clinical testing. We conclude by outlining technical developments to be expected in the future. If needle-free myocardial T1ρ mapping is shown to improve patient diagnosis and prognosis, and can be effectively integrated in cardiovascular practice, it will fulfill its potential as an essential component of a cardiac magnetic resonance examination.
Collapse
Affiliation(s)
- Aurelien Bustin
- IHU LIRYC, Electrophysiology and Heart Modeling Institute, Université de Bordeaux, INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, Avenue du Haut Lévêque, 33604, Pessac, France.
- Department of Cardiovascular Imaging, Hôpital Cardiologique du Haut-Lévêque, CHU de Bordeaux, Avenue de Magellan, 33604, Pessac, France.
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
| | | | - Ruud B van Heeswijk
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Hubert Cochet
- IHU LIRYC, Electrophysiology and Heart Modeling Institute, Université de Bordeaux, INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, Avenue du Haut Lévêque, 33604, Pessac, France
- Department of Cardiovascular Imaging, Hôpital Cardiologique du Haut-Lévêque, CHU de Bordeaux, Avenue de Magellan, 33604, Pessac, France
| | - Matthias Stuber
- IHU LIRYC, Electrophysiology and Heart Modeling Institute, Université de Bordeaux, INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, Avenue du Haut Lévêque, 33604, Pessac, France
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Center for Biomedical Imaging (CIBM), Lausanne, Switzerland
| |
Collapse
|
23
|
Sharma R, Vijay A, Chattopadhayay S, Mukherjee A, Talukdar P. Self-assembled anion channel formation by bis(1,3-propanediol)-linked meta-dipropynylbenzene-based small molecules. Chem Commun (Camb) 2023; 59:3602-3605. [PMID: 36883913 DOI: 10.1039/d2cc05155a] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Two self-assembled barrel-rosette ion channels have been developed using bis(1,3-propanediol)-linked m-dipropynylbenzene-based molecules. The system with an additional amide arm acted as a better channel compared to that having an ester arm. The amide-linked channel displayed substantial channel activity and excellent chloride selectivity in the lipid bilayer membranes. Molecular dynamics simulation studies confirmed efficient hydrogen-bonded self-assembly of the amide-linked bis(1,3-propanediol)-based molecules in the lipid bilayer membrane and the detection of chloride recognition in the cavity.
Collapse
Affiliation(s)
- Rashmi Sharma
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Dr Homi Bhabha Road, Pashan, Pune 411008, Maharashtra, India.
| | - Amal Vijay
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Dr Homi Bhabha Road, Pashan, Pune 411008, Maharashtra, India.
| | - Sandip Chattopadhayay
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Dr Homi Bhabha Road, Pashan, Pune 411008, Maharashtra, India.
| | - Arnab Mukherjee
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Dr Homi Bhabha Road, Pashan, Pune 411008, Maharashtra, India.
| | - Pinaki Talukdar
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Dr Homi Bhabha Road, Pashan, Pune 411008, Maharashtra, India.
| |
Collapse
|
24
|
Hill-type pH probes. Anal Bioanal Chem 2023:10.1007/s00216-023-04515-y. [PMID: 36624196 DOI: 10.1007/s00216-023-04515-y] [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: 11/08/2022] [Revised: 12/23/2022] [Accepted: 01/03/2023] [Indexed: 01/11/2023]
Abstract
Sensitive detection of the minute and yet pathologically significant pH variation is important and in fact challenging for the conventional pH probes following the Henderson-Hasselbalch equation, i.e., HH-type probes. A paradigm shift to Hill-type pH probes is ongoing. Bestowed by their positive cooperative acid-base chemistry, their pH-responsive profile follows the Hill equation, which exhibits a narrower acid/base transition width than HH-type probes and warrants a higher detection sensitivity. A polymer-based Hill-type pH-responsive material was first developed. More recently, there emerged several distinct small-molecular approaches to achieve Hill-type pH-responsive profiles. They complement the polymer-based sensing materials in applications where membrane permeability is a concern. In this trends article, we rationalize the molecular origins of their positive cooperativity in pH sensing and highlight some interesting proof-of-concept applications. We also discussed future directions of this dynamic research area.
Collapse
|
25
|
Ferrero JM, Gonzalez-Ascaso A, Matas JFR. The mechanisms of potassium loss in acute myocardial ischemia: New insights from computational simulations. Front Physiol 2023; 14:1074160. [PMID: 36923288 PMCID: PMC10009276 DOI: 10.3389/fphys.2023.1074160] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 02/13/2023] [Indexed: 03/03/2023] Open
Abstract
Acute myocardial ischemia induces hyperkalemia (accumulation of extracellular potassium), a major perpetrator of lethal reentrant ventricular arrhythmias. Despite considerable experimental efforts to explain this pathology in the last decades, the intimate mechanisms behind hyperkalemia remain partially unknown. In order to investigate these mechanisms, we developed a novel computational model of acute myocardial ischemia which couples a) an electrophysiologically detailed human cardiomyocyte model that incorporates modifications to account for ischemia-induced changes in transmembrane currents, with b) a model of cardiac tissue and extracellular K + transport. The resulting model is able to reproduce and explain the triphasic time course of extracellular K + concentration within the ischemic zone, with values of [ K + ] o close to 14 mmol/L in the central ischemic zone after 30 min. In addition, the formation of a [ K + ] o border zone of approximately 1.2 cm 15 min after the onset of ischemia is predicted by the model. Our results indicate that the primary rising phase of [ K + ] o is mainly due to the imbalance between K + efflux, that increases slightly, and K + influx, that follows a reduction of the NaK pump activity by more than 50%. The onset of the plateau phase is caused by the appearance of electrical alternans (a novel mechanism identified by the model), which cause an abrupt reduction in the K + efflux. After the plateau, the secondary rising phase of [ K + ] o is caused by a subsequent imbalance between the K + influx, which continues to decrease slowly, and the K + efflux, which remains almost constant. Further, the study shows that the modulation of these mechanisms by the electrotonic coupling is the main responsible for the formation of the ischemic border zone in tissue, with K + transport playing only a minor role. Finally, the results of the model indicate that the injury current established between the healthy and the altered tissue is not sufficient to depolarize non-ischemic cells within the healthy tissue.
Collapse
Affiliation(s)
- Jose M Ferrero
- Centro de Investigacion e Innovacion en Bioingenieria, Universitat Politecnica de Valencia, Valencia, Spain
| | - Ana Gonzalez-Ascaso
- Centro de Investigacion e Innovacion en Bioingenieria, Universitat Politecnica de Valencia, Valencia, Spain.,Dipartimento di Chimica, Materiali e Ingegneria Chimica "Giulio Natta", Politecnico di Milano, Milan, Italy
| | - Jose F Rodriguez Matas
- Dipartimento di Chimica, Materiali e Ingegneria Chimica "Giulio Natta", Politecnico di Milano, Milan, Italy
| |
Collapse
|
26
|
Milliken AS, Ciesla JH, Nadtochiy SM, Brookes PS. Distinct effects of intracellular vs. extracellular acidic pH on the cardiac metabolome during ischemia and reperfusion. J Mol Cell Cardiol 2023; 174:101-114. [PMID: 36481511 PMCID: PMC9868090 DOI: 10.1016/j.yjmcc.2022.11.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/22/2022] [Accepted: 11/25/2022] [Indexed: 12/10/2022]
Abstract
Tissue ischemia results in intracellular pH (pHIN) acidification, and while metabolism is a known driver of acidic pHIN, less is known about how acidic pHIN regulates metabolism. Furthermore, acidic extracellular (pHEX) during early reperfusion confers cardioprotection, but how this impacts metabolism is unclear. Herein we employed LCMS based targeted metabolomics to analyze perfused mouse hearts exposed to: (i) control perfusion, (ii) hypoxia, (iii) ischemia, (iv) enforced acidic pHIN, (v) control reperfusion, and (vi) acidic pHEX (6.8) reperfusion. Surprisingly little overlap was seen between metabolic changes induced by hypoxia, ischemia, and acidic pHIN. Acidic pHIN elevated metabolites in the top half of glycolysis, and enhanced glutathione redox state. Meanwhile, acidic pHEX reperfusion induced substantial metabolic changes in addition to those seen in control reperfusion. This included elevated metabolites in the top half of glycolysis, prevention of purine nucleotide loss, and an enhancement in glutathione redox state. These data led to hypotheses regarding potential roles for methylglyoxal inhibiting the mitochondrial permeability transition pore, and for acidic inhibition of ecto-5'-nucleotidase, as potential mediators of cardioprotection by acidic pHEX reperfusion. However, neither hypothesis was supported by subsequent experiments. In contrast, analysis of cardiac effluents revealed complex effects of pHEX on metabolite transport, suggesting that mildly acidic pHEX may enhance succinate release during reperfusion. Overall, each intervention had distinct and overlapping metabolic effects, suggesting acidic pH is an independent metabolic regulator regardless which side of the cell membrane it is imposed.
