1
|
Atta A, Salem MM, Reda A, Mohamed TM. "Carnosine-Niosomal Delivery System for Targeted Cancer Therapy". Cell Biochem Biophys 2025; 83:1495-1520. [PMID: 39656368 DOI: 10.1007/s12013-024-01626-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2024] [Indexed: 05/20/2025]
Abstract
Cancer is considered to be among the main causes of death worldwide. Treatment options for cancer are numerous. The type of cancer and its stage of progression determine which kind of treatment is needed. Nanomedicine is a new field for the treatment of various diseases. Pharmaceutical nanocarriers can be fabricated from various materials such as polymers, metals, or lipid-based surfactants. Carnosine-loaded niosomes have emerged as a promising approach in targeted cancer therapy, offering potential advantages over conventional treatments such as chemotherapy and radiation, by improving drug delivery specificity and reducing side effects. The study demonstrates that the encapsulation of carnosine in niosomes enhances its stability and bioavailability, leading to a significant increase in anticancer efficacy. These findings suggest that niosome technology can serve as an effective delivery system for carnosine, potentially transforming its use in cancer treatment and paving the way for future research in targeted therapies. Nanomaterials provide a good delivery system for this method of treatment. It's used in the treatment and diagnosis of diseases. Numerous investigations have been conducted on nanoscale vesicular systems, such as the most recent generations of vesicular nanocarriers, liposomes, and niosomes. Lipophilic and hydrophilic bioactive chemicals are transported via the niosomes in a vesicle. Since niosomes are composed of non-ionic surfactants mixed with cholesterol or other amphiphilic substances, they have a wide range of applications. The therapy of cancer with carnosine-loaded niosomes is one of these uses. The body synthesizes carnosine, a histidine-containing dipeptide, by enzymatically mixing L-histidine and β-alanine. With its antioxidant activities, Carnosine is considered a drug that can reduce and treat cancerous cells and many other therapeutic applications.
Collapse
Affiliation(s)
- Amira Atta
- Biochemistry Division- Chemistry Department- Faculty of Science- Tanta University, Tanta, 31527, Egypt
| | - Maha M Salem
- Biochemistry Division- Chemistry Department- Faculty of Science- Tanta University, Tanta, 31527, Egypt.
| | - Ahmed Reda
- Biochemistry Division- Chemistry Department- Faculty of Science- Tanta University, Tanta, 31527, Egypt
| | - Tarek M Mohamed
- Biochemistry Division- Chemistry Department- Faculty of Science- Tanta University, Tanta, 31527, Egypt
| |
Collapse
|
2
|
Chen Y, Xiao D, Li X. Lactylation and Central Nervous System Diseases. Brain Sci 2025; 15:294. [PMID: 40149815 PMCID: PMC11940311 DOI: 10.3390/brainsci15030294] [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: 01/22/2025] [Revised: 03/01/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
As the final product of glycolysis, lactate serves as an energy substrate, metabolite, and signaling molecule in various diseases and mediates lactylation, an epigenetic modification that occurs under both physiological and pathological conditions. Lactylation is a crucial mechanism by which lactate exerts its functions, participating in vital biological activities such as glycolysis-related cellular functions, macrophage polarization, and nervous system regulation. Lactylation links metabolic regulation to central nervous system (CNS) diseases, such as traumatic brain injury, Alzheimer's disease, acute ischemic stroke, and schizophrenia, revealing the diverse functions of lactylation in the CNS. In the future, further exploration of lactylation-associated enzymes and proteins is needed to develop specific lactylation inhibitors or activators, which could provide new tools and strategies for the treatment of CNS diseases.
Collapse
Affiliation(s)
- Ye Chen
- Department of Emergency Medicine, West China Second University Hospital, Sichuan University, Chengdu 610041, China; (Y.C.); (D.X.)
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu 610041, China
| | - Dongqiong Xiao
- Department of Emergency Medicine, West China Second University Hospital, Sichuan University, Chengdu 610041, China; (Y.C.); (D.X.)
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu 610041, China
| | - Xihong Li
- Department of Emergency Medicine, West China Second University Hospital, Sichuan University, Chengdu 610041, China; (Y.C.); (D.X.)
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu 610041, China
| |
Collapse
|
3
|
Farkas E, Rose CR. A dangerous liaison: Spreading depolarization and tissue acidification in cerebral ischemia. J Cereb Blood Flow Metab 2025; 45:201-218. [PMID: 39535276 PMCID: PMC12000947 DOI: 10.1177/0271678x241289756] [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: 03/28/2024] [Revised: 08/30/2024] [Accepted: 08/30/2024] [Indexed: 11/16/2024]
Abstract
Brain pH is precisely regulated, and pH transients associated with activity are rapidly restored under physiological conditions. During ischemia, the brain's ability to buffer pH changes is rapidly depleted. Tissue oxygen deprivation causes a shift from aerobic to anaerobic metabolism and the accumulation of lactic acid and protons. Although the degree of tissue acidosis resulting from ischemia depends on the severity of the ischemia, spreading depolarization (SD) events emerge as central elements to determining ischemic tissue acidosis. A marked decrease in tissue pH during cerebral ischemia may exacerbate neuronal injury, which has become known as acidotoxicity, in analogy to excitotoxicity. The cellular pathways underlying acidotoxicity have recently been described in increasing detail. The molecular structure of acid or base carriers and acidosis-activated ion channels, the precise (dys)homeostatic conditions under which they are activated, and their possible role in severe ischemia have been addressed. The expanded understanding of acidotoxic mechanisms now provides an opportunity to reevaluate the contexts that lead to acidotoxic injury. Here, we review the specific cellular pathways of acidotoxicity and demonstrate that SD plays a central role in activating the molecular machinery leading to acid-induced damage. We propose that SD is a key contributor to acidotoxic injury in cerebral ischemia.
Collapse
Affiliation(s)
- Eszter Farkas
- Hungarian Centre of Excellence for Molecular Medicine – University of Szeged, Cerebral Blood Flow and Metabolism Research Group, Szeged, Hungary
- Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Christine R Rose
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
4
|
Huang Z, Shi B, Mu X, Qiao S, Xiao G, Wang Y, Xu Y. Construction of a Dataset for All Expressed Transcripts for Alzheimer's Disease Research. Brain Sci 2024; 14:1180. [PMID: 39766379 PMCID: PMC11674848 DOI: 10.3390/brainsci14121180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
Accurate identification and functional annotation of splicing isoforms and non-coding RNAs (lncRNAs), alongside full-length protein-encoding transcripts, are critical for understanding gene (mis)regulation and metabolic reprogramming in Alzheimer's disease (AD). This study aims to provide a comprehensive and accurate transcriptome resource to improve existing AD transcript databases. Background/Objectives: Gene mis-regulation and metabolic reprogramming play a key role in AD, yet existing transcript databases lack accurate and comprehensive identification of splicing isoforms and lncRNAs. This study aims to generate a refined transcriptome dataset, expanding the understanding of AD onset and progression. Methods: Publicly available RNA-seq data from pre-AD and AD tissues were utilized. Advanced bioinformatics tools were applied to assemble and annotate full-length transcripts, including splicing isoforms and lncRNAs, with an emphasis on correcting errors and enhancing annotation accuracy. Results: A significantly improved transcriptome dataset was generated, which includes detailed annotations of splicing isoforms and lncRNAs. This dataset expands the scope of existing AD transcript databases and provides new insights into the molecular mechanisms underlying AD. The findings demonstrate that the refined dataset captures more relevant details about AD progression compared to publicly available data. Conclusions: The newly developed transcriptome resource and the associated analysis tools offer a valuable contribution to AD research, providing deeper insights into the disease's molecular mechanisms. This work supports future research into gene regulation and metabolic reprogramming in AD and serves as a foundation for exploring novel therapeutic targets.
Collapse
Affiliation(s)
- Zhenyu Huang
- College of Computer Science and Technology, Jilin University, Changchun 130012, China; (Z.H.); (G.X.)
- Systems Biology Lab for Metabolic Reprogramming, Department of Human Genetics and Cell Biology, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (B.S.); (X.M.); (S.Q.)
| | - Bocheng Shi
- Systems Biology Lab for Metabolic Reprogramming, Department of Human Genetics and Cell Biology, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (B.S.); (X.M.); (S.Q.)
- School of Mathematics, Jilin University, Changchun 130012, China
| | - Xuechen Mu
- Systems Biology Lab for Metabolic Reprogramming, Department of Human Genetics and Cell Biology, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (B.S.); (X.M.); (S.Q.)
- School of Mathematics, Jilin University, Changchun 130012, China
| | - Siyu Qiao
- Systems Biology Lab for Metabolic Reprogramming, Department of Human Genetics and Cell Biology, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (B.S.); (X.M.); (S.Q.)
| | - Gangyi Xiao
- College of Computer Science and Technology, Jilin University, Changchun 130012, China; (Z.H.); (G.X.)
| | - Yan Wang
- College of Computer Science and Technology, Jilin University, Changchun 130012, China; (Z.H.); (G.X.)
| | - Ying Xu
- Systems Biology Lab for Metabolic Reprogramming, Department of Human Genetics and Cell Biology, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (B.S.); (X.M.); (S.Q.)
| |
Collapse
|
5
|
Bouyer PG, Salameh AI, Zhou Y, Kolba TN, Boron WF. Effects of extracellular metabolic acidosis and out-of-equilibrium CO 2/HCO 3 - solutions on intracellular pH in cultured rat hippocampal neurons. Front Physiol 2024; 15:1434359. [PMID: 39444753 PMCID: PMC11496273 DOI: 10.3389/fphys.2024.1434359] [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: 05/17/2024] [Accepted: 08/28/2024] [Indexed: 10/25/2024] Open
Abstract
Metabolic acidosis (MAc)-an extracellular pH (pHo) decrease caused by a [HCO3 -]o decrease at constant [CO2]o-usually causes intracellular pH (pHi) to fall. Here we determine the extent to which the pHi decrease depends on the pHo decrease vs the concomitant [HCO3 -]o decrease. We use rapid-mixing to generate out-of-equilibrium CO2/HCO3 - solutions in which we stabilize [CO2]o and [HCO3 -]o while decreasing pHo (pure acidosis, pAc), or stabilize [CO2]o and pHo while decreasing [HCO3 -]o (pure metabolic/down, pMet↓). Using the fluorescent dye 2',7'-bis-2-carboxyethyl)-5(and-6)carboxyfluorescein (BCECF) to monitor pHi in rat hippocampal neurons in primary culture, we find that-in naïve neurons-the pHi decrease caused by MAc is virtually the sum of those caused by pAc (∼70%) + pMet↓ (∼30%). However, if we impose a first challenge (MAc1, pAc1, or pMet↓1), allow the neurons to recover, and then impose a second challenge (MAc2, pAc2, or pMet↓2), we find that pAc/pMet↓ additivity breaks down. In a twin-challenge protocol in which challenge #2 is MAc, the pHo and [HCO3 -]o decreases during challenge #1 must be coincident in order to mimic the effects of MAc1 on MAc2. Conversely, if challenge #1 is MAc, then the pHo and [HCO3 -]o decreases during challenge #2 must be coincident in order for MAc1 to produce its physiological effects during the challenge #2 period. We conclude that the history of challenge #1 (MAc1, pAc1, or pMet↓1)-presumably as detected by one or more acid-base sensors-has a major impact on the pHi response during challenge #2 (MAc2, pAc2, or pMet↓2).
Collapse
Affiliation(s)
- Patrice G. Bouyer
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, United States
- Department of Biology, Valparaiso University, Valparaiso, IN, United States
| | - Ahlam I. Salameh
- Preclinical Sciences Division, Kent State University College of Podiatric Medicine, Independence, OH, United States
- Department of Physiology & Biophysics Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Yuehan Zhou
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, United States
- Department of Physiology & Biophysics Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Tiffany N. Kolba
- Department of Mathematics & Statistics, Valparaiso University, Valparaiso, IN, United States
| | - Walter F. Boron
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, United States
- Department of Physiology & Biophysics Case Western Reserve University School of Medicine, Cleveland, OH, United States
| |
Collapse
|
6
|
Su Y, Chen J, Hu J, Qian C, Ma J, Brynjolfsson S, Fu W. Manipulation of ion/electron carrier genes in the model diatom Phaeodactylum tricornutum enables its growth under lethal acidic stress. iScience 2024; 27:110482. [PMID: 39758278 PMCID: PMC11700652 DOI: 10.1016/j.isci.2024.110482] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 05/14/2024] [Accepted: 07/08/2024] [Indexed: 01/07/2025] Open
Abstract
A major obstacle to exploiting industrial flue gas for microalgae cultivation is the unfavorable acidic environment. We previously identified three upregulated genes in the low-pH-adapted model diatom Phaeodactylum tricornutum: ferredoxin (PtFDX), cation/proton antiporter (PtCPA), and HCO3 - transporter (PtSCL4-2). Here, we individually overexpressed these genes in P. tricornutum to investigate their respective roles in resisting acidic stress (pH 5.0). The genetic modifications enabled positive growths of transgenic strains under acidic stress that completely inhibited the growth of the wild-type strain. Physiological studies indicated improved photosynthesis and reduced oxidative stress in the transgenic strains. Transcriptomes of the PtCPA- and PtSCL4-2-overexpressing transgenics showed widespread upregulation of various transmembrane transporters, which could help counteract excessive external protons. This work highlights ion/electron carrier genes' role in enhancing diatom resistance to acidic stress, providing insights into phytoplankton adaptation to ocean acidification and a strategy for biological carbon capture and industrial flue gas CO2 utilization.
