1
|
Martinez ML, Nan K, Bao Z, Bacchetti R, Yuan S, Tyler J, Guezennec XL, Bard FA, Rainero E. Novel kinase regulators of extracellular matrix internalisation identified by high-content screening modulate invasive carcinoma cell migration. PLoS Biol 2024; 22:e3002930. [PMID: 39666682 PMCID: PMC11637276 DOI: 10.1371/journal.pbio.3002930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 11/06/2024] [Indexed: 12/14/2024] Open
Abstract
The interaction between cancer cells and the extracellular matrix (ECM) plays a pivotal role in tumour progression. While the extracellular degradation of ECM proteins has been well characterised, ECM endocytosis and its impact on cancer cell progression, migration, and metastasis is poorly understood. ECM internalisation is increased in invasive breast cancer cells, suggesting it may support invasiveness. However, current high-throughput approaches mainly focus on cells grown on plastic in 2D, making it difficult to apply these to the study of ECM dynamics. Here, we developed a high-content screening assay to study ECM uptake, based on the of use automated ECM coating for the generation of highly homogeneous ECM a pH-sensitive dye to image ECM trafficking in live cells. We identified that mitogen-activated protein kinase (MAPK) family members, MAP3K1 and MAPK11 (p38β), and the protein phosphatase 2 (PP2) subunit PPP2R1A were required for the internalisation of ECM-bound α2β1 integrin. Mechanistically, we show that down-regulation of the sodium/proton exchanger 1 (NHE1), an established macropinocytosis regulator and a target of p38, mediated ECM macropinocytosis. Moreover, disruption of α2 integrin, MAP3K1, MAPK11, PPP2R1A, and NHE1-mediated ECM internalisation significantly impaired cancer cell migration and invasion in 2D and 3D culture systems. Of note, integrin-bound ECM was targeted for lysosomal degradation, which was required for cell migration on cell-derived matrices. Finally, α2β1 integrin and MAP3K1 expression were significantly up-regulated in pancreatic tumours and correlated with poor prognosis in pancreatic cancer patients. Strikingly, MAP3K1, MAPK11, PPP2R1A, and α2 integrin expression were higher in chemotherapy-resistant tumours in breast cancer patients. Our results identified the α2β1 integrin/p38 signalling axis as a novel regulator of ECM endocytosis, which drives invasive migration and tumour progression, demonstrating that our high-content screening approach has the capability of identifying novel regulators of cancer cell invasion.
Collapse
Affiliation(s)
- Montserrat Llanses Martinez
- School of Biosciences, University of Sheffield, Western Bank, Sheffield, United Kingdom
- Institute of Molecular and Cell Biology, Singapore, Singapore
| | - Keqian Nan
- School of Biosciences, University of Sheffield, Western Bank, Sheffield, United Kingdom
| | - Zhe Bao
- School of Biosciences, University of Sheffield, Western Bank, Sheffield, United Kingdom
| | - Rachele Bacchetti
- School of Biosciences, University of Sheffield, Western Bank, Sheffield, United Kingdom
| | - Shengnan Yuan
- School of Biosciences, University of Sheffield, Western Bank, Sheffield, United Kingdom
| | - Joe Tyler
- School of Biosciences, University of Sheffield, Western Bank, Sheffield, United Kingdom
| | | | - Frederic A. Bard
- Institute of Molecular and Cell Biology, Singapore, Singapore
- Centre de Recherche en Cancérologie de Marseille, CRCM, Marseille, France
| | - Elena Rainero
- School of Biosciences, University of Sheffield, Western Bank, Sheffield, United Kingdom
| |
Collapse
|
2
|
Kratochvil HT, Watkins LC, Mravic M, Thomaston JL, Nicoludis JM, Somberg NH, Liu L, Hong M, Voth GA, DeGrado WF. Transient water wires mediate selective proton transport in designed channel proteins. Nat Chem 2023; 15:1012-1021. [PMID: 37308712 PMCID: PMC10475958 DOI: 10.1038/s41557-023-01210-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 04/19/2023] [Indexed: 06/14/2023]
Abstract
Selective proton transport through proteins is essential for forming and using proton gradients in cells. Protons are conducted along hydrogen-bonded 'wires' of water molecules and polar side chains, which, somewhat surprisingly, are often interrupted by dry apolar stretches in the conduction pathways, inferred from static protein structures. Here we hypothesize that protons are conducted through such dry spots by forming transient water wires, often highly correlated with the presence of the excess protons in the water wire. To test this hypothesis, we performed molecular dynamics simulations to design transmembrane channels with stable water pockets interspersed by apolar segments capable of forming flickering water wires. The minimalist designed channels conduct protons at rates similar to viral proton channels, and they are at least 106-fold more selective for H+ over Na+. These studies inform the mechanisms of biological proton conduction and the principles for engineering proton-conductive materials.
Collapse
Affiliation(s)
- Huong T Kratochvil
- Department of Pharmaceutical Chemistry, University of California-San Francisco, San Francisco, CA, USA.
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Laura C Watkins
- Department of Chemistry, Chicago Center for Theoretical Chemistry, Institute for Biophysical Dynamics and James Franck Institute, The University of Chicago, Chicago, IL, USA
- Kemper Insurance, Chicago, IL, USA
| | - Marco Mravic
- Department of Pharmaceutical Chemistry, University of California-San Francisco, San Francisco, CA, USA
- Department of Integrative Structural and Computational Biology Scripps Research Institute, La Jolla, CA, USA
| | - Jessica L Thomaston
- Department of Pharmaceutical Chemistry, University of California-San Francisco, San Francisco, CA, USA
| | - John M Nicoludis
- Department of Pharmaceutical Chemistry, University of California-San Francisco, San Francisco, CA, USA
- Genentech, San Francisco, CA, USA
| | - Noah H Somberg
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Mei Hong
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Gregory A Voth
- Department of Chemistry, Chicago Center for Theoretical Chemistry, Institute for Biophysical Dynamics and James Franck Institute, The University of Chicago, Chicago, IL, USA.
| | - William F DeGrado
- Department of Pharmaceutical Chemistry, University of California-San Francisco, San Francisco, CA, USA.
| |
Collapse
|
3
|
Abdella S, Abid F, Youssef SH, Kim S, Afinjuomo F, Malinga C, Song Y, Garg S. pH and its applications in targeted drug delivery. Drug Discov Today 2023; 28:103414. [PMID: 36273779 DOI: 10.1016/j.drudis.2022.103414] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 08/09/2022] [Accepted: 10/17/2022] [Indexed: 02/02/2023]
Abstract
Physiologic pH is vital for the normal functioning of tissues and varies in different parts of the body. The varying pH of the body has been exploited to design pH-sensitive smart oral, transdermal and vaginal drug delivery systems (DDS). The DDS demonstrated promising results in hard-to-treat diseases such as cancer and Helicobacter pylori infection. In some cases, a change in pH of tissues or body fluids has also been employed as a useful diagnostic biomarker. This paper aims to comprehensively review the development and applications of pH-sensitive DDS as well as recent advances in the field.
Collapse
Affiliation(s)
- Sadikalmahdi Abdella
- Centre for Pharmaceutical Innovation (CPI), University of South Australia, Adelaide, SA 5000, Australia; Department of Pharmacology and Clinical Pharmacy, College of Health Sciences, Addis Ababa University, Zambia St, Addis Ababa, Ethiopia
| | - Fatima Abid
- Centre for Pharmaceutical Innovation (CPI), University of South Australia, Adelaide, SA 5000, Australia
| | - Souha H Youssef
- Centre for Pharmaceutical Innovation (CPI), University of South Australia, Adelaide, SA 5000, Australia
| | - Sangseo Kim
- Centre for Pharmaceutical Innovation (CPI), University of South Australia, Adelaide, SA 5000, Australia
| | - Franklin Afinjuomo
- Centre for Pharmaceutical Innovation (CPI), University of South Australia, Adelaide, SA 5000, Australia
| | - Constance Malinga
- Centre for Pharmaceutical Innovation (CPI), University of South Australia, Adelaide, SA 5000, Australia
| | - Yunmei Song
- Centre for Pharmaceutical Innovation (CPI), University of South Australia, Adelaide, SA 5000, Australia
| | - Sanjay Garg
- Centre for Pharmaceutical Innovation (CPI), University of South Australia, Adelaide, SA 5000, Australia.
| |
Collapse
|
4
|
Bhattarai RS, Bariwal J, Kumar V, Hao C, Deng S, Li W, Mahato RI. pH-sensitive nanomedicine of novel tubulin polymerization inhibitor for lung metastatic melanoma. J Control Release 2022; 350:569-583. [PMID: 36037976 PMCID: PMC10322201 DOI: 10.1016/j.jconrel.2022.08.023] [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: 04/19/2022] [Revised: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 10/14/2022]
Abstract
Microtubule binding agents such as paclitaxel and vincristine have activity in metastatic melanoma. However, even responsive tumors develop resistance, highlighting the need to investigate new drug molecules. Here, we showed that a new compound, CH-2-102, developed by our group, has high anti-tumor efficacy in human and murine melanoma cells. We confirmed that CH-2-102 robustly suppresses the microtubule polymerization process by directly interacting with the colchicine binding site. Our results unveil that CH-2-102 suppresses microtubule polymerization and subsequently induces G2 phase cell arrest as one of the possible mechanisms. Notably, CH-2-102 maintains its efficacy even in the paclitaxel resistance melanoma cells due to different binding sites and a non-Pgp substrate. We developed a pH-responsive drug-polymer Schiff bases linker for high drug loading into nanoparticles (NPs). Our CH-2-102 conjugated NPs induced tumor regression more effectively than Abraxane® (Nab-paclitaxel, N-PTX), free drug, and non-sensitive NPs in B16-F10 cell-derived lung metastasis mouse model. Furthermore, our results suggest that the formulation has a high impact on the in vivo efficacy of the drug and warrants further investigation in other cancers, particularly taxane resistant. In conclusion, the microtubule polymerization inhibitor CH-2-102 conjugated pH-responsive NPs induce tumor regression in lung metastasis melanoma mice, suggesting it may be an effective strategy for treating metastatic melanoma.