Collapse
Affiliation(s)
- Alexander S Milliken
- Department of Pharmacology and Physiology, University of Rochester Medical Center, USA
| | - Jessica H Ciesla
- Department of Biochemistry, University of Rochester Medical Center, USA
| | - Sergiy M Nadtochiy
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, USA
| | - Paul S Brookes
- Department of Pharmacology and Physiology, University of Rochester Medical Center, USA; Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, USA.
| |
Collapse
|
27
|
Wilson AD, Richards MA, Curtis MK, Gunadasa-Rohling M, Monterisi S, Loonat AA, Miller JJ, Ball V, Lewis A, Tyler DJ, Moshnikova A, Andreev OA, Reshetnyak YK, Carr C, Swietach P. Acidic environments trigger intracellular H+-sensing FAK proteins to re-balance sarcolemmal acid-base transporters and auto-regulate cardiomyocyte pH. Cardiovasc Res 2022; 118:2946-2959. [PMID: 34897412 PMCID: PMC9648823 DOI: 10.1093/cvr/cvab364] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 12/08/2021] [Indexed: 11/12/2022] Open
Abstract
AIMS In cardiomyocytes, acute disturbances to intracellular pH (pHi) are promptly corrected by a system of finely tuned sarcolemmal acid-base transporters. However, these fluxes become thermodynamically re-balanced in acidic environments, which inadvertently causes their set-point pHi to fall outside the physiological range. It is unclear whether an adaptive mechanism exists to correct this thermodynamic challenge, and return pHi to normal. METHODS AND RESULTS Following left ventricle cryo-damage, a diffuse pattern of low extracellular pH (pHe) was detected by acid-sensing pHLIP. Despite this, pHi measured in the beating heart (13C NMR) was normal. Myocytes had adapted to their acidic environment by reducing Cl-/HCO3- exchange (CBE)-dependent acid-loading and increasing Na+/H+ exchange (NHE1)-dependent acid-extrusion, as measured by fluorescence (cSNARF1). The outcome of this adaptation on pHi is revealed as a cytoplasmic alkalinization when cells are superfused at physiological pHe. Conversely, mice given oral bicarbonate (to improve systemic buffering) had reduced myocardial NHE1 expression, consistent with a needs-dependent expression of pHi-regulatory transporters. The response to sustained acidity could be replicated in vitro using neonatal ventricular myocytes incubated at low pHe for 48 h. The adaptive increase in NHE1 and decrease in CBE activities was linked to Slc9a1 (NHE1) up-regulation and Slc4a2 (AE2) down-regulation. This response was triggered by intracellular H+ ions because it persisted in the absence of CO2/HCO3- and became ablated when acidic incubation media had lower chloride, a solution manoeuvre that reduces the extent of pHi-decrease. Pharmacological inhibition of FAK-family non-receptor kinases, previously characterized as pH-sensors, ablated this pHi autoregulation. In support of a pHi-sensing role, FAK protein Pyk2 (auto)phosphorylation was reduced within minutes of exposure to acidity, ahead of adaptive changes to pHi control. CONCLUSIONS Cardiomyocytes fine-tune the expression of pHi-regulators so that pHi is at least 7.0. This autoregulatory feedback mechanism defines physiological pHi and protects it during pHe vulnerabilities.
Collapse
Affiliation(s)
- Abigail D Wilson
- Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, UK
| | - Mark A Richards
- Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, UK
| | - M Kate Curtis
- Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, UK
| | - Mala Gunadasa-Rohling
- Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, UK
| | - Stefania Monterisi
- Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, UK
| | - Aminah A Loonat
- Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, UK
| | - Jack J Miller
- Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, UK
- Department of Physics, Clarendon Laboratory, University of Oxford, Parks Road, Oxford OX1 3PU, UK
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Radcliffe Department of Medicine, Level 0, John Radcliffe Hospital, Headington, Oxford OX3 9DU, UK
| | - Vicky Ball
- Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, UK
| | - Andrew Lewis
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Radcliffe Department of Medicine, Level 0, John Radcliffe Hospital, Headington, Oxford OX3 9DU, UK
| | - Damian J Tyler
- Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, UK
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Radcliffe Department of Medicine, Level 0, John Radcliffe Hospital, Headington, Oxford OX3 9DU, UK
| | - Anna Moshnikova
- Physics Department, University of Rhode Island, 2 Lippitt Rd, Kingston, RI 02881, USA
| | - Oleg A Andreev
- Physics Department, University of Rhode Island, 2 Lippitt Rd, Kingston, RI 02881, USA
| | - Yana K Reshetnyak
- Physics Department, University of Rhode Island, 2 Lippitt Rd, Kingston, RI 02881, USA
| | - Carolyn Carr
- Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, UK
| | - Pawel Swietach
- Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, UK
| |
Collapse
|
28
|
Prag HA, Aksentijevic D, Dannhorn A, Giles AV, Mulvey JF, Sauchanka O, Du L, Bates G, Reinhold J, Kula-Alwar D, Xu Z, Pellerin L, Goodwin RJA, Murphy MP, Krieg T. Ischemia-Selective Cardioprotection by Malonate for Ischemia/Reperfusion Injury. Circ Res 2022; 131:528-541. [PMID: 35959683 PMCID: PMC9426742 DOI: 10.1161/circresaha.121.320717] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Inhibiting SDH (succinate dehydrogenase), with the competitive inhibitor malonate, has shown promise in ameliorating ischemia/reperfusion injury. However, key for translation to the clinic is understanding the mechanism of malonate entry into cells to enable inhibition of SDH, its mitochondrial target, as malonate itself poorly permeates cellular membranes. The possibility of malonate selectively entering the at-risk heart tissue on reperfusion, however, remains unexplored. METHODS C57BL/6J mice, C2C12 and H9c2 myoblasts, and HeLa cells were used to elucidate the mechanism of selective malonate uptake into the ischemic heart upon reperfusion. Cells were treated with malonate while varying pH or together with transport inhibitors. Mouse hearts were either perfused ex vivo (Langendorff) or subjected to in vivo left anterior descending coronary artery ligation as models of ischemia/reperfusion injury. Succinate and malonate levels were assessed by liquid chromatography-tandem mass spectrometry LC-MS/MS, in vivo by mass spectrometry imaging, and infarct size by TTC (2,3,5-triphenyl-2H-tetrazolium chloride) staining. RESULTS Malonate was robustly protective against cardiac ischemia/reperfusion injury, but only if administered at reperfusion and not when infused before ischemia. The extent of malonate uptake into the heart was proportional to the duration of ischemia. Malonate entry into cardiomyocytes in vivo and in vitro was dramatically increased at the low pH (≈6.5) associated with ischemia. This increased uptake of malonate was blocked by selective inhibition of MCT1 (monocarboxylate transporter 1). Reperfusion of the ischemic heart region with malonate led to selective SDH inhibition in the at-risk region. Acid-formulation greatly enhances the cardioprotective potency of malonate. CONCLUSIONS Cardioprotection by malonate is dependent on its entry into cardiomyocytes. This is facilitated by the local decrease in pH that occurs during ischemia, leading to its selective uptake upon reperfusion into the at-risk tissue, via MCT1. Thus, malonate's preferential uptake in reperfused tissue means it is an at-risk tissue-selective drug that protects against cardiac ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Hiran A. Prag
- Department of Medicine (H.A.P., A.V.G., J.F.M., O.S., D.K.-A., M.P.M., T.K.), University of Cambridge, United Kingdom.,MRC Mitochondrial Biology Unit (H.A.P., A.V.G., G.B., J.R., M.M.P.), University of Cambridge, United Kingdom
| | - Dunja Aksentijevic
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (D.A.)
| | - Andreas Dannhorn
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, United Kingdom (A.D., R.J.A.G.)
| | - Abigail V. Giles
- Department of Medicine (H.A.P., A.V.G., J.F.M., O.S., D.K.-A., M.P.M., T.K.), University of Cambridge, United Kingdom.,MRC Mitochondrial Biology Unit (H.A.P., A.V.G., G.B., J.R., M.M.P.), University of Cambridge, United Kingdom.,Laboratory of Cardiac Energetics, National Heart, Lung and Blood Institute, Bethesda, MD (A.V.G.)
| | - John F. Mulvey
- Department of Medicine (H.A.P., A.V.G., J.F.M., O.S., D.K.-A., M.P.M., T.K.), University of Cambridge, United Kingdom
| | - Olga Sauchanka
- Department of Medicine (H.A.P., A.V.G., J.F.M., O.S., D.K.-A., M.P.M., T.K.), University of Cambridge, United Kingdom
| | - Luping Du
- Department of Physiology and Pathophysiology, Tianjin Medical University, China (L.D., Z.X.)
| | - Georgina Bates
- MRC Mitochondrial Biology Unit (H.A.P., A.V.G., G.B., J.R., M.M.P.), University of Cambridge, United Kingdom
| | - Johannes Reinhold
- MRC Mitochondrial Biology Unit (H.A.P., A.V.G., G.B., J.R., M.M.P.), University of Cambridge, United Kingdom.,Faculty of Medicine and Health Sciences, University of East Anglia, Norwich Research Park (J.R.)