Collapse
Affiliation(s)
- Yixi Su
- Ocean College, Zhejiang University, Zhoushan, Zhejiang 316021, China
- Center for Systems Biology and Faculty of Industrial Engineering, School of Engineering and Natural Sciences, University of Iceland, 101 Reykjavík, Iceland
| | - Jiwei Chen
- Ocean College, Zhejiang University, Zhoushan, Zhejiang 316021, China
| | - Jingyan Hu
- Ocean College, Zhejiang University, Zhoushan, Zhejiang 316021, China
| | - Cheng Qian
- Ocean College, Zhejiang University, Zhoushan, Zhejiang 316021, China
- School of Biological Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Jiahao Ma
- Ocean College, Zhejiang University, Zhoushan, Zhejiang 316021, China
| | - Sigurður Brynjolfsson
- Center for Systems Biology and Faculty of Industrial Engineering, School of Engineering and Natural Sciences, University of Iceland, 101 Reykjavík, Iceland
| | - Weiqi Fu
- Ocean College, Zhejiang University, Zhoushan, Zhejiang 316021, China
- Center for Systems Biology and Faculty of Industrial Engineering, School of Engineering and Natural Sciences, University of Iceland, 101 Reykjavík, Iceland
| |
Collapse
|
7
|
Rieck J, Derst C, Veh RW. Polyamines (PAs) but not small peptides with closely spaced positively charged groups interact with DNA and RNA, but they do not represent a relevant buffer system at physiological pH values. PLoS One 2024; 19:e0304658. [PMID: 39052628 PMCID: PMC11271873 DOI: 10.1371/journal.pone.0304658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 05/15/2024] [Indexed: 07/27/2024] Open
Abstract
Polyamines (PAs) including putrescine (PUT), spermidine (SPD) and spermine (SPM) are small, versatile molecules with two or more positively charged amino groups. Despite their importance for almost all forms of life, their specific roles in molecular and cellular biology remain partly unknown. The molecular structures of PAs suggest two presumable biological functions: (i) as potential buffer systems and (ii) as interactants with poly-negatively charged molecules like nucleic acids. The present report focuses on the question, whether the molecular structures of PAs are essential for such functions, or whether other simple molecules like small peptides with closely spaced positively charged side chains might be suitable as well. Consequently, we created titration curves for PUT, SPD, and SPM, as well as for oligolysines like tri-, tetra-, and penta-lysine. None of the molecules provided substantial buffering capacity at physiological intracellular pH values. Apparently, the most important mechanism for intracellular pH homeostasis in neurons is not a buffer system but is provided by the actions of the sodium-hydrogen and the bicarbonate-chloride antiporters. In a similar approach we investigated the interaction with DNA by following the extinction at 260 nm when titrating DNA with the above molecules. Again, PUT and tri-lysine were not able to interact with herring sperm DNA, while SPD and SPM were. Obviously, the presence of several positively charged groups on its own is not sufficient for the interaction with nucleic acids. Instead, the precise spacing of these groups is necessary for biological activity.
Collapse
Affiliation(s)
- Julian Rieck
- Institut für Zell- und Neurobiologie, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Christian Derst
- Institut für Zell- und Neurobiologie, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Rüdiger W. Veh
- Institut für Zell- und Neurobiologie, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
8
|
Ping H, Ding D, Zhu G, Wang J, Zhang J. Advancements in the application of nanotechnology for the management of epileptic seizures. ACTA EPILEPTOLOGICA 2024; 6:23. [PMID: 40217331 PMCID: PMC11960228 DOI: 10.1186/s42494-024-00171-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 06/23/2024] [Indexed: 04/15/2025] Open
Abstract
Epilepsy is a common yet complex neurological disorder. Historically, antiseizure medications (ASMs) have faced challenges in crossing the blood-brain barrier (BBB) and targeting the epileptogenic zone, creating a bottleneck in seizure management. Certain nanomaterials can facilitate drug penetration through the BBB and enable stimulus-responsive drug release, thereby enhancing targeted and efficient drug utilization while reducing adverse reactions in other brain tissues and peripherally. This article reviews the current researches on stimulus-responsive nanosystems applicable in antiepileptic therapy, as well as nanotechnology applications that improve the brain delivery of ASMs.
Collapse
Affiliation(s)
- Honglu Ping
- Department of Neurology, Huashan Hospital, Fudan University, 12 Wulumuqi Middle Road, Shanghai, 200040, China
| | - Ding Ding
- Department of Neurology, Huashan Hospital, Fudan University, 12 Wulumuqi Middle Road, Shanghai, 200040, China
| | - Guoxing Zhu
- Department of Neurology, Huashan Hospital, Fudan University, 12 Wulumuqi Middle Road, Shanghai, 200040, China
| | - Jianhong Wang
- Department of Neurology, Huashan Hospital, Fudan University, 12 Wulumuqi Middle Road, Shanghai, 200040, China.
| | - Jun Zhang
- Department of Radiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, National Center for Neurological Disorders, Fudan University, Wulumuqi Middle Road No.12, Shanghai, 200040, China.
| |
Collapse
|
9
|
Theparambil SM, Begum G, Rose CR. pH regulating mechanisms of astrocytes: A critical component in physiology and disease of the brain. Cell Calcium 2024; 120:102882. [PMID: 38631162 PMCID: PMC11423562 DOI: 10.1016/j.ceca.2024.102882] [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: 03/05/2024] [Revised: 03/28/2024] [Accepted: 03/28/2024] [Indexed: 04/19/2024]
Abstract
Strict homeostatic control of pH in both intra- and extracellular compartments of the brain is fundamentally important, primarily due to the profound impact of free protons ([H+]) on neuronal activity and overall brain function. Astrocytes, crucial players in the homeostasis of various ions in the brain, actively regulate their intracellular [H+] (pHi) through multiple membrane transporters and carbonic anhydrases. The activation of astroglial pHi regulating mechanisms also leads to corresponding alterations in the acid-base status of the extracellular fluid. Notably, astrocyte pH regulators are modulated by various neuronal signals, suggesting their pivotal role in regulating brain acid-base balance in both health and disease. This review presents the mechanisms involved in pH regulation in astrocytes and discusses their potential impact on extracellular pH under physiological conditions and in brain disorders. Targeting astrocytic pH regulatory mechanisms represents a promising therapeutic approach for modulating brain acid-base balance in diseases, offering a potential critical contribution to neuroprotection.
Collapse
Affiliation(s)
- Shefeeq M Theparambil
- Faculty of Health and Medicine, Department of Biomedical and Life Sciences, Lancaster University, Lancaster, LA1 4YW, Lancaster, UK.
| | - Gulnaz Begum
- Department of Neurology, The Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA
| | - Christine R Rose
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| |
Collapse
|
10
|
Chávez JC, Carrasquel-Martínez G, Hernández-Garduño S, Matamoros Volante A, Treviño CL, Nishigaki T, Darszon A. Cytosolic and Acrosomal pH Regulation in Mammalian Sperm. Cells 2024; 13:865. [PMID: 38786087 PMCID: PMC11120249 DOI: 10.3390/cells13100865] [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: 04/19/2024] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
As in most cells, intracellular pH regulation is fundamental for sperm physiology. Key sperm functions like swimming, maturation, and a unique exocytotic process, the acrosome reaction, necessary for gamete fusion, are deeply influenced by pH. Sperm pH regulation, both intracellularly and within organelles such as the acrosome, requires a coordinated interplay of various transporters and channels, ensuring that this cell is primed for fertilization. Consistent with the pivotal importance of pH regulation in mammalian sperm physiology, several of its unique transporters are dependent on cytosolic pH. Examples include the Ca2+ channel CatSper and the K+ channel Slo3. The absence of these channels leads to male infertility. This review outlines the main transport elements involved in pH regulation, including cytosolic and acrosomal pH, that participate in these complex functions. We present a glimpse of how these transporters are regulated and how distinct sets of them are orchestrated to allow sperm to fertilize the egg. Much research is needed to begin to envision the complete set of players and the choreography of how cytosolic and organellar pH are regulated in each sperm function.
Collapse
Affiliation(s)
- Julio C. Chávez
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología (IBT), Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, Morelos, Mexico; (J.C.C.); (G.C.-M.)
| | - Gabriela Carrasquel-Martínez
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología (IBT), Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, Morelos, Mexico; (J.C.C.); (G.C.-M.)
- CITMER, Medicina Reproductiva, México City 11520, Mexico
| | - Sandra Hernández-Garduño
- Departamento de Morfología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México (UNAM), México City 04510, Mexico;
| | - Arturo Matamoros Volante
- Department of Electrical and Computer Engineering and School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA;
| | - Claudia L. Treviño
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología (IBT), Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, Morelos, Mexico; (J.C.C.); (G.C.-M.)
| | - Takuya Nishigaki
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología (IBT), Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, Morelos, Mexico; (J.C.C.); (G.C.-M.)
| | - Alberto Darszon
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología (IBT), Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, Morelos, Mexico; (J.C.C.); (G.C.-M.)
| |
Collapse
|
11
|
Lin J, Rivadeneira AP, Ye Y, Ryu C, Parvin S, Jang K, Garraway SM, Choi I. Sodium Bicarbonate Decreases Alcohol Consumption in Mice. Int J Mol Sci 2024; 25:5006. [PMID: 38732226 PMCID: PMC11084513 DOI: 10.3390/ijms25095006] [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: 03/06/2024] [Revised: 04/26/2024] [Accepted: 04/28/2024] [Indexed: 05/13/2024] Open
Abstract
We previously reported that mice with low neuronal pH drink more alcohol, demonstrating the importance of pH for alcohol reward and motivation. In this study, we tested whether systemic pH affects alcohol consumption and if so, whether it occurs by changing the alcohol reward. C57BL/6J mice were given NaHCO3 to raise their blood pH, and the animals' alcohol consumption was measured in the drinking-in-the-dark and two-bottle free choice paradigms. Alcohol consumption was also assessed after suppressing the bitterness of NaHCO3 with sucrose. Alcohol reward was evaluated using a conditioned place preference. In addition, taste sensitivity was assessed by determining quinine and sucrose preference. The results revealed that a pH increase by NaHCO3 caused mice to decrease their alcohol consumption. The decrease in high alcohol contents (20%) was significant and observed at different ages, as well as in both males and females. Alcohol consumption was also decreased after suppressing NaHCO3 bitterness. Oral gavage of NaHCO3 did not alter quinine and sucrose preference. In the conditioned place preference, NaHCO3-treated mice spent less time in the alcohol-injected chamber. Conclusively, the results show that raising systemic pH with NaHCO3 decreases alcohol consumption, as it decreases the alcohol reward value.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Inyeong Choi
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA; (J.L.); (Y.Y.); (C.R.); (S.P.); (K.J.); (S.M.G.)
| |
Collapse
|
12
|
Schulte S, Decker D, Nowduri B, Gries M, Christmann A, Meyszner A, Rabe H, Saumer M, Schäfer KH. Improving morphological and functional properties of enteric neuronal networks in vitro using a novel upside-down culture approach. Am J Physiol Gastrointest Liver Physiol 2024; 326:G567-G582. [PMID: 38193168 PMCID: PMC11376983 DOI: 10.1152/ajpgi.00170.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 12/05/2023] [Accepted: 12/21/2023] [Indexed: 01/10/2024]
Abstract
The enteric nervous system (ENS) comprises millions of neurons and glia embedded in the wall of the gastrointestinal tract. It not only controls important functions of the gut but also interacts with the immune system, gut microbiota, and the gut-brain axis, thereby playing a key role in the health and disease of the whole organism. Any disturbance of this intricate system is mirrored in an alteration of electrical functionality, making electrophysiological methods important tools for investigating ENS-related disorders. Microelectrode arrays (MEAs) provide an appropriate noninvasive approach to recording signals from multiple neurons or whole networks simultaneously. However, studying isolated cells of the ENS can be challenging, considering the limited time that these cells can be kept vital in vitro. Therefore, we developed an alternative approach cultivating cells on glass samples with spacers (fabricated by photolithography methods). The spacers allow the cells to grow upside down in a spatially confined environment while enabling acute consecutive recordings of multiple ENS cultures on the same MEA. Upside-down culture also shows beneficial effects on the growth and behavior of enteric neural cultures. The number of dead cells was significantly decreased, and neural networks showed a higher resemblance to the myenteric plexus ex vivo while producing more stable signals than cultures grown in the conventional way. Overall, our results indicate that the upside-down approach not only allows to investigate the impact of neurological diseases in vitro but could also offer insights into the growth and development of the ENS under conditions much closer to the in vivo environment.NEW & NOTEWORTHY In this study, we devised a novel approach for culturing and electrophysiological recording of the enteric nervous system using custom-made glass substrates with spacers. This allows to turn cultures of isolated myenteric plexus upside down, enhancing the use of the microelectrode array technique by allowing recording of multiple cultures consecutively using only one chip. In addition, upside-down culture led to significant improvements in the culture conditions, resulting in a more in vivo-like growth.
Collapse
Affiliation(s)
- Steven Schulte
- Department of Informatics and Microsystems Technology, University of Applied Sciences Kaiserslautern, Zweibrücken, Germany
| | - Dominique Decker
- Department of Informatics and Microsystems Technology, University of Applied Sciences Kaiserslautern, Zweibrücken, Germany
| | - Bharat Nowduri
- Department of Informatics and Microsystems Technology, University of Applied Sciences Kaiserslautern, Zweibrücken, Germany
| | - Manuela Gries
- Department of Informatics and Microsystems Technology, University of Applied Sciences Kaiserslautern, Zweibrücken, Germany
| | - Anne Christmann
- Department of Informatics and Microsystems Technology, University of Applied Sciences Kaiserslautern, Zweibrücken, Germany
| | - Antoine Meyszner
- Department of Informatics and Microsystems Technology, University of Applied Sciences Kaiserslautern, Zweibrücken, Germany
| | - Holger Rabe
- Department of Informatics and Microsystems Technology, University of Applied Sciences Kaiserslautern, Zweibrücken, Germany
| | - Monika Saumer
- Department of Informatics and Microsystems Technology, University of Applied Sciences Kaiserslautern, Zweibrücken, Germany
| | - Karl-Herbert Schäfer
- Department of Informatics and Microsystems Technology, University of Applied Sciences Kaiserslautern, Zweibrücken, Germany
| |
Collapse
|
13
|
Nohesara S, Abdolmaleky HM, Thiagalingam S. Potential for New Therapeutic Approaches by Targeting Lactate and pH Mediated Epigenetic Dysregulation in Major Mental Diseases. Biomedicines 2024; 12:457. [PMID: 38398057 PMCID: PMC10887322 DOI: 10.3390/biomedicines12020457] [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: 01/29/2024] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 02/25/2024] Open
Abstract
Multiple lines of evidence have shown that lactate-mediated pH alterations in the brains of patients with neuropsychiatric diseases such as schizophrenia (SCZ), Alzheimer's disease (AD) and autism may be attributed to mitochondrial dysfunction and changes in energy metabolism. While neuronal activity is associated with reduction in brain pH, astrocytes are responsible for rebalancing the pH to maintain the equilibrium. As lactate level is the main determinant of brain pH, neuronal activities are impacted by pH changes due to the binding of protons (H+) to various types of proteins, altering their structure and function in the neuronal and non-neuronal cells of the brain. Lactate and pH could affect diverse types of epigenetic modifications, including histone lactylation, which is linked to histone acetylation and DNA methylation. In this review, we discuss the importance of pH homeostasis in normal brain function, the role of lactate as an essential epigenetic regulatory molecule and its contributions to brain pH abnormalities in neuropsychiatric diseases, and shed light on lactate-based and pH-modulating therapies in neuropsychiatric diseases by targeting epigenetic modifications. In conclusion, we attempt to highlight the potentials and challenges of translating lactate-pH-modulating therapies to clinics for the treatment of neuropsychiatric diseases.