Collapse
Affiliation(s)
- Rajan S Bhattarai
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jitender Bariwal
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Virender Kumar
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Chen Hao
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Shanshan Deng
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Wei Li
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
5
|
Bernardazzi C, Sheikh IA, Xu H, Ghishan FK. The Physiological Function and Potential Role of the Ubiquitous Na +/H + Exchanger Isoform 8 (NHE8): An Overview Data. Int J Mol Sci 2022; 23:ijms231810857. [PMID: 36142772 PMCID: PMC9501935 DOI: 10.3390/ijms231810857] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/08/2022] [Accepted: 09/15/2022] [Indexed: 11/16/2022] Open
Abstract
The Na+/H+ exchanger transporters (NHE) play an important role in various biologic processes including Na+ absorption, intracellular pH homeostasis, cell volume regulation, proliferation, and apoptosis. The wide expression pattern and cellular localization of NHEs make these proteins pivotal players in virtually all human tissues and organs. In addition, recent studies suggest that NHEs may be one of the primeval transport protein forms in the history of life. Among the different isoforms, the most well-characterized NHEs are the Na+/H+ exchanger isoform 1 (NHE1) and Na+/H+ exchanger isoform 3 (NHE3). However, Na+/H+ exchanger isoform 8 (NHE8) has been receiving attention based on its recent discoveries in the gastrointestinal tract. In this review, we will discuss what is known about the physiological function and potential role of NHE8 in the main organ systems, including useful overviews that could inspire new studies on this multifaceted protein.
Collapse
|
6
|
Fernandez MA, Bah F, Ma L, Lee Y, Schmidt M, Welch E, Morrow EM, Young-Pearse TL. Loss of endosomal exchanger NHE6 leads to pathological changes in tau in human neurons. Stem Cell Reports 2022; 17:2111-2126. [PMID: 36055242 PMCID: PMC9481919 DOI: 10.1016/j.stemcr.2022.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 08/03/2022] [Accepted: 08/03/2022] [Indexed: 12/09/2022] Open
Abstract
Disruption of endolysosomal and autophagy-lysosomal systems is increasingly implicated in neurodegeneration. Sodium-proton exchanger 6 (NHE6) contributes to the maintenance of proper endosomal pH, and loss-of function mutations in the X-linked NHE6 lead to Christianson syndrome (CS) in males. Neurodegenerative features of CS are increasingly recognized, with postmortem and clinical data implicating a role for tau. We generated cortical neurons from NHE6 knockout (KO) and isogenic wild-type control human induced pluripotent stem cells. We report elevated phosphorylated and sarkosyl-insoluble tau in NHE6 KO neurons. We demonstrate that NHE6 KO leads to lysosomal and autophagy dysfunction involving reduced lysosomal number and protease activity, diminished autophagic flux, and p62 accumulation. Finally, we show that treatment with trehalose or rapamycin, two enhancers of autophagy-lysosomal function, each partially rescue this tau phenotype. We provide insight into the neurodegenerative processes underlying NHE6 loss of function and into the broader role of the endosome-lysosome-autophagy network in neurodegeneration.
Collapse
Affiliation(s)
- Marty A Fernandez
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Fatmata Bah
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Li Ma
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA; Center for Translational Neuroscience, Carney Institute for Brain Science and Brown Institute for Translational Science (BITS), Brown University, Providence, RI 02912, USA
| | - YouJin Lee
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA; Center for Translational Neuroscience, Carney Institute for Brain Science and Brown Institute for Translational Science (BITS), Brown University, Providence, RI 02912, USA
| | - Michael Schmidt
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA; Center for Translational Neuroscience, Carney Institute for Brain Science and Brown Institute for Translational Science (BITS), Brown University, Providence, RI 02912, USA
| | - Elizabeth Welch
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Eric M Morrow
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA; Center for Translational Neuroscience, Carney Institute for Brain Science and Brown Institute for Translational Science (BITS), Brown University, Providence, RI 02912, USA; Department of Psychiatry and Human Behavior, Warren Alpert Medical School of Brown University, Providence, RI 02912, USA.
| | - Tracy L Young-Pearse
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
7
|
Shinn J, Kwon N, Lee SA, Lee Y. Smart pH-responsive nanomedicines for disease therapy. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2022; 52:427-441. [PMID: 35573320 PMCID: PMC9083479 DOI: 10.1007/s40005-022-00573-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 04/20/2022] [Indexed: 02/06/2023]
Abstract
Background Currently nanomedicines are the focus of attention from researchers and clinicians because of the successes of lipid-nanoparticles-based COVID-19 vaccines. Nanoparticles improve existing treatments by providing a number of advantages including protection of cargo molecules from external stresses, delivery of drugs to target tissues, and sustained drug release. To prevent premature release-related side effects, stable drug loading in nanoformulations is required, but the increased stability of the formulation could also lead to a poor drug-release profile at the target sites. Thus, researchers have exploited differences in a range of properties (e.g., enzyme levels, pH, levels of reduced glutathione, and reactive oxygen species) between non-target and target sites for site-specific release of drugs. Among these environmental stimuli, pH gradients have been widely used to design novel, responsive nanoparticles. Area covered In this review, we assess drug delivery based on pH-responsive nanoparticles at the levels of tissues (tumor microenvironment, pH ~ 6.5) and of intracellular compartments (endosome and lysosome, pH 4.5-6.5). Upon exposure to these pH stimuli, pH-responsive nanoparticles respond with physicochemical changes to their material structure and surface characteristics. These changes include swelling, dissociation, or surface charge switching, in a manner that favors drug release at the target site (the tumor microenvironment region and the cytosol followed by endosomal escape) rather than the surrounding tissues. Expert opinion Lastly, we consider the challenges involved in the development of pH-responsive nanomedicines.
Collapse
Affiliation(s)
- Jongyoon Shinn
- College of Pharmacy, Ewha Womans University, Seoul, 03760 South Korea
| | - Nuri Kwon
- College of Pharmacy, Ewha Womans University, Seoul, 03760 South Korea
| | - Seon Ah Lee
- College of Pharmacy, Ewha Womans University, Seoul, 03760 South Korea
| | - Yonghyun Lee
- College of Pharmacy, Ewha Womans University, Seoul, 03760 South Korea
| |
Collapse
|
8
|
Behl T, Kaur D, Sehgal A, Singh S, Makeen HA, Albratty M, Abdellatif AAH, Dachani SR, Bungau S. Exploring the potential role of rab5 protein in endo-lysosomal impairment in Alzheimer's disease. Biomed Pharmacother 2022; 148:112773. [PMID: 35245734 DOI: 10.1016/j.biopha.2022.112773] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/18/2022] [Accepted: 02/27/2022] [Indexed: 11/02/2022] Open
Abstract
Growing evidence suggests that neuronal dysfunction in the endo-lysosomal and autophagic processes contributes to the onset and progression of neurodegenerative diseases such as Alzheimer's disease (AD). Since they are the primary cellular systems involved in the production and clearance of aggregated amyloid plaques, endo-lysosomal or autophagic equilibrium must be maintained throughout life. As a result, variations in the autophagic and endo-lysosomal torrent, as a measure of degenerative function in these sections or pathways, may have a direct impact on disease-related processes, such as Aß clearance from the brain and interneuronal deposition of Aß and tau aggregates, thus disrupting synaptic plasticity. The discovery of several chromosomal factors for Alzheimer's disease that are clinically linked to regulation of the endocytic pathway, including protein aggregation and removal, supports the theory that the endo-lysosomal/autophagic torrent is more susceptible to impairment, especially as people age, thus catalysing the onset of disease. Although the role of endo-lysosomal/autophagic dysfunction in neurodegeneration has progressed in recent years, the field remains underdeveloped. Because of its possible therapeutic implications in Alzheimer's disease, further study is needed to explain the possibilities for effective autophagy regulation.
Collapse
Affiliation(s)
- Tapan Behl
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India.
| | - Dapinder Kaur
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India
| | - Aayush Sehgal
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India
| | - Sukhbir Singh
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India
| | - Hafiz A Makeen
- Pharmacy Practice Research Unit, Clinical Pharmacy, Department, College of Pharmacy, Jazan University, P.O. Box-114, Jazan 45142, Saudi Arabia
| | - Mohammed Albratty
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia
| | - Ahmed A H Abdellatif
- Department of Pharmaceutics, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
| | - Sudharshan Reddy Dachani
- Department of Pharmacy Practice & Pharmacology, College of Pharmacy, Shaqra University, Al-Dawadmi Campus, Al-Dawadmi 11961, Saudi Arabia
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania.
| |
Collapse
|
9
|
Seo N, Nakamura J, Kaneda T, Tateno H, Shimoda A, Ichiki T, Furukawa K, Hirabayashi J, Akiyoshi K, Shiku H. Distinguishing functional exosomes and other extracellular vesicles as a nucleic acid cargo by the anion-exchange method. J Extracell Vesicles 2022; 11:e12205. [PMID: 35289089 PMCID: PMC8920962 DOI: 10.1002/jev2.12205] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 01/15/2022] [Accepted: 03/01/2022] [Indexed: 12/13/2022] Open
Abstract
The development of a new large‐scale purification protocol is required for research on the reliable bioactivity and drug discovery of extracellular vesicles (EVs). To address this issue, herein, we propose an effective method for preparing high‐performance exosomes (EXOs) by using an anion‐exchange method. Cytotoxic T‐lymphocyte (CTL) EVs from 4 L of culture supernatant through a 220 nm cut‐off filter are divided into two populations at a deproteinization rate of over 99.97%, which are eluted at low (0.15 M–0.3 M) and high (0.3 M–0.5 M) NaCl concentrations (approximately 2 × 1012 and 1.5 × 1012 particles, respectively) through the anion‐exchange column chromatography. The former are abundant in EXO proteins, including late endosome‐associated proteins and rab‐family and integrin‐family proteins, and functional micro (mi) RNAs, and have bioactivity for preventing tumour metastasis by depleting mesenchymal cell populations in the primary tumour lesions. By contrast, the latter is microvesicle (MV)‐like particles including DNA, core histone and ribosomal proteins, and GC‐rich miRNAs with unknown function, and are easily phagocytosed by mannose receptor+ Kupffer cells. Thus, the anion‐exchange method is suitable for the large‐scale separation of bioactive EXOs and MV‐like EVs as a cargo for dangerous nucleic acids at high‐purity.