| | - Duvaraka Kula-Alwar
- Department of Medicine (H.A.P., A.V.G., J.F.M., O.S., D.K.-A., M.P.M., T.K.), University of Cambridge, United Kingdom
| | - Zhelong Xu
- Department of Physiology and Pathophysiology, Tianjin Medical University, China (L.D., Z.X.)
| | - Luc Pellerin
- Département de Physiologie, Université de Lausanne, Switzerland (L.P.).,Centre de Résonance Magnétique des Systèmes Biologiques, UMR5536 CNRS, LabEx TRAIL-IBIO, Université de Bordeaux, France (L.P.).,Inserm U1313, Université et CHU de Poitiers, France (L.P.)
| | - Richard J. A. Goodwin
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, United Kingdom (A.D., R.J.A.G.).,Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom (R.J.A.G.)
| | - Michael P. Murphy
- Department of Medicine (H.A.P., A.V.G., J.F.M., O.S., D.K.-A., M.P.M., T.K.), University of Cambridge, United Kingdom.,MRC Mitochondrial Biology Unit (H.A.P., A.V.G., G.B., J.R., M.M.P.), University of Cambridge, United Kingdom
| | - Thomas Krieg
- Department of Medicine (H.A.P., A.V.G., J.F.M., O.S., D.K.-A., M.P.M., T.K.), University of Cambridge, United Kingdom
| |
Collapse
|
29
|
Creighton JV, de Souza Gonçalves L, Artioli GG, Tan D, Elliott-Sale KJ, Turner MD, Doig CL, Sale C. Physiological Roles of Carnosine in Myocardial Function and Health. Adv Nutr 2022; 13:1914-1929. [PMID: 35689661 PMCID: PMC9526863 DOI: 10.1093/advances/nmac059] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/25/2022] [Accepted: 06/08/2022] [Indexed: 01/28/2023] Open
Abstract
Carnosine is a pleiotropic histidine-containing dipeptide synthesized from β-alanine and l-histidine, with the intact dipeptide and constituent amino acids being available from the diet. The therapeutic application of carnosine in myocardial tissue is promising, with carnosine playing a potentially beneficial role in both healthy and diseased myocardial models. This narrative review discusses the role of carnosine in myocardial function and health, including an overview of the metabolic pathway of carnosine in the myocardial tissue, the roles carnosine may play in the myocardium, and a critical analysis of the literature, focusing on the effect of exogenous carnosine and its precursors on myocardial function. By so doing, we aim to identify current gaps in the literature, thereby identifying considerations for future research.
Collapse
Affiliation(s)
- Jade V Creighton
- Musculoskeletal Physiology Research Group, Sport, Health, and Performance Enhancement (SHAPE) Research Centre, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham, United Kingdom
| | | | - Guilherme G Artioli
- Department of Life Sciences, Manchester Metropolitan University, Manchester, United Kingdom
| | - Di Tan
- Natural Alternatives International, Inc., Carlsbad, CA, USA
| | - Kirsty J Elliott-Sale
- Musculoskeletal Physiology Research Group, Sport, Health, and Performance Enhancement (SHAPE) Research Centre, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham, United Kingdom,Department of Sport and Exercise Sciences, Institute of Sport, Manchester Metropolitan University, Manchester, United Kingdom
| | - Mark D Turner
- Centre for Diabetes, Chronic Diseases, and Ageing, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham, United Kingdom
| | - Craig L Doig
- Centre for Diabetes, Chronic Diseases, and Ageing, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham, United Kingdom
| | | |
Collapse
|
30
|
Di Mattía RA, Díaz Zegarra LA, Valverde CA, Blanco PG, Jaquenod De Giusti C, Portiansky EL, Aiello EA, Orlowski A. In vivo Overexpression of Electrogenic Sodium/Bicarbonate Cotransporter (NBCe1) by AAV9 Modifies the Cardiac Action Potential and the QT Interval in Mice. Front Cardiovasc Med 2022; 9:862118. [PMID: 35548416 PMCID: PMC9082548 DOI: 10.3389/fcvm.2022.862118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/23/2022] [Indexed: 11/29/2022] Open
Abstract
Cardiac cells depend on specific sarcolemmal ion transporters to assure the correct intracellular pH regulation. The sodium/bicarbonate cotransporter (NBC) is one of the major alkalinizing mechanisms. In the heart two different NBC isoforms have been described: the electroneutral NBCn1 (1Na+:1HCO3-) and the electrogenic NBCe1 (1Na+:2HCO3-). NBCe1 generates an anionic repolarizing current that modulates the action potential duration (APD). In addition to regulating the pH, the NBC is a source of sodium influx. It has been postulated that NBC could play a role in the development of hypertrophy. The aim of this research was to study the contribution of NBCe1 in heart electrophysiology and in the development of heart hypertrophy in an in vivo mouse model with overexpression of NBCe1. Heart NBCe1 overexpression was achieved by a recombinant cardiotropic adeno-associated virus (AAV9) and was evidenced by western-blot and qPCR. AAV9-mCherry was used as a transduction control. NBCe1 overexpression fails to increase heart growth. Patch clamp and electrocardiogram were performed. We observed a reduction on both, ventricular myocytes APD and electrocardiogram QT interval corrected by cardiac rate, emphasizing for the first time NBCe1 relevance for the electrical activity of the heart.
Collapse
Affiliation(s)
- Romina A. Di Mattía
- Centro de Investigaciones Cardiovasculares “Dr. Horacio E. Cingolani, ” Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, La Plata, Argentina
| | - Leandro A. Díaz Zegarra
- Centro de Investigaciones Cardiovasculares “Dr. Horacio E. Cingolani, ” Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, La Plata, Argentina
| | - Carlos A. Valverde
- Centro de Investigaciones Cardiovasculares “Dr. Horacio E. Cingolani, ” Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, La Plata, Argentina
| | - Paula G. Blanco
- Centro de Fisiología Reproductiva y Métodos Complementarios de Diagnóstico, Facultad de Ciencias Veterinarias, Universidad Nacional de La Plata-CONICET, La Plata, Argentina
| | - Carolina Jaquenod De Giusti
- Centro de Investigaciones Cardiovasculares “Dr. Horacio E. Cingolani, ” Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, La Plata, Argentina
| | - Enrique L. Portiansky
- Laboratorio de Análisis de Imágenes, Facultad de Ciencias Veterinarias, Universidad Nacional de La Plata-CONICET, La Plata, Argentina
| | - Ernesto A. Aiello
- Centro de Investigaciones Cardiovasculares “Dr. Horacio E. Cingolani, ” Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, La Plata, Argentina
- *Correspondence: Ernesto A. Aiello
| | - Alejandro Orlowski
- Centro de Investigaciones Cardiovasculares “Dr. Horacio E. Cingolani, ” Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, La Plata, Argentina
- Alejandro Orlowski
| |
Collapse
|
31
|
Hulikova A, Park KC, Loonat AA, Gunadasa-Rohling M, Curtis MK, Chung YJ, Wilson A, Carr CA, Trafford AW, Fournier M, Moshnikova A, Andreev OA, Reshetnyak YK, Riley PR, Smart N, Milne TA, Crump NT, Swietach P. Alkaline nucleoplasm facilitates contractile gene expression in the mammalian heart. Basic Res Cardiol 2022; 117:17. [PMID: 35357563 PMCID: PMC8971196 DOI: 10.1007/s00395-022-00924-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 03/04/2022] [Accepted: 03/11/2022] [Indexed: 01/31/2023]
Abstract
Cardiac contractile strength is recognised as being highly pH-sensitive, but less is known about the influence of pH on cardiac gene expression, which may become relevant in response to changes in myocardial metabolism or vascularization during development or disease. We sought evidence for pH-responsive cardiac genes, and a physiological context for this form of transcriptional regulation. pHLIP, a peptide-based reporter of acidity, revealed a non-uniform pH landscape in early-postnatal myocardium, dissipating in later life. pH-responsive differentially expressed genes (pH-DEGs) were identified by transcriptomics of neonatal cardiomyocytes cultured over a range of pH. Enrichment analysis indicated "striated muscle contraction" as a pH-responsive biological process. Label-free proteomics verified fifty-four pH-responsive gene-products, including contractile elements and the adaptor protein CRIP2. Using transcriptional assays, acidity was found to reduce p300/CBP acetylase activity and, its a functional readout, inhibit myocardin, a co-activator of cardiac gene expression. In cultured myocytes, acid-inhibition of p300/CBP reduced H3K27 acetylation, as demonstrated by chromatin immunoprecipitation. H3K27ac levels were more strongly reduced at promoters of acid-downregulated DEGs, implicating an epigenetic mechanism of pH-sensitive gene expression. By tandem cytoplasmic/nuclear pH imaging, the cardiac nucleus was found to exercise a degree of control over its pH through Na+/H+ exchangers at the nuclear envelope. Thus, we describe how extracellular pH signals gain access to the nucleus and regulate the expression of a subset of cardiac genes, notably those coding for contractile proteins and CRIP2. Acting as a proxy of a well-perfused myocardium, alkaline conditions are permissive for expressing genes related to the contractile apparatus.