Collapse
Affiliation(s)
- Shabnam Nohesara
- Department of Medicine (Biomedical Genetics), Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA;
| | - Hamid Mostafavi Abdolmaleky
- Department of Medicine (Biomedical Genetics), Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA;
- Nutrition/Metabolism Laboratory, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Sam Thiagalingam
- Department of Medicine (Biomedical Genetics), Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA;
- Department of Pathology & Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
14
|
Wu J, Jin M, Tran Q, Kim M, Kim SI, Shin J, Park H, Shin N, Kang H, Shin HJ, Lee SY, Cui SB, Lee CJ, Lee WH, Kim DW. Employing the sustained-release properties of poly(lactic-co-glycolic acid) nanoparticles to reveal a novel mechanism of sodium-hydrogen exchanger 1 in neuropathic pain. Transl Res 2024; 263:53-72. [PMID: 37678757 DOI: 10.1016/j.trsl.2023.09.003] [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: 05/15/2023] [Revised: 08/16/2023] [Accepted: 09/01/2023] [Indexed: 09/09/2023]
Abstract
Neuropathic pain is caused by injury or disease of the somatosensory system, and its course is usually chronic. Several studies have been dedicated to investigating neuropathic pain-related targets; however, little attention has been paid to the persistent alterations that these targets, some of which may be crucial to the pathophysiology of neuropathic pain. The present study aimed to identify potential targets that may play a crucial role in neuropathic pain and validate their long-term impact. Through bioinformatics analysis of RNA sequencing results, we identified Slc9a1 and validated the reduced expression of sodium-hydrogen exchanger 1 (NHE1), the protein that Slc9a1 encodes, in the spinal nerve ligation (SNL) model. Colocalization analysis revealed that NHE1 is primarily co-localized with vesicular glutamate transporter 2-positive neurons. In vitro experiments confirmed that poly(lactic-co-glycolic acid) nanoparticles loaded with siRNA successfully inhibited NHE1 in SH-SY5Y cells, lowered intracellular pH, and increased intracellular calcium concentrations. In vivo experiments showed that sustained suppression of spinal NHE1 expression by siRNA-loaded nanoparticles resulted in delayed hyperalgesia in naïve and SNL model rats, whereas amiloride-induced transient suppression of NHE1 expression yielded no significant changes in pain sensitivity. We identified Slc9a1, which encodes NHE1, as a key gene in neuropathic pain. Utilizing the sustained release properties of nanoparticles enabled us to elucidate the chronic role of decreased NHE1 expression, establishing its significance in the mechanisms of neuropathic pain.
Collapse
Affiliation(s)
- Junhua Wu
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Neurology, Yanji Hospital, Yanji, China
| | - Meiling Jin
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Quangdon Tran
- Molecular Biology Laboratory, Department of Medical Laboratories, Hai Phong International Hospital, Hai Phong City, Vietnam
| | - Minwoo Kim
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Song I Kim
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Juhee Shin
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Hyewon Park
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Nara Shin
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Hyunji Kang
- Center for Cognition and Sociality, Life Science Cluster, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Hyo Jung Shin
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Sun Yeul Lee
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anesthesia and Pain Medicine, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Song-Biao Cui
- Department of Neurology, Affiliated Hospital of Yanbian University, Yanji, China
| | - C Justin Lee
- Center for Cognition and Sociality, Life Science Cluster, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Won Hyung Lee
- Department of Anesthesia and Pain Medicine, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Dong Woon Kim
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea.
| |
Collapse
|
15
|
Maurer M, Lazaridis T. Transmembrane β-Barrel Models of α-Synuclein Oligomers. J Chem Inf Model 2023; 63:7171-7179. [PMID: 37963823 DOI: 10.1021/acs.jcim.3c00997] [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: 11/16/2023]
Abstract
The aggregation of α-synuclein is implicated in a number of neurodegenerative diseases, such as Parkinson's and Multiple System Atrophy, but the role of these aggregates in disease development is not clear. One possible mechanism of cytotoxicity is the disturbance or permeabilization of cell membranes by certain types of oligomers. However, no high-resolution structure of such membrane-embedded complexes has ever been determined. Here we construct and evaluate putative transmembrane β-barrels formed by this protein. Examination of the α-synuclein sequence reveals two regions that could form membrane-embedded β-hairpins: 64-92 (the NAC), and 35-56, which harbors many familial Parkinson's mutations. The stability of β-barrels formed by these hairpins is examined first in implicit membrane pores and then by multimicrosecond all-atom simulations. We find that a NAC region barrel remains stably inserted and hydrated for at least 10 μs. A 35-56 barrel remains stably inserted in the membrane but dehydrates and collapses if all His50 are neutral or if His50 is replaced by Q. If half of the His50 are doubly protonated, the barrel takes an oval shape but remains hydrated for at least 10 μs. Possible implications of these findings for α-synuclein pathology are discussed.
Collapse
Affiliation(s)
- Manuela Maurer
- Department of Chemistry & Biochemistry, City College of New York/CUNY, 160 Convent Ave, New York, New York 10031, United States
| | - Themis Lazaridis
- Department of Chemistry & Biochemistry, City College of New York/CUNY, 160 Convent Ave, New York, New York 10031, United States
| |
Collapse
|
16
|
Fasham J, Huebner AK, Liebmann L, Khalaf-Nazzal R, Maroofian R, Kryeziu N, Wortmann SB, Leslie JS, Ubeyratna N, Mancini GMS, van Slegtenhorst M, Wilke M, Haack TB, Shamseldin HE, Gleeson JG, Almuhaizea M, Dweikat I, Abu-Libdeh B, Daana M, Zaki MS, Wakeling MN, McGavin L, Turnpenny PD, Alkuraya FS, Houlden H, Schlattmann P, Kaila K, Crosby AH, Baple EL, Hübner CA. SLC4A10 mutation causes a neurological disorder associated with impaired GABAergic transmission. Brain 2023; 146:4547-4561. [PMID: 37459438 PMCID: PMC10629776 DOI: 10.1093/brain/awad235] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/19/2023] [Accepted: 06/06/2023] [Indexed: 11/09/2023] Open
Abstract
SLC4A10 is a plasma-membrane bound transporter that utilizes the Na+ gradient to drive cellular HCO3- uptake, thus mediating acid extrusion. In the mammalian brain, SLC4A10 is expressed in principal neurons and interneurons, as well as in epithelial cells of the choroid plexus, the organ regulating the production of CSF. Using next generation sequencing on samples from five unrelated families encompassing nine affected individuals, we show that biallelic SLC4A10 loss-of-function variants cause a clinically recognizable neurodevelopmental disorder in humans. The cardinal clinical features of the condition include hypotonia in infancy, delayed psychomotor development across all domains and intellectual impairment. Affected individuals commonly display traits associated with autistic spectrum disorder including anxiety, hyperactivity and stereotyped movements. In two cases isolated episodes of seizures were reported in the first few years of life, and a further affected child displayed bitemporal epileptogenic discharges on EEG without overt clinical seizures. While occipitofrontal circumference was reported to be normal at birth, progressive postnatal microcephaly evolved in 7 out of 10 affected individuals. Neuroradiological features included a relative preservation of brain volume compared to occipitofrontal circumference, characteristic narrow sometimes 'slit-like' lateral ventricles and corpus callosum abnormalities. Slc4a10 -/- mice, deficient for SLC4A10, also display small lateral brain ventricles and mild behavioural abnormalities including delayed habituation and alterations in the two-object novel object recognition task. Collapsed brain ventricles in both Slc4a10-/- mice and affected individuals suggest an important role of SLC4A10 in the production of the CSF. However, it is notable that despite diverse roles of the CSF in the developing and adult brain, the cortex of Slc4a10-/- mice appears grossly intact. Co-staining with synaptic markers revealed that in neurons, SLC4A10 localizes to inhibitory, but not excitatory, presynapses. These findings are supported by our functional studies, which show the release of the inhibitory neurotransmitter GABA is compromised in Slc4a10-/- mice, while the release of the excitatory neurotransmitter glutamate is preserved. Manipulation of intracellular pH partially rescues GABA release. Together our studies define a novel neurodevelopmental disorder associated with biallelic pathogenic variants in SLC4A10 and highlight the importance of further analyses of the consequences of SLC4A10 loss-of-function for brain development, synaptic transmission and network properties.
Collapse
Affiliation(s)
- James Fasham
- RILD Wellcome Wolfson Centre, University of Exeter Medical School, Royal Devon University Healthcare NHS Foundation Trust, Exeter EX2 5DW, UK
- Peninsula Clinical Genetics Service, Royal Devon University Healthcare NHS Foundation Trust, Exeter EX2 5DW, UK
| | - Antje K Huebner
- Institute of Human Genetics, Jena University Hospital, Friedrich Schiller Universität, 07747 Jena, Germany
| | - Lutz Liebmann
- Institute of Human Genetics, Jena University Hospital, Friedrich Schiller Universität, 07747 Jena, Germany
| | - Reham Khalaf-Nazzal
- Department of Biomedical Sciences, Faculty of Medicine, Arab American University of Palestine, Jenin, P227, Palestine
| | - Reza Maroofian
- Molecular and Clinical Sciences Institute, St. George’s University of London, London SW17 0RE, UK
| | - Nderim Kryeziu
- Institute of Human Genetics, Jena University Hospital, Friedrich Schiller Universität, 07747 Jena, Germany
| | - Saskia B Wortmann
- University Children’s Hospital, Salzburger Landeskliniken (SALK) and Paracelsus Medical University (PMU), 5020 Salzburg, Austria
- Amalia Children’s Hospital, Radboudumc, 6525 GA Nijmegen, The Netherlands
- Institute of Human Genetics, Technische Universität München, 80333 Munich, Germany
| | - Joseph S Leslie
- RILD Wellcome Wolfson Centre, University of Exeter Medical School, Royal Devon University Healthcare NHS Foundation Trust, Exeter EX2 5DW, UK
| | - Nishanka Ubeyratna
- RILD Wellcome Wolfson Centre, University of Exeter Medical School, Royal Devon University Healthcare NHS Foundation Trust, Exeter EX2 5DW, UK
| | - Grazia M S Mancini
- Department of Clinical Genetics, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| | | | - Martina Wilke
- Department of Clinical Genetics, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Tobias B Haack
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, 72076 Tübingen, Germany
| | - Hanan E Shamseldin
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh 11564, Saudi Arabia
| | - Joseph G Gleeson
- Rady Children’s Institute for Genomic Medicine, San Diego, CA 92123, USA
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Mohamed Almuhaizea
- Department of Neuroscience, King Faisal Specialist Hospital and Research Center, Riyadh 11564, Saudi Arabia
| | - Imad Dweikat
- Department of Biomedical Sciences, Faculty of Medicine, Arab American University of Palestine, Jenin, P227, Palestine
| | - Bassam Abu-Libdeh
- Department of Pediatrics and Genetics, Makassed Hospital and Al-Quds University, East Jerusalem, 95908, Palestine
| | - Muhannad Daana
- Department of Pediatrics, Arab Women’s Union Hospital, Nablus, P400, Palestine
| | - Maha S Zaki
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Matthew N Wakeling
- RILD Wellcome Wolfson Centre, University of Exeter Medical School, Royal Devon University Healthcare NHS Foundation Trust, Exeter EX2 5DW, UK
| | - Lucy McGavin
- Department of Radiology, Derriford Hospital, Plymouth PL6 8DH, UK
| | - Peter D Turnpenny
- RILD Wellcome Wolfson Centre, University of Exeter Medical School, Royal Devon University Healthcare NHS Foundation Trust, Exeter EX2 5DW, UK
- Peninsula Clinical Genetics Service, Royal Devon University Healthcare NHS Foundation Trust, Exeter EX2 5DW, UK
| | - Fowzan S Alkuraya
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh 11564, Saudi Arabia
| | - Henry Houlden
- Molecular and Clinical Sciences Institute, St. George’s University of London, London SW17 0RE, UK
| | - Peter Schlattmann
- Institute for Medical Statistics, Computer Science and Data Science, Jena University Hospital, 07747 Jena, Germany
| | - Kai Kaila
- Molecular and Integrative Biosciences, University of Helsinki, 00014 Helsinki, Finland
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
| | - Andrew H Crosby
- RILD Wellcome Wolfson Centre, University of Exeter Medical School, Royal Devon University Healthcare NHS Foundation Trust, Exeter EX2 5DW, UK
| | - Emma L Baple
- RILD Wellcome Wolfson Centre, University of Exeter Medical School, Royal Devon University Healthcare NHS Foundation Trust, Exeter EX2 5DW, UK
- Peninsula Clinical Genetics Service, Royal Devon University Healthcare NHS Foundation Trust, Exeter EX2 5DW, UK
| | - Christian A Hübner
- Institute of Human Genetics, Jena University Hospital, Friedrich Schiller Universität, 07747 Jena, Germany
- Center for Rare Diseases, Jena University Hospital, Friedrich Schiller Universität, 07747 Jena, Germany
| |
Collapse
|
17
|
Abstract
Gap junctions between neurons of the brain are thought to be present in only certain cell types, and they mostly connect dendrites, somata, and axons. Synapses with gap junctions serve bidirectional metabolic and electrical coupling between connected neuronal compartments. Although plasticity of electrical synapses has been described, recent evidence of the presence of silent, but activatable, gap junctions suggests that electrical nodes in a neuronal circuit can be added or suppressed by changes in the synaptic microenvironment. This opens the possibility of reconfiguration of neuronal ensembles in response to activity. Moreover, the coexistence of gap junctions in a glutamatergic synapse may add electric and metabolic coupling to a neuronal aggregate and may serve to constitute primed ensembles within a higher-order neural network. The interaction of chemical with electrical synapses should be further explored to find, especially, emerging properties of neuronal ensembles. It will be worth to reexamine in a new light the "functional" implications of the "anatomic" concepts: "continuity" and "contiguity," which were championed by Golgi and Ramón y Cajal, respectively. In any case, exploring the versatility of the gap junctions will likely enrich the heuristic aspects of the neural and network postulates.