Collapse
Affiliation(s)
- Naohiro Seo
- Department of Personalized Cancer Immunotherapy, Mie University Graduate School of Medicine, Mie, Japan.,Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo, Japan
| | - Junko Nakamura
- Department of Personalized Cancer Immunotherapy, Mie University Graduate School of Medicine, Mie, Japan
| | - Tsuguhiro Kaneda
- Department of Personalized Cancer Immunotherapy, Mie University Graduate School of Medicine, Mie, Japan
| | - Hiroaki Tateno
- Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo, Japan.,Research Center for Medical Glycoscience, National Institute of Advanced Industrial Science and Technology, Ibaraki, Japan
| | - Asako Shimoda
- Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo, Japan.,Department of Polymer Chemistry, Graduate School of Engineering, Katsura Int'tech Center, Kyoto University, Kyoto, Japan
| | - Takanori Ichiki
- Department of Materials Engineering, School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Koichi Furukawa
- Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo, Japan.,Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Aichi, Japan
| | - Jun Hirabayashi
- Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo, Japan.,Institute for Glyco-core Research (iGCORE), Nagoya University, Aichi, Japan
| | - Kazunari Akiyoshi
- Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo, Japan.,Department of Polymer Chemistry, Graduate School of Engineering, Katsura Int'tech Center, Kyoto University, Kyoto, Japan
| | - Hiroshi Shiku
- Department of Personalized Cancer Immunotherapy, Mie University Graduate School of Medicine, Mie, Japan
| |
Collapse
|
10
|
Sivils A, Yang F, Wang JQ, Chu XP. Acid-Sensing Ion Channel 2: Function and Modulation. MEMBRANES 2022; 12:membranes12020113. [PMID: 35207035 PMCID: PMC8880099 DOI: 10.3390/membranes12020113] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 01/08/2023]
Abstract
Acid-sensing ion channels (ASICs) have an important influence on human physiology and pathology. They are members of the degenerin/epithelial sodium channel family. Four genes encode at least six subunits, which combine to form a variety of homotrimers and heterotrimers. Of these, ASIC1a homotrimers and ASIC1a/2 heterotrimers are most widely expressed in the central nervous system (CNS). Investigations into the function of ASIC1a in the CNS have revealed a wealth of information, culminating in multiple contemporary reviews. The lesser-studied ASIC2 subunits are in need of examination. This review will focus on ASIC2 in health and disease, with discussions of its role in modulating ASIC function, synaptic targeting, cardiovascular responses, and pharmacology, while exploring evidence of its influence in pathologies such as ischemic brain injury, multiple sclerosis, epilepsy, migraines, drug addiction, etc. This information substantiates the ASIC2 protein as a potential therapeutic target for various neurological, psychological, and cerebrovascular diseases.
Collapse
Affiliation(s)
| | | | | | - Xiang-Ping Chu
- Correspondence: ; Tel.: +1-816-235-2248; Fax: +1-816-235-6517
| |
Collapse
|
11
|
Williamson M, Moustaid-Moussa N, Gollahon L. The Molecular Effects of Dietary Acid Load on Metabolic Disease (The Cellular PasaDoble: The Fast-Paced Dance of pH Regulation). FRONTIERS IN MOLECULAR MEDICINE 2021; 1:777088. [PMID: 39087082 PMCID: PMC11285710 DOI: 10.3389/fmmed.2021.777088] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/27/2021] [Indexed: 08/02/2024]
Abstract
Metabolic diseases are becoming more common and more severe in populations adhering to western lifestyle. Since metabolic conditions are highly diet and lifestyle dependent, it is suggested that certain diets are the cause for a wide range of metabolic dysfunctions. Oxidative stress, excess calcium excretion, inflammation, and metabolic acidosis are common features in the origins of most metabolic disease. These primary manifestations of "metabolic syndrome" can lead to insulin resistance, diabetes, obesity, and hypertension. Further complications of the conditions involve kidney disease, cardiovascular disease, osteoporosis, and cancers. Dietary analysis shows that a modern "Western-style" diet may facilitate a disruption in pH homeostasis and drive disease progression through high consumption of exogenous acids. Because so many physiological and cellular functions rely on acid-base reactions and pH equilibrium, prolonged exposure of the body to more acids than can effectively be buffered, by chronic adherence to poor diet, may result in metabolic stress followed by disease. This review addresses relevant molecular pathways in mammalian cells discovered to be sensitive to acid - base equilibria, their cellular effects, and how they can cascade into an organism-level manifestation of Metabolic Syndromes. We will also discuss potential ways to help mitigate this digestive disruption of pH and metabolic homeostasis through dietary change.
Collapse
Affiliation(s)
- Morgan Williamson
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, United States
| | - Naima Moustaid-Moussa
- Department of Nutrition Sciences, Texas Tech University, Lubbock, TX, United States
- Obesity Research Institute, Texas Tech University, Lubbock, TX, United States
| | - Lauren Gollahon
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, United States
- Department of Nutrition Sciences, Texas Tech University, Lubbock, TX, United States
- Obesity Research Institute, Texas Tech University, Lubbock, TX, United States
| |
Collapse
|
12
|
FLIM-Based Intracellular and Extracellular pH Measurements Using Genetically Encoded pH Sensor. BIOSENSORS-BASEL 2021; 11:bios11090340. [PMID: 34562930 PMCID: PMC8468847 DOI: 10.3390/bios11090340] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/07/2021] [Accepted: 09/10/2021] [Indexed: 02/04/2023]
Abstract
The determination of pH in live cells and tissues is of high importance in physiology and cell biology. In this report, we outline the process of the creation of SypHerExtra, a genetically encoded fluorescent sensor that is capable of measuring extracellular media pH in a mildly alkaline range. SypHerExtra is a protein created by fusing the previously described pH sensor SypHer3s with the neurexin transmembrane domain that targets its expression to the cytoplasmic membrane. We showed that with excitation at 445 nm, the fluorescence lifetime of both SypHer3s and SypHerExtra strongly depend on pH. Using FLIM microscopy in live eukaryotic cells, we demonstrated that SypHerExtra can be successfully used to determine extracellular pH, while SypHer3s can be applied to measure intracellular pH. Thus, these two sensors are suitable for quantitative measurements using the FLIM method, to determine intracellular and extracellular pH in a range from pH 7.5 to 9.5 in different biological systems.
Collapse
|
13
|
Pottanam Chali S, Hüwel S, Rentmeister A, Ravoo BJ. Self-Assembled Cationic Polypeptide Supramolecular Nanogels for Intracellular DNA Delivery. Chemistry 2021; 27:12198-12206. [PMID: 34125454 PMCID: PMC8457085 DOI: 10.1002/chem.202101924] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Indexed: 12/14/2022]
Abstract
Supramolecular nanogels are an emerging class of polymer nanocarriers for intracellular delivery, due to their straightforward preparation, biocompatibility, and capability to spontaneously encapsulate biologically active components such as DNA. A completely biodegradable three-component cationic supramolecular nanogel was designed exploiting the multivalent host-guest interaction of cyclodextrin and adamantane attached to a polypeptide backbone. While cyclodextrin was conjugated to linear poly-L-lysine, adamantane was grafted to linear as well as star shaped poly-L-lysine. Size control of nanogels was obtained with the increase in the length of the host and guest polymer. Moreover, smaller nanogels were obtained using the star shaped polymers because of the compact nature of star polymers compared to linear polymers. Nanogels were loaded with anionic model cargoes, pyranine and carboxyfluorescein, and their enzyme responsive release was studied using protease trypsin. Confocal microscopy revealed successful transfection of mammalian HeLa cells and intracellular release of pyranine and plasmid DNA, as quantified using a luciferase assay, showing that supramolecular polypeptide nanogels have significant potential in gene therapy applications.
Collapse
Affiliation(s)
- Sharafudheen Pottanam Chali
- Organic Chemistry Institute and Centre for Soft NanoscienceWestfälische Wilhelms-Universität MünsterCorrensstrasse 3648149MünsterGermany
| | - Sabine Hüwel
- Institute of BiochemistryWestfälische Wilhelms-Universität MünsterCorrensstrasse 3648149MünsterGermany
| | - Andrea Rentmeister
- Institute of BiochemistryWestfälische Wilhelms-Universität MünsterCorrensstrasse 3648149MünsterGermany
| | - Bart Jan Ravoo
- Organic Chemistry Institute and Centre for Soft NanoscienceWestfälische Wilhelms-Universität MünsterCorrensstrasse 3648149MünsterGermany
| |
Collapse
|
14
|
Lai SSM, Ng KY, Koh RY, Chok KC, Chye SM. Endosomal-lysosomal dysfunctions in Alzheimer's disease: Pathogenesis and therapeutic interventions. Metab Brain Dis 2021; 36:1087-1100. [PMID: 33881723 DOI: 10.1007/s11011-021-00737-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/08/2021] [Indexed: 12/14/2022]
Abstract
The endosomal-lysosomal system mediates the process of protein degradation through endocytic pathway. This system consists of early endosomes, late endosomes, recycling endosomes and lysosomes. Each component in the endosomal-lysosomal system plays individual crucial role and they work concordantly to ensure protein degradation can be carried out functionally. Dysregulation in the endosomal-lysosomal system can contribute to the pathogenesis of neurodegenerative diseases such as Alzheimer's disease (AD). In AD endosomal-lysosomal abnormalities are the earliest pathological features to note and hence it is important to understand the involvement of endosomal-lysosomal dysfunction in the pathogenesis of AD. In-depth understanding of this dysfunction can allow development of new therapeutic intervention to prevent and treat AD.