Collapse
Affiliation(s)
- Alzbeta Hulikova
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - Kyung Chan Park
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - Aminah A Loonat
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - Mala Gunadasa-Rohling
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - M Kate Curtis
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - Yu Jin Chung
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - Abigail Wilson
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - Carolyn A Carr
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - Andrew W Trafford
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - Marjorie Fournier
- Department of Biochemistry, Advanced Proteomics Facility, University of Oxford, Oxford, UK
| | - Anna Moshnikova
- Physics Department, University of Rhode Island, 2 Lippitt Rd, Kingston, RI, 02881, USA
| | - Oleg A Andreev
- Physics Department, University of Rhode Island, 2 Lippitt Rd, Kingston, RI, 02881, USA
| | - Yana K Reshetnyak
- Physics Department, University of Rhode Island, 2 Lippitt Rd, Kingston, RI, 02881, USA
| | - Paul R Riley
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - Nicola Smart
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - Thomas A Milne
- MRC Molecular Haematology Unit, Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, NIHR Oxford Biomedical Research Centre Haematology Theme, University of Oxford, Oxford, UK
| | - Nicholas T Crump
- MRC Molecular Haematology Unit, Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, NIHR Oxford Biomedical Research Centre Haematology Theme, University of Oxford, Oxford, UK
| | - Pawel Swietach
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK.
| |
Collapse
|
32
|
Wu X, Li Y, Maienschein-Cline M, Feferman L, Wu L, Hong L. RNA-Seq Analyses Reveal Roles of the HVCN1 Proton Channel in Cardiac pH Homeostasis. Front Cell Dev Biol 2022; 10:860502. [PMID: 35372367 PMCID: PMC8967321 DOI: 10.3389/fcell.2022.860502] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 02/16/2022] [Indexed: 12/13/2022] Open
Abstract
The voltage-gated proton channel HVCN1 is a member of the voltage-gated ion channel family. HVCN1 channel controls acid extrusion and regulates pH homeostasis in various cell types. Recent evidence indicated that the HVCN1 channel was associated with cardiac function. To investigate the role of HVCN1 in cardiac myocytes, we performed an RNA sequencing analysis of murine hearts and showed that HVCN1 null hearts exhibited a differential transcriptome profile compared with wild-type hearts. The RNA-seq data indicating impaired pH homeostasis in HVCN1 null hearts were the downregulated NADPH oxidoreductases (NOXs) and decreased expression of Cl−/HCO3− exchanger, indicating HVCN1 is a regulator of gene transcriptional networks controlling NOX signaling and CO2 homeostasis in the heart. Additionally, HVCN1 null hearts exhibited differential expression of cardiac ion channels, suggesting a potential role of HVCN1 in cardiac electrophysiological remodeling. The study highlights the importance of HVCN1 in cardiac function and may present a novel target associated with heart diseases.
Collapse
Affiliation(s)
- Xin Wu
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Yawei Li
- Department of Preventive Medicine, Northwestern University, Chicago, IL, United States
| | - Mark Maienschein-Cline
- Research Informatics Core, Research Resources Center, University of Illinois at Chicago, Chicago, IL, United States
| | - Leonid Feferman
- Research Informatics Core, Research Resources Center, University of Illinois at Chicago, Chicago, IL, United States
| | - Longjun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
| | - Liang Hong
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
- *Correspondence: Liang Hong,
| |
Collapse
|
33
|
Lyu Y, Thai PN, Ren L, Timofeyev V, Jian Z, Park S, Ginsburg KS, Overton J, Bossuyt J, Bers DM, Yamoah EN, Chen-Izu Y, Chiamvimonvat N, Zhang XD. Beat-to-beat dynamic regulation of intracellular pH in cardiomyocytes. iScience 2022; 25:103624. [PMID: 35005560 PMCID: PMC8718820 DOI: 10.1016/j.isci.2021.103624] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/10/2021] [Accepted: 12/10/2021] [Indexed: 11/20/2022] Open
Abstract
The mammalian heart beats incessantly with rhythmic mechanical activities generating acids that need to be buffered to maintain a stable intracellular pH (pHi) for normal cardiac function. Even though spatial pHi non-uniformity in cardiomyocytes has been documented, it remains unknown how pHi is regulated to match the dynamic cardiac contractions. Here, we demonstrated beat-to-beat intracellular acidification, termed pHi transients, in synchrony with cardiomyocyte contractions. The pHi transients are regulated by pacing rate, Cl-/HCO3 - transporters, pHi buffering capacity, and β-adrenergic signaling. Mitochondrial electron-transport chain inhibition attenuates the pHi transients, implicating mitochondrial activity in sculpting the pHi regulation. The pHi transients provide dynamic alterations of H+ transport required for ATP synthesis, and a decrease in pHi may serve as a negative feedback to cardiac contractions. Current findings dovetail with the prevailing three known dynamic systems, namely electrical, Ca2+, and mechanical systems, and may reveal broader features of pHi handling in excitable cells.
Collapse
Affiliation(s)
- Yankun Lyu
- Department of Internal Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Phung N. Thai
- Department of Internal Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Lu Ren
- Department of Internal Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Valeriy Timofeyev
- Department of Internal Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Zhong Jian
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
| | - Seojin Park
- Department of Physiology and Cell Biology, University of Nevada, Reno, Reno, NV 89557, USA
| | - Kenneth S. Ginsburg
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
| | - James Overton
- Department of Internal Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Julie Bossuyt
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
| | - Donald M. Bers
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
| | - Ebenezer N. Yamoah
- Department of Physiology and Cell Biology, University of Nevada, Reno, Reno, NV 89557, USA
| | - Ye Chen-Izu
- Department of Internal Medicine, University of California, Davis, Davis, CA 95616, USA
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616, USA
| | - Nipavan Chiamvimonvat
- Department of Internal Medicine, University of California, Davis, Davis, CA 95616, USA
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
- Department of Veterans Affairs, Northern California Health Care System, Mather, CA 95655, USA
| | - Xiao-Dong Zhang
- Department of Internal Medicine, University of California, Davis, Davis, CA 95616, USA
- Department of Veterans Affairs, Northern California Health Care System, Mather, CA 95655, USA
| |
Collapse
|
34
|
Sharma R, Vijay A, Mukherjee A, Talukdar P. Bis(cholyl)-based chloride channels with oxalamide and hydrazide selectivity filters. Org Biomol Chem 2022; 20:2054-2058. [DOI: 10.1039/d1ob02028e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We report the development of supramolecular bis(cholyl) ion channels by using oxalamide and hydrazide as selectivity filters. The hydrazide system displayed superior chloride transport activity than oxalamide via the formation...
Collapse
|
35
|
Ottolia M, John S, Hazan A, Goldhaber JI. The Cardiac Na + -Ca 2+ Exchanger: From Structure to Function. Compr Physiol 2021; 12:2681-2717. [PMID: 34964124 DOI: 10.1002/cphy.c200031] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Ca2+ homeostasis is essential for cell function and survival. As such, the cytosolic Ca2+ concentration is tightly controlled by a wide number of specialized Ca2+ handling proteins. One among them is the Na+ -Ca2+ exchanger (NCX), a ubiquitous plasma membrane transporter that exploits the electrochemical gradient of Na+ to drive Ca2+ out of the cell, against its concentration gradient. In this critical role, this secondary transporter guides vital physiological processes such as Ca2+ homeostasis, muscle contraction, bone formation, and memory to name a few. Herein, we review the progress made in recent years about the structure of the mammalian NCX and how it relates to function. Particular emphasis will be given to the mammalian cardiac isoform, NCX1.1, due to the extensive studies conducted on this protein. Given the degree of conservation among the eukaryotic exchangers, the information highlighted herein will provide a foundation for our understanding of this transporter family. We will discuss gene structure, alternative splicing, topology, regulatory mechanisms, and NCX's functional role on cardiac physiology. Throughout this article, we will attempt to highlight important milestones in the field and controversial topics where future studies are required. © 2021 American Physiological Society. Compr Physiol 12:1-37, 2021.