Collapse
Affiliation(s)
- Rafael Gutiérrez
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| |
Collapse
|
18
|
Wu A, Lee D, Xiong WC. Lactate Metabolism, Signaling, and Function in Brain Development, Synaptic Plasticity, Angiogenesis, and Neurodegenerative Diseases. Int J Mol Sci 2023; 24:13398. [PMID: 37686202 PMCID: PMC10487923 DOI: 10.3390/ijms241713398] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/21/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
Neural tissue requires a great metabolic demand despite negligible intrinsic energy stores. As a result, the central nervous system (CNS) depends upon a continuous influx of metabolic substrates from the blood. Disruption of this process can lead to impairment of neurological functions, loss of consciousness, and coma within minutes. Intricate neurovascular networks permit both spatially and temporally appropriate metabolic substrate delivery. Lactate is the end product of anaerobic or aerobic glycolysis, converted from pyruvate by lactate dehydrogenase-5 (LDH-5). Although abundant in the brain, it was traditionally considered a byproduct or waste of glycolysis. However, recent evidence indicates lactate may be an important energy source as well as a metabolic signaling molecule for the brain and astrocytes-the most abundant glial cell-playing a crucial role in energy delivery, storage, production, and utilization. The astrocyte-neuron lactate-shuttle hypothesis states that lactate, once released into the extracellular space by astrocytes, can be up-taken and metabolized by neurons. This review focuses on this hypothesis, highlighting lactate's emerging role in the brain, with particular emphasis on its role during development, synaptic plasticity, angiogenesis, and disease.
Collapse
Affiliation(s)
- Anika Wu
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (A.W.); (D.L.)
| | - Daehoon Lee
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (A.W.); (D.L.)
- Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA
| | - Wen-Cheng Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (A.W.); (D.L.)
- Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA
| |
Collapse
|
19
|
Giha HA. Hidden chronic metabolic acidosis of diabetes type 2 (CMAD): Clues, causes and consequences. Rev Endocr Metab Disord 2023; 24:735-750. [PMID: 37380824 DOI: 10.1007/s11154-023-09816-2] [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] [Accepted: 06/09/2023] [Indexed: 06/30/2023]
Abstract
Interpretation of existing data revealed that chronic metabolic acidosis is a pathognomic feature for type 2 diabetes (T2D), which is described here as "chronic metabolic acidosis of T2D (CMAD)" for the first time. The biochemical clues for the CMAD are summarised in the following; low blood bicarbonate (high anionic gap), low pH of interstitial fluid and urine, and response to acid neutralization, while the causes of extra protons are worked out to be; mitochondrial dysfunction, systemic inflammation, gut microbiota (GM), and diabetic lung. Although, the intracellular pH is largely preserved by the buffer system and ion transporters, a persistent systemic mild acidosis leaves molecular signature in cellular metabolism in diabetics. Reciprocally, there are evidences that CMAD contributes to the initiation and progression of T2D by; reducing insulin production, triggering insulin resistance directly or via altered GM, and inclined oxidative stress. The details about the above clues, causes and consequences of CMAD are obtained by searching literature spanning between 1955 and 2022. Finally, the molecular bases of CMAD are discussed in details by interpretation of an up-to-date data and aid of well constructed diagrams, with a conclusion unravelling that CMAD is a major player in T2D pathophysiology. To this end, the CMAD disclosure offers several therapeutic potentials for prevention, delay or attenuation of T2D and its complications.
Collapse
Affiliation(s)
- Hayder A Giha
- Medical Biochemistry and Molecular Biology, Khartoum, Sudan.
| |
Collapse
|
20
|
Fang X, Lu Y, Fu Y, Liu Z, Kermode AG, Qiu W, Ling L, Liu C. Cerebrospinal Fluid Chloride Is Associated with Disease Activity of Relapsing-Remitting Multiple Sclerosis: A Retrospective Cohort Study. Brain Sci 2023; 13:924. [PMID: 37371400 DOI: 10.3390/brainsci13060924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 05/27/2023] [Accepted: 06/04/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Blood-brain barrier dysfunction in active multiple sclerosis (MS) lesions leads to pathological changes in the cerebrospinal fluid (CSF). This study aimed to investigate the possible association between routine CSF findings, especially CSF chloride, at the time of the first lumbar puncture and the relapse risk and disability progression of relapsing-remitting MS (RRMS). METHODS This retrospective study included 77 patients with RRMS at the MS Center of our institution from January 2012 to December 2020. The Anderson and Gill (AG) model and Spearman correlation analysis were used to explore predictors of relapse and disability during follow-up. RESULTS In the multivariate AG model, patients with elevated CSF chloride level (hazard ratio [HR], 1.1; 95% confidence interval [CI]: 1.06-1.22; p = 0.001) had a high risk of MS relapse. Using median values of CSF chloride (123.2 mmol/L) as a cut-off, patients with CSF chloride level ≥ 123.2 mmol/L had a 120% increased relapse risk compared with those with CSF chloride level < 123.2 mmol/L (HR = 2.20; 95% CI: 1.19-4.05; p = 0.012). CONCLUSIONS Elevated CSF chloride levels might be a biologically unfavorable predictive factor for disease relapse in RRMS.
Collapse
Affiliation(s)
- Xingwei Fang
- Faculty of Medical Statistics, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Yaxin Lu
- Clinical Data Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Yongmei Fu
- Emergency Department, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Zifeng Liu
- Clinical Data Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Allan G Kermode
- Perron Institute, University of Western Australia, Nedlands, WA 6009, Australia
| | - Wei Qiu
- Neurology Department, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Li Ling
- Faculty of Medical Statistics, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
- Clinical Research Design Division, Clinical Research Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Chunxin Liu
- Emergency Department, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| |
Collapse
|
21
|
Panigrahi R, Krishnan R, Singh JS, Padinhateeri R, Kumar A. SUMO1 hinders α-Synuclein fibrillation by inducing structural compaction. Protein Sci 2023; 32:e4632. [PMID: 36974517 PMCID: PMC10108436 DOI: 10.1002/pro.4632] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 03/17/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023]
Abstract
Small Ubiquitin-like Modifier 1 (SUMO1) is an essential protein for many cellular functions, including regulation, signaling, etc., achieved by a process known as SUMOylation, which involves covalent attachment of SUMO1 to target proteins. SUMO1 also regulates the function of several proteins via non-covalent interactions involving the hydrophobic patch in the target protein identified as SUMO Binding or Interacting Motif (SBM/SIM). Here, we demonstrate a crucial functional potential of SUMO1 mediated by its non-covalent interactions with α-Synuclein, a protein responsible for many neurodegenerative diseases called α-Synucleinopathies. SUMO1 hinders the fibrillation of α-Synuclein, an intrinsically disordered protein (IDP) that undergoes a transition to β-structures during the fibrillation process. Using a plethora of biophysical techniques, we show that SUMO1 transiently binds to the N-terminus region of α-Synuclein non-covalently and causes structural compaction, which hinders the self-association process and thereby delays the fibrillation process. On the one hand, this study demonstrates an essential functional role of SUMO1 protein concerning neurodegeneration; it also illustrates the commonly stated mechanism that IDPs carry out multiple functions by structural adaptation to suit specific target proteins, on the other. Residue-level details about the SUMO1-α-Synuclein interaction obtained here also serve as a reliable approach for investigating the detailed mechanisms of IDP functions.
Collapse
Affiliation(s)
- Rajlaxmi Panigrahi
- Department of Biosciences and BioengineeringIndian Institute of Technology (IIT) BombayMumbaiMaharashtraIndia
| | - Rakesh Krishnan
- Department of Biosciences and BioengineeringIndian Institute of Technology (IIT) BombayMumbaiMaharashtraIndia
| | - Jai Shankar Singh
- Department of Biosciences and BioengineeringIndian Institute of Technology (IIT) BombayMumbaiMaharashtraIndia
| | - Ranjith Padinhateeri
- Department of Biosciences and BioengineeringIndian Institute of Technology (IIT) BombayMumbaiMaharashtraIndia
| | - Ashutosh Kumar
- Department of Biosciences and BioengineeringIndian Institute of Technology (IIT) BombayMumbaiMaharashtraIndia
| |
Collapse
|
22
|
Hatch RJ, Berecki G, Jancovski N, Li M, Rollo B, Jafar-Nejad P, Rigo F, Kaila K, Reid CA, Petrou S. Carbogen-Induced Respiratory Acidosis Blocks Experimental Seizures by a Direct and Specific Inhibition of Na V1.2 Channels in the Axon Initial Segment of Pyramidal Neurons. J Neurosci 2023; 43:1658-1667. [PMID: 36732074 PMCID: PMC10010452 DOI: 10.1523/jneurosci.1387-22.2022] [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: 07/18/2022] [Revised: 11/01/2022] [Accepted: 12/05/2022] [Indexed: 02/04/2023] Open
Abstract
Brain pH is a critical factor for determining neuronal activity, with alkalosis increasing and acidosis reducing excitability. Acid shifts in brain pH through the breathing of carbogen (5% CO2/95% O2) reduces seizure susceptibility in animal models and patients. The molecular mechanisms underlying this seizure protection remain to be fully elucidated. Here, we demonstrate that male and female mice exposed to carbogen are fully protected from thermogenic-triggered seizures. Whole-cell patch-clamp recordings revealed that acid shifts in extracellular pH (pHo) significantly reduce action potential firing in CA1 pyramidal neurons but did not alter firing in hippocampal inhibitory interneurons. In real-time dynamic clamp experiments, acidification reduced simulated action potential firing generated in hybrid model neurons expressing the excitatory neuron predominant NaV1.2 channel. Conversely, acidification had no effect on action potential firing in hybrid model neurons expressing the interneuron predominant NaV1.1 channel. Furthermore, knockdown of Scn2a mRNA in vivo using antisense oligonucleotides reduced the protective effects of carbogen on seizure susceptibility. Both carbogen-mediated seizure protection and the reduction in CA1 pyramidal neuron action potential firing by low pHo were maintained in an Asic1a knock-out mouse ruling out this acid-sensing channel as the underlying molecular target. These data indicate that the acid-mediated reduction in excitatory neuron firing is mediated, at least in part, through the inhibition of NaV1.2 channels, whereas inhibitory neuron firing is unaffected. This reduction in pyramidal neuron excitability is the likely basis of seizure suppression caused by carbogen-mediated acidification.SIGNIFICANCE STATEMENT Brain pH has long been known to modulate neuronal excitability. Here, we confirm that brain acidification reduces seizure susceptibility in a mouse model of thermogenic seizures. Extracellular acidification reduced excitatory pyramidal neuron firing while having no effect on interneuron firing. Acidification also reduced dynamic clamp firing in cells expressing the NaV1.2 channel but not in cells expressing NaV1.1 channels. In vivo knockdown of Scn2a mRNA reduced seizure protection of acidification. In contrast, acid-mediated seizure protection was maintained in the Asic1a knock-out mouse. These data suggest NaV1.2 channel as an important target for acid-mediated seizure protection. Our results have implications on how natural variations in pH can modulate neuronal excitability and highlight potential antiseizure drug development strategies based on the NaV1.2 channel.
Collapse
Affiliation(s)
- Robert J Hatch
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Géza Berecki
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Nikola Jancovski
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Melody Li
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Ben Rollo
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria 3052, Australia
| | | | - Frank Rigo
- Ionis Pharmaceuticals, Carlsbad, California 92008
| | - Kai Kaila
- Molecular and Integrative Biosciences and Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland
| | - Christopher A Reid
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Steven Petrou
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria 3052, Australia
| |
Collapse
|
23
|
Pandey AK, Buchholz CR, Nathan Kochen N, Pomerantz WCK, Braun AR, Sachs JN. pH Effects Can Dominate Chemical Shift Perturbations in 1H, 15N-HSQC NMR Spectroscopy for Studies of Small Molecule/α-Synuclein Interactions. ACS Chem Neurosci 2023; 14:800-808. [PMID: 36749138 DOI: 10.1021/acschemneuro.2c00782] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
1H,15N-Heteronuclear Single Quantum Coherence (HSQC) NMR is a powerful technique that has been employed to characterize small-molecule interactions with intrinsically disordered monomeric α-Synuclein (aSyn). We report how solution pH can impact the interpretation of aSyn HSQC NMR spectra and demonstrate that small-molecule formulations (e.g., complexation with acidic salts) can lower sample pH and confound interpretation of drug binding and concomitant protein structural changes. Through stringent pH control, we confirm that several previously identified compounds (EGCG, Baicalin, and Dopamine (DOPA)) as well as a series of potent small-molecule inhibitors of aSyn pathology (Demeclocycline, Ro90-7501, and (±)-Bay K 8644) are capable of direct target engagement of aSyn. Previously, DOPA-aSyn interactions have been shown to elicit a dramatic chemical shift perturbation (CSP) localized to aSyn's H50 at low DOPA concentrations then expanding to aSyn's acidic C-terminal residues at increasing DOPA levels. Interestingly, this CSP profile mirrors our pH titration, where a small reduction in pH affects H50 CSP, and large pH changes induce robust C-terminal CSP. In contrast, under tightly controlled pH 5.0, DOPA induces significant CSPs observed at both ionizable and nonionizable residues. These results suggest that previous interpretations of DOPA-aSyn interactions were conflated with pH-induced CSP, highlighting the need for stringent pH control to minimize potential false-positive interpretations of ligand interactions in HSQC NMR experiments. Furthermore, DOPA's preferential interaction with aSyn under acidic pH represents a novel understanding of DOPA-aSyn interactions that may provide insight into the potential gain of toxic function of aSyn misfolding in α-synucleinopathies.