Collapse
Affiliation(s)
- Shereen Shi Min Lai
- School of Health Science, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Khuen Yen Ng
- School of Pharmacy, Monash University Malaysia, 47500, Selangor, Malaysia
| | - Rhun Yian Koh
- Division of Biomedical Science and Biotechnology, School of Health Science, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Kian Chung Chok
- School of Health Science, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Soi Moi Chye
- Division of Biomedical Science and Biotechnology, School of Health Science, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia.
| |
Collapse
|
15
|
Pizzagalli MD, Bensimon A, Superti‐Furga G. A guide to plasma membrane solute carrier proteins. FEBS J 2021; 288:2784-2835. [PMID: 32810346 PMCID: PMC8246967 DOI: 10.1111/febs.15531] [Citation(s) in RCA: 186] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 08/07/2020] [Accepted: 08/17/2020] [Indexed: 12/13/2022]
Abstract
This review aims to serve as an introduction to the solute carrier proteins (SLC) superfamily of transporter proteins and their roles in human cells. The SLC superfamily currently includes 458 transport proteins in 65 families that carry a wide variety of substances across cellular membranes. While members of this superfamily are found throughout cellular organelles, this review focuses on transporters expressed at the plasma membrane. At the cell surface, SLC proteins may be viewed as gatekeepers of the cellular milieu, dynamically responding to different metabolic states. With altered metabolism being one of the hallmarks of cancer, we also briefly review the roles that surface SLC proteins play in the development and progression of cancer through their influence on regulating metabolism and environmental conditions.
Collapse
Affiliation(s)
- Mattia D. Pizzagalli
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Ariel Bensimon
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Giulio Superti‐Furga
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
- Center for Physiology and PharmacologyMedical University of ViennaAustria
| |
Collapse
|
16
|
Bahou C, Szijj PA, Spears RJ, Wall A, Javaid F, Sattikar A, Love EA, Baker JR, Chudasama V. A Plug-and-Play Platform for the Formation of Trifunctional Cysteine Bioconjugates that also Offers Control over Thiol Cleavability. Bioconjug Chem 2021; 32:672-679. [PMID: 33710874 PMCID: PMC8154211 DOI: 10.1021/acs.bioconjchem.1c00057] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/04/2021] [Indexed: 02/08/2023]
Abstract
Linkers that enable the site-selective synthesis of chemically modified proteins are of great interest to the field of chemical biology. Homogenous bioconjugates often show advantageous pharmacokinetic profiles and consequently increased efficacy in vivo. Cysteine residues have been exploited as a route to site-selectively modify proteins, and many successfully approved therapeutics make use of cysteine directed conjugation reagents. However, commonly used linkers, including maleimide-thiol conjugates, are not stable to the low concentrations of thiol present in blood. Furthermore, only a few cysteine-targeting reagents enable the site-selective attachment of multiple functionalities: a useful tool in the fields of theranostics and therapeutic blood half-life extension. Herein, we demonstrate the application of the pyridazinedione motif to enable site-selective attachment of three functionalities to a protein bearing a single cysteine residue. Extending upon previously documented dual modification work, here we demonstrate that by exploiting a bromide leaving group as an additional reactive point on the pyridazinedione scaffold, a thiol or aniline derivative can be added to a protein, post-conjugation. Thiol cleavability appraisal of the resultant C-S and C-N linked thio-bioconjugates demonstrated C-S functionalized linkers to be cleavable and C-N functionalized linkers to be noncleavable when incubated in an excess of glutathione. The plug-and-play trifunctional platform was exemplified by attaching clinically relevant motifs: biotin, fluorescein, a polyethylene glycol chain, and a model peptide. This platform provides a rare opportunity to combine up to three functionalities on a protein in a site-selective fashion. Furthermore, by selecting the use of a thiol or an amine for functionalization, we provide unique control over linker cleavability toward thiols, allowing this novel linker to be applied in a range of physiological environments.
Collapse
Affiliation(s)
- Calise Bahou
- Department
of Chemistry, University College London, 20 Gordon Street, WC1H OAJ, London, United Kingdom
| | - Peter A. Szijj
- Department
of Chemistry, University College London, 20 Gordon Street, WC1H OAJ, London, United Kingdom
| | - Richard J. Spears
- Department
of Chemistry, University College London, 20 Gordon Street, WC1H OAJ, London, United Kingdom
| | - Archie Wall
- Department
of Chemistry, University College London, 20 Gordon Street, WC1H OAJ, London, United Kingdom
| | - Faiza Javaid
- Department
of Chemistry, University College London, 20 Gordon Street, WC1H OAJ, London, United Kingdom
| | - Afrah Sattikar
- LifeArc,
Accelerator Building, SBC Open Innovation Campus, SG1 2FX, Stevenage, United Kingdom
| | - Elizabeth A. Love
- LifeArc,
Accelerator Building, SBC Open Innovation Campus, SG1 2FX, Stevenage, United Kingdom
| | - James R. Baker
- Department
of Chemistry, University College London, 20 Gordon Street, WC1H OAJ, London, United Kingdom
| | - Vijay Chudasama
- Department
of Chemistry, University College London, 20 Gordon Street, WC1H OAJ, London, United Kingdom
- Research
Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-004 Lisbon, Portugal
| |
Collapse
|
17
|
Sengupta P, Bose D, Chatterjee S. The Molecular Tête-à-Tête between G-Quadruplexes and the i-motif in the Human Genome. Chembiochem 2021; 22:1517-1537. [PMID: 33355980 DOI: 10.1002/cbic.202000703] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/16/2020] [Indexed: 12/22/2022]
Abstract
G-Quadruplex (GQ) and i-motif structures are the paradigmatic examples of nonclassical tetrastranded nucleic acids having multifarious biological functions and widespread applications in therapeutics and material science. Recently, tetraplexes emerged as promising anticancer targets due to their structural robustness, gene-regulatory roles, and predominant distribution at specific loci of oncogenes. However, it is arguable whether the i-motif evolves in the complementary single-stranded region after GQ formation in its opposite strand and vice versa. In this review, we address the prerequisites and significance of the simultaneous and/or mutually exclusive formation of GQ and i-motif structures at complementary and sequential positions in duplexes in the cellular milieu. We discussed how their dynamic interplay Sets up cellular homeostasis and exacerbates carcinogenesis. The review gives insights into the spatiotemporal formation of GQ and i-motifs that could be harnessed to design different types of reporter systems and diagnostic platforms for potential bioanalytical and therapeutic intervention.
Collapse
Affiliation(s)
- Pallabi Sengupta
- Department of Biophysics, Bose Institute, Centenary Campus, P-1/12, C.I.T. Scheme VIIM, Kankurgachi, Kolkata, 700054, West Bengal, India
| | - Debopriya Bose
- Department of Biophysics, Bose Institute, Centenary Campus, P-1/12, C.I.T. Scheme VIIM, Kankurgachi, Kolkata, 700054, West Bengal, India
| | - Subhrangsu Chatterjee
- Department of Biophysics, Bose Institute, Centenary Campus, P-1/12, C.I.T. Scheme VIIM, Kankurgachi, Kolkata, 700054, West Bengal, India
| |
Collapse
|
18
|
Baker S, Petukh M. Effect of pH on the Ability of N-Terminal Domain of Human NPC1 to Recognize, Bind, and Transfer Cholesterol. ACS OMEGA 2020; 5:29222-29230. [PMID: 33225153 PMCID: PMC7676335 DOI: 10.1021/acsomega.0c03983] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/21/2020] [Indexed: 06/11/2023]
Abstract
Niemann-Pick type C1 (NPC1) is a large multidomain transmembrane protein essential for transporting cholesterol (CLR) from late endosomes and lysosomes to the endoplasmic reticulum and other cellular compartments. The lumen-facing N-terminal domain (NTD), involved in direct binding of CLR, is expected to have an optimum activity at acidic pH = 4.5. Here, we show that acidic pH is vital for the functionality of NPC1(NTD) and should be taken into account when studying the protein activity. We applied evolutionary, structural, and physicochemical analyses to decipher the consequences of a change in pH from acidic (pH = 4.5) to neutral (pH = 7.2) on the structural integrity of the NTD and its ability to bind CLR. We revealed that the change in pH from 4.5 to 7.2 increases the potential energy of the protein in both apo- and holo-states making the system less energetically favorable. At neutral pH, the flexibility of the protein in the apo-state is decreased caused by the alteration of specific interactions, which in turn might have a high impact on ligand recognition and binding. In contrast, neutral pH significantly exaggerates the flexibility of the protein with bound CLR that causes a partial exposure of the ligand to the water phase and its mislocation inside the ligand-binding pocket, which might obstruct CLR translocation through the membrane.