Collapse
Affiliation(s)
- Michela Ottolia
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Scott John
- Department of Medicine (Cardiology), UCLA, Los Angeles, California, USA
| | - Adina Hazan
- Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, California, USA
| | - Joshua I Goldhaber
- Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
36
|
Chung YJ, Park KC, Tokar S, Eykyn TR, Fuller W, Pavlovic D, Swietach P, Shattock MJ. Off-target effects of sodium-glucose co-transporter 2 blockers: empagliflozin does not inhibit Na+/H+ exchanger-1 or lower [Na+]i in the heart. Cardiovasc Res 2021; 117:2794-2806. [PMID: 33135077 PMCID: PMC8683707 DOI: 10.1093/cvr/cvaa323] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/28/2020] [Accepted: 11/02/2020] [Indexed: 12/17/2022] Open
Abstract
AIMS Emipagliflozin (EMPA) is a potent inhibitor of the renal sodium-glucose co-transporter 2 (SGLT2) and an effective treatment for type-2 diabetes. In patients with diabetes and heart failure, EMPA has cardioprotective effects independent of improved glycaemic control, despite SGLT2 not being expressed in the heart. A number of non-canonical mechanisms have been proposed to explain these cardiac effects, most notably an inhibitory action on cardiac Na+/H+ exchanger 1 (NHE1), causing a reduction in intracellular [Na+] ([Na+]i). However, at resting intracellular pH (pHi), NHE1 activity is very low and its pharmacological inhibition is not expected to meaningfully alter steady-state [Na+]i. We re-evaluate this putative EMPA target by measuring cardiac NHE1 activity. METHODS AND RESULTS The effect of EMPA on NHE1 activity was tested in isolated rat ventricular cardiomyocytes from measurements of pHi recovery following an ammonium pre-pulse manoeuvre, using cSNARF1 fluorescence imaging. Whereas 10 µM cariporide produced near-complete inhibition, there was no evidence for NHE1 inhibition with EMPA treatment (1, 3, 10, or 30 µM). Intracellular acidification by acetate-superfusion evoked NHE1 activity and raised [Na+]i, reported by sodium binding benzofuran isophthalate (SBFI) fluorescence, but EMPA did not ablate this rise. EMPA (10 µM) also had no significant effect on the rate of cytoplasmic [Na+]i rise upon superfusion of Na+-depleted cells with Na+-containing buffers. In Langendorff-perfused mouse, rat and guinea pig hearts, EMPA did not affect [Na+]i at baseline nor pHi recovery following acute acidosis, as measured by 23Na triple quantum filtered NMR and 31P NMR, respectively. CONCLUSIONS Our findings indicate that cardiac NHE1 activity is not inhibited by EMPA (or other SGLT2i's) and EMPA has no effect on [Na+]i over a wide range of concentrations, including the therapeutic dose. Thus, the beneficial effects of SGLT2i's in failing hearts should not be interpreted in terms of actions on myocardial NHE1 or intracellular [Na+].
Collapse
Affiliation(s)
- Yu Jin Chung
- British Heart Foundation Centre of Research Excellence, King’s College London, The Rayne Institute, St Thomas’ Hospital, Lambeth Palace Road, London SE1 7EH, UK
| | - Kyung Chan Park
- Department of Physiology, Anatomy and Genetics, Parks Road, Oxford, OX1 3PT, UK
| | - Sergiy Tokar
- British Heart Foundation Centre of Research Excellence, King’s College London, The Rayne Institute, St Thomas’ Hospital, Lambeth Palace Road, London SE1 7EH, UK
| | - Thomas R Eykyn
- British Heart Foundation Centre of Research Excellence, King’s College London, The Rayne Institute, St Thomas’ Hospital, Lambeth Palace Road, London SE1 7EH, UK
- School of Biomedical Engineering and Imaging Sciences, King’s College London, The Rayne Institute, St Thomas' Hospital, Lambeth Palace Road, London SE1 7EH, UK
| | - William Fuller
- Institute of Cardiovascular & Medical Sciences, Sir James Black Building, University of Glasgow, University Avenue, Glasgow G12 8QQ, UK
| | - Davor Pavlovic
- Institute for Cardiovascular Sciences, University of Birmingham, Wolfson Drive, Birmingham B15 2TT, UK
| | - Pawel Swietach
- Department of Physiology, Anatomy and Genetics, Parks Road, Oxford, OX1 3PT, UK
| | - Michael J Shattock
- British Heart Foundation Centre of Research Excellence, King’s College London, The Rayne Institute, St Thomas’ Hospital, Lambeth Palace Road, London SE1 7EH, UK
| |
Collapse
|
37
|
Yang J, Yu G, Sessler JL, Shin I, Gale PA, Huang F. Artificial transmembrane ion transporters as potential therapeutics. Chem 2021. [DOI: 10.1016/j.chempr.2021.10.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
38
|
Ciocci Pardo A, González Arbeláez LF, Fantinelli JC, Álvarez BV, Mosca SM, Swenson ER. Myocardial and mitochondrial effects of the anhydrase carbonic inhibitor ethoxzolamide in ischemia-reperfusion. Physiol Rep 2021; 9:e15093. [PMID: 34806317 PMCID: PMC8606860 DOI: 10.14814/phy2.15093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/21/2021] [Accepted: 09/21/2021] [Indexed: 11/24/2022] Open
Abstract
We have previously demonstrated that inhibition of extracellularly oriented carbonic anhydrase (CA) isoforms protects the myocardium against ischemia-reperfusion injury. In this study, our aim was to assess the possible further contribution of CA intracellular isoforms examining the actions of the highly diffusible cell membrane permeant inhibitor of CA, ethoxzolamide (ETZ). Isolated rat hearts, after 20 min of stabilization, were assigned to the following groups: (1) Nonischemic control: 90 min of perfusion; (2) Ischemic control: 30 min of global ischemia and 60 min of reperfusion (R); and (3) ETZ: ETZ at a concentration of 100 μM was administered for 10 min before the onset of ischemia and then during the first 10 min of reperfusion. In additional groups, ETZ was administered in the presence of SB202190 (SB, a p38MAPK inhibitor) or chelerythrine (Chel, a protein kinase C [PKC] inhibitor). Infarct size, myocardial function, and the expression of phosphorylated forms of p38MAPK, PKCε, HSP27, and Drp1, and calcineurin Aβ content were assessed. In isolated mitochondria, the Ca2+ response, Ca2+ retention capacity, and membrane potential were measured. ETZ decreased infarct size by 60%, improved postischemic recovery of myocardial contractile and diastolic relaxation increased P-p38MAPK, P-PKCε, P-HSP27, and P-Drp1 expression, decreased calcineurin content, and normalized calcium and membrane potential parameters measured in isolated mitochondria. These effects were significantly attenuated when ETZ was administered in the presence of SB or Chel. These data show that ETZ protects the myocardium and mitochondria against ischemia-reperfusion injury through p38MAPK- and PKCε-dependent pathways and reinforces the role of CA as a possible target in the management of acute cardiac ischemic diseases.
Collapse
Affiliation(s)
- Alejandro Ciocci Pardo
- Centro de Investigaciones Cardiovasculares ¨Dr Horacio E Cingolani¨CCT‐CONICETFacultad de Ciencias MédicasUniversidad Nacional de La PlataLa PlataBuenos AiresArgentina
| | - Luisa F. González Arbeláez
- Centro de Investigaciones Cardiovasculares ¨Dr Horacio E Cingolani¨CCT‐CONICETFacultad de Ciencias MédicasUniversidad Nacional de La PlataLa PlataBuenos AiresArgentina
| | - Juliana C. Fantinelli
- Centro de Investigaciones Cardiovasculares ¨Dr Horacio E Cingolani¨CCT‐CONICETFacultad de Ciencias MédicasUniversidad Nacional de La PlataLa PlataBuenos AiresArgentina
| | - Bernardo V. Álvarez
- Centro de Investigaciones Cardiovasculares ¨Dr Horacio E Cingolani¨CCT‐CONICETFacultad de Ciencias MédicasUniversidad Nacional de La PlataLa PlataBuenos AiresArgentina
- Present address:
Department of BiochemistryMembrane Protein Disease Research GroupUniversity of AlbertaEdmontonAlbertaT6G 2H7Canada
| | - Susana M. Mosca
- Centro de Investigaciones Cardiovasculares ¨Dr Horacio E Cingolani¨CCT‐CONICETFacultad de Ciencias MédicasUniversidad Nacional de La PlataLa PlataBuenos AiresArgentina
| | - Erik R. Swenson
- Department of Medicine, Pulmonary and Critical Care MedicineVA Puget Sound Health Care SystemUniversity of WashingtonSeattleWashingtonUSA
| |
Collapse
|
39
|
Du L, Zahra A, Jia M, Wang Q, Wu J. Understanding the Functional Expression of Na+-Coupled SLC4 Transporters in the Renal and Nervous Systems: A Review. Brain Sci 2021; 11:1276. [PMID: 34679341 PMCID: PMC8534249 DOI: 10.3390/brainsci11101276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/16/2021] [Accepted: 09/23/2021] [Indexed: 11/25/2022] Open
Abstract
Acid-base homeostasis is crucial for numerous physiological processes. Na+/HCO3- cotransporters (NBCs) belong to the solute carrier 4 (SLC4) family, which regulates intracellular pH as well as HCO3- absorption and secretion. However, knowledge of the structural functions of these proteins remains limited. Electrogenic NBC (NBCe-1) is thought to be the primary factor promoting the precise acid-base equilibrium in distinct cell types for filtration and reabsorption, as well as the function of neurons and glia. NBC dysregulation is strongly linked to several diseases. As such, the need for special drugs that interfere with the transmission function of NBC is becoming increasingly urgent. In this review, we focus on the structural and functional characteristics of NBCe1, and discuss the roles of NBCe1 in the kidney, central nervous system (CNS), and related disorders, we also summarize the research on NBC inhibitors. NBCe1 and the related pathways should be further investigated, so that new medications may be developed to address the related conditions.