Collapse
Affiliation(s)
- Anil K Pandey
- Dept. of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Caroline R Buchholz
- Dept. of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Noah Nathan Kochen
- Dept. of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - William C K Pomerantz
- Dept. of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States.,Dept. of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Anthony R Braun
- Dept. of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Jonathan N Sachs
- Dept. of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
24
|
Menzikov SA, Zaichenko DM, Moskovtsev AA, Morozov SG, Kubatiev AA. Zinc Inhibits the GABA AR/ATPase during Postnatal Rat Development: The Role of Cysteine Residue. Int J Mol Sci 2023; 24:ijms24032764. [PMID: 36769085 PMCID: PMC9917249 DOI: 10.3390/ijms24032764] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/24/2023] [Accepted: 01/26/2023] [Indexed: 02/04/2023] Open
Abstract
Zinc ions (Zn2+) are concentrated in various brain regions and can act as a neuromodulator, targeting a wide spectrum of postsynaptic receptors and enzymes. Zn2+ inhibits the GABAARs, and its potency is profoundly affected by the subunit composition and neuronal developmental stage. Although the extracellular amino acid residues of the receptor's hetero-oligomeric structure are preferred for Zn2+ binding, there are intracellular sites that, in principle, could coordinate its potency. However, their role in modulating the receptor function during postembryonic development remains unclear. The GABAAR possesses an intracellular ATPase that enables the energy-dependent anion transport via a pore. Here, we propose a mechanistic and molecular basis for the inhibition of intracellular GABAAR/ATPase function by Zn2+ in neonatal and adult rats. The enzymes within the scope of GABAAR performance as Cl-ATPase and then as Cl-, HCO3-ATPase form during the first week of postnatal rat development. In addition, we have shown that the Cl-ATPase form belongs to the β1 subunit, whereas the β3 subunit preferably possesses the Cl-, HCO3-ATPase activity. We demonstrated that a Zn2+ with variable efficacy inhibits the GABAAR as well as the ATPase activities of immature or mature neurons. Using fluorescence recording in the cortical synaptoneurosomes (SNs), we showed a competitive association between Zn2+ and NEM in parallel changes both in the ATPase activity and the GABAAR-mediated Cl- and HCO3- fluxes. Finally, by site-directed mutagenesis, we identified in the M3 domain of β subunits the cysteine residue (C313) that is essential for the manifestation of Zn2+ potency.
Collapse
|
25
|
Martinez Ramirez CE, Ruiz-Pérez G, Stollenwerk TM, Behlke C, Doherty A, Hillard CJ. Endocannabinoid signaling in the central nervous system. Glia 2023; 71:5-35. [PMID: 36308424 PMCID: PMC10167744 DOI: 10.1002/glia.24280] [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: 02/01/2022] [Revised: 09/02/2022] [Accepted: 09/29/2022] [Indexed: 11/07/2022]
Abstract
It is hard to overestimate the influence of the endocannabinoid signaling (ECS) system on central nervous system (CNS) function. In the 40 years since cannabinoids were found to trigger specific cell signaling cascades, studies of the ECS system continue to cause amazement, surprise, and confusion! CB1 cannabinoid receptors are expressed widely in the CNS and regulate cell-cell communication via effects on the release of both neurotransmitters and gliotransmitters. CB2 cannabinoid receptors are difficult to detect in the CNS but seem to "punch above their weight" as compounds targeting these receptors have significant effects on inflammatory state and behavior. Positive and negative allosteric modulators for both receptors have been identified and examined in preclinical studies. Concentrations of the endocannabinoid ligands, N-arachidonoylethanolamine and 2-arachidonoylglycerol (2-AG), are regulated by a combination of enzymatic synthesis and degradation and inhibitors of these processes are available and making their way into clinical trials. Importantly, ECS regulates many essential brain functions, including regulation of reward, anxiety, inflammation, motor control, and cellular development. While the field is on the cusp of preclinical discoveries providing impactful clinical and therapeutic insights into many CNS disorders, there is still much to be learned about this remarkable and versatile modulatory system.
Collapse
Affiliation(s)
- César E Martinez Ramirez
- Neuroscience Research Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Gonzalo Ruiz-Pérez
- Neuroscience Research Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Todd M Stollenwerk
- Neuroscience Research Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Christina Behlke
- Neuroscience Research Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Ashley Doherty
- Neuroscience Research Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Cecilia J Hillard
- Neuroscience Research Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
26
|
Michaiel AM, Bernard A. Neurobiology and changing ecosystems: Toward understanding the impact of anthropogenic influences on neurons and circuits. Front Neural Circuits 2022; 16:995354. [PMID: 36569799 PMCID: PMC9769128 DOI: 10.3389/fncir.2022.995354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/16/2022] [Indexed: 12/02/2022] Open
Abstract
Rapid anthropogenic environmental changes, including those due to habitat contamination, degradation, and climate change, have far-reaching effects on biological systems that may outpace animals' adaptive responses. Neurobiological systems mediate interactions between animals and their environments and evolved over millions of years to detect and respond to change. To gain an understanding of the adaptive capacity of nervous systems given an unprecedented pace of environmental change, mechanisms of physiology and behavior at the cellular and biophysical level must be examined. While behavioral changes resulting from anthropogenic activity are becoming increasingly described, identification and examination of the cellular, molecular, and circuit-level processes underlying those changes are profoundly underexplored. Hence, the field of neuroscience lacks predictive frameworks to describe which neurobiological systems may be resilient or vulnerable to rapidly changing ecosystems, or what modes of adaptation are represented in our natural world. In this review, we highlight examples of animal behavior modification and corresponding nervous system adaptation in response to rapid environmental change. The underlying cellular, molecular, and circuit-level component processes underlying these behaviors are not known and emphasize the unmet need for rigorous scientific enquiry into the neurobiology of changing ecosystems.
Collapse
|
27
|
Shin YS, Fomina N, Johnson C, Rocznik T, Ahmad H, Staley RPA, Weller J, Lang C. Toward Rapid and Automated Immunoassays: Using a Localized Electrochemical pH Modulation Platform to Perform a Single-Step Immunoassay. Anal Chem 2022; 94:13171-13180. [PMID: 36099239 DOI: 10.1021/acs.analchem.2c02686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
An electrochemical platform for generating and controlling a localized pH microenvironment on demand is proposed by employing a closed-loop control algorithm based on an iridium oxide pH sensor input. We use a combination of solution-borne quinones and galvanostatic excitation on a prepatterned indium tin oxide (ITO) working electrode to modulate pH within a very well confined, small volume of solution close to the electrode surface. We demonstrate that the rate of pH change can be controlled at up to 2 pH s-1 with an excellent repeatability (±0.004). The desired pH microenvironment can be stably maintained for longer than 2 h within ±0.0012 pH. As a high-impact application of the platform technology, we propose a single-step immunoassay and demonstrate its utility in measuring C-reactive protein (CRP), a critical inflammatory marker in various conditions such as myocardial infarction and even SARS-Cov-2. Utilizing pH modulation technology along with pH-sensitive fluorescence dye simplifies the immunoassay process into a single-step, where a mixture of all of the reagents is incubated only for 1 h without any washing steps or the need to change solution. This simplified immunoassay process minimizes the hands-on time of the end-user and thus decreases technician-driven errors. Moreover, the absence of complicated liquid-handling hardware makes it more suitable and attractive for an ultracompact platform to ultimately be used in a point-of-care diagnostic assay.
Collapse
Affiliation(s)
- Young Shik Shin
- Robert Bosch LLC, Research & Technology Center North America, 384 Santa Trinita Avenue, Sunnyvale, California 94085, United States
| | - Nadezda Fomina
- Robert Bosch LLC, Research & Technology Center North America, 384 Santa Trinita Avenue, Sunnyvale, California 94085, United States
| | - Christopher Johnson
- Robert Bosch LLC, Research & Technology Center North America, 384 Santa Trinita Avenue, Sunnyvale, California 94085, United States
| | - Thomas Rocznik
- Robert Bosch LLC, Research & Technology Center North America, 384 Santa Trinita Avenue, Sunnyvale, California 94085, United States
| | - Habib Ahmad
- Robert Bosch LLC, Research & Technology Center North America, 384 Santa Trinita Avenue, Sunnyvale, California 94085, United States
| | - Rachel Patricia-Andrea Staley
- Robert Bosch LLC, Research & Technology Center North America, 384 Santa Trinita Avenue, Sunnyvale, California 94085, United States
| | - Juliane Weller
- Robert Bosch LLC, Research & Technology Center North America, 384 Santa Trinita Avenue, Sunnyvale, California 94085, United States
| | - Christoph Lang
- Robert Bosch LLC, Research & Technology Center North America, 384 Santa Trinita Avenue, Sunnyvale, California 94085, United States
| |
Collapse
|
28
|
Wang J, Zahra A, Wang Y, Wu J. Understanding the Physiological Role of Electroneutral Na+-Coupled HCO3− Cotransporter and Its Therapeutic Implications. Pharmaceuticals (Basel) 2022; 15:ph15091082. [PMID: 36145304 PMCID: PMC9505461 DOI: 10.3390/ph15091082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/28/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
Acid–base homeostasis is critical for proper physiological function and pathology. The SLC4 family of HCO3− transmembrane cotransporters is one of the HCO3− transmembrane transport carriers responsible for cellular pH regulation and the uptake or secretion of HCO3− in epithelial cells. NBCn1 (SLC4A7), an electroneutral Na+/HCO3− cotransporter, is extensively expressed in several tissues and functions as a cotransporter for net acid extrusion after cellular acidification. However, the expression and activity level of NBCn1 remain elusive. In addition, NBCn1 has been involved in numerous other cellular processes such as cell volume, cell death/survival balance, transepithelial transport, as well as regulation of cell viability. This review aims to give an inclusive overview of the most recent advances in the research of NBCn1, emphasizing the basic features, regulation, and tissue-specific physiology as well as the development and application of potent inhibitors of NBCn1 transporter in cancer therapy. Research and development of targeted therapies should be carried out for NBCn1 and its associated pathways.
Collapse
Affiliation(s)
- Jingjing Wang
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
| | - Aqeela Zahra
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
| | - YunFu Wang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan 442000, China
| | - Jianping Wu
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan 442000, China
- Correspondence:
| |
Collapse
|
29
|
Pai VP, Cooper BG, Levin M. Screening Biophysical Sensors and Neurite Outgrowth Actuators in Human Induced-Pluripotent-Stem-Cell-Derived Neurons. Cells 2022; 11:cells11162470. [PMID: 36010547 PMCID: PMC9406775 DOI: 10.3390/cells11162470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/26/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
All living cells maintain a charge distribution across their cell membrane (membrane potential) by carefully controlled ion fluxes. These bioelectric signals regulate cell behavior (such as migration, proliferation, differentiation) as well as higher-level tissue and organ patterning. Thus, voltage gradients represent an important parameter for diagnostics as well as a promising target for therapeutic interventions in birth defects, injury, and cancer. However, despite much progress in cell and molecular biology, little is known about bioelectric states in human stem cells. Here, we present simple methods to simultaneously track ion dynamics, membrane voltage, cell morphology, and cell activity (pH and ROS), using fluorescent reporter dyes in living human neurons derived from induced neural stem cells (hiNSC). We developed and tested functional protocols for manipulating ion fluxes, membrane potential, and cell activity, and tracking neural responses to injury and reinnervation in vitro. Finally, using morphology sensor, we tested and quantified the ability of physiological actuators (neurotransmitters and pH) to manipulate nerve repair and reinnervation. These methods are not specific to a particular cell type and should be broadly applicable to the study of bioelectrical controls across a wide range of combinations of models and endpoints.
Collapse
Affiliation(s)
- Vaibhav P. Pai
- Allen Discovery Center at Tufts University, Medford, MA 02155, USA
| | - Ben G. Cooper
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Michael Levin
- Allen Discovery Center at Tufts University, Medford, MA 02155, USA
- Correspondence:
| |
Collapse
|
30
|
Zhang WT, Chao THH, Yang Y, Wang TW, Lee SH, Oyarzabal EA, Zhou J, Nonneman R, Pegard NC, Zhu H, Cui G, Shih YYI. Spectral fiber photometry derives hemoglobin concentration changes for accurate measurement of fluorescent sensor activity. CELL REPORTS METHODS 2022; 2:100243. [PMID: 35880016 PMCID: PMC9308135 DOI: 10.1016/j.crmeth.2022.100243] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 04/08/2022] [Accepted: 06/08/2022] [Indexed: 12/22/2022]
Abstract
Fiber photometry is an emerging technique for recording fluorescent sensor activity in the brain. However, significant hemoglobin absorption artifacts in fiber photometry data may be misinterpreted as sensor activity changes. Because hemoglobin exists widely in the brain, and its concentration varies temporally, such artifacts could impede the accuracy of photometry recordings. Here we present use of spectral photometry and computational methods to quantify photon absorption effects by using activity-independent fluorescence signals, which can be used to derive oxy- and deoxy-hemoglobin concentration changes. Although these changes are often temporally delayed compared with the fast-responding fluorescence spikes, we found that erroneous interpretation may occur when examining pharmacology-induced sustained changes and that sometimes hemoglobin absorption could flip the GCaMP signal polarity. We provide hemoglobin-based correction methods to restore fluorescence signals and compare our results with other commonly used approaches. We also demonstrated the utility of spectral fiber photometry for delineating regional differences in hemodynamic response functions.