Collapse
|
19
|
Zhang H, Tian X, Lu X, Xu D, Guo Y, Dong Z, Li Y, Ma Y, Chen C, Yang Y, Yang M, Yang Y, Liu F, Zhou R, He M, Xiao F, Wang X. TMEM25 modulates neuronal excitability and NMDA receptor subunit NR2B degradation. J Clin Invest 2020; 129:3864-3876. [PMID: 31424425 DOI: 10.1172/jci122599] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 06/24/2019] [Indexed: 12/24/2022] Open
Abstract
The expression of the transmembrane protein 25 gene (Tmem25) is strongly influenced by glutamate ionotropic receptor kainate type subunit 4, and its function remains unknown. Here, we showed that TMEM25 was primarily localized to late endosomes in neurons. Electrophysiological experiments suggested that the effects of TMEM25 on neuronal excitability were likely mediated by N-methyl-d-aspartate receptors. TMEM25 affected the expression of the N-methyl-d-aspartate receptor NR2B subunit and interacted with NR2B, and both were colocalized to late endosome compartments. TMEM25 induced acidification changes in lysosome compartments and accelerated the degradation of NR2B. Furthermore, TMEM25 expression was decreased in brain tissues from patients with epilepsy and epileptic mice. TMEM25 overexpression attenuated the behavioral phenotypes of epileptic seizures, whereas TMEM25 downregulation exerted the opposite effect. These results provide some insights into TMEM25 biology in the brain and the functional relationship between TMEM25 and epilepsy.
Collapse
Affiliation(s)
- Haiqing Zhang
- Department of Neurology, First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Xin Tian
- Department of Neurology, First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Xi Lu
- Department of Neurology, First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Demei Xu
- Department of Neurology, First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Yi Guo
- Department of Neurology, First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Zhifang Dong
- Ministry of Education Key Laboratory of Child Development and Disorders and Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yun Li
- Department of Neurology, First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Yuanlin Ma
- Department of Neurology, First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Chengzhi Chen
- School of Public Health and Management, Research Center for Medicine and Social Development, Chongqing Medical University, Chongqing, China
| | - Yong Yang
- Department of Neurology, First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Min Yang
- Department of Neurology, First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Yi Yang
- Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing, China
| | - Feng Liu
- Department of Neurology, First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Ruijiao Zhou
- Department of Neurology, First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Miaoqing He
- Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing, China
| | - Fei Xiao
- Department of Neurology, First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Xuefeng Wang
- Department of Neurology, First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China.,Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
20
|
Song Q, Meng B, Xu H, Mao Z. The emerging roles of vacuolar-type ATPase-dependent Lysosomal acidification in neurodegenerative diseases. Transl Neurodegener 2020; 9:17. [PMID: 32393395 PMCID: PMC7212675 DOI: 10.1186/s40035-020-00196-0] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 04/23/2020] [Indexed: 12/15/2022] Open
Abstract
Background Lysosomes digest extracellular material from the endocytic pathway and intracellular material from the autophagic pathway. This process is performed by the resident hydrolytic enzymes activated by the highly acidic pH within the lysosomal lumen. Lysosome pH gradients are mainly maintained by the vacuolar (H+) ATPase (or V-ATPase), which pumps protons into lysosomal lumen by consuming ATP. Dysfunction of V-ATPase affects lysosomal acidification, which disrupts the clearance of substrates and leads to many disorders, including neurodegenerative diseases. Main body As a large multi-subunit complex, the V-ATPase is composed of an integral membrane V0 domain involved in proton translocation and a peripheral V1 domain catalyzing ATP hydrolysis. The canonical functions of V-ATPase rely on its H+-pumping ability in multiple vesicle organelles to regulate endocytic traffic, protein processing and degradation, synaptic vesicle loading, and coupled transport. The other non-canonical effects of the V-ATPase that are not readily attributable to its proton-pumping activity include membrane fusion, pH sensing, amino-acid-induced activation of mTORC1, and scaffolding for protein-protein interaction. In response to various stimuli, V-ATPase complex can reversibly dissociate into V1 and V0 domains and thus close ATP-dependent proton transport. Dysregulation of pH and lysosomal dysfunction have been linked to many human diseases, including neurodegenerative disorders such as Alzheimer disease, Parkinson’s disease, amyotrophic lateral sclerosis as well as neurodegenerative lysosomal storage disorders. Conclusion V-ATPase complex is a universal proton pump and plays an important role in lysosome acidification in all types of cells. Since V-ATPase dysfunction contributes to the pathogenesis of multiple neurodegenerative diseases, further understanding the mechanisms that regulate the canonical and non-canonical functions of V-ATPase will reveal molecular details of disease process and help assess V-ATPase or molecules related to its regulation as therapeutic targets.
Collapse
Affiliation(s)
- Qiaoyun Song
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA.,Department of Reproductive Genetics, Hebei General Hospital, Shijiazhuang, Hebei Province, 050051, People's Republic of China.,Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Bo Meng
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA.,Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Haidong Xu
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA.,Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Zixu Mao
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA. .,Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
21
|
Kumar R, Tang Q, Müller SA, Gao P, Mahlstedt D, Zampagni S, Tan Y, Klingl A, Bötzel K, Lichtenthaler SF, Höglinger GU, Koeglsperger T. Fibroblast Growth Factor 2-Mediated Regulation of Neuronal Exosome Release Depends on VAMP3/Cellubrevin in Hippocampal Neurons. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1902372. [PMID: 32195080 PMCID: PMC7080514 DOI: 10.1002/advs.201902372] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 12/11/2019] [Indexed: 05/06/2023]
Abstract
Extracellular vesicles (EVs) are endogenous membrane-derived vesicles that shuttle bioactive molecules between glia and neurons, thereby promoting neuronal survival and plasticity in the central nervous system (CNS) and contributing to neurodegenerative conditions. Although EVs hold great potential as CNS theranostic nanocarriers, the specific molecular factors that regulate neuronal EV uptake and release are currently unknown. A combination of patch-clamp electrophysiology and pH-sensitive dye imaging is used to examine stimulus-evoked EV release in individual neurons in real time. Whereas spontaneous electrical activity and the application of a high-frequency stimulus induce a slow and prolonged fusion of multivesicular bodies (MVBs) with the plasma membrane (PM) in a subset of cells, the neurotrophic factor basic fibroblast growth factor (bFGF) greatly increases the rate of stimulus-evoked MVB-PM fusion events and, consequently, the abundance of EVs in the culture medium. Proteomic analysis of neuronal EVs demonstrates bFGF increases the abundance of the v-SNARE vesicle-associated membrane protein 3 (VAMP3, cellubrevin) on EVs. Conversely, knocking-down VAMP3 in cultured neurons attenuates the effect of bFGF on EV release. The results determine the temporal characteristics of MVB-PM fusion in hippocampal neurons and reveal a new function for bFGF signaling in controlling neuronal EV release.
Collapse
Affiliation(s)
- Rohit Kumar
- Department of Translational NeurodegenerationGerman Centre for Neurodegenerative DiseasesFeodor‐Lynen‐Str. 1781377MunichGermany
- Department of NeurologyLudwig Maximilian UniversityMarchioninistr. 1581377MunichGermany
- Graduate Program for Experimental MedicineFaculty of MedicineTechnical University of MunichIsmaninger Straße 2281675MünchenGermany
| | - Qilin Tang
- Department of Translational NeurodegenerationGerman Centre for Neurodegenerative DiseasesFeodor‐Lynen‐Str. 1781377MunichGermany
- Department of NeurologyLudwig Maximilian UniversityMarchioninistr. 1581377MunichGermany
| | - Stephan A. Müller
- Department of NeuroproteomicsGerman Centre for Neurodegenerative DiseasesFeodor‐Lynen‐Str. 1781377MunichGermany
| | - Pan Gao
- Department of Translational NeurodegenerationGerman Centre for Neurodegenerative DiseasesFeodor‐Lynen‐Str. 1781377MunichGermany
| | - Diana Mahlstedt
- Department of Translational NeurodegenerationGerman Centre for Neurodegenerative DiseasesFeodor‐Lynen‐Str. 1781377MunichGermany
- Graduate Program for Experimental MedicineFaculty of MedicineTechnical University of MunichIsmaninger Straße 2281675MünchenGermany
| | - Sofia Zampagni
- Department of Translational NeurodegenerationGerman Centre for Neurodegenerative DiseasesFeodor‐Lynen‐Str. 1781377MunichGermany
| | - Yi Tan
- Department of Translational NeurodegenerationGerman Centre for Neurodegenerative DiseasesFeodor‐Lynen‐Str. 1781377MunichGermany
- Graduate Program for Experimental MedicineFaculty of MedicineTechnical University of MunichIsmaninger Straße 2281675MünchenGermany
| | - Andreas Klingl
- Plant Development and Electron MicroscopyDepartment of Biology IBiocenterLudwig Maximilian UniversityGroßhaderner Str. 282152Planegg‐MartinsriedGermany
| | - Kai Bötzel
- Department of NeurologyLudwig Maximilian UniversityMarchioninistr. 1581377MunichGermany
| | - Stefan F. Lichtenthaler
- Department of NeuroproteomicsGerman Centre for Neurodegenerative DiseasesFeodor‐Lynen‐Str. 1781377MunichGermany
- NeuroproteomicsKlinikum rechts der IsarInstitute for Advanced StudyTechnical University of MunichIsmaninger Straße 2281675MunichGermany
| | - Günter U. Höglinger
- Department of Translational NeurodegenerationGerman Centre for Neurodegenerative DiseasesFeodor‐Lynen‐Str. 1781377MunichGermany
- Department of Neurology (OE 7210)Hannover Medical SchoolCarl‐Neuberg‐Str. 130625HannoverGermany
- Department of NeurologyTechnical University of MunichIsmaninger Str. 2281675MunichGermany
| | - Thomas Koeglsperger
- Department of Translational NeurodegenerationGerman Centre for Neurodegenerative DiseasesFeodor‐Lynen‐Str. 1781377MunichGermany
- Department of NeurologyLudwig Maximilian UniversityMarchioninistr. 1581377MunichGermany
| |
Collapse
|
22
|
Fernández M, Shamsabadi A, Chudasama V. Fine-tuning thio-pyridazinediones as SMDC scaffolds (with intracellular thiol release via a novel self-immolative linker). Chem Commun (Camb) 2020; 56:1125-1128. [PMID: 31894778 DOI: 10.1039/c9cc08744c] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Herein we report the synthesis of a library of thioalkyl- and thioaryl-pyridazinediones for thiol-based self-immolative release of cargo. A bisthioaryl-pyridazinedione is shown to be stable to serum protein albumin but unstable in intracellular conditions. A derivatised analogue underwent self-immolative degradation in cellular thiol conditions as evidenced by LC-MS/release of a turn-on fluorescence fluorophore; versatility of the thiol-pyridazinedione is demonstrated through synthesis of SMDC precursors that contain three different functional groups on the same central molecule.