Collapse
Affiliation(s)
- Le Du
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China; (L.D.); (A.Z.)
| | - Aqeela Zahra
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China; (L.D.); (A.Z.)
| | - Meng Jia
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (M.J.); (Q.W.)
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100070, China
- National Clinical Research Center for Neurological Disease, Beijing 100070, China
| | - Qun Wang
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (M.J.); (Q.W.)
- National Clinical Research Center for Neurological Disease, Beijing 100070, China
| | - Jianping Wu
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China; (L.D.); (A.Z.)
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (M.J.); (Q.W.)
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100070, China
- National Clinical Research Center for Neurological Disease, Beijing 100070, China
- Health Science Center, Yangtze University, Jingzhou 434023, China
| |
Collapse
|
40
|
Nolly MB, Vargas LA, Correa MV, Lofeudo JM, Pinilla AO, Rueda JOV, Guerrero-Gimenez ME, Swenson ER, Damiani MT, Alvarez BV. Carbonic anhydrase IX and hypoxia-inducible factor 1 attenuate cardiac dysfunction after myocardial infarction. Pflugers Arch 2021; 473:1273-1285. [PMID: 34231059 DOI: 10.1007/s00424-021-02592-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 01/11/2023]
Abstract
Myocardial infarction (MI) is one of the leading causes of death worldwide. Prognosis and mortality rate are directly related to infarct size and post-infarction pathological heart remodeling, which can lead to heart failure. Hypoxic MI-affected areas increase the expression of hypoxia-inducible factor (HIF-1), inducing infarct size reduction and improving cardiac function. Hypoxia translocates HIF-1 to the nucleus, activating carbonic anhydrase IX (CAIX) transcription. CAIX regulates myocardial intracellular pH, critical for heart performance. Our objective was to investigate CAIX participation and relation with sodium bicarbonate transporters 1 (NBC1) and HIF-1 in cardiac remodeling after MI. We analyzed this pathway in an "in vivo" rat coronary artery ligation model and isolated cardiomyocytes maintained under hypoxia. Immunohistochemical studies revealed an increase in HIF-1 levels after 2 h of infarction. Similar results were observed in 2-h infarcted cardiac tissue (immunoblotting) and in hypoxic cardiomyocytes with a nuclear distribution (confocal microscopy). Immunohistochemical studies showed an increase CAIX in the infarcted area at 2 h, mainly distributed throughout the cell and localized in the plasma membrane at 24 h. Similar results were observed in 2 h in infarcted cardiac tissue (immunoblotting) and in hypoxic cardiomyocytes (confocal microscopy). NBC1 expression increased in cardiac tissue after 2 h of infarction (immunoblotting). CAIX and NBC1 interaction increases in cardiac tissue subjected to MI for 2h when CAIX is present (immunoprecipitation). These results suggest that CAIX interacts with NBC1 in our infarct model as a mechanism to prevent acidic damage in hypoxic tissue, making it a promising therapeutic target.
Collapse
Affiliation(s)
- Mariela Beatriz Nolly
- Laboratorio de Bioquímica e Inmunidad, IMBECU-CONICET-UNCuyo, Instituto de Bioquímica y Biotecnología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, 5500, Mendoza, Argentina.
| | - Lorena Alejandra Vargas
- Centro de Investigaciones Cardiovasculares, CIC-CONICET, Facultad de Medicina, Universidad Nacional de La Plata, La Plata, 1900, Buenos Aires, Argentina
| | - María Verónica Correa
- Comisión de Investigaciones Científicas de la Provincia de Buenos Aires, CIC-PBA, La Plata, 1900, Buenos Aires, Argentina
| | - Juan Manuel Lofeudo
- Centro de Investigaciones Cardiovasculares, CIC-CONICET, Facultad de Medicina, Universidad Nacional de La Plata, La Plata, 1900, Buenos Aires, Argentina
| | - Andrés Oscar Pinilla
- Centro de Investigaciones Cardiovasculares, CIC-CONICET, Facultad de Medicina, Universidad Nacional de La Plata, La Plata, 1900, Buenos Aires, Argentina
| | - Jorge Omar Velez Rueda
- Centro de Investigaciones Cardiovasculares, CIC-CONICET, Facultad de Medicina, Universidad Nacional de La Plata, La Plata, 1900, Buenos Aires, Argentina
| | - Martin E Guerrero-Gimenez
- Laboratorio de Oncología, IMBECU-CONICET-UNCuyo, Instituto de Bioquímica y Biotecnología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, 5500, Mendoza, Argentina
| | - Erik Richard Swenson
- Medical Service, VA Puget Sound Health Care System, University of Washington, Seattle, WA, USA
| | - Maria Teresa Damiani
- Laboratorio de Bioquímica e Inmunidad, IMBECU-CONICET-UNCuyo, Instituto de Bioquímica y Biotecnología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, 5500, Mendoza, Argentina
| | - Bernardo Victor Alvarez
- Centro de Investigaciones Cardiovasculares, CIC-CONICET, Facultad de Medicina, Universidad Nacional de La Plata, La Plata, 1900, Buenos Aires, Argentina
- Department of Biochemistry, Membrane Protein Disease Research Group, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
| |
Collapse
|
41
|
Abstract
The hydrogen ion concentration ([H+]) in intracellular cytoplasmic fluid (ICF) must be maintained in a narrow range in all species for normal protein functions. Thus, mechanisms regulating ICF are of fundamental biological importance. Studies on the regulation of ICF [H+] have been hampered by use of pH notation, failure to consider the roles played by differences in the concentration of strong ions (strong ion difference, SID), the conservation of mass, the principle of electrical neutrality, and that [H+] and bicarbonate ions [HCO3-] are dependent variables. This argument is based on the late Peter Stewart's physical-chemical analysis of [H+] regulation reported in this journal nearly forty years ago (Stewart. 1983. Can. J. Physiol. Pharmacol. 61: 1444-1461. Doi:10.1139/y83-207). We start by outlining the principles of Stewart's analysis and then provide a general understanding of its significance for regulation of ICF [H+]. The system may initially appear complex, but it becomes evident that changes in SID dominate regulation of [H+]. The primary strong ions are Na+, K+, and Cl-, and a few organic strong anions. The second independent variable, partial pressure of carbon dioxide (PCO2), can easily be assessed. The third independent variable, the activity of intracellular weak acids ([Atot]), is much more complex but largely plays a modifying role. Attention to these principles will potentially provide new insights into ICF pH regulation.
Collapse
Affiliation(s)
- Sheldon Magder
- Department of Critical Care, McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC H4A 3J1, Canada
- Department of Critical Care, McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC H4A 3J1, Canada
| | - Alexandr Magder
- Department of Critical Care, McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC H4A 3J1, Canada
- Department of Critical Care, McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC H4A 3J1, Canada
| | - Gordan Samoukovic
- Department of Critical Care, McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC H4A 3J1, Canada
- Department of Critical Care, McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC H4A 3J1, Canada
| |
Collapse
|
42
|
Murphy E, Eisner DA. Does the cardioprotective effect of Empagliflozin involve inhibition of the sodium-proton exchanger? Cardiovasc Res 2021; 117:2696-2698. [PMID: 34021316 DOI: 10.1093/cvr/cvab137] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Affiliation(s)
- Elizabeth Murphy
- National Heart, Lung and Blood Institute, NIH, Bethesda, MD, USA
| | - David A Eisner
- Unit of Cardiac Physiology Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| |
Collapse
|
43
|
Fan C, Zhu B, Zhang X, Bi C, Zhang D, Zong Z, Fan Y. Highly Stable Acid-Induced Emission-Enhancing Cd-MOFs: Synthesis, Characterization, and Detection of Glutamic Acid in Water and Fe Ions in Acid. Inorg Chem 2021; 60:6339-6348. [PMID: 33866780 DOI: 10.1021/acs.inorgchem.1c00017] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Two novel 3D fluorescent metal-organic frameworks (MOFs), [Cd(L)(bbibp)]n (1) and [Cd(L)(bbibp)0.5]n (2), where H2L = 4,4'-(4,4'-bipyridine-2,6-diyl)dibenzoic acid and bbibp = 4,4'-bis(benzoimidaz-1-yl)biphenyl, were acquired through a conventional method and characterized via IR spectra, single-crystal X-ray diffraction, elemental analysis, thermogravimetric analysis, powder X-ray diffraction (PXRD), scanning electron microscopy, N2 adsorption-desorption isotherms, and X-ray photoelectron spectroscopy (XPS). The crystal framework of Cd-MOF 1 remained stable in the range of pH = 1.0-12.0. Interestingly, the emission peak of 1 showed a red shift and exhibited a fluorescence turn-on effect in an acidic environment. X-ray diffraction measurement revealed that the crystal structure of 1 remained unchanged after immersion in a pH = 1.0 solution. In addition, Cd-MOFs 1 and 2 displayed fluorescent quenching to l-glutamic acid with high sensitivity and selectivity. Meanwhile, 1 showed high selectivity in recognizing Fe3+ under acidic conditions, which made 1 capable of detecting Fe3+ in acidic industrial wastewater. Finally, the fluorescent sensing mechanism was carefully studied by PXRD, transient fluorescence lifetime, XPS, and UV spectroscopy.