Collapse
Affiliation(s)
- Wei-Ting Zhang
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Tzu-Hao Harry Chao
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yue Yang
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Tzu-Wen Wang
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sung-Ho Lee
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Esteban A. Oyarzabal
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jingheng Zhou
- In Vivo Neurobiology Group, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC 27709, USA
| | - Randy Nonneman
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Nicolas C. Pegard
- Department of Applied Physical Sciences, UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hongtu Zhu
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Guohong Cui
- In Vivo Neurobiology Group, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC 27709, USA
| | - Yen-Yu Ian Shih
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
31
|
Molecular Mechanisms of Epilepsy: The Role of the Chloride Transporter KCC2. J Mol Neurosci 2022; 72:1500-1515. [PMID: 35819636 DOI: 10.1007/s12031-022-02041-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 06/07/2022] [Indexed: 10/17/2022]
Abstract
Epilepsy is a neurological disease characterized by abnormal or synchronous brain activity causing seizures, which may produce convulsions, minor physical signs, or a combination of symptoms. These disorders affect approximately 65 million people worldwide, from all ages and genders. Seizures apart, epileptic patients present a high risk to develop neuropsychological comorbidities such as cognitive deficits, emotional disturbance, and psychiatric disorders, which severely impair quality of life. Currently, the treatment for epilepsy includes the administration of drugs or surgery, but about 30% of the patients treated with antiepileptic drugs develop time-dependent pharmacoresistence. Therefore, further investigation about epilepsy and its causes is needed to find new pharmacological targets and innovative therapeutic strategies. Pharmacoresistance is associated to changes in neuronal plasticity and alterations of GABAA receptor-mediated neurotransmission. The downregulation of GABA inhibitory activity may arise from a positive shift in GABAA receptor reversal potential, due to an alteration in chloride homeostasis. In this paper, we review the contribution of K+-Cl--cotransporter (KCC2) to the alterations in the Cl- gradient observed in epileptic condition, and how these alterations are coupled to the increase in the excitability.
Collapse
|
32
|
Xue D, Wei C, Zhou Y, Wang K, Zhou Y, Chen C, Li Y, Sheng L, Lu B, Zhu Z, Cai W, Ning X, Li S, Qi T, Pi J, Lin S, Yan G, Huang Y, Yin W. TRIOL Inhibits Rapid Intracellular Acidification and Cerebral Ischemic Injury: The Role of Glutamate in Neuronal Metabolic Reprogramming. ACS Chem Neurosci 2022; 13:2110-2121. [PMID: 35770894 DOI: 10.1021/acschemneuro.2c00119] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
As one of the key injury incidents, tissue acidosis in the brain occurs very quickly within several minutes upon the onset of ischemic stroke. Glutamate, an excitatory amino acid inducing neuronal excitotoxicity, has been reported to trigger the decrease in neuronal intracellular pH (pHi) via modulating proton-related membrane transporters. However, there remains a lack of clarity on the possible role of glutamate in neuronal acidosis via regulating metabolism. Here, we show that 200 μM glutamate treatment quickly promotes glycolysis and inhibits mitochondrial oxidative phosphorylation of primary cultured neurons within 15 min, leading to significant cytosolic lactate accumulation, which contributes to the rapid intracellular acidification and neuronal injury. The reprogramming of neuronal metabolism by glutamate is dependent on adenosine monophosphate-activated protein kinase (AMPK) signaling since the inhibition of AMPK activation by its selective inhibitor compound C significantly reverses these deleterious events in vitro. Moreover, 5α-androst-3β,5α,6β-TRIOL (TRIOL), a neuroprotectant we previously reported, can also remarkably reverse intracellular acidification and alleviate neuronal injury through the inhibition of AMPK signaling. Furthermore, TRIOL remarkably reduced the infarct volume and attenuated neurologic impairment in acute ischemic stroke models of middle cerebral artery occlusion in vivo. In summary, we reveal a novel role of glutamate in rapid intracellular acidification injury resulting from glutamate-induced lactate accumulation through AMPK-mediated neuronal reprogramming. Moreover, inhibition of the quick drop in neuronal pHi by TRIOL significantly reduces the cerebral damages, suggesting that it is a promising drug candidate for ischemic stroke.
Collapse
Affiliation(s)
- DongDong Xue
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - CaiLv Wei
- Department of Molecular Biology and Biochemistry, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - YueHan Zhou
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Kai Wang
- University College London, London WC1E 6BT, U.K
| | - YuWei Zhou
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Chen Chen
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yuan Li
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - LongXiang Sheng
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - BingZheng Lu
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Zhu Zhu
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Wei Cai
- Department of Molecular Biology and Biochemistry, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - XinPeng Ning
- Department of Molecular Biology and Biochemistry, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - ShengLong Li
- Department of Molecular Biology and Biochemistry, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - TianYu Qi
- Department of Molecular Biology and Biochemistry, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - JiaKai Pi
- Guangzhou Foreign Language School, Guangzhou 511400, China
| | - SuiZhen Lin
- Guangzhou Cellprotek Pharmaceutical Co., Ltd., Guangzhou 510663, China
| | - GuangMei Yan
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - YiJun Huang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Wei Yin
- Department of Molecular Biology and Biochemistry, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| |
Collapse
|
33
|
Jin X, Zhou M, Chen S, Li D, Cao X, Liu B. Effects of pH alterations on stress- and aging-induced protein phase separation. Cell Mol Life Sci 2022; 79:380. [PMID: 35750966 PMCID: PMC9232405 DOI: 10.1007/s00018-022-04393-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/26/2022] [Accepted: 05/21/2022] [Indexed: 01/18/2023]
Abstract
Upon stress challenges, proteins/RNAs undergo liquid–liquid phase separation (LLPS) to fine-tune cell physiology and metabolism to help cells adapt to adverse environments. The formation of LLPS has been recently linked with intracellular pH, and maintaining proper intracellular pH homeostasis is known to be essential for the survival of organisms. However, organisms are constantly exposed to diverse stresses, which are accompanied by alterations in the intracellular pH. Aging processes and human diseases are also intimately linked with intracellular pH alterations. In this review, we summarize stress-, aging-, and cancer-associated pH changes together with the mechanisms by which cells regulate cytosolic pH homeostasis. How critical cell components undergo LLPS in response to pH alterations is also discussed, along with the functional roles of intracellular pH fluctuation in the regulation of LLPS. Further studies investigating the interplay of pH with other stressors in LLPS regulation and identifying protein responses to different pH levels will provide an in-depth understanding of the mechanisms underlying pH-driven LLPS in cell adaptation. Moreover, deciphering aging and disease-associated pH changes that influence LLPS condensate formation could lead to a deeper understanding of the functional roles of biomolecular condensates in aging and aging-related diseases.
Collapse
Affiliation(s)
- Xuejiao Jin
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Lin'an, Hangzhou, 311300, China
| | - Min Zhou
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Lin'an, Hangzhou, 311300, China
| | - Shuxin Chen
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Lin'an, Hangzhou, 311300, China
| | - Danqi Li
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Lin'an, Hangzhou, 311300, China
| | - Xiuling Cao
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Lin'an, Hangzhou, 311300, China.
| | - Beidong Liu
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Lin'an, Hangzhou, 311300, China. .,Department of Chemistry and Molecular Biology, University of Gothenburg, Medicinaregatan 9C, 413 90, Goteborg, Sweden. .,Center for Large-Scale Cell-Based Screening, Faculty of Science, University of Gothenburg, Medicinaregatan 9C, 413 90, Goteborg, Sweden.
| |
Collapse
|
34
|
Batool Z, Wang M, Chen J, Ma M, Chen F. Regulation of physiological pH and consumption of potential food ingredients for maintaining homeostasis and metabolic function: An overview. FOOD REVIEWS INTERNATIONAL 2022. [DOI: 10.1080/87559129.2022.2062379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Zahra Batool
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen, China
- Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, China
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Mingfu Wang
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen, China
- Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, China
| | - Jiehua Chen
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen, China
- Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, China
| | - Meihu Ma
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Feng Chen
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen, China
- Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, China
| |
Collapse
|
35
|
Bose K, Maity A, Ngo KH, Vandana JJ, Shneider NA, Phan AT. Formation of RNA G-wires by G4C2 repeats associated with ALS and FTD. Biochem Biophys Res Commun 2022; 610:113-118. [DOI: 10.1016/j.bbrc.2022.03.162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 03/31/2022] [Indexed: 11/29/2022]
|
36
|
Harguindey S, Alfarouk K, Polo Orozco J, Reshkin SJ, Devesa J. Hydrogen Ion Dynamics as the Fundamental Link between Neurodegenerative Diseases and Cancer: Its Application to the Therapeutics of Neurodegenerative Diseases with Special Emphasis on Multiple Sclerosis. Int J Mol Sci 2022; 23:ijms23052454. [PMID: 35269597 PMCID: PMC8910484 DOI: 10.3390/ijms23052454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 02/17/2022] [Accepted: 02/21/2022] [Indexed: 02/01/2023] Open
Abstract
The pH-related metabolic paradigm has rapidly grown in cancer research and treatment. In this contribution, this recent oncological perspective has been laterally assessed for the first time in order to integrate neurodegeneration within the energetics of the cancer acid-base conceptual frame. At all levels of study (molecular, biochemical, metabolic, and clinical), the intimate nature of both processes appears to consist of opposite mechanisms occurring at the far ends of a physiopathological intracellular pH/extracellular pH (pHi/pHe) spectrum. This wide-ranging original approach now permits an increase in our understanding of these opposite processes, cancer and neurodegeneration, and, as a consequence, allows us to propose new avenues of treatment based upon the intracellular and microenvironmental hydrogen ion dynamics regulating and deregulating the biochemistry and metabolism of both cancer and neural cells. Under the same perspective, the etiopathogenesis and special characteristics of multiple sclerosis (MS) is an excellent model for the study of neurodegenerative diseases and, utilizing this pioneering approach, we find that MS appears to be a metabolic disease even before an autoimmune one. Furthermore, within this paradigm, several important aspects of MS, from mitochondrial failure to microbiota functional abnormalities, are analyzed in depth. Finally, and for the first time, a new and integrated model of treatment for MS can now be advanced.
Collapse
Affiliation(s)
- Salvador Harguindey
- Division of Oncology, Institute of Clinical Biology and Metabolism, 01004 Vitoria, Spain;
- Correspondence: ; Tel.: +34-629-047-141
| | - Khalid Alfarouk
- Institute of Endemic Diseases, University of Khartoum, Khartoum 11111, Sudan;
| | - Julián Polo Orozco
- Division of Oncology, Institute of Clinical Biology and Metabolism, 01004 Vitoria, Spain;
| | - Stephan J Reshkin
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, 70125 Bari, Italy;
| | - Jesús Devesa
- Scientific Direction, Foltra Medical Centre, 15886 Teo, Spain;
| |
Collapse
|
37
|
Ponomareva D, Petukhova E, Bregestovski P. Simultaneous Monitoring of pH and Chloride (Cl -) in Brain Slices of Transgenic Mice. Int J Mol Sci 2021; 22:13601. [PMID: 34948398 PMCID: PMC8708776 DOI: 10.3390/ijms222413601] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 11/16/2022] Open
Abstract
Optosensorics is the direction of research possessing the possibility of non-invasive monitoring of the concentration of intracellular ions or activity of intracellular components using specific biosensors. In recent years, genetically encoded proteins have been used as effective optosensory means. These probes possess fluorophore groups capable of changing fluorescence when interacting with certain ions or molecules. For monitoring of intracellular concentrations of chloride ([Cl-]i) and hydrogen ([H+] i) the construct, called ClopHensor, which consists of a H+- and Cl--sensitive variant of the enhanced green fluorescent protein (E2GFP) fused with a monomeric red fluorescent protein (mDsRed) has been proposed. We recently developed a line of transgenic mice expressing ClopHensor in neurons and obtained the map of its expression in different areas of the brain. The purpose of this study was to examine the effectiveness of transgenic mice expressing ClopHensor for estimation of [H+]i and [Cl-]i concentrations in neurons of brain slices. We performed simultaneous monitoring of [H+]i and [Cl-]i under different experimental conditions including changing of external concentrations of ions (Ca2+, Cl-, K+, Na+) and synaptic stimulation of Shaffer's collaterals of hippocampal slices. The results obtained illuminate different pathways of regulation of Cl- and pH equilibrium in neurons and demonstrate that transgenic mice expressing ClopHensor represent a reliable tool for non-invasive simultaneous monitoring of intracellular Cl- and pH.
Collapse
Affiliation(s)
- Daria Ponomareva
- Institut de Neurosciences des Systèmes, Aix-Marseille University, INSERM, INS, 13005 Marseille, France;
- Institute of Neurosciences, Kazan State Medical University, 420111 Kazan, Russia;
- Department of Normal Physiology, Kazan State Medical University, 420111 Kazan, Russia
| | - Elena Petukhova
- Institute of Neurosciences, Kazan State Medical University, 420111 Kazan, Russia;
- Department of Normal Physiology, Kazan State Medical University, 420111 Kazan, Russia
| | - Piotr Bregestovski
- Institut de Neurosciences des Systèmes, Aix-Marseille University, INSERM, INS, 13005 Marseille, France;
- Institute of Neurosciences, Kazan State Medical University, 420111 Kazan, Russia;
- Department of Normal Physiology, Kazan State Medical University, 420111 Kazan, Russia
| |
Collapse
|
38
|
Kilb W. When Are Depolarizing GABAergic Responses Excitatory? Front Mol Neurosci 2021; 14:747835. [PMID: 34899178 PMCID: PMC8651619 DOI: 10.3389/fnmol.2021.747835] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/28/2021] [Indexed: 11/13/2022] Open
Abstract
The membrane responses upon activation of GABA(A) receptors critically depend on the intracellular Cl− concentration ([Cl−]i), which is maintained by a set of transmembrane transporters for Cl−. During neuronal development, but also under several pathophysiological conditions, the prevailing expression of the Cl− loader NKCC1 and the low expression of the Cl− extruder KCC2 causes elevated [Cl−]i, which result in depolarizing GABAergic membrane responses. However, depolarizing GABAergic responses are not necessarily excitatory, as GABA(A) receptors also reduces the input resistance of neurons and thereby shunt excitatory inputs. To summarize our knowledge on the effect of depolarizing GABA responses on neuronal excitability, this review discusses theoretical considerations and experimental studies illustrating the relation between GABA conductances, GABA reversal potential and neuronal excitability. In addition, evidences for the complex spatiotemporal interaction between depolarizing GABAergic and glutamatergic inputs are described. Moreover, mechanisms that influence [Cl−]i beyond the expression of Cl− transporters are presented. And finally, several in vitro and in vivo studies that directly investigated whether GABA mediates excitation or inhibition during early developmental stages are summarized. In summary, these theoretical considerations and experimental evidences suggest that GABA can act as inhibitory neurotransmitter even under conditions that maintain substantial depolarizing membrane responses.