Collapse
|
23
|
Kurata T, Rajendran V, Fan S, Ohta T, Numata M, Fushida S. NHE5 regulates growth factor signaling, integrin trafficking, and degradation in glioma cells. Clin Exp Metastasis 2019; 36:527-538. [PMID: 31595389 PMCID: PMC6834540 DOI: 10.1007/s10585-019-10001-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 09/30/2019] [Indexed: 11/26/2022]
Abstract
Na+/H+ exchanger 5 (NHE5) is enriched in neurons and cycles between recycling endosomes and plasma membranes and transports protons to the endosomal lumen as well as to the extracellular space. Although NHE5 expression is undetectable in normal astrocytes, C6 glioma cells express NHE5 at an elevated level. Using C6 cells as a model, here we demonstrate that NHE5 has an important role in tumor growth and tumor cell proliferation and invasion. Glioma xenografts originating from NHE5-knockdown cells exhibited significantly slower growth than those from NHE1-knockdown cells and control cells. Histological characterization of the migration front of NHE5-knockdown tumors revealed a less invasive and less proliferative appearance than NHE1-knockdown and control tumors. NHE5-knockdown but not NHE1-knockdown led to downregulation of fetal bovine serum (FBS)-induced MET and EGFR signaling. Moreover, depletion of NHE5 but not NHE1 reduced the ability of cells to spread on collagen. We found that NHE5 depletion greatly abrogated endocytic recycling and the protein stability of β1-integrin, which in part accounted for the defective cell adhesion, spreading, and invasion of NHE5-knockdown cells.
Collapse
Affiliation(s)
- Toru Kurata
- Department of Gastroenterological Surgery, Kanazawa University Graduate School of Medical Science, 13-1 Takara-machi, Kanazawa, 920-8641, Japan
| | - Vinotheni Rajendran
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Steven Fan
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Tetsuo Ohta
- Department of Gastroenterological Surgery, Kanazawa University Graduate School of Medical Science, 13-1 Takara-machi, Kanazawa, 920-8641, Japan
| | - Masayuki Numata
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Sachio Fushida
- Department of Gastroenterological Surgery, Kanazawa University Graduate School of Medical Science, 13-1 Takara-machi, Kanazawa, 920-8641, Japan.
| |
Collapse
|
24
|
Todorov G, Cunha C. Hypothesis: Regulation of neuroplasticity may involve I-motif and G-quadruplex DNA formation modulated by epigenetic mechanisms. Med Hypotheses 2019; 127:129-135. [PMID: 31088636 DOI: 10.1016/j.mehy.2019.04.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 04/04/2019] [Accepted: 04/08/2019] [Indexed: 01/12/2023]
Abstract
Recent studies demonstrated the existence in vivo of various functional DNA structures that differ from the double helix. The G-quadruplex (G4) and intercalated motif (I-motif or IM) DNA structures are formed as knots where, correspondingly, guanines or cytosines on the same strand of DNA bind to each other. There are grounds to believe that G4 and IM sequences play a significant role in regulating gene expression considering their tendency to be found in or near regulatory sites (such as promoters, enhancers, and telomeres) as well as the correlation between the prevalence of G4 or IM conformations and specific phases of cell cycle. Notably, G4 and IM capable sequences tend to be found on the opposite strands of the same DNA site with at most one of the two structures formed at any given time. The recent evidence that K+, Mg2+ concentrations directly affect IM formation (and likely G4 formation indirectly) lead us to believe that these structures may play a major role in synaptic plasticity of neurons, and, therefore, in a variety of central nervous system (CNS) functions including memory, learning, habitual behaviors, pain perception and others. Furthermore, epigenetic mechanisms, which have an important role in synaptic plasticity and memory formation, were also shown to influence formation and stability of G4s and IMs. Our hypothesis is that non-canonical DNA and RNA structures could be an integral part of neuroplasticity control via gene expression regulation at the level of transcription, translation and splicing. We propose that the regulatory activity of DNA IM and G4 structures is modulated by DNA methylation/demethylation of the IM and/or G4 sequences, which facilitates the switch between canonical and non-canonical conformation. Other neuronal mechanisms interacting with the formation and regulatory activity of non-canonical DNA and RNA structures, particularly G4, IM and triplexes, may involve microRNAs as well as ion and proton fluxes. We are proposing experiments in acute brain slices and in vivo to test our hypothesis. The proposed studies would provide new insights into fundamental neuronal mechanisms in health and disease and potentially open new avenues for treating mental health disorders.
Collapse
Affiliation(s)
- German Todorov
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, NY, USA
| | - Catarina Cunha
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, NY, USA.
| |
Collapse
|
25
|
Clark AM, Ponniah K, Warden MS, Raitt EM, Yawn AC, Pascal SM. pH-Induced Folding of the Caspase-Cleaved Par-4 Tumor Suppressor: Evidence of Structure Outside of the Coiled Coil Domain. Biomolecules 2018; 8:biom8040162. [PMID: 30518159 PMCID: PMC6316887 DOI: 10.3390/biom8040162] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/20/2018] [Accepted: 11/22/2018] [Indexed: 12/13/2022] Open
Abstract
Prostate apoptosis response-4 (Par-4) is a 38 kDa largely intrinsically disordered tumor suppressor protein that functions in cancer cell apoptosis. Par-4 down-regulation is often observed in cancer while up-regulation is characteristic of neurodegenerative conditions such as Alzheimer’s disease. Cleavage of Par-4 by caspase-3 activates tumor suppression via formation of an approximately 25 kDa fragment (cl-Par-4) that enters the nucleus and inhibits Bcl-2 and NF-ƙB, which function in pro-survival pathways. Here, we have investigated the structure of cl-Par-4 using biophysical techniques including circular dichroism (CD) spectroscopy, dynamic light scattering (DLS), and intrinsic tyrosine fluorescence. The results demonstrate pH-dependent folding of cl-Par-4, with high disorder and aggregation at neutral pH, but a largely folded, non-aggregated conformation at acidic pH.
Collapse
Affiliation(s)
- Andrea M Clark
- Department of Chemistry and Biochemistry, Old Dominion University, Norfolk, VA 23529, USA.
| | - Komala Ponniah
- Department of Chemistry and Biochemistry, Old Dominion University, Norfolk, VA 23529, USA.
| | - Meghan S Warden
- Department of Chemistry and Biochemistry, Old Dominion University, Norfolk, VA 23529, USA.
| | - Emily M Raitt
- Department of Chemistry and Biochemistry, Old Dominion University, Norfolk, VA 23529, USA.
| | - Andrea C Yawn
- Department of Chemistry and Biochemistry, Old Dominion University, Norfolk, VA 23529, USA.
| | - Steven M Pascal
- Department of Chemistry and Biochemistry, Old Dominion University, Norfolk, VA 23529, USA.
| |
Collapse
|
26
|
Petukh M, Zhulin IB. Comparative study of the effect of disease causing and benign mutations in position Q92 on cholesterol binding by the NPC1 n-terminal domain. Proteins 2018; 86:1165-1175. [PMID: 30183109 DOI: 10.1002/prot.25597] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 08/27/2018] [Accepted: 08/31/2018] [Indexed: 11/06/2022]
Abstract
The Niemann-Pick type C1 (NPC1) protein is a large transmembrane protein located in lysosomes/endosomes. NPC1 binds cholesterol (CLR) and transports it to cellular membrane and endoplasmic reticulum. Mutations in NPC1 cause Niemann-Pick type C (NPC) disease, a rare autosomal disorder characterized by intracellular accumulations of CLR and subsequent neurodegeneration leading to premature death. Among known disease-causing mutations in NPC1, Q92R is the one that is located in the N-terminal cholesterol-binding domain [NTD]. Here we study the effect of the mutation on the ability of NPC1 (NTD) to bind and retain CLR in the binding pocket using structural analysis. We compare characteristics of the Q92R and Q92S mutant type (MT) protein, which is predicted to be benign. We provide detailed investigation of the CLR-NPC1 (NTD) binding process; and propose the mechanism, by which Q92R mutation causes NPC disease. We show that although Q92 residue neither directly participates in catalytic activity of the NPC1 (NTD), nor defines its CLR-binding specificity - it is important for the overall protein structure as well as for providing favorable electrostatic environment for CLR transfer. Our results suggest that a negative electrostatic potential of the CLR binding site (the S-opening) might promote NPC2 interaction with NPC1 (NTD) and/or proper CLR orientation and its enforced transfer. We show that in contrast to the benign Q92S mutation, Q92R significantly reduces electrostatic potential around S-opening, and thus likely affects NPC1 (NTD)-NPC2 interaction and/or CLR transfer from NPC2 to NPC1.