Collapse
Affiliation(s)
- Chuanbin Fan
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao, Shandong 266100, P. R. China
| | - Bin Zhu
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao, Shandong 266100, P. R. China
| | - Xia Zhang
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao, Shandong 266100, P. R. China
| | - Caifeng Bi
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao, Shandong 266100, P. R. China
| | - Dongmei Zhang
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao, Shandong 266100, P. R. China
| | - Ziao Zong
- School of Laboratory Medicine, Youjiang Medical University for Nationalities, Baise, Guangxi 533000, China
| | - Yuhua Fan
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao, Shandong 266100, P. R. China
| |
Collapse
|
44
|
Fernandez-Caggiano M, Eaton P. Heart failure-emerging roles for the mitochondrial pyruvate carrier. Cell Death Differ 2021; 28:1149-1158. [PMID: 33473180 PMCID: PMC8027425 DOI: 10.1038/s41418-020-00729-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/14/2020] [Accepted: 12/27/2020] [Indexed: 01/30/2023] Open
Abstract
The mitochondrial pyruvate carrier (MPC) is the entry point for the glycolytic end-product pyruvate to the mitochondria. MPC activity, which is controlled by its abundance and post-translational regulation, determines whether pyruvate is oxidised in the mitochondria or metabolised in the cytosol. MPC serves as a crucial metabolic branch point that determines the fate of pyruvate in the cell, enabling metabolic adaptations during health, such as exercise, or as a result of disease. Decreased MPC expression in several cancers limits the mitochondrial oxidation of pyruvate and contributes to lactate accumulation in the cytosol, highlighting its role as a contributing, causal mediator of the Warburg effect. Pyruvate is handled similarly in the failing heart where a large proportion of it is reduced to lactate in the cytosol instead of being fully oxidised in the mitochondria. Several recent studies have found that the MPC abundance was also reduced in failing human and mouse hearts that were characterised by maladaptive hypertrophic growth, emulating the anabolic scenario observed in some cancer cells. In this review we discuss the evidence implicating the MPC as an important, perhaps causal, mediator of heart failure progression.
Collapse
Affiliation(s)
- Mariana Fernandez-Caggiano
- grid.4868.20000 0001 2171 1133The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ UK
| | - Philip Eaton
- grid.4868.20000 0001 2171 1133The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ UK
| |
Collapse
|
45
|
Priya B, Mahajan V, Kumar N. Xanthene-based Fluorescence Turn-on Probe for Highly Acidic pH Range in Aqueous Solution. J Fluoresc 2021; 31:853-860. [PMID: 33768472 DOI: 10.1007/s10895-021-02723-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/17/2021] [Indexed: 11/26/2022]
Abstract
A xanthene-based probe, Xanth-NPr, is developed as a molecular system that exhibits sensitivity for the highly acidic environments with fluorescence turn-on behavior. Xanth-NPr is designed on the principle of photoinduced electron transfer (PET), which controls the fluorescence profile of the probe. The structure of Xanth-NPr contains the dipropylaniline group as a PET promoting unit. Xanth-NPr exhibited quenched fluorescence as long as it is present in neutral or moderately acidic conditions. However, in the highly acidic pH range, it displayed a strong red-colored fluorescence at 592 nm as the protonation of dipropylaniline moiety inhibits the PET process. A model probe Xanth-M without any PET promoting unit was also synthesized. The model probe along with theoretical calculations was employed to explain the role of the PET process in regulating the fluorescence behavior of Xanth-NPr. Xanth-NPr showed linear fluorescence response as a function of pH in the range of 1 to 4.1 with the pKa value of 2.72. Likewise, its fluorescence profile is not altered by the presence of biologically relevant cations.
Collapse
Affiliation(s)
- Bhanu Priya
- Department of Chemical Sciences, IKG-Punjab Technical University, Kapurthala, 144603, Punjab, India
| | - Vibha Mahajan
- Department of Chemical Sciences, IKG-Punjab Technical University, Kapurthala, 144603, Punjab, India
| | - Naresh Kumar
- Department of Chemistry, SRM University, Delhi-NCR, Sonepat, 131029, Haryana, India.
| |
Collapse
|
46
|
Kreitmeier KG, Tarnowski D, Nanadikar MS, Baier MJ, Wagner S, Katschinski DM, Maier LS, Sag CM. CaMKII δ Met281/282 oxidation is not required for recovery of calcium transients during acidosis. Am J Physiol Heart Circ Physiol 2021; 320:H1199-H1212. [PMID: 33449853 DOI: 10.1152/ajpheart.00040.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 12/15/2020] [Accepted: 01/11/2021] [Indexed: 12/31/2022]
Abstract
CaMKII is needed for the recovery of Ca2+ transients during acidosis but also mediates postacidic arrhythmias. CaMKIIδ can sustain its activity following Met281/282 oxidation. Increasing cytosolic Na+ during acidosis as well as postacidic pH normalization should result in prooxidant conditions within the cell favoring oxidative CaMKIIδ activation. We tested whether CaMKIIδ activation through Met281/282 oxidation is involved in recovery of Ca2+ transients during acidosis and promotes cellular arrhythmias post-acidosis. Single cardiac myocytes were isolated from a well-established mouse model in which CaMKIIδ was made resistant to oxidative activation by knock-in replacement of two oxidant-sensitive methionines (Met281/282) with valines (MM-VV). MM-VV myocytes were exposed to extracellular acidosis (pHo 6.5) and compared to wild type (WT) control cells. Full recovery of Ca2+ transients was observed in both WT and MM-VV cardiac myocytes during late-phase acidosis. This was associated with comparably enhanced sarcoplasmic reticulum Ca2+ load and preserved CaMKII specific phosphorylation of phospholamban at Thr17 in MM-VV myocytes. CaMKII was phosphorylated at Thr287, but not Met281/282 oxidized. In line with this, postacidic cellular arrhythmias occurred to a similar extent in WT and MM-VV cells, whereas inhibition of CaMKII using AIP completely prevented recovery of Ca2+ transients during acidosis and attenuated postacidic arrhythmias in MM-VV cells. Using genetically altered cardiomyocytes with cytosolic expression of redox-sensitive green fluorescent protein-2 coupled to glutaredoxin 1, we found that acidosis has a reductive effect within the cytosol of cardiac myocytes despite a significant acidosis-related increase in cytosolic Na+. Our study shows that activation of CaMKIIδ through Met281/282 oxidation is neither required for recovery of Ca2+ transients during acidosis nor relevant for postacidic arrhythmogenesis in isolated cardiac myocytes. Acidosis reduces the cytosolic glutathione redox state of isolated cardiac myocytes despite a significant increase in cytosolic Na+. Pharmacological inhibition of global CaMKII activity completely prevents recovery of Ca2+ transients and protects from postacidic arrhythmias in MM-VV myocytes, which confirms the relevance of CaMKII in the context of acidosis.NEW & NOTEWORTHY The current study shows that activation of CaMKIIδ through Met281/282 oxidation is neither required for CaMKII-dependent recovery of Ca2+ transients during acidosis nor relevant for the occurrence of postacidic cellular arrhythmias. Despite a usually prooxidant increase in cytosolic Na+, acidosis reduces the cytosolic glutathione redox state within cardiac myocytes. This novel finding suggests that oxidation of cytosolic proteins is less likely to occur during acidosis.
Collapse
Affiliation(s)
- K G Kreitmeier
- Department of Internal Medicine II, University Medical Center Regensburg, Germany
- Department of Internal Medicine III, University Medical Center Regensburg, Germany
| | - D Tarnowski
- Department of Internal Medicine II, University Medical Center Regensburg, Germany
| | - M S Nanadikar
- Institute for Cardiovascular Physiology, Georg August University, Göttingen, Germany
| | - M J Baier
- Department of Internal Medicine II, University Medical Center Regensburg, Germany
| | - S Wagner
- Department of Internal Medicine II, University Medical Center Regensburg, Germany
| | - D M Katschinski
- Institute for Cardiovascular Physiology, Georg August University, Göttingen, Germany
| | - L S Maier
- Department of Internal Medicine II, University Medical Center Regensburg, Germany
| | - C M Sag
- Department of Internal Medicine II, University Medical Center Regensburg, Germany
| |
Collapse
|
47
|
Fill M, Gillespie D. Simulating cardiac Ca 2+ release units: effects of RyR cluster size and Ca 2+ buffers on diastolic Ca 2+ leak. Pflugers Arch 2021; 473:435-446. [PMID: 33608799 DOI: 10.1007/s00424-021-02539-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/27/2021] [Accepted: 02/05/2021] [Indexed: 10/22/2022]
Abstract
Leak of Ca2+ out of the cardiac sarcoplasmic reticulum (SR) via ryanodine receptors (RyRs) during diastole is vital to regulate SR Ca2+ levels. This leak can become deleterious when large spontaneous RyR-mediated Ca2+ release events evoke proarrhythmic Ca2+ waves that can lead to delayed after-depolarizations. Here, we model diastolic SR Ca2+ leak at individual SR Ca2+ release sites using computer simulations of RyR arrays like those in the dyadic cleft. The results show that RyR arrays size has a significant effect on SR Ca2+ leak, with bigger arrays producing larger and more frequent Ca2+ release events. Moreover, big RyR arrays are more susceptible to small changes in the levels of dyadic Ca2+ buffers. Such changes in buffering shift Ca2+ leak from small Ca2+ release events (involving few open RyRs) to larger events (with many open RyRs). Moreover, by analyzing a large parameter space of possible buffering and SR Ca2+ loads, we find further evidence for the hypothesis that SR Ca2+ leak by RyR arrays can undergo a sudden phase transition.