Collapse
Affiliation(s)
- Werner Kilb
- Institute of Physiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
39
|
Dynamics of cortical oxygenation during immediate adaptation to extrauterine life. Sci Rep 2021; 11:22041. [PMID: 34764396 PMCID: PMC8586152 DOI: 10.1038/s41598-021-01674-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 11/02/2021] [Indexed: 11/09/2022] Open
Abstract
The neonatal transition involves physiological modifications as a consequence of the complexity of the perinatal period. Various strategies can be used to attain the same level of postnatal cerebral oxygenation, depending on the status of the infant at birth. We evaluated such strategies by recording 20 full-term newborns by near-infrared spectroscopy during the first 10 min of life. The acid–base status at birth revealed two clustered profiles of cerebral oxygenation dynamics. Lower pH and base excess and higher lactate levels were associated with more rapid attainment of the 95% maximal tissue oxygenation index value. These results suggest that metabolic mechanisms drive initial cerebral oxygenation dynamics during this critical period. These results confirm the capacity of newborns to develop multiple strategies to protect the brain.
Collapse
|
40
|
Feghhi T, Hernandez RX, Stawarski M, Thomas CI, Kamasawa N, Lau AWC, Macleod GT. Computational modeling predicts ephemeral acidic microdomains in the glutamatergic synaptic cleft. Biophys J 2021; 120:5575-5591. [PMID: 34774503 DOI: 10.1016/j.bpj.2021.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/21/2021] [Accepted: 11/05/2021] [Indexed: 10/19/2022] Open
Abstract
At chemical synapses, synaptic vesicles release their acidic contents into the cleft, leading to the expectation that the cleft should acidify. However, fluorescent pH probes targeted to the cleft of conventional glutamatergic synapses in both fruit flies and mice reveal cleft alkalinization rather than acidification. Here, using a reaction-diffusion scheme, we modeled pH dynamics at the Drosophila neuromuscular junction as glutamate, ATP, and protons (H+) were released into the cleft. The model incorporates bicarbonate and phosphate buffering systems as well as plasma membrane calcium-ATPase activity and predicts substantial cleft acidification but only for fractions of a millisecond after neurotransmitter release. Thereafter, the cleft rapidly alkalinizes and remains alkaline for over 100 ms because the plasma membrane calcium-ATPase removes H+ from the cleft in exchange for calcium ions from adjacent pre- and postsynaptic compartments, thus recapitulating the empirical data. The extent of synaptic vesicle loading and time course of exocytosis have little influence on the magnitude of acidification. Phosphate but not bicarbonate buffering is effective at suppressing the magnitude and time course of the acid spike, whereas both buffering systems are effective at suppressing cleft alkalinization. The small volume of the cleft levies a powerful influence on the magnitude of alkalinization and its time course. Structural features that open the cleft to adjacent spaces appear to be essential for alleviating the extent of pH transients accompanying neurotransmission.
Collapse
Affiliation(s)
- Touhid Feghhi
- Department of Physics, College of Science, Florida Atlantic University, Boca Raton, Florida
| | - Roberto X Hernandez
- Integrative Biology & Neuroscience Graduate Program, Florida Atlantic University, Boca Raton, Florida; International Max Planck Research School for Brain and Behavior, Jupiter, Florida; Jupiter Life Sciences Initiative, Florida Atlantic University, Jupiter, Florida
| | - Michal Stawarski
- Wilkes Honors College, Florida Atlantic University, Jupiter, Florida
| | - Connon I Thomas
- Electron Microscopy Core Facility, Max Planck Florida Institute, Jupiter, Florida
| | - Naomi Kamasawa
- Electron Microscopy Core Facility, Max Planck Florida Institute, Jupiter, Florida
| | - A W C Lau
- Department of Physics, College of Science, Florida Atlantic University, Boca Raton, Florida
| | - Gregory T Macleod
- Jupiter Life Sciences Initiative, Florida Atlantic University, Jupiter, Florida; Wilkes Honors College, Florida Atlantic University, Jupiter, Florida; Brain Institute, Florida Atlantic University, Jupiter, Florida; Institute for Human Health & Disease Intervention, Florida Atlantic University, Jupiter, Florida.
| |
Collapse
|
41
|
Mokrushin AA. Optimization of the Acidic–Alkaline Composition of the Incubation Medium for Long-Term and Reversible Cryopreservation of Brain Slices of Nonhibernating Animals. Biophysics (Nagoya-shi) 2021. [DOI: 10.1134/s0006350921050134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
42
|
Zhao W, Zhang L, Ermilov LG, Colmenares Aguilar MG, Linden DR, Eisenman ST, Romero MF, Farrugia G, Sha L, Gibbons SJ. Bicarbonate ion transport by the electrogenic Na + /HCO 3- cotransporter, NBCe1, is required for normal electrical slow-wave activity in mouse small intestine. Neurogastroenterol Motil 2021; 33:e14149. [PMID: 33837991 PMCID: PMC8485339 DOI: 10.1111/nmo.14149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 02/26/2021] [Accepted: 03/21/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Normal gastrointestinal motility depends on electrical slow-wave activity generated by interstitial cells of Cajal (ICC) in the tunica muscularis of the gastrointestinal tract. A requirement for HCO3- in extracellular solutions used to record slow waves indicates a role for HCO3- transport in ICC pacemaking. The Slc4a4 gene transcript encoding the electrogenic Na+ /HCO3- cotransporter, NBCe1, is enriched in mouse small intestinal myenteric region ICC (ICC-MY) that generate slow waves. This study aimed to determine how extracellular HCO3- concentrations affect electrical activity in mouse small intestine and to determine the contribution of NBCe1 activity to these effects. METHODS Immunohistochemistry and sharp electrode electrical recordings were used. KEY RESULTS The NBCe1 immunoreactivity was localized to ICC-MY of the tunica muscularis. In sharp electrode electrical recordings, removal of HCO3- from extracellular solutions caused significant, reversible, depolarization of the smooth muscle and a reduction in slow-wave amplitude and frequency. In 100 mM HCO3- , the muscle hyperpolarized and slow wave amplitude and frequency increased. The effects of replacing extracellular Na+ with Li+ , an ion that does not support NBCe1 activity, were similar to, but larger than, the effects of removing HCO3- . There were no additional changes to electrical activity when HCO3- was removed from Li+ containing solutions. The Na+ /HCO3- cotransport inhibitor, S-0859 (30µM) significantly reduced the effect of removing HCO3- on electrical activity. CONCLUSIONS & INFERENCES These studies demonstrate a major role for Na+ /HCO3- cotransport by NBCe1 in electrical activity of mouse small intestine and indicated that regulation of intracellular acid:base homeostasis contributes to generation of normal pacemaker activity in the gastrointestinal tract.
Collapse
Affiliation(s)
- Wenchang Zhao
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Rochester, Minnesota, USA.,Physiology and Biomedical Engineering, Rochester, Minnesota, USA.,Neuroendocrine Pharmacology, China Medical University, Shenyang, Liaoning Province, P. R. China
| | - Liwen Zhang
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Rochester, Minnesota, USA.,Physiology and Biomedical Engineering, Rochester, Minnesota, USA.,Neuroendocrine Pharmacology, China Medical University, Shenyang, Liaoning Province, P. R. China
| | - Leonid G. Ermilov
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Rochester, Minnesota, USA.,Physiology and Biomedical Engineering, Rochester, Minnesota, USA
| | - Maria Gabriela Colmenares Aguilar
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Rochester, Minnesota, USA.,Physiology and Biomedical Engineering, Rochester, Minnesota, USA
| | - David R. Linden
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Rochester, Minnesota, USA.,Physiology and Biomedical Engineering, Rochester, Minnesota, USA
| | - Seth T. Eisenman
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Rochester, Minnesota, USA.,Physiology and Biomedical Engineering, Rochester, Minnesota, USA
| | - Michael F. Romero
- Physiology and Biomedical Engineering, Rochester, Minnesota, USA.,Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Gianrico Farrugia
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Rochester, Minnesota, USA.,Physiology and Biomedical Engineering, Rochester, Minnesota, USA
| | - Lei Sha
- Neuroendocrine Pharmacology, China Medical University, Shenyang, Liaoning Province, P. R. China.,Corresponding Authors: Simon J Gibbons, Ph.D., Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, 55905, USA. . Telephone: +1 507 284 9652, Lei Sha, M.D., China Medical University, 77 Pu He Road, Shenbei New District, Shenyang, Liaoning Province, P. R. China, 110122, , . Telephone: +86 18900911003
| | - Simon J. Gibbons
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Rochester, Minnesota, USA.,Physiology and Biomedical Engineering, Rochester, Minnesota, USA.,Corresponding Authors: Simon J Gibbons, Ph.D., Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, 55905, USA. . Telephone: +1 507 284 9652, Lei Sha, M.D., China Medical University, 77 Pu He Road, Shenbei New District, Shenyang, Liaoning Province, P. R. China, 110122, , . Telephone: +86 18900911003
| |
Collapse
|
43
|
Decreased Brain pH and Pathophysiology in Schizophrenia. Int J Mol Sci 2021; 22:ijms22168358. [PMID: 34445065 PMCID: PMC8395078 DOI: 10.3390/ijms22168358] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/30/2021] [Accepted: 07/30/2021] [Indexed: 12/26/2022] Open
Abstract
Postmortem studies reveal that the brain pH in schizophrenia patients is lower than normal. The exact cause of this low pH is unclear, but increased lactate levels due to abnormal energy metabolism appear to be involved. Schizophrenia patients display distinct changes in mitochondria number, morphology, and function, and such changes promote anaerobic glycolysis, elevating lactate levels. pH can affect neuronal activity as H+ binds to numerous proteins in the nervous system and alters the structure and function of the bound proteins. There is growing evidence of pH change associated with cognition, emotion, and psychotic behaviors. Brain has delicate pH regulatory mechanisms to maintain normal pH in neurons/glia and extracellular fluid, and a change in these mechanisms can affect, or be affected by, neuronal activities associated with schizophrenia. In this review, we discuss the current understanding of the cause and effect of decreased brain pH in schizophrenia based on postmortem human brains, animal models, and cellular studies. The topic includes the factors causing decreased brain pH in schizophrenia, mitochondria dysfunction leading to altered energy metabolism, and pH effects on the pathophysiology of schizophrenia. We also review the acid/base transporters regulating pH in the nervous system and discuss the potential contribution of the major transporters, sodium hydrogen exchangers (NHEs), and sodium-coupled bicarbonate transporters (NCBTs), to schizophrenia.
Collapse
|
44
|
Chebotarev AS, Pochechuev MS, Lanin AA, Kelmanson IV, Kotova DA, Fetisova ES, Panova AS, Bilan DS, Fedotov AB, Belousov VV, Zheltikov AM. Enhanced-contrast two-photon optogenetic pH sensing and pH-resolved brain imaging. JOURNAL OF BIOPHOTONICS 2021; 14:e202000301. [PMID: 33205577 DOI: 10.1002/jbio.202000301] [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: 07/22/2020] [Revised: 10/29/2020] [Accepted: 11/16/2020] [Indexed: 06/11/2023]
Abstract
We present experiments on cell cultures and brain slices that demonstrate two-photon optogenetic pH sensing and pH-resolved brain imaging using a laser driver whose spectrum is carefully tailored to provide the maximum contrast of a ratiometric two-photon fluorescence readout from a high-brightness genetically encoded yellow-fluorescent-protein-based sensor, SypHer3s. Two spectrally isolated components of this laser field are set to induce two-photon-excited fluorescence (2PEF) by driving SypHer3s through one of two excitation pathways-via either the protonated or deprotonated states of its chromophore. With the spectrum of the laser field accurately adjusted for a maximum contrast of these two 2PEF signals, the ratio of their intensities is shown to provide a remarkably broad dynamic range for pH measurements, enabling high-contrast optogenetic deep-brain pH sensing and pH-resolved 2PEF imaging within a vast class of biological systems, ranging from cell cultures to the living brain.
Collapse
Affiliation(s)
- Artem S Chebotarev
- Physics Department, International Laser Center, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Matvei S Pochechuev
- Physics Department, International Laser Center, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Aleksandr A Lanin
- Physics Department, International Laser Center, M.V. Lomonosov Moscow State University, Moscow, Russia
- Russian Quantum Center, Moscow, Russia
- Kazan Quantum Center, A.N. Tupolev Kazan National Research Technical University, Kazan, Russia
| | - Ilya V Kelmanson
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - Daria A Kotova
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Elena S Fetisova
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Anastasiya S Panova
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Dmitry S Bilan
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - Andrei B Fedotov
- Physics Department, International Laser Center, M.V. Lomonosov Moscow State University, Moscow, Russia
- Russian Quantum Center, Moscow, Russia
- Kazan Quantum Center, A.N. Tupolev Kazan National Research Technical University, Kazan, Russia
- National University of Science and Technology "MISiS,", Moscow, Russia
| | - Vsevolod V Belousov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies of the Federal Medical Biological Agency, Moscow, Russia
| | - Aleksei M Zheltikov
- Physics Department, International Laser Center, M.V. Lomonosov Moscow State University, Moscow, Russia
- Russian Quantum Center, Moscow, Russia
- Kazan Quantum Center, A.N. Tupolev Kazan National Research Technical University, Kazan, Russia
- Department of Physics and Astronomy, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
45
|
Intricacies of GABA A Receptor Function: The Critical Role of the β3 Subunit in Norm and Pathology. Int J Mol Sci 2021; 22:ijms22031457. [PMID: 33535681 PMCID: PMC7867123 DOI: 10.3390/ijms22031457] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 12/23/2022] Open
Abstract
Neuronal intracellular chloride ([Cl−]i) is a key determinant in γ-aminobutyric acid type A (GABA)ergic signaling. γ-Aminobutyric acid type A receptors (GABAARs) mediate both inhibitory and excitatory neurotransmission, as the passive fluxes of Cl− and HCO3− via pores can be reversed by changes in the transmembrane concentration gradient of Cl−. The cation–chloride co-transporters (CCCs) are the primary systems for maintaining [Cl−]i homeostasis. However, despite extensive electrophysiological data obtained in vitro that are supported by a wide range of molecular biological studies on the expression patterns and properties of CCCs, the presence of ontogenetic changes in [Cl−]i—along with the consequent shift in GABA reversal potential—remain a subject of debate. Recent studies showed that the β3 subunit possesses properties of the P-type ATPase that participates in the ATP-consuming movement of Cl− via the receptor. Moreover, row studies have demonstrated that the β3 subunit is a key player in GABAAR performance and in the appearance of serious neurological disorders. In this review, we discuss the properties and driving forces of CCCs and Cl−, HCO3−ATPase in the maintenance of [Cl−]i homeostasis after changes in upcoming GABAAR function. Moreover, we discuss the contribution of the β3 subunit in the manifestation of epilepsy, autism, and other syndromes.