Collapse
Affiliation(s)
- Marharyta Petukh
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee.,Computational Sciences and Engineering Division, Oak Ridge National Laboratory, Oak Ridge, Tennessee
| | - Igor B Zhulin
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee.,Computational Sciences and Engineering Division, Oak Ridge National Laboratory, Oak Ridge, Tennessee.,Center for Bioinformatics, Pavlov First Saint Petersburg State Medical University, Saint Petersburg, Russia
| |
Collapse
|
27
|
Soto E, Ortega-Ramírez A, Vega R. Protons as Messengers of Intercellular Communication in the Nervous System. Front Cell Neurosci 2018; 12:342. [PMID: 30364044 PMCID: PMC6191491 DOI: 10.3389/fncel.2018.00342] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 09/14/2018] [Indexed: 12/18/2022] Open
Abstract
In this review, evidence demonstrating that protons (H+) constitute a complex, regulated intercellular signaling mechanisms are presented. Given that pH is a strictly regulated variable in multicellular organisms, localized extracellular pH changes may constitute significant signals of cellular processes that occur in a cell or a group of cells. Several studies have demonstrated that the low pH of synaptic vesicles implies that neurotransmitter release is always accompanied by the co-release of H+ into the synaptic cleft, leading to transient extracellular pH shifts. Also, evidence has accumulated indicating that extracellular H+ concentration regulation is complex and implies a source of protons in a network of transporters, ion exchangers, and buffer capacity of the media that may finally establish the extracellular proton concentration. The activation of membrane transporters, increased production of CO2 and of metabolites, such as lactate, produce significant extracellular pH shifts in nano- and micro-domains in the central nervous system (CNS), constituting a reliable signal for intercellular communication. The acid sensing ion channels (ASIC) function as specific signal sensors of proton signaling mechanism, detecting subtle variations of extracellular H+ in a range varying from pH 5 to 8. The main question in relation to this signaling system is whether it is only synaptically restricted, or a volume modulator of neuron excitability. This signaling system may have evolved from a metabolic activity detection mechanism to a highly localized extracellular proton dependent communication mechanism. In this study, evidence showing the mechanisms of regulation of extracellular pH shifts and of the ASICs and its function in modulating the excitability in various systems is reviewed, including data and its role in synaptic neurotransmission, volume transmission and even segregated neurotransmission, leading to a reliable extracellular signaling mechanism.
Collapse
Affiliation(s)
- Enrique Soto
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | | | - Rosario Vega
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| |
Collapse
|
28
|
Bonnet U, Bingmann D, Speckmann EJ, Wiemann M. Aging is associated with a mild acidification in neocortical human neurons in vitro. J Neural Transm (Vienna) 2018; 125:1495-1501. [PMID: 29995171 DOI: 10.1007/s00702-018-1904-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 07/06/2018] [Indexed: 11/30/2022]
Abstract
The intracellular pH (pHi) in the cytosol of mammalian central neurons is tightly regulated and small pHi-fluctuations are deemed to modulate inter-/intracellular signaling, excitability, and synaptic plasticity. The resting pHi of young rodent hippocampal pyramidal neurons is known to decrease alongside aging for about 0.1 pH-units. There is no information about the relationship between age and pHi of human central neurons. We addressed this knowledge gap using 26 neocortical slices from 12 patients (1-56-years-old) who had undergone epilepsy surgery. For fluorometric recordings, the slice-neurons were loaded with the pHi-sensitive dye BCECF-AM. We found that the pyramidal cells' resting pHi (n = 26) descended linearly alongside aging (r = - 0.71, p < 0.001). This negative relationship persisted, when the sample was confined to specific brain regions (i.e., middle temporal gyrus, 23 neurons, r = - 0.68, p < 0.001) or pathologies (i.e., hippocampus sclerosis, 8 neurons, r = - 0.78, p = 0.02). Specifically, neurons (n = 9, pHi 7.25 ± 0.12) from young children (1.5 ± 0.46-years-old) were significantly more alkaline than neurons from adults (n = 17, 38.53 ± 12.38 years old, pHi 7.08 ± 0.07, p < 0.001). Although the samples were from patients with different pathologies the results were in line with those from the rodent hippocampal pyramidal neurons. Like a hormetin, the age-related mild pHi-decrease might contribute to neuroprotection, e.g., via limiting excitotoxicity. On the other hand, aging cortical neurons could become more vulnerable to metabolic overstress by a successive pHi-decrease. Certainly, its impact for the dynamics in short and long-term synaptic plasticity and, ultimately, learning and memory provides a challenge for further research.
Collapse
Affiliation(s)
- Udo Bonnet
- Department of Psychiatry, Psychotherapy, and Psychosomatic Medicine, Evangelisches Krankenhaus Castrop-Rauxel, Academic Teaching Hospital of the University Duisburg-Essen, Castrop-Rauxel, Germany. .,Department of Psychiatry and Psychotherapy, Faculty of Medicine, LVR-Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| | - Dieter Bingmann
- Institute of Physiology, University of Duisburg-Essen, Essen, Germany
| | | | - Martin Wiemann
- Institute of Physiology, University of Duisburg-Essen, Essen, Germany.,IBE R&D gGmbH, Institute for Lung Health, 48149, Münster, Germany
| |
Collapse
|
29
|
Chen X, Wang X, Tang L, Wang J, Shen C, Liu J, Lu S, Zhang H, Kuang Y, Fei J, Wang Z. Nhe5 deficiency enhances learning and memory via upregulating Bdnf/TrkB signaling in mice. Am J Med Genet B Neuropsychiatr Genet 2017; 174:828-838. [PMID: 28981195 DOI: 10.1002/ajmg.b.32600] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 07/10/2017] [Accepted: 09/05/2017] [Indexed: 11/11/2022]
Abstract
Nhe5, a Na+ /H+ exchanger, is predominantly expressed in brain tissue and is proposed to act as a negative regulator of dendritic spine growth. Up to now, its physiological function in vivo remains unclear. Here we show that Nhe5-deficient mice exhibit markedly enhanced learning and memory in Morris water maze, novel object recognition, and passive avoidance task. Meanwhile, the pre- and post-synaptic components, synaptophysin (Syn) and post-synaptic density 95 (PSD95) expression levels were found increased in hippocampal regions lacking of Nhe5, suggesting a possible alterations in neuronal synaptic structure and function in Nhe5-/- mice. Further study reveals that Nhe5 deficiency leads to higher Bdnf expression levels, followed by increased phosphorylated TrkB and PLCγ levels, indicating that Bdnf/TrkB signaling is activated due to Nhe5 deficiency. Moreover, the corresponding brain regions of Nhe5-/- mice display elevated ERK/CaMKII/CREB phosphorylation levels. Taken together, these findings uncover a novel physiological function of Nhe5 in regulating learning and memory, further implying Nhe5 as a potential therapeutic target for improving cognition.
Collapse
Affiliation(s)
- Xuejiao Chen
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine of Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China.,Shanghai Research Center for Model Organisms, Shanghai, China
| | - Xiyi Wang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine of Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China.,Shanghai Research Center for Model Organisms, Shanghai, China
| | - Lingyun Tang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine of Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Jinjin Wang
- Shanghai Research Center for Model Organisms, Shanghai, China
| | - Chunling Shen
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine of Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China.,Shanghai Research Center for Model Organisms, Shanghai, China
| | - Jianbing Liu
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine of Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China.,Shanghai Research Center for Model Organisms, Shanghai, China
| | - Shunyuan Lu
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine of Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Hongxin Zhang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine of Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Ying Kuang
- Shanghai Research Center for Model Organisms, Shanghai, China
| | - Jian Fei
- Shanghai Research Center for Model Organisms, Shanghai, China
| | - Zhugang Wang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine of Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China.,Shanghai Research Center for Model Organisms, Shanghai, China.,Department of Medical Genetics, E-Institutes of Shanghai Universities, SJTUSM, Shanghai, China
| |
Collapse
|
30
|
High-Affinity Functional Fluorescent Ligands for Human β-Adrenoceptors. Sci Rep 2017; 7:12319. [PMID: 28951558 PMCID: PMC5614969 DOI: 10.1038/s41598-017-12468-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 09/08/2017] [Indexed: 01/14/2023] Open
Abstract
Visualization of the G-protein coupled receptor (GPCR) is of great importance for studying its function in a native cell. We have synthesized a series of red-emitting fluorescent probes targeting β-adrenergic receptor (βAR) that are compatible with confocal and Stimulated Emission Depletion (STED) microscopy as well as with Time-Resolved Fluorescence Resonance Energy Transfer (TR-FRET) binding assay in living cells. The probe based on the agonist BI-167107 and fluorescent dye KK114 demonstrates nanomolar binding affinity and up to nine-fold β2AR selectivity over β1AR. Carazolol-derived probes are fluorogenic and allow no-wash imaging experiments. STED microscopy of β2ARs stained at the native expression level on pancreatic CAPAN cells provides two-fold improvement in lateral optical resolution over confocal mode and reveals the formation of receptor microdomains. These probes retain their functional (agonist or antagonist) properties, allowing simultaneous modulation of cyclic adenosine monophosphate (cAMP) levels and receptor internalization as well as imaging receptor localization.
Collapse
|
31
|
Chiacchiaretta M, Latifi S, Bramini M, Fadda M, Fassio A, Benfenati F, Cesca F. Neuronal hyperactivity causes Na +/H + exchanger-induced extracellular acidification at active synapses. J Cell Sci 2017; 130:1435-1449. [PMID: 28254883 DOI: 10.1242/jcs.198564] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 02/28/2017] [Indexed: 12/12/2022] Open
Abstract
Extracellular pH impacts on neuronal activity, which is in turn an important determinant of extracellular H+ concentration. The aim of this study was to describe the spatio-temporal dynamics of extracellular pH at synaptic sites during neuronal hyperexcitability. To address this issue we created ex.E2GFP, a membrane-targeted extracellular ratiometric pH indicator that is exquisitely sensitive to acidic shifts. By monitoring ex.E2GFP fluorescence in real time in primary cortical neurons, we were able to quantify pH fluctuations during network hyperexcitability induced by convulsant drugs or high-frequency electrical stimulation. Sustained hyperactivity caused a pH decrease that was reversible upon silencing of neuronal activity and located at active synapses. This acidic shift was not attributable to the outflow of synaptic vesicle H+ into the cleft nor to the activity of membrane-exposed H+ V-ATPase, but rather to the activity of the Na+/H+-exchanger. Our data demonstrate that extracellular synaptic pH shifts take place during epileptic-like activity of neural cultures, emphasizing the strict links existing between synaptic activity and synaptic pH. This evidence may contribute to the understanding of the physio-pathological mechanisms associated with hyperexcitability in the epileptic brain.