Collapse
Affiliation(s)
- Michael Fill
- Department of Physiology and Biophysics, Rush University Medical Center, Chicago, IL, USA
| | - Dirk Gillespie
- Department of Physiology and Biophysics, Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
48
|
The electrogenic sodium bicarbonate cotransporter and its roles in the myocardial ischemia-reperfusion induced cardiac diseases. Life Sci 2021; 270:119153. [PMID: 33539911 DOI: 10.1016/j.lfs.2021.119153] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 01/06/2021] [Accepted: 01/22/2021] [Indexed: 12/19/2022]
Abstract
Cardiac tissue ischemia/hypoxia increases glycolysis and lactic acid accumulation in cardiomyocytes, leading to intracellular metabolic acidosis. Sodium bicarbonate cotransporters (NBCs) play a vital role in modulating intracellular pH and maintaining sodium ion concentrations in cardiomyocytes. Cardiomyocytes mainly express electrogenic sodium bicarbonate cotransporter (NBCe1), which has been demonstrated to participate in myocardial ischemia/reperfusion (I/R) injury. This review outlines the structural and functional properties of NBCe1, summarizes the signaling pathways and factors that may regulate the activity of NBCe1, and reviews the roles of NBCe1 in the pathogenesis of I/R-induced cardiac diseases. Further studies revealing the regulatory mechanisms of NBCe1 activity should provide novel therapeutic targets for preventing I/R-induced cardiac diseases.
Collapse
|
49
|
Shaul D, Azar A, Sapir G, Uppala S, Nardi-Schreiber A, Gamliel A, Sosna J, Gomori JM, Katz-Brull R. Correlation between lactate dehydrogenase/pyruvate dehydrogenase activities ratio and tissue pH in the perfused mouse heart: A potential noninvasive indicator of cardiac pH provided by hyperpolarized magnetic resonance. NMR IN BIOMEDICINE 2021; 34:e4444. [PMID: 33258527 DOI: 10.1002/nbm.4444] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 10/05/2020] [Accepted: 10/24/2020] [Indexed: 06/12/2023]
Abstract
Cardiovascular diseases account for more than 30% of all deaths worldwide and many could be ameliorated with early diagnosis. Current cardiac imaging modalities can assess blood flow, heart anatomy and mechanical function. However, for early diagnosis and improved treatment, further functional biomarkers are needed. One such functional biomarker could be the myocardium pH. Although tissue pH is already determinable via MR techniques, and has been since the early 1990s, it remains elusive to use practically. The objective of this study was to explore the possibility to evaluate cardiac pH noninvasively, using in-cell enzymatic rates of hyperpolarized [1-13 C]pyruvate metabolism (ie, moles of product produced per unit time) determined directly in real time using magnetic resonance spectroscopy in a perfused mouse heart model. As a gold standard for tissue pH we used 31 P spectroscopy and the chemical shift of the inorganic phosphate (Pi) signal. The nonhomogenous pH distribution of the perfused heart was analyzed using a multi-parametric analysis of this signal, thus taking into account the heterogeneous nature of this characteristic. As opposed to the signal ratio of hyperpolarized [13 C]bicarbonate to [13 CO2 ], which has shown correlation to pH in other studies, we investigated here the ratio of two intracellular enzymatic rates: lactate dehydrogenase (LDH) and pyruvate dehydrogenase (PDH), by way of determining the production rates of [1-13 C]lactate and [13 C]bicarbonate, respectively. The enzyme activities determined here are intracellular, while the pH determined using the Pi signal may contain an extracellular component, which could not be ruled out. Nevertheless, we report a strong correlation between the tissue pH and the LDH/PDH activities ratio. This work may pave the way for using the LDH/PDH activities ratio as an indicator of cardiac intracellular pH in vivo, in an MRI examination.
Collapse
Affiliation(s)
- David Shaul
- Department of Radiology, Hadassah Medical Center, Hebrew University of Jerusalem, The Faculty of Medicine, Jerusalem, Israel
| | - Assad Azar
- Department of Radiology, Hadassah Medical Center, Hebrew University of Jerusalem, The Faculty of Medicine, Jerusalem, Israel
| | - Gal Sapir
- Department of Radiology, Hadassah Medical Center, Hebrew University of Jerusalem, The Faculty of Medicine, Jerusalem, Israel
| | - Sivaranjan Uppala
- Department of Radiology, Hadassah Medical Center, Hebrew University of Jerusalem, The Faculty of Medicine, Jerusalem, Israel
| | - Atara Nardi-Schreiber
- Department of Radiology, Hadassah Medical Center, Hebrew University of Jerusalem, The Faculty of Medicine, Jerusalem, Israel
| | - Ayelet Gamliel
- Department of Radiology, Hadassah Medical Center, Hebrew University of Jerusalem, The Faculty of Medicine, Jerusalem, Israel
| | - Jacob Sosna
- Department of Radiology, Hadassah Medical Center, Hebrew University of Jerusalem, The Faculty of Medicine, Jerusalem, Israel
| | - J Moshe Gomori
- Department of Radiology, Hadassah Medical Center, Hebrew University of Jerusalem, The Faculty of Medicine, Jerusalem, Israel
| | - Rachel Katz-Brull
- Department of Radiology, Hadassah Medical Center, Hebrew University of Jerusalem, The Faculty of Medicine, Jerusalem, Israel
| |
Collapse
|
50
|
Kandilci HB, Richards MA, Fournier M, Şimşek G, Chung YJ, Lakhal-Littleton S, Swietach P. Cardiomyocyte Na +/H + Exchanger-1 Activity Is Reduced in Hypoxia. Front Cardiovasc Med 2021; 7:617038. [PMID: 33585583 PMCID: PMC7873356 DOI: 10.3389/fcvm.2020.617038] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 12/29/2020] [Indexed: 12/30/2022] Open
Abstract
Fully-activated Na+/H+ exchanger-1 (NHE1) generates the cardiomyocyte's largest trans-membrane extrusion of H+ ions for an equimolar influx of Na+ ions. This has the desirable effect of clearing excess intracellular acidity, but comes at a large energetic premium because the exchanged Na+ ions must ultimately be extruded by the sodium pump, a process that consumes the majority of the heart's non-contractile ATP. We hypothesize that the state of NHE1 activation depends on metabolic resources, which become limiting in periods of myocardial hypoxia. To test this functionally, NHE1 activity was measured in response to in vitro and in vivo hypoxic treatments. NHE1 flux was interrogated as a function of intracellular pH by fluorescence imaging of rodent ventricular myocytes loaded with pH-sensitive dyes BCECF or cSNARF1. Anoxic superfusates promptly inhibited NHE1, tracking the time-course of mitochondrial depolarization. Mass spectrometry of NHE1 immuno-precipitated from Langendorff-perfused anoxic hearts identified Tyr-581 dephosphorylation and Tyr-561 phosphorylation. The latter residue is part of the domain that interacts with phosphatidylinositol 4,5-bisphosphate (PIP2), a membrane lipid that becomes depleted under metabolic inhibition. Tyr-561 phosphorylation is expected to electrostatically weaken this activatory interaction. To test if a period of hypoxia produces a persistent inhibition of NHE1, measurements under normoxia were performed on myocytes that had been incubated in 2% O2 for 4 h. NHE1 activity remained inhibited, but the effect was ablated in the presence of Dasatinib, an inhibitor of Abl/Src-family tyrosine kinases. Chronic tissue hypoxia in vivo, attained in a mouse model of anemic hypoxia, also resulted in persistently slower NHE1. In summary, we show that NHE1 responds to oxygen, a physiologically-relevant metabolic regulator, ostensibly to divert ATP for contraction. We describe a novel mechanism of NHE1 inhibition that may be relevant in cardiac disorders featuring altered oxygen metabolism, such as myocardial ischemia and reperfusion injury.
Collapse
Affiliation(s)
- Hilmi Burak Kandilci
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, United Kingdom.,Department of Biophysics, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Mark A Richards
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, United Kingdom
| | - Marjorie Fournier
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Gül Şimşek
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, United Kingdom.,Department of Biophysics, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Yu Jin Chung
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, United Kingdom
| | - Samira Lakhal-Littleton
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, United Kingdom
| | - Pawel Swietach
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|