Collapse
|
46
|
Hwang SM, Lee JY, Park CK, Kim YH. The Role of TRP Channels and PMCA in Brain Disorders: Intracellular Calcium and pH Homeostasis. Front Cell Dev Biol 2021; 9:584388. [PMID: 33585474 PMCID: PMC7876282 DOI: 10.3389/fcell.2021.584388] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 01/06/2021] [Indexed: 12/19/2022] Open
Abstract
Brain disorders include neurodegenerative diseases (NDs) with different conditions that primarily affect the neurons and glia in the brain. However, the risk factors and pathophysiological mechanisms of NDs have not been fully elucidated. Homeostasis of intracellular Ca2+ concentration and intracellular pH (pHi) is crucial for cell function. The regulatory processes of these ionic mechanisms may be absent or excessive in pathological conditions, leading to a loss of cell death in distinct regions of ND patients. Herein, we review the potential involvement of transient receptor potential (TRP) channels in NDs, where disrupted Ca2+ homeostasis leads to cell death. The capability of TRP channels to restore or excite the cell through Ca2+ regulation depending on the level of plasma membrane Ca2+ ATPase (PMCA) activity is discussed in detail. As PMCA simultaneously affects intracellular Ca2+ regulation as well as pHi, TRP channels and PMCA thus play vital roles in modulating ionic homeostasis in various cell types or specific regions of the brain where the TRP channels and PMCA are expressed. For this reason, the dysfunction of TRP channels and/or PMCA under pathological conditions disrupts neuronal homeostasis due to abnormal Ca2+ and pH levels in the brain, resulting in various NDs. This review addresses the function of TRP channels and PMCA in controlling intracellular Ca2+ and pH, which may provide novel targets for treating NDs.
Collapse
Affiliation(s)
- Sung-Min Hwang
- Gachon Pain Center, Department of Physiology, Gachon University College of Medicine, Incheon, South Korea
| | - Ji Yeon Lee
- Gil Medical Center, Department of Anesthesiology and Pain Medicine, Gachon University, Incheon, South Korea
| | - Chul-Kyu Park
- Gachon Pain Center, Department of Physiology, Gachon University College of Medicine, Incheon, South Korea
| | - Yong Ho Kim
- Gachon Pain Center, Department of Physiology, Gachon University College of Medicine, Incheon, South Korea
| |
Collapse
|
47
|
Esmaile SC, Bezerra KS, de Oliveira Campos DM, da Silva MK, Neto JXL, Manzoni V, Fulco UL, Oliveira JIN. Quantum binding energy features of the drug olmesartan bound to angiotensin type-1 receptors in the therapeutics of stroke. NEW J CHEM 2021. [DOI: 10.1039/d1nj03975j] [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 investigated the binding energies of 105 residues within a 10 Å pocket radius, predicted the energetic relevance of olmesartan regions, and the influence of individual protein segments on OLM -AT1 binding.
Collapse
Affiliation(s)
- Stephany Campanelli Esmaile
- Departamento de Biofísica e Farmacologia, Universidade Federal do Rio Grande do Norte, 59072-970, Natal, RN, Brazil
| | - Katyanna Sales Bezerra
- Departamento de Biofísica e Farmacologia, Universidade Federal do Rio Grande do Norte, 59072-970, Natal, RN, Brazil
| | | | - Maria Karolaynne da Silva
- Departamento de Biofísica e Farmacologia, Universidade Federal do Rio Grande do Norte, 59072-970, Natal, RN, Brazil
| | - José Xavier Lima Neto
- Departamento de Biofísica e Farmacologia, Universidade Federal do Rio Grande do Norte, 59072-970, Natal, RN, Brazil
| | - Vinicius Manzoni
- Instituto de Física, Universidade Federal de Alagoas, 57072-970, Maceio, AL, Brazil
| | - Umberto Laino Fulco
- Departamento de Biofísica e Farmacologia, Universidade Federal do Rio Grande do Norte, 59072-970, Natal, RN, Brazil
| | - Jonas Ivan Nobre Oliveira
- Departamento de Biofísica e Farmacologia, Universidade Federal do Rio Grande do Norte, 59072-970, Natal, RN, Brazil
| |
Collapse
|
48
|
Li Y, Ritzel RM, He J, Cao T, Sabirzhanov B, Li H, Liu S, Wu LJ, Wu J. The voltage-gated proton channel Hv1 plays a detrimental role in contusion spinal cord injury via extracellular acidosis-mediated neuroinflammation. Brain Behav Immun 2021; 91:267-283. [PMID: 33039662 PMCID: PMC7749852 DOI: 10.1016/j.bbi.2020.10.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/27/2020] [Accepted: 10/06/2020] [Indexed: 10/23/2022] Open
Abstract
Tissue acidosis is an important secondary injury process in the pathophysiology of traumatic spinal cord injury (SCI). To date, no studies have examined the role of proton extrusion as mechanism of pathological acidosis in SCI. In the present study, we hypothesized that the phagocyte-specific proton channel Hv1 mediates hydrogen proton extrusion after SCI, contributing to increased extracellular acidosis and poor long-term outcomes. Using a contusion model of SCI in adult female mice, we demonstrated that tissue pH levels are markedly lower during the first week after SCI. Acidosis was most evident at the injury site, but also extended into proximal regions of the cervical and lumbar cord. Tissue reactive oxygen species (ROS) levels and expression of Hv1 were significantly increased during the week of injury. Hv1 was exclusively expressed in microglia within the CNS, suggesting that microglia contribute to ROS production and proton extrusion during respiratory burst. Depletion of Hv1 significantly attenuated tissue acidosis, NADPH oxidase 2 (NOX2) expression, and ROS production at 3 d post-injury. Nanostring analysis revealed decreased gene expression of neuroinflammatory and cytokine signaling markers in Hv1 knockout (KO) mice. Furthermore, Hv1 deficiency reduced microglia proliferation, leukocyte infiltration, and phagocytic oxidative burst detected by flow cytometry. Importantly, Hv1 KO mice exhibited significantly improved locomotor function and reduced histopathology. Overall, these data suggest an important role for Hv1 in regulating tissue acidosis, NOX2-mediated ROS production, and functional outcome following SCI. Thus, the Hv1 proton channel represents a potential target that may lead to novel therapeutic strategies for SCI.
Collapse
Affiliation(s)
- Yun Li
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Rodney M Ritzel
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Junyun He
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Tuoxin Cao
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Boris Sabirzhanov
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Hui Li
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Simon Liu
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA.
| | - Junfang Wu
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA; University of Maryland Center to Advance Chronic Pain Research, University of Maryland, Baltimore, MD 21201, USA.
| |
Collapse
|
49
|
Colmenares Aguilar MG, Mazzone A, Eisenman ST, Strege PR, Bernard CE, Holmes HL, Romero MF, Farrugia G, Gibbons SJ. Expression of the regulated isoform of the electrogenic Na +/HCO 3- cotransporter, NBCe1, is enriched in pacemaker interstitial cells of Cajal. Am J Physiol Gastrointest Liver Physiol 2021; 320:G93-G107. [PMID: 33112159 PMCID: PMC8112189 DOI: 10.1152/ajpgi.00255.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Interstitial cells of Cajal (ICCs) generate electrical slow waves, which are required for normal gastrointestinal motility. The mechanisms for generation of normal pacemaking are not fully understood. Normal gastrointestinal contractility- and electrical slow-wave activity depend on the presence of extracellular HCO3-. Previous transcriptional analysis identified enrichment of mRNA encoding the electrogenic Na+/HCO3- cotransporter (NBCe1) gene (Slc4a4) in pacemaker myenteric ICCs in mouse small intestine. We aimed to determine the distribution of NBCe1 protein in ICCs of the mouse gastrointestinal tract and to identify the transcripts of the Slc4a4 gene in mouse and human small intestinal tunica muscularis. We determined the distribution of NBCe1 immunoreactivity (NBCe1-IR) by immunofluorescent labeling in mouse and human tissues. In mice, NBCe1-IR was restricted to Kit-positive myenteric ICCs of the stomach and small intestine and submuscular ICCs of the large intestine, that is, the slow wave generating subset of ICCs. Other subtypes of ICCs were NBCe1-negative. Quantitative real-time PCR identified >500-fold enrichment of Slc4a4-207 and Slc4a4-208 transcripts ["IP3-receptor-binding protein released by IP3" (IRBIT)-regulated isoforms] in Kit-expressing cells isolated from KitcreERT2/+, Rpl22tm1.1Psam/Sj mice and from single GFP-positive ICCs from Kittm1Rosay mice. Human jejunal tunica muscularis ICCs were also NBCe1-positive, and SLC4A4-201 and SLC4A4-204 RNAs were >300-fold enriched relative to SLC4A4-202. In summary, NBCe1 protein expressed in ICCs with electrical pacemaker function is encoded by Slc4a4 gene transcripts that generate IRBIT-regulated isoforms of NBCe1. In conclusion, Na+/HCO3- cotransport through NBCe1 contributes to the generation of pacemaker activity in subsets of ICCs.NEW & NOTEWORTHY In this study, we show that the electrogenic Na+/HCO3- cotransporter, NBCe1/Slc4a4, is expressed in subtypes of interstitial cells of Cajal (ICCs) responsible for electrical slow wave generation throughout the mouse gastrointestinal tract and is absent in other types of ICCs. The transcripts of Slc4a4 expressed in mouse ICCs and human gastrointestinal smooth muscle are the regulated isoforms. This indicates a key role for HCO3- transport in generation of gastrointestinal motility patterns.
Collapse
Affiliation(s)
| | - Amelia Mazzone
- 1Enteric NeuroScience Program, Division of
Gastroenterology and Hepatology, Mayo Clinic,
Rochester, Minnesota
| | - Seth T. Eisenman
- 1Enteric NeuroScience Program, Division of
Gastroenterology and Hepatology, Mayo Clinic,
Rochester, Minnesota
| | - Peter R. Strege
- 1Enteric NeuroScience Program, Division of
Gastroenterology and Hepatology, Mayo Clinic,
Rochester, Minnesota
| | - Cheryl E. Bernard
- 1Enteric NeuroScience Program, Division of
Gastroenterology and Hepatology, Mayo Clinic,
Rochester, Minnesota
| | - Heather L. Holmes
- 2Physiology and Biomedical Engineering, Nephrology and
Hypertension, Mayo Clinic College of Medicine and
Science, Rochester, Minnesota
| | - Michael F. Romero
- 2Physiology and Biomedical Engineering, Nephrology and
Hypertension, Mayo Clinic College of Medicine and
Science, Rochester, Minnesota
| | - Gianrico Farrugia
- 1Enteric NeuroScience Program, Division of
Gastroenterology and Hepatology, Mayo Clinic,
Rochester, Minnesota,3Department of Physiology and Biomedical Engineering,
Mayo Clinic, Rochester, Minnesota
| | - Simon J. Gibbons
- 1Enteric NeuroScience Program, Division of
Gastroenterology and Hepatology, Mayo Clinic,
Rochester, Minnesota
| |
Collapse
|
50
|
Turaev AV, Isaakova EA, Severov VV, Bogomazova AN, Zatsepin TS, Sardushkin MV, Aralov AV, Lagarkova MA, Pozmogova GE, Varizhuk AM. Genomic DNA i-motifs as fast sensors responsive to near-physiological pH microchanges. Biosens Bioelectron 2020; 175:112864. [PMID: 33309217 DOI: 10.1016/j.bios.2020.112864] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 11/11/2020] [Accepted: 11/24/2020] [Indexed: 02/06/2023]
Abstract
We report the design of robust sensors for measuring intracellular pH, based on the native DNA i-motifs (iMs) found in neurodegeneration- or carcinogenesis-related genes. Those iMs appear to be genomic regulatory elements and might modulate transcription in response to pH stimuli. Given their intrinsic sensitivity to minor pH changes within the physiological range, such noncanonical DNA structures can be used as sensor core elements without additional modules other than fluorescent labels or quenchers. We focused on several iMs that exhibited fast folding/unfolding kinetics. Using stopped-flow techniques and FRET-melting/annealing assays, we confirmed that the rates of temperature-driven iM-ssDNA transitions correlate with the rates of the pH-driven transitions. Thus, we propose FRET-based hysteresis analysis as an express method for selecting sensors with desired kinetic characteristics. For the leading fast-response sensor, we optimized the labelling scheme and performed intracellular calibration. Unlike the commonly used small-molecule pH indicators, that sensor was transferred efficiently to cell nuclei. Considering its favourable kinetic characteristics, the sensor can be used for monitoring proton dynamics in the nucleus. These results argue that the 'genome-inspired' design is a productive approach to the development of biocompatible molecular tools.
Collapse
Affiliation(s)
- Anton V Turaev
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Ekaterina A Isaakova
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Vjacheslav V Severov
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Alexandra N Bogomazova
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, 119435, Russia; Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Timofei S Zatsepin
- Skolkovo Institute of Science and Technology, Moscow Oblast, 143026, Russia
| | - Makar V Sardushkin
- Mendeleev University of Chemical Technology of Russia, Moscow, 125047, Russia
| | - Andrey V Aralov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia
| | - Maria A Lagarkova
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, 119435, Russia; Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Galina E Pozmogova
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Anna M Varizhuk
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, 119435, Russia; Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, 119435, Russia; Engelhardt Institute of Molecular Biology, Moscow, 119991, Russia.
| |
Collapse
|