Collapse
Affiliation(s)
- Martina Chiacchiaretta
- Center for Synaptic Neuroscience and Technology, Fondazione Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, Genova 16132, Italy.,Department of Experimental Medicine, University of Genova, Viale Benedetto XV 3, Genova 16132, Italy
| | - Shahrzad Latifi
- Center for Synaptic Neuroscience and Technology, Fondazione Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, Genova 16132, Italy
| | - Mattia Bramini
- Center for Synaptic Neuroscience and Technology, Fondazione Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, Genova 16132, Italy
| | - Manuela Fadda
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV 3, Genova 16132, Italy
| | - Anna Fassio
- Center for Synaptic Neuroscience and Technology, Fondazione Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, Genova 16132, Italy.,Department of Experimental Medicine, University of Genova, Viale Benedetto XV 3, Genova 16132, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Fondazione Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, Genova 16132, Italy.,Department of Experimental Medicine, University of Genova, Viale Benedetto XV 3, Genova 16132, Italy
| | - Fabrizia Cesca
- Center for Synaptic Neuroscience and Technology, Fondazione Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, Genova 16132, Italy
| |
Collapse
|
32
|
González-Garrido A, Vega R, Mercado F, López IA, Soto E. Acid-Sensing Ion Channels Expression, Identity and Role in the Excitability of the Cochlear Afferent Neurons. Front Cell Neurosci 2015; 9:483. [PMID: 26733809 PMCID: PMC4686812 DOI: 10.3389/fncel.2015.00483] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 11/30/2015] [Indexed: 12/21/2022] Open
Abstract
Acid-sensing ion channels (ASICs) are activated by an increase in the extracellular proton concentration. There are four genes (ASIC1-4) that encode six subunits, and they are involved in diverse neuronal functions, such as mechanosensation, learning and memory, nociception, and modulation of retinal function. In this study, we characterize the ASIC currents of spiral ganglion neurons (SGNs). These ASIC currents are primarily carried by Na(+), exhibit fast activation and desensitization, display a pH50 of 6.2 and are blocked by amiloride, indicating that these are ASIC currents. The ASIC currents were further characterized using several pharmacological tools. Gadolinium and acetylsalicylic acid reduced these currents, and FMRFamide, zinc (at high concentrations) and N,N,N',N'-tetrakis-(2-piridilmetil)-ethylenediamine increased them, indicating that functional ASICs are composed of the subunits ASIC1, ASIC2, and ASIC3. Neomycin and streptomycin reduced the desensitization rate of the ASIC current in SGNs, indicating that ASICs may contribute to the ototoxic action of aminoglycosides. RT-PCR of the spiral ganglion revealed significant expression of all ASIC subunits. By immunohistochemistry the expression of the ASIC1a, ASIC2a, ASIC2b, and ASIC3 subunits was detected in SGNs. Although only a few SGNs exhibited action potential firing in response to an acidic stimulus, protons in the extracellular solution modulated SGN activity during sinusoidal stimulation. Our results show that protons modulate the excitability of SGNs via ASICs.
Collapse
Affiliation(s)
| | - Rosario Vega
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla Puebla, Mexico
| | - Francisco Mercado
- Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz México D.F., Mexico
| | - Iván A López
- Department of Head and Neck Surgery, David Geffen School of Medicine, University of California Los Angeles, CA, USA
| | - Enrique Soto
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla Puebla, Mexico
| |
Collapse
|
33
|
Hu YB, Dammer EB, Ren RJ, Wang G. The endosomal-lysosomal system: from acidification and cargo sorting to neurodegeneration. Transl Neurodegener 2015; 4:18. [PMID: 26448863 PMCID: PMC4596472 DOI: 10.1186/s40035-015-0041-1] [Citation(s) in RCA: 403] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 09/22/2015] [Indexed: 12/13/2022] Open
Abstract
The endosomal-lysosomal system is made up of a set of intracellular membranous compartments that dynamically interconvert, which is comprised of early endosomes, recycling endosomes, late endosomes, and the lysosome. In addition, autophagosomes execute autophagy, which delivers intracellular contents to the lysosome. Maturation of endosomes and/or autophagosomes into a lysosome creates an unique acidic environment within the cell for proteolysis and recycling of unneeded cellular components into usable amino acids and other biomolecular building blocks. In the endocytic pathway, gradual maturation of endosomes into a lysosome and acidification of the late endosome are accompanied by vesicle trafficking, protein sorting and targeted degradation of some sorted cargo. Two opposing sorting systems are operating in these processes: the endosomal sorting complex required for transport (ESCRT) supports targeted degradation and the retromer supports retrograde retrieval of certain cargo. The endosomal-lysosomal system is emerging as a central player in a host of neurodegenerative diseases, demonstrating potential roles which are likely to be revealed in pathogenesis and for viable therapeutic strategies. Here we focus on the physiological process of endosomal-lysosomal maturation, acidification and sorting systems along the endocytic pathway, and further discuss relationships between abnormalities in the endosomal-lysosomal system and neurodegenerative diseases, especially Alzheimer’s disease (AD).
Collapse
Affiliation(s)
- Yong-Bo Hu
- Department of Neurology & Neuroscience Institute, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Eric B Dammer
- Department of Biochemistry, Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Ru-Jing Ren
- Department of Neurology & Neuroscience Institute, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Gang Wang
- Department of Neurology & Neuroscience Institute, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| |
Collapse
|
34
|
The cellular and molecular progression of mitochondrial dysfunction induced by 2,4-dinitrophenol in developing zebrafish embryos. Differentiation 2015; 89:51-69. [PMID: 25771346 DOI: 10.1016/j.diff.2015.01.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 01/22/2015] [Accepted: 01/27/2015] [Indexed: 12/18/2022]
Abstract
The etiology of mitochondrial disease is poorly understood. Furthermore, treatment options are limited, and diagnostic methods often lack the sensitivity to detect disease in its early stages. Disrupted oxidative phosphorylation (OXPHOS) that inhibits ATP production is a common phenotype of mitochondrial disorders that can be induced in zebrafish by exposure to 2,4-dinitrophenol (DNP), a FDA-banned weight-loss agent and EPA-regulated environmental toxicant, traditionally used in research labs as an uncoupler of OXPHOS. Despite the DNP-induced OXPHOS inhibition we observed using in vivo respirometry, the development of the DNP-treated and control zebrafish were largely similar during the first half of embryogenesis. During this period, DNP-treated embryos induced gene expression of mitochondrial and nuclear genes that stimulated the production of new mitochondria and increased glycolysis to yield normal levels of ATP. DNP-treated embryos were incapable of sustaining this mitochondrial biogenic response past mid-embryogenesis, as shown by significantly lowered ATP production and ATP levels, decreased gene expression, and the onset of developmental defects. Examining neural tissues commonly affected by mitochondrial disease, we found that DNP exposure also inhibited motor neuron axon arbor outgrowth and the proper formation of the retina. We observed and quantified the molecular and physiological progression of mitochondrial dysfunction during development with this new model of OXPHOS dysfunction, which has great potential for use in diagnostics and therapies for mitochondrial disease.
Collapse
|
35
|
Zheng JC, Tham CT, Keatings K, Fan S, Liou AYC, Numata Y, Allan D, Numata M. Secretory Carrier Membrane Protein (SCAMP) deficiency influences behavior of adult flies. Front Cell Dev Biol 2014; 2:64. [PMID: 25478561 PMCID: PMC4235465 DOI: 10.3389/fcell.2014.00064] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 10/17/2014] [Indexed: 12/21/2022] Open
Abstract
Secretory Carrier Membrane Proteins (SCAMPs) are a group of tetraspanning integral membrane proteins evolutionarily conserved from insects to mammals and plants. Mammalian genomes contain five SCAMP genes SCAMP1-SCAMP5 that regulate membrane dynamics, most prominently membrane-depolarization and Ca2+-induced regulated secretion, a key mechanism for neuronal and neuroendocrine signaling. However, the biological role of SCAMPs has remained poorly understood primarily owing to the lack of appropriate model organisms and behavior assays. Here we generate Drosophila Scamp null mutants and show that they exhibit reduced lifespan and behavioral abnormalities including impaired climbing, deficiency in odor associated long-term memory, and a susceptibility to heat-induced seizures. Neuron-specific restoration of Drosophila Scamp rescues all Scamp null behavioral phenotypes, indicating that the phenotypes are due to loss of neuronal Scamp. Remarkably, neuronal expression of human SCAMP genes rescues selected behavioral phenotypes of the mutants, suggesting the conserved function of SCAMPs across species. The newly developed Drosophila mutants present the first evidence that genetic depletion of SCAMP at the organismal level leads to varied behavioral abnormalities, and the obtained results indicate the importance of membrane dynamics in neuronal functions in vivo.
Collapse
Affiliation(s)
- JiaLin C Zheng
- Department of Biochemistry and Molecular Biology, University of British Columbia Vancouver, BC, Canada
| | - Chook Teng Tham
- Department of Biochemistry and Molecular Biology, University of British Columbia Vancouver, BC, Canada
| | - Kathleen Keatings
- Department of Biochemistry and Molecular Biology, University of British Columbia Vancouver, BC, Canada
| | - Steven Fan
- Department of Biochemistry and Molecular Biology, University of British Columbia Vancouver, BC, Canada
| | - Angela Yen-Chun Liou
- Department of Biochemistry and Molecular Biology, University of British Columbia Vancouver, BC, Canada
| | - Yuka Numata
- Department of Biochemistry and Molecular Biology, University of British Columbia Vancouver, BC, Canada
| | - Douglas Allan
- Department of Cellular and Physiological Sciences, University of British Columbia Vancouver, BC, Canada
| | - Masayuki Numata
- Department of Biochemistry and Molecular Biology, University of British Columbia Vancouver, BC, Canada
| |
Collapse